WO2003062206A2 - Small molecule modulators of the 5-ht2a serotonin receptor useful for the prophylaxis and treatment of disorders related thereto - Google Patents

Small molecule modulators of the 5-ht2a serotonin receptor useful for the prophylaxis and treatment of disorders related thereto Download PDF

Info

Publication number
WO2003062206A2
WO2003062206A2 PCT/US2003/002059 US0302059W WO03062206A2 WO 2003062206 A2 WO2003062206 A2 WO 2003062206A2 US 0302059 W US0302059 W US 0302059W WO 03062206 A2 WO03062206 A2 WO 03062206A2
Authority
WO
WIPO (PCT)
Prior art keywords
phenyl
methyl
bromo
pyrazol
urea
Prior art date
Application number
PCT/US2003/002059
Other languages
French (fr)
Other versions
WO2003062206A3 (en
Inventor
Bradley Teegarden
Keith Drouet
Honnappa Jayakumar
William Thomsen
Paul Maffuid
Katie Elwell
Richard Foster
Michael Lawless
Qian Liu
Julian Smith
Konrad Feichtinger
Robert C. Glen
Nigel R. A. BEELELY
Original Assignee
Arena Pharmaceuticals, 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 Arena Pharmaceuticals, Inc. filed Critical Arena Pharmaceuticals, Inc.
Priority to EP03705889A priority Critical patent/EP1509505A2/en
Publication of WO2003062206A2 publication Critical patent/WO2003062206A2/en
Publication of WO2003062206A3 publication Critical patent/WO2003062206A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/02Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings
    • C07D231/10Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D231/12Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/02Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings
    • C07D231/10Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D231/14Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/02Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings
    • C07D231/10Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D231/14Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D231/16Halogen atoms or nitro radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/04Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond

Definitions

  • the present invention relates to certain pyrazole derivatives of Formula (I) and pharmaceutical compositions thereof that modulate the activity of the 5-HT 2A serotonin receptor.
  • Compounds and pharmaceutical compositions thereof are directed to methods useful in the prophylaxis or treatment of reducing platelet aggreagation, coronary artery disease, myocardial infarction, transient ischemic attack, angina, stroke, atrial fibrillation, reducing the risk of blood clot formation, asthma or symptoms thereof, agitation or a symptom, behavioral disorders, drug induced psychosis, excitative psychosis, Gilles de la Tourette's syndrome, manic disorder, organic or NOS psychosis, psychotic disorder, psychosis, acute schizophrenia, chronic schizophrenia and NOS schizophrenia and related disorders.
  • the present invention also relates to the method of prophylaxis or treatment of 5-HT 2A serotonin receptor mediated disorders in combination with a dopamine D2 receptor antagonist such as haloperidol, administered separately or together.
  • a dopamine D2 receptor antagonist such as haloperidol
  • G protein-coupled receptors share a common structural motif. All these receptors have seven sequences of between 22 to 24 hydrophobic amino acids that form seven alpha helices, each of which spans the membrane. The transmembrane helices are joined by strands of amino acids having a larger loop between the fourth and fifth transmembrane helix on the extracellular side ofthe membrane. Another larger loop, composed pi ⁇ rnarily of hydrophilic amino acids, joins transmembrane helices five and six on the intracellular side of the membrane. The carboxy tenninus of the receptor lies intracellularly with the amino teiminus in the extracellular space.
  • G protein-coupled receptors exist in the cell membrane in equiUbrium between two different states or conformations: an "inactive" state and an "active" state. As shown schematically in Figure 2, a receptor in an inactive state is unable to link to the intracellular transduction pathway to produce a biological response. Changing the receptor conformation to the active state allows linkage to the transduction pathway and produces a biological response.
  • a receptor may be stabilized in an active state by an endogenous ligand or an exogenous agonist ligand.
  • Recent discoveries such as, including but not exclusively limited to, modifications to the amino acid sequence of the receptor provide means other than ligands to stabilize the active state conformation. These means effectively stabilize the receptor in an active state by simulating the effect of a ligand binding to the receptor. Stabilization by such ligand-independent means is termed "constitutive receptor activation.”
  • Serotouin receptors [0004] Receptors for serotonin (5-hyc_oxyrrypt_mine, 5-HT) are an important class of G protein-coupled receptors. Serotonin is thought to play a role in processes related to learning and memory, sleep, thermoregulation, mood, motor activity, pain, sexual and aggressive behaviors, appetite, neurodegenerative regulation, and biological rhythms. Not surprisingly, serotonin is linked to pathophysiological conditions such as anxiety, depression, obsessive-compulsive disorders, schizophrenia, suicide, autism, migraine, emesis, alcoholism, and neurodegenerative disorders.
  • these types of therapeutics can generally be divided into two classes, the "typical” and the “atypical.” Both have anti- psychotic effects, but the typicals also include concomitant motor-related side effects (extra pyramidal syndromes, e.g., lip-smacking, tongue darting, locomotor movement, etc). Such side effects are thought to be associated with the compounds interacting with other receptors, such as the human dopamine D2 receptor in the nigro-striatal pathway. Therefore, an atypical treatment is preferred. Haloperidol is considered a typical anti-psychotic, and clozapine is considered an atypical anti-psychotic.
  • Serotonin receptors are divided into seven subfamilies, referred to as 5-HT1 through
  • 5-HT7 inclusive. These subfamilies are further divided into subtypes.
  • the 5-HT2 subfamily is divided into three receptor subtypes: 5-HT 2A , 5-HT 2B , and 5-HT 2 c.
  • the human 5-HT 2 c receptor was first isolated and cloned in 1987, and the human 5-HT 2A receptor was first isolated and cloned in 1990. These two receptors are thought to be the site of action of hallucinogenic drugs.
  • antagonists to the 5-HT 2A and 5-HT 2C receptors are believed to be useful in treating depression, anxiety, psychosis, and eating disorders. [0006] U.S.
  • Patent Number 4,985,352 describes the isolation, characterization, and expression of a functional cDNA clone encoding the entire human 5-HT1C receptor (now known as the 5-HT 2C receptor).
  • U.S. Patent Number 5,661,012 describes the isolation, characterization, and expression of a functional cDNA clone encoding the entire human 5-HT 2A receptor.
  • Casey describes a mutation of the cysteine residue at position 322 of the rat 5-HT 2A receptor to lysine (C322K), glutamine (C322Q), and arginine (C322R) which reportedly led to constitutive activation.
  • Herrick-Davis 1 and Herrick-Davis 2 describe mutations of the serine residue at position 312 of the rat 5-HT 2C receptor to phenylalanine (S312F) and lysine (S312K), which reportedly led to constitutive activation.
  • the present invention relates to non-endogenous, constitutively activated forms ofthe human 5-HT 2A and human 5-HT 2C receptors and various uses of such receptors. Further disclosed are small molecule modulators of these receptors. Most preferably, these modulators have inverse agonist characteristics at the receptor.
  • the present invention discloses nucleic acid molecules and the proteins for three non-endogenous, constitutively activated human serotonin receptors, referred to herein as, AP-1, AP-3, and AP-4.
  • the AP-1 receptor is a constitutively active form of the human 5- HT 2 c receptor created by an S31 OK point mutation.
  • the AP-3 receptor is a constitutively active form ofthe human 5-HT2A receptor whereby the intracellular loop 3 (IC3) portion and the cytoplasmic-tail portion of the endogenous human 5-HT2A receptor have been replaced with the IC3 portion and the cytoplasmic-tail portion of the human 5-HT 2C receptor.
  • the AP-4 receptor is a constitutively active form ofthe human 5-HT2A receptor whereby (1) the region ofthe intracellular third loop between the proline of the transmembrane 5 region (TM5) and the proline of TM6 of the endogenous human 5- HT 2A receptor has been replaced with the corresponding region of the human 5-HT 2C receptor (including a S310K point mutation); and (2) the cytoplasmic-tail portion ofthe endogenous human 5- HT 2A receptor has been replaced with the cytoplasmic-tail portion of the endogenous human 5-HT 2C receptor.
  • the invention also provides assays that may be used to directly identify candidate compounds as agonists, partial agonists or inverse agonists to non-endogenous, constitutively activated human serotonin receptors; such candidate compounds can then be utilized in pharmaceutical composition(s) for treatment of diseases and disorders which are related to the human 5-HT 2A and/or human 5-HT 2C receptors.
  • the invention also provides compounds that exhibit high selectivity 5-HT 2A activity.
  • Ri is H, halogens, NR 5 R 6 , OH or OR 7 , wherein
  • R 5 and R_ are independently H, or C 1-6 alkyl, or C 2 . 6 alkenyl, or cycloalkyl, or aryl, or CH 2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from CF 3 , CC1 3 , Me, N0 2 , OH, OMe, OEt, CONR_R 9 , NR_R 9 , NHCOCH 3 , OCF 3 , SMe, COOR 10 , S0 3 R 8 , SO_NR_R 9 , COMe, COEt, CO-lower alkyl, SCF 3 , CN, C 2 .
  • R 5 and Re may form part of a 5, 6 or 7 membered cyclic structure which may be either saturated or unsaturated and that may contain up to four heteroatoms selected from O, N or S and said cyclic structure may be optionally substituted by up to four substituents in any position independently selected from CF 3 , CC1 3 , Me, N0 2 , OH, OMe, OEt, OCF 3 , SMe, COOR 10 , S0 2 NR_R 9 , SO 3 R 10 , NHCOCH 3 , COEt, COMe, or halogen;
  • R 8 and R 9 are independently a H, or C ⁇ _ 6 alkyl, or C 2 . 6 alkenyl, or cycloalkyl, or aryl, or CH 2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from halogen, CF 3 , OCF 3 ,
  • Rio is H or C ⁇ -6 alkyl;
  • R 7 is H or C 1-6 alkyl;
  • R 2 is H, straight chain or branched C 1-6 alkyl, C 2-6 alkenyl, or cycloalkyl;
  • R 3 is halogen, carboxy, CN, alkoxycarbonyl, straight chain or branched C ⁇ _ 6 alkyl, C 2- 6 alkenyl, C 2 . 6 alkynyl, cycloalkyl, aryl or heteroaryl, wherein each of said alkoxycarbonyl, straight chain or branched C ⁇ _ 6 alkyl, C 2 .
  • 6 alkenyl, C 2 _ 6 alkynyl, cycloalkyl, aryl or heteroaryl groups can be optionally substituted with up to four substituents in any position selected from OH, ORio, NR 8 R 9 , halogen, -C(p) 3j or -0-C(p) 3 where p is halogen, and said cycloalkyl, aryl or heteroaryl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position;
  • R 4 is C ⁇ - 6 alkyl, C 2 . ⁇ alkenyl, or cycloalkyl;
  • Rn and R ⁇ 2 are each independently H, straight chain or branched C ⁇ . 6 alkyl, C 2-6 alkenyl, or cycloalkyl;
  • R- 1 6 and R 17 are each independently H, halogen, CN, NR 8 R 9 , COOR 10 , SR 10 , straight chain or branched C . . . alkyl, C 2 . 6 alkenyl, C 2-6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl, wherein each of said alkoxycarbonyl, straight chain or branched C ⁇ _ 6 alkyl, C 2 . 6 alkenyl, C 2 .
  • 6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl groups can be optionally substituted with up to four substituents in any position selected from N0 2 , OR 10 , NR_R 9 , halogen, -C(p) 3> or -0-C(p) 3 where p is halogen, and said cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position;
  • L 2 is -0-Q 2 wherein Q 2 is straight chain or branched C ⁇ . 6 alkyl, C 2 . 6 alkenyl, C 2-6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl, wherein each of said cycloalkyl, aryl, alkylaryl, or arylalkyl groups can be optionally substituted with up to four substituents in any position selected from N0 2 , OR 7 , halogen, -C(p) 3 ⁇ or -0-C(p) 3 where p is halogen, or an aliphatic or aromatic heterocycle, and said cycloalkyl, aryl, alkylaryl, and arylalkyl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position; or a pharmaceutically acceptable salt.
  • bold typeface indicates the location of the mutation in the non- endogenous, constitutively activated receptor relative to the corresponding endogenous receptor.
  • Figure 1 shows a generalized structure of a G protein-coupled receptor with the numbers assigned to the transmembrane helices, the intracellular loops, and the extracellular loops.
  • Figure 2 schematically shows the active and inactive states for a typical G protein- coupled receptor and the linkage ofthe active state to the second messenger transduction pathway.
  • Figure 3a provides the nucleic acid sequence of the endogenous human 5-HT 2A receptor (SEQ.TD.NO.:22).
  • Figure 3b provides the corresponding amino acid sequence ofthe endogenous human 5-HT2A receptor (SEQ._D.NO.:23).
  • Figure 4a provides the nucleic acid sequence of the endogenous human 5-HT 2C receptor (SEQ.ID.NO.:24).
  • Figure 4b provides the corresponding amino acid sequence ofthe endogenous human
  • 5-HT 2C receptor SEQ.ID.NO.:25.
  • Figure 5a provides the nucleic acid sequence of a constitutively active form of the human 5-HT 2C receptor ("AP-1 cDNA" - SEQ.ID.NO.:26).
  • Figure 5b provides the corresponding amino acid sequence ofthe AP-1 cDNA ("AP-
  • Figure 6a provides the nucleic acid sequence of a constitutively active form of the human 5-HT 2A receptor whereby the IC3 portion and the cytoplasmic-tail portion of the endogenous 5-HT 2A receptor have been replaced with the IC3 portion and the cytoplasmic-tail portion of the human 5-HT 2C receptor ("AP-3 cDNA" - SEQ.ID.NO.:28).
  • Figure 6b provides the corresponding amino acid sequence ofthe AP-3 cDNA CAP ⁇
  • Figure 6c provides a schematic representation of AP-3, where die dashed-lines represent the portion obtained from the human 5-HT 2C receptor.
  • Figure 7a provides the nucleic acid sequence of a constitutively active form of the human 5-HT 2A receptor whereby (1) the region o the between the proline of TM5 and the proline of TM6 of the endogenous human 5-HT 2A receptor has been replaced with the corresponding region of the human 5-HT 2C receptor (including a S310K point mutation); and (2) the cytoplasmic-tail portion of the endogenous 5-HT 2A receptor has been replaced with the cytoplasmic-tail portion of the endogenous human 5-HT 2C receptor ("AP-4 cDNA" - SEQ.ED.NO.:30).
  • Figure 7b provides the corresponding amino acid sequence ofthe AP-4 cDNA ("AP-
  • Figure 7c provides a schematic representation of the mutated 5-HT 2A receptor of
  • Figure 8 is a representation of the preferred vector, pCMV, used herein.
  • Figure 9 is a diagram illustrating (1) enhanced ( 35 S)GTP ⁇ S binding to membranes prepared from COS cells expressing the endogenous human 5-HT 2C receptor in response to serotonin, and (2) inhibition by mianserin using wheatgerm agglutinin scintillation proximity beads.
  • the concentration of ( 35 S)GTP">S was held constant at 0.3 nM, and the concentration of GDP was held at 1 ⁇ M.
  • the concentration ofthe membrane protein was 12.5 ⁇ g.
  • Figure 10 is a diagram showing serotonin stimulation of ( 35 S)GTP ⁇ S binding to membranes expressing AP-1 receptors in 293T cells and the inhibition by 30 ⁇ M mianserin on WallacTM scintistrips.
  • FIGs 11A and UB are diagrams showing the effects of protein concentration on ( 35 S)GTP-yS binding in membranes prepared from 293T cells transfected with the endogenous human 5-HT 2C receptors and AP-1 receptors compared to cells transfected with the control vector (pCMV) alone in the absence ( Figure 11 A) and presence ( Figure 11B) of 10 ⁇ M serotonin.
  • the radiolableled concentration of ( 35 S)GTP' S was held constant at 0.3 nM, and the GDP concentration was held constant at 1 ⁇ M.
  • the assay was perfonned on 96-well format on WallacTM scintistrips.
  • Figure 12 provides bar-graph comparisons of inositol tris-phosphate ("IP3") production between the endogenous human 5-HT 2A receptor and AP-2, a mutated form of the receptor.
  • IP3 inositol tris-phosphate
  • Figure 13 provides bar-graph comparisons of inositol tris-phosphate ("IP3") production between the endogenous human 5-HT2A receptor and AP-4, a mutated form of the receptor.
  • IP3 inositol tris-phosphate
  • Figure 14 provides bar graph comparisons of IP3 production between the endogenous human 5-HT2A receptor and AP-3, a mutated form ofthe receptor.
  • Figure 15 provides bar-graph comparisons of IP3 production between the endogenous human 5-HT 2C receptor and AP-1.
  • Figures 16A-C provides representative autoradiograms showing displacement of I 125 -
  • Figures 17A-C show in vivo response of animals to Compound 2 exposure.
  • AGONISTS shall mean moieties that activate the intracellular response when they bind to the receptor, or enhance GTP binding to membranes.
  • AMINO ACID ABBREVIATIONS used herein are set out in TABLE 1 :
  • PARTIAL AGONISTS shall mean moieties which activate the intracellular response when they bind to the receptor to a lesser degree/extent than do agonists, or enhance GTP binding to membranes to a lesser degree/extent than do agonists.
  • ANTAGONIST shall mean moieties that competitively bind to the receptor at the same site as the agonists but which do not activate the intracellular response initiated by the active form of the receptor, and can thereby inhibit the intracellular responses by agonists or partial agonists. ANTAGONISTS do not diminish the baseline intracellular response in the absence of an agonist or partial agonist.
  • CANDIDATE COMPOUND shall mean a molecule (for example, and not limitation, a chemical compound) which is amenable to a screening technique.
  • COMPOSITION shall mean a material comprising at least two compounds or two components; for example, and not limitation, a Pharmaceutical Composition is a Composition.
  • COMPOUND EFFICACY shall mean a measurement of the ability of a compound to inhibit or stimulate receptor functionality, as opposed to receptor binding affinity.
  • CONSTITUTIVELY ACTIVATED RECEPTOR shall mean a receptor subject to constitutive receptor activation.
  • CONSTITUTIVE RECEPTOR ACTIVATION shall mean stabilization of a receptor in the active state by means other than binding of the receptor with its endogenous ligand or a chemical equivalent thereof.
  • CONTACT or CONTACTING shall mean bringing at least two moieties together, whether in an in vitro system or an in vivo system.
  • ENDOGENOUS shall mean a material that a mammal naturally produces.
  • ENDOGENOUS in reference to, for example and not limitation, the term "receptor” shall mean that which is naturally produced by a mammal (for example, and not limitation, a human) or a virus.
  • NON-ENDOGENOUS in this context shall mean that which is not naturally produced by a mammal (for example, and not limitation, a human) or a virus.
  • a receptor which is not constitutively active in its endogenous form, but when manipulated becomes constitutively active, is most preferably referred to herein as a "non-endogenous, constitutively activated receptor.”
  • Both terms can be utilized to describe both "in vivo" and "in vitro" systems.
  • the endogenous or non-endogenous receptor may be in reference to an in vitro screening system.
  • screening of a candidate compound by means of an in vivo system is viable.
  • INHIBIT or INHIBITING in relationship to the term “response” shall mean that a response is decreased or prevented in the presence of a compound as opposed to in the absence of the compound.
  • INVERSE AGONISTS shall mean moieties that bind the endogenous form of the receptor or to the constitutively activated form ofthe receptor, and which inhibit the baseline intracellular response initiated by the active form of the receptor below the normal base level of activity which is observed in the absence of agonists or partial agonists, or decrease GTP binding to membranes.
  • the baseline intracellular response is inhibited in the presence ofthe inverse agonist by at least 30%, more preferably by at least 50%, and most preferably by at least 75%, as compared with the baseline response in the absence ofthe inverse agonist.
  • LIGAND shall mean an endogenous, naturally occurring molecule specific for an endogenous, naturally occurring receptor.
  • MODULATE or MODULATING shall mean to refer to an increase or decrease in the amount, quality, response or effect of a particular activity, function or molecule.
  • Compounds which modulate/capable of modulating the 5-HT 2A activity include agonists, inverse agonists, antagonists, inhibitors, activators, and compounds which directly or indirectly affect regulation ofthe 5-HT 2A activity.
  • PHARMACEUTICAL COMPOSITION shall mean a composition comprising at least one active ingredient, whereby the composition is amenable to investigation for a specified, efficacious outcome in a mammal (for example, and not limitation, a human).
  • a mammal for example, and not limitation, a human.
  • STIMULATE or STIMULATING in relationship to the term “response” shall mean that a response is increased in the presence of a compound as opposed to in the absence ofthe compound.
  • the compounds disclosed herein are believed to be useful in the treatment of several additional diseases and disorders, and in the amelioration of symptoms thereof. Without limitation, these include the following:
  • Antiplatelet agents are prescribed for a variety of conditions. For example, in coronary artery disease they are used to help prevent myocardial infarction or stroke in patients who are at risk of developing obstructive blood clots (e.g., coronary thrombosis).
  • a myocardial infarction heart attack
  • the heart muscle does not receive enough oxygen-rich blood as a result of a blockage in the coronary blood vessels. If taken while an attack is in progress or ⁇ nmediately afterward (preferably within 30 minutes), antiplatelets can reduce the damage to the heart.
  • a transient ischemic attack (“TIA” or "mini-stroke”) is a brief imterruption of oxygen flow to the brain due to decreased blood flow through arteries, usually due to an obstructing blood clot.
  • Antiplatelet drugs have been found to be effective in preventing TIAs.
  • Angina is a temporary and often recurring chest pain, pressure or discomfort caused by inadequate oxygen-rich blood flow (ischemia) to some parts of the heart.
  • ischemia oxygen-rich blood flow
  • antiplatelet therapy can reduce the effects of angina and the risk of myocardial infarction.
  • Stroke is an event in which the brain does not receive enough oxygen-rich blood, usually due to blockage of a cerebral blood vessel by a blood clot. In high-risk patients, taking antiplatelets regularly has been found to prevent the formation blood clots that cause first or second strokes.
  • Angioplasty is a catheter based technique used to open arteries obstructed by a blood clot. Whether or not stenting is performed immediately after this procedure to keep the artery open, antiplatelets can reduce the risk of forming additional blood clots following the procedure(s).
  • Coronary bypass surgery is a surgical procedure in which an artery or vein is taken from elsewhere in the body and grafted to a blocked coronary artery, rerouting blood around the blockage and through the newly attached vessel. After the procedure, antiplatelets can reduce the risk of secondary blood clots.
  • Atrial fibrillation is the most common type of sustained irregular heart rhythm
  • Atrial fibrillation affects about two million Americans every year. In atrial fibrillation, the atria (the heart's upper chambers) rapidly fire electrical signals that cause them to quiver rather than contract normally. The result is an abnormally fast and highly irregular heartbeat. When given after an episode of atrial fibrillation, antiplatelets can reduce the risk of blood clots forming in the heart and traveling to the brain (embolism).
  • 5-HT 2A receptors are expressed on smooth muscle of blood vessels and 5-HT secreted by activated platelets causes vasoconstriction as well as activation of additional platelets during clotting.
  • 5-HT 2A inverse agonist will inhibit platelet aggregation and thus be a potential treatment as an antiplatelet therapy. See Satimura, K, et al., Clin Cardiol 2002 Jan. 25 (l):28-32; and Wilson, H.C et al., Thromb Haemost 1991 Sep 2;66(3):355-60.
  • the 5-HT 2A inverse agonists disclosed herein provide beneficial improvement in microcirculation to patients in need of antiplatelet therapy by antagonizing the vasoconstrictive products of the aggregating platelets in, for example and not limitation, the indications described above. Accordingly, in some embodiments, the present invention provides methods for reducing platelet aggregation in a patient in need thereof comprising administering to said patient a composition comprising a 5-HT 2A inverse agonist disclosed herein.
  • the present invention provides methods for treating coronary artery disease, myocardial infarction, transient ischemic attack, angina, stroke, atrial fibrillation, or a symptom of any of the foregoing in a patient in need of said treatment, comprising administering to said patient a composition comprising a 5-HT 2A inverse agonist disclosed herein.
  • the present invention provides methods for reducing risk of blood clot formation in a angioplasty or coronary bypass surgery patient, or a patient suffering from atrial fibrillation, comprising administering to a said patient a composition comprising a 5-HT 2A inverse agonist disclosed herein at a time where such risk exists.
  • the present invention provides methods for treating asthma in a patient in need of said treatment, comprising administering to said patient a composition comprising a 5-HT 2A inverse agonist disclosed herein.
  • methods for treating a symptom of asthma in a patient in need of said treatment comprising administering to said patient a composition comprising a 5-HT 2A inverse agonist disclosed herein.
  • Agitation is a well-recognized behavioral syndrome with a range of symptoms, including hostility, extreme excitement, poor impulse control, tension and uncooperativeness (See Cohen-Mansfield J, and Billig, N., (1986), Agitated Behaviors in the Elderly. I. A Conceptual Review. J Am Geriatr Soc 34(10): 711-721).
  • Alzheimer's disease is a common occurrence in the elderly and often associated with dementia such as those caused by Alzheimer's disease, Lewy Body, Parkinson's, and Huntington's, which are degenerative diseases of the nervous system and by diseases that affect blood vessels, such as stroke, or multi-infarct dementia, which is caused by multiple strokes in the brain can also induce dementia.
  • Alzheimer's disease accounts for approximately 50 to 70% of all dementias (See Koss E, et al., (1997), Assessing patterns of agitation in Alzheimer's disease patients with the Cohen-Mansfield Agitation Inventory. The Alzheimer's Disease Cooperative Study. Alzheimer Dis Assoc Disord ll(suppl 2):S45-S50).
  • Agitated behaviors can also be manifested in cognitively intact elderly people and by those with psychiatric disorders other than dementia
  • the present invention provides methods for treating agitation in a patient in need of such treatment comprising administering to said patient a composition comprising a 5-HT 2A inverse agonist disclosed herein.
  • the agitation is due to a psychiatric disorder other than dementia.
  • the present invention provides methods for treatment of agitation or a symptom thereof in a patient suffering from dementia comprising administering to said patient a composition comprising a 5-HT 2A inverse agonist disclosed herein.
  • the dementia is due to a degenerative disease of the nervous system, for example and without limitation, Alzheimers disease, Lewy Body, Parkinson's disease, and Huntington's disease, or dementia due to diseases that affect blood vessels, including with out limitation stroke and multi-infarct dementia.
  • methods are provided for treating agitation or a symptom thereof in a patient in need of such treatment, where the patient is a cognitively intact elderly patient, comprising administering to said patient a composition comprising a 5-HT 2A inverse agonist disclosed herein.
  • Schizophrenia is a psychopathic disorder of unknown origin, which usually appears for the first time in early adulthood and is marked by a number of characteristics, psychotic symptoms, progression, phasic development and deterioration in social behavior and professional capability in the region below the highest level ever attained.
  • Characteristic psychotic symptoms are disorders of thought content (multiple, fragmentary, incoherent, implausible or simply delusional contents or ideas of doctrine) and of mentality (loss of association, flight of imagination, incoherence up to incomprehensibility), as well as disorders of perceptibility (hallucinations), of emotions (superficial or inadequate emotions), of self-perception, of intentions and impulses, of interhuman relationships, and finally psychomotoric disorders (such as catatonia). Other symptoms are also associated with this disorder. (See, American Statistical and Diagnostic Handbook).
  • Haloperidol is a potent dopamine D2 receptor antagonist. It is widely prescribed for acute schizophrenic symptoms, and is very effective for the positive symptoms of schizophrenia. However, Haldol is not effective for the negative symptoms of schizophrenia and may actually induce negative symptoms as well as cognitive dysfunction. In accordance with some methods of the invention, adding a 5-HT 2A inverse agonist concomitantly with Haldol will provide benefits including the ability to use a lower dose of Haldol without losing its effects on positive symptoms, while reducing or eliminating its inductive effects on negative symptoms, and prolonging relapse to the patient's next schizophrenic event.
  • Haloperidol is used for treatment of a variety of behavioral disorders, drug induced psychosis, excitative psychosis, Gilles de la Tourette's syndrome, manic disorders, psychosis (organic and NOS), psychotic disorder, psychosis, schizophrenia (acute, chronic and NOS). Further uses include in the treatment of infantile autism, huntington's chorea, and nausea and vomiting from chemotherapy and chemotherapeutic antibodies. Administration of 5-HT 2A inverse agonists disclosed herein with haloperidol also will provide benefits in these indications.
  • the present invention provides methods for treating a behavioral disorder, drug induced psychosis, excitative psychosis, Gilles de la Tourette's syndrome, manic disorders, psychosis (organic and NOS), psychotic disorder, psychosis, schizophrenia (acute, chronic and NOS) comprising administering to said patient a dopamine D2 receptor antagonist and a 5-HT 2A inverse agonist disclosed herein.
  • the present invention provides methods for treating a behavioral disorder, drug induced psychosis, excitative psychosis, Gilles de la Tourette's syndrome, manic disorders, psychosis (organic and NOS), psychotic disorder, psychosis, schizophrenia (acute, chronic and NOS) comprising administering to said patient haloperidol and a 5-HT 2A inverse agonist disclosed herein.
  • the present invention provides methods for treating infantile autism, huntington's chorea, or nausea and vomiting from chemotherapy or chemotherapeutic antibodies comprising administering to said patient a dopamine D2 receptor antagonist and a 5-HT 2A inverse agonist disclosed herein.
  • the present invention provides methods for treating infantile autism, huntington's chorea, or nausea and vomiting from chemotherapy or chemotherapeutic antibodies comprising administering to said patient haloperidol and a 5-HT 2A inverse agonist disclosed herein.
  • the present invention provides methods for treating schizophrenia in a patient in need of said treatment comprising administering to said patient a dopamine D2 receptor antagonist and a 5-HT 2A inverse agonist disclosed herein.
  • the dopamine D2 receptor antagonist is haloperidol.
  • the administration of the dopamine D2 receptor antagonist can be concomitant with administration ofthe 5-HT 2A inverse agonist, or they can be administered at different times. Those of skill in the art will easily be able to determine appropriate dosing regimes for the most efficacious reduction or elimination of deleterions haloperidol effects.
  • haloperidol and the 5-HT 2A inverse agonist are administered in a single dosage form, and in other embodiments, they are administered in separate dosage forms.
  • the present invention further provides methods of alleviating negative symptoms of schizophrenia induced by the administration of haloperidol to a patient suffering from said schizophrenia, comprising administering to said patient a 5-HT 2A inverse agonist as disclosed herein.
  • sequence information regarding the non-endogenous, constitutively active human 5-HT 2A and 5-HT 2 c receptors are referred to by identifiers as set forth in TABLE 2:
  • AP-2 a mutation analogous to that reported by Casey (C322K) was utilized in the human 5-HT 2A receptor and is referred to herein as AP-2.
  • AP-2 did not lead to sufficient constitutive activation to allow for utilization in screening techniques.
  • Herrick-Davis 1 or Herrick-Davis 2 is of limited predictive value relative to the manipulation ofthe human 5-HT 2 c receptor. Consequently, the ability to make reasonable predictions about the effects of mutations to the rat 5-HT receptors vis-a- vis the corresponding human receptors is not possible. Nonetheless, this unfortunate lack of reasonable predictability provides the opportunity for others to discover mutations to the human 5-HT receptors that provide evidence of constitutive activation.
  • the present invention is based upon the desire of defining mutated sequences ofthe human serotonin receptors 5-HT 2A and 5-HT 2C whereby such mutated versions ofthe expressed receptor are constitutively active. These constitutively active receptors allow for, inter alia, screening candidate compounds.
  • G protein receptor When a G protein receptor becomes constitutively active, it binds to a G protein (Gq, Gs,
  • candidate compounds are identified using the "generic" G protein-coupled receptor assay (i.e. an assay to select compounds that are agonists, partial agonists, or inverse agonists), further screening to confirm that the compounds have interacted at the receptor site is preferred.
  • a compound identified by the "generic” assay may not bind to the receptor, but may instead merely "uncouple" the G protein from the intracellular domain.
  • Lysergic acid diethylamide is a well-known agonist ofthe 5-HT 2A and 5-HT 2C receptors, while mesulergine is a well-known antagonist to the 5-HT 2C receptor. Accordingly, in most preferred embodiments, an agonist (LSD) and/or antagonist (mesulergine) competitive binding assay(s) is used to further screen those compounds selected from the "generic" assay for confirmation of serotonin receptor binding.
  • IP3 accumulation can be used to confirm constitutive activation of these types of Gq coupled receptors (see Herrick-Davis-1).
  • IP3 accumulation assays can be used to further screen those compounds selected from an agonist and/or antagonist competitive binding assay. .
  • Candidate compounds selected for further development can be formulated into pharmaceutical compositions using techniques well known to those in the art. Suitable pharmaceutically- acceptable carriers are available to those in the art; for example, see Remington's Pharmaceutical Sciences, 16 th Edition, 1980, Mack Publishing Co., (Oslo et al., eds.).
  • PCR was performed using either TaqPlusTM precision polymerase (Stratagene) or rTthTM polymerase (Perkin Elmer) with the buffer systems provided by the manufacturers, 0.25 ⁇ M of each primer, and 0.2 mM of each ofthe four (4) nucleotides.
  • the cycle condition was 30 cycles of 94°C for 1 minute, 57 °C for 1 minute and 72 °C for 2 minutes.
  • the 1.5 kb PCR fragment was digested with Xho I and Xba I and subcloned into the Sal I-Xba I site of pBluescript.
  • the cDNA containing a S310K mutation (AP-1 cDNA) in the third intracellular loop , of the human 5-HT 2C receptor was constructed by replacing the Sty I restriction fragment containing amino acid 310 with synthetic double stranded oligonucleotides encoding the desired mutation.
  • the sense strand sequence utilized had the following sequence:
  • the cDNA encoding endogenous human 5-HT 2A receptor was obtained by RT-PCR using human brain poly-A + RNA; a 5 ' primer from the 5 ' untranslated region with a Xho I restriction site:
  • the resulting cDNA clones were fully sequenced and found to encode two amino acid changes from the published sequences.
  • the first change is a T25N mutation in the N-terminal extracellular domain and the second change is an H452Y mutation.
  • These mutations are likely to represent sequence polymorphisms rather than PCR errors since the cDNA clones having the same two mutations were derived from two independent PCR procedures using Taq polymerase from two different commercial sources (TaqPlusTM Stratagene and rTthTM Perkin Elmer).
  • the cDNA containing the point mutation C322K in the third intracellular loop was constructed by using the Sph I restriction enzyme site, which encompasses amino acid 322.
  • a primer containing the C322K mutation: 5'-CAAAGAAAGTACTGGGCATCGTCTTCTTCCT-3' (SEQ.ro.NO.:7) was used along with the primer from the 3' untranslated region set forth above as SEQ.LD.NO.:6.
  • the resulting PCR fragment was then used to replace the 3' end ofthe wild type 5-HT 2A cDNA by the T4 polymerase blunted Sph I site.
  • PCR was performed using pfu polymerase (Stratagene) with the buffer system provided by the manufacturer and 10% DMSO, 0.25 mM of each primer, 0.5mM of each of the 4 nucleotides.
  • the cycle conditions were 25 cycles of 94°C for 1 minute, 60°C for 1 minute, and 72°C for 1 minute.
  • the human 5-HT 2A cDNA with intracellular loop 3 (IC3) or IC3 and cytoplasmic tail replaced by the corresponding human 5-HT 2C cDNA was constructed using PCR-based mutagenesis.
  • (a) Replacement of IC3 Loop [0104] The IC3 loop of human 5-HT 2A cDNA was first replaced with the corresponding human 5-HT 2C cDNA. Two separate PCR procedures were performed to generate the two fragments, Fragment A and Fragment B, that fuse the 5-HT 2C IC3 loop to the transmembrane 6 (TM6) of 5-HT 2A .
  • the 237 bp PCR fragment, Fragment A, containing 5-HT 2C IC3 and the initial 13 bp of 5-HT 2A TM6 was amplified by using the following primers:
  • Second round PCR was performed using Fragment A and Fragment B as co-templates with SEQ.ID.NO.:8 and SEQ.ID.NO.:ll (it is noted that the sequences for SEQ.ID.NOS.:6 and 11 are the same) as primers.
  • the resulting 740 bp PCR fragment, Fragment C contained the IC3 loop of human 5-HT 2C fused to TM6 through the end of the cytoplasmic tail of human 5-HT 2A .
  • PCR was performed using pfuTM polymerase (Stratagene) with the buffer system provided by the manufacturer, and 10% DMSO, 0.25 mM of each primer, and 0.5 mM of each ofthe four (4) nucleotides.
  • the cycle conditions were 25 cycles of 94 °C for 1 minute, 57 °C (1st round PCR) or 60 °C (2nd round PCR) for 1 minute, and 72 °C for 1 minute (1st round PCR) or 90 seconds (2nd round PCR). [0107] To generate a PCR fragment containing a fusion junction between the human 5-HT 2A
  • TM5 and the IC3 loop of 5-HT 2C four (4) primers were used.
  • the two external primers, derived from human 5-HT 2A had the following sequences: 5'-CGTGTCTCTCCTTACTTCA-3' (SEQ.ID.NO.:12)
  • the first internal primer utilized was an antisense strand containing the initial 13 bp of IC3 of 5-HT 2C followed by the terminal 23 bp derived from TM5 of 5-HT 2A : 5'-TCGGCGCAGTACTTTGATAGTTAGAAAGTAGGTGAT-3' (SEQ.ID.NO.:13)
  • the second internal primer was a sense strand containing the te ⁇ ninal 14 bp derived from TM5 of 5-HT 2A followed by the initial 24 bp derived from IC3 of 5-HT 2C :
  • PCR was performed using endogenous human 5-HT 2A and a co-template, Fragment
  • PCR was performed using a sense primer containing the C-te ⁇ ninal 22 bp of TM7 of endogenous human 5-HT 2A followed by the initial 21 bp ofthe cytoplasmic tail of endogenous human 5-HT 2C : 5'-TTCAGCAGTCAACCCACTAGTCTATACTCTGTTCAACAAAATT-3 ' (SEQ.LO.NO.:15)
  • the antisense primer was derived from the 3 ' untranslated region of endogenous human 5-HT 2C : 5'-ATTTCTAGACATATGTAGCTTGTACCGT-3' (SEQ.ID.NO.:16).
  • the resulting PCR fragment, Fragment D contained the last 22 bp of endogenous human 5-HT 2A TM7 fused to the cytoplasmic tail of endogenous human 5-HT 2C .
  • Second round PCR was performed using Fragment D and the co-template was endogenous human 5-HT 2A that was previously digested with Ace I to avoid undesired amplification.
  • the antisense primer used was SEQ.ID.NO.:16 (the sequences for SEQ.LD.NOS.:16 and 2 are the same) and the sense primer used was derived from endogenous human 5-HT 2A : 5 '-ATCACCTACTTTCTAACTA-3 ' (SEQ.ID.NO.:17).
  • PCR conditions were as set forth in Example lB3(a) for the first round PCR, except that the annealing temperature was 48 °C and the extension time was 90 seconds.
  • the resulting 710 bp PCR product was digested with Apa I and Xba I and used to replace the corresponding Apa I-Xba I fragment of either (a) endogenous human 5-HT 2A , or (b) 5-HT 2A with 2C IC3 to generate (a) endogenous human 5-HT 2A with endogenous human 5-HT 2 c cytoplasmic tail and (b) AP-3, respectively.
  • This mutant was created by replacement ofthe region of endogenous human 5-HT 2A from amino acid 247, the middle of TM5 right after Pro 246 , to amino acid 337, the middle of TM6 just before Pro 338 , by the corresponding region of AP-1 cDNA.
  • the junction in TM5 is referred to as the "2 A-2C junction”
  • the junction in TM6 is referred to as the "2C-2A junction.”
  • PCR fragments containing the desired hybrid junctions were generated.
  • the 5' fragment of 561 bp containing the 2A-2C junction in TM5 was generated by PCR using endogenous human 5-HT 2A as template, SEQ.EO.NO.:12 as the sense primer, and the antisense primer was derived from 13 bp of 5-HT 2C followed by 20 bp of 5-HT 2A sequence: 5'-CCATAATCGTCAGGGGAATGAAAAATGACACAA-3' (SEQ.ED.NO.:18)
  • the middle fragment of the 323 bp contains endogenous human 5-HT 2C sequence derived from the middle of TM5 to the middle of TM6, flanked by 13 bp of 5-HT 2A sequences from the 2A-2C junction and the 2C-2A junction.
  • This middle fragment was generated by using AP-1 cDNA as a template, a sense primer containing 13 bp of 5-HT2A followed by 20 bp of 5-HT 2C sequences across the 2A-2C junction and having the sequence:
  • the 3' fragment of 487 bp containing the 2C-2A junction was generated by PCR using endogenous human 5-HT 2A as a template and a sense primer having the following sequence from the 2C-2A junction: 5'-GATCATGTGGTGCCCTTTCTTCATCACAAACAT-3' (SEQ.ED.NO.:21) and the antisense primer was SEQ.ID.NO.:6 (see note above regarding SEQ.ID.NOS.:6 and 11).
  • the M3' PCR used the middle and M3' PCR fragment described above as the co- template, SEQ.ID.NO.:19 as the sense primer and SEQ.ED.NO.:6 (see note above regarding SEQ.ID.NOS.:6 and 11) as the antisense primer, and generated a 784 bp amplification product.
  • the final round of PCR was performed using the 857 bp and 784 bp fragments from the second round PCR as the co-template, and SEQ.ID.NO.:12 and SEQ.ID.NO.:6 (see note above regarding SEQ._O.NOS.:6 and 11) as the sense and the antisense primer, respectively.
  • the 1.32 kb amplification product from the final round of PCR was digested with Pst I and Eco RI. Then resulting 1 kb Pst I-Eco RI fragment was used to replace the corresponding fragment ofthe endogenous human 5-HT 2A to generate mutant 5-HT 2A with 5-HT 2C : C310K/IC3.
  • the Apa I-Xba fragment of AP3 was used to replace the corresponding fragment in mutant 5-HT 2A with 5-HT 2C : C310K/IC3 to generate AP4.
  • the vector utilized be pCMV.
  • This vector was deposited with the American Type Culture Collection (ATCC) on October 13, 1998 (10801 University Boulevard., Manassas, VA 20110-2209 USA) under the provisions ofthe Budapest Treaty for the International Recognition ofthe Deposit of Microorganisms for the Purpose of Patent Procedure. The DNA was tested by the ATCC and determined to be viable. The ATCC has assigned the following deposit number to pCMV: ATCC #203351. See Figure 8.
  • tube A was prepared by mixing 20 ⁇ g DNA (e.g., pCMV vector; pCMV vector AP-1 cDNA, etc.) in 1.2 ml serum free DMEM (Irvine Scientific, Irvine, CA); tube B was prepared by rnixing 120 ⁇ l hpofectarnine (Gibco BRL) in 1.2 ml serum free DMEM. Tubes A and B were then admixed by inversions (several times), followed by incubation at room temperature for 30-45 min. The admixture is referred to as the "transfection mixture".
  • Plated COS-7 cells were washed with IX PBS, followed by addition of 10 ml serum free DMEM. 2.4 ml of the transfection mixture was then added to the cells, followed by incubation for 4 hrs at 37°C/5% C0 2 . The transfection mixture was then removed by aspiration, followed by the addition of 25 ml of DMEM/10% Fetal Bovine Serum. Cells were then incubated at 37°C/5% C0 2 . After 72 hr incubation, cells were then harvested and utilized for analysis.
  • (a) ( 35 S)GTP ⁇ S binding is generically applicable to all G protein-coupled receptors; and (b) ( 35 S)GTP ⁇ S binding is proximal at the membrane surface, thereby making it less likely to pick-up molecules which affect the intracellular cascade.
  • the assay utilizes the ability of G protein-coupled receptors to stimulate ( 35 S)GTP ⁇ S binding to membranes expressing the relevant receptors. Therefore, the assay may be used to directly screen compounds at the disclosed serotonin receptors.
  • Figure 9 demonstrates the utility of a scintillation proximity assay to monitor the binding of ( 35 S)GTP ⁇ S to membranes expressing, e.g., the endogenous human 5-HT 2 c receptor expressed in COS cells.
  • a preferred protocol for the assay is such that the assay was incubated in 20 mM HEPES, pH 7.4, binding buffer with 0.3 nM ( 35 S)GTP ⁇ S and 12.5 ⁇ g membrane protein and 1 ⁇ M GDP for 30 minutes.
  • Wheatgerm agglutinin beads 25 ⁇ l; Amersham
  • the tubes were then centrifuged at 1500 x g for 5 minutes at room temperature and then counted in a scintillation counter.
  • serotonin which as the endogenous ligand activates the 5-HT 2C receptor, stimulated ( 35 S)GTP ⁇ S binding to the membranes in a concentration dependant manner.
  • the stimulated binding was completely inhibited by 30 ⁇ M mianserin, a compound considered as a classical 5-HT 2C antagonist, but also known as a 5-HT 2C inverse agonist.
  • this assay measures agonist-induced binding of ( 35 S)GTP ⁇ S to membranes and can be routinely used to measure constitutive activity of receptors
  • the present cost of wheatgerm agglutinin beads may be prohibitive.
  • a less costly but equally applicable alternative also meets the needs of large-scale screening.
  • Flash plates and WallacTM scintistrips may be used to format a high throughput ( 35 S)GTP ⁇ S binding assay. This technique allows one to monitor the tritiated ligand binding to the receptor while simultaneously monitoring the efficacy via ( 35 S)GTP ⁇ S binding. This is possible because the WallacTM beta counter can switch energy windows to analyze both tritium and 35 S-labeled probes.
  • this assay may be used for detecting of other types of membrane activation events that result in receptor activation.
  • the assay may be used to monitor 32 P phosphorylation of a variety of receptors (including G protein-coupled and tyrosine kinase receptors).
  • the bound ( 35 S)GTP ⁇ S or the 32 P- phosphorylated receptor will activate the scintillant coated on the wells.
  • Use of Scinti ® strips (WallacTM) demonstrate this principle.
  • this assay may be used for measuring ligand binding to receptors using radiolabeled ligands. In a similar manner, the radiolabeled bound ligand is centrifuged to the bottom of the well and activates the scintillant.
  • the ( 35 S)GTP ⁇ S assay results parallel the results obtained in traditional second messenger assays of receptors.
  • Figure 11A and Figure 11B demonstrate the enhanced binding of ( 35 S)GTP ⁇ S to membranes prepared from 293T cells expressing the control vector alone, the native human 5-HT 2 c receptor or the AP-1 receptor was observed (data not shown).
  • the total protein concentration used in the assay affects the total amount of ( 35 S)GTP ⁇ S binding for each receptor.
  • the c.p.m. differential between the CMV transfected and the constitutively active mutant receptor increased from approximately 1000 c.p.m at 10 ⁇ g/well to approximately 6-8000 c.p.m. at 75 ⁇ g/well protein concentration, as shown in Figure 11.
  • the AP-1 receptor showed the highest level of constitutive activation followed by the wild type receptor, which also showed enhanced ( 35 S)GTP ⁇ S binding above basal. This is consistent with the ability of the endogenous human 5-HT 2C receptor to accumulate intracellular IP3 in the absence of 5-HT stimulation (Example 5) and is also consistent with published data claiming that the endogenous human 5-HT 2C receptor has a high natural basal activity. Therefore, the AP-1 receptor demonstrates that constitutive activity may be measured by proximal ( 3S S)GTP ⁇ S binding events at the membrane interface.
  • Membranes were prepared from transfected COS-7 cells (see Example 2) by homogenization in 20 mM HEPES and 10 mM EDTA, pH 7.4 and centrifuged at 49,000 x g for 15 min. The pellet was resuspended in 20 mM HEPES and 0.1 mM EDTA, pH 7.4, homogenized for 10 sec. using a Polytron homogenizer (Brinkman) at 5000 rpm and centrifuged at 49,000 x g for 15 min. The final pellet was resuspended in 20 mM HEPES and 10 mM MgCl 2 , pH 7.4, homogenized for 10 sec. using polytron homogenizer (Brinkman) at 5000 rpm.
  • COS-7 and 293 Cells [0132] On day one, COS-7 cells or 293 cells were plated onto 24 well plates, usually lxl 0 5 cells/well or 2xl0 5 cells/well, respectively. On day two, the cells were transfected by first mixing 0.25 ug DNA (see Example 2) in 50 ⁇ l serum-free DMEM/well and then 2 ⁇ l lipofectamine in 50 ⁇ l serum-free DMEM/well.
  • transfection media The solutions (“transfection media”) were gentiy mixed and incubated for 15-30 minutes at room temperature. The cells were washed with 0.5 ml PBS and then 400 ⁇ l of serum free media was mixed with the transfection media and added to the cells. The cells were then incubated for 3-4 hours at 37°C/5%C0 2 . Then the transfection media was removed and replaced with 1 ml/well of regular growth media. On day 3, the media was removed and the cells were washed with 0.5 ml PBS.
  • tube A was prepared by mixing 20 ⁇ g DNA (e.g., pCMV vector; pCMV vector AP-1 cDNA, etc.) in 1.2 ml serum free DMEM (Irvine Scientific, Irvine, CA); tube B was prepared by mixing 120 ⁇ l lipofectamine (Gibco BRL) in 1.2 ml serum free DMEM. Tubes A and B were then admixed by inversions (several times), followed by incubation at room temperature for 30-45 min.
  • DNA e.g., pCMV vector; pCMV vector AP-1 cDNA, etc.
  • tube B was prepared by mixing 120 ⁇ l lipofectamine (Gibco BRL) in 1.2 ml serum free DMEM. Tubes A and B were then admixed by inversions (several times), followed by incubation at room temperature for 30-45 min.
  • the admixture is referred to as the "transfection mixture”.
  • Plated 293 cells were washed with 1XPBS, followed by addition of 10ml serum free DMEM.
  • 2.4 ml of the transfection mixture was then added to die cells, followed by incubation for 4 hrs at 37°C/5% C0 2 .
  • On day 3 cells were trypsinized and counted, followed by plating of lxlO 6 cells/well (poly D-lysine treated 12-well plates). Cells were permitted to adhere to the wells, followed by one wash with lxPBS. Thereafter, 0.5 ⁇ Ci 3 H-inositol in 1ml inositol-free DMEM was added per well. Protocol B.
  • the cells were washed with 0.5 ml PBS and then 0.45 ml of assay medium was added containing inositol-free/serum free media, 10 ⁇ M pargyline, 10 mM lithium chloride, or 0.4 ml of assay medium and 50 ⁇ l of lOx ketanserin (ket) to a final concentration of 10 ⁇ M.
  • the cells were then incubated for 30 minutes at 37° C. Then the cells were washed with 0.5 ml PBS and 200 ⁇ l of fresh/ice cold stop solution (IM KOH; 18 mM Na-borate; 3.8 mM EDTA) was added/well.
  • IM KOH fresh/ice cold stop solution
  • the solution was kept on ice for 5-10 minutes or until the cells were lysed and then neutralized by 200 ⁇ l of fresh/ice cold neutralization sol. (7.5 % HCL).
  • the lysate was then transferred into 1.5 ml rnicro- centrifuge tubes and 1 ml of chloroform/methanol (1:2) was added/tube.
  • the solution was vortexed for 15 seconds and the upper phase was applied to a Biorad AG1-X8 anion exchange resin ( 100-200 mesh). The resin was washed with water and 0.9 ml of the upper phase was loaded onto the column.
  • the column was washed with 10 ml of 5 mM myo-inositol and 10 ml of 5 mM Na-borate/60mM Na- formate.
  • the inositol trisphosphates were eluted into scintillation vials containing 10 ml of scintillation cocktail with 2 ml of 0.1 M formic acid 1 M ammonium formate.
  • the columns were regenerated by washing with 10 ml of 0.1 M formic acid/3M ammonium formate and rinsed twice with dd H 2 0 and stored at room temperature in water. Results are discussed below.
  • Figure 12 is an illustration of IP3 production from the human 5-HT 2A receptor which was mutated using the same point mutation as set forth in Casey, which rendered the rat receptor constitutively active.
  • the results represented in Figure 12 support die position that when the point mutation shown to activate the rat receptor is introduced into the human receptor, little activation of the receptor is obtained that would allow for appropriate screening of candidate compounds, with the response being only moderately above that of the endogenous human 5-HT 2A receptor. Generally, a response of at least 2X above that ofthe endogenous response is preferred.
  • Figure 13 provides an illustration comparing IP3 production from endogenous 5-HT 2A receptor and the AP4 mutation.
  • the results illustrated in Figure 13 support the position that when the novel mutation disclosed herein is utilized, a robust response of constitutive IP3 accumulation is obtained (e.g., over 2X that ofthe endogenous receptor).
  • Figure 14 provides an illustration of IP3 production from AP3.
  • the results illustrated in Figure 14 support the position fliat when the novel mutation disclosed herein is utilized, a robust response of constitutive IP3 accumulation is obtained.
  • Figure 15 provides bar-graph comparisons of IP3 accumulation between endogenous human 5-HT 2C receptor and AP-1. Note that the endogenous receptor has a high degree of natural constitutive activity relative to the control CMV transfected cells (i.e., the endogenous receptor appears to be constitutively activated).
  • transiently expressing constitutively active mutant human 5-HT 2C receptor AP-1 were incubated with binding buffer (20 mM HEPES, pH 7.4, 100 mM NaCl, 20 mM MgCl 2 ° 6H,0, 0.2% saponin, and 0.2 mM ascobate), GDP(1 ⁇ M) and compound in a 96-well plate format for a period of 60 rninutes at ambient room temperature. Plates were then centrifuged at 4,000 rpm for 15 minutes followed by aspiration of the reaction mixture and counting for 1 minute in a WallacTM MicroBeta plate scintillation counter.
  • Example 7 SCREENING OF SELECTED COMPOUNDS TO CONFIRM RECEPTOR BINDING: AP-1 [0141]
  • the candidate compounds identified from Example 7 were then screened using the assay protocol of Example 4 (mesulergine), using the AP-1 mutant receptor.
  • IC 50 (nM) values were determined; five of the nearly 120 compounds of Example 7 were determined to have potent binding affinity for the receptor. Results are summarized in TABLE 4. TABLE 4
  • the "primary" screen designed to directly identify human 5-HT 2A /5-HT 2C receptor inverse agonists consisted of a membrane-based GTP ⁇ S binding assay utilizing membranes prepared from COS7 cells transiently transfected with AP-1 human receptor.
  • Candidate compounds (lO ⁇ M final assay concentration) directly identified as inhibiting receptor-mediated increases in GTP ⁇ S binding by greater than 50-75% (arbitrary cut-off value) were considered active "hits”.
  • Primary assay hits were then re-tested in the same assay to recorifirm their inverse agonist activity.
  • This protocol is substantially the same as set forth above in Example 6.
  • Primary screening assays measuring GTP ⁇ S binding to membranes prepared from COS7 cells transiently transfected with human mutated 5-HT 2C receptor (AP-1) were used to directly identify inverse agonists in screening libraries (Tripos, Inc.).
  • Candidate compound screens were performed in a total assay volume of 200 ⁇ l using scintillant-coated Wallac ScintistripTM plates.
  • the primary assay was comprised of the following chemicals (at indicated final assay concentrations): 20 mM HEPES, pH 7.4, 100 mM NaCl, 20 mM MgCl 2 , 0.2% saponin, 0.2 mM ascorbic acid, 1 ⁇ M GDP, 0.3 nM GTP ⁇ 35 S, and 12.5 ⁇ g of the above defined membranes. Incubations were performed for 60 minutes at ambient room temperature. The binding assay incubation was temiinated by centrifugation of assay plates at 4,000 rpm for 15 minutes, followed by rapid aspiration ofthe reaction mixture and counting in a Wallac MicroBetaTM scintillation counter.
  • Radioligand binding competition experiments were performed in a total assay volume of 200 ⁇ l using standard 96-well microtiter plates.
  • the final assay ingredients consisted of assay buffer (20 mM HEPES and 10 mM MgCl 2 ), InM ( 3 H) mesulergine, and 50 ⁇ g of membranes (COS7 with AP-1 as defined above).
  • Nonspecific ( 3 H) mesulergine binding was defined in the presence of 100 ⁇ M mianserin. Incubations were performed for 1 hour at 37°C.
  • Receptor bound radioligand was resolved from free radioligand by rapid filtration ofthe assay mixture over a Wallac FiltermatTM Type B filter, followed by washing with ice-cold assay buffer using a SkatronTM cell harvester. Radioactivity was counted using a Wallac 1205 BetaPlateTM counter. Each assay plate contained five negative control wells (membranes expressing receptor and no candidate compound addition) and three positive control wells (each containing 100 ⁇ M mianserin). For one concentration tests, candidate compounds were diluted into assay buffer and screened at a final concentration of 10 ⁇ M, in triplicate. For IC 50 determinations, candidate compounds were diluted in assay buffer and eight different concentrations were evaluated, in triplicate. A total of 16 wells were designated for an eight concentration mianserin dose response curve evaluation for both assays.
  • Radioligand binding competition experiments were performed in a total assay volume of 200 ⁇ l using standard 96-well microtiter plates.
  • the final assay ingredients comprised assay buffer (20 mM HEPES and lOmM MgCl 2 ), InM ( 3 H)LSD, and 50 ⁇ g of the above-defined membranes (COS7 with AP-1).
  • Nonspecific ( 3 H)LSD binding was defined in the presence of 100 ⁇ M serotonin. Incubations were performed for 1 hour at 37° C.
  • Receptor bound radioligand was resolved from free radioligand by rapid filtration of the assay mixture over a Wallac FiltermatTM Type B filter, followed by washing with ice-cold assay buffer using a SkatronTM cell harvester. Radioactivity was counted using a Wallac 1205 BetaPlateTM counter. Each assay plate contained five negative control wells (membranes expressing receptor and no candidate compound addition) and three positive control wells (containing 100 ⁇ M mianserin). For one concentration tests, candidate compounds were diluted into assay buffer and screened at a final concentration of 10 ⁇ M in triplicate. For IC 50 determinations, candidate compounds were diluted in assay buffer and eight different concentrations were evaluated in triplicate. A total of 16 wells were designated for an eight concentration serotonin dose response curve evaluation for both assays. EXAMPLE 9D
  • Candidate compound identified in the assays of Examples 9A-9C were then evaluated for inositol phosphate accumulation, following the protocol of Example 5 (COS7 cells expressing human mutated 5-HT 2A receptor, AP-3), modified as follows: tube A was prepared by mixing 16 ⁇ g DNA (e.g., pCMV vector; pCMV vector AP-1 cDNA, etc.) in 1.0 ml serum free DMEM (Irvine Scientific, Irvine, CA); tube B was prepared by mixing 60 ⁇ l lipofectamine (Gibco BRL) in 1.0 ml serum free DMEM.
  • DNA e.g., pCMV vector; pCMV vector AP-1 cDNA, etc.
  • tube B was prepared by mixing 60 ⁇ l lipofectamine (Gibco BRL) in 1.0 ml serum free DMEM.
  • Tubes A and B were then admixed by inversions (several times), followed by incubation at room temperature for 30 min. The admixture is referred to as die "transfection mixture”. Plated 293 cells were washed with 10 ml Serum Free DMEM, followed by addition of 11 ml Serum Free DMEM. 2.0 ml ofthe transfection mixture was then added to the cells, followed by incubation for 5 hrs at 37° C/5% C0 2 . On day 3, cells were trypsinized and counted, followed by plating of lxlO 6 cells/well (12-well plates). Cells were permitted to adhere to the wells for 8 hrs, followed by one wash with lxPBS. Thereafter, 0.5 ⁇ Ci 3 H-inositol in 1ml inositol-free DMEM was added per well.
  • the cells were washed with 1.5 ml PBS and then 0.9 ml of assay medium was added containing inositol-free/serum free media, 10 ⁇ M pargyline, 10 mM lithium chloride, for 5min in 37°C/5% C0 2 followed by lOO ⁇ l addition of candidate compound diluted in the same material. The cells were then incubated for 120 minutes at 37° C. Then the cells were washed witii 1.5 ml PBS and 200 ⁇ l of fresh/icecold stop solution (IM KOH; 18 mM Na-borate; 3.8 mM EDTA) was added/well.
  • IM KOH fresh/icecold stop solution
  • the solution was kept on ice for 5-10 minutes or until the cells were lysed and then neutralized by 200 ⁇ l of fresh/ice cold neutralization sol. (7.5 % HCL).
  • the lysate was then transferred into 1.5 ml micro-centrifuge tubes and 1 ml of chloroform/methanol (1:2) was added/tube.
  • the solution was vortexed for 15 seconds and the upper phase was applied to a Biorad AG1-X8 anion exchange resin (100-200 mesh).
  • the resin was washed with water and 0.9 ml ofthe upper phase was loaded onto the column.
  • the column was washed with 10 ml of 5 mM myo-inositol and 10 ml of 5 mM Na-borate/60mM Na-formate.
  • the inositol trisphosphates were eluted into scintillation vials containing 10 ml of scintillation cocktail with 2 ml of 0.1 M formic acid/ 1 M ammonium formate.
  • the columns were regenerated by washing with 10 ml of 0.1 M formic acid/3M ammonium formate and rinsed twice with ddH 2 0 and stored at room temperature in water.
  • This series of compounds exhibits highly selective 5-HT 2A activity. Accordingly, in one aspect of die invention, a series of compounds possessing 5-HT 2A receptor activity that are useful as inverse agonists at such receptors is designated by the general Formula (I):
  • Ri is H, halogens, NR 5 R_, OH or OR 7 , wherein
  • R5 and Re are independently H, or C ⁇ _ 6 alkyl, or C 2-6 alkenyl, or cycloalkyl, or aryl, or CH 2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from CF 3 , CC1 3 , Me, N0 2 , OH, OMe, OEt, CONRgR . , NRsR . , NHCOCH 3 , OCF 3 , SMe, COOR 10 , S0 3 R 8 , S0 2 NR 8 R 9 , COMe, COEt, CO-lower alkyl, SCF 3 , CN, C 2 .
  • R 5 and R ⁇ may form part of a 5, 6 or 7 membered cyclic structure which may be either saturated or unsaturated and that may contain up to four heteroatoms selected from O, N or S and said cyclic structure may be optionally substituted by up to four substituents in any position independently selected from CF 3 , CC1 3 , Me, N0 2 , OH, OMe, OEt, OCF 3 , SMe, COOR 10 , S0 2 NR_R 9 , SO 3 R ⁇ 0 , NHCOCH 3 , COEt, COMe, or halogen;
  • R 8 and R 9 are independently a H, or C ⁇ _ 6 alkyl, or C 2 _ 6 alkenyl, or cycloalkyl, or aryl, or CH 2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from halogen, CF 3 , OCF 3 , OEt, CC1 3 ,
  • R 7 is H or C ⁇ alkyl; ii) R 2 is H, straight chain or branched C ⁇ __ alkyl, C 2-6 alkenyl, or cycloalkyl; iii) R 3 is halogen, carboxy, CN, alkoxycarbonyl, straight chain or branched C ⁇ -6 alkyl, C 2- ⁇ alkenyl, C 2 . 6 alkynyl, cycloalkyl, aryl or heteroaryl, wherein each of said alkoxycarbonyl, straight chain or branched C_._ alkyl, C 2 . 6 alkenyl, C 2 . 6 alkynyl, cycloalkyl, aryl or heteroaryl groups can be optionally substituted with up to four substituents in any position selected from OH, OR 10 , NRsRg, halogen,
  • R 4 is Cue alkyl, C 2-6 alkenyl, or cycloalkyl;
  • Rn and R ⁇ 2 are each independently H, straight chain or branched C ⁇ - 6 alkyl, C 2-6 alkenyl, or cycloalkyl;
  • R ⁇ 3 , R 14 , Ri5, Ri ⁇ and R are each independently H, halogen, CN,
  • NR 8 R 9 COORio, SRio, straight chain or branched C ⁇ -6 alkyl, C_. 6 alkenyl, C 2 . 6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl, wherein each of said alkoxycarbonyl, straight chain or branched C ⁇ _ 6 alkyl, C 2 . 6 alkenyl, C 2 .
  • 6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl groups can be optionally substituted with up to four substituents in any position selected from N0 2 , ORio, NR_R 9 , halogen, -C(p) 3 ⁇ or -0-C(p) 3 where p is halogen, and said cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl groups can alternatively or additionally be optionally substituted witii up to four alkyl substituents in any position;
  • L_ is -O-Q 2 wherein Q 2 is straight chain or branched C ⁇ -6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl, wherein each of said cycloalkyl, aryl, alkylaryl, or arylalkyl groups can be optionally substituted with up to four substituents in any position selected from N0 2 , OR .
  • An aryl moiety can be a 5 or 6 membered aromatic heterocyclic ring (containing up to 4 hetero atoms independently selected from N, O, or S) or a 6 membered aromatic non-heterocyclic ring or a polycycle.
  • C ⁇ -6 alkyl moieties can be straight chain or branched; optionally substituted C ⁇ _ 6 alkyl moieties can be straight chain or branched; C 2 . 6 alkenyl moieties can be straight chain or branched; and optionally substituted C 2 . 6 alkenyl moieties can be straight chain or branched.
  • suitable C ⁇ -6 alkyl groups include but are not limited to methyl, ethyl, n-propyl, i-propyl, n-butyl, and t-butyl.
  • Halogens are typically F, CI, Br, and I.
  • Examples of 5 or 6 membered ring moieties include, but are not restricted to, phenyl, furanyl, thienyl, imidazolyl, pyridyl, pyrrolyl, oxazolyl, isoxazolyl, triazolyl, pyrazolyl, tetrazolyl, thiazolyl and isothiazoyl.
  • Examples of polycycle moieties include, but are not restricted to, naphthyl, benzothiazolyl, benzofuranyl, benzimidazolyl, quinolyl, isoquinolyl, indolyl, quinoxalinyl, quinazolinyl and benzothienyl.
  • B is L b q is 1, m is 0, and n is 0.
  • B is L q is 1, m is 1, and n is 0.
  • B is L b q is 1, m is 0, and n is 1.
  • B is L q is 0, m is 0, and n is 0.
  • B is L 2 .
  • Ri is H, CI, F, dimethylamino, pyrrolidin-1-yl, morpholin-1-yl, 4-methylpiperazin-l-yl, OH or OCH 3 .
  • R 2 is H and R 4 is methyl of each of the foregoing embodiments of the compounds of Formula (I),
  • R ⁇ 3 , R 14 , R 15 , Ri 6 and Rn are each independently H, F, CI, Br, CN, dimethylamino, ethoxycarbonyl, methylthio, methyl, ethyl, isopropyl, trifluoromethyl, trifluoromethoxy, methoxy, NH 2 or N0 2 .
  • R 2 is H and R4 is methyl.
  • B is L 2 .
  • R 4 is methyl.
  • R 2 is H.
  • R 2 is H or lower allcyl(C ⁇ _ ⁇ );
  • R 3 is lower alkyl (C ⁇ _ 6 ), or halogen
  • R 4 is lower alkyl ( . 6 );
  • X is eitiier Oxygen or Sulfur
  • R 15 is H or lower alkyl(C ⁇ _();
  • Ri 6 and Rn are independently d . 6 alkyl, or C 2 . 6 alkenyl, or cycloalkyl, or aryl, or CH 2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from CF 3 , CC1 3 , Me, N0 2 , OH, OMe, OEt, C0NR I8 R 19 ,
  • NR 1S R 19 NHCOCH 3j OCF 3 , SMe, COOR 20 , SO 3 R 20 , S0 2 NR 18 R 19 , COMe, COEt, CO-lower alkyl, SCF 3 , CN, C 2 . 6 alkenyl, H, halogens, C M alkoxy, C 3-6 cycloalkyl, C ⁇ . ⁇ alkyl, and aryl;
  • Ris and R ⁇ 9 are independently a H or C ⁇ -6 alkyl, or C 2 . 6 alkenyl, or cycloalkyl, or aryl, or CH 2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from CF 3 , CC1 3 , Me, N0 2 , OH, OMe, OEt, CONR 21 R 22 , NR 21 R 22 , NHCOCH 3 , OCF 3 , SMe, C00R 23 , S0 3 R 23 , S0 2 NR 2 ⁇ R 22 , COMe, COEt, CO-lower alkyl, SCF 3 , CN, C 2 .
  • R !8 and R ⁇ 9 may form part of a 5, 6 or 7 membered cyclic structure which may be either saturated or unsaturated and that may contain up to four heteroatoms selected from O, N or S and said cyclic structure may be optionally substituted by up to four substituents in any position independently selected from CF 3 , CC1 3 , Me, N0 2 , OH, OMe, OEt, OCF 3 , SMe, COOR 23 , S0 2 NR 21 R 22 , S0 3 R 23 , NHCOCH 3 , COEt, COMe, or halogen;
  • R 20 and R 23 are each independently selected from H or C ⁇ _ 6 alkyl;
  • R 2 ⁇ and R 22 are each independently are independently a H, or C) .
  • an aryl moiety can be a 5 or 6 membered aromatic heterocyclic ring (containing up to 4 hetero atoms independently selected from N, 0, or S) or a 6 membered aromatic non-heterocyclic ring or a polycycle;
  • Ci- ⁇ alkyl groups can be straight chain or branched; optionally substituted C ⁇ . 6 alkyl moieties can be straight chain or branched; C 2 . 6 alkenyl moieties can be straight chain or branched; and optionally substituted C 2 .6 alkenyl moieties can be straight chain or branched.
  • suitable Ci- ⁇ alkyl groups include but are not limited to methyl, ethyl, n- propyl, i-propyl, n-butyl, and t-butyl.
  • Halogens are typically F, CI, Br, and I.
  • Examples of 5 or 6 membered ring moieties include, but are not restricted to, phenyl, furanyl, thienyl, imidazolyl, pyridyl, pyrrolyl, oxazolyl, isoxazolyl, friazolyl, pyrazolyl, tefrazolyl, thiazolyl and isothiazoyl.
  • Examples of polycycle moieties include, but are not restricted to, naphthyl, benzothiazolyl, benzofuranyl, benzimidazolyl, quinolyl, isoquinolyl, indolyl, quinoxalinyl, quinazolinyl and benzothienyl.
  • R 3 is F, CI, Br, I, C ⁇ -6 straight chain or branched alkyl, C 3 . 8 cycloalkyl, C 4-9 alkylcycloalkyl, or C 2 . 6 alkenyl;
  • X is O or S
  • Y is NR 15 Ri6, or (CH 2 ) m R 17 , or 0(CH 2 ) n R ; m is an integer between 0 and 4, inclusive; n is an integer between 0 and 4, inclusive;
  • R 4 is H, C ⁇ - 8 straight chain or branched alkyl, C 3-8 cycloalkyl, C 4-9 alkylcycloalkyl, or C 2 . 8 alkenyl;
  • R 2 and R ⁇ 5 a is each independently selected from H, C ⁇ _ 8 straight chain or branched alkyl, C 3 . 8 cycloalkyl, C 4-9 alkylcycloalkyl, or C 2-8 alkenyl;
  • Ri 6 and R 17 is each independently selected from: C_._ straight chain or branched alkyl, C 2-8 alkenyl or cycloalkyl, or alkylcycloalkyl, or aryl, or CH 2 aryl, wherein each moiety within said C ⁇ -8 straight chain or branched alkyl, C 2 .
  • alkenyl or cycloalkyl, or alkylcycloalkyl, or aryl or CH 2 aryl may be optionally substituted by up to four substituents in any position, whereby each substituent is independently selected from: H, F, CI, Br, I, R 20 , CF 3 , CF 2 R 7 , CF 2 CF 2 , CC1 3 , CC1 2 R 7 , CC1 2 CC1 2 R 7 , NR 18 R 19 , NR ⁇ 8 COR 20 , NR 18 SO 2 R 20 , OR 20 , OCF 3 , OCF 2 R 20 , OCF 2 CF 2 R 20 , OCOR 20 , OSO 2 R 20 , OPO(OR 20 ) 2 , SR 20 , SCF 3 , SCF 2 R 20 , SCF 2 CF 2 R 20 , SCOR 20 , SO 3 R 20 , S0 2 NR 1S R 19 , PO(OR 20 ) 3 , PO(
  • R 20 is H, Ci-s straight chain or branched alkyl, C 3-8 cycloalkyl, C 4-9 alkylcycloalkyl, C 2-8 alkenyl, aryl or alkylaryl; R ⁇ 8 and R ⁇ 9 are each independently selected from H, C ⁇ -8 straight chain or branched alkyl, C 2 .
  • An aryl moiety can be a 5 or 6 membered aromatic heterocyclic ring (containing up to
  • C . .. alkyl groups include but are not limited to methyl, ethyl, n-propyl, i-propyl, n-butyl, and t-butyl.
  • Examples of 5 or 6 membered ring moieties include, but are not restricted to, phenyl, furanyl, thienyl, imidazolyl, pyridyl, pyrrolyl, oxazolyl, isoxazolyl, friazolyl, pyrazolyl, tefrazolyl, thiazolyl, and isothiazolyl.
  • Examples of polycycle moieties include, but are not restricted to, naphthyl, benzothiazolyl, benzofuranyl, benzirnidazolyl, quinolyl, isoquinolyl, indolyl, quinoxalinyl, quinazolinyl, and benzothienyl.
  • X is O or S;
  • R 4 is H or CH 3 ;
  • Rs and R 20 are each independently selected from H, C ⁇ -8 straight chain or branched alkyl, C 3 . 8 cycloalkyl, C 4-9 alkylcycloalkyl, or C 2-8 alkenyl;
  • R 15 is H, F, CI, Br, I, R 20 , CF 3 , CF 2 R 20 , CF 2 CF 2 , CC1 3 , CC1 2 R 20 , CC1 2 CC1 2 R 20 , NR 18 R 19 ,
  • R 18 and R !9 are each independently selected from H, C ⁇ -8 straight chain or branched alkyl, C 2 . 8 alkenyl or cycloalkyl, or alkylcycloalkyl, or aryl, or CH 2 aryl, wherein each moiety within said C ⁇ _ 8 straight chain or branched alkyl, C 2-8 alkenyl or cycloalkyl, or alkylcycloalkyl, or aryl or CH 2 aryl may be optionally substituted by up to four substituents in any position, whereby each substituent is independently selected from: F, CI, Br, I, CF 3 , CC1 3) CH 3 , C 2 H 5 , C 3 H 7 , C 4 H 9 , NH 2 , NHCH 3 , N(CH 3 ) 2 , NHC 2 H 5 , N(C 2 H 5 ) 2 , NHC 3 H 7 , N(C 3 H 7 ) 2 , NHC 4 H 9 ,
  • R 13 , Ri 4 , R 15 , Ri 6 and R 17 each independently selected from the following: F, CI, Br, I, CF 3 , CC1 3 , CH 3 , C 2 H 5 , C 3 H 7 , C 4 H 9 , NH 2 , NHCH 3; N(CH 3 ) 2 , NHC 2 H 5 , N(C 2 H 5 ) 2 , NHC 3 H 7 , N(C 3 H 7 ) 2 , NHC 4 H 9 , N(C 4 H 9 ) 2 , NHCOH, NHCOCH 3 , NHCOC 2 H 5 , NHCOC 3 H 7 , NHCOC 4 H 9 , NHS0 2 CH 3 , NHS0 2 C 2 H 5 , NHS0 2 C 3 H 7 , NHS0 2 C 4 H 9 , OH, OCH 3 , OC 2 H 5 , OC 3 H 7 , OC 4 H 7 , OC 4 H 9 , OC 5 H 9 , OC 5
  • a more preferred series of compounds possessing 5-HT 2A receptor activity that are useful as inverse agonists at such receptors is designated by the general Formula (B):
  • R 2 is H or lower alkyl (C 1-4 );
  • R 3 is Me, or Et, or halogen
  • X is either Oxygen or Sulfur
  • Y is NR 1 5R16, or (CH 2 ) m R 17 , or 0(CH 2 ) n R 17 ;
  • Ri 5 is H or lower alkyl(C ); Ri 6 and Rn are independently C ⁇ _ 6 alkyl, or C 2 . 6 alkenyl, or cycloalkyl, or aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from CF 3 , CC1 3 , Me, N0 2 , OH, OMe, OEt, CONR 18 R ⁇ 9 , NR ⁇ 8 R 19 , NHCOCH 3 , OCF 3 , SMe, COOR 20 , SO 3 R 20 , S0 2 NR ⁇ 8 R 19 , COMe, COEt, CO-lower alkyl, SCF 3 , CN, C 2-6 alkenyl, H, halogens, C__ ⁇ alkoxy, C 3-6 cycloalkyl, C ⁇ -6 alkyl, aryl and aryloxy wherein each of the C 1-4 alkoxy, C 3 .6 cycloalkyl
  • Ris and R ⁇ 9 are independently a H or C ⁇ _ 6 alkyl, or C 2 . 6 alkenyl, or cycloalkyl, or aryl, or CH 2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from CF 3 , CC1 3 , Me, N0 2 , OH, OMe, OEt, CONR 21 R 22 , NR 21 R 22 , NHCOCH3, OCF 3 , SMe, COOR 23 , S0 3 R 23 , S0 2 NR_ ⁇ R 22 , COMe, COEt, CO-lower alkyl, SCF 3 , CN, C 2 . 6 alkenyl, H, halogens, Q.
  • C 3-6 cycloalkyl, C ⁇ -6 alkyl, and aryl or R ⁇ 8 and R ⁇ 9 may form part of a 5, 6 or 7 membered cyclic structure which may be either saturated or unsaturated and that may contain up to four heteroatoms selected from O, N or S and said cyclic structure may be optionally substituted by up to four substituents in any position independently selected from CF 3 , CC1 3 , Me, N0 2 ,
  • R 20 and R 23 are each independently selected from H or C ⁇ . 6 alkyl;
  • R 21 and R 22 are each independently are independently a H, or C 1 . 6 alkyl, or C 2- 6 alkenyl, or cycloalkyl, or aryl, or CH 2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from halogen, CF 3 , OCF 3 , OEt, CC1 3 , Me, N0 2 , OH, OMe, SMe, COMe, CN, COOR 20 , SO 3 R 20 , COEt, NHCOCH 3 , or aryl;
  • C 1 . 6 alkyl moieties can be straight chain or branched; optionally substituted C ⁇ _ 6 alkyl moieties can be straight chain or branched:
  • C 2-s alkenyl moieties can be straight chain or branched; and optionally substituted C 2 _6 alkenyl moieties can be straight chain or branched.
  • C ⁇ _ 6 alkyl groups include but are not limited to methyl, ethyl, n- propyl, i-propyl, n-butyl, and t-butyl.
  • Halogens are typically F, Cl, Br, and I.
  • Examples of 5 or 6 membered ring moieties include, but are not restricted to, phenyl, furanyl, thienyl, imidazolyl, pyridyl, pyrrolyl, oxazolyl, isoxazolyl, friazolyl, pyrazolyl, tefrazolyl, thiazolyl and isothiazoyl.
  • Examples of polycycle moieties include, but are not restricted to, naphthyl, benzothiazolyl, benzofuranyl, benzimidazolyl, quinolyl, isoquinolyl, indolyl, quinoxahnyl, quinazolinyl and benzothienyl.
  • R 2 and R ⁇ 5 are H;
  • R 3 is Br;
  • X is O;
  • R 4 is Me.
  • R ⁇ 6 is preferably 4-trifluoromethoxyphenyl , 4- trifluoromethoxybenzyl, 4-chlorophenyl or 4-fluorophenyl.
  • Certain preferred compounds are: Compound 7 N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)(((4-frifluoromethoxy)phenyl)an nocarboxamide
  • Inositol phosphate accumulation assays evidence the activity of test compounds. Both single concentration percentages of control values and IC 0 determinations indicate activity.
  • the column legends have the following meanings: IP 3 % Control: The values in this column reflect an IP Accumulation Assay where the test compounds were evaluated at one concentration of 10 ⁇ M. For these assays, the compound was diluted into inositol-free Dulbecco's Eagle Media containing 10 ⁇ M pargyline and 10 mM LiCl and tested at a final assay concentration of 10 ⁇ M, in triplicate. The percent control value was calculated based on the control in which no test compound was added.
  • IPT AP-3 ICsn nM The values in this column reflect an IP accumulation assay in which the test compound was evaluated at several different concentrations whereby an IC 50 could be detennined. This column corresponds to the column appearing in the tables above which is labeled: Inositol Phosphate Accumulation, AP-3, IC 50 Value ( ⁇ M).
  • WT 5-HTTA LSD ICsn nM The values in this column reflect a competitive binding assay using LSD. This column corresponds to the column appearing in the tables above which is labeled: Competitive Binding, WT 5-HT 2A) (( 3 H)LSD), IC 50 Value ( ⁇ M).
  • R 2 is H.
  • R 3 is Br.
  • X is O.
  • R 4 is Me.
  • R 17 is 4-methoxyphenyl or tertiary butyl.
  • 5-HT 2A receptor was further confirmed by the following:
  • Lysergic acid diethylamide is a potent 5-HT 2A receptor and dopamine D2 receptor ligand.
  • An indication of the selectivity of compounds for either or both of these receptors involves displacement of radiolabeled-bound LSD from pre-treated brain sections.
  • radiolabeled I 125 -LSD NN Life Sciences, Boston, Mass., Catalogue number NEX-199 was utilized; spiperone (RBI, Natick, Mass. Catalogue number s-128) a 5-HT 2A receptor and dopamine D2 receptor antagonist, was also utilized.
  • Buffer consisted of 50 nanomolar TRIS-HCl, pH 7.4.
  • Brain sections were incubated in (a) Buffer plus 1 nanomolar I i25 -LSD; (b) Buffer plus 1 nanomolar I 125 -LSD and 1 micromolar spiperone; or Buffer plus 1 nanomolar I 125 -LSD and 1 micromolar Compound 1 for 30 minutes at room temperature. Sections were then washed 2x 10 minutes at 4 ° C. in Buffer, followed by 20 seconds in distilled H 2 0. Slides were then air-dried.
  • Figures 16A-C provide representative autoradiographic sections from this study.
  • Figure 16A evidences darker bands (derived from I 125 -LSD binding) primarily in both die fourth layer of the cerebral cortex (primarily 5-HT 2A receptors), and the caudate nucleus (primarily dopamine D2 receptors and some 5-HT 2A receptors).
  • spiperone which is a 5-HT 2 and dopamine D2 antagonist, displaces the I 125 -LSD from these receptors on both the cortex and the caudate.
  • Compound 1 appears to selectively displace the I 125 - LSD from the cortex (5-HT 2A ) and not the caudate (dopamine D2).
  • a third series of compounds having 5-HT 2A receptor activity is represented by a class
  • R 2 is H.
  • R 3 is Br.
  • X is O.
  • R 4 is Me.
  • R is preferably 4-trifluoromethoxyphenyl, or thiophene, or
  • a 5-HT 2A receptor antagonist or inverse agonist is expected to decrease amphetamine- stimulated locomotion without affecting baseline locomotion. See, for example, Soresnon, et al, 266(2) J. Pharmacol. Exp. Ther. 684 (1993). Based upon the foregoing information, Compound 2 is a potent inverse agonist at the human 5-HT 2A receptor. For the following study, the following parameters and protocol were utilized:
  • a San Diego Instruments Flex Field apparatus was used to quantify baseline and amphetamine-stimulated locomotor activity. This apparatus consists of four 16" x 16" clear plastic open fields. Photocell arrays (16 in each dimension) interfaced with a personal computer to automatically quantify activity. Several measures of activity can be assessed witii the apparatus, including total photocell beam breaks. Animals (vehicle control and Compound treated) were injected s.c. 30 minutes prior to initiation of analysis. Following this 30 minute period, animals were placed individually into an open field and baseline activity was assessed for 30 minutes (habituation phase).
  • R 2 is H or lower alkyl(C );
  • R 3 is Me, or Et, or halogen;
  • X is either Oxygen or Sulfur;
  • Y is NR 15 R 16 , or (CH2) m Rn, or 0(CH 2 ) n R 17 ;
  • R 15 is H or lower alkyl(C M );
  • R 18 and R ⁇ 9 are independently a H or C ⁇ -6 alkyl, or C 2-6 alkenyl, or cycloalkyl, or aryl, or CH 2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from CF 3 , CC1 3 , Me, N0 2 , OH, OMe, OEt, CONR 21 R 22 , NR 2 ⁇ R 22 , NHCOCH 3 , OCF 3 , SMe, C00R 23 , S0 3 R 23 , S0 2 NR 2 ⁇ R 22 , COMe, COEt, CO-lower alkyl, SCF 3 , CN, C 2-5 alkenyl, H, halogens, C..
  • Ris and R 19 may form part of a 5, 6 or 7 membered cyclic structure which may be either saturated or unsaturated and that may contain up to four heteroatoms selected from O, N or S and said cyclic structure may be optionally substituted by up to four substituents in any position independently selected from CF 3 , CC1 3 , Me, N0 2 , OH, OMe, OEt, OCF 3 , SMe, COOR 7 , S0 2 NR 2 ⁇ R 22 , S0 3 R 23 , NHCOCH 3 , COEt, COMe, or halogen;
  • Rn is C ⁇ - 6 alkyl, or C 2 . 6 alkenyl, or cycloalkyl, or aryl, or CH 2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from CF 3 , CC1 3 , Me, N0 2 , OH, OMe, OEt, CONR 21 R 22 , NR_ ⁇ R 22 , NHCOCH,, OCF 3 , SMe, COOR 23 , S0 3 R 23 , S0 2 NR 21 R 22 , COMe, COEt, CO-lower alkyl, SCF 3 , CN, C 2 .
  • R 20 and R 23 may be independently selected from H or C ⁇ -6 alkyl;
  • R 21 and R 22 are independently a H, or C alkyl, or C 2-6 alkenyl, or cycloalkyl, or aryl, or CH 2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from halogen, CF 3 , OCF3, OEt, CC1 3 , Me, N0 2 , OH, OMe, SMe, COMe, CN, COOR 20 , SO 3 R 20 , COEt, NHCOCH 3 , or aryl.
  • An aryl moiety can be a 5 or 6 membered aromatic heterocyclic ring (containing up to 4 hetero atoms independently selected from N, O, or S) or a 6 membered aromatic non-heterocyclic ring or a polycycle;
  • C ⁇ -6 alkyl moieties can be straight chain or branched; optionally substituted C ⁇ _ 6 alkyl moieties can be straight chain or branched; C 2-6 alkenyl moieties can be straight chain or branched; and optionally substituted C 2 . 6 alkenyl moieties can be straight chain or branched;
  • suitable C ⁇ -6 alkyl groups include but are not limited to methyl, ethyl, n-propyl, i-propyl, n-butyl, and t-butyl.
  • Halogens are typically F, CI, Br, and I.
  • Examples of 5 or 6 membered ring moieties include, but are not restricted to, phenyl, furanyl, thienyl, imidazolyl, pyridyl, pyrrolyl, oxazolyl, isoxazolyl, friazolyl, pyrazolyl, tefrazolyl, thiazolyl and isothiazolyl.
  • Examples of polycycle moieties include, but are not restricted to, naphthyl, benzothiazolyl, benzofuranyl, benzimidazolyl, quinolyl, isoquinolyl, indolyl, quinoxahnyl, quinazolinyl and benzothienyl.
  • Some embodiments of the invention are compounds of Formula (V) and have one of the structures below.
  • X is O or S; P is H or CH 3 ;
  • R 8 and R 30 are each independently selected from H, C ⁇ -8 straight chain or branched alkyl, C 3-8 cycloalkyl, C 4-9 alkylcycloalkyl, or C 2 . 8 alkenyl;
  • R 15 is H, F, CI, Br, I, R 20 , CF 3 , CC1 3 , NR 18 R 19 , NR 30 COR 20 , NR 30 SO 2 R 20 , OR 20 , OCF 3 ,
  • R ⁇ S and said adjacent position can together be selected from SCH 2 S, SCH 2 CH 2 S, OCH 2 0, or OCH 2 CH 2 0 to form a bi-cyclic structure;
  • R 20 is H, C ⁇ -8 straight chain or branched alkyl, C 3 . 8 cycloalkyl, C 4-9 alkylcycloalkyl, C 2-8 alkenyl, aryl or alkylaryl;
  • Ris and Ri are each independently selected from: H, C ⁇ _ 8 straight chain or branched alkyl, C 2- 8 alkenyl or cycloalkyl, or alkylcycloalkyl, or aryl or CH 2 aryl wherein each moiety within said C ⁇ -8 straight chain or branched alkyl, C 2 .
  • alkenyl or cycloalkyl, or alkylcycloalkyl, or aryl or CH 2 aryl may be optionally substituted by up to four substituents in any position, whereby each substituent is independently selected from: F, CI, Br, I, CF 3 , CC1 3 , CH 3 , C 2 H 5 , C 3 H 7 , C 4 H 9 , NH 2 , NHCH 3 , N(CH 3 ) 2 , NHC 2 H 5 , N(C 2 H 5 ) 2 , NHC 3 H 7 , N(C 3 H 7 ) 2 , NHC 4 H 9 , N(C 4 H 9 ) 2 , NHCOH, NHCOCH 3 , NHCOC 2 H 5 , NHCOC 3 H 7 , NHCOC 4 H 9 , NHS0 2 CH 3 , NHS0 2 C 2 H 5 , NHS0 2 C 3 H 7 , NHS0 2 C 4 H 9 , OH, OCH 3 , OC
  • Ar is a phenyl ring optionally substituted with up to five groups selected from tiie group consisting of halogen, OR 7 , OH, NR_R 9 , carboxy, CN, alkoxycarbonyl, straight chain or branched C ⁇ -6 alkyl -C(p) 3 ⁇ or -0-C(p) 3 where p is halogen;
  • R 8 and R 9 are independently a H, or C ⁇ -6 alkyl, or C 2 . 6 alkenyl, or cycloalkyl, or aryl, or CH 2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from halogen, CF 3 , OCF 3 ,
  • R 8 and R 9 may form part of a 5, 6 or 7 membered cyclic structure which may be either saturated or unsaturated and that may contain up to four heteroatoms selected from O, N or S and said cyclic structure may be optionally substituted by up to four substituents in any position independently selected from CF 3 , CC1 3 , Me, N0 2 ,
  • R 7 is H or C 1-6 alkyl
  • Rio is H or C ⁇ -6 alkyl
  • R 2 is H, straight chain or branched C ⁇ -6 alkyl, C 2 . 6 alkenyl, or cycloalkyl;
  • R 3 is halogen, carboxy, CN, alkoxycarbonyl, straight chain or branched C M alkyl, C 2 _ 6 alkenyl, C 2 solicit6 alkynyl, cycloalkyl, aryl or heteroaryl, wherein each of said alkoxycarbonyl, straight chain or branched -6 alkyl, C 2 . 6 alkenyl, C 2 .
  • 6 alkynyl, cycloalkyl, aryl or heteroaryl groups can be optionally substituted with up to four substituents in any position selected from OH, ORio, NR 8 R 9 , halogen, -C(p) 3> or -O- C(p) 3 where p is halogen, and said cycloalkyl, aryl or heteroaryl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position;
  • R is Ci -6 alkyl, C 2 . 6 alkenyl, or cycloalkyl;
  • Rn and R l2 are each independently H, straight chain or branched C ⁇ _ 6 alkyl, C 2-6 alkenyl, or cycloalkyl;
  • Ri 3 , R ⁇ , R 1 5, Ri ⁇ and R are each independently H, halogen, CN, NR 8 R 9 , COORio, SRio, straight chain or branched C 1-6 alkyl, C 2 . 6 alkenyl, C 2 . 6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl, wherein each of said alkoxycarbonyl, straight chain or branched C ⁇ _ 6 alkyl, C 2 .
  • 6 alkenyl, C 2-6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl groups can be optionally substituted with up to four substituents in any position selected from N0 2 , OR 10 , NR 8 R 9 , halogen, -C(p) 3 , or -0-C(p) 3 where p is halogen, and said cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position;
  • L 2 is -0-Q 2 wherein Q 2 is straight chain or branched - ⁇ alkyl
  • the present invention further provides methods for modulating die activity of a human 5-HT 2A serotonin receptor by contacting the receptor with a compound of formula:
  • Ri is H, halogens, NR 5 R_, OH or OR 7 , wherein
  • R 5 and Re are independently H, or C ⁇ -6 alkyl, or C 2-6 alkenyl, or cycloalkyl, or aryl, or CH 2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from CF 3 , CC1 3 , Me, N0 2 , OH, OMe, OEt, CONR 8 R 9 , NR 8 R 9 , NHCOCH 3 , OCF 3 , SMe, COOR 10 , S0 3 R 8 ,
  • 6 cycloalkyl, Q .6 alkyl, or aryl groups may be further optionally substituted by up to four substituents in any position independently selected from CF 3 , CC1 3 , Me, N0 2 , OH, OMe, OEt, CONR 8 R 9 , NR 8 R 9 , NHCOCH 3 , OCF 3 , SMe, COOR 10 , S0 2 NR 8 R 9 , SO 3 R 10 , COMe,
  • COEt CO-lower alkyl, SCF 3 , CN, C 2 . 6 alkenyl, H, halogens, C M alkoxy, C 3 . 6 cycloalkyl, C 1-6 alkyl, and aryl; or
  • R 5 and ⁇ may form part of a 5, 6 or 7 membered cyclic structure which may be either saturated or unsaturated and that may contain up to four heteroatoms selected from O, N or S and said cyclic structure may be optionally substituted by up to four substituents in any position independently selected from CF 3 , CC1 3 , Me, N0 2 , OH, OMe, OEt, OCF 3 , SMe, COOR 10 , S0 2 NRsR 9 , SO 3 R ⁇ 0 , NHCOCH 3 , COEt, COMe, or halogen; R 8 and R 9 are independently a H, or C .
  • Rio is H or C M alkyl;
  • R 7 is H or C ⁇ _6 alkyl;
  • R 2 is H, straight chain or branched C ⁇ -6 alkyl, C 2 . 6 alkenyl, or cycloalkyl;
  • R 3 is halogen, carboxy, CN, alkoxycarbonyl, straight chain or branched Ci . alkyl, C 2- 6 alkenyl, C 2 . 6 alkynyl, cycloalkyl, aryl or heteroaryl, wherein each of said alkoxycarbonyl, straight chain or branched C e alkyl, C 2 .
  • 6 alkenyl, C 2 _6 alkynyl, cycloalkyl, aryl or heteroaryl groups can be optionally substituted with up to four substituents in any position selected from OH, OR 10 , NR ⁇ R 9 , halogen,
  • R 4 is C ⁇ _6 alkyl, C 2-6 alkenyl, or cycloalkyl;
  • L is:
  • R u and R ⁇ 2 are each independently H, straight chain or branched C M alkyl, C 2-6 alkenyl, or cycloalkyl;
  • R 13 , R 14 , R 15 , R 16 and R 17 are each independently H, halogen, CN,
  • NR 8 R 9 , COOR JO , SR 10 straight chain or branched C ⁇ -6 alkyl, C 2 . 6 alkenyl, C 2 . 6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl, wherein each of said alkoxycarbonyl, straight chain or branched C ⁇ _ 6 alkyl, C 2 . 6 alkenyl, C 2 .
  • 6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl groups can be optionally substituted with up to four substituents in any position selected from N0 2 , ORio, NR 8 R 9 , halogen, -C(p) 3j or -0-C(p) 3 where p is halogen, and said cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position;
  • Q 2 is straight chain or branched _ 6 alkyl
  • An aryl moiety can be a 5 or 6 membered aromatic heterocyclic ring (containing up to
  • Also provided by the present invention are methods for modulating the activity of a human 5-HT 2A serotonin receptor by contacting the receptor with a compound of formula:
  • R 2 is H or lower alkyl(C ⁇ _ ⁇ );
  • R 3 is lower alkyl (C ⁇ . 6 ), or halogen;
  • R 4 is lower alkyl (C ⁇ _ 6 );
  • X is eidier Oxygen or Sulfur
  • R 1 6 and Rn are independently C ⁇ _ 6 alkyl, or C 2 . ⁇ alkenyl, or cycloalkyl, or aryl, or CH 2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from CF 3 , CC1 3 , Me, N0 2 , OH, OMe, OEt, CONR 18 R ⁇ 9 , NR 18 R 19 , NHCOCH 3 , OCF 3 , SMe, COOR 20 , SO 3 R 20 , S0 2 NR 18 R 19 , COMe, COEt, CO-lower alkyl, SCF 3 , CN, C 2 . 6 alkenyl, H, halogens, C alkoxy, C 3-6 cycloalkyl, d_6 alkyl, and aryl;
  • Ris and R 19 are independently a H or C ⁇ -6 alkyl, or C 2 . 6 alkenyl, or cycloalkyl, or aryl, or CH 2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from CF 3 , CC1 3 , Me, N0 2 , OH, OMe, OEt, C0NR 21 R 22 , NR 21 R 22 , NHCOCH 3 , OCF 3 , SMe, C00R 23 , S0 3 R 23 , S0 2 NR 2 ⁇ R 22 , COMe, COEt, CO-lower alkyl, SCF 3 , CN, C 2-6 alkenyl, H, halogens, .
  • Ris and R ⁇ 9 may form part of a 5, 6 or 7 membered cyclic structure which may be either saturated or unsaturated and that may contain up to four heteroatoms selected from O, N or S and said cyclic structure may be optionally substituted by up to four substituents in any position independently selected from CF 3 , CC1 3 , Me, N0 2 ,
  • R 20 and R 23 are each independently selected from H or C ⁇ _ 6 alkyl;
  • R 2 ⁇ and R 22 are each independently are independently a H, or C 1-6 alkyl, or C 2- 6 alkenyl, or cycloalkyl, or aryl, or CH 2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from halogen, CF 3 , OCF 3 , OEt, CC1 3 , Me, N0 2 , OH, OMe, SMe, COMe, CN, COOR 20 , SO 3 R 20 , COEt, NHCOCH 3 , or aryl; an aryl moiety can be a 5 or 6 membered aromatic heterocyclic ring (containing up to 4 hetero atoms independently selected from N, O, or S) or a 6 membered aromatic non-heterocyclic ring or a polycycle.
  • the present invention further provides methods for modulating the activity of a human 5-HT 2A serotonin receptor by contacting the receptor with a compound of formula:
  • R 3 is F, Cl, Br, I, C 1-6 straight chain or branched alkyl, C 3-8 cycloalkyl, C 4 . 9 alkylcycloalkyl, or C 2 . 6 alkenyl; Xis O or S;
  • Y is NR15R16, or (CH 2 ) m R 17 , or 0(CH 2 ) n R 17; m is an integer between 0 and 4, inclusive; n is an integer between 0 and 4, inclusive; is H, C ⁇ -8 straight chain or branched alkyl, C 3-8 cycloalkyl, C 4-9 alkylcycloalkyl, or C 2-8 alkenyl;
  • R 2 and R 15 a is each independently selected from H, C ⁇ . g straight chain or branched alkyl, C 3 . 8 cycloalkyl, C 4-9 alkylcycloalkyl, or C 2-8 alkenyl;
  • R J6 and Rn is each independently selected from: C ⁇ . 8 straight chain or branched alkyl, C 2-8 alkenyl or cycloalkyl, or alkylcycloalkyl, or aryl, or CH 2 aryl, wherein each moiety within said C 1-8 straight chain or branched alkyl, C 2 .
  • alkenyl or cycloalkyl, or alkylcycloalkyl, or aryl or CH 2 aryl may be optionally substituted by up to four substituents in any position, whereby each substituent is independently selected from: H, F, Cl, Br, I, R 20 , CF 3 , CF 2 R 7 , CF 2 CF 2 , CC1 3 , CC1 2 R 7 , CC1 2 CC1 2 R 7 , NR 18 R 19 , NR 18 COR 20 , NR ⁇ 8 SO 2 R 20 , OR 20 , OCF 3 , OCF 2 R 20 , OCF 2 CF 2 R 20 , OCOR 20 , OSO 2 R 20 , OPO(OR 20 ) 2 , SR 20 , SCF 3 , SCF 2 R 20 , SCF 2 CF 2 R 20 , SCOR 20 , SO 3 R 20 , S0 2 NR ⁇ 8 R ⁇ 9 , PO(OR 20 ) 3 , PO(
  • R 20 is H, C ⁇ _ 8 straight chain or branched alkyl, C 3 . 8 cycloalkyl, C 4-9 alkylcycloalkyl, C 2 . 8 alkenyl, aryl or alkylaryl; Ris and R 19 are each independently selected from H, C ⁇ -8 straight chain or branched alkyl, C 2 . 8 alkenyl or cycloalkyl, or alkylcycloalkyl, or aryl, or CH 2 aryl, wherein each moiety within said C ⁇ -8 straight chain or branched alkyl, C 2 .
  • alkenyl or cycloalkyl, or alkylcycloalkyl, or aryl or CH 2 aryl may be optionally substituted by up to four substituents in any position, whereby each substituent is independently selected from: F, Cl, Br, I, CF 3 , CC1 3 , CH 3 , C 2 H 5 , C 3 H 7 , C 4 H 9 , NH 2 , NHCH 3 , N(CH 3 ) 2 , NHC 2 H 5 , N(C 2 H 5 ) 2 , NHC 3 H 7 , N(C 3 H 7 ) 2 , NHC 4 H 9 , N(C 4 H 9 ) 2 , NHCOH, NHCOCH . , NHC0C 2 H 5 ,
  • OCOC 4 H 9 OS0 2 CH 3 , OS0 2 C 2 H 5 , OS0 2 C 3 H 7 , OS0 2 C 4 H 9 , SH, SCH 3 , SC 2 H 5 , SC 3 H 7 , SC 4 H 7 , SGd ,
  • Ris or R ]9 may together form part of a 5, 6 or 7 membered cyclic stracture, with said stracture being saturated or unsaturated, and further with said stracture containing up to four heteroatoms selected from O, N or S, and further wherein each moiety within said cyclic structure being optionally substituted by up to four substituents in any position independently selected from: F, Cl, Br, I, CF 3 , CC1 3 , CH 3 , C 2 H 5 , C 3 H 7 , C 4 H 9 , NH 2 , NHCH 3; N(CH 3 ) 2 , NHC 2 H 5 , N(C 2 H 5 ) 2 , NHC 3 H 7 , N(C 3 H 7 ) 2 , NHC 4 H 9 , N(C 4 H 9 ) 2 , NHCOH, NHCOCH 3 , NHCOC 2 H 5 , NHCOC 3 H 7 , NHCOQH,, NHS0 2 CH 3 , NHS
  • An aryl moiety can be a 5 or 6 membered aromatic heterocyclic ring (containing up to
  • Also provided by the present invention are methods for modulating the activity of a human 5-HT 2A serotonin receptor by contacting the receptor with a compound of formula:
  • X is O or S;
  • P is H or CH 3 ;
  • R 8 and R 20 are each independently selected from H, C ⁇ -8 straight chain or branched alkyl, C 3-8 cycloalkyl, C 4 . 9 alkylcycloalkyl, or C 2 . 8 alkenyl;
  • R 15 is H, F, Cl, Br, I, R 20 , CF 3 , CF 2 R 20 , CF 2 CF 2 , CC1 3 , CC1 2 R 20 , CC1 2 CC1 2 R 20 , NR 18 R 19 , NR 19 COR 20 , NR 19 SO 2 R 20 , OR 20 , OCF 3 , OCF 2 R 20 , OCF 2 CF 2 R 20 , OCOR 20 , OSO 2 R 20 , OPO(OR 20 ) 2 , SR 20 , SCF 3 , SCF 2 R 20 , SCF 2 CF 2 R 20 , SCOR 20 , SO 3 R 20 , S0 2 NR 18 R ⁇ 9 , PO(OR 20 ) 3 , PO(OR 20 ) 2 R 20 , N0 2 , CN, CNR 20 (NR 18 R ⁇ 9 ), CNR 19 (SR 20 ), COOR 20 , COSR 20 , CONR ⁇ 8 R ⁇ 9 , with the pro
  • Ris and R ⁇ 9 are each independently selected from H, C]. s straight chain or branched alkyl, C 2 . 8 alkenyl or cycloalkyl, or alkylcycloalkyl, or aryl, or CH 2 aryl, wherein each moiety within said C ⁇ _ 8 straight chain or branched alkyl, C 2-8 alkenyl or cycloalkyl, or alkylcycloalkyl, or aryl or CH 2 aryl may be optionally substituted by up to four substituents in any position, whereby each substituent is independently selected from: F, Cl, Br, I, CF 3 , CC1_, CH 3 , C 2 H 5 , C 3 H 7 , C 4 H 9 , NH 2 , NHCH 3 , N(CH 3 ) 2 ,
  • OCOC 4 H 9 OS0 2 CH 3 , OS0 2 C 2 H 5 , OS0 2 C 3 H 7 , OS0 2 C 4 H 9 , SH, SCH 3 , SC 2 H 5 , SC 3 H 7 , SC 4 H 7 , SC 4 H 9 ,
  • R 13 , R ⁇ 4 , R 15 , Ri 6 and R each independently selected from the following: F, Cl, Br, I, CF 3 , CC1 3 , CH 3 , C 2 H 5 , C 3 H 7 , C 4 H 9 , NH 2 , NHCH 3 , N(CH 3 ) 2 , NHC 2 H 5 , N(C 2 H 5 ) 2 , NHC 3 H 7 , N(C 3 H 7 ) 2 ,
  • NHC 4 H 9 N(C 4 H 9 ) 2 , NHCOH, NHCOCH 3 , NHCOC 2 H 5 , NHC0C 3 H 7 , NHCOC 4 H 9 , NHS0 2 CH 3 ,
  • An aryl moiety can be a 5 or 6 membered aromatic heterocyclic ring (containing up to 4 hetero atoms independently selected from N, O, or S) or a 6 membered aromatic non-heterocyclic ring or a polycycle.
  • the present invention further provides methods for modulating the activity of a human 5-HT 2A serotonin receptor by contacting the receptor with a compound of formula:
  • R 2 is H or lower alkyl (C M );
  • R 3 is Me, or Et, or halogen;
  • X is either Oxygen or Sulfur;
  • Y is NR ⁇ 5 R 16 , or (CH 2 ) m R 17 , or 0(CH 2 ) n R 17 ;
  • R 4 is lower alkyl (C ⁇ _ 6 );
  • R 16 and R are independently C M alkyl, or C 2 . 6 alkenyl, or cycloalkyl, or aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from CF 3 , CC1 3 , Me, N0 2 , OH, OMe, OEt, C0NR 18 R ⁇ 9 , NR ⁇ 8 R ⁇ 9 , NHCOCH 3 , OCF 3 , SMe, COOR 20 , SO 3 R 20 , S0 2 NR 18 R 19 , COMe, COEt, CO-lower alkyl, SCF 3 , CN, C 2 .
  • Ris and R 19 are independently a H or -6 alkyl, or C 2 . 6 alkenyl, or cycloalkyl, or aryl, or CH 2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from CF 3 , CC1 3 , Me, N0 2 , OH, OMe, OEt, CONR 2 ⁇ R 22 , NR 21 R 22 , NHCOCH3, OCF 3 , SMe, COOR 23 , S0 3 R 23 , S0 2 NR 2 ⁇ R 22 , COMe, COEt, CO-lower alkyl, SCF 3 , CN, C 2 . 6 alkenyl, H, halogens, Q.
  • C 3 . 6 cycloalkyl, C alkyl, and aryl or R 18 and R i9 may form part of a 5, 6 or 7 membered cyclic structure which may be either saturated or unsaturated and that may contain up to four heteroatoms selected from O, N or S and said cyclic structure may be "optionally substituted by up to four substituents in any position independently selected from CF 3 , CC1 3 , Me, N0 2 , OH, OMe, OEt, OCF 3 , SMe, COOR 7 , S0 2 NR 24 R 25 , S0 3 R 26 , NHCOCH 3 , COEt, COMe, or halogen;
  • R 20 and R 23 are each independently selected from H or C !-6 alkyl;
  • R 21 and R 22 are each independently are independently a H, or C 1-5 alkyl, or C 2- 6 alkenyl, or cycloalkyl, or aryl, or CH 2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from halogen, CF 3 , OCF 3 , OEt, CC1 3 , Me, N0 2 , OH, OMe, SMe, COMe, CN, COOR 20 , SO 3 R 20 , COEt, NHCOCH 3 , or aryl.
  • An aryl moiety can be a 5 or 6 membered aromatic hetero-cyclic ring (containing up to 4 hetero atoms independently selected from N, O, or S) or a 6 membered aromatic non-heterocyclic ring or a polycycle.
  • the present invention further provides methods for modulating the activity of a human 5-HT 2A serotonin receptor by contacting the receptor with a compound of formula:
  • Ar is a phenyl ring optionally substituted with up to five groups selected from the group consisting of halogen, OR 7 , OH, NRs 9 , carboxy, CN, alkoxycarbonyl, straight chain or branched C M alkyl -C(p) 3j or -0-C(p) 3 where p is halogen;
  • Rs and R 9 are independentiy a H, or C M alkyl, or C 2-6 alkenyl, or cycloalkyl, or aryl, or CH 2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from halogen, CF 3 , OCF 3 , OEt, CC1 3 , Me, N0 2 , OH, OMe, SMe, COMe, CN, COOR 10 , SO 3 R 10 , COEt, NHCOCH 3 , or aryl; or
  • R 8 and R 9 may form part of a 5, 6 or 7 membered cyclic structure which may be either saturated or unsaturated and that may contain up to four heteroatoms selected from 0, N or S and said cyclic structure may be optionally substituted by up to four substituents in any position independently selected from CF 3 , CC1 3 , Me, N0 2 , OH, OMe, OEt, OCF 3 , SMe, COORio, SOzNRsRc,, SO 3 R 10 , NHCOCH 3 , COEt, COMe, or halogen;
  • R 7 is H or C alkyl; Rio is H or CM alkyl;
  • R 2 is H, straight chain or branched C alkyl, C 2 . 6 alkenyl, or cycloalkyl; iv) R 3 is halogen, carboxy, CN, alkoxycarbonyl, straight chain or branched CM alkyl, C 2 _
  • R 4 is C alkyl, C 2 . 6 alkenyl, or cycloalkyl;
  • R n and R 12 are each independently H, straight chain or branched C alkyl, C 2 . 6 alkenyl, or cycloalkyl; wherein:
  • R ⁇ 3 , R ⁇ 4 , Ris, Ri 6 and R are each independently H, halogen, CN, NR 8 R 9 , COORio, SRio, straight chain or branched C ⁇ -6 alkyl, C -6 alkenyl, C 2 . 6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl, wherein each of said alkoxycarbonyl, straight chain or branched C M alkyl, C 2 . 6 alkenyl, C 2 .
  • 6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl groups can be optionally substituted with up to four substituents in any position selected from N0 2 , ORio, NRsR . , halogen, -C(p) 3; or -0-C(p) 3 where p is halogen, and said cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position;
  • Q 2 is straight chain or branched C M alkyl, C 2 . 6 alkenyl, C 2 . ⁇ alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl, wherein each of said cycloalkyl, aryl, alkylaryl, or arylalkyl groups can be optionally substituted with up to four substituents in any position selected from N0 2 , OR 7 , halogen, -C(p) 3> or -0-C(p) 3 where p is halogen, or an aliphatic or aromatic heterocycle, and said cycloalkyl, aryl, alkylaryl, and arylalkyl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position; or a pharmaceutically acceptable salt.
  • the compounds are selected from compounds and combinations and subcombinations of compounds selected from the group consisting of:
  • the present invention provides the foregoing compounds, and combinations and subcombinations thereof.
  • the present invention provides compounds useful as inverse agonists for 5-HT 2A receptors stracturally represented as follows:
  • compositions comprising each of the compounds ofthe invention.
  • Odier preferred compounds provided by the present invention and useful in the methods disclosed herein include, but are not limited to:
  • R 3 is Br or Cl; t is lower alkyl; R n isH,F,ClorCF 3 ;
  • R 12 is H, F or Cl
  • Specifically preferred compounds of Formula (LX) above include the following:
  • the amine 12-1 can be converted to the corresponding isocyanate 12-4 by die action of phosgene or a suitable phosgene equivalent, e.g. friphosgene, in an inert solvent such as a halocarbon in the presence of an organic base such as triethylamine (i.e., TEA) or diisopropylethylamine (i.e., DIEA).
  • phosgene or a suitable phosgene equivalent e.g. friphosgene
  • Isocyanate 12-4 reacts with amines of the general formula 12-5 in an analogous fashion to that described above in Scheme 12-1 to give urea 12-6.
  • This approach allows for diverse groups to be introduced for the R 2 or R 3 group based on the starting amine 12-5 (Scheme 12-2).
  • Arnines of general formula 12-5 are also readily converted to activated isocyanate equivalents of general fonnula 12-8 by the sequential action of carbonyldiimidazole and metiiyl iodide in tefrahydrofuran and acetonitrile respectively (R.A. Batey et al, Tetrahedron Lett., (1998), 39, 6267-6270.) Reaction of 12-8 with amine 12-1 in an inert solvent such as a halocarbon would yield the requisite ureas of general formula 12-3 (Scheme 12-4).
  • Amine 12-1 may be monomethylated according to the procedure of J. Barluenga et al,
  • Carbamates of general formula 12-12 can be obtained in a similar manner via a variety of synthetic manipulations, all of which would be familiar to one skilled in the art.
  • the reaction of amine 12-1 witii chloroformates (see Org. Syn. Coll. Vol. IN, (1963), 780) of general formula 12-13 in an inert solvent such as ether or halocarbon in the presence of a tertiary base such as friethylamine or ethylisopropylamine readily yields the requisite carbamates of general formula 12-12 (Scheme 12-6).
  • Chloroformates of general formula 12-13 not commercially available may be readily prepared from the corresponding alcohol 12-15 in an inert solvent such as toluene, chlorocarbon or etiier by the action of excess phosgene (see Org. Syn. Coll. Vol. Ill, (1955), 167) (Scheme 12-9).
  • Amide compounds of the general formula 12-16 can be obtained via a variety of synthetic manipulations, all of which would be familiar to one skilled in the art.
  • the reaction of amine 12-1 with acid chlorides (see Org. Syn. Coll. N, (1973), 336) of general formula 12-17 to yield the desired amides 12-16 is readily achieved in an inert solvent such as chloroform or dichloromethane in the presence of an organic base such as friethylamine or emyldiisopropylamine (Scheme 12-10).
  • the corresponding acids of general formula 12-19 may be coupled with dicyclohexylcarbodiimide (DCC)/hydroxybenzofriazole (HOBT) (see W. Konig et al, Chem. Ber., (1970), 103, 788) or hydroxybenzotriazole (HOBT)/2-(lH-benzotriazole-l-yl)-l, 1,3,3- tetramethyluronium hexafluorophosphate (HBTU) (see M.
  • DCC dicyclohexylcarbodiimide
  • HOBT hydroxybenzofriazole
  • HBTU 1,3,3- tetramethyluronium hexafluorophosphate
  • a third aspect of the present invention provides a compound of Formula (I) or a solvate or physiologically functional derivative thereof for use as a tiierapeutic agent, specifically as a modifier ofthe activity of the serotonin 5-HT 2A receptor.
  • Modifiers ofthe activity ofthe serotonin 5- HT 2A receptor are believed to be of potential use for the treatment or prophylaxis of CNS, gastrointestinal, cardiovascular, and inflammatory disorders.
  • Compounds of die Formula (I) may be adieri concludedred by oral, sublingual, parenteral, rectal, or topical administration.
  • physiologically acceptable salts of the compounds may also be formed and used as therapeutic agents.
  • Different amounts of the compounds of Formula (I) will be required to achieve the desired biological effect. The amount will depend on factors such as the specific compound, the use for which it is intended, the means of adminisfration, and the condition of the treated individual.
  • a typical dose may be expected to fall in the range of 0.001 to 200 mg per kilogram of body weight of die treated individual. Unit does may contain from 1 to 200 mg of the compounds of Formula (1) and may be administered one or more times a day, individually or in multiples.
  • the salt or solvate of a compound of Formulas (I) the dose is based on the cation (for salts) or the unsolvated compound.
  • a fourth aspect of the present invention provides pharmaceutical compositions, comprising at least one compound of Formula (I) and/or an acceptable salt or solvate thereof (e.g., a pharmaceutically acceptable salt or solvate) as an active ingredient combined with at least one carrier or excipient (e.g., pharmaceutical carrier or excipient).
  • Pharmaceutical compositions may be used in die treatment of clinical conditions for which a modifier of the activity of the serotonin 5-HT 2A receptor is indicated, particularly where the active ingredient is preferentially selective for the 5-HT 2A receptor over the 5-HT 2A receptor, and most particularly where the active ingredient is also an inverse agonist at the 5-HT 2A receptor.
  • At least one compound of Formula (I) may be combined with the carrier in either solid or liquid form in a unit dose formulation.
  • the pharmaceutical carrier must be compatible with the other ingredients in tiie composition and must be tolerated by the individual recipient.
  • Other physiologically active ingredients may be inco ⁇ orated into the pharmaceutical composition of the invention if desired, and if such ingredients are compatible with the other ingredients in the composition.
  • Formulations may be prepared by any suitable method, typically by uniformly mixing the active compound(s) with liquids or finely divided solid carriers, or both, in the required proportions, and then, if necessary, forming the resulting mixture into a desired shape.
  • liquid preparations for oral administration may be in the form of solutions, emulsions, aqueous or oily suspensions, and syrups.
  • the oral preparations may be in the form of dry powder that can be reconstituted with water or another suitable liquid vehicle before use. Additional additives such as suspending or emulsifying agents, non-aqueous vehicles (including edible oils), preservatives, and flavorings and colorants may be added to the liquid preparations.
  • Parenteral dosage forms may be prepared by dissolving the compound ofthe invention in a suitable liquid vehicle and filter sterilizing the solution before filling and sealing an appropriate vial or ampoule. These are just a few examples ofthe many appropriate methods well known in the art for preparing dosage forms.
  • the fifth aspect of the present invention provides for the use of a compound of formula (I) in the preparation of a medicament for the treatment of a medical condition for which a modifier ofthe activity ofthe serotonin 5-HT 2A receptor is indicated.
  • the sixth aspect of the present invention provides for a method of treatment of a clinical condition of a mammal, such as a human, for which a modifier of the activity of the serotonin 5-HT 2A receptor is indicated, which comprises the administration to the mammal of a therapeutically effective amount of a compound of Formula (I) or a physiologically acceptable salt, solvate, or physiologically functional derivative thereof.
  • Mass spectra were recorded on a Micromass PlatformTM LC with Gilson HPLC.
  • Infra-red spectra were recorded on a Nicolet AvatarTM 360 FT-IR. Melting points were recorded on a Electrothermal IA9200TM apparatus and are uncorrected. Proton nuclear magnetic resonance spectra were recorded on a BrukerTM 300MHz machine. Chemical shifts are given with respect to teframethylsilane. In the text the following abbreviations are used; s (singlet), d (doublet), t (triplet), m (multiplet) or combinations thereof. Chemical shifts are quoted in parts per million (ppm) and with coupling constants in Hertz.
  • HPLC (250 ⁇ L; GF 254 ).
  • HPLC was recorded either on a HP ChemstationTM 1100 HPLC using a Hichrom 3.5 C18 reverse phase column (50mm x 2.1mm i.d.). Linear gradient elution over 5 minutes - 95% water (+0.1% TFA) / 5% acetonitrile (+0.05% TFA) down to 5% water / 95% acetonitrile. Flow rate 0.8mL/min (Method A); or on a Hichrom 3.5 C18 reverse phase column (100mm x 3.2mm i.d.).
  • HPLC retention time 3.28 mins (Method A).
  • Tic Rf 0.4 (EtOAc/hexane).
  • This compound is commercially available from Maybridge Chemical Company, Catalog No. KM04515, under the name N-(3-(4-bromo-l-methylpyrazol-3-yl)phenyl)(((4- trifluoromethoxy)phenyl) amino)carboxamide. :
  • IR: v raax 1684, 1592, 1510, 1253, 1217, 1157, 987, 798, 700 cm "1 .

Abstract

The present invention relates to certain pyrazole derivatives of Formula (I) and pharmaceutical compositions thereof that modulate the activity of the 5-HT2A serotonin receptor. Formula (I): Compounds and pharmaceutical compositions thereof are directed to methods useful in the prophylaxis or treatment of reducing platelet aggreagation, coronary artery disease, myocardial infarction, transient ischemic attack, angina, stroke, atrial fibrillation, reducing the risk of blood clot formation, asthma or symptoms thereof, agitation or a symptom, behavioral disorders, drug induced psychosis, excitative psychosis, Gilles de la Tourette's syndrome, manic disorder, organic or NOS psychosis, psychotic disorder, psychosis, acute schizophrenia, chronic schizophrenia and NOS schizophrenia and related disorders. The present invention also relates to the method of prophylaxis or treatment of 5-HT2A serotonin receptor mediated disorders in combination with a dopamine D2 receptor antagonist such as haloperidol, administered separately or together.

Description

SMALL MOLECULE MODULATORS OF THE 5-HT2A SEROTONIN RECEPTOR USEFUL FOR THE PROPHYLAXIS AND TREATMENT OF DISORDERS
RELATED THERETO
FIELD OF THE INVENTION The present invention relates to certain pyrazole derivatives of Formula (I) and pharmaceutical compositions thereof that modulate the activity of the 5-HT2A serotonin receptor. Compounds and pharmaceutical compositions thereof are directed to methods useful in the prophylaxis or treatment of reducing platelet aggreagation, coronary artery disease, myocardial infarction, transient ischemic attack, angina, stroke, atrial fibrillation, reducing the risk of blood clot formation, asthma or symptoms thereof, agitation or a symptom, behavioral disorders, drug induced psychosis, excitative psychosis, Gilles de la Tourette's syndrome, manic disorder, organic or NOS psychosis, psychotic disorder, psychosis, acute schizophrenia, chronic schizophrenia and NOS schizophrenia and related disorders.
The present invention also relates to the method of prophylaxis or treatment of 5-HT2A serotonin receptor mediated disorders in combination with a dopamine D2 receptor antagonist such as haloperidol, administered separately or together.
BACKGROUND OF THE INVENTION I. G protein-coupled receptors [0001] G protein-coupled receptors share a common structural motif. All these receptors have seven sequences of between 22 to 24 hydrophobic amino acids that form seven alpha helices, each of which spans the membrane. The transmembrane helices are joined by strands of amino acids having a larger loop between the fourth and fifth transmembrane helix on the extracellular side ofthe membrane. Another larger loop, composed pi±rnarily of hydrophilic amino acids, joins transmembrane helices five and six on the intracellular side of the membrane. The carboxy tenninus of the receptor lies intracellularly with the amino teiminus in the extracellular space. It is thought that the loop joining helices five and six, as well as, the carboxy terminus, interact with the G protein. Currently, Gq, Gs, Gi and Go are G proteins that have been identified. The general structure of G protein-coupled receptors is shown in Figure 1. [0002] Under physiological conditions, G protein-coupled receptors exist in the cell membrane in equiUbrium between two different states or conformations: an "inactive" state and an "active" state. As shown schematically in Figure 2, a receptor in an inactive state is unable to link to the intracellular transduction pathway to produce a biological response. Changing the receptor conformation to the active state allows linkage to the transduction pathway and produces a biological response.
[0003] A receptor may be stabilized in an active state by an endogenous ligand or an exogenous agonist ligand. Recent discoveries such as, including but not exclusively limited to, modifications to the amino acid sequence of the receptor provide means other than ligands to stabilize the active state conformation. These means effectively stabilize the receptor in an active state by simulating the effect of a ligand binding to the receptor. Stabilization by such ligand-independent means is termed "constitutive receptor activation."
Serotouin receptors [0004] Receptors for serotonin (5-hyc_oxyrrypt_mine, 5-HT) are an important class of G protein-coupled receptors. Serotonin is thought to play a role in processes related to learning and memory, sleep, thermoregulation, mood, motor activity, pain, sexual and aggressive behaviors, appetite, neurodegenerative regulation, and biological rhythms. Not surprisingly, serotonin is linked to pathophysiological conditions such as anxiety, depression, obsessive-compulsive disorders, schizophrenia, suicide, autism, migraine, emesis, alcoholism, and neurodegenerative disorders. With respect to on anti-psychotic treatment approaches focused on the serotonin receptors, these types of therapeutics can generally be divided into two classes, the "typical" and the "atypical." Both have anti- psychotic effects, but the typicals also include concomitant motor-related side effects (extra pyramidal syndromes, e.g., lip-smacking, tongue darting, locomotor movement, etc). Such side effects are thought to be associated with the compounds interacting with other receptors, such as the human dopamine D2 receptor in the nigro-striatal pathway. Therefore, an atypical treatment is preferred. Haloperidol is considered a typical anti-psychotic, and clozapine is considered an atypical anti-psychotic.
[0005] Serotonin receptors are divided into seven subfamilies, referred to as 5-HT1 through
5-HT7, inclusive. These subfamilies are further divided into subtypes. For example, the 5-HT2 subfamily is divided into three receptor subtypes: 5-HT2A, 5-HT2B, and 5-HT2c. The human 5-HT2c receptor was first isolated and cloned in 1987, and the human 5-HT2A receptor was first isolated and cloned in 1990. These two receptors are thought to be the site of action of hallucinogenic drugs. Additionally, antagonists to the 5-HT2A and 5-HT2C receptors are believed to be useful in treating depression, anxiety, psychosis, and eating disorders. [0006] U.S. Patent Number 4,985,352 describes the isolation, characterization, and expression of a functional cDNA clone encoding the entire human 5-HT1C receptor (now known as the 5-HT2C receptor). U.S. Patent Number 5,661,012 describes the isolation, characterization, and expression of a functional cDNA clone encoding the entire human 5-HT2A receptor.
[0007] Mutations of the endogenous forms of the rat 5-HT2A and rat 5-HT2C receptors have been reported to lead to constitutive activation of these receptors (5-HT2A: Casey, C. et al. (1996) Society for Neuroscience Abstracts, 22:699.10, hereinafter "Casey"; 5-HT2 : Herrick-Davis, K., and Teitler, M. (1996) Society for Neuroscience Abstracts, 22:699.18, hereinafter "Herrick-Davis 1"; and Herrick-Davis, K. et al. (1997) J. Neurochemistry 69(3): 1138, hereinafter "Herrick-Davis-2"). Casey describes a mutation of the cysteine residue at position 322 of the rat 5-HT2A receptor to lysine (C322K), glutamine (C322Q), and arginine (C322R) which reportedly led to constitutive activation. Herrick-Davis 1 and Herrick-Davis 2 describe mutations of the serine residue at position 312 of the rat 5-HT2C receptor to phenylalanine (S312F) and lysine (S312K), which reportedly led to constitutive activation.
SUMMARY OF THE INVENTION
[0008] The present invention relates to non-endogenous, constitutively activated forms ofthe human 5-HT2A and human 5-HT2C receptors and various uses of such receptors. Further disclosed are small molecule modulators of these receptors. Most preferably, these modulators have inverse agonist characteristics at the receptor.
[0009] More specifically, the present invention discloses nucleic acid molecules and the proteins for three non-endogenous, constitutively activated human serotonin receptors, referred to herein as, AP-1, AP-3, and AP-4. The AP-1 receptor is a constitutively active form of the human 5- HT2c receptor created by an S31 OK point mutation. The AP-3 receptor is a constitutively active form ofthe human 5-HT2A receptor whereby the intracellular loop 3 (IC3) portion and the cytoplasmic-tail portion of the endogenous human 5-HT2A receptor have been replaced with the IC3 portion and the cytoplasmic-tail portion of the human 5-HT2C receptor. The AP-4 receptor is a constitutively active form ofthe human 5-HT2A receptor whereby (1) the region ofthe intracellular third loop between the proline of the transmembrane 5 region (TM5) and the proline of TM6 of the endogenous human 5- HT2A receptor has been replaced with the corresponding region of the human 5-HT2C receptor (including a S310K point mutation); and (2) the cytoplasmic-tail portion ofthe endogenous human 5- HT2A receptor has been replaced with the cytoplasmic-tail portion of the endogenous human 5-HT2C receptor. [0010] The invention also provides assays that may be used to directly identify candidate compounds as agonists, partial agonists or inverse agonists to non-endogenous, constitutively activated human serotonin receptors; such candidate compounds can then be utilized in pharmaceutical composition(s) for treatment of diseases and disorders which are related to the human 5-HT2A and/or human 5-HT2C receptors.
[0011] The invention also provides compounds that exhibit high selectivity 5-HT2A activity.
More specifically, the compounds possessing 5-HT2A receptor activity are designated by the general Formula (I):
Figure imgf000006_0001
(I) wherein: i) Ri is H, halogens, NR5R6, OH or OR7, wherein
R5 and R_ are independently H, or C1-6 alkyl, or C2.6 alkenyl, or cycloalkyl, or aryl, or CH2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02, OH, OMe, OEt, CONR_R9, NR_R9, NHCOCH3, OCF3, SMe, COOR10, S03R8, SO_NR_R9, COMe, COEt, CO-lower alkyl, SCF3, CN, C2.6 alkenyl, H, halogens, CM alkoxy, C3_6 cycloalkyl, Cι-6 alkyl, and aryl wherein each of the C3.6 cycloalkyl, .6 alkyl, or aryl groups may be further optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02, OH, OMe, OEt, CONRsR9, NR8R9, NHCOCH3, OCF3, SMe, COOR10, S02NR8R9, SO3R10, COMe, COEt, CO-lower alkyl, SCF3, CN, C2.6 alkenyl, H, halogens, CM alkoxy, C3.6 cycloalkyl, Cι_6 alkyl, and aryl; or
R5 and Re may form part of a 5, 6 or 7 membered cyclic structure which may be either saturated or unsaturated and that may contain up to four heteroatoms selected from O, N or S and said cyclic structure may be optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02, OH, OMe, OEt, OCF3, SMe, COOR10, S02NR_R9, SO3R10, NHCOCH3, COEt, COMe, or halogen; R8 and R9 are independently a H, or Cι_6 alkyl, or C2.6 alkenyl, or cycloalkyl, or aryl, or CH2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from halogen, CF3, OCF3,
OEt, CC13, Me, N02, OH, OMe, SMe, COMe, CN, COOR10, SO3R10, COEt, NHCOCHs, or aryl;
Rio is H or Cι-6 alkyl; R7 is H or C1-6 alkyl; ii) R2 is H, straight chain or branched C1-6 alkyl, C2-6 alkenyl, or cycloalkyl; iii) R3 is halogen, carboxy, CN, alkoxycarbonyl, straight chain or branched Cι_6 alkyl, C2- 6 alkenyl, C2.6 alkynyl, cycloalkyl, aryl or heteroaryl, wherein each of said alkoxycarbonyl, straight chain or branched Cι_6 alkyl, C2.6 alkenyl, C2_6 alkynyl, cycloalkyl, aryl or heteroaryl groups can be optionally substituted with up to four substituents in any position selected from OH, ORio, NR8R9, halogen, -C(p)3j or -0-C(p)3 where p is halogen, and said cycloalkyl, aryl or heteroaryl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position; iv) R4 is Cι-6 alkyl, C2.β alkenyl, or cycloalkyl; v) A is C(=0), C(=S) or S02;
Figure imgf000007_0001
q is O or 1; m is 0 or 1 ; n is O or 1;
Rn and Rι2 are each independently H, straight chain or branched Cι.6 alkyl, C2-6 alkenyl, or cycloalkyl;
Figure imgf000007_0002
wherein:
Ri3» RI43 Ri5> R-16 and R17 are each independently H, halogen, CN, NR8R9, COOR10, SR10, straight chain or branched C... alkyl, C2.6 alkenyl, C2-6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl, wherein each of said alkoxycarbonyl, straight chain or branched Cι_6 alkyl, C2.6 alkenyl, C2.6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl groups can be optionally substituted with up to four substituents in any position selected from N02, OR10, NR_R9, halogen, -C(p)3> or -0-C(p)3 where p is halogen, and said cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position;
L2 is -0-Q2 wherein Q2 is straight chain or branched Cι.6 alkyl, C2.6 alkenyl, C2-6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl, wherein each of said cycloalkyl, aryl, alkylaryl, or arylalkyl groups can be optionally substituted with up to four substituents in any position selected from N02, OR7, halogen, -C(p) or -0-C(p)3 where p is halogen, or an aliphatic or aromatic heterocycle, and said cycloalkyl, aryl, alkylaryl, and arylalkyl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position; or a pharmaceutically acceptable salt. [0012] These and other aspects of the invention disclosed herein will be set forth in greater detail as the patent disclosure proceeds.
BRIEF DESCRIPTION OF THE DRAWINGS
[0013] In the following figures, bold typeface indicates the location of the mutation in the non- endogenous, constitutively activated receptor relative to the corresponding endogenous receptor.
[0014] Figure 1 shows a generalized structure of a G protein-coupled receptor with the numbers assigned to the transmembrane helices, the intracellular loops, and the extracellular loops.
[0015] Figure 2 schematically shows the active and inactive states for a typical G protein- coupled receptor and the linkage ofthe active state to the second messenger transduction pathway.
[0016] Figure 3a provides the nucleic acid sequence of the endogenous human 5-HT2A receptor (SEQ.TD.NO.:22).
[0017] Figure 3b provides the corresponding amino acid sequence ofthe endogenous human 5-HT2A receptor (SEQ._D.NO.:23). [0018] Figure 4a provides the nucleic acid sequence of the endogenous human 5-HT2C receptor (SEQ.ID.NO.:24).
[0019] Figure 4b provides the corresponding amino acid sequence ofthe endogenous human
5-HT2C receptor (SEQ.ID.NO.:25).
[0020] Figure 5a provides the nucleic acid sequence of a constitutively active form of the human 5-HT2C receptor ("AP-1 cDNA" - SEQ.ID.NO.:26).
[0021] Figure 5b provides the corresponding amino acid sequence ofthe AP-1 cDNA ("AP-
1" - SEQ.ED.NO.:27).
[0022] Figure 6a provides the nucleic acid sequence of a constitutively active form of the human 5-HT2A receptor whereby the IC3 portion and the cytoplasmic-tail portion of the endogenous 5-HT2A receptor have been replaced with the IC3 portion and the cytoplasmic-tail portion of the human 5-HT2C receptor ("AP-3 cDNA" - SEQ.ID.NO.:28).
[0023] Figure 6b provides the corresponding amino acid sequence ofthe AP-3 cDNA CAP¬
S'' - SEQ.ED.NO.:29).
[0024] Figure 6c provides a schematic representation of AP-3, where die dashed-lines represent the portion obtained from the human 5-HT2C receptor.
[0025] Figure 7a provides the nucleic acid sequence of a constitutively active form of the human 5-HT2A receptor whereby (1) the region o the between the proline of TM5 and the proline of TM6 of the endogenous human 5-HT2A receptor has been replaced with the corresponding region of the human 5-HT2C receptor (including a S310K point mutation); and (2) the cytoplasmic-tail portion of the endogenous 5-HT2A receptor has been replaced with the cytoplasmic-tail portion of the endogenous human 5-HT2C receptor ("AP-4 cDNA" - SEQ.ED.NO.:30).
[0026] Figure 7b provides the corresponding amino acid sequence ofthe AP-4 cDNA ("AP-
4" - SEQJD.NO.:31).
[0027] Figure 7c provides a schematic representation of the mutated 5-HT2A receptor of
Figure 7b where the dashed-lines represent the portion obtained from the human 5-HT2C receptor.
[0028] Figure 8 is a representation of the preferred vector, pCMV, used herein. [0029] Figure 9 is a diagram illustrating (1) enhanced (35S)GTPτS binding to membranes prepared from COS cells expressing the endogenous human 5-HT2C receptor in response to serotonin, and (2) inhibition by mianserin using wheatgerm agglutinin scintillation proximity beads. The concentration of (35S)GTP">S was held constant at 0.3 nM, and the concentration of GDP was held at 1 μM. The concentration ofthe membrane protein was 12.5 μg.
[0030] Figure 10 is a diagram showing serotonin stimulation of (35S)GTPτS binding to membranes expressing AP-1 receptors in 293T cells and the inhibition by 30 μM mianserin on Wallac™ scintistrips.
[0031] Figures 11A and UB are diagrams showing the effects of protein concentration on (35S)GTP-yS binding in membranes prepared from 293T cells transfected with the endogenous human 5-HT2C receptors and AP-1 receptors compared to cells transfected with the control vector (pCMV) alone in the absence (Figure 11 A) and presence (Figure 11B) of 10 μM serotonin. The radiolableled concentration of (35S)GTP' S was held constant at 0.3 nM, and the GDP concentration was held constant at 1 μM. The assay was perfonned on 96-well format on Wallac™ scintistrips.
[0032] Figure 12 provides bar-graph comparisons of inositol tris-phosphate ("IP3") production between the endogenous human 5-HT2A receptor and AP-2, a mutated form of the receptor.
[0033] Figure 13 provides bar-graph comparisons of inositol tris-phosphate ("IP3") production between the endogenous human 5-HT2A receptor and AP-4, a mutated form of the receptor.
[0034] Figure 14 provides bar graph comparisons of IP3 production between the endogenous human 5-HT2A receptor and AP-3, a mutated form ofthe receptor.
[0035] Figure 15 provides bar-graph comparisons of IP3 production between the endogenous human 5-HT2C receptor and AP-1.
[0036] Figures 16A-C provides representative autoradiograms showing displacement of I125-
LSD from brain sections by spiperone and Compound 1.
[0037] Figures 17A-C show in vivo response of animals to Compound 2 exposure. DEFINITIONS
[0038] The scientific literature that has evolved around receptors has adopted a number of terms to refer to ligands having various effects on receptors. For clarity and consistency, the following definitions will be used throughout this patent document. To the extent that these definitions conflict with other definitions for these terms, the following definitions shall control.
[0039] AGONISTS shall mean moieties that activate the intracellular response when they bind to the receptor, or enhance GTP binding to membranes.
[0040] AMINO ACID ABBREVIATIONS used herein are set out in TABLE 1 :
TABLE 1
Figure imgf000011_0001
[0041] PARTIAL AGONISTS shall mean moieties which activate the intracellular response when they bind to the receptor to a lesser degree/extent than do agonists, or enhance GTP binding to membranes to a lesser degree/extent than do agonists.
[0042] ANTAGONIST shall mean moieties that competitively bind to the receptor at the same site as the agonists but which do not activate the intracellular response initiated by the active form of the receptor, and can thereby inhibit the intracellular responses by agonists or partial agonists. ANTAGONISTS do not diminish the baseline intracellular response in the absence of an agonist or partial agonist.
[0043] CANDIDATE COMPOUND shall mean a molecule (for example, and not limitation, a chemical compound) which is amenable to a screening technique.
[0044] COMPOSITION shall mean a material comprising at least two compounds or two components; for example, and not limitation, a Pharmaceutical Composition is a Composition.
[0045] COMPOUND EFFICACY shall mean a measurement of the ability of a compound to inhibit or stimulate receptor functionality, as opposed to receptor binding affinity.
[0046] CONSTITUTIVELY ACTIVATED RECEPTOR shall mean a receptor subject to constitutive receptor activation.
[0047] CONSTITUTIVE RECEPTOR ACTIVATION shall mean stabilization of a receptor in the active state by means other than binding of the receptor with its endogenous ligand or a chemical equivalent thereof.
[0048] CONTACT or CONTACTING shall mean bringing at least two moieties together, whether in an in vitro system or an in vivo system.
[0049] ENDOGENOUS shall mean a material that a mammal naturally produces.
ENDOGENOUS in reference to, for example and not limitation, the term "receptor" shall mean that which is naturally produced by a mammal (for example, and not limitation, a human) or a virus.
[0050] In contrast, the term NON-ENDOGENOUS in this context shall mean that which is not naturally produced by a mammal (for example, and not limitation, a human) or a virus. For example, and not limitation, a receptor which is not constitutively active in its endogenous form, but when manipulated becomes constitutively active, is most preferably referred to herein as a "non-endogenous, constitutively activated receptor." Both terms can be utilized to describe both "in vivo" and "in vitro" systems. For example, and not a limitation, in a screening approach, the endogenous or non-endogenous receptor may be in reference to an in vitro screening system. As a further example and not hmitation, where the genome of a mammal has been manipulated to include a non-endogenous constitutively activated receptor, screening of a candidate compound by means of an in vivo system is viable.
[0051] INHIBIT or INHIBITING, in relationship to the term "response" shall mean that a response is decreased or prevented in the presence of a compound as opposed to in the absence of the compound.
[0052] INVERSE AGONISTS shall mean moieties that bind the endogenous form of the receptor or to the constitutively activated form ofthe receptor, and which inhibit the baseline intracellular response initiated by the active form of the receptor below the normal base level of activity which is observed in the absence of agonists or partial agonists, or decrease GTP binding to membranes. Preferably, the baseline intracellular response is inhibited in the presence ofthe inverse agonist by at least 30%, more preferably by at least 50%, and most preferably by at least 75%, as compared with the baseline response in the absence ofthe inverse agonist.
[0053] LIGAND shall mean an endogenous, naturally occurring molecule specific for an endogenous, naturally occurring receptor.
[0054] As used herein, the terms MODULATE or MODULATING shall mean to refer to an increase or decrease in the amount, quality, response or effect of a particular activity, function or molecule. For example, Compounds which modulate/capable of modulating the 5-HT2A activity include agonists, inverse agonists, antagonists, inhibitors, activators, and compounds which directly or indirectly affect regulation ofthe 5-HT2A activity.
[0055] PHARMACEUTICAL COMPOSITION shall mean a composition comprising at least one active ingredient, whereby the composition is amenable to investigation for a specified, efficacious outcome in a mammal (for example, and not limitation, a human). Those of ordinary skill in the art will understand and appreciate the techniques appropriate for determining whether an active ingredient has a desired efficacious outcome based upon the needs ofthe artisan.
[0056] STIMULATE or STIMULATING, in relationship to the term "response" shall mean that a response is increased in the presence of a compound as opposed to in the absence ofthe compound. DETAILED DESCRIPTION
[0057] In addition to the foregoing beneficial uses for the modulators of 5-HT2A receptor activity disclosed herein, the compounds disclosed herein are believed to be useful in the treatment of several additional diseases and disorders, and in the amelioration of symptoms thereof. Without limitation, these include the following:
USE OF THE COMPOUNDS OF THE INVENTION 1. Antiplatelet Therapies (5-HT2A mediated platelet aggregation): [0058] Antiplatelet agents (antiplatelets) are prescribed for a variety of conditions. For example, in coronary artery disease they are used to help prevent myocardial infarction or stroke in patients who are at risk of developing obstructive blood clots (e.g., coronary thrombosis).
[0059] In a myocardial infarction (heart attack), the heart muscle does not receive enough oxygen-rich blood as a result of a blockage in the coronary blood vessels. If taken while an attack is in progress or ήnmediately afterward (preferably within 30 minutes), antiplatelets can reduce the damage to the heart.
[0060] A transient ischemic attack ("TIA" or "mini-stroke") is a brief imterruption of oxygen flow to the brain due to decreased blood flow through arteries, usually due to an obstructing blood clot. Antiplatelet drugs have been found to be effective in preventing TIAs.
[0061] Angina is a temporary and often recurring chest pain, pressure or discomfort caused by inadequate oxygen-rich blood flow (ischemia) to some parts of the heart. In patients with angina, antiplatelet therapy can reduce the effects of angina and the risk of myocardial infarction.
[0062] Stroke is an event in which the brain does not receive enough oxygen-rich blood, usually due to blockage of a cerebral blood vessel by a blood clot. In high-risk patients, taking antiplatelets regularly has been found to prevent the formation blood clots that cause first or second strokes.
[0063] Angioplasty is a catheter based technique used to open arteries obstructed by a blood clot. Whether or not stenting is performed immediately after this procedure to keep the artery open, antiplatelets can reduce the risk of forming additional blood clots following the procedure(s). [0064] Coronary bypass surgery is a surgical procedure in which an artery or vein is taken from elsewhere in the body and grafted to a blocked coronary artery, rerouting blood around the blockage and through the newly attached vessel. After the procedure, antiplatelets can reduce the risk of secondary blood clots.
[0065] Atrial fibrillation is the most common type of sustained irregular heart rhythm
(arrythmia). Atrial fibrillation affects about two million Americans every year. In atrial fibrillation, the atria (the heart's upper chambers) rapidly fire electrical signals that cause them to quiver rather than contract normally. The result is an abnormally fast and highly irregular heartbeat. When given after an episode of atrial fibrillation, antiplatelets can reduce the risk of blood clots forming in the heart and traveling to the brain (embolism).
[0066] 5-HT2A receptors are expressed on smooth muscle of blood vessels and 5-HT secreted by activated platelets causes vasoconstriction as well as activation of additional platelets during clotting. There is evidence that a 5-HT2A inverse agonist will inhibit platelet aggregation and thus be a potential treatment as an antiplatelet therapy. See Satimura, K, et al., Clin Cardiol 2002 Jan. 25 (l):28-32; and Wilson, H.C et al., Thromb Haemost 1991 Sep 2;66(3):355-60.
[0067] The 5-HT2A inverse agonists disclosed herein provide beneficial improvement in microcirculation to patients in need of antiplatelet therapy by antagonizing the vasoconstrictive products of the aggregating platelets in, for example and not limitation, the indications described above. Accordingly, in some embodiments, the present invention provides methods for reducing platelet aggregation in a patient in need thereof comprising administering to said patient a composition comprising a 5-HT2A inverse agonist disclosed herein. In further embodiments, the present invention provides methods for treating coronary artery disease, myocardial infarction, transient ischemic attack, angina, stroke, atrial fibrillation, or a symptom of any of the foregoing in a patient in need of said treatment, comprising administering to said patient a composition comprising a 5-HT2A inverse agonist disclosed herein.
[0068] In further embodiments, the present invention provides methods for reducing risk of blood clot formation in a angioplasty or coronary bypass surgery patient, or a patient suffering from atrial fibrillation, comprising administering to a said patient a composition comprising a 5-HT2A inverse agonist disclosed herein at a time where such risk exists.
2. Asthma
[0069] It has been suggested that 5-HT (5-hydroxytryptamine) plays a role in the pathophysiology of acute asthma. See Cazzola, M. and Matera, M.G., TIPS, 2000, 21, 13; and De Bie, JJ. et al., British J. Pharm., 1998, 124, 857-864. The compounds of the present invention disclosed herein are useful in the treatment of asthma, and the treatment of the symptoms thereof. Accordingly, in some embodiments, the present invention provides methods for treating asthma in a patient in need of said treatment, comprising administering to said patient a composition comprising a 5-HT2A inverse agonist disclosed herein. In further embodiments, methods are provided for treating a symptom of asthma in a patient in need of said treatment, comprising administering to said patient a composition comprising a 5-HT2A inverse agonist disclosed herein.
3. Agitation
[0070] Agitation is a well-recognized behavioral syndrome with a range of symptoms, including hostility, extreme excitement, poor impulse control, tension and uncooperativeness (See Cohen-Mansfield J, and Billig, N., (1986), Agitated Behaviors in the Elderly. I. A Conceptual Review. J Am Geriatr Soc 34(10): 711-721).
[0071] Agitation is a common occurrence in the elderly and often associated with dementia such as those caused by Alzheimer's disease, Lewy Body, Parkinson's, and Huntington's, which are degenerative diseases of the nervous system and by diseases that affect blood vessels, such as stroke, or multi-infarct dementia, which is caused by multiple strokes in the brain can also induce dementia. Alzheimer's disease accounts for approximately 50 to 70% of all dementias (See Koss E, et al., (1997), Assessing patterns of agitation in Alzheimer's disease patients with the Cohen-Mansfield Agitation Inventory. The Alzheimer's Disease Cooperative Study. Alzheimer Dis Assoc Disord ll(suppl 2):S45-S50).
[0072] An estimated five percent of people aged 65 and older and up to 20 percent of those aged 80 and older are affected by dementia. Of these sufferers, nearly half exhibit behavioral disturbances, such as agitation, wandering and violent outbursts.
[0073] Agitated behaviors can also be manifested in cognitively intact elderly people and by those with psychiatric disorders other than dementia
[0074] Agitation is often treated with antipsychotic medications such as haloperidol in nursing home and other assisted care settings. There is emerging evidence that agents acting at the 5- HT2A receptors in the brain have the effects of reducing agitation in patients, including Alzheimer's dementia (See Katz, I.R., et al., J Clin Psychiatry 1999 Feb., 60(2):107-115; and Street, J.S., et al., Arch Gen Psychiatry 2000 Oct., 57(10):968-976). [0075] The compounds ofthe invention disclosed herein are useful for treating agitation and symptoms thereof. Thus, in some embodiments, the present invention provides methods for treating agitation in a patient in need of such treatment comprising administering to said patient a composition comprising a 5-HT2A inverse agonist disclosed herein. In some embodiments, the agitation is due to a psychiatric disorder other than dementia. In some embodiments, the present invention provides methods for treatment of agitation or a symptom thereof in a patient suffering from dementia comprising administering to said patient a composition comprising a 5-HT2A inverse agonist disclosed herein. In some embodiments of such methods, the dementia is due to a degenerative disease of the nervous system, for example and without limitation, Alzheimers disease, Lewy Body, Parkinson's disease, and Huntington's disease, or dementia due to diseases that affect blood vessels, including with out limitation stroke and multi-infarct dementia. In some embodiments, methods are provided for treating agitation or a symptom thereof in a patient in need of such treatment, where the patient is a cognitively intact elderly patient, comprising administering to said patient a composition comprising a 5-HT2A inverse agonist disclosed herein.
4. Add-on therapy to Haloperidol in the treatment of schizophrenia and other disorders: [0076] Schizophrenia is a psychopathic disorder of unknown origin, which usually appears for the first time in early adulthood and is marked by a number of characteristics, psychotic symptoms, progression, phasic development and deterioration in social behavior and professional capability in the region below the highest level ever attained. Characteristic psychotic symptoms are disorders of thought content (multiple, fragmentary, incoherent, implausible or simply delusional contents or ideas of persecution) and of mentality (loss of association, flight of imagination, incoherence up to incomprehensibility), as well as disorders of perceptibility (hallucinations), of emotions (superficial or inadequate emotions), of self-perception, of intentions and impulses, of interhuman relationships, and finally psychomotoric disorders (such as catatonia). Other symptoms are also associated with this disorder. (See, American Statistical and Diagnostic Handbook).
[0077] Haloperidol (Haldol) is a potent dopamine D2 receptor antagonist. It is widely prescribed for acute schizophrenic symptoms, and is very effective for the positive symptoms of schizophrenia. However, Haldol is not effective for the negative symptoms of schizophrenia and may actually induce negative symptoms as well as cognitive dysfunction. In accordance with some methods of the invention, adding a 5-HT2A inverse agonist concomitantly with Haldol will provide benefits including the ability to use a lower dose of Haldol without losing its effects on positive symptoms, while reducing or eliminating its inductive effects on negative symptoms, and prolonging relapse to the patient's next schizophrenic event. [0078] Haloperidol is used for treatment of a variety of behavioral disorders, drug induced psychosis, excitative psychosis, Gilles de la Tourette's syndrome, manic disorders, psychosis (organic and NOS), psychotic disorder, psychosis, schizophrenia (acute, chronic and NOS). Further uses include in the treatment of infantile autism, huntington's chorea, and nausea and vomiting from chemotherapy and chemotherapeutic antibodies. Administration of 5-HT2A inverse agonists disclosed herein with haloperidol also will provide benefits in these indications.
[0079] In some embodiments, the present invention provides methods for treating a behavioral disorder, drug induced psychosis, excitative psychosis, Gilles de la Tourette's syndrome, manic disorders, psychosis (organic and NOS), psychotic disorder, psychosis, schizophrenia (acute, chronic and NOS) comprising administering to said patient a dopamine D2 receptor antagonist and a 5-HT2A inverse agonist disclosed herein.
[0080] In some embodiments, the present invention provides methods for treating a behavioral disorder, drug induced psychosis, excitative psychosis, Gilles de la Tourette's syndrome, manic disorders, psychosis (organic and NOS), psychotic disorder, psychosis, schizophrenia (acute, chronic and NOS) comprising administering to said patient haloperidol and a 5-HT2A inverse agonist disclosed herein.
[0081] In some embodiments, the present invention provides methods for treating infantile autism, huntington's chorea, or nausea and vomiting from chemotherapy or chemotherapeutic antibodies comprising administering to said patient a dopamine D2 receptor antagonist and a 5-HT2A inverse agonist disclosed herein.
[0082] In some embodiments, the present invention provides methods for treating infantile autism, huntington's chorea, or nausea and vomiting from chemotherapy or chemotherapeutic antibodies comprising administering to said patient haloperidol and a 5-HT2A inverse agonist disclosed herein.
[0083] In further embodiments, the present invention provides methods for treating schizophrenia in a patient in need of said treatment comprising administering to said patient a dopamine D2 receptor antagonist and a 5-HT2A inverse agonist disclosed herein. Preferably, the dopamine D2 receptor antagonist is haloperidol.
[0084] The administration of the dopamine D2 receptor antagonist can be concomitant with administration ofthe 5-HT2A inverse agonist, or they can be administered at different times. Those of skill in the art will easily be able to determine appropriate dosing regimes for the most efficacious reduction or elimination of deleterions haloperidol effects. In some embodiments, haloperidol and the 5-HT2A inverse agonist are administered in a single dosage form, and in other embodiments, they are administered in separate dosage forms.
[0085] The present invention further provides methods of alleviating negative symptoms of schizophrenia induced by the administration of haloperidol to a patient suffering from said schizophrenia, comprising administering to said patient a 5-HT2A inverse agonist as disclosed herein.
PARTICULARLY PREFERRED MUTATIONS
[0086] For convenience, the sequence information regarding the non-endogenous, constitutively active human 5-HT2A and 5-HT2c receptors are referred to by identifiers as set forth in TABLE 2:
TABLE 2
Figure imgf000019_0001
As will be discussed in greater detail below, a mutation analogous to that reported by Casey (C322K) was utilized in the human 5-HT2A receptor and is referred to herein as AP-2. However, AP-2 did not lead to sufficient constitutive activation to allow for utilization in screening techniques.
INTRODUCTION
[0087] While it is sometimes possible to make predictions as to the effect of nucleic acid manipulation from one species to another, this is not always the case. The results reported by Casey suggest that a point mutation in the rat 5-HT2A receptor evidences constitutive activation of the mutated receptor. Casey reports that the C322K mutation was approximately four fold more active than the native rat 5-HT2A receptor. However, for purposes of a most preferred use, i.e., screening of candidate compounds, this corresponding mutation in the human 5-HT2A receptor had little discernable effect in evidencing constitutive activation ofthe human receptor. This, of course, creates the reasonable conclusion that the information reported in Herrick-Davis 1 or Herrick-Davis 2 is of limited predictive value relative to the manipulation ofthe human 5-HT2c receptor. Consequently, the ability to make reasonable predictions about the effects of mutations to the rat 5-HT receptors vis-a- vis the corresponding human receptors is not possible. Nonetheless, this unfortunate lack of reasonable predictability provides the opportunity for others to discover mutations to the human 5-HT receptors that provide evidence of constitutive activation.
[0088] Therefore, the present invention is based upon the desire of defining mutated sequences ofthe human serotonin receptors 5-HT2A and 5-HT2C whereby such mutated versions ofthe expressed receptor are constitutively active. These constitutively active receptors allow for, inter alia, screening candidate compounds.
[0089] What has been discovered and disclosed herein is that substantial activation of the human 5-HT2A receptor can be obtained by "domain swapping," i.e., by switching the third intracellular domain ofthe 5-HT2A receptor with the third intracellular domain ofthe 5-HT2C receptor. Additionally, swapping the cytoplasmic tail of the two receptors further increases the IP3 response. Furthermore, mutation of the serine at position 310 to lysine (S310K) of the human 5-HT2C receptor leads to constitutive activation.
[0090] What follows is a most preferred approach to identification of candidate compounds; those in the art will readily appreciate that the particular order of screening approaches, and/or whether or not to utilize certain of these approaches, is a matter of choice. Thus, the order presented below, set for presentational efficiency and for indication of the most preferred approach utilized in screening candidate compounds, is not intended, nor is to be construed, as a limitation on the disclosure, or any claims to follow.
GENERIC G PROTEIN-COUPLED RECEPTOR SCREENING ASSAY TECHNIQUES
[0091] When a G protein receptor becomes constitutively active, it binds to a G protein (Gq, Gs,
Gi, Go) and stimulates the binding of GTP to the G protein. The G protein then acts as a GTPase and slowly hydrolyzes the GTP to GDP, whereby the receptor, under normal conditions, becomes deactivated. However, constitutively activated receptors continue to exchange GDP to GTP. A non- hydrolyzable analog of GTP, (35S)GTPγS, can be used to monitor enhanced binding to membranes which express constitutively activated receptors. It is reported that (35S)GTPyS can be used to monitor G protein coupling to membranes in the absence and presence of ligand. An example of this monitoring, among other examples well-known and available to those in the art, was reported by Traynor and Nahorski in 1995. The preferred use of this assay system is for initial screening of candidate compounds because the system is generically applicable to all G protein-coupled receptors regardless ofthe particular G protein that interacts with the intracellular domain ofthe receptor. .
CONFIRMATION OF G PROTEIN-COUPLED RECEPTOR SITE SCREENING ASSAY TECHNIQUES
[0092] Once candidate compounds are identified using the "generic" G protein-coupled receptor assay (i.e. an assay to select compounds that are agonists, partial agonists, or inverse agonists), further screening to confirm that the compounds have interacted at the receptor site is preferred. For example, a compound identified by the "generic" assay may not bind to the receptor, but may instead merely "uncouple" the G protein from the intracellular domain. Thus, by further screening those candidate compounds, which have been identified using a "generic" assay in an agonist and/or antagonist competitive binding assay, further refinement in the selection process is provided.
[0093] Lysergic acid diethylamide (LSD) is a well-known agonist ofthe 5-HT2A and 5-HT2C receptors, while mesulergine is a well-known antagonist to the 5-HT2C receptor. Accordingly, in most preferred embodiments, an agonist (LSD) and/or antagonist (mesulergine) competitive binding assay(s) is used to further screen those compounds selected from the "generic" assay for confirmation of serotonin receptor binding.
SPECIFIED G PROTEIN ASSAY TECHNIQUES
[0094] The art-accepted physiologically mediated pathway for the human 5-HT2A and 5-
HT2C receptors is via Gq. Intracellular accumulation of IP3 can be used to confirm constitutive activation of these types of Gq coupled receptors (see Herrick-Davis-1). As a result, "IP3 accumulation" assays can be used to further screen those compounds selected from an agonist and/or antagonist competitive binding assay. .
PHARMACEUTICAL COMPOSITIONS [0095] Candidate compounds selected for further development can be formulated into pharmaceutical compositions using techniques well known to those in the art. Suitable pharmaceutically- acceptable carriers are available to those in the art; for example, see Remington's Pharmaceutical Sciences, 16th Edition, 1980, Mack Publishing Co., (Oslo et al., eds.).
EXAMPLES [0096] The following examples are presented for purposes of elucidation, and not limitation, of the present invention. While specific nucleic acid and amino acid sequences are disclosed herein, those of ordinary skill in the art are credited with the ability to make minor modifications to these sequences while achieving the same or substantially similar results reported below. It is intended that equivalent, non-endogenous, constitutively activated human serotonin receptor sequences include those having eighty-five percent (85%) homology, more preferably having ninety percent (90%) homology, and most preferably having ninety-five percent (95%) homology to the disclosed and claimed sequences all fall within the scope of any claims appended hereto.
EXAMPLE 1 GENERATION OF NON-ENDOGENOUS, CONSTITUTIVELY ACTIVATED HUMAN SEROTONIN RECEPTORS 5-HT2C AND 5-HT2A
A. Construction of constitutively active 5-HT2C receptor cDNA 1. Endogenous Human 5-HT2C [0097] The cDNA encoding endogenous human 5-HT2C receptor was obtained from human brain ρoly-A+ RNA by RT-PCR. The 5' and 3' primers were derived from the 5' and 3' untranslated regions and contained the following sequences: . 5'-GACCTCGAGGTTGCTTAAGACTGAAGCA-3' (SEQ.ID.NO.:l) 5'-ATTTCTAGACATATGTAGCTTGTACCGT-3' (SEQ.ID.NO.:2) PCR was performed using either TaqPlus™ precision polymerase (Stratagene) or rTth™ polymerase (Perkin Elmer) with the buffer systems provided by the manufacturers, 0.25 μM of each primer, and 0.2 mM of each ofthe four (4) nucleotides. The cycle condition was 30 cycles of 94°C for 1 minute, 57 °C for 1 minute and 72 °C for 2 minutes. The 1.5 kb PCR fragment was digested with Xho I and Xba I and subcloned into the Sal I-Xba I site of pBluescript.
[0098] The derived cDNA clones were fully sequenced and found to correspond to published sequences. 2. AP-1 cDNA
[0099] The cDNA containing a S310K mutation (AP-1 cDNA) in the third intracellular loop, of the human 5-HT2C receptor was constructed by replacing the Sty I restriction fragment containing amino acid 310 with synthetic double stranded oligonucleotides encoding the desired mutation. The sense strand sequence utilized had the following sequence:
5 '-CTAGGGGCACCATGCAGGCTATCAACAATGAAAGAAAAGCTAAGAAAGTC-3 ' (SEQ._D.NO.:3) and the antisense strand sequence utilized had the following sequence: 5'-CAAGGACTITCTTAGCTTTTCTTrCATTGTTGATAGCCTGCATGGTGCCC-3' (SEQ.π>.NO.:4).
B. Construction of constitutively active 5-HT2A receptor cDNA
1. Endogenous Human 5-HT2A
[0100] The cDNA encoding endogenous human 5-HT2A receptor was obtained by RT-PCR using human brain poly-A+ RNA; a 5 ' primer from the 5 ' untranslated region with a Xho I restriction site:
5'-GACCTCGAGTCCTTCTACACCTCATC-3' (SEQ.ID.NO.:5) and a 3' primer from the 3' untranslated region containing an Xba I site: 5'-TGCTCTAGATTCCAGATAGGTGAAAA CTTG-3' (SEQJD.NO.:6). PCR was performed using either TaqPlus™ precision polymerase (Stratagene) or rTth™ polymerase (Perkin Elmer) with the buffer systems provided by the manufacturers, 0.25 μM of each primer, and 0.2 mM of each ofthe four (4) nucleotides. The cycle condition was 30 cycles of 94°C for 1 minute, 57 °C for 1 minute, and 72 °C for 2 minutes. H e 1.5 kb PCR fragment was digested with Xba I and subcloned into the Eco RV-Xba I site of pBluescript.
[0101] The resulting cDNA clones were fully sequenced and found to encode two amino acid changes from the published sequences. The first change is a T25N mutation in the N-terminal extracellular domain and the second change is an H452Y mutation. These mutations are likely to represent sequence polymorphisms rather than PCR errors since the cDNA clones having the same two mutations were derived from two independent PCR procedures using Taq polymerase from two different commercial sources (TaqPlus™ Stratagene and rTth™ Perkin Elmer).
2. Human 5-HT2A (C322K; AP-2)
[0102] The cDNA containing the point mutation C322K in the third intracellular loop was constructed by using the Sph I restriction enzyme site, which encompasses amino acid 322. For the PCR procedure, a primer containing the C322K mutation: 5'-CAAAGAAAGTACTGGGCATCGTCTTCTTCCT-3' (SEQ.ro.NO.:7) was used along with the primer from the 3' untranslated region set forth above as SEQ.LD.NO.:6. The resulting PCR fragment was then used to replace the 3' end ofthe wild type 5-HT2A cDNA by the T4 polymerase blunted Sph I site. PCR was performed using pfu polymerase (Stratagene) with the buffer system provided by the manufacturer and 10% DMSO, 0.25 mM of each primer, 0.5mM of each of the 4 nucleotides. The cycle conditions were 25 cycles of 94°C for 1 minute, 60°C for 1 minute, and 72°C for 1 minute.
3. AP-3 cDNA
[0103] The human 5-HT2A cDNA with intracellular loop 3 (IC3) or IC3 and cytoplasmic tail replaced by the corresponding human 5-HT2C cDNA was constructed using PCR-based mutagenesis. (a) Replacement of IC3 Loop [0104] The IC3 loop of human 5-HT2A cDNA was first replaced with the corresponding human 5-HT2C cDNA. Two separate PCR procedures were performed to generate the two fragments, Fragment A and Fragment B, that fuse the 5-HT2C IC3 loop to the transmembrane 6 (TM6) of 5-HT2A. The 237 bp PCR fragment, Fragment A, containing 5-HT2C IC3 and the initial 13 bp of 5-HT2A TM6 was amplified by using the following primers:
5'-CCGCTCGAGTACTGCGCCGACAAGCTTTGAT-3 ' (SEQ.ID.NO.:8) 5'-CGATGCCCAGCACTTTCGAAGCTTTTCTTTCATTGTTG-3'(SEQ.ID.NO.:9) The template used was human 5-HT2c cDNA.
[0105] The 529 bp PCR fragment, Fragment B, containing the C-tenninal 13 bp of IC3 from
5-HT2C and die C-teiminal of 5-HT2A starting at beginning of TM6, was amplified by using the following primers:
5'-AAAAGCTTCGAAAGTGCTGGGCATCGTCTTCTTCCT-3' (SEQ.IO.NO.:10) 5'-TGCTCTAGATTCCAGATAGGTGAAAACTTG-3' (SEQ.ID.NO.:ll) The template used was human 5-HT2A cDNA.
[0106] Second round PCR was performed using Fragment A and Fragment B as co-templates with SEQ.ID.NO.:8 and SEQ.ID.NO.:ll (it is noted that the sequences for SEQ.ID.NOS.:6 and 11 are the same) as primers. The resulting 740 bp PCR fragment, Fragment C, contained the IC3 loop of human 5-HT2C fused to TM6 through the end of the cytoplasmic tail of human 5-HT2A. PCR was performed using pfu™ polymerase (Stratagene) with the buffer system provided by the manufacturer, and 10% DMSO, 0.25 mM of each primer, and 0.5 mM of each ofthe four (4) nucleotides. The cycle conditions were 25 cycles of 94 °C for 1 minute, 57 °C (1st round PCR) or 60 °C (2nd round PCR) for 1 minute, and 72 °C for 1 minute (1st round PCR) or 90 seconds (2nd round PCR). [0107] To generate a PCR fragment containing a fusion junction between the human 5-HT2A
TM5 and the IC3 loop of 5-HT2C, four (4) primers were used. The two external primers, derived from human 5-HT2A, had the following sequences: 5'-CGTGTCTCTCCTTACTTCA-3' (SEQ.ID.NO.:12)
[0108] The other primer used was SEQ.ID.NO.:6 (see note above regarding
SEQ.ED.NOS.:6 and 11). The first internal primer utilized was an antisense strand containing the initial 13 bp of IC3 of 5-HT2C followed by the terminal 23 bp derived from TM5 of 5-HT2A: 5'-TCGGCGCAGTACTTTGATAGTTAGAAAGTAGGTGAT-3' (SEQ.ID.NO.:13)
[0109] The second internal primer was a sense strand containing the teπninal 14 bp derived from TM5 of 5-HT2A followed by the initial 24 bp derived from IC3 of 5-HT2C:
5'-TTCTAACTATCAAAGTACTGCGCCGACAAGCTTTGATG-3' (SEQ.ID.NO.:14).
[0110] PCR was performed using endogenous human 5-HT2A and a co-template, Fragment
C, in a 50 ml reaction volume containing IX pfu buffer, 10% DMSO, 0.5 mM of each ofthe four (4) nucleotides, 0.25 mM of each external primer (SEQ.ED.NOS.:ll and 12), 0.06 mM of each internal primer (SEQ.LD.NOS.:13 and 14) and 1.9 units of pfu polymerase (Stratagene). The cycle conditions were 25 cycles of 94°C for 1 minute, 52°C for 1 minute, and 72 °C for 2 minutes and 10 seconds. The 1.3 kb PCR product was then gel purified and digested with Pst I and Eco RI. The resulting 1 kb Pstl- Eco RI fragment was used to replace the corresponding fragment in the endogenous human 5-HT2A sequence to generate the mutant 5-HT2A sequence encoding the IC3 loop of 5-HT2C.
(b) Replacement of the cytoplasmic tail
[0111] To replace the cytoplasmic tail of 5-HT2A with that of 5-HT2C, PCR was performed using a sense primer containing the C-teπninal 22 bp of TM7 of endogenous human 5-HT2A followed by the initial 21 bp ofthe cytoplasmic tail of endogenous human 5-HT2C: 5'-TTCAGCAGTCAACCCACTAGTCTATACTCTGTTCAACAAAATT-3 ' (SEQ.LO.NO.:15) The antisense primer was derived from the 3 ' untranslated region of endogenous human 5-HT2C: 5'-ATTTCTAGACATATGTAGCTTGTACCGT-3' (SEQ.ID.NO.:16).
[0112] The resulting PCR fragment, Fragment D, contained the last 22 bp of endogenous human 5-HT2A TM7 fused to the cytoplasmic tail of endogenous human 5-HT2C. Second round PCR was performed using Fragment D and the co-template was endogenous human 5-HT2A that was previously digested with Ace I to avoid undesired amplification. The antisense primer used was SEQ.ID.NO.:16 (the sequences for SEQ.LD.NOS.:16 and 2 are the same) and the sense primer used was derived from endogenous human 5-HT2A: 5 '-ATCACCTACTTTCTAACTA-3 ' (SEQ.ID.NO.:17).
[0113] PCR conditions were as set forth in Example lB3(a) for the first round PCR, except that the annealing temperature was 48 °C and the extension time was 90 seconds. The resulting 710 bp PCR product was digested with Apa I and Xba I and used to replace the corresponding Apa I-Xba I fragment of either (a) endogenous human 5-HT2A, or (b) 5-HT2A with 2C IC3 to generate (a) endogenous human 5-HT2A with endogenous human 5-HT2c cytoplasmic tail and (b) AP-3, respectively.
4. AP-4 cDNA
[0114] This mutant was created by replacement ofthe region of endogenous human 5-HT2A from amino acid 247, the middle of TM5 right after Pro246, to amino acid 337, the middle of TM6 just before Pro338, by the corresponding region of AP-1 cDNA. For convenience, the junction in TM5 is referred to as the "2 A-2C junction," and the junction in TM6 is referred to as the "2C-2A junction."
[0115] Three PCR fragments containing the desired hybrid junctions were generated. The 5' fragment of 561 bp containing the 2A-2C junction in TM5 was generated by PCR using endogenous human 5-HT2A as template, SEQ.EO.NO.:12 as the sense primer, and the antisense primer was derived from 13 bp of 5-HT2C followed by 20 bp of 5-HT2A sequence: 5'-CCATAATCGTCAGGGGAATGAAAAATGACACAA-3' (SEQ.ED.NO.:18)
[0116] The middle fragment of the 323 bp contains endogenous human 5-HT2C sequence derived from the middle of TM5 to the middle of TM6, flanked by 13 bp of 5-HT2A sequences from the 2A-2C junction and the 2C-2A junction. This middle fragment was generated by using AP-1 cDNA as a template, a sense primer containing 13 bp of 5-HT2A followed by 20 bp of 5-HT2C sequences across the 2A-2C junction and having the sequence:
and an antisense primer containing 13 bp of 5-HT2A followed by 20 bp of 5-HT2C sequences across the 2C-2A junction and having the sequence:
5'-TGATGAAGAAAGGGCACCACATGATCAGAAACA-3' (SEQ.ro.NO.:20).
The 3' fragment of 487 bp containing the 2C-2A junction was generated by PCR using endogenous human 5-HT2A as a template and a sense primer having the following sequence from the 2C-2A junction: 5'-GATCATGTGGTGCCCTTTCTTCATCACAAACAT-3' (SEQ.ED.NO.:21) and the antisense primer was SEQ.ID.NO.:6 (see note above regarding SEQ.ID.NOS.:6 and 11).
[0117] Two second round PCR reactions were performed separately to link the 5' and middle fragment (5'M PCR) and the middle and 3' fragment (M3' PCR). The 5'M PCR co-template used was the 5' and middle PCR fragment as described above, the sense primer was SEQ.ID.NO.:12 and the antisense primer was SEQ.ED.NO.:20. The 5'M PCR procedure resulted in an 857 bp PCR fragment.
[0118] The M3' PCR used the middle and M3' PCR fragment described above as the co- template, SEQ.ID.NO.:19 as the sense primer and SEQ.ED.NO.:6 (see note above regarding SEQ.ID.NOS.:6 and 11) as the antisense primer, and generated a 784 bp amplification product. The final round of PCR was performed using the 857 bp and 784 bp fragments from the second round PCR as the co-template, and SEQ.ID.NO.:12 and SEQ.ID.NO.:6 (see note above regarding SEQ._O.NOS.:6 and 11) as the sense and the antisense primer, respectively. The 1.32 kb amplification product from the final round of PCR was digested with Pst I and Eco RI. Then resulting 1 kb Pst I-Eco RI fragment was used to replace the corresponding fragment ofthe endogenous human 5-HT2A to generate mutant 5-HT2A with 5-HT2C: C310K/IC3. The Apa I-Xba fragment of AP3 was used to replace the corresponding fragment in mutant 5-HT2A with 5-HT2C: C310K/IC3 to generate AP4.
EXAMPLE 2 RECEPTOR EXPRESSION
A. pCMV
[0119] Although a variety of expression vectors are available to those in the art, for purposes of utilization for both the endogenous and non-endogenous receptors discussed herein, it is most preferred that the vector utilized be pCMV. This vector was deposited with the American Type Culture Collection (ATCC) on October 13, 1998 (10801 University Blvd., Manassas, VA 20110-2209 USA) under the provisions ofthe Budapest Treaty for the International Recognition ofthe Deposit of Microorganisms for the Purpose of Patent Procedure. The DNA was tested by the ATCC and determined to be viable. The ATCC has assigned the following deposit number to pCMV: ATCC #203351. See Figure 8.
B. Transfection procedure
[0120] For the generic assay ((35S)GTPγS; Example 3) and the antagonist binding assay
(mesulergine; Example 4), transfection of COS-7 or 293T cells was accomplished using the following protocol.
[0121] On day one, 5xl06 COS-7 cells or lxlO7 293T cells per 150mm plate were plated out.
On day two, two reaction tubes were prepared (the proportions to follow for each tube are per plate): tube A was prepared by mixing 20 μg DNA (e.g., pCMV vector; pCMV vector AP-1 cDNA, etc.) in 1.2 ml serum free DMEM (Irvine Scientific, Irvine, CA); tube B was prepared by rnixing 120 μl hpofectarnine (Gibco BRL) in 1.2 ml serum free DMEM. Tubes A and B were then admixed by inversions (several times), followed by incubation at room temperature for 30-45 min. The admixture is referred to as the "transfection mixture". Plated COS-7 cells were washed with IX PBS, followed by addition of 10 ml serum free DMEM. 2.4 ml of the transfection mixture was then added to the cells, followed by incubation for 4 hrs at 37°C/5% C02. The transfection mixture was then removed by aspiration, followed by the addition of 25 ml of DMEM/10% Fetal Bovine Serum. Cells were then incubated at 37°C/5% C02. After 72 hr incubation, cells were then harvested and utilized for analysis.
EXAMPLE 3
GTP MEMBRANE BINDING SCINTILLATION PROXIMITY ASSAY
[0122] The advantages of using (35S)GTPγS binding to measure constitutive activation are that: '
(a) (35S)GTPγS binding is generically applicable to all G protein-coupled receptors; and (b) (35S)GTPγS binding is proximal at the membrane surface, thereby making it less likely to pick-up molecules which affect the intracellular cascade. The assay utilizes the ability of G protein-coupled receptors to stimulate (35S)GTPγS binding to membranes expressing the relevant receptors. Therefore, the assay may be used to directly screen compounds at the disclosed serotonin receptors.
[0123] Figure 9 demonstrates the utility of a scintillation proximity assay to monitor the binding of (35S)GTPγS to membranes expressing, e.g., the endogenous human 5-HT2c receptor expressed in COS cells. In brief, a preferred protocol for the assay is such that the assay was incubated in 20 mM HEPES, pH 7.4, binding buffer with 0.3 nM (35S)GTPγS and 12.5 μg membrane protein and 1 μM GDP for 30 minutes. Wheatgerm agglutinin beads (25 μl; Amersham) were then added and the mixture was incubated for another 30 minutes at room temperature. The tubes were then centrifuged at 1500 x g for 5 minutes at room temperature and then counted in a scintillation counter. As shown in FIG. 9, serotonin, which as the endogenous ligand activates the 5-HT2C receptor, stimulated (35S)GTPγS binding to the membranes in a concentration dependant manner. The stimulated binding was completely inhibited by 30 μM mianserin, a compound considered as a classical 5-HT2C antagonist, but also known as a 5-HT2C inverse agonist.
[0124] Although this assay measures agonist-induced binding of (35S)GTPγS to membranes and can be routinely used to measure constitutive activity of receptors, the present cost of wheatgerm agglutinin beads may be prohibitive. A less costly but equally applicable alternative also meets the needs of large-scale screening. Flash plates and Wallac™ scintistrips may be used to format a high throughput (35S)GTPγS binding assay. This technique allows one to monitor the tritiated ligand binding to the receptor while simultaneously monitoring the efficacy via (35S)GTPγS binding. This is possible because the Wallac™ beta counter can switch energy windows to analyze both tritium and 35S-labeled probes.
[0125] Also, this assay may be used for detecting of other types of membrane activation events that result in receptor activation. For example, the assay may be used to monitor 32P phosphorylation of a variety of receptors (including G protein-coupled and tyrosine kinase receptors). When the membranes are centrifuged to the bottom of the well, the bound (35S)GTPγS or the 32P- phosphorylated receptor will activate the scintillant coated on the wells. Use of Scinti® strips (Wallac™) demonstrate this principle. Additionally, this assay may be used for measuring ligand binding to receptors using radiolabeled ligands. In a similar manner, the radiolabeled bound ligand is centrifuged to the bottom of the well and activates the scintillant. The (35S)GTPγS assay results parallel the results obtained in traditional second messenger assays of receptors.
[0126] As shown in Figure 10, serotonin stimulates the binding of (35S)GTPγS to the endogenous human 5-HT2C receptor, while mianserin inhibits this response; furthermore, mianserin acts as a partial inverse agonist by inhibiting the basal constitutive binding of (35S)GTPγS to membranes expressing the endogenous human 5-HT2c receptor. As expected, there is no agonist response in the absence of GDP since there is no GDP present to exchange for (35S)GTPγS. Not only does this assay system demonstrate the response of the native 5-HT2C receptor, but it also measures the constitutive activation of other receptors.
[0127] Figure 11A and Figure 11B demonstrate the enhanced binding of (35S)GTPγS to membranes prepared from 293T cells expressing the control vector alone, the native human 5-HT2c receptor or the AP-1 receptor was observed (data not shown). The total protein concentration used in the assay affects the total amount of (35S)GTPγS binding for each receptor. The c.p.m. differential between the CMV transfected and the constitutively active mutant receptor increased from approximately 1000 c.p.m at 10 μg/well to approximately 6-8000 c.p.m. at 75 μg/well protein concentration, as shown in Figure 11.
[0128] The AP-1 receptor showed the highest level of constitutive activation followed by the wild type receptor, which also showed enhanced (35S)GTPγS binding above basal. This is consistent with the ability of the endogenous human 5-HT2C receptor to accumulate intracellular IP3 in the absence of 5-HT stimulation (Example 5) and is also consistent with published data claiming that the endogenous human 5-HT2C receptor has a high natural basal activity. Therefore, the AP-1 receptor demonstrates that constitutive activity may be measured by proximal (3SS)GTPγS binding events at the membrane interface.
EXAMPLE 4
SEROTONIN RECEPTOR AGONIST/ANTAGONIST COMPETITIVE BINDING ASSAY
[0129] Membranes were prepared from transfected COS-7 cells (see Example 2) by homogenization in 20 mM HEPES and 10 mM EDTA, pH 7.4 and centrifuged at 49,000 x g for 15 min. The pellet was resuspended in 20 mM HEPES and 0.1 mM EDTA, pH 7.4, homogenized for 10 sec. using a Polytron homogenizer (Brinkman) at 5000 rpm and centrifuged at 49,000 x g for 15 min. The final pellet was resuspended in 20 mM HEPES and 10 mM MgCl2, pH 7.4, homogenized for 10 sec. using polytron homogenizer (Brinkman) at 5000 rpm.
[0130] Assays were performed in triplicate 200 μl volumes in 96 well plates. Assay buffer
(20 mM HEPES and 10 mM MgCl2, pH 7.4) was used to dilute membranes, 3H-LSD, 3H-mesulergine, serotonin (used to define non-specific for LSD binding) and mianserin (used to define non-specific for mesulergine binding). Final assay concentrations consisted of 1 nM 3H-LSD or 1 nM 3H-mesulergine, 50 μg membrane protein and 100 μm serotonin or mianserin. LSD assays were incubated for 1 hr at 37° C, while mesulergine assays were incubated for 1 hr at room temperature. Assays were terminated by rapid filtration onto Wallac Filtermat Type B with ice cold binding buffer using Skatron cell harvester. The radioactivity was determined in a Wallac 1205 BetaPlate counter.
EXAMPLE 5
INTRACELLULAR IP3 ACCUMULATION ASSAY
[0131] For the IP3 accumulation assay, a transfection protocol different from the protocol set forth in Example 2 was utilized. In the following example, the protocols used for days 1-3 were slightly different for the data generated for Figures 12 and 14 and for Figures 13 and 15; the protocol for day 4 was the same for all conditions. A. COS-7 and 293 Cells [0132] On day one, COS-7 cells or 293 cells were plated onto 24 well plates, usually lxl 05 cells/well or 2xl05 cells/well, respectively. On day two, the cells were transfected by first mixing 0.25 ug DNA (see Example 2) in 50 μl serum-free DMEM/well and then 2 μl lipofectamine in 50 μl serum-free DMEM/well. The solutions ("transfection media") were gentiy mixed and incubated for 15-30 minutes at room temperature. The cells were washed with 0.5 ml PBS and then 400 μl of serum free media was mixed with the transfection media and added to the cells. The cells were then incubated for 3-4 hours at 37°C/5%C02. Then the transfection media was removed and replaced with 1 ml/well of regular growth media. On day 3, the media was removed and the cells were washed with 0.5 ml PBS. Then 0.5 ml inositol-free/serum-free media (GIBCO BRL) was added to each well with 0.25 μCi of 3H-myo-inositol/well and the cells were incubated for 16-18 hours overnight at 37°C/5% C02. Protocol A.
B. 293 Cells
[0133] On day one, lxl 07 293 cells per 150 mm plate were plated out. On day two, two reaction tubes were prepared (the proportions to follow for each tube are per plate): tube A was prepared by mixing 20 μg DNA (e.g., pCMV vector; pCMV vector AP-1 cDNA, etc.) in 1.2 ml serum free DMEM (Irvine Scientific, Irvine, CA); tube B was prepared by mixing 120 μl lipofectamine (Gibco BRL) in 1.2 ml serum free DMEM. Tubes A and B were then admixed by inversions (several times), followed by incubation at room temperature for 30-45 min. The admixture is referred to as the "transfection mixture". Plated 293 cells were washed with 1XPBS, followed by addition of 10ml serum free DMEM. 2.4 ml of the transfection mixture was then added to die cells, followed by incubation for 4 hrs at 37°C/5% C02. On day 3, cells were trypsinized and counted, followed by plating of lxlO6 cells/well (poly D-lysine treated 12-well plates). Cells were permitted to adhere to the wells, followed by one wash with lxPBS. Thereafter, 0.5 μCi 3H-inositol in 1ml inositol-free DMEM was added per well. Protocol B.
[0134] On day 4, the cells were washed with 0.5 ml PBS and then 0.45 ml of assay medium was added containing inositol-free/serum free media, 10 μM pargyline, 10 mM lithium chloride, or 0.4 ml of assay medium and 50 μl of lOx ketanserin (ket) to a final concentration of 10 μM. The cells were then incubated for 30 minutes at 37° C. Then the cells were washed with 0.5 ml PBS and 200 μl of fresh/ice cold stop solution (IM KOH; 18 mM Na-borate; 3.8 mM EDTA) was added/well. The solution was kept on ice for 5-10 minutes or until the cells were lysed and then neutralized by 200 μl of fresh/ice cold neutralization sol. (7.5 % HCL). The lysate was then transferred into 1.5 ml rnicro- centrifuge tubes and 1 ml of chloroform/methanol (1:2) was added/tube. The solution was vortexed for 15 seconds and the upper phase was applied to a Biorad AG1-X8 anion exchange resin ( 100-200 mesh). The resin was washed with water and 0.9 ml of the upper phase was loaded onto the column. The column was washed with 10 ml of 5 mM myo-inositol and 10 ml of 5 mM Na-borate/60mM Na- formate. The inositol trisphosphates were eluted into scintillation vials containing 10 ml of scintillation cocktail with 2 ml of 0.1 M formic acid 1 M ammonium formate. The columns were regenerated by washing with 10 ml of 0.1 M formic acid/3M ammonium formate and rinsed twice with dd H20 and stored at room temperature in water. Results are discussed below. [0135] Figure 12 is an illustration of IP3 production from the human 5-HT2A receptor which was mutated using the same point mutation as set forth in Casey, which rendered the rat receptor constitutively active. The results represented in Figure 12, support die position that when the point mutation shown to activate the rat receptor is introduced into the human receptor, little activation of the receptor is obtained that would allow for appropriate screening of candidate compounds, with the response being only moderately above that of the endogenous human 5-HT2A receptor. Generally, a response of at least 2X above that ofthe endogenous response is preferred.
[0136] Figure 13 provides an illustration comparing IP3 production from endogenous 5-HT2A receptor and the AP4 mutation. The results illustrated in Figure 13 support the position that when the novel mutation disclosed herein is utilized, a robust response of constitutive IP3 accumulation is obtained (e.g., over 2X that ofthe endogenous receptor).
[0137] Figure 14 provides an illustration of IP3 production from AP3. The results illustrated in Figure 14 support the position fliat when the novel mutation disclosed herein is utilized, a robust response of constitutive IP3 accumulation is obtained.
[0138] Figure 15 provides bar-graph comparisons of IP3 accumulation between endogenous human 5-HT2C receptor and AP-1. Note that the endogenous receptor has a high degree of natural constitutive activity relative to the control CMV transfected cells (i.e., the endogenous receptor appears to be constitutively activated).
Example 6
Screening of Compounds Known to Have 5-HT_c Antagonist Activity Against Non-Endogenous, Constitutively Activated Human Serotonin Receptor: AP-1
[0139] A final concentration of 12.5 μg membranes prepared from COS7 cells (see Example
2) transiently expressing constitutively active mutant human 5-HT2C receptor AP-1 were incubated with binding buffer (20 mM HEPES, pH 7.4, 100 mM NaCl, 20 mM MgCl2° 6H,0, 0.2% saponin, and 0.2 mM ascobate), GDP(1 μM) and compound in a 96-well plate format for a period of 60 rninutes at ambient room temperature. Plates were then centrifuged at 4,000 rpm for 15 minutes followed by aspiration of the reaction mixture and counting for 1 minute in a Wallac™ MicroBeta plate scintillation counter. A series of compounds known to possess reported 5-HT2c antagonist activity were determined to be active in the (35S)GTP- S binding assay using AP-1. IC50 determinations were made for these commercially available compounds (RBI, Natick, Mass.). Results are summarized in TABLE 3. For each determination, eight concentrations of test compounds were tested in triplicate. The negative control in these experiments consisted of AP-1 receptor without test compound addition, and the positive control consisted of 12.5 μg/well of COS7 cell membranes expressing the CMV promoter without expressed AP-1 receptor.
TABLE 3
Figure imgf000033_0001
The IC50 results confirm that the seven tested compounds showed antagonist activity at the AP-1 receptor.
EXAMPLE 7
SCREENING OF CANDIDATE COMPOUNDS AGAINST NON-ENDOGENOUS, CONSTITUTIVELY ACTIVATED HUMAN SEROTONIN RECEPTORS: AP-1
[0140] Approximately 5,500 candidate compounds (Tripos, Inc., St. Louis, MO) were screened using the assay protocol of Example 3 (with AP-1 mutant receptor) for identification as inverse agonists against the receptor; for this assay, an arbitrary cut-off of at least 50% inhibition was established for identification of inverse agonists. Approximately 120 of these compounds evidenced at least 50% inhibition of (35S) GTPγS binding at 10 μM candidate compound (data not shown).
EXAMPLE 8
SCREENING OF SELECTED COMPOUNDS TO CONFIRM RECEPTOR BINDING: AP-1 [0141] The candidate compounds identified from Example 7 were then screened using the assay protocol of Example 4 (mesulergine), using the AP-1 mutant receptor. IC50 (nM) values were determined; five of the nearly 120 compounds of Example 7 were determined to have potent binding affinity for the receptor. Results are summarized in TABLE 4. TABLE 4
Figure imgf000034_0001
EXAMPLE 9A
GENERAL SCREENING PARADIGM: SELECTION OF PRE-CLLNICAL CANDIDATE LEADS
[0142] The "primary" screen designed to directly identify human 5-HT2A/5-HT2C receptor inverse agonists consisted of a membrane-based GTPγS binding assay utilizing membranes prepared from COS7 cells transiently transfected with AP-1 human receptor. Candidate compounds (lOμM final assay concentration) directly identified as inhibiting receptor-mediated increases in GTPγS binding by greater than 50-75% (arbitrary cut-off value) were considered active "hits". Primary assay hits were then re-tested in the same assay to recorifirm their inverse agonist activity. If primary assay hits were reconfirmed active (50% or greater inhibition), and therefore directly identified as, e.g., an inverse agonist, one of two approaches were available: (a) so-called "directed libraries" could be created, i.e., additional candidate compounds were synthesized based upon the structures of the reconfirmed hits (geared towards, e.g., improvement in the characteristics ofthe compounds) whereby the directed library compounds were then evaluated for the ability to compete for radioligand binding to both mutant 5-HT2C (AP-1) and endogenous 5-HT2A receptors, or (b) the reconfirmed hits were then evaluated for the ability to compete for radioligand binding to both mutant 5-HT2C (AP-1) and endogenous 5-HT2A receptors. Thus, when approach (a) was used, because these directed library candidate compounds were based upon the structures of compounds that were directly identified from the membrane-based GTPγS binding assay, the directed library compounds were not re-tested in the membrane-based GTPγS binding assay but rather were then confirmed via the radioligand binding analysis. The radioligand binding analysis tests were initially performed at lOμM test compound in triplicate and if the compound inhibited radiolabeled binding by 50% or more, the analysis was followed by eight concentration competition curves to determine Ki values. The last step in secondary assay evaluation was to determine if test compounds were capable of inhibiting AP-3 receptor- mediated accumulation of inositol phosphates (e.g., IP3). This final assay confirms that the directly identified compounds retained inverse agonist properties.
Example 9B CONSTITUTIVELY ACTIVATED HUMAN 5-HT2C RECEPTOR (AP-1)
MEDIATED FACILITATION OF GTPγS BINDING TO COS7 MEMBRANES
[0143] This protocol is substantially the same as set forth above in Example 6. Primary screening assays measuring GTPγS binding to membranes prepared from COS7 cells transiently transfected with human mutated 5-HT2C receptor (AP-1) were used to directly identify inverse agonists in screening libraries (Tripos, Inc.). Candidate compound screens were performed in a total assay volume of 200 μl using scintillant-coated Wallac Scintistrip™ plates. The primary assay was comprised of the following chemicals (at indicated final assay concentrations): 20 mM HEPES, pH 7.4, 100 mM NaCl, 20 mM MgCl2, 0.2% saponin, 0.2 mM ascorbic acid, 1 μM GDP, 0.3 nM GTPγ35S, and 12.5 μg of the above defined membranes. Incubations were performed for 60 minutes at ambient room temperature. The binding assay incubation was temiinated by centrifugation of assay plates at 4,000 rpm for 15 minutes, followed by rapid aspiration ofthe reaction mixture and counting in a Wallac MicroBeta™ scintillation counter.
[0144] Primary screening of candidate compounds initially involved testing of 72 test compounds per assay plate (96-well plates were utilized), at a final assay concentration of 10 μM candidate compound, in single replicates. A total of sixteen wells of each plate were dedicated for an eight concentration clozapine (a confirmed 5-HT2C/2A inverse agonist) dose response curve (duplicate determinations at each concentration). Finally, a total of five assay wells of each plate were dedicated to define the negative control (AP-1 receptor expressing membranes without addition of candidate compounds) and three wells from each plate to define the positive control (membranes without AP-1 receptor) .
[0145] Reconfirmation experiments involve re-testing candidate compounds in the same assay described above, except that candidate compounds were evaluated in triplicate, thus allowing evaluation of 24 compounds per 96-well assay plate. Similar to the primary assay plates, an eight concentration clozapine dose response curve (duplicate deteπriinations at each concentration) and the same negative and positive control wells were also included within each 96-well plate. Example 9C(1)
COMPETITION STUDIES FOR DIRECTLY IDENTIFIED COMPOUNDS: MUTATED HUMAN 5-HT2C RECEPTOR (AP-1)
[0146] Radioligand binding competition experiments were performed in a total assay volume of 200 μl using standard 96-well microtiter plates. The final assay ingredients consisted of assay buffer (20 mM HEPES and 10 mM MgCl2), InM (3H) mesulergine, and 50 μg of membranes (COS7 with AP-1 as defined above). Nonspecific (3H) mesulergine binding was defined in the presence of 100 μM mianserin. Incubations were performed for 1 hour at 37°C. Receptor bound radioligand was resolved from free radioligand by rapid filtration ofthe assay mixture over a Wallac Filtermat™ Type B filter, followed by washing with ice-cold assay buffer using a Skatron™ cell harvester. Radioactivity was counted using a Wallac 1205 BetaPlate™ counter. Each assay plate contained five negative control wells (membranes expressing receptor and no candidate compound addition) and three positive control wells (each containing 100 μM mianserin). For one concentration tests, candidate compounds were diluted into assay buffer and screened at a final concentration of 10 μM, in triplicate. For IC50 determinations, candidate compounds were diluted in assay buffer and eight different concentrations were evaluated, in triplicate. A total of 16 wells were designated for an eight concentration mianserin dose response curve evaluation for both assays.
EXAMPLE 9C(2)
COMPETITION STUDIES WILD TYPE HUMAN 5-HT2A RECEPTOR [0147] Radioligand binding competition experiments were performed in a total assay volume of 200 μl using standard 96-well microtiter plates. The final assay ingredients comprised assay buffer (20 mM HEPES and lOmM MgCl2), InM (3H)LSD, and 50 μg of the above-defined membranes (COS7 with AP-1). Nonspecific (3H)LSD binding was defined in the presence of 100 μM serotonin. Incubations were performed for 1 hour at 37° C. Receptor bound radioligand was resolved from free radioligand by rapid filtration of the assay mixture over a Wallac Filtermat™ Type B filter, followed by washing with ice-cold assay buffer using a Skatron™ cell harvester. Radioactivity was counted using a Wallac 1205 BetaPlate™ counter. Each assay plate contained five negative control wells (membranes expressing receptor and no candidate compound addition) and three positive control wells (containing 100 μM mianserin). For one concentration tests, candidate compounds were diluted into assay buffer and screened at a final concentration of 10 μM in triplicate. For IC50 determinations, candidate compounds were diluted in assay buffer and eight different concentrations were evaluated in triplicate. A total of 16 wells were designated for an eight concentration serotonin dose response curve evaluation for both assays. EXAMPLE 9D
RECEPTOR-MEDIATED INOSITOL PHOSPHATE ACCUMULATION
[0148] Candidate compound identified in the assays of Examples 9A-9C were then evaluated for inositol phosphate accumulation, following the protocol of Example 5 (COS7 cells expressing human mutated 5-HT2A receptor, AP-3), modified as follows: tube A was prepared by mixing 16 μg DNA (e.g., pCMV vector; pCMV vector AP-1 cDNA, etc.) in 1.0 ml serum free DMEM (Irvine Scientific, Irvine, CA); tube B was prepared by mixing 60 μl lipofectamine (Gibco BRL) in 1.0 ml serum free DMEM. Tubes A and B were then admixed by inversions (several times), followed by incubation at room temperature for 30 min. The admixture is referred to as die "transfection mixture". Plated 293 cells were washed with 10 ml Serum Free DMEM, followed by addition of 11 ml Serum Free DMEM. 2.0 ml ofthe transfection mixture was then added to the cells, followed by incubation for 5 hrs at 37° C/5% C02. On day 3, cells were trypsinized and counted, followed by plating of lxlO6 cells/well (12-well plates). Cells were permitted to adhere to the wells for 8 hrs, followed by one wash with lxPBS. Thereafter, 0.5 μCi 3H-inositol in 1ml inositol-free DMEM was added per well.
[0149] On day 4, the cells were washed with 1.5 ml PBS and then 0.9 ml of assay medium was added containing inositol-free/serum free media, 10 μM pargyline, 10 mM lithium chloride, for 5min in 37°C/5% C02 followed by lOOμl addition of candidate compound diluted in the same material. The cells were then incubated for 120 minutes at 37° C. Then the cells were washed witii 1.5 ml PBS and 200 μl of fresh/icecold stop solution (IM KOH; 18 mM Na-borate; 3.8 mM EDTA) was added/well. The solution was kept on ice for 5-10 minutes or until the cells were lysed and then neutralized by 200 μl of fresh/ice cold neutralization sol. (7.5 % HCL). The lysate was then transferred into 1.5 ml micro-centrifuge tubes and 1 ml of chloroform/methanol (1:2) was added/tube. The solution was vortexed for 15 seconds and the upper phase was applied to a Biorad AG1-X8 anion exchange resin (100-200 mesh). The resin was washed with water and 0.9 ml ofthe upper phase was loaded onto the column. The column was washed with 10 ml of 5 mM myo-inositol and 10 ml of 5 mM Na-borate/60mM Na-formate. The inositol trisphosphates were eluted into scintillation vials containing 10 ml of scintillation cocktail with 2 ml of 0.1 M formic acid/ 1 M ammonium formate. The columns were regenerated by washing with 10 ml of 0.1 M formic acid/3M ammonium formate and rinsed twice with ddH20 and stored at room temperature in water.
[0150] Following this round of assaying, candidate compounds having an IC50 value of less than 10 μM were considered as potential leads for the development of pharmaceutical compositions. SCREENENG CANDIDATE COMPOUNDS
[0151] Following the protocols set forth above, one compound, Compound 7 (Example 8, supra) evidenced the following results as shown in TABLE 4A:
TABLE 4A
Figure imgf000038_0002
[0152] Based upon these results, structure activity analysis of the Compound 7 compound suggested that a series of derivatives of 3-(4-bromo-2-methylpyrazole-3-yl)phenylamine would exhibit similar 5-HT2A activity and selectivity. A series of derivatives of 3-(4-bromo-2- metbylpyrazole-3-yl)phenylamine were synthesized. These "directed" library compounds (Tripos, Inc.) were then analyzed in accordance with die protocols of Examples 9c(l), 9c(2) and 9d.
[0153] This series of compounds exhibits highly selective 5-HT2A activity. Accordingly, in one aspect of die invention, a series of compounds possessing 5-HT2A receptor activity that are useful as inverse agonists at such receptors is designated by the general Formula (I):
Figure imgf000038_0001
(I) wherein: i) Ri is H, halogens, NR5R_, OH or OR7, wherein
R5 and Re are independently H, or Cι_6 alkyl, or C2-6 alkenyl, or cycloalkyl, or aryl, or CH2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02, OH, OMe, OEt, CONRgR., NRsR., NHCOCH3, OCF3, SMe, COOR10, S03R8, S02NR8R9, COMe, COEt, CO-lower alkyl, SCF3, CN, C2.6 alkenyl, H, halogens, CM alkoxy, C3.6 cycloalkyl, Cι_6 alkyl, and aryl wherein each of the C3-6 cycloalkyl, _6 alkyl, or aryl groups may be further optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02, OH, OMe, OEt,
CONRsR9, NRsR9, NHCOCH3, OCF3, SMe, COOR10, S02NR8R9, SO3R10, COMe, COEt, CO-lower alkyl, SCF3, CN, C2.6 alkenyl, H, halogens, C alkoxy, C3.6 cycloalkyl, Cι._ alkyl, and aryl; or
R5 and R^ may form part of a 5, 6 or 7 membered cyclic structure which may be either saturated or unsaturated and that may contain up to four heteroatoms selected from O, N or S and said cyclic structure may be optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02, OH, OMe, OEt, OCF3, SMe, COOR10, S02NR_R9, SO30, NHCOCH3, COEt, COMe, or halogen; R8 and R9 are independently a H, or Cι_6 alkyl, or C2_6 alkenyl, or cycloalkyl, or aryl, or CH2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from halogen, CF3, OCF3, OEt, CC13, Me, N02, OH, OMe, SMe, COMe, CN, COOR10, SO3R10, COEt, NHCOCH3, or aryl; R10 is H or Cι_6 alkyl;
R7 is H or C^ alkyl; ii) R2 is H, straight chain or branched Cι__ alkyl, C2-6 alkenyl, or cycloalkyl; iii) R3 is halogen, carboxy, CN, alkoxycarbonyl, straight chain or branched Cι-6 alkyl, C2- β alkenyl, C2.6 alkynyl, cycloalkyl, aryl or heteroaryl, wherein each of said alkoxycarbonyl, straight chain or branched C_._ alkyl, C2.6 alkenyl, C2.6 alkynyl, cycloalkyl, aryl or heteroaryl groups can be optionally substituted with up to four substituents in any position selected from OH, OR10, NRsRg, halogen,
-C(p)3j or -0-C(p)3 where p is halogen, and said cycloalkyl, aryl or heteroaryl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position; iv) R4 is Cue alkyl, C2-6 alkenyl, or cycloalkyl; v) A is C(=0), C(=S) or S02;
Figure imgf000039_0001
Figure imgf000040_0001
q is O or 1; m is O or 1; n is O or 1;
Rn and Rι2 are each independently H, straight chain or branched Cι-6 alkyl, C2-6 alkenyl, or cycloalkyl;
Figure imgf000040_0002
wherein: Rι3, R14, Ri5, Riβ and R are each independently H, halogen, CN,
NR8R9, COORio, SRio, straight chain or branched Cι-6 alkyl, C_.6 alkenyl, C2.6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl, wherein each of said alkoxycarbonyl, straight chain or branched Cι_6 alkyl, C2.6 alkenyl, C2.6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl groups can be optionally substituted with up to four substituents in any position selected from N02, ORio, NR_R9, halogen, -C(p) or -0-C(p)3 where p is halogen, and said cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl groups can alternatively or additionally be optionally substituted witii up to four alkyl substituents in any position;
L_ is -O-Q2 wherein Q2 is straight chain or branched Cι-6 alkyl, C2_6 alkenyl, C2_6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl, wherein each of said cycloalkyl, aryl, alkylaryl, or arylalkyl groups can be optionally substituted with up to four substituents in any position selected from N02, OR. , halogen, -C(p)3, or -0-C(p)3 where p is halogen, or an aliphatic or aromatic heterocycle, and said cycloalkyl, aryl, alkylaryl, and arylalkyl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position; or a pharmaceutically acceptable salt. [0154] An aryl moiety can be a 5 or 6 membered aromatic heterocyclic ring (containing up to 4 hetero atoms independently selected from N, O, or S) or a 6 membered aromatic non-heterocyclic ring or a polycycle. Cι-6 alkyl moieties can be straight chain or branched; optionally substituted Cι_6 alkyl moieties can be straight chain or branched; C2.6 alkenyl moieties can be straight chain or branched; and optionally substituted C2.6 alkenyl moieties can be straight chain or branched. Examples of suitable Cι-6 alkyl groups include but are not limited to methyl, ethyl, n-propyl, i-propyl, n-butyl, and t-butyl. Halogens are typically F, CI, Br, and I. Examples of 5 or 6 membered ring moieties include, but are not restricted to, phenyl, furanyl, thienyl, imidazolyl, pyridyl, pyrrolyl, oxazolyl, isoxazolyl, triazolyl, pyrazolyl, tetrazolyl, thiazolyl and isothiazoyl. Examples of polycycle moieties include, but are not restricted to, naphthyl, benzothiazolyl, benzofuranyl, benzimidazolyl, quinolyl, isoquinolyl, indolyl, quinoxalinyl, quinazolinyl and benzothienyl.
[0155] In some embodiments of die foregoing Formula (I), compounds of Example 13, Experiments 1-43, infra, and combinations or subcombinations thereof, are included.
[0156] In some embodiments of the foregoing Formula (I), compounds of Example 13,
Experiments 1-43, infra, and combinations or subcombinations thereof, are not included.
[0157] In some embodiments of the compounds of Fonnula (I), B is Lb q is 1, m is 0, and n is 0. In further embodiments of the compounds of Formula (I), B is L q is 1, m is 1, and n is 0. In furtiier embodiments of the compounds of Formula (I), B is Lb q is 1, m is 0, and n is 1. In further embodiments of die compounds of Formula (I), B is L q is 0, m is 0, and n is 0.
[0158] In some embodiments of the compounds of Formula (I), B is L2. In further embodiments of the compounds of Formula (I), A, is C(=0). In further embodiments of the compounds of Formula (I), A, is C(=S). In further embodiments ofthe compounds of Formula (I), A, is S02.
[0159] In further embodiments of the compounds of Formula (I), Ri is H, CI, F, dimethylamino, pyrrolidin-1-yl, morpholin-1-yl, 4-methylpiperazin-l-yl, OH or OCH3.
[0160] In some embodiments ofthe compounds of Formula (I), R3 is CI, Br, I, -COOCH3, 2- hydroxyethyl, 2-(dimethylamino)ethyl, vinyl, ethyl, phenyl, 4-methoxyphenyl, 3-methoxyphenyl, 4- fluorophenyl, 4-frifluoromethoxyphenyl, thiophenyl, carboxy, cyclopropyl, -CCH, -CH=CH-CCH, or
CN.
[0161] In some embodiments ofthe compounds of Formula (I), where B is Lb q is 1, m is 0, n is 0 and A is C(=0), Ri is H, CI, F, dimethylamino, pyrrolidin-1-yl, mo holin-1-yl, 4- methylpiperazin-1-yl, OH or OCH3. [0162] In some embodiments ofthe compounds of Formula (I), where B is Lb q is 1, m is 0, n is 0 and A is C(=0), R3 is CI, Br, I, -COOCH3, 2-hydroxyethyl, 2-(dime%lamino)ethyl, vinyl, ethyl, phenyl, 4-methoxyphenyl, 3-methoxyphenyl, 4-fluorophenyl, 4-trifluoromethoxyphenyl, thiophenyl, carboxy, cyclopropyl, -CCH, -CH=CH-CCH, or CN.
[0163] In some embodiments ofthe compounds of Formula (I), where B is Lb q is 1, m is 1, n is 0 and A is C(=0), Ri is H, CI, F, dimethylamino, pyrrolidin-1-yl, morpholin-1-yl, 4- methylpiperazin-1-yl, OH or OCH3.
[0164] In some embodiments ofthe compounds of Formula (I), where B is Lh q is 1, m is 1, and n is 0, R3 is CI, Br, I, -COOCH3, 2-hydroxyethyl, 2-(dimetiιylamino)ethyl, vinyl, ethyl, phenyl, 4- methoxyphenyl, 3-methoxyphenyl, 4-fluorophenyl, 4-trifluoromethoxyphenyl, thiophenyl, carboxy, cyclopropyl, -CCH, -CH=CH-CCH, or CN.
[0165] In some embodiments ofthe compounds of Formula (I), where B is Lb q is 1, m is 0, n is 1 and A is C(=0), Ri is H, CI, F, dimethylamino, pyrrolidin-1-yl, morpholin-1-yl, 4- methylpiperazin-1-yl, OH or OCH3.
[0166] In some embodiments of the compounds of Formula (I), where B is Lb q is 1 , m is 0, and n is 1, R3 is CI, Br, I, -COOCH3, 2-hydroxyethyl, 2-(dimetbylamino)ethyl, vinyl, ethyl, phenyl, 4- methoxyphenyl, 3-methoxyphenyl, 4-fluorophenyl, 4-trifluoromethoxyphenyl, thiophenyl, carboxy, cyclopropyl, -CCH, -CH=CH-CCH, or CN.
[0167] In some of each of Hie foregoing embodiments ofthe compounds of Formula (I), R2 is H and R4 is methyl of each of the foregoing embodiments of the compounds of Formula (I), Rι3, R14, R15, Ri6 and Rn are each independently H, F, CI, Br, CN, dimethylamino, ethoxycarbonyl, methylthio, methyl, ethyl, isopropyl, trifluoromethyl, trifluoromethoxy, methoxy, NH2 or N02.
[0168] In some embodiments of the compounds of Formula (I), where B is L q is 0, m is 0, and n is 0, A is S02. In some of such embodiments, R2 is H and R4 is methyl. In still further such embodiments, R3 is CI, Br, I, -COOCH3, 2-hydroxyethyl, 2-(dimethylamino)ethyl, vinyl, ethyl, phenyl, 4-methoxyphenyl, 3-methoxyphenyl, 4-fluorophenyl, 4-trifluoromethoxyphenyl, tiiiophenyl, carboxy, cyclopropyl, -CCH, -CH=CH-CCH, or CN.
[0169] In some embodiments of the compounds of Formula (I), B is L2. In some such embodiments, A is (C=0). In further such embodiments, R4 is methyl. In still further such embodiments, R2 is H. In further such embodiments, Qi is ethyl, 4-nitrophenyl, allyl, 4-methylphenyl, isopropyl, butyl, 2-isopropyl-5-methylcyclohexyl, benzyl, 3-bromophenyl, 4-fluorophenyl, 2- metiioxyphenyl, 2-chlorophenyl, -C(CH3)=CH, l-(N-pyridyl)ethyl, or 9-fluoreneylmethyl. In further such embodiments, Rl is H, CI, F, dimethylamino, pyrrolidin-1-yl, morpholin-1-yl, 4- methylpiperazin-1-yl, OH or OCH3; and R3 is CI, Br, I, -COOCH3, 2-hydroxyethyl, 2- (dimetbylamino)ethyl, vinyl, ethyl, phenyl, 4-methoxyphenyl, 3-methoxyphenyl, 4-fluorophenyl, 4- trifluoromethoxyphenyl, thiophenyl, carboxy, cyclopropyl, -CCH, -CH=CH-CCH, or CN.
[0170] In another aspect of the invention, a series of compounds possessing 5-HT2A receptor activity that are useful as inverse agonists at such receptors is designated by Formula (A):
Figure imgf000043_0001
wherein:
R2 is H or lower allcyl(Cι_ι);
R3 is lower alkyl (Cι_6), or halogen;
R4 is lower alkyl ( .6);
X is eitiier Oxygen or Sulfur;
Y is NR15R,6, or (CH2)mR17, or 0(CH2)nR17; m=0-4 n=0-4
R15 is H or lower alkyl(Cι_();
Ri6 and Rn are independently d .6 alkyl, or C2.6 alkenyl, or cycloalkyl, or aryl, or CH2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02, OH, OMe, OEt, C0NRI8R19,
NR1SR19, NHCOCH3j OCF3, SMe, COOR20, SO3R20, S02NR18R19, COMe, COEt, CO-lower alkyl, SCF3, CN, C2.6 alkenyl, H, halogens, CM alkoxy, C3-6 cycloalkyl, Cι.β alkyl, and aryl;
Ris and Rι9 are independently a H or Cι-6 alkyl, or C2.6 alkenyl, or cycloalkyl, or aryl, or CH2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02, OH, OMe, OEt, CONR21R22, NR21R22, NHCOCH3, OCF3, SMe, C00R23, S03R23, S02NR2ιR22, COMe, COEt, CO-lower alkyl, SCF3, CN, C2.6 alkenyl, H, halogens, C 4 alkoxy, C3.6 cycloalkyl, Cι.6 alkyl, and aryl; or R!8 and Rι9 may form part of a 5, 6 or 7 membered cyclic structure which may be either saturated or unsaturated and that may contain up to four heteroatoms selected from O, N or S and said cyclic structure may be optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02, OH, OMe, OEt, OCF3, SMe, COOR23, S02NR21R22, S03R23, NHCOCH3, COEt, COMe, or halogen;
R20 and R23 are each independently selected from H or Cι_6 alkyl; R2ι and R22 are each independently are independently a H, or C) . alkyl, or C2- 6 alkenyl, or cycloalkyl, or aryl, or CH2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from halogen, CF3, OCF3, OEt, CC13, Me, N02, OH, OMe, SMe, COMe, CN, COOR20, SO3R20, COEt, NHCOCH3, or aryl; an aryl moiety can be a 5 or 6 membered aromatic heterocyclic ring (containing up to 4 hetero atoms independently selected from N, 0, or S) or a 6 membered aromatic non-heterocyclic ring or a polycycle;
-6 alkyl moieties can be straight chain or branched; optionally substituted Cι.6 alkyl moieties can be straight chain or branched; C2.6 alkenyl moieties can be straight chain or branched; and optionally substituted C2.6 alkenyl moieties can be straight chain or branched. [0171] Examples of suitable Ci-β alkyl groups include but are not limited to methyl, ethyl, n- propyl, i-propyl, n-butyl, and t-butyl.
[0172] Halogens are typically F, CI, Br, and I.
[0173] Examples of 5 or 6 membered ring moieties include, but are not restricted to, phenyl, furanyl, thienyl, imidazolyl, pyridyl, pyrrolyl, oxazolyl, isoxazolyl, friazolyl, pyrazolyl, tefrazolyl, thiazolyl and isothiazoyl. Examples of polycycle moieties include, but are not restricted to, naphthyl, benzothiazolyl, benzofuranyl, benzimidazolyl, quinolyl, isoquinolyl, indolyl, quinoxalinyl, quinazolinyl and benzothienyl.
[0174] In some embodiments ofthe foregoing genus (A), compounds of Example 13,
Experiments 1-43, infra, and combinations or subcombinations thereof, are included.
[0175] In some embodiments ofthe foregoing genus (A), compounds of Example 13,
Experiments 1-43, infra, and combinations or subcombinations thereof, are not included. [0176] In some preferred embodiments, compounds possessing 5-HT2A receptor activity that are useful as inverse agonists at such receptors are designated by the general Formula Al:
Figure imgf000045_0001
(Al) wherein:
R3 is F, CI, Br, I, Cι-6 straight chain or branched alkyl, C3.8 cycloalkyl, C4-9 alkylcycloalkyl, or C2.6 alkenyl;
X is O or S;
Y is NR15Ri6, or (CH2)mR17, or 0(CH2)nR ; m is an integer between 0 and 4, inclusive; n is an integer between 0 and 4, inclusive;
R4 is H, Cι-8 straight chain or branched alkyl, C3-8 cycloalkyl, C4-9 alkylcycloalkyl, or C2.8 alkenyl;
R2 and Rι5 ais each independently selected from H, Cι_8 straight chain or branched alkyl, C3.8 cycloalkyl, C4-9 alkylcycloalkyl, or C2-8 alkenyl;
Ri6 and R17 is each independently selected from: C_._ straight chain or branched alkyl, C2-8 alkenyl or cycloalkyl, or alkylcycloalkyl, or aryl, or CH2aryl, wherein each moiety within said Cι-8 straight chain or branched alkyl, C2.s alkenyl or cycloalkyl, or alkylcycloalkyl, or aryl or CH2aryl may be optionally substituted by up to four substituents in any position, whereby each substituent is independently selected from: H, F, CI, Br, I, R20, CF3, CF2R7, CF2CF2, CC13, CC12R7, CC12CC12R7, NR18R19, NRι8COR20, NR18SO2R20, OR20, OCF3, OCF2R20, OCF2CF2R20, OCOR20, OSO2R20, OPO(OR20)2, SR20, SCF3, SCF2R20, SCF2CF2R20, SCOR20, SO3R20, S02NR1SR19, PO(OR20)3, PO(OR20)2R20, N02, CN, CNR20(NRls9), CNR18(SR20), COOR20, COSR20, CONRι89, with the proviso that when Rι6 or R17 contains an aryl ring substituted at at least two adjacent positions on said aryl ring, then said two adjacent positions can together be selected from SCH2S, SCH2CH2S, OCH20, or OCH2CH20 to form a bi-cyclic structure;
R20 is H, Ci-s straight chain or branched alkyl, C3-8 cycloalkyl, C4-9 alkylcycloalkyl, C2-8 alkenyl, aryl or alkylaryl; Rι8 and Rι9 are each independently selected from H, Cι-8 straight chain or branched alkyl, C2.8 alkenyl or cycloalkyl, or alkylcycloalkyl, or aryl, or CH2aryl, wherein each moiety within said Cι_8 straight chain or branched alkyl, C2-8 alkenyl or cycloalkyl, or alkylcycloalkyl, or aryl or CH2aryl may be optionally substituted by up to four substituents in any position, whereby each substituent is independently selected from: F, CI, Br, I, CF3, CCl3j CH3, C2H5, C3H7, C4H9, NH2, NHCH3> N(CH3)2, NHC2H5, N(C2H5)2, NHC3H7, N(C3H7)2, NHC4H9, N(C4H9)2, NHCOH, NHCOCH3, NHCOC2H5, NHCOC3H7, NHCOC4H9, NHS02CH3, NHS02C2H5, NHS02C3H7, NHS02C4H9, OH, OCH3, OC2H5, OC3H7, OC4H7, OC4H9, OC5H9, OC5H„, OC6H„, OC63, OCF3, OCOCH3j OCOC2H5, OCOC3H7, OCOC4H9) OS02CH3, OS02C2H5, OS02C3H7j OS02C4H9, SH, SCH3, SC2H5, SC3H7, SC4H7, SC4H9, SC5H9, SC5HU, SC6H„, SC6H13, SCF3, SCOCH3, SCOC2H5, SCOC3H7, SCOC4H9, S03CH3, S03C2H5, S03C3H7, S03C4H9, S02NH, S02NH2, S02NHCH3> S02N(CH3)2, S02NHC2H5, S02N(C2H5)2, S02NHC3H7, S02N(C3H7)2, S02NHC4H9, S02N(C4H9)2, N02, CN, COOCH3, COOC2H5, C00C3H7, C00C4H9, COSCH3, C0SC2H5, C0SC3H7) COSC4H9, CONH2, CONHCH3, CON(CH3)2, CONHC2H5, CON(C2H5)2, CONHC3H7, CON(C3H7)2, CONHC4H9, CON(C4H9)2, and with the proviso that when either or both of Ri8 or R19 contain an aryl ring substituted at at least two adjacent positions on said aryl ring, then said two adjacent positions can together be selected from SCH2S, SCH2CH2S, OCH20, or OCH2CH20 to form a bi-cyclic structure; or RJ8 or Rι9 may together fonn part of a 5, 6 or 7 membered cyclic structure, with said structure being saturated or unsaturated, and further with said structure containing up to four heteroatoms selected from O, N or S, and further wherein each moiety within said cyclic structure being optionally substituted by up to four substituents in any position independently selected from: F, CI, Br, I, CF3, CC13, CH3, C2H5, C3H7, C4H9, NH2, NHCH3, N(CH3)2, NHC2H5, N(C2H5)2, NHC3H7, N(C3H7)2, NHC B,, N(C4H9)2, NHCOH, NHCOCH3, NHCOC2H5, NHCOC3H7, NHCOC4H9, NHS02CH3, NHS02C2H5, NHS02C3H7, NHS02C4H9, OH, OCH3, OC2H5, OC3H7, OC4H7, OC4H9, OC5H9, OC5Hn, OC6Hu, OC63, OCF3, OCOCH3, OCOC2H5, OCOC3H7, OCOC4H9, OS02CH3, OS02C2H5, OS02C3H , OS02C4H9, SH, SCH3, SC2H5, SC3H7, SC4H7, SC4H9, SCsH9, SC5H11, SCβHn, SCgHι3„ SCF3, SCOCH3, SCOC2H5, SCOC3H7, SCOC4H9, S03CH3, S03C2H5, S03C3H7, S03C4H9, S02NH, S02NH2; S02NHCH3, S02N(CH3)2, S02NHC2H5, S02N(C2H5)2, S02NHC3H7, S02N(C3H7)2, S02NHC4H9, S02N(C4H9)2, N02, CN, COOCH3, C00C2H5, C00C3H7, C00C4H9, COSCH3, C0SC2H5, COSC3H7, C0SC4H9, CONH2, CONHCH3> CON(CH3)2, CONHC2H5, CON(C2H5)2, CONHC3H7, CON(C3H7)2, CONHC4H9, CON(C4H9)2, and with the proviso that wherein when Rι8 or Ri9 form an aryl ring substituted at at least two adjacent positions on said aryl ring, then said two adjacent positions can together be selected from SCH2S, SCH2CH2S, OCH20, or OCH2CH20 to form a bi-cyclic structure.
[0177] An aryl moiety can be a 5 or 6 membered aromatic heterocyclic ring (containing up to
4 hetero atoms independently selected from N, O, or S) or a 6 membered aromatic non-heterocyclic ring or a polycycle;
Examples of suitable C... alkyl groups include but are not limited to methyl, ethyl, n-propyl, i-propyl, n-butyl, and t-butyl.
[0178] Examples of 5 or 6 membered ring moieties include, but are not restricted to, phenyl, furanyl, thienyl, imidazolyl, pyridyl, pyrrolyl, oxazolyl, isoxazolyl, friazolyl, pyrazolyl, tefrazolyl, thiazolyl, and isothiazolyl. Examples of polycycle moieties include, but are not restricted to, naphthyl, benzothiazolyl, benzofuranyl, benzirnidazolyl, quinolyl, isoquinolyl, indolyl, quinoxalinyl, quinazolinyl, and benzothienyl.
[0179] In some embodiments of the foregoing genus (A), compounds of Experiments 1-43, infra, and combinations or subcombinations thereof, are included.
[0180] In some embodiments of the foregoing genus (A), compounds of Experiments 1-43, infra, and combinations or subcombinations thereof, are not included.
[0181] Preferred compounds falling within the scope of general Formula (A) as Class (1) compounds where Y = Nι56 and has the Formula (A2):
Figure imgf000047_0001
wherein:
X is O or S; R4 is H or CH3;
Rs and R20 are each independently selected from H, Cι-8 straight chain or branched alkyl, C3.8 cycloalkyl, C4-9 alkylcycloalkyl, or C2-8 alkenyl; R15 is H, F, CI, Br, I, R20, CF3, CF2R20, CF2CF2, CC13, CC12R20, CC12CC12R20, NR18R19,
NRi9COR20, NR19SO2R20, OR20, OCF3, OCF2R20, OCF2CF2R20, OCOR20, OSO2R20, OPO(OR20)2, SR20, SCF3, SCF2R20, SCF2CF2R20, SCOR20, SO3R20, S02NR18R19, PO(OR20)3, PO(OR20)2R20, N02, CN, CNR20(NRι89), CNR19(SR20), COOR20, COSR20, CONRι89, with the proviso that when a position adjacent to R15 is substituted, then R15 and said adjacent position can together be selected from SCH2S, SCH2CH2S, OCH20, or OCH2CH20 to form a bi-cyclic structure;
R18 and R!9 are each independently selected from H, Cι-8 straight chain or branched alkyl, C2.8 alkenyl or cycloalkyl, or alkylcycloalkyl, or aryl, or CH2aryl, wherein each moiety within said Cι_8 straight chain or branched alkyl, C2-8 alkenyl or cycloalkyl, or alkylcycloalkyl, or aryl or CH2aryl may be optionally substituted by up to four substituents in any position, whereby each substituent is independently selected from: F, CI, Br, I, CF3, CC13) CH3, C2H5, C3H7, C4H9, NH2, NHCH3, N(CH3)2, NHC2H5, N(C2H5)2, NHC3H7, N(C3H7)2, NHC4H9, N(C4H9)2, NHCOH, NHCOCH3, NHC0C2H5, NHCOC3H7, NHCOQH., NHS02CH3, NHS02C2H5, NHS02C3H7, NHS02C4H9, OH, OCH3, OC2H5, OC3H7, OC4H7, OC4H9, OC5H9, OC5H„, OC6H„, OC6H13, OCF3, OCOCH3, OCOC2H5, OCOC3H7, OCOC4H9, OS02CH3, OS02C2H3, OS02C3H7, OS02C4H9, SH, SCH3, SC2H5, SC3H7, SC4H7, SQH,, SC5H9, SC5H„, SC6H„, SC6H13„ SCF3, SCOCH3, SCOC2H5, SCOC3H7, SCOC4H9, SO3CH3, S03C2H5, S03C3H7, S03C4H9, S02NH, S02NH2, S02NHCH3, S02N(CH3)2, S02NHC2H5, S02N(C2H5)2, S02NHC3H7, S02N(C3H7)2, S02NHC4H9, S02N(C4H9)2, N02, CN, COOCH3, C00C2H5, C00C3H7, C00C4H9, COSCH3, C0SC2H5, COSC3H7, C0SC4H9, CONH2, CONHCH3; CON(CH3)2, CONHC2H5, CON(C2H5)2, CONHC3H7, CON(C3H7)2, CONHC4H9, CON(C4H9)2, and witii the proviso that when either or botii of RιS or Rι9 contain an aryl ring substituted at at least two adjacent positions on said aryl ring, then said two adjacent positions can together be selected from SCH2S, SCH2CH2S, OCH20, or OCH2CH20 to form a bi-cyclic structure;
R13, Ri4, R15, Ri6 and R17 each independently selected from the following: F, CI, Br, I, CF3, CC13, CH3, C2H5, C3H7, C4H9, NH2, NHCH3; N(CH3)2, NHC2H5, N(C2H5)2, NHC3H7, N(C3H7)2, NHC4H9, N(C4H9)2, NHCOH, NHCOCH3, NHCOC2H5, NHCOC3H7, NHCOC4H9, NHS02CH3, NHS02C2H5, NHS02C3H7, NHS02C4H9, OH, OCH3, OC2H5, OC3H7, OC4H7, OC4H9, OC5H9, OC5Hu, OC6H„, OC6H13, OCF3, OCOCH3, OCOC2H5, OCOC3H7, OCOC4H9, OS02CH3, OS02C2H5, 0S02C3H7, 0S02C4H9, SH, SCH3, SC2Hs, SC3H , SC4H , SCφHg, SC5H9, SC5H11, SCβHπ, SCgHι3„ SCF3, SCOCH3, SCOC2H5, SCOC3H7, SC0C4H9, S03CH3, S03C2H5, S03C3H7, S03C4H9, S02NH, S02NH2, S02NHCH3> S02N(CH3)2, S02NHC2H5) S02N(C2H5)2, S02NHC3H7, S02N(C3H7)2, S02NHC4H9, S02N(C4H9)2, N02, CN, COOCH3, C00C2H5, C00C3H7, C00C4H9, COSCH3, COSC2H5, COSC3H7, COSC4H9, CONH2, CONHCH3, CON(CH3)2, CONHC2H5, CON(C2H5)2, CONHC3H7, CON(C3H7)2, CONHC4H9, CON(C4H9)2, and with the proviso that when any two adjacent positions of Rι3, Rι4, Rι5, Rι6 and R17are substituted, said two adjacent positions can together be further selected from SCH2S, SCH2CH2S, OCH20, or OCH2CH20 to form a bi-cyclic structure; and with the proviso that at least one of Rι3, Rw, Rι5, Riβ and Rι7must be H.
[0182] In some embodiments ofthe foregoing genus (A), class (1), compounds of Example
13, Experiments 1-43, infra, and combinations or subcombinations thereof, are included.
[0183] In some embodiments ofthe foregoing genus (A), class (1), compounds of Example
13, Experiments 1-43, infra, and combinations or subcombinations thereof, are not included.
[0184] A more preferred series of compounds possessing 5-HT2A receptor activity that are useful as inverse agonists at such receptors is designated by the general Formula (B):
Figure imgf000049_0001
wherein:
R2 is H or lower alkyl (C1-4);
R3 is Me, or Et, or halogen;
X is either Oxygen or Sulfur;
Y is NR15R16, or (CH2)mR17, or 0(CH2)nR17;
R4 is lower alkyl (Cι-6); m=0-4 n=0-4
Ri5 is H or lower alkyl(C ); Ri6 and Rn are independently Cι_6 alkyl, or C2.6 alkenyl, or cycloalkyl, or aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02, OH, OMe, OEt, CONR189, NRι8R19, NHCOCH3, OCF3, SMe, COOR20, SO3R20, S02NRι8R19, COMe, COEt, CO-lower alkyl, SCF3, CN, C2-6 alkenyl, H, halogens, C__ι alkoxy, C3-6 cycloalkyl, Cι-6 alkyl, aryl and aryloxy wherein each of the C1-4 alkoxy, C3.6 cycloalkyl, Cι-6 alkyl, aryl and aryloxy groups may be further optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02, OH, OMe, OEt, CONR21R22, NR21R22, NHCOCH3, OCF3, SMe, COOR23, S03R23, S02NR2ιR22, COMe, COEt, CO-lower alkyl, SCF3, CN, C2.6 alkenyl, H, halogens, CM alkoxy, C3.6 cycloalkyl, Cι.6 alkyl, and aryl;
Ris and Rι9 are independently a H or Cι_6 alkyl, or C2.6 alkenyl, or cycloalkyl, or aryl, or CH2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02, OH, OMe, OEt, CONR21R22, NR21R22, NHCOCH3, OCF3, SMe, COOR23, S03R23, S02NR_ιR22, COMe, COEt, CO-lower alkyl, SCF3, CN, C2.6 alkenyl, H, halogens, Q.
4 alkoxy, C3-6 cycloalkyl, Cι-6 alkyl, and aryl or Rι8 and Rι9 may form part of a 5, 6 or 7 membered cyclic structure which may be either saturated or unsaturated and that may contain up to four heteroatoms selected from O, N or S and said cyclic structure may be optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02,
OH, OMe, OEt, OCF3, SMe, COOR7, S02NR_4R25, S03R26, NHCOCH3, COEt, COMe, or halogen;
R20 and R23 are each independently selected from H or Cι.6 alkyl; R21 and R22 are each independently are independently a H, or C1.6 alkyl, or C2- 6 alkenyl, or cycloalkyl, or aryl, or CH2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from halogen, CF3, OCF3, OEt, CC13, Me, N02, OH, OMe, SMe, COMe, CN, COOR20, SO3R20, COEt, NHCOCH3, or aryl; C1.6 alkyl moieties can be straight chain or branched; optionally substituted Cι_6 alkyl moieties can be straight chain or branched:
C2-s alkenyl moieties can be straight chain or branched; and optionally substituted C2_6 alkenyl moieties can be straight chain or branched.
[0185] Examples of suitable Cι_6 alkyl groups include but are not limited to methyl, ethyl, n- propyl, i-propyl, n-butyl, and t-butyl.
[0186] Halogens are typically F, Cl, Br, and I.
[0187] Examples of 5 or 6 membered ring moieties include, but are not restricted to, phenyl, furanyl, thienyl, imidazolyl, pyridyl, pyrrolyl, oxazolyl, isoxazolyl, friazolyl, pyrazolyl, tefrazolyl, thiazolyl and isothiazoyl. Examples of polycycle moieties include, but are not restricted to, naphthyl, benzothiazolyl, benzofuranyl, benzimidazolyl, quinolyl, isoquinolyl, indolyl, quinoxahnyl, quinazolinyl and benzothienyl.
[0188] In some embodiments of the foregoing genus (B), compounds of Example 13,
Experiments 1-43, infra, and combinations or subcombinations thereof, are included.
[0189] In some embodiments of the foregoing genus (B), compounds of Example 13,
Experiments 1-43, infra, and combinations or subcombinations thereof, are not included.
[0190] Exemplary compounds of general Formula (A), Class (1) are set forth below. Based upon in vivo data developed (as set forth below), Compounds 8 and 9 are particularly preferred.
[0191] A first series of compounds having 5-HT2A receptor activity is represented by a class (I) of compounds of Fonnula (B) wherein Y=NRι56 and is Formula (Bl):
Figure imgf000051_0001
(Bl) wherein:
Preferably R2 and Rι5 are H; Preferably R3 is Br; Preferably X is O;
Preferably R4 is Me.
In some embodiments of the foregoing genus (B), class (1), compounds of Example 13, Experiments 1-43, infra, and combinations or subcombinations thereof, are included.
[0192] In some embodiments of the foregoing genus (B), class (1), compounds of Example
13, Experiments 1-43, infra, and combinations or subcombinations thereof, are not included.
[0193] In reference to Formula (Bl) Rι6 is preferably 4-trifluoromethoxyphenyl , 4- trifluoromethoxybenzyl, 4-chlorophenyl or 4-fluorophenyl.
[0194] Certain preferred compounds are: Compound 7 N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)(((4-frifluoromethoxy)phenyl)an nocarboxamide
Figure imgf000052_0001
Compound 10 N-(3-(4-bromo-2-methylpyrazol-3-yl)-phenyl)(((4-trifluoromethoxy)phenylmethyl)
Aminocarboxamide
Figure imgf000052_0002
[0195] These two compounds demonstrated the following activities using the assay protocols defined in the Examples above:
TABLE 5
Figure imgf000052_0003
[0196] Additional compounds are set forth below. Inositol phosphate accumulation assays evidence the activity of test compounds. Both single concentration percentages of control values and IC 0 determinations indicate activity. In the tables below the column legends have the following meanings: IP3 % Control: The values in this column reflect an IP Accumulation Assay where the test compounds were evaluated at one concentration of 10 μM. For these assays, the compound was diluted into inositol-free Dulbecco's Eagle Media containing 10 μM pargyline and 10 mM LiCl and tested at a final assay concentration of 10 μM, in triplicate. The percent control value was calculated based on the control in which no test compound was added. IPT AP-3 ICsn nM: The values in this column reflect an IP accumulation assay in which the test compound was evaluated at several different concentrations whereby an IC50 could be detennined. This column corresponds to the column appearing in the tables above which is labeled: Inositol Phosphate Accumulation, AP-3, IC50 Value (μM).
WT 5-HTTA LSD ICsn nM: The values in this column reflect a competitive binding assay using LSD. This column corresponds to the column appearing in the tables above which is labeled: Competitive Binding, WT 5-HT2A) ((3H)LSD), IC50 Value (μM).
[0197] Compounds listed in each ofthe following tables reference the structures hnmediately preceding the table. A "dash" in the table indicates that no value was determined.
TABLE6
Figure imgf000054_0001
Figure imgf000054_0002
Figure imgf000055_0001
Figure imgf000056_0002
TABLE 7
Figure imgf000056_0001
Figure imgf000056_0003
TABLE8
Figure imgf000057_0001
Figure imgf000057_0002
Figure imgf000058_0002
Figure imgf000058_0001
Compound Name
Compound IPs WT
No. Rl5 Rl4 Rl3 Rl6 Rs AP-3 5-HT2A
LSD
ICso nM ICso nM
N -(3 -(4-bromo-2-methylpyrazol-3 -yl)phenyl)((2-(4- methoxyphenyl)etiιyl)an ino)carboxamide
Compound OMe H H H H 32 61 38 [0198] A second series of compounds having 5-HT2A receptor activity is represented by a class (II) of compounds of formula (B) wherein Y= 0(CH2)n7.
Figure imgf000059_0001
(B2) wherein:
Preferably R2 is H. Preferably R3 is Br. Preferably X is O. Preferably R4 is Me.
Preferably when n = 0, R17 is 4-methoxyphenyl or tertiary butyl.
In some embodiments of the foregoing genus (B), class (II), compounds of Example 13, Experiments 1-43, infra, and combinations or subcombinations thereof, are included.
[0199] In some embodiments of the foregoing genus (B), class (II) compounds of Example
13, Experiments 1-43, infra, and combinations or subcombinations thereof, are not included.
[0200] Certain preferred compounds are: .
Compound 1 N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)(4-methoxyphenoxy)carboxamide
Figure imgf000059_0002
Compound 39 (tert-butoxy)-N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)carboxamide
Figure imgf000060_0001
[0201] Compounds 1 and 39 demonstrated the following activity as, illustrated in TABLE 10:
TABLE 10
Figure imgf000060_0002
[0202] In addition to the assays discussed above, the specific activity of Compound 1 at the
5-HT2A receptor was further confirmed by the following:
In Vitro Binding of 5-HT,A Receptor Animals:
[0203] Animals (Sprague-Dawley rats) were sacrificed and brains were rapidly dissected and frozen in isopentane maintained at -42° C. Horizontal sections were prepared on a cryostat and maintained at -20 ° C.
LSD Displacement Protocol: [0204] Lysergic acid diethylamide (LSD) is a potent 5-HT2A receptor and dopamine D2 receptor ligand. An indication of the selectivity of compounds for either or both of these receptors involves displacement of radiolabeled-bound LSD from pre-treated brain sections. For these studies, radiolabeled I125-LSD (NEN Life Sciences, Boston, Mass., Catalogue number NEX-199) was utilized; spiperone (RBI, Natick, Mass. Catalogue number s-128) a 5-HT2A receptor and dopamine D2 receptor antagonist, was also utilized. Buffer consisted of 50 nanomolar TRIS-HCl, pH 7.4. [0205] Brain sections were incubated in (a) Buffer plus 1 nanomolar Ii25-LSD; (b) Buffer plus 1 nanomolar I125-LSD and 1 micromolar spiperone; or Buffer plus 1 nanomolar I125-LSD and 1 micromolar Compound 1 for 30 minutes at room temperature. Sections were then washed 2x 10 minutes at 4 ° C. in Buffer, followed by 20 seconds in distilled H20. Slides were then air-dried.
[0206] After drying, sections were apposed to x-ray film (Kodak Hyperfilm) and exposed for
4 days.
Analysis:
[0207] Figures 16A-C provide representative autoradiographic sections from this study.
Figure 16A evidences darker bands (derived from I125 -LSD binding) primarily in both die fourth layer of the cerebral cortex (primarily 5-HT2A receptors), and the caudate nucleus (primarily dopamine D2 receptors and some 5-HT2A receptors). As can be seen from Figure 16B, spiperone, which is a 5-HT2 and dopamine D2 antagonist, displaces the I125-LSD from these receptors on both the cortex and the caudate. As can be further seen from Figure 16C, Compound 1 appears to selectively displace the I125- LSD from the cortex (5-HT2A) and not the caudate (dopamine D2).
[0208] A third series of compounds having 5-HT2A receptor activity is represented by a class
(III) of compounds of Formula (B3) wherein Y=(CH2)m7:
Figure imgf000061_0001
(B3) wherein
Preferably R2 is H. Preferably R3 is Br.
Preferably X is O. Preferably R4 is Me. [0209] Preferably when m= 0, R is preferably 4-trifluoromethoxyphenyl, or thiophene, or
4-chlorophenyl.
[0210] In some embodiments ofthe foregoing genus (B), class (III), compounds of Example
13, Experiments 1-43, infra, and combinations or subcombinations thereof, are included. [0211] In some embodiments ofthe foregoing genus (B), class (III) compounds of Example
13, Experiments 1-43, infra, and combinations or subcombinations thereof, are not included.
[0212] Certain preferred compounds are:
Compound 40 (m = 0, R2 = H, Ri7 = 4-trifluoromethoxyphenyl)
N-[3-(4-Bromo-2-methyl-2H-pyτazol-3-yl)-phenyl]-4-frifluoromethoxy-benzamide
Figure imgf000062_0001
Compound 2 (m=0, R2= H, Rn = thiophene) Thiophene-2-carboxylic acid [3-(4-bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]amide
Figure imgf000062_0002
Compound 41 (m=0, R2= H, R = chlorophenyl) N-[3 -(4-Bromo-2-methyl-2H-pyrazol-3 -yl)-phenyl] -4-chloro-benzamide
Figure imgf000062_0003
[0213] These three compounds demonstrated die following activities:
TABLE 11
Figure imgf000063_0001
In Vivo Analysis of Compound 2 [0214] In addition to the in vitro assays shown in the above table, the in vivo response of animals to Compound 2 is demonstrated by the following.
[0215] A 5-HT2A receptor antagonist or inverse agonist is expected to decrease amphetamine- stimulated locomotion without affecting baseline locomotion. See, for example, Soresnon, et al, 266(2) J. Pharmacol. Exp. Ther. 684 (1993). Based upon the foregoing information, Compound 2 is a potent inverse agonist at the human 5-HT2A receptor. For the following study, the following parameters and protocol were utilized:
Animals. Vehicle
[0216] Adult male Sprague-Dawley rats were utilized for these studies. Animals were housed in groups of 2-3 in hanging plastic cages with food and water available at all times. Animals were weighed and handled for at least one day prior to surgery and throughout the studies. For these studies, Vehicle consisted of 90% ethanol (100%) and 10% water.
Amphetamine-Stimulated Locomotor Activity: Assessment and Apparatus
[0217] A San Diego Instruments Flex Field apparatus was used to quantify baseline and amphetamine-stimulated locomotor activity. This apparatus consists of four 16" x 16" clear plastic open fields. Photocell arrays (16 in each dimension) interfaced with a personal computer to automatically quantify activity. Several measures of activity can be assessed witii the apparatus, including total photocell beam breaks. Animals (vehicle control and Compound treated) were injected s.c. 30 minutes prior to initiation of analysis. Following this 30 minute period, animals were placed individually into an open field and baseline activity was assessed for 30 minutes (habituation phase). Following baseline, animals were removed, injected with d-amphetamine sulfate (1.0 mg/kg) and immediately returned to the open field for 150 minutes, in order to follow the time course (10 minute intervals) of amphetamine-stimulated locomotor activity.
Dosing
TABLE 12
Figure imgf000064_0001
Analysis [0218] Results, based upon the number of recorded photobeam breaks (mean.+/-sem), are presented in Figures 17A-C. As supported by Figures 17A, B and C, a general "inverted U" shaped pattern was observed (see, generally, Sahgal, A. "Practical behavioural neuroscience: problems, pitfalls and suggestions" pp 1-8, 5 in Behavioral Neuroscience: A Practical Approach. Volume 1 A. Sahgal (Ed.) 1993, IRL Press, New York). As Figure 17 also indicates, with exception ofthe highest dose (10 mg/kg), in vivo, the tested doses of Compound 2 evidenced a decrease in the amphetamine- stimulated locomotion, consistent with a 5-HT2A receptor antagonist or inverse agonist.
[0219] Additional series of compounds of Formula (B) wherein Y=(CH2)m7 are set forth below in TABLE 13.
TABLE 13
Figure imgf000065_0001
Figure imgf000065_0002
[0220] Based upon the discovery of the specific inverse agonist activity of the above identified compounds at the 5-HT2A receptor, a novel class of compounds has been identified which exhibits said activity. Accordingly, in the second aspect of the invention, there is provided a novel compound of formula (C):
Figure imgf000066_0001
wherein:
R2 is H or lower alkyl(C ); R3 is Me, or Et, or halogen;
X is either Oxygen or Sulfur; Y is NR15R16, or (CH2)m Rn, or 0(CH2)n R17; R4 is lower alkyl (Cι.6); m=0-4; n=0-4;
R15 is H or lower alkyl(CM);
Ri6 is a Cι-6 alkyl, or C2-6 alkenyl, or cycloalkyl, or (CH2)k aryl group (k=l-4), preferably k=l, and each said group may be optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02, OH, OMe, OEt, CONR18R19, NRι8R19, NHCOCH3, OCF3, SMe, COOR20, SO3R20, S02NR18R19, COMe, COEt, CO-lower alkyl, SCF3, CN, C2.6 alkenyl, H, halogens, CM alkoxy, C3-6 cycloalkyl, d_6 alkyl, aryl and aryloxy wherein each of the C3-6 cycloalkyl, Cι_6 alkyl, aryl and aryloxy may be further substituted by up to four substitutents in any position independently selected from CF3, CCI3, Me, N02, OH, OMe, OEt, CONR18R19, NR18R19, NHCOCH3, OCF3, SMe, COOR20, SO3R20, S02NR18R19, COMe, COEt, CO-lower alkyl, SCF3, CN, C2.6 alkenyl, H, halogens, C alkoxy, C3.6 cycloalkyl, Cι_6 alkyl, and aryl;
R18 and Rι9 are independently a H or Cι-6 alkyl, or C2-6 alkenyl, or cycloalkyl, or aryl, or CH2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02, OH, OMe, OEt, CONR21R22, NR2ιR22, NHCOCH3, OCF3, SMe, C00R23, S03R23, S02NR2ιR22, COMe, COEt, CO-lower alkyl, SCF3, CN, C2-5 alkenyl, H, halogens, C.. 4 alkoxy, C3.6 cycloalkyl, Cι__ alkyl, and aryl wherein each of C3.6 cycloalkyl, Cι_6 alkyl, and aryl groups may be further optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02, OH, OMe, OEt, CONR2ιR22, NR21R22, NHCOCH3, OCF3, SMe, COOR23, S03R23, S02NR21R22, COMe, COEt, CO-lower alkyl, SCF3, CN, C2.6 alkenyl, H, halogens, CM alkoxy, C3.6 cycloalkyl, Cι_6 alkyl, and aryl; or Ris and R19 may form part of a 5, 6 or 7 membered cyclic structure which may be either saturated or unsaturated and that may contain up to four heteroatoms selected from O, N or S and said cyclic structure may be optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02, OH, OMe, OEt, OCF3, SMe, COOR7, S02NR2ιR22, S03R23, NHCOCH3, COEt, COMe, or halogen;
Rn is Cι-6 alkyl, or C2.6 alkenyl, or cycloalkyl, or aryl, or CH2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02, OH, OMe, OEt, CONR21R22, NR_ιR22, NHCOCH,, OCF3, SMe, COOR23, S03R23, S02NR21R22, COMe, COEt, CO-lower alkyl, SCF3, CN, C2.6 alkenyl, H, halogens, CM alkoxy, C3.6 cycloalkyl, -6 alkyl, and aryl wherein each of C3._ cycloalkyl, Cι_6 alkyl, and aryl groups may be further optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02, OH,
OMe, OEt, C0NR21R22, NR21R22, NHCOCH3, OCF3, SMe, C00R23, S03R23, S02NR21R22, COMe, COEt, CO-lower alkyl, SCF3, CN, C2.6 alkenyl, H, halogens, C alkoxy, C3.6 cycloalkyl, Cι-6 alkyl, and aryl;
R20 and R23 may be independently selected from H or Cι-6 alkyl; R21 and R22 are independently a H, or C alkyl, or C2-6 alkenyl, or cycloalkyl, or aryl, or CH2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from halogen, CF3, OCF3, OEt, CC13, Me, N02, OH, OMe, SMe, COMe, CN, COOR20, SO3R20, COEt, NHCOCH3, or aryl. An aryl moiety can be a 5 or 6 membered aromatic heterocyclic ring (containing up to 4 hetero atoms independently selected from N, O, or S) or a 6 membered aromatic non-heterocyclic ring or a polycycle;
-6 alkyl moieties can be straight chain or branched; optionally substituted Cι_6 alkyl moieties can be straight chain or branched; C2-6 alkenyl moieties can be straight chain or branched; and optionally substituted C2.6 alkenyl moieties can be straight chain or branched; Examples of suitable Cι-6 alkyl groups include but are not limited to methyl, ethyl, n-propyl, i-propyl, n-butyl, and t-butyl.
[0221] Halogens are typically F, CI, Br, and I.
[0222] Examples of 5 or 6 membered ring moieties include, but are not restricted to, phenyl, furanyl, thienyl, imidazolyl, pyridyl, pyrrolyl, oxazolyl, isoxazolyl, friazolyl, pyrazolyl, tefrazolyl, thiazolyl and isothiazolyl. Examples of polycycle moieties include, but are not restricted to, naphthyl, benzothiazolyl, benzofuranyl, benzimidazolyl, quinolyl, isoquinolyl, indolyl, quinoxahnyl, quinazolinyl and benzothienyl.
[0223] In some embodiments of the foregoing genus (C), compounds of Example 13, Experiments 1-43, infra, and combinations or subcombinations thereof, are included.
[0224] In some embodiments of the foregoing genus (C), compounds of Example,
Experiments 1-43, infra, and combinations or subcombinations thereof, are not included.
[0225] Some embodiments of the invention are compounds of Formula (V) and have one of the structures below.
TABLE 14
Figure imgf000068_0001
(V)
Figure imgf000068_0002
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001
Figure imgf000072_0001
Figure imgf000073_0001
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0002
[0226] Also provided in accordance with the present invention are compounds useful as inverse agonists for 5-HT2A receptors having the structure:
Figure imgf000077_0001
wherein:
X is O or S; P is H or CH3;
R8 and R30 are each independently selected from H, Cι-8 straight chain or branched alkyl, C3-8 cycloalkyl, C4-9 alkylcycloalkyl, or C2.8 alkenyl; R15 is H, F, CI, Br, I, R20, CF3, CC13, NR18R19, NR30COR20, NR30SO2R20, OR20, OCF3,
OCOR20, OSO2R20, SR20, SCF3, SCOR20, SO3R20, S02NR8R9, N02, CN, COOR20, COSR20, or CONRι8R19, with the proviso that when a position adjacent to Rι5 is substituted, then RιS and said adjacent position can together be selected from SCH2S, SCH2CH2S, OCH20, or OCH2CH20 to form a bi-cyclic structure;
R20 is H, Cι-8 straight chain or branched alkyl, C3.8 cycloalkyl, C4-9 alkylcycloalkyl, C2-8 alkenyl, aryl or alkylaryl;
Ris and Ri. are each independently selected from: H, Cι_8 straight chain or branched alkyl, C2- 8 alkenyl or cycloalkyl, or alkylcycloalkyl, or aryl or CH2aryl wherein each moiety within said Cι-8 straight chain or branched alkyl, C2.8 alkenyl or cycloalkyl, or alkylcycloalkyl, or aryl or CH2aryl may be optionally substituted by up to four substituents in any position, whereby each substituent is independently selected from: F, CI, Br, I, CF3, CC13, CH3, C2H5, C3H7, C4H9, NH2, NHCH3, N(CH3)2, NHC2H5, N(C2H5)2, NHC3H7, N(C3H7)2, NHC4H9, N(C4H9)2, NHCOH, NHCOCH3, NHCOC2H5, NHCOC3H7, NHCOC4H9, NHS02CH3, NHS02C2H5, NHS02C3H7, NHS02C4H9, OH, OCH3, OC2H5, OC3H7, OC4H7, OC4H9, OC5H9, OC5Hu, OC6Hn, OC63, OCF3, OCOCH3, OCOC2H5, OCOC3H7, OCOC4H9, OS02CH3, OS02C2H5, OS02C3H7, OS02C4H9, SH, SCH3, SC2H5, SC3H7, SC4H7, SC^, SC5H9, SC5HU, SC6H„, SC6HI3, SCF3, SCOCH3, SC0C2H5, SC0C3H7, SCOC4H9, S03CH3, S03C2H5, S03C3H7, S03C4H9, S02NH, S02NH2, S02NHCH3) S02N(CH3)2, S02NHC2H5, S02N(C2H5)2, S02NHC3H7, S02N(C3H7)2, S02NHC4H9, S02N(C4H9)2, N02, CN, C00CH3, C00C2H5, C00C3H7, COOC .H9, COSCH3, COSC2H5, C0SC3H7, C0SC4H9, CONH2, CONHCH3, CON(CH3)2, CONHC2H5, CON(C2H5)2, CONHC3H7, CON(C3H7)2, CONHC4H9, CON(C4H9)2, witii die proviso that when either of R18 or R19 contain an aryl ring substituted at at least two adjacent positions on said aryl ring, then said two adjacent positions can together be selected from SCH2S, SCH2CH2S, OCH20, or OCH2CH20 to form a bicyclic stracture; Rι3, Rι4, R15, Ri6 and Rn are each independently selected from the following: F, CI, Br, I, CF3, CC13, CH3, C2H5, C3H7, C4H9, NH2, NHCH3; N(CH3)2, NHC2H5, N(C2H5)2, NHC3H7, N(C3H7)2, NHC4H9, N(C4H9)2, NHCOH, NHCOCH3, NHC0C2H5, NHC0C3H7, NHCOC4H9, NHS02CH3, NHS02C2H5, NHS02C3H7, NHS02C4H9, OH, OCH3, OC2H5, OC3H7, 0C4H7, 0C4H9, 0C5H9, OC5H11, OC6H„, OC6H13, OCF3, OCOCH3, OCOC2H5, OCOC3H7, OCOC4H9, OS02CH3, OS02C2H5, 0S02C H7, 0S02C4H9, SH, SCH3, SC2H5, SC3H7, SC4H7, SC4H , SC5H9, SC5H11, SCgHn, SCβHι3, SCF3, SCOCH3, SC0C2H5, SC0C3H7, SCOQH. , S03CH3, S03C2H5, S03C3H7, S03C4H9, S02NH, S02NH2, S02NHCH3, S02N(CH3)2, S02NHC2H5, S02N(C2H5)2, S02NHC3H7, S02N(C3H7)2, S02NHC4H9, S02N(C4H9)2, N02, CN, COOCH3, COOC2H5, C00C3H7, C00C4H9, COSCH3, COSC2H5, COSC3H7, COSC4H9, CONH2, CONHCH3, CON(CH3)2, CONHC2H5, CON(C2H5)2, CONHC3H7, CON(C3H7)2, CONHC4H9, CON(C4H9)2, and with the proviso that when any two adjacent positions of Rι3, Rι , R15, R]6 and Rn are substituted, said two adjacent positions can together be further selected from SCH2S, SCH2CH2S, OCH20, or OCH2CH20 to form a bi-cyclic structure; with the proviso that at least two of Rι3, R1 , R15, Rι6 and R17must be H.
[0227] In some embodiments of the foregoing genus, compounds of Example 13, Experiments 1-43, infra, and combinations or subcombinations thereof, are included.
[0228] In some embodiments of the foregoing genus, compounds of Example 13,
Experiments 1-43, infra, and combinations or subcombinations thereof, are not included. [0229] In some embodiments, compounds possessing 5-HT2A receptor activity that are useful as inverse agonists at such receptors are designated by the general Formula (XV):
Figure imgf000079_0001
(XV) wherein: i) Ar is a phenyl ring optionally substituted with up to five groups selected from tiie group consisting of halogen, OR7, OH, NR_R9, carboxy, CN, alkoxycarbonyl, straight chain or branched Cι-6 alkyl -C(p) or -0-C(p)3 where p is halogen;
R8 and R9 are independently a H, or Cι-6 alkyl, or C2.6 alkenyl, or cycloalkyl, or aryl, or CH2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from halogen, CF3, OCF3,
OEt, CC13, Me, N02, OH, OMe, SMe, COMe, CN, COOR10, SO3R10, COEt,
NHCOCH3, or aryl; or
R8 and R9 may form part of a 5, 6 or 7 membered cyclic structure which may be either saturated or unsaturated and that may contain up to four heteroatoms selected from O, N or S and said cyclic structure may be optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02,
OH, OMe, OEt, OCF3, SMe, COOR10) SO^RsRs, SO3R10, NHCOCH3, COEt,
COMe, or halogen; R7 is H or C1-6 alkyl;
Rio is H or Cι-6 alkyl;
ii) R2 is H, straight chain or branched Cι-6 alkyl, C2.6 alkenyl, or cycloalkyl; iii) R3 is halogen, carboxy, CN, alkoxycarbonyl, straight chain or branched CM alkyl, C2_ 6 alkenyl, C2„6 alkynyl, cycloalkyl, aryl or heteroaryl, wherein each of said alkoxycarbonyl, straight chain or branched -6 alkyl, C2.6 alkenyl, C2.6 alkynyl, cycloalkyl, aryl or heteroaryl groups can be optionally substituted with up to four substituents in any position selected from OH, ORio, NR8R9, halogen, -C(p)3> or -O- C(p)3 where p is halogen, and said cycloalkyl, aryl or heteroaryl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position; iv) R is Ci -6 alkyl, C2.6 alkenyl, or cycloalkyl; v) A is C(=0), C(=S) or S02;
Figure imgf000080_0001
q is O or 1; m is 0 or 1; n is 0 or 1 ;
Rn and Rl2 are each independently H, straight chain or branched Cι_6 alkyl, C2-6 alkenyl, or cycloalkyl;
Figure imgf000080_0002
wherein:
Ri3, Rι , R15, Riβ and R are each independently H, halogen, CN, NR8R9, COORio, SRio, straight chain or branched C1-6 alkyl, C2.6 alkenyl, C2.6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl, wherein each of said alkoxycarbonyl, straight chain or branched Cι_6 alkyl, C2.6 alkenyl, C2-6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl groups can be optionally substituted with up to four substituents in any position selected from N02, OR10, NR8R9, halogen, -C(p)3, or -0-C(p)3 where p is halogen, and said cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position;
L2 is -0-Q2 wherein Q2 is straight chain or branched -β alkyl,
C2_6 alkenyl, C2.6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl, wherein each of said cycloalkyl, aryl, alkylaryl, or arylalkyl groups can be optionally substituted with up to four substituents in any position selected from N02, OR7, halogen, -C(p)3j or -0-C(p)3 where p is halogen, or an aliphatic or aromatic heterocycle, and said cycloalkyl, aryl, alkylaryl, and arylalkyl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position; or a pharmaceutically acceptable salt.
Methods
[0230] " The present invention further provides methods for modulating die activity of a human 5-HT2A serotonin receptor by contacting the receptor with a compound of formula:
Figure imgf000081_0001
(I) wherein: i) Ri is H, halogens, NR5R_, OH or OR7, wherein
R5 and Re are independently H, or Cι-6 alkyl, or C2-6 alkenyl, or cycloalkyl, or aryl, or CH2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02, OH, OMe, OEt, CONR8R9, NR8R9, NHCOCH3, OCF3, SMe, COOR10, S03R8,
S02NR8R9, COMe", COEt, CO-lower alkyl, SCF3, CN, C2.6 alkenyl, H, halogens, CM alkoxy, C3.6 cycloalkyl, Cι_6 alkyl, and aryl wherein each of the C3.6 cycloalkyl, Q .6 alkyl, or aryl groups may be further optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02, OH, OMe, OEt, CONR8R9, NR8R9, NHCOCH3, OCF3, SMe, COOR10, S02NR8R9, SO3R10, COMe,
COEt, CO-lower alkyl, SCF3, CN, C2.6 alkenyl, H, halogens, CM alkoxy, C3.6 cycloalkyl, C1-6 alkyl, and aryl; or
R5 and ^ may form part of a 5, 6 or 7 membered cyclic structure which may be either saturated or unsaturated and that may contain up to four heteroatoms selected from O, N or S and said cyclic structure may be optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02, OH, OMe, OEt, OCF3, SMe, COOR10, S02NRsR9, SO30, NHCOCH3, COEt, COMe, or halogen; R8 and R9 are independently a H, or C .6 alkyl, or C2-6 alkenyl, or cycloalkyl, or aryl, or CH2 aryl group and each said group may be optionally substituted by up to four substituents in any position independentiy selected from halogen, CF3, OCF3,
OEt, CC13, Me, N02, OH, OMe, SMe, COMe, CN, COOR10, SO30, COEt, NHCOCH3, or aryl;
Rio is H or CM alkyl; R7 is H or Cι_6 alkyl; ii) R2 is H, straight chain or branched Cι-6 alkyl, C2.6 alkenyl, or cycloalkyl; iii) R3 is halogen, carboxy, CN, alkoxycarbonyl, straight chain or branched Ci . alkyl, C2- 6 alkenyl, C2.6 alkynyl, cycloalkyl, aryl or heteroaryl, wherein each of said alkoxycarbonyl, straight chain or branched C e alkyl, C2.6 alkenyl, C2_6 alkynyl, cycloalkyl, aryl or heteroaryl groups can be optionally substituted with up to four substituents in any position selected from OH, OR10, NRϋR9, halogen,
-C(p)3j or -0-C(p)3 where p is halogen, and said cycloalkyl, aryl or heteroaryl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position; iv) R4 is Cι_6 alkyl, C2-6 alkenyl, or cycloalkyl; v) A is C(=0), C(=S) or S02;
Figure imgf000082_0001
L, is:
Figure imgf000082_0002
q is 0 or 1 ; m is 0 or 1; n is 0 or 1 ; Ru and Rι2 are each independently H, straight chain or branched CM alkyl, C2-6 alkenyl, or cycloalkyl;
Figure imgf000082_0003
wherein: R13, R14, R15, R16 and R17 are each independently H, halogen, CN,
NR8R9, COORJO, SR10, straight chain or branched Cι-6 alkyl, C2.6 alkenyl, C2.6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl, wherein each of said alkoxycarbonyl, straight chain or branched Cι_6 alkyl, C2.6 alkenyl, C2.6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl groups can be optionally substituted with up to four substituents in any position selected from N02, ORio, NR8R9, halogen, -C(p)3j or -0-C(p)3 where p is halogen, and said cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position;
is -0-Q2 wherein Q2 is straight chain or branched _6 alkyl,
C2.s alkenyl, C2._ alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl, wherein each of said cycloalkyl, aryl, alkylaryl, or arylalkyl groups can be optionally substituted with up to four substituents in any position selected from N02, OR7, halogen, -C(p)3, or -0-C(p)3 where p is halogen, or an aliphatic or aromatic heterocycle, and said cycloalkyl, aryl, alkylaryl, and arylalkyl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position; or a pharmaceutically acceptable salt.
[0231] An aryl moiety can be a 5 or 6 membered aromatic heterocyclic ring (containing up to
4 hetero atoms independently selected from N, O, or S) or a 6 membered aromatic non-heterocyclic ring or a polycycle.
[0232] In some embodiments of the foregoing genus, compounds of Example 13,
Experiments 1-43, infra, and combinations or subcombinations thereof, are included.
[0233] In some embodiments of the foregoing genus, compounds of Example 13,
Experiments 1-43, infra, and combinations or subcombinations thereof, are not included.
[0234] Also provided by the present invention are methods for modulating the activity of a human 5-HT2A serotonin receptor by contacting the receptor with a compound of formula:
Figure imgf000084_0001
(A) wherein:
R2 is H or lower alkyl(Cι_ι); R3 is lower alkyl (Cι.6), or halogen; R4 is lower alkyl (Cι_6);
X is eidier Oxygen or Sulfur; Y is NR15Ri6, or (CH2)mR17, or 0(CH2)nR17; m=0-4 n=0-4 R15 is H or lower alkyl(CM);
R16 and Rn are independently Cι_6 alkyl, or C2.β alkenyl, or cycloalkyl, or aryl, or CH2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02, OH, OMe, OEt, CONR189, NR18R19, NHCOCH3, OCF3, SMe, COOR20, SO3R20, S02NR18R19, COMe, COEt, CO-lower alkyl, SCF3, CN, C2.6 alkenyl, H, halogens, C alkoxy, C3-6 cycloalkyl, d_6 alkyl, and aryl;
Ris and R19 are independently a H or Cι-6 alkyl, or C2.6 alkenyl, or cycloalkyl, or aryl, or CH2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02, OH, OMe, OEt, C0NR21R22, NR21R22, NHCOCH3, OCF3, SMe, C00R23, S03R23, S02NR2ιR22, COMe, COEt, CO-lower alkyl, SCF3, CN, C2-6 alkenyl, H, halogens, .
4 alkoxy, C3.6 cycloalkyl, Cι.6 alkyl, and aryl; or Ris and Rι9 may form part of a 5, 6 or 7 membered cyclic structure which may be either saturated or unsaturated and that may contain up to four heteroatoms selected from O, N or S and said cyclic structure may be optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02,
OH, OMe, OEt, OCF3, SMe, C00R23, S02NR21R22, S03R23, NHCOCH3, COEt, COMe, or halogen;
R20 and R23 are each independently selected from H or Cι_6 alkyl; R2ι and R22 are each independently are independently a H, or C1-6 alkyl, or C2- 6 alkenyl, or cycloalkyl, or aryl, or CH2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from halogen, CF3, OCF3, OEt, CC13, Me, N02, OH, OMe, SMe, COMe, CN, COOR20, SO3R20, COEt, NHCOCH3, or aryl; an aryl moiety can be a 5 or 6 membered aromatic heterocyclic ring (containing up to 4 hetero atoms independently selected from N, O, or S) or a 6 membered aromatic non-heterocyclic ring or a polycycle.
[0235] In some embodiments of the foregoing genus, compounds of Example 13, Experiments 1-43, infra, and combinations or subcombinations thereof, are included.
[0236] In some embodiments of the foregoing genus, compounds of Example 13,
Experiments 1-43, infra, and combinations or subcombinations thereof, are not included.
[0237] The present invention further provides methods for modulating the activity of a human 5-HT2A serotonin receptor by contacting the receptor with a compound of formula:
Figure imgf000085_0001
wherein:
R3 is F, Cl, Br, I, C1-6 straight chain or branched alkyl, C3-8 cycloalkyl, C4.9 alkylcycloalkyl, or C2.6 alkenyl; Xis O or S;
Y is NR15R16, or (CH2)mR17, or 0(CH2)nR17; m is an integer between 0 and 4, inclusive; n is an integer between 0 and 4, inclusive; is H, Cι-8 straight chain or branched alkyl, C3-8 cycloalkyl, C4-9 alkylcycloalkyl, or C2-8 alkenyl;
R2 and R15 ais each independently selected from H, Cι.g straight chain or branched alkyl, C3.8 cycloalkyl, C4-9 alkylcycloalkyl, or C2-8 alkenyl; RJ6 and Rn is each independently selected from: Cι.8 straight chain or branched alkyl, C2-8 alkenyl or cycloalkyl, or alkylcycloalkyl, or aryl, or CH2aryl, wherein each moiety within said C1-8 straight chain or branched alkyl, C2.8 alkenyl or cycloalkyl, or alkylcycloalkyl, or aryl or CH2aryl may be optionally substituted by up to four substituents in any position, whereby each substituent is independently selected from: H, F, Cl, Br, I, R20, CF3, CF2R7, CF2CF2, CC13, CC12R7, CC12CC12R7, NR18R19, NR18COR20, NRι8SO2R20, OR20, OCF3, OCF2R20, OCF2CF2R20, OCOR20, OSO2R20, OPO(OR20)2, SR20, SCF3, SCF2R20, SCF2CF2R20, SCOR20, SO3R20, S02NRι89, PO(OR20)3, PO(OR20)2R20, N02, CN, CNR20(NR18R19), CNR18(SR20), COOR20, COSR20, CONR18R19, with the proviso that when Rι6 or R17 contains an aryl ring substituted at at least two adjacent positions on said aryl ring, then said two adjacent positions can together be selected from SCH2S, SCH2CH2S, OCH20, or OCH2CH20 to form a bi-cyclic stracture;
R20 is H, Cι_8 straight chain or branched alkyl, C3.8 cycloalkyl, C4-9 alkylcycloalkyl, C2.8 alkenyl, aryl or alkylaryl; Ris and R19 are each independently selected from H, Cι-8 straight chain or branched alkyl, C2.8 alkenyl or cycloalkyl, or alkylcycloalkyl, or aryl, or CH2aryl, wherein each moiety within said Cι-8 straight chain or branched alkyl, C2.8 alkenyl or cycloalkyl, or alkylcycloalkyl, or aryl or CH2aryl may be optionally substituted by up to four substituents in any position, whereby each substituent is independently selected from: F, Cl, Br, I, CF3, CC13, CH3, C2H5, C3H7, C4H9, NH2, NHCH3, N(CH3)2, NHC2H5, N(C2H5)2, NHC3H7, N(C3H7)2, NHC4H9, N(C4H9)2, NHCOH, NHCOCH., NHC0C2H5,
NHCOC3H7, NHCOC4H9, NHS02CH3, NHS02C2H5, NHS02C3H7, NHS02C4H9, OH, OCH3, OC2H5,
OC3H7, OC4H7, OC4H9, OC5H9, OC5H„, OC6H„, OC6H13, OCF3, OCOCH3, OCOC2H5, OCOC3H7,
OCOC4H9, OS02CH3, OS02C2H5, OS02C3H7, OS02C4H9, SH, SCH3, SC2H5, SC3H7, SC4H7, SGd ,
SC5H9, SC5H„, SC6Hn, SC6Hi3„ SCF3, SCOCH3, SC0C2H5, SCOC3H7, SCOG,H9, S03CH3, S03C2H5, S03C3H7, S03C4H9, S02NH, S02NH2, S02NHCH3, S02N(CH3)2, S02NHC2H5,
S02N(C2H5)2, S02NHC3H7, S02N(C3H7)2, S02NHC4H9, S02N(C4H9)2, N02, CN, COOCH3,
C00C2H5; COOC3H7, C00C4H9, COSCH3, C0SC2H5, COSC3H7, COSC4H9, CONH2, CONHCH3,
CON(CH3)2, CONHC2H5, CON(C2H5)2, CONHC3H7, CON(C3H7)2, CONHC4H9, CON(C4H9)2, and with the proviso that when either or both of Rι8 or Rι9 contain an aryl ring substituted at at least two adjacent positions on said aryl ring, then said two adjacent positions can together be selected from SCH2S, SCH2CH2S, OCH20, or OCH2CH20 to form a bi-cyclic structure; or
Ris or R]9 may together form part of a 5, 6 or 7 membered cyclic stracture, with said stracture being saturated or unsaturated, and further with said stracture containing up to four heteroatoms selected from O, N or S, and further wherein each moiety within said cyclic structure being optionally substituted by up to four substituents in any position independently selected from: F, Cl, Br, I, CF3, CC13, CH3, C2H5, C3H7, C4H9, NH2, NHCH3; N(CH3)2, NHC2H5, N(C2H5)2, NHC3H7, N(C3H7)2, NHC4H9, N(C4H9)2, NHCOH, NHCOCH3, NHCOC2H5, NHCOC3H7, NHCOQH,, NHS02CH3, NHS02C2H5, NHS02C3H7, NHS02C4H9, OH, OCH3, OC2H5, OC3H7, OGJH?, OC4H9, OC5H9, OC5H„, OC6H„, OC6Hi3, OCF3, OCOCH3, OCOC2H5, OCOC3H7, OCOC4H9, OS02CH3, OS02C2H5, 0S02C3H7, OS02C4H9, SH, SCH3, SQ2H5, SC3H7, SC4H7, SGdlg, SCsH9, SC5H11, SCgHn, SCgHι3„ SCF3, SCOCH3, SCOC2H5, SCOC3H7, SCOC4H9, S03CH3, S03C2H5, S03C3H7, S03C4H9, S02NH, S02NH2, S02NHCH3, S02N(CH3)2, S02NHC2H5, S02N(C2H5)2, S02NHC3H7, S02N(C3H7)2, S02NHC4H9, S02N(C4H9)2, N02, CN, COOCH3, C00C2H5, C00C3H7, COOC4H9, COSCH3, COSC2H5) COSC3H7, COSC4H9, CONH2, CONHCH3, CON(CH3)2, CONHC2H5, CON(C2H5)2, CONHC3H7, CON(C3H7)2, CONHC4H9, CON(C4H9)2, and with the proviso that wherein when Rι8 or Rι9 form an aryl ring substituted at at least two adjacent positions on said aryl ring, then said two adjacent positions can together be selected from SCH2S, SCH2CH2S, OCH20, or OCH2CH20 to form a bi-cyclic structure.
[0238] An aryl moiety can be a 5 or 6 membered aromatic heterocyclic ring (containing up to
4 hetero atoms independently selected from N, O, or S) or a 6 membered aromatic non-heterocyclic ring or a polycycle.
[0239] In some embodiments of the foregoing genus, compounds of Example 13,
Experiments 1-43, infra, and combinations or subcombinations thereof, are included.
[0240] In some embodiments of die foregoing genus, compounds of Example 13,
Experiments 1-43, infra, and combinations or subcombinations thereof, are not included.
[0241] Also provided by the present invention are methods for modulating the activity of a human 5-HT2A serotonin receptor by contacting the receptor with a compound of formula:
Figure imgf000088_0001
(A2) wherein:
X is O or S; P is H or CH3; R8 and R20 are each independently selected from H, Cι-8 straight chain or branched alkyl, C3-8 cycloalkyl, C4.9 alkylcycloalkyl, or C2.8 alkenyl;
R15 is H, F, Cl, Br, I, R20, CF3, CF2R20, CF2CF2, CC13, CC12R20, CC12CC12R20, NR18R19, NR19COR20, NR19SO2R20, OR20, OCF3, OCF2R20, OCF2CF2R20, OCOR20, OSO2R20, OPO(OR20)2, SR20, SCF3, SCF2R20, SCF2CF2R20, SCOR20, SO3R20, S02NR189, PO(OR20)3, PO(OR20)2R20, N02, CN, CNR20(NR189), CNR19(SR20), COOR20, COSR20, CONRι89, with the proviso that when a position adjacent to Rι5 is substituted, then Rι5 and said adjacent position can together be selected from SCH2S, SCH2CH2S, OCH20, or
OCH2CH20 to form a bi-cyclic stracture;
Ris and Rι9 are each independently selected from H, C].s straight chain or branched alkyl, C2.8 alkenyl or cycloalkyl, or alkylcycloalkyl, or aryl, or CH2aryl, wherein each moiety within said Cι_8 straight chain or branched alkyl, C2-8 alkenyl or cycloalkyl, or alkylcycloalkyl, or aryl or CH2aryl may be optionally substituted by up to four substituents in any position, whereby each substituent is independently selected from: F, Cl, Br, I, CF3, CC1_, CH3, C2H5, C3H7, C4H9, NH2, NHCH3, N(CH3)2,
NHC2H5, N(C2H5)2, NHC3H7, N(C3H7)2, NHG,H9, N(C4H9)2, NHCOH, NHCOCH3, NHCOC2H5, NHCOC3H7, NHCOC4H9, NHS02CH3, NHS02C2H5, NHS02C3H7, NHS02C4H9, OH, OCH3, OC2H5,
OC3H7, OC4H7, OC4H9, OC5H9, OC5H„, OC6Hn, OC63, OCF3, OCOCH3, OCOC2H5, OCOC3H7,
OCOC4H9, OS02CH3, OS02C2H5, OS02C3H7, OS02C4H9, SH, SCH3, SC2H5, SC3H7, SC4H7, SC4H9,
SC5H9, SC5H„, SC6Hιι, SC6H13„ SCF3, SCOCH3, SCOC2H5, SC0C3H7, SCOC4H., S03CH3,
S03C2H5, S03C3H7, S03C4H9, S02NH, S02NH2, S02NHCH3j S02N(CH3)2) S02NHC2H5, S02N(C2H5)2, S02NHC3H7, S02N(C3H7)2, S02NHC4H9, S02N(C4H9)2, N02, CN, COOCH3,
COOC2H5, COOC3H7, COOC4H9, COSCH3, COSC2H5, COSC3H7, COSC4H9, CONH2, CONHCH3,
CON(CH3)2, CONHC2H5, CON(C2H5)2, CONHC3H7, CON(C3H7)2, CONHC4H9, CON(C4H9)2, and with the proviso that when either or both of R18 or Rι9 contain an aryl ring substituted at at least two adjacent positions on said aryl ring, then said two adjacent positions can together be selected from SCH2S, SCH2CH2S, OCH20, or OCH2CH20 to form a bi-cyclic structure;
R13, Rι4, R15, Ri6 and R each independently selected from the following: F, Cl, Br, I, CF3, CC13, CH3, C2H5, C3H7, C4H9, NH2, NHCH3, N(CH3)2, NHC2H5, N(C2H5)2, NHC3H7, N(C3H7)2,
NHC4H9, N(C4H9)2, NHCOH, NHCOCH3, NHCOC2H5, NHC0C3H7, NHCOC4H9, NHS02CH3,
NHS02C2H5, NHS02C3H7, NHS02C4H9, OH, OCH3, 0C2H5j OC3H7, OC4H7, 0C4H9, OC5H9,
OC5H„, OC6Hn, OC6H13, OCF3, OCOCH3, OCOC2H5, OCOC3H7, OCOC4H9, OS02CH3, OS02C2H5,
OS02C3H7, OS02C4H9, SH, SCH3, SC2Hs, SC3H7, SC4H7, SC4H9, SCsH9, SC5H11, SCβHn, SC6H13,, SCF3, SCOCH3, SCOC2H5, SCOC3H7, SCOC4H9, S03CH3, S03C2H5, S03C3H7, S03C4H9, S02NH,
S02NH2, S02NHCH3> S02N(CH3)2, S02NHC2H5, S02N(C2H5)2, S02NHC3H7, S02N(C3H7)2,
S02NHC4H9, S02N(C4H9)2, N02, CN, COOCH3, C00C2H5, COOC3H7, COOC4H9, COSCH3,
COSC2H5, COSC3H7, COSC4H9, CONH2, CONHCH3> CON(CH3)2, CONHC2H5, CON(C2H5)2,
CONHC3H7, CON(C3H7)2, CONHC4H9, CON(C4H9)2, and with the proviso that when any two adjacent positions of R13, Rι , R15, Rι6 and R re substituted, said two adjacent positions can together be further selected from SCH2S,
SCH2CH2S, OCH20, or OCH2CH20 to form a bi-cyclic structure; and with the proviso that at least one of Rι3, Rι , R15, R16 and Rn ustbe H.
[0242] An aryl moiety can be a 5 or 6 membered aromatic heterocyclic ring (containing up to 4 hetero atoms independently selected from N, O, or S) or a 6 membered aromatic non-heterocyclic ring or a polycycle.
[0243] In some embodiments of the foregoing genus, compounds of Example 13,
Experiments 1-43, infra, and combinations or subcombinations thereof, are included.
[0244] In some embodiments of the foregoing genus, compounds of Example 13, Experiments 1-43, infra, and combinations or subcombinations thereof, are not included.
[0245] The present invention further provides methods for modulating the activity of a human 5-HT2A serotonin receptor by contacting the receptor with a compound of formula:
Figure imgf000090_0001
(B) wherein:
R2 is H or lower alkyl (CM); R3 is Me, or Et, or halogen; X is either Oxygen or Sulfur;
Y is NRι5R16, or (CH2)mR17, or 0(CH2)nR17; R4 is lower alkyl (Cι_6); m=0-4 n=0-4 R15 is H or lower alkyl(CM);
R16 and R are independently CM alkyl, or C2.6 alkenyl, or cycloalkyl, or aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02, OH, OMe, OEt, C0NR189, NRι89, NHCOCH3, OCF3, SMe, COOR20, SO3R20, S02NR18R19, COMe, COEt, CO-lower alkyl, SCF3, CN, C2.6 alkenyl, H, halogens, C alkoxy, C3._ cycloalkyl, Cι-6 alkyl, aryl and aryloxy wherein each of the CM alkoxy, C3.6 cycloalkyl, -6 alkyl, aryl and aryloxy groups may be further optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02, OH, OMe, OEt, CONR_ιR22, NR2ιR22, NHCOCH3, OCF3, SMe, COOR23, S03R23, S02NR21R22, COMe, COEt, CO-lower alkyl, SCF3, CN, C2.6 alkenyl, H, halogens, Cμ4 alkoxy, C3.β cycloalkyl, C alkyl, and aryl;
Ris and R19 are independently a H or -6 alkyl, or C2.6 alkenyl, or cycloalkyl, or aryl, or CH2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02, OH, OMe, OEt, CONR2ιR22, NR21R22, NHCOCH3, OCF3, SMe, COOR23, S03R23, S02NR2ιR22, COMe, COEt, CO-lower alkyl, SCF3, CN, C2.6 alkenyl, H, halogens, Q.
4 alkoxy, C3.6 cycloalkyl, C alkyl, and aryl or R18 and Ri9 may form part of a 5, 6 or 7 membered cyclic structure which may be either saturated or unsaturated and that may contain up to four heteroatoms selected from O, N or S and said cyclic structure may be "optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02, OH, OMe, OEt, OCF3, SMe, COOR7, S02NR24R25, S03R26, NHCOCH3, COEt, COMe, or halogen;
R20 and R23 are each independently selected from H or C!-6 alkyl; R21 and R22 are each independently are independently a H, or C1-5 alkyl, or C2- 6 alkenyl, or cycloalkyl, or aryl, or CH2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from halogen, CF3, OCF3, OEt, CC13, Me, N02, OH, OMe, SMe, COMe, CN, COOR20, SO3R20, COEt, NHCOCH3, or aryl.
[0246] An aryl moiety can be a 5 or 6 membered aromatic hetero-cyclic ring (containing up to 4 hetero atoms independently selected from N, O, or S) or a 6 membered aromatic non-heterocyclic ring or a polycycle.
[0247] In some embodiments of the foregoing genus, compounds of Example 13,
Experiments 1-43, infra, and combinations or subcombinations thereof, are included.
[0248] In some embodiments of the foregoing genus, compounds of Example 13,
Experiments 1-43, infra, and combinations or subcombinations thereof, are not included.
[0249] The present invention further provides methods for modulating the activity of a human 5-HT2A serotonin receptor by contacting the receptor with a compound of formula:
Figure imgf000091_0001
(XV) wherein: ii) Ar is a phenyl ring optionally substituted with up to five groups selected from the group consisting of halogen, OR7, OH, NRs 9, carboxy, CN, alkoxycarbonyl, straight chain or branched CM alkyl -C(p)3j or -0-C(p)3 where p is halogen; Rs and R9 are independentiy a H, or CM alkyl, or C2-6 alkenyl, or cycloalkyl, or aryl, or CH2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from halogen, CF3, OCF3, OEt, CC13, Me, N02, OH, OMe, SMe, COMe, CN, COOR10, SO3R10, COEt, NHCOCH3, or aryl; or
R8 and R9 may form part of a 5, 6 or 7 membered cyclic structure which may be either saturated or unsaturated and that may contain up to four heteroatoms selected from 0, N or S and said cyclic structure may be optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02, OH, OMe, OEt, OCF3, SMe, COORio, SOzNRsRc,, SO3R10, NHCOCH3, COEt, COMe, or halogen;
R7 is H or C alkyl; Rio is H or CM alkyl;
ii) R2 is H, straight chain or branched C alkyl, C2.6 alkenyl, or cycloalkyl; iv) R3 is halogen, carboxy, CN, alkoxycarbonyl, straight chain or branched CM alkyl, C2_
6 alkenyl, C2.6 alkynyl, cycloalkyl, aryl or heteroaryl, wherein each of said alkoxycarbonyl, straight chain or branched C1-6 alkyl, C2_β alkenyl, C2.6 alkynyl, cycloalkyl, aryl or heteroaryl groups can be optionally substituted with up to four substituents in any position selected from OH, ORio, NRsR9, halogen, -C(p)3; or -O-
C(p)3 where p is halogen, and said cycloalkyl, aryl or heteroaryl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position; iv) R4 is C alkyl, C2.6 alkenyl, or cycloalkyl; v) A is C(=0), C(=S) or S02;
Figure imgf000092_0001
q is O or 1; m is O or 1; n is O or 1;
Rn and R12 are each independently H, straight chain or branched C alkyl, C2.6 alkenyl, or cycloalkyl;
Figure imgf000092_0002
Figure imgf000093_0001
wherein:
3, Rι4, Ris, Ri6 and R are each independently H, halogen, CN, NR8R9, COORio, SRio, straight chain or branched Cι-6 alkyl, C -6 alkenyl, C2.6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl, wherein each of said alkoxycarbonyl, straight chain or branched CM alkyl, C2.6 alkenyl, C2.6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl groups can be optionally substituted with up to four substituents in any position selected from N02, ORio, NRsR. , halogen, -C(p)3; or -0-C(p)3 where p is halogen, and said cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position;
1^ is -O-Q2 wherein Q2 is straight chain or branched CM alkyl, C2.6 alkenyl, C2.β alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl, wherein each of said cycloalkyl, aryl, alkylaryl, or arylalkyl groups can be optionally substituted with up to four substituents in any position selected from N02, OR7, halogen, -C(p)3> or -0-C(p)3 where p is halogen, or an aliphatic or aromatic heterocycle, and said cycloalkyl, aryl, alkylaryl, and arylalkyl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position; or a pharmaceutically acceptable salt.
[0250] In some embodiments of the foregoing methods, the compounds are selected from compounds and combinations and subcombinations of compounds selected from the group consisting of:
Figure imgf000093_0002
Compound 1 Compound 2
Figure imgf000094_0001
Compound 8 Compound 9
Figure imgf000094_0002
Compound 10 Compound 11
Figure imgf000094_0003
Compound 12 Compound 13
Figure imgf000094_0004
Compound 14 Compound 15
Figure imgf000094_0005
Compound 16 Compound 17
Figure imgf000095_0001
Compound 18 Compound 19
Figure imgf000095_0002
Compound 20 Compound 21
Figure imgf000095_0003
Compound 24 Compound 25
Figure imgf000095_0004
Compound 26 Compound 27
Figure imgf000096_0001
Compound 28 Compound 29
Figure imgf000096_0002
Compound 30 Compound 31
Figure imgf000096_0003
Compound 32
Figure imgf000096_0005
Figure imgf000096_0004
Compound 34 Compound 35
Figure imgf000097_0001
Compound 36 Compound 37
Figure imgf000097_0002
Compound 38 Compound 39
Figure imgf000097_0003
Compound 40 Compound 41
Figure imgf000097_0004
Compound 42 Compound 43
Figure imgf000097_0005
Compound 44 Compound 45
Figure imgf000098_0001
Compound 46 Compound 47
[0251] In some embodiments, the present invention provides the foregoing compounds, and combinations and subcombinations thereof.
[0252] In some preferred embodiments, the present invention provides compounds useful as inverse agonists for 5-HT2A receptors stracturally represented as follows:
Figure imgf000098_0002
Figure imgf000099_0001
[0253] Also provided by the present invention are compositions comprising each of the compounds ofthe invention.
[0254] Odier preferred compounds provided by the present invention and useful in the methods disclosed herein, include, but are not limited to:
Figure imgf000099_0002
Compound 48 Compound 49
Figure imgf000099_0003
Compound 50 Compound 51
Figure imgf000099_0004
Compound 52 Compound 53
Figure imgf000100_0001
Compound 54 Compound 55
Figure imgf000100_0002
Compound 56 Compound 57
Figure imgf000100_0003
Compound 58 Compound 59
Figure imgf000100_0004
Compound 60 Compound 61
Figure imgf000101_0001
Compound 62 Compound 63
Figure imgf000101_0002
Compound 64 Compound 65
Figure imgf000101_0003
Compound 66 Compound 67
Figure imgf000101_0004
Compound 68 Compound 69
Figure imgf000101_0005
Compound 70 Compound 71
Figure imgf000102_0001
Compound 72 Compound 73
Figure imgf000102_0002
Compound 74 Compound 75
Figure imgf000102_0003
Compound 76 Compound 77
Figure imgf000102_0004
Compound 78 Compound 79
Figure imgf000103_0001
Compound 80 Compound 81
Figure imgf000103_0002
Compound 82 Compound 83
Figure imgf000103_0003
Compound 84 Compound 85
Figure imgf000103_0004
Compound 86 Compound 87
Figure imgf000104_0001
Compound 88 Compound 89
Figure imgf000104_0002
Compound 90 Compound 91
Figure imgf000104_0003
Compound 92 Compound 93
Figure imgf000104_0004
Compound 94 Compound 95
Figure imgf000105_0001
Compound 96 Compound 97
Figure imgf000105_0002
Compound 98 Compound 99
Figure imgf000105_0003
Compound 100 Compound 101
Figure imgf000105_0004
Compound 102 Compound 103
Figure imgf000106_0001
Compound 104 Compound 105
Figure imgf000106_0002
Compound 106 Compound 107
Figure imgf000106_0003
Compound 108 Compound 109
Figure imgf000106_0004
Compound 110 Compound 111
Figure imgf000107_0001
Compound 112 Compound 113
Figure imgf000107_0002
Compound 114 Compound 115
Figure imgf000107_0003
Compound 116 Compound 117
Figure imgf000107_0004
Compound 118 Compound 119
Figure imgf000108_0001
Compound 120 Compound 121
Figure imgf000108_0002
Compound 122 Compound 123
Figure imgf000108_0003
Compound 124 Compound 125
Figure imgf000108_0004
Compound 126 Compound 127
Figure imgf000109_0001
Compound 128 Compound 129
Figure imgf000109_0002
Compound 130 Compound 131
Figure imgf000109_0003
Compound 132 Compound 133
Figure imgf000109_0004
Compound 134 Compound 135
10
Figure imgf000110_0001
Compound 136 Compound 137
Figure imgf000110_0002
Compound 138 Compound 139
Figure imgf000110_0003
Compound 140 Compound 141
Figure imgf000110_0004
Compound 142 Compound 143
Figure imgf000111_0001
Compound 144 Compound 145
Figure imgf000111_0002
Compound 146 Compound 147
Figure imgf000111_0003
Compound 148 Compound 149
Figure imgf000111_0004
Compound 150 Compound 151
Figure imgf000112_0001
Compound 152 Compound 153
Figure imgf000112_0002
Compound 154 Compound 155
Figure imgf000112_0003
Compound 156 Compound 157
Figure imgf000112_0004
Compound 158 Compound 159
Figure imgf000112_0005
Compound 160 Compound 161
Figure imgf000113_0001
Compound 162 Compound 163
Figure imgf000113_0002
Compound 164
[0255] Other preferred compounds of the present invention include compounds of the following group:
Figure imgf000113_0003
l-[3-(4-Bromo-2-methyl-2H-pyrazol- 1 -(3 ,4-Bis-trifluoromethyl-phenyl)- 3-yl)-phenyl]-3-(2-chloro-4- 3-[3-(4-bromo-2-mefhyl-2H- frifluoromef__yl-phenyl)-urea pyrazol-3-yl)-phenyl]-urea
Figure imgf000113_0004
3-[3-(4-Bromo-2-methyl-2H- l-[3-(4-Bromo-2-methyl-2H-pyrazol- pyrazol-3-yl)-phenyl]-l-(4-fluoro- 3-yl)-phenyl]-3-(3-fluoro-4- phenyl)- 1 -methyl-urea trifluoromethyl-phenyl)-urea
Figure imgf000114_0001
3-[3-(4-Bromo-2-mefhyl-2H- 3-[3-(4-Chloro-2-methyl-2H- pyrazol-3 -yl)-phenyl] - 1 -(4-chloropyrazol-3 -yl)-phenyl] - 1 -(4- phenyl)- 1 -methyl-urea chloro-phenyl)- 1 -methyl-urea
Figure imgf000114_0002
3-[3-(4-Chloro-2-methyl-2H- pyrazol-3 -yl)-phenyl]- 1 -(4- fluoro-phenyl)- 1 -methyl-urea
[0256] In some embodiments of each of the genera described herein, compounds of Example
13, Experiments 1-43, infra, and combinations or subcombinations thereof, are included.
[0257] In some embodiments of each ofthe genera described herein, compounds of Example
13, Experiments 1-43, infra, and combinations or subcombinations thereof, are not included.
[0258] Still other preferred compounds falling within die scope of the general Formula (I) have the Formula LX:
Figure imgf000114_0003
(LX) wherein:
R3 is Br or Cl; t is lower alkyl;
Figure imgf000114_0004
RnisH,F,ClorCF3;
R12 is H, F or Cl;
and combinations and subcombinations thereof.
Specifically preferred compounds of Formula (LX) above include the following:
Figure imgf000115_0001
Compound 165 Compound 166
Figure imgf000115_0002
Compound 167 Compound 168
Figure imgf000115_0003
Compound 169 Compound 170
Figure imgf000115_0004
Compound 171 Compound 172
Figure imgf000116_0001
Compound 173 Compound 174
Figure imgf000116_0002
Compound 175 Compound 176
Figure imgf000116_0003
Compound 177 Compound 178
Figure imgf000116_0004
Compound 179 Compound 180
Figure imgf000117_0001
Compound 181 Compound 182
[0260] The IC50 values for Compounds 165-182 ranged between 8 to 158 nM in the IP3 AP-3 assay with their corresponding values shown below:
Figure imgf000117_0002
EXAMPLE 12
GENERAL SYNTHETIC APPROACHES
[0261] The compounds disclosed in this invention may be readily prepared in accordance to a variety of synthetic manipulations, all of which would be familiar to one skilled in the art.
[0262] Compounds of general Formula (I) can be obtained via a variety of synthetic routes all of which would be familiar to one skilled in the art. The reaction of isocyanates with amines is a commonly practiced method for the formation of ureas (see Org. Syn. Coll. Vol. V, (1973), 555). Amine 12-1, 3-(4-bromo-2-methylpyrazole-3-yl)phenylamine (sold commercially as 3-(4-bromo-l- methylpyrazole-3-yl)phenylamine, by Maybridge Chemical Company, Catalog No. KM01978, CAS No. 175201-77-1) reacts readily with isocyanates 12-2 in inert solvents such as halocarbons to yield the desired ureas of Formula 12-3 as shown in Scheme 12-1.
Figure imgf000118_0001
12-1 12-3
Scheme 12-1
[0263] Alternatively the amine 12-1 can be converted to the corresponding isocyanate 12-4 by die action of phosgene or a suitable phosgene equivalent, e.g. friphosgene, in an inert solvent such as a halocarbon in the presence of an organic base such as triethylamine (i.e., TEA) or diisopropylethylamine (i.e., DIEA). Isocyanate 12-4 reacts with amines of the general formula 12-5 in an analogous fashion to that described above in Scheme 12-1 to give urea 12-6. This approach allows for diverse groups to be introduced for the R2 or R3 group based on the starting amine 12-5 (Scheme 12-2).
Figure imgf000119_0001
12-1 12-4 12-6 Scheme 12-2
[0264] Alternatively wherein the isocyanate of general formula 12-2 is not commercially available it can be prepared from the corresponding amine of general formula 12-7 in an analogous procedure to that described above for the preparation of 12-1. Reaction of ti ese isocyanates with 12-1 would again yield the requisite ureas of general formula 12-3 (Scheme 12-3).
Figure imgf000119_0002
12-1 12-3 dieme 12-3
[0265] Arnines of general formula 12-5 are also readily converted to activated isocyanate equivalents of general fonnula 12-8 by the sequential action of carbonyldiimidazole and metiiyl iodide in tefrahydrofuran and acetonitrile respectively (R.A. Batey et al, Tetrahedron Lett., (1998), 39, 6267-6270.) Reaction of 12-8 with amine 12-1 in an inert solvent such as a halocarbon would yield the requisite ureas of general formula 12-3 (Scheme 12-4).
Figure imgf000120_0001
Scheme 12-4
[0266] Amine 12-1 may be monomethylated according to the procedure of J. Barluenga et al,
J. Chem. Soc., Chem. Commun., (1984), 20, 1334-1335, or alkylated according to the procedure of P. Marchini et al, J. Org. Chem., (1975), 40(23), 3453-3456, to yield compounds of general formula 12- 9 wherein R1 = lower alkyl. Substituted amine 12-9 may be allowed to react with an isocyanate equivalent, either 12-2 or 12-8, in a similar manner as described herein to give urea 12-10 or 12-11 respectively (Scheme 12-5).
Figure imgf000121_0001
12-11
Scheme 12-5
[0267] Carbamates of general formula 12-12 can be obtained in a similar manner via a variety of synthetic manipulations, all of which would be familiar to one skilled in the art. The reaction of amine 12-1 witii chloroformates (see Org. Syn. Coll. Vol. IN, (1963), 780) of general formula 12-13 in an inert solvent such as ether or halocarbon in the presence of a tertiary base such as friethylamine or ethylisopropylamine readily yields the requisite carbamates of general formula 12-12 (Scheme 12-6).
Figure imgf000121_0002
Scheme 12-6 [0268] Analogously, amines of general formula 12-9 react similarly with chloroformates 12-
13 to yield d e requisite carbamates ofthe general formula 12-14 (Scheme 12-7).
Figure imgf000122_0001
Scheme 12-7
[0269] An alternate route employs the ready reaction of an alcohol with an isocyanate. Thus isocyanate 12-4 described previously reacts readily with alcohols 12-15 in an aprotic solvent such as ether or chlorocarbon to yield the desired carbamates of general formula 12-12 (Scheme 12-8). :
Figure imgf000122_0002
12-4 12-12
Scheme 12-8
[0270] Chloroformates of general formula 12-13 not commercially available may be readily prepared from the corresponding alcohol 12-15 in an inert solvent such as toluene, chlorocarbon or etiier by the action of excess phosgene (see Org. Syn. Coll. Vol. Ill, (1955), 167) (Scheme 12-9).
O
Phosgene
HO(CH2)nR4 - O CIl A" ^,0(CH2)nR4
12-15 12-13
Scheme 12-9
[0271] Amide compounds of the general formula 12-16 can be obtained via a variety of synthetic manipulations, all of which would be familiar to one skilled in the art. The reaction of amine 12-1 with acid chlorides (see Org. Syn. Coll. N, (1973), 336) of general formula 12-17 to yield the desired amides 12-16 is readily achieved in an inert solvent such as chloroform or dichloromethane in the presence of an organic base such as friethylamine or emyldiisopropylamine (Scheme 12-10).
Figure imgf000123_0001
Scheme 12-10
[0272] In an identical fashion amines of general fonnula 12-9 would react with acid chlorides 12-17 to yield the desired amides 12-18 (Scheme 12-11).
Figure imgf000123_0002
Scheme 12-11
[0273] Alternatively the corresponding acids of general formula 12-19 may be coupled with dicyclohexylcarbodiimide (DCC)/hydroxybenzofriazole (HOBT) (see W. Konig et al, Chem. Ber., (1970), 103, 788) or hydroxybenzotriazole (HOBT)/2-(lH-benzotriazole-l-yl)-l, 1,3,3- tetramethyluronium hexafluorophosphate (HBTU) (see M. Bematowicz et al., Tetrahedron Lett., (1989), 30, 4645) as condensing agents in dimelhylformarnide or chloroform with amines 12-1 and 12-9 respectively to give amides 12-16 (Schemes 12-12) and 12-18 (Scheme 12-13) respectively as described above.
Figure imgf000124_0001
Scheme 12-12
Figure imgf000124_0002
12-9 12-18
Scheme 12-13
[0274] The acids of general formula 12-18 are readily converted to the corresponding acid chlorides 12-19 by the action of thionyl chloride or oxalyl chloride in the presence of catalytic dimethylformamide:
0 O
1 SOCI2 or (COCI)2 11 HO ^(CH2)mR4 — ► HO/^(CH2)mR4
DMF cat.
12-19 12-18
Scheme 12-14
[0275] A third aspect of the present invention provides a compound of Formula (I) or a solvate or physiologically functional derivative thereof for use as a tiierapeutic agent, specifically as a modifier ofthe activity of the serotonin 5-HT2A receptor. Modifiers ofthe activity ofthe serotonin 5- HT2A receptor are believed to be of potential use for the treatment or prophylaxis of CNS, gastrointestinal, cardiovascular, and inflammatory disorders. Compounds of die Formula (I) may be adiriinistered by oral, sublingual, parenteral, rectal, or topical administration. In addition to the neutral forms of compounds of Formula (I) by appropriate addition of an ionizable substituent, which does not alter the receptor specificity of the compound, physiologically acceptable salts of the compounds may also be formed and used as therapeutic agents. Different amounts of the compounds of Formula (I) will be required to achieve the desired biological effect. The amount will depend on factors such as the specific compound, the use for which it is intended, the means of adminisfration, and the condition of the treated individual. A typical dose may be expected to fall in the range of 0.001 to 200 mg per kilogram of body weight of die treated individual. Unit does may contain from 1 to 200 mg of the compounds of Formula (1) and may be administered one or more times a day, individually or in multiples. In tiie case of the salt or solvate of a compound of Formulas (I), the dose is based on the cation (for salts) or the unsolvated compound.
[0276] A fourth aspect of the present invention provides pharmaceutical compositions, comprising at least one compound of Formula (I) and/or an acceptable salt or solvate thereof (e.g., a pharmaceutically acceptable salt or solvate) as an active ingredient combined with at least one carrier or excipient (e.g., pharmaceutical carrier or excipient). Pharmaceutical compositions may be used in die treatment of clinical conditions for which a modifier of the activity of the serotonin 5-HT2A receptor is indicated, particularly where the active ingredient is preferentially selective for the 5-HT2A receptor over the 5-HT2A receptor, and most particularly where the active ingredient is also an inverse agonist at the 5-HT2A receptor. At least one compound of Formula (I) may be combined with the carrier in either solid or liquid form in a unit dose formulation. The pharmaceutical carrier must be compatible with the other ingredients in tiie composition and must be tolerated by the individual recipient. Other physiologically active ingredients may be incoφorated into the pharmaceutical composition of the invention if desired, and if such ingredients are compatible with the other ingredients in the composition. Formulations may be prepared by any suitable method, typically by uniformly mixing the active compound(s) with liquids or finely divided solid carriers, or both, in the required proportions, and then, if necessary, forming the resulting mixture into a desired shape.
[0277] Conventional excipients, such as binding agents, fillers, acceptable wetting agents, tabletting lubricants, and disintegrants may be used in tablets and capsules for oral administration. Liquid preparations for oral administration may be in the form of solutions, emulsions, aqueous or oily suspensions, and syrups. Alternatively, the oral preparations may be in the form of dry powder that can be reconstituted with water or another suitable liquid vehicle before use. Additional additives such as suspending or emulsifying agents, non-aqueous vehicles (including edible oils), preservatives, and flavorings and colorants may be added to the liquid preparations. Parenteral dosage forms may be prepared by dissolving the compound ofthe invention in a suitable liquid vehicle and filter sterilizing the solution before filling and sealing an appropriate vial or ampoule. These are just a few examples ofthe many appropriate methods well known in the art for preparing dosage forms.
[0278] The fifth aspect of the present invention provides for the use of a compound of formula (I) in the preparation of a medicament for the treatment of a medical condition for which a modifier ofthe activity ofthe serotonin 5-HT2A receptor is indicated.
[0279] The sixth aspect of the present invention provides for a method of treatment of a clinical condition of a mammal, such as a human, for which a modifier of the activity of the serotonin 5-HT2A receptor is indicated, which comprises the administration to the mammal of a therapeutically effective amount of a compound of Formula (I) or a physiologically acceptable salt, solvate, or physiologically functional derivative thereof.
EXPERIMENTAL DATA EXAMPLE 13
Preparation and Analysis of Compounds Experiments 13.1 - 13.43
[0280] Mass spectra were recorded on a Micromass Platform™ LC with Gilson HPLC.
Infra-red spectra were recorded on a Nicolet Avatar™ 360 FT-IR. Melting points were recorded on a Electrothermal IA9200™ apparatus and are uncorrected. Proton nuclear magnetic resonance spectra were recorded on a Bruker™ 300MHz machine. Chemical shifts are given with respect to teframethylsilane. In the text the following abbreviations are used; s (singlet), d (doublet), t (triplet), m (multiplet) or combinations thereof. Chemical shifts are quoted in parts per million (ppm) and with coupling constants in Hertz.
[0281] Thin layer chromatography was carried out using aluminium backed silica plates
(250μL; GF254). HPLC was recorded either on a HP Chemstation™ 1100 HPLC using a Hichrom 3.5 C18 reverse phase column (50mm x 2.1mm i.d.). Linear gradient elution over 5 minutes - 95% water (+0.1% TFA) / 5% acetonitrile (+0.05% TFA) down to 5% water / 95% acetonitrile. Flow rate 0.8mL/min (Method A); or on a Hichrom 3.5 C18 reverse phase column (100mm x 3.2mm i.d.). Linear gradient elution over 11 minutes - 95% water (+0.1% TFA) / 5% acetonitrile (+0.05% TFA) down to 5% water / 95% acetonitrile. Flow rate lmL/min (Method B). Samples were routinely monitored at 254nM unless otherwise stated.
[0282] All reagents were purchased from commercial sources. Experiment 13.1
Preparation and Analysis of Compound 1
N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)(4-methoxyphenoxy)carboxamide
[0283] To 4-methoxyphenylchloroformate (19 mg, 0.10 mmol) in CH2C12 (0.5 mL) was added dropwise a solution of 3-(3-anιinophenyl)-4-bromo-2-me1hylpyrazole (25 mg, 0.10 mmol) and friethylamine (14 μL, 0.10 mmol) in CH2C12 (0.5 mL). The mixture was stirred for 16 h and concentrated. Chromatography on flash silica (40% EtOAc/hexane) gave the title compound as a colourless solid (21 mg, 52%), m.p. 140.3-141.8oC. (EtOAc/hexane).
IR: vmax =1748, 1592, 1504, 1412, 1190, 835, 764, 676 cm."1 MS (ES+): m/z (%)=404 (M+H 81Br, 100), 402 (M+H 79Br, 90).
Η-NMR (CD3 OD): δ =3.80 (3H, s, CH3), 3.81 (3H, s, CH3), 6.91-6.98 (2H, m, ArH), 7.07-7.18 (3H, m, ArH), 7.42-7.53 (4H, m, ArH).
[0284] HPLC: retention time 3.28 mins (Method A). Tic: Rf 0.4 (EtOAc/hexane).
Experiment 13.2
Preparation and Analysis of Compound 2
N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)(2-thienyl)carboxamide
[0285] To thiophene-2-carbonyl chloride (11 μL, 0.09 mmol) in CH2C12 (1 mL) was added dropwise a solution of 3-(3-aminophenyl)-4-bromo-2-methylpyrazole (25 mg, 0.09 mmol) and friethylamine (14 μL, 0.09 mmol) in CH2C12 (0.5 mL). The reaction mixture was stirred for 16 h and concentrated. Chromatography on flash silica (50% EtOAc/hexane) gave the title compound as a colourless solid (24 mg, 68%), m.p. 127.8-128.6°C (EtOAc/hexane). :
MS (ES+): m/z (%)=364 (M+H 81Br, 96), 362 (M+H 79Br, 100).
'H-NMR (CD3 OD): δ =3.81 (3H, s, CH3), 7.19 (2H, m, ArH), 7.48-7.58 (2H, m, ArH), 7.68-7.83 (3H, m, ArH), 7.93 (IH, dd, J=1.0, 3.8, ArH).
[0286] HPLC: retention time 3.12 min (Method A). TLC: Rf 0.30 (30% EtOAc/hexane). Experiment 13.3
Preparation and Analysis of Compound 7
N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)(((4-frifluoromethoxy)phenyl) an ino)carboxamide
[0287] This compound is commercially available from Maybridge Chemical Company, Catalog No. KM04515, under the name N-(3-(4-bromo-l-methylpyrazol-3-yl)phenyl)(((4- trifluoromethoxy)phenyl) amino)carboxamide. :
Experiment 13.4
Preparation and Analysis of Compound 10 N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)(((4- frifluoromethoxy)phenyl)methyl)amino)carboxamide
[0288] To a stirred solution of friphosgene (12 mg, 0.04 mmol) in CH2C12 (0.5 mL) was added dropwise a solution of 3-(3-aminophenyl)-4-bromo-2-methylpyrazole (30 mg, 0.12 mmol) and friethylamine (33 μL, 0.24 mmol) in CH2C12 (0.5 mL). After 1 h, 4- (frifluoromethoxy)ben_ylamine (23 mg, 0.12 mmol) was added. The reaction mixture was stirred for 16 h and concenfrated. Chromatography on flash silica (75% EtOAc/hexane) gave the title compound as a colourless solid (38 mg, 68%), m.p. 144.6-145.8 °C (EtOAc/hexane).
IR: ■ vmax =1626, 1558, 1278, 1160, 969, 871, 789, 703 cm-1 MS (ES+): m/z (%)=471 (M+H 81Br, 91), 469 (M+H 79Br, 100). __-NMR (CD3 OD): δ =3.81 (3H, s, CH3), 4.42 (2H, s, CH2), 7.06 (IH, d, J=7.1 ,
ArH), 7.24 (2H, d, J=8.4, ArH), 7.37-7.52 (6H, m, ArH).
[0289] HPLC: retention time 3.06 mins (Method A). Tic: Rf 0.5 (EtOAc).
Experiment 13.5 Preparation and Analysis of Compound 39
((3 -(4-bromo-2-methylpyrazol-3 -yl)phenyl)amino)-N-(l , 1 -dimetiiylethoxy)carboxamide
[0290] To di-tert-butyl dicarbonate (36 mg, 0.17 mmol) in methanol (1 mL) was added dropwise a solution of 3-(3-aminophenyl)-4-bromo-2-methylpyrazole (42 mg, 0.17 mmol) in methanol (1 mL). The mixture was stirred for 16 h and concenfrated. Chromatography on flash silica (40% EtOAc/hexane) gave the title compound as a colourless solid (29 mg, 49%) (EtOAc/hexane).
MS (CI-): m/z (%)=352 (M-H 81Br, 100), 350 (M-H 79Br, 96). Η-NMR (DMSO d6): δ =1.46 (9H, s, 3.times. CH3), 3.73 (3H, s, CH3), 7.07 (IH, m, ArH), 7.42 (IH, t, J=7.7, ArH), 7.53-7.60 (2H, m, ArH), 7.64 (IH, s, ArH), 9.57 (IH, s, NH).
[0291] HPLC: retention time 7.15 min (Method B).
Experiment 13.6
Preparation and Analysis of Compound 40
N-(3-(4-bromo-2-metbylpyrazol-3-yl)phenyl)(4-frifluoromethoxyphenyl)carboxamide
[0292] To 4-(frifluoromethoxy)benzoyl chloride (19 μL, 0.12 mmol) in CH2C12 (1 mL) was added dropwise a solution of 3-(3-aminophenyl)-4-bromo-2-methylpyrazole (30 mg, 0.12 mmol) and friediylamine (17 μL, 0.12 mmol) in CH2C12 (0.5 mL). The reaction mixture was stirred for 16 h and concentrated. Chromatography on flash silica (50% EtOAc/hexane) gave the title compound as a colourless solid (40 mg, 76%), m.p. 138.6-139.6°C (EtOAc/hexane).
MS (ES+): m/z (%)=442 (M+H 81Br, 93), 440 (M+H 79Br, 100). Η-NMR (DMSO d_): δ=3.79 (3H, s, CH3), 7.27 (IH, m, ArH), 7.45-7.60 (3H, m, ArH), 7.65 (IH, s, ArH), 7.87 (2H, m, ArH), 8.09 (2H, m, ArH), 10.51 (IH, s, NH).
[0293] HPLC: retention time 3.60 min (Method A). TLC: Rf 0.40 (50% EtOAc/hexane).
Experiment 13.7
Preparation and Analysis of Compound 41 N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)(4-chloroρhenyl)carboxamide
[0294] To 4-chlorobenzoyl chloride (15 mg, 0.08 mmol) in CH2C12 (1 mL) was added dropwise a solution of 3-(3-aminophenyl)-4-bromo-2-methylpyrazole (21 mg, 0.08 mmol) and friethylamine (12 μL, 0.08 mmol) in CH2C12 (0.5 mL). The mixture was stirred for 16 h and concentrated. Chromatography on flash silica (50% EtOAc/hexane) gave the title compound as a colourless solid (23 mg, 72%), m.ρ. 184.4-184.8 °C (EtOAc/hexane).
MS (ES+): m/z (%)=394 (M+H 81Br 37C1, 34), 392 (M+H 79Br 37C1 (81Br 35C1), 100), 390 (M+H 79Br 35Cl, 67).
Η-NMR (DMSO ds): δ =3.79 (3H, s, CH3), 7.25 (IH, d, J=7.9, ArH), 7.51-7.6 (3H, m, ArH), 7.69 (IH, s, ArH), 7.90 (2H, m, ArH), 8.00 (2H, m, ArH), 10.51 (IH, s, NH).
[0295] HPLC: retention time 3.40 min (Method A). TLC: Rf 0.35 (50% EtOAc/hexane). Experiment 13.8
Preparation and Analysis of Compound 42
N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)-2-(4-(frifluorometiιoxy)phenyl)acetamide
[0296] A solution of 3-(3-aminophenyl)-4-bromo-2-methylpyrazole(35 mg, 0.14 mmol) and friethylamine (23 μL, 0.17 mmol) in DMF (0.5 mL) was added in one portion to a stirred solution of 4-trifluoromethoxyphenylacetic acid (31 mg, 0.14 mmol), HBTU (53 mg, 0.14 mmol) and HOBT (19 mg, 0.14 mmol) in DMF (1 mL). The mixture was heated at 70 °C for 24 h and then quenched with aqueous sodium bicarbonate solution. Ethyl acetate was added and the organic phase separated, washed with water (.times.3), brine, dried (MgS04) and evaporated.
[0297] Chromatography on flash silica (50% EtOAc/hexane) gave the title compound as a colourless solid (43 mg, 68%). m.p. 141.2-142.5 oC (EtOAc/hexane).
IR: vraax =1684, 1592, 1510, 1253, 1217, 1157, 987, 798, 700 cm"1.
MS (ES+): m/z (%)=456 (M+H 81Br, 100), 454 (M+H 79Br, 94).
Η-NMR (DMSO d6): δ =3.72 (2H, s, CH2), 3.75 (3H, s, CH3), 7.17 (IH, d, J=7.7, ArH), 7.33 (2H, d, J=8.7, ArH), 7.38-7.51 (3H, m, ArH), 7.62-7.73 (3H, m, ArH), 10.44 (IH, s, NH).
[0298] HPLC: retention time 3.52 min (Metliod A).
Experiment 13.9
Preparation and Analysis of Compound 43 N-(3 -(4-bromo-2-methylpyrazol-3 -yl)phenyl)-2-(3 -fluorophenyl)acetamide
[0299] A mixture of 3-(3-arninophenyl)-4-bromo-2-methylpyrazole (30 mg, 0.12 mmol), 3- fluorophenylacetic acid (18 mg, 0.12 mmol), 1-hydroxybenzotriazole hydrate (16 mg, 0.12 mmol) and 2-(lH-benzotriazole-l-yl)-l,l,3,3-teframetiιyluronium hexafluoro-phosphate (46 mg, 0.12 mmol) were dissolved in chloroform (1.5 ml). N,N-Diisopropylethylamine (0.02 ml, 0.13 mmol) was added and the mixture stirred at room temperature for 16 h. The reaction mixture was then poured into brine and the organic layer washed with further brine, dried over magnesium sulphate and then concentrated in vacuo. The crude product was purified by column chromatography (ethyl acetate-toluene, 1:1), giving the title compound (12 mg, 26%). Rf 0.41 (ethyl acetate-toluene, 1:1).
MS (AP+): m/z (%)=390 (M-H 81Br 100), 388 (M-H 79Br, 100). Η-NMR (CDC13): δ = 3.77 (2H, s), 3.83 (3H, s), 7.02-7.20 (4H, m), 7.54 (IH, s),
7.60-7.63 (IH, m).
[0300] HPLC (Method B): retention time 7.07 min (100%). Experiment 13.10
Preparation and Analysis of Compound 44
N-(3-(4-bromo-2-ιnethylpyrazol-3-yl)phenyl)-2-(3-methoxyphenyl)acetamide
[0301] A solution of 3-methoxyphenylacetyl chloride (0.02 ml, 0.12 mmol) in dichloromethane (0.75ml) was added dropwise at 0 °C to a solution of 3-(3-aminophenyl)-4-bromo-2- methylpyrazole (30 mg, 0.12 mmol) and friethylamine (0.02 ml, 0.13 mmol) in dichloromethane (0.75 ml). The resulting mixture was stirred at room temperature for 16 h and then poured into brine. The organic layer was washed with more brine then dried over magnesium sulphate and concenfrated in vacuo. The crude product was purified by column chromatography (ethyl acetate-toluene, 1:1), giving the title compound (9 mg, 19%). Rf 0.30 (ethyl acetate-toluene, 1:1).
MS (AP+): m/z (%)=402 (M+H 81Br, 100), 400 (M+H 79Br, 95). •H-NMR (CDC13): δ = 3.76 (2H, s), 3.82 (3H, s), 3.85 (3H, s), 6.84-6.90 (3H, m), 7.07-7.44 (5H, m), 7.53 (IH, s), 7.60 (IH, br s).
[0302] HPLC (Method B): retention time 8.62 min (97.09%). δH (CDC13)
Experiment 13.11
Preparation and Analysis of Compound 45
N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)-2-(2-fluorophenyl)acetamide
[0303] A mixture of 3-(3-arninophenyl)-4-bromo-2-methylρyrazole (30 mg, 0.12 mmol), 2- fluorophenylacetic acid (18 mg, 0.12 mmol), 1-hydroxybenzotriazole hydrate (16 mg, 0.12 mmol) and 2-(lH-benzofriazole-l-yl)-l,l,3,3-teframethyluronium hexafluoro-phosphate (46 mg, 0.12 mmol) were dissolved in chloroform (1.5 ml). N,N-Diisopropylethylamine (0.02 ml, 0.13mmol) was added and die mixture stirred at room temperature for 16 h. The reaction mixture was then poured into brine and the organic layer washed witii further brine, dried over magnesium sulphate and then concenfrated in vacuo. The crude product was purified by column chromatography (ethyl acetate-toluene, 1:1), giving the title compound (15 mg, 32%). Rf 0.52 (ethyl acetate-toluene, 1:1).
MS (AP+): m/z (%)=390 (M+H 81Br, 100), 388 (M+H 79Br, 100).
Η-NMR (CDC13): δ = 3.79 (2H, s), 3.83 (3H, s), 7.11-7.23 (3H, m), 7.30-7.55 (6H, m), 7.61-7.64 (lH, m).
[0304] HPLC (Metiiod B): retention time 7.28 min (100%). Experiment 13.12
Preparation and Analysis of Compound 46
N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)-2-(4-nifrophenyl)acetamide
[0305] A mixture of 3-(3-aminophenyl)-4-bromo-2-methylpyrazole(30 mg, 0.12 mmol), 4- nifrophenylacetic acid (22 mg, 0.12 mmol), 1-hydroxybenzotriazole hydrate (16 mg, 0.12 mmol) and 2-(lH-benzotriazole-l-yl)-l,l,3,3-teframethyluronium hexafluorophosphate (46 mg, 0.12 mmol) were dissolved in chloroform (1.5 ml). N,N-Diisopropylethylamine (0.02 ml, 0.13 mmol) was added and the mixture stirred at room temperature for 16 h. The reaction mixture was then poured into brine and the organic layer washed with further brine, dried over magnesium sulphate and then concentrated in vacuo. The crude product was purified by column chromatography (ethyl acetate-toluene, 1:1), giving the title compound (9 mg, 18%). Rf 0.19 (ethyl acetate-toluene, 1:1).
MS (AP+): m/z (%)=417 (M+H 81Br, 100), 415 (M+H 79Br, 100). Η-NMR (CDC13): δ = 3.83 (3H, s), 3.87 (2H, s), 7.18-7.23 (IH, m), 7.42-7.65 (7H, m), 8.22-8.30 (2H, m).
[0306] HPLC (Method B): retention time 7.22 min (94.30%).
Experiment 13.13
Preparation and Analysis of Compound 47
N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)-2-(2-metiioxyphenyl)acetamide
[0307] A mixture of 3-(3-arninophenyl)-4-bromo-2-methylpyrazole(30 mg, 0.12 mmol), 2- methoxyphenylacetic acid (20 mg, 0.12 mmol), 1-hydroxybenzotriazole hydrate (16 mg, 0.12 mmol) and 2-(lH-benzofriazole-l-yl)-l,l,3,3-tetramethyluronium hexafluorophosphate (46 mg, 0.12 mmol) were dissolved in chloroform (1.5 ml). N,N-Diisopropylethylamine (0.02 ml, 0.13 mmol) was added and the mixture stirred at room temperature for 16 h. The reaction mixture was then poured into brine and the organic layer washed with further brine, dried over magnesium sulphate and tiien concentrated in vacuo. The crade product was purified by column chromatography (chloroform-methanol, 99:1), giving the title compound (18 mg, 38%) as a colourless solid. Rf 0.65 (chloroform-metiianol, 98:2). MS (AP-): m/z (%)=400 (M-H 81Br, 90), 398 (M-H 79Br, 100). Η-NMR(CDC13) δ=3.76 (2H, s), 3.83 (3H, s), 3.98 (3H, s), 6.97-7.06 (2H, m), 7.11- 7.16 (IH, m), 7.31-7.50 (4H, m), 7.53 (IH, s), 7.57-7.60 (IH, m), 7.91 (IH, br s).
[0308] HPLC (Method B): retention time 7.16 min (100%). [0309] One or the other (as indicated) of the two following synthetic protocols was used to generate each ofthe compounds below:
Protocol A:
[0310] To an isocyanate (1 mmol) in CH2C12 (4 mL) was added dropwise a solution of 3-(3- aminophenyl)-4-bromo-2-methylpyrazole (1 mmol) in CH2C12 (4 mL). The mixture was stirred for 16 hours and concenfrated. Chromatography on flash silica (20%-80% EtOAc/hexane) followed by recrystallisation gave the pure urea.
Protocol B:
[0311] To a stirred solution of friphosgene (0.33 mmol) in CH2C12 (4 mL) was added dropwise a solution of 3-(3-arninophenyl)-4-bromo-2-methylpyrazole(l mmol) and triethylamine (2 mmol) in CH2C12 (4 mL). After 1 hour, an aniline was added (1 mmol). The reaction mixture was stirred for 16 hours and concentrated. Chromatography on flash silica (20%-80% EtOAc/hexane) followed by recrystallisation gave die pure urea. :
Experiment 13.14
Preparation and Analysis of Compound 11
N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)((4-memylthiophenyl)andno)carboxamide
[0312] (Protocol A) - 4-(methylthio)phenyl isocyanate; colorless solid (EtOAc/hexane).
MS (ES+): m/z (%) = 419 (M+H 81Br, 100), 417 (M+H 79Br, 94). !H-NMR (MeOH d4): d = 2.42 (3H, s, SCH3), 3.81 (3H, s, NCH3), 7.06 (IH, m, ArH), 7.22 (2H, m, ArH), 7.37 (2H, m, ArH), 7.42-7.61 (4H, m, ArH).
[0313] HPLC: retention time 3.35 min (Method A).
Experiment 13.15
Preparation and Analysis of Compound 8
N-(3-(4-bromo-2-memylpyrazol-3-yl)phenyl)((4-chlorophenyl)anιmo)carboxamide
[0314] (Protocol A) - 4-chlorophenyl isocyanate; colorless solid (EtOAc/hexane). MS (ES+): m/z (%) = 409 (M+H 81Br 37C1, 19), 407 (M+H 79Br 37C1 (81Br 35C1), 100), 405 (M+H 79Br 35Cl, 81).
Η-NMR (MeOH d4): d = 3.81 (3H, s, CH3), 7.07 (1H, m, ArH), 7.23 (2H, m, ArH), 7.36-7.60 (6H, m, ArH).
[0315] HPLC: retention time 3.42 min (Method A).
Experiment 13.16
Preparation and Analysis of Compound 9
((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)an mo)-N-(4-fluorophenyl)carboxarnide
[0316] (Protocol A) - 4-fluorophenyl isocyanate; colorless solid (EtOAc/hexane).
MS (ES+): m/z (%) = 391 (M+H 81Br, 96), 389 (M+H 79Br, 100). 'H-NMR (MeOH d4): δ = 3.81 (3H, s, CH3), 6.93-7.11 (3H, m, ArH), 7.37-7.61 (6H, m, ArH).
[0317] HPLC: retention time 3.11 min.
Experiment 13.17
Preparation and Analysis of Compound 12
((3-(4-bromo-2-methylpyτa_ol-3-yl)phenyl)antino)-N-(2-(frifluorome1boxy)phenyl)carboxamide
[0318] (Protocol A) - 2-(frifluoromethoxy)phenyl isocyanate; colorless solid (EtOAc/hexane).
MS (ES+): m/z (%) = 457 (M+H 81Br, 100), 455 (M+H 79Br, 95).
Η-NMR (DMSO d6): δ = 3.79 (3H, s, CH3), 7.06-7.18 (2H, m, ArH), 7.38-7.49 (2H, m, ArH), 7.51-7.62 (2H, m, ArH), 7.65 (IH, m, ArH), 7.71 (IH, s, ArH), 8.24 (IH, dd, J=l.l, 8.2, ArH), 8.56 (IH, s, NH), 9.49 (IH, s, NH).
[0319] HPLC: retention time 3.40 min.
Experiment 13.18
Preparation and Analysis of Compound 13 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)an_no)-N-(2-nifrophenyl)carboxamide
[0320] (Protocol A) - 2-nifrophenyl isocyanate; yellow solid (EtOAc/hexane).
MS (ES+): m/z (%) = 418 (M+H 81Br, 98), 416 (M+H 79Br, 100). !H-NMR (DMSO d6): δ = 3.79 (3H, s, NCH3), 7.14 (IH, m, ArH), 7.24 (IH, m, ArH), 7.50 (IH, t, J=7.7, ArH), 7.60 (2H, m, ArH), 7.67 (IH, s, ArH), 7.71 (IH, s, ArH), 8.10 (IH, m, ArH), 8.29 (IH, m, ArH), 9.65 (IH, s, NH), 10.09 (IH, s, NH).
[0321] HPLC: retention time 3.10 min (Method A).
Experiment 13.19
Preparation and Analysis of Compound 14 ((3-(4-bromo-2-me1hylpyrazol-3-yl)phenyl)amino)-N-(4-methoxyphenyl)carboxaιnide
[0322] (Protocol A) - 4-methoxyphenyl isocyanate; colorless solid (EtOAc/hexane).
MS (ES+): m/z (%) = 403 (M+H 81Br, 100), 401 (M+H 79Br, 96). •H-NMR (DMSO d6): δ = 3.71 (3H, s, OCH3), 3.79 (3H, s, NCH3), 6.87 (2H, d, J=8.9, ArH), 7.06 (IH, d, J=7.5, ArH), 7.39 (2H, d, J=8.9, ArH), 7.45-7.61 (3H, m, ArH),
7.65 (IH, s, ArH), 8.52 (IH, s, NH), 8.84 (IH, s, NH).
[0323] HPLC: retention time 3.08 min.
Experiment 13.20 Preparation and Analysis of Compound 15
((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)anιino)-N-(2-methylphenyl)carboxamide
[0324] (Protocol A) - o-tolyl isocyanate; colorless solid (EtOAc/hexane).
MS (ES+): m/z (%) = 387 (M+H 81Br, 94), 385 (M+H 79Br, 100). !H-NMR (MeOH d,): δ = 2.29 (3H, s, CH3), 3.81 (3H, s, NCH3), 7.03 (IH, dt,
J=l.l,7.5, ArH), 7.09 (IH, dt, J=l.l, 7.5, ArH), 7.13-7.22 (2H, m, ArH), 7.45 (IH, t, J=7.9, ArH), 7.49-7.57 (2H, m, ArH), 7.60-7.68 (2H, m, ArH).
[0325] HPLC: retention tune 2.96 min. Experiment 13.21
Preparation and Analysis of Compound 16
((3-(4-bromo-2-methylpyr_?ol-3-yl)phenyl)amino)-N-(4-(1rifluoromethyl)phenyl)carboxarnide
[0326] (Protocol A) - 4-(trifluoromethyl)phenyl isocyanate; colorless solid (EtOAc/hexane).
MS (ES+): m/z (%) = 441 (M+H 81Br, 94), 439 (M+H 79Br, 100). Η-NMR (MeOH d4): δ = 3.82 (3H, s, CH3), 7.04-7.16 (3H, m, ArH), 7.20-7.47 (6H, m, ArH).
[0327] HPLC: retention time 3.56 min. :
Experiment 13.22
Preparation and Analysis of Compound 17
((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)ammo)-N-(3-chlorophenyl)carboxamide
[0328] (Protocol A) - 3-chlorophenyl isocyanate; colorless solid (EtOAc/hexane).
MS (ES+): m/z (%) = 409 (M+H 81Br 37C1, 26), 407 (M+H 79Br 37C1 (81Br 35C1), 100), 405 (M+H 79Br 35Cl, 70).
Η-NMR (MeOH d4): δ = 3.81 (3H, s, NCH3), 7.04 (IH, m, ArH), 7.10 (IH, m, ArH), 7.28 (2H, m, ArH), 7.47 (IH, t, J=7.8, ArH), 7.55 (IH, m, ArH), 7.63 (IH, m, ArH), 7.68 (IH, s, ArH), 7.73 (IH, m, ArH), 9.04 (2H, s, NH).
[0329] HPLC: retention time 3.20 min (Metliod A).
Experiment 13.23
Preparation and Analysis of Compound 18 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)an_mo)-N-(2-chlorophenyl)carboxamide
[0330] (Protocol A) - 2-chlorophenyl isocyanate; colorless solid (EtOAc/hexane).
MS (ES+): m/z (%) = 409 (M+H 81Br 37C1, 24), 407 (M+H 79Br 37C1 (81Br 35C1), 100), 405 (M+H 79Br 35Cl, 72). Η-NMR (MeOH d4): δ = 3.81 (3H, s, NCH3), 7.03 (IH, m, ArH), 7.11 (IH, m, ArH),
7.28 (IH, m, ArH), 7.35-7.53 (3H, m, ArH), 7.55 (IH, s, ArH), 7.62 (IH, m, ArH), 8.11 (IH, m, ArH). [0331] HPLC: retention time 3.13 min.
Experiment 13.24
Preparation and Analysis of Compound 19 ((3 -(4-bromo-2-methylpyrazol-3 -yl)phenyl)arnmo)-N-(4-(methylethyl)phenyl)carboxamide
[0332] (Protocol A) - 4-isopropylphenyl isocyanate; colorless solid (THF/hexane).
MS (ES+): m/z (%) = 415 (M+H 81Br, 100), 413 (M+H 79Br, 92). Η-NMR (MeOH d4): δ = 1.23 (6H, d, J=6.8, 2xCH3), 2.86 (IH, septet, J=6.8, CH), 3.82 (3H, s, NCH3), 7.09 (IH, m, ArH), 7.16 (2H, d, J=7.6, ArH), 7.31 (2H, d, J=7.6, ArH),
7.42-7.51 (2H, m, ArH), 7.54 (IH, s, ArH), 7.59 (IH, m, ArH).
[0333] HPLC: retention time 3.66 min.
Experiment 13.25 Preparation and Analysis of Compound 20
((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(3-methoxyphenyl)carboxamide
[0334] (Protocol A) — 3-methoxyphenyl isocyanate; colorless solid (EtOAc/hexane).
MS (ES+): m/z (%) = 403 (M+H 81Br, 100), 401 (M+H 79Br, 96). Η-NMR (MeOH d4): δ = 3.73 (3H, s, OCH3), 3.81 (3H, s, NCH3), 6.59 (IH, m,
ArH), 6.91 (IH, m, ArH), 7.08 (IH, m, ArH), 7.14 (2H, m, ArH), 7.39-7.61 (4H, m, ArH).
[0335] HPLC: retention time 2.90 min.
Experiment 13.26 Preparation and Analysis of Compound 21
((3 -(4-bromo-2-methylpyrazol-3 -yl)phenyl)amino)-N-(3 -methylphenyl)carboxamide
[0336] (Protocol A) - m-tolyl isocyanate; : colorless solid (EtOAc/hexane).
MS (ES+): m/z (%) = 387 (M+H 81Br, 100), 385 (M+H 79Br, 96). Η-NMR (DMSO d6): δ = 2.26 (3H, s, CH3), 3.76 (3H, s, NCH3), 6.79 (IH, m, ArH),
7.06-7.22 (3H, m, ArH), 7.29 (IH, m, ArH), 7.43-7.62 (3H, m, ArH), 7.68 (IH, s, ArH), 8.65 (1H, S, NH), 8.89 (1H, S, NH). [0337] HPLC: retention time 3.05 min (Method A).
Experiment 13.27
Preparation and Analysis of Compound 22 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-methyl-N-(4-
(frifluoromethoxy)phenyl)-carboxamide
[0338] (Protocol B) - N-methyl-4-(trifluoromethoxy)aniline; pale yellow solid (EtOAc/hexane).
MS (ES+): m/z (%) = 471 (M+H 81Br, 88), 469 (M+H 79Br, 100). Η-NMR (MeOH d4): δ = 3.35 (3H, s, NCH3), 3.81 (3H, s, NCH3), 7.09 (IH, m,
ArH), 7.25-7.51 (8H, m, ArH).
[0339] HPLC: retention time 3.56 min (Metiiod A).
Experiment 13.28 Preparation and Analysis of Compound 23
N-(4-(tert-butyl)phenyl)((3-(4-bromo-2-memylpyrazol-3-yl)phenyl)amiiio)carboxamide
(Protocol B) - 4-tert-butylaniline;
[0340] colorless solid (EtOAc/hexane).
MS (ES+): m/z (%) = 429 (M+H 81Br, 98), 427 (M+H 79Br, 100). 'H-NMR (DMSO d6): δ = 1.27 (9H, s, 3xCH3), 3.79 (3H, s, NCH3), 7.07 (IH, d,
J=7.5, ArH), 7.29 (2H, d, J=8.7, ArH), 7.37 (2H, d, J=8.7, ArH), 7.45 (IH, t, J=7.5, ArH), 7.51-7.60 (2H, m, ArH), 7.66 (IH, s, ArH), 8.65 (IH, s, NH), 8.83 (IH, s, NH).
[0341] HPLC: retention time 3.77 min.
Experiment 13.29
Preparation and Analysis of Compound 24
N-(4-(dimethylammo)phenyl)((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)ammo)carboxarnide (Protocol B) - N^V-dimethyl-/>-phenylenediamine;
[0342] colorless solid (EtOAc/hexane). MS (ES+): m/z (%) = 416 (M+H 81Br, 96), 414 (M+H 79Br, 100). Η-NMR (DMSO ds): δ = 2.86 (6H, s, NCH3), 3.80 (3H, s, NCH3), 6.80 (2H, m, ArH), 7.09 (IH, d, J=7.7, ArH), 7.28 (2H, m, ArH), 7.42 (IH, t, J=7.8, ArH), 7.52 (IH, m, ArH), 7.59 (IH, s, ArH), 7.67 (IH, s, ArH), 8.45 (IH, s, NH), 8.75 (IH, s, NH).
[0343] HPLC: retention time 2.07 min (Method A).
Experiment 13.30
Preparation and Analysis of Compound 25
N-(3,5-dichloro-4-methylphenyl)((3-(4-bromo-2-methylρyrazol-3-yl)phenyl)amino) carboxamide (Protocol B) - 3,5-dichloro-4-methylphenylamine;
[0344] colorless solid (EtOAc/hexane).
MS (ES+): m/z (%) = 457 (M+H, 35), 455 (M+H, 100), 453 (M+H, 65).
Η-NMR (DMSO d6): δ = 2.32 (3H, s, CH3), 3.79 (3H, s, NCH3), 7.11 (IH, d, J=7.4, ArH), 7.46 (IH, t, J=7.8, ArH), 7.50-7.64 (4H, m, ArH), 7.68 (IH, s, ArH), 9.02 (IH, s, NH), 9.09 (lH, s, NH).
[0345] HPLC: retention time 3.66 min.
Experiment 13.31
Preparation and Analysis of Compound 26
((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amiiio)-N-(4-(frifluorometiiylthio)phenyl) carboxamide (Protocol B) - 4-(frifluoromethylthio)aniline;
[0346] colorless solid (EtOAc/hexane).
MS (ES+): m/z (%) = 473 (M+H 81Br, 100), 471 (M+H 79Br, 94). Η-NMR (DMSO d6): δ = 3.81 (3H, s, NCH3), 7.11 (IH, d, J=7.5, ArH), 7.47 (IH, t, J=7.9, ArH), 7.51-7.63 (6H, m, ArH), 7.66 (IH, s, ArH), 9.03 (IH, s, NH), 9.16 (IH, s, NH). HPLC: retention time 3.76 min.
Experiment 13.32
Preparation and Analysis of Compound 31
((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(cyclohexyl)carboxarnide
[0347] (Protocol B) - cyclohexylamine; : colorless solid, m.p. 155.5-156.3°C (EtOAc/hexane).
MS (ES+): m/z (%) = 379 (M+H 81Br, 93), 377 (M+H 79Br, 100). 'H-NMR (DMSO d6): δ = 1.07-1.34 (5H, m, 5xCH), 1.52 (IH, m, CH), 1.63 (2H, m, 2xCH), 1.76 (2H, m, 2xCH), 3.48 (IH, m, NCH), 3.74 (3H, s, CH3), 6.15 (IH, d, J=7.8, ArH), 6.98 (IH, d, J=7.5, ArH), 7.32-7.43 (2H, m, ArH), 7.51 (IH, m, NH), 7.62 (IH, s, ArH), 8.50 (lH, s, NH).
[0348] HPLC: retention time 3.16 min (Method A). :
TLC: retention factor 0.35 (50% EtOAc/hexane).
Experiment 13.33
Preparation and Analysis of Compound 32 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)aιnmo)-N-φhenylmethyl)carboxamide
[0349] (Protocol B) - benzylamine; colorless solid, m.p. 144.5-146.2°C (EtOAc/hexane). IR: max = 1622, 1565, 1467, 1374, 1239, 973, 802, 752, 695 cm"1. MS (ES+): m/z (%) = 387 (M+H 81Br, 89), 385 (M+H 79Br, 100). 1H-NMR (CD3OD): δ = 3.81 (3H, s, CH3), 4.40 (2H, s, CH2), 7.05 (IH, m, ArH),
7.19-7.51 (9H, m, ArH).
[0350] HPLC: retention time 3.06 min (Method A).
Experiment 13.34 Preparation and Analysis of Compound 27
((3-(4-bromo-2-memylpyrazol-3-yl)phenyl)arnmo)-N-(2-fluorophenyl)carboxamide
[0351] (Protocol A) - 2-fluorophenyl isocyanate; colorless solid (DCM/hexane).
MS (ES+): m/z (%) = 391 (M+H 81Br, 100), 389 (M+H 79Br, 90). Η-NMR (MeOH d4): δ = 3.79 (3H, s, NCH3), 7.00-7.11 (4H, m, ArH), 7.40-7.56
(3H, m, ArH), 7.61 (IH, m, ArH), 8.09 (IH, m, ArH).
[0352] HPLC: retention time 3.01 min.
Experiment 13.35 Preparation and Analysis of Compound 28
2-(((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)-amino)carbonylan_no)benzamide [0353] (Protocol B) - 2-aminobenzarnide; colorless solid (EtOAc/hexane).
MS (ES+): m/z (%) = 399 (M+H -17 81Br, 100), 397 (M+H - 17 79Br, 94). Η-NMR (DMSO d6): δ = 3.79 (3H, s, NCH3), 6.93-7.10 (2H, m, ArH), 7.45 (2H, t, J=7.8, ArH), 7.59-7.72 (5H, m, ArH), 8.22 (2H, m), 9.92 (IH, s, NH), 10.69 (IH, s, NH).
[0354] HPLC: retention time 2.88 min.
Experiment 13.36
Preparation and Analysis of Compound 29 ((3 -(4-bromo-2-methylpyrazol-3 -yl)phenyl)amino)-N-(4-cyanophenyl)carboxamide
[0355] (Protocol B) - 4-arninobenzonitrile; colorless solid (EtOAc/hexane).
MS (ES+): m/z (%) = 398 (M+H 81Br, 100), 396 (M+H 9Br, 96). !H-NMR (MeOH d4): δ = 3.81 (3H, s, NCH3), 7.12 (IH, m, ArH), 7.46-7.57 (3H, m, ArH), 7.62-7.69 (5H, m, ArH).
[0356] HPLC: retention time 3.12 min.
Experiment 13.37
Preparation and Analysis of Compound 30 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)armno)-N-(2-cyanophenyl)carboxamide
[0357] (Protocol B) - 2-aminobenzonitrile; colorless solid (EtOAc/hexane).
MS (ES+): m/z (%) = 398 (M+H 81Br, 95), 396 (M+H 79Br, 100). :H-NMR (CDC13): δ = 3.79 (3H, s, CH3), 7.13-7.28 (2H, m, ArH), 7.49 (IH, t, J=7.8, ArH), 7.57 (IH, m, ArH), 7.62 (IH, m, ArH), 7.65-7.71 (2H, m, ArH), 7.78 (IH, m, ArH),
8.07 (IH, d, J=8.6, ArH), 8.83 (IH, s, NH), 9.62 (IH, s, NH).
[0358] HPLC: retention time 3.05 min (Method A).
Experiment 13.38 Preparation and Analysis of Compound 33
((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)anι o)-N-(4-fluorophenylmethyl)carboxamide [0359] (Protocol B) - 4-fluorobenzylamine; colorless solid, m.p. 185.5-186.6°C (EtOAc/hexane). MS (ES+): m/z (%) = 405 (M+H 81Br, 97), 403 (M+H 79Br, 100). 'H-NMR (DMSO d6): δ = 3.75 (3H, s, CH3), 4.28 (2H, d, J=6.0, CH2), 6.73 (IH, t, J^'9, NH), 7.01 (IH, d, J=7.5, ArH), 7.10-7.18 (2H, m, ArH), 7.27-7.41 (4H, m, ArH), 7.56
(IH, s, ArH), 7.62 (IH, s, ArH), 8.82 (IH, s, NH).
[0360] HPLC: retention time 3.10 min (Method A).
[0361] TLC: retention factor 0.25 (50% EtOAc hexane).
Experiment 13.39
Preparation and Analysis of Compound 34
((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(3,4- dimethoxyphenylmethyl)carboxamide
[0362] (Protocol B) - 3 ,4-dimethoxybenzylamine: colorless solid, m.p. 174.9-175.5°C (EtOAc/hexane).
MS (CI+): m/z (%) = 447 (M+H 81Br, 100), 445 (M+H 79Br, 92). 'H-NMR (DMSO d6): δ = 3.71 (3H, s, CH3), 3.73 (3H, s, CH3), 3.76 (3H, s, CH3), 4.22 (2H, d, J=5.8, CH2), 6.62 (IH, t, J=5.7, NH), 6.80 (IH, m, ArH), 6.89 (2H, m, ArH), 6.98 (IH, m, ArH), 7.36-7.51 (3H, m, ArH), 7.63 (IH, s, ArH), 8.76 (IH, s, NH).
[0363] HPLC: retention time 2.86 min (Method A).
[0364] TLC: retention factor 0.20 (50% EtOAc/hexane).
Experiment 13.40
Preparation and Analysis of Compound 35 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)arnino)-N-(3,4,5- tιimetiιoxyphenylme1hyl)carboxamide
[0365] (Protocol B) - 3,4,5-trimethoxybenzylamine; colorless solid (EtOAc/hexane). MS (CI+): m/z (%) = 477 (M+H 81Br, 100), 475 (M+H 79Br, 95). Η-NMR (DMSO d6): δ = 3.63 (3H, s, OCH3), 3.75 (9H, s, 3xCH3), 4.21 (IH, d, J=5.9, CH2), 6.61 (2H, s, ArH), 6.65 (IH, t, J=5.9, NH), 6.99 (IH, m, ArH), 7.40 (IH, t, J=7.7, ArH), 7.45 (IH, m, ArH), 7.56 (IH, m, ArH), 7.64 (IH, s, ArH), 8.77 (IH, s, NH).
[0366] HPLC: retention time 5.91 min (Method B).
[0367] TLC: retention factor 0.50 (50% EtOAc/hexane).
Experiment 13.41
Preparation and Analysis of Compound 36
N-(3-(4-bromo-2-methylρyrazol-3-yl)phenyl)-(((2-methylphenyl)methyl)aιrrino)carboxarnide
[0368] (Protocol B) - 2-metiιylbenzylamine; colorless solid (EtOAc/hexane).
MS (CI+): m/z (%) = 401 (M+H 81Br, 96), 399 (M+H 79Br, 100).
'H-NMR (DMSO d6): δ = 2.28 (3H, s, CH3), 3.76 (3H, s, NCH3), 4.28 (IH, d, J=5.8, CH2), 6.60 (IH, t, J=5.8, NH), 7.01 (IH, m, ArH), 7.15 (3H, m, ArH), 7.24 (IH, m, ArH), 7.38-7.50 (2H, m, ArH), 7.57 (IH, m, ArH), 7.65 (IH, s, ArH), 8.77 (IH, s, NH).
[0369] HPLC: retention time 2.74 min (Method A).
[0370] TLC: retention factor 0.20 (50% EtOAc/hexane).
Experiment 13.42 Preparation and Analysis of Compound 37
((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)aιmno
[0371] (Protocol B) - 4-methoxybenzylamine; colorless solid (EtOAc/hexane).
MS (CI+): m/z (%) = 417 (M+H 81Br, 94), 415 (M+H 79Br, 100). 'H-NMR (DMSO d6): δ = 3.72 (3H, s, CH3), 3.77 (3H, s, NCH3), 4.22 (IH, d, J=5.9,
CH2), 6.62 (IH, t, J=5.9, NH), 6.90 (2H, d, J=8.8, ArH), 7.00 (IH, m, ArH), 7.23 (2H, d, J=8.8, ArH), 7.39 (IH, t, J=7.8, ArH), 7.43 (IH, m, ArH), 7.56 (IH, m, ArH), 7.64 (IH, s, ArH), 8.73 (lH, s, NH).
[0372] HPLC: retention time 6.41 min (Method B).
[0373] TLC: retention factor 0.25 (50% EtOAc/hexane). Experiment 13.43
Preparation and Analysis of Compound 38
((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)ammo)-N-(2-(4-methoxy)phenylethyl)carboxarnide
[0374] (Protocol B) - 2-(4-methoxyphenyl)ethylamine; colorless solid (EtOAc/hexane).
MS (ES+): m/z (%) = 431 (M+H 81Br, 95), 429 (M+H 79Br, 100). Η-NMR (DMSO d6): δ = 2.68 (2H, t, J=7.1, CH2), 3.31 (2H, m, CH2), 3.71 (3H, s, CH3), 3.77 (3H, s, CH3), 6.16 (IH, t, J=5.8, NH), 6.87 (2H, d, J=8.6, ArH), 6.99 (IH, dt, J=1.4, 7.3, ArH), 7.16 (2H, d, J=8.6, ArH), 7.33-7.48 (2H, m, ArH), 7.52 (IH, m, ArH), 7.63
(IH, s, ArH), 8.71 (IH, s,NH).
[0375] HPLC: retention time 6.62 min (Method B).
EXAMPLE 14
Experiments 14.1 - 14.19
General Synthetic Strategies, Preparation and Analysis of Compounds
[0376] Compounds of certain embodiments of the invention were prepared according to the procedures below.
General Synthetic Strategies
[0377] Tributyltin pyrazole 14-9 was synthesized from pyrazole 14-12 in 80 % chemical yield. Pyrazole 14-12 was directly lithiated with LDA at -78 °C followed by quenching with Bu3SnCl. Tributyltin pyrazole 14-9 is fairly stable on silica gel and can be stored at room temperature without any decomposition for up to one month in the air. Zinc pyraole 14-10 was also formed under lithiation with LDA or /z-BuLi at -78 °C followed by quenching with ZnCl2 in high yields. Zinc pyraole 14-10 was freshly synthesized for coupling reactions (Scheme 14-1).
Figure imgf000145_0001
14-10 Scheme 14-1
[0378] 1,3-Dinifrobenzene 14-13 was commercially available and iodinated with I2 and
H2S04 (oleum) to afford iodobenzene 14-14 in 68 % yield. Iodobenzene 14-14 was coupled with fributyltin pyrazole 14-9 under 7 % PdCl2(PPh3)2 to afford coupled product 14-15 in 85 % yield which could also be separable by recrystallization in ethanol and synthesize in quantity (i.e., 20g scale). Regiospecific bromination of 14-16 occurred smoothly under Br2 in dichloromethane at room temperature (Scheme 14-2).
Figure imgf000145_0002
Scheme 14-2
[0379] A number of reagents have been used for selective reduction of nifroaromatic compounds, the most common being alcoholic basic hydrogen sulfϊde, alcoholic ammonium sulphide, and alcoholic sodium polysulphide. However, the product amines from these reactions are generally obtained in low yields and are often difficult to purify. The hydrosulfide-induced reaction was investigated intensively by Idoux and is superior to the previously reported method. Based on this result, dinifro 14-16 was reduced to aniline 14-17 with NaSH in CH3OH; 2.7 equivalents of NaSH were needed to complete the reaction. Double reduced compound was also detected on the LCMS, but it was less than 5 % (Scheme 14-3).
[0380] Aniline 14-17 was also fluorinated under HF'Pyr and NaN02 to furnish 14-18 in 90
% yield. 14-18 could be reduced with SnCl2 as a usual way to form flouroaniline 14-19 in 91 % yield. The precursor 14-19 was coupled with various isocyanates, acyl halides and sulfonyl halides to form urea, amide and sulfonamide type molecules respectively in good yields (Scheme 14-3).
Figure imgf000146_0001
Urea type molecules Amide type molecules Sulfonamide type molecule
s
Figure imgf000146_0002
Scheme 14-3
[0381] The following contains the list of representative fluorinated compounds synthesized
(TABLE 14-1).
Table 14-1 Compounds ofthe invention containing a fluoride functionality.
Figure imgf000147_0001
Compound 96 Compound 100 Compound 101
Figure imgf000147_0002
Compound 98 Compound 97 Compound 103
Figure imgf000147_0003
Compound 183
[0382] In addition, as shown in Scheme 14-3, aniline 14-17 was also alkylated with 2 equivalents of CH3I and NaH at room temperature in 86 % yield. Finally, 14-17 was reduced with SnCl2*H20 in EtOH at a reflux condition to furnish the arninomethylated precursor 14-21 in 85 % yield (Scheme 14-4).
Figure imgf000147_0004
Scheme 14-4
[0383] Aniline 14-21 was coupled with various isocyanates, acyl chlorides and sulfonyl chlorides to form aminodimethylated compound ofthe invention in high yields (TABLE 14-2). Table 14-2 Compounds ofthe invention containing N, N-dimethylamino fuctionality.
Figure imgf000148_0001
Compound 113 Compound 129 Compound 136
Figure imgf000148_0002
Compound 105 Compound 119 Compound 144
[0384] The amino group on aniline 14-17 was transformed into a hydroxy group under
H2S04 and NaN02 to give phenol 14-22 in a 57 % yield. Phenol 14-22 was methylated with Mel and NaH in DMF followed by reduction witii SnCl2 «H20 in EtOH providing aniline 14-24 in good yields (Scheme 14-5).
Figure imgf000148_0003
Scheme 14-5
[0385] Aniline 14-24 was coupled with various isocyanates, acyl chlorides and sulfonyl chlorides to furnish the compounds ofthe invention containing a methoxy group (TABLE 14-3). Table 14-3 Compounds ofthe invention containing methoxy fuctionality.
Figure imgf000149_0001
Compound 152 Compound 158 Compound 163
Figure imgf000149_0002
Compound 162
[0386] An introduction of a hydroxy group on the aromatic ring was also attempted. Phenol
14-22 was protected with TBS-C1 in pyridine at room temperature followed by reduction with SnCl2 «H20 in EtOH to afford silyletiier 14-25 in high yield. After reduction with SnCl2, aniline 14-26 was coupled with 4-chlorophenyl isocyanate at room temperature and deprotected with TBAF in situ to furnish Compound 157 of the invention in 89 % yield after column chlomatography on silica gel (Scheme 14-6).
Figure imgf000149_0003
Figure imgf000149_0004
Figure imgf000149_0005
Compound 157
Scheme 14-6 [0387] The list of compounds of the invention containing a hydroxy functionality is shown below (TABLE 14-4).
Table 14-4 Compounds ofthe invention containing hydroxy functionality
Figure imgf000150_0001
Compound 161 Compound 150
[0388] Several attempts for infroduction of cycloamines on the aromatic group have been made. The direct substitution reaction of various cyclic amines under mild conditions gave desired products. For these reactions, DMSO is the choice of solvents (Scheme 14-7).
Figure imgf000150_0002
Scheme 14-7 [0389] Fluoro phenyl 14-18 was treated with pyrrolidine in DMSO to afford 14-27 in 93% yield. Subsequently, pyrrolidine 14-27 was reduced with four equivalents of SnCl2* 2H20 in EtOH to furnish the precursor 14-28 in 91 % yield. N-Methyl piperazine and moφholine were introduced to the aromatic group in the same manner to achive the precursors 14-29 and 14-30 respectively. With these precursors available, the coupling reactions were compelted with various isocyanates, acyl chlorides and sulfonyl chlorides. TABLES 14-5, 14-6 and 14-7 show additional compounds of the invention.
Table 14-5 Compounds ofthe invention containing pyrrolidine fuctionality.
Figure imgf000151_0001
Compound 125 Compound 141 Compound 115
Figure imgf000151_0002
Compound 184 Compound 122 Compound 145
Figure imgf000151_0003
Compound 109 Compound 148 Compound 118 Table 14-5 Continued
Figure imgf000152_0001
Compound 135 Compound 140
Table 14-6 Compounds ofthe invention containing ./V-methyl piperazine fuctionality.
Figure imgf000152_0002
Compound 117 Compound 124 Compound 133
Figure imgf000152_0003
Compound 116 Compound 147 Compound 112
Figure imgf000152_0004
Compound 127 Compound 132 Table 14-7 Compounds ofthe invention containing morpholine fuctionality.
Figure imgf000153_0001
Figure imgf000153_0002
Compound 107
Figure imgf000153_0003
Experiment 14.1 Preparation and Analysis of Compound
1,3-Dinifro iodobenzene 14-14:
Figure imgf000153_0004
[0390] 1,3-Dinitobenzene (10 g, 59.5 mmol) and I2 (7.5 g, 29.5 mmol) were dissolved in
20% oleum (75.9 g) and heated to 170 °C for 45 min. The reaction mixture was maintained at 170-180 °C for lhr. The temperature was lowered to room temperature. The rriixture was poured into crashed ice (500 g) and stirred for 2 hrs. The solid material was filtered and washed with H20 (500 mL). The solid was dissolved in EtOAc (250 mL) and washed with H20 (150 mL x 2). The EtOAc was dried over MgS04 and concenfrated under vaccum to afford to the desired product 14-14 in 62 % yield (10.8 g). Rf = 0.52 (EtOAc/Hex = 1/4). Experiment 14.2
Preparation and Analysis of Compound
Pyrazole 14-15
Figure imgf000154_0001
[0391] Iodobenzen 14-14 (2 g, 6.8 mmol) was dissolved in anhydrous THF (20 mL) and treated with fributyltin pyrazole 14-9 (2.5 g, 6.8 mmol, see Scheme 14-1) and PdCl2(PPh3)2 (0.48 g, 0.68 mmol). The mixture was heated to reflux. After refluxing for 12hrs, the reaction mixture was cooled to room temperature and concenfrated under vaccum. The residue was dissolved in EtOAc (50 mL) and washed with H20 (50 mL x 2). The EtOAc was dried over MgS04 and concenfrated under vaccum. The crude compound was recrystallized in EtOH (30 mL) to afford the desired coupled product 14-15 (1.41 g, 84 %). Rf = 0.5 (EtOAc/Hex = 1/3).
Experiment 14.3
Preparation and Analysis of Compound
Bromo pyrazole 14-16
Figure imgf000154_0002
[0392] To a solution of 14-15 (0.4 g, 1.6 mmol) in dichloromethane (5 mL), was added dropwise Br2 (91 μL, 1.77 mmol) in dichloromethane (1 mL) for 5 min at 0 °C. After addition of Br2, the reaction mixture was warmed to room temperature and stirred for 2 hrs. The reaction mixture was washed with H20 (10 mL) and friturated with EtOH to afford yellowish crystals ofthe desired product (0.46 g, 92 %) Rf = 0.72 (EtOAc/Hex = 1/2). Experiment 14.4
Preparation and Analysis of Compound
Aniline 14-17
Figure imgf000155_0001
[0393] Dinifrophenyl 14-16 (0.6 g, 1.8 mmol) was dissolved in MeOH (40 L) and toluene
(10 mL) and heated to refluxing. NaSH (262 mg, 4.68 mmol) in MeOH (10 mL) was added into the solution for 45 min. The reaction mixture was stirred for 30 min at the same temperature and cooled to room temperature. The reaction mixture was concenfrated under vaccum and dissolved in EtOAc (20 mL). The EtOAc was washed with H20 (20 mL x 2) and dried over MgS0 . The crude product was purified over column cliromatography (Rf = 0.51, EtOAc/Hex = 1/1) to afford to a yellow solid as the desired product 14-17 (487 mg, 91 %).
Experiment 14.5
Preparation and Analysis of Compound Fluorophenyl pyrazole 14-18
Figure imgf000155_0002
[0394] Aniline 14-17 (0.3 g, 1.01 mmol) was added into HF-Pyr (2.6 mL) at 0 °C and warmed to room temperature. After stirring for 30 min, the solution was cooled to -30 °C and NaN02 (77 mg, 1.11 mmol) added portionwise for 10 min. The reaction mixture was maintained at -30 °C for 30 min. The mixture was then heated to 145 °C and stirred for 10 min. The reaction mixture was cooled to room temperature and quenched with crashed ice (10 mL). The crude product was extracted with EtOAc (20 mL), dried over MgS04 and concentrated under vaccum. The crude material was purified by column chromatograph (EtOAc/Hex = 1/1, Rf = 0.85) to afford a yellow crystal as desired product 14-17 (275 mg, 91%). Experiment 14.6
Preparation and Analysis of Compound
Fluoro aniline 14-19
Figure imgf000156_0001
[0395] A 5 mL round-bottom flask was charged with 100 mg of 14-18 (0.33 mmol) and
SnCl2*2H20 (0.3 g, 1.33 mmol) and dissolved in EtOH (3 mL). The mixture was refluxed for 1.5 hrs and cooled to room temperature. The reaction was under concentrated under vaccum and exfracted with EtOAc (20 mL x 2) and H20 (20 mL). The EtOAc was dried over MgS04, concenfrated under vaccum and purified over silica gel (EtOAc/Hex = 1/1, R = 0.41) to afford a white crystal of the desired product 14-19 (79 mg, 89%).
Experiment 14.7
Preparation and Analysis of Compound
Dimethyl aniline 14-20
Figure imgf000156_0002
[0396] Aniline 14-17 (1.0 g, 3.36 mmol) was dissolved in DMF (5 mL) in a 10 mL round- bottom flask. The solution was cooled to 0 °C and treated with 60 % NaH (404 mg, 10.08 mmol). The reaction mixture was maintained at the same temperature for 30 min and warmed to room temperature. The πiixture was quenched with 100 μL of EtOH and exfracted with EtOAc (20 mL). The EtOAc was dried over MgS0 and concenfrated under vaccum. The residue was purified over silica gel (EA/Hex = 1/1, Rf = 0.81) to afford yellow solids as the desired product 14-20 (858 mg, Experiment 14.8
Preparation and Analysis of Compound
Aniline 14-21
Figure imgf000157_0001
[0397] A 5 mL round-bottom flask was charged with 103 mg of 14-20 (0.33 mmol) and
SnCl2«2H20 (0.3 g, 1.33 mmol) and dissolved in EtOH (3 mL). The mixture was refluxed for 1.5 hrs and cooled to room temperature. The reaction was under concentrated under vaccum and extracted with EtOAc (20 mL x 2) and H20 (20 mL). The EtOAc was dried over MgS04, concentrated under vaccum and purified over silica gel (EtOAc/Hex = 1/1, Rf = 0.41) to afford a white crystal as desired product 14-21 (82 mg, 85%).
Experiment 14.9
Preparation and Analysis of Compound
Phenol 14-22
Figure imgf000157_0002
[0398] Aniline 14-17 (2 g, 6.73 mmol) was dissolved in H2S04 (5 mL) and H20 (5 mL) at 0
°C. The mixture was warmed to room temperature and stirred for 30 min. The solution was treated with NaN02 (557 mg, 808 mmol) at -30 °C and stirred for 1 hr. The reaction mixture was heated to 100 °C and stirred for 1 hr. The resulting mixture was cooled to room temperature and quenched with crashed ice (10 g). The mixture was extracted with EtOAc (20 mL x 2). The EtOAc was dried over MgS04 and concentrated under vaccum. The residue was purified over silica gel (EtOAc/Hex = 1/1, Rf = 0.52) to afford the white crystals of desired product 14-22 (1.13 g, 57%). Experiment 14.10
Preparation and Analysis of Compound
Anisole 14-23
Figure imgf000158_0001
[0399] To a cooled solution of phenol 14-22 (657 mg, 2.2 mmol) in DMF (5 mL) was freated with 60 % NaH (106 mg, 2.65 mmol). The resulting mixture was stirred at 0 °C and freated with CH3I (165 μL, 2.65 mmol). The reaction mixture was stirred for 1 hr and warmed to room temperature. The mixture was poured into H20 (10 mL) and exfracted with EtOAc (10 mL x 2). The EtOAc was dried over MgS04 and concentrated under vaccum. The residue was purified over silica gel (EtOAc/Hex = 1/1 , Rf = 0.75) to afford desired anisole 14-23 (629 mg, 92%).
Experiment 14.11
Preparation and Analysis of Compound
Aniline 14-24
Figure imgf000158_0002
[0400] A 25 mL round-bottom flask wad charged with 103 mg of anisole 14-23 (2.08 mmol) and SnCl2*2H20 (2.13 g, 8.36 mmol) and dissolved in EtOH (10 mL). The mixture was refluxed for 1.5 hrs and cooled to room temperature. The reaction was concentrated under vaccum and exfracted with EtOAc (30 mL x 2) and H20 (30 mL). The EtOAc was dried over MgS04, concenfrated under vaccum and purified over silica gel (EtOAc/Hex = 1/1, Rf = 0.41) to afford a white crystal of desired product 14-24 (489 mg, 84 %). Experiment 14.12
Preparation and Analysis of Compound
Silyl ether 14-25
Figure imgf000159_0001
[0401] To a solution of phenol 14-22 (lg, 3.38 mmol) dissolved in pyridine (3 mL) was added TBS-C1 (611 mg, 4.05 mmol) and the resulting solution was allowed to stir for 10 hrs. The reaction mixture was poured into H20 (20 mL) and exfracted with EtOAc (20 mL x 2). The EtOAc was washed witii 1.0 N HCl (10 mL) dried over MgS04 and concentrated under vaccum to afford crude product 14-25 (1.36 g, 96%). The crade product was used for the next step without furtiier purification (EtOAc/Hex = 1/1, Rf = 0.89).
Experiment 14.13
Preparation and Analysis of Compound
Aniline 14-26
Figure imgf000159_0002
[0402] A 25 mL round-bottom flask was charged with 1.0 g of 14-25 (2.4 mmol) and
SnCl2«2H20 (2.19 g, 9.7 mmol) and dissolved in EtOH (10 mL). The mixture was refluxed for 1.5 hrs and cooled to room temperature. The reaction was concenfrated under vaccum and exfracted with EtOAc (30 mL x 2) and H20 (30 mL). The EtOAc was dried over MgS04, concentrated under vaccum and purified over silica gel (EtOAc/Hex = 1/1, Rf = 0.41) to afford a white crystal of desired product 14-26 (798 mg, 87 %). Experiment 14.14
Preparation and Analysis of Compound
Pyrrolidine 14-27
Figure imgf000160_0001
[0403] Fluorophenyl pyrazole 14-18 (2.6 g, 8.67 mmol) was directly weighed into a 50 mL round-bottom flask and dissolved in DMSO (10 mL). The solution was treated with pyrrolidine (2.2 mL, 26 mmol) and heated to 80 °C. After stirring for 2 hrs, tiie reaction was poured into H20 (50 mL) and extracted with EtOAc (20 mL x 2). The EtOAc was washed with 1.0 N HCl (10 mL), dried over MgS0 and concenfrated under vaccum. The residue was purified over silica gel to afford the desired product 14-27 (2.77 g, 93%).
Experiment 14.15
Preparation and Analysis of Compound
Aniline 14-28
Figure imgf000160_0002
[0404] A 50 mL round-bottom flask was charged with 2.0 of Pyrrolidine 14-27 (5.7 mmol) and SnCl2*2H20 (5.8 g, 22.8 mmol) charged dissolved in EtOH (30 mL). The mixture was refluxed for 1.5 hrs and cooled to room temperature. The reaction was concentrated under vaccum and extracted with EtOAc (50 mL x 2) and H20 (50 mL). The EtOAc was dried over MgS04, concentrated under vaccum and purified over silica gel (EtOAc/Hex = 1/1, Rf = 0.41) to afford a white crystal of desire product 14-28 (1.66 g, 91 %).: Experiment 14.16
Preparation and Analysis of Compound
N-Methylpiperazine 14-29
Figure imgf000161_0001
[0405] Fluorophenyl pyrazole 14-18 (2.6 g, 8.67 mmol) was directly weighed into a 50 mL round-bottom flask and dissolved in DMSO (10 mL). The solution was freated with 4- methylpiperazine (1.5 mL, 17.3 mmol) and heated to 80 °C. After stirring for 2 hrs, the reaction was poured into H20 (50 mL) and extracted with EtOAc (20 mL x 2). The EtOAc was washed with 1.0 N HCl (10 mL), dried over MgS04 and concentrated under vaccum. The crade product was used for the next step without furtiier purification. A 100 mL round-bottom flask was charged with the crade compound and SnCl2*2H20 (8.86 g, 34.7 mmol) and dissolved in EtOH (70 mL). The mixture was refluxed for 1.5 hrs and cooled to room temperature. The reaction was concentrated under vaccum and exfracted with EtOAc (50 mL x 2) and H20 (50 mL). The EtOAc was dried over MgS04, concentrated under vaccum and purified over silica gel EtOAc (Hex = 1/1, Rf = 0.38) to afford a white crystal of desired product 14-29 (2.12 g, 69.7 % in 2 steps).
Experiment 14.17
Preparation and Analysis of Compound
Moφholine 14-30
Figure imgf000161_0002
[0406] Fluorophenyl pyrazole 14-18 (509 mg, 1.69 mmol) was directly weighed into a 5 mL round-bottom flask and dissolved in DMSO (3 mL). The solution was freated with moφholine (295 μL, 3.38 mmol) and heated to 80 °C. After stirring for 2 hrs, the reaction was poured into H20 (10 mL) and exfracted with EtOAc (10 mL x 2). The EtOAc was washed with 1.0 N HCl (5 mL), dried over MgS04 and concenfrated under vaccum. The curde product was used for the next step without further purification. A 25 mL round-bottom flask was charged with the crude compound and SnCl2«2H20 (1.72 g, 6.79 mmol) and dissolved in EtOH (20 mL). The mixture was refluxed for 1.5 hrs and cooled to room temperature. The reaction was concenfrated under vaccum and extracted with EtOAc (10 mL x 2) and H20 (10 mL). The EtOAc was dried over MgS04, concenfrated under vaccum and purified over silica gel (EtOAc/Hex = 1/1, Rf = 0.45) to afford a white crystal of desired product 14-30 (415 mg, 73.5 % in 2 steps).
Experiment 14.18 Preparation and Analysis of Compound
Compound 158
Figure imgf000162_0001
Typical coupling reaction of the precursors and isocyanates:
[0407] To a 1 mL oven dried vial, was aniline 14-24 (80 mg, 0.28 mmol, see Experiment 14- 11 and Scheme 14-5) dissolved in dichloroethane (0.3 mL) and freated with p-chlorophenyl isocyanate (53 mg, 0.34 mmol) at room temperature.
[0408] After stirring for 12 hrs, the solid was filtered, washed with dichloroethane (0.5 mL) and dried in vacuo to afford Compound 158 (90.1 mg, 75%).
Experiment 14.19
Preparation and Analysis of Compound
Compound 100
Figure imgf000162_0002
Typical coupling reaction of the precursors and sulfonyl halides or acyl halides
[0409] To a 1 mL oven dried vial, was aniline 14-19 (50 mg, 0.17 mmol, see Experiment 14-
6 and Scheme 14-3) dissolved in dichloromethane (0.3 mL) and freated with p-fluorophenyl sulfonylchloride (40 mg, 0.20 mmol) and Et3N (28 μL, 0.21 mmol) at 0 °C. The reaction was warmed to room temperature and maintained for 4 hrs. The mixture was poured into H20 (10 mL) and exfracted with EtOAc (20 mL x 2). The EtOAc was dried over MgS04, concentrated under vaccum and purified over silica gel (EtOAc/Hex = 1/2, Rf = 0.52) to afford Compound 100 as a white crystal (58 mg, 85%).:
EXAMPLE 15
Experiments 15.1 - 15.22
General Synthetic Strategies, Preparation and Analysis of Compounds
[0410] Compounds of certain embodiments of the invention were prepared according to the procedures below. The following abbreviations are employed herein: List of Abbreviations:
DME Dimethoxyethylene
THF Tefrahydrofuran
Pd2(dba)3 Tris(dibenzylideneacetone)dipalladium NCS N-Chlorosuccinimide
ACC l,l-Azobis(cyclohexanecarbonitrile)
DIEA Diisopropylethylamine
DMSO Dimethylsulfoxide
Hex Hexanes EtOAc Ethyl Acetate
GENERALSYNTHETIC STRATEGIES
[0411] N-Methyl pyrazole was lithiated with n-BuLi in THF at -78°C. The lithiated pyrazole was then exchanged with I2 to form iodopyrazole 15-1 in 78% yield. Suzuki coupling of 1 with 3-nifrophenylboronic acid provided coupled product 15-2 in 67-98% yield (Scheme 15-1).
Figure imgf000163_0001
15-2
Scheme 15-1 [0412] Pyrazole 15-2 was brominated at 0°C to provide 15-3 in 68% yield. Intermediate 15-
3 was then reduced with iron in 1:1 acetic acid/ethanol to afford aniline 15-4 in 80% yield. Aniline 15-4 was subsequently coupled with 2,4-difluorophenylisocyanate at room temperature to provide Compound 173 in 95% yield (Method A, Scheme 15-2).
Method A
Figure imgf000164_0001
Scheme 15-2
[0413] Separately, Pyrazole 15-2 was chlorinated at 85°C with NCS providing intermediate chloropyrazole 15-5 in 87% yield. Intermediate 15-5 was then reduced with H2/Pd-C to provide aniline 15-6 in 88% yield. Aniline 15-6 was subsequently freated with 3,4-difluorophenylisocyanate at room temperature to provide Compound 171 in 85% yield (Method B, Scheme 15-3).
Method B
Figure imgf000164_0002
Compound 171
Scheme 15-3
[0414] The following compounds shown below were made in a similar manner as described above herein for Compound 173 and Compound 171.
Figure imgf000165_0001
(X)
Figure imgf000165_0002
Figure imgf000166_0001
PREPARATION AND ANALYSIS OF COMPOUNDS
Experiment 15.1 Preparation and Analysis of Compound
5-Iodo-l-N-methylpyrazole 15-1
[0415] N-Methylpyrazole (10.0 mL, 120.6 mmol) was dissolved in THF (250 mL) and freated with n-BuLi (75.6 mL, 120.6 mmol) at -78°C under dry Ν2. The reaction was stirred for 30 min at -78°C. The reaction mixture was freated at -78°C with I2 (30.7 gm, 120.6 mmol) in THF (200 mL). The reaction was allowed to warm to room temperature and stirred an additional 3 hours. The rnixture was quenched by the addition of NFL.C1 (200 mL) stirring for 30 min. Extraction with EtOAc (3 x 100 mL) then washing of the organic phase with a 10% solution of Na2S203 removed die excess I2. The combined organic layers were then dried over Na2S04, filtered and the solvent removed under reduced pressure to provide 19.6g of 5-Iodo-l-N-methylpyrazole 15-1 in 78% yield (the material was used as crade in the next step): LCMS m/z 209 MH\ *H NMR (400 MHz. CDC13) σ 7.46 (d, J = 2 Hz, IH), 6.4 (d, J = 2 Hz, IH), 3.9 (s, 3H). Experiment 15.2
Preparation and Analysis of Compound
Pyrazole 15-2 (Route-1)
[0416] The intermediate iodopyrazole 15-1 (588 mg, 2.82 mmol) was dissolved in DME (5 mL) and H20 (500 μL). To this solution was added 3-nitrophenylboronic acid (943 mg, 5.65 mmol), Cs2C03 (920 mg, 2.82 mmol) and Pd(Ph3P)4 (555 mg, 0.48 mmol). The mixture was degassed with argon and heated to 80°C for 12 hours. After cooling to room temperature, brine (30 mL) was added and the mixture exfracted with EtOAc (3 x 30 mL). The combined organic layers were dried over Na2S04, filtered and tiie solvent removed under reduced pressure. The residue was purified by flash chromatography (Si02, Hexanes/EtOAc gradient elution) to provide 562 mg (98%) of pyrazole 15-2 as a pale yellow solid: LCMS m/z 204 MH1". _ϊ NMR (400 MHz. CDC13) σ 8.25 (s, IH), 8.22 (dd, J=7.8, 1.0 Hz, IH), 7.74 (d, J=7.6 Hz, IH), 7.64 (dd, J=8.0, 8.0 Hz, IH ), 7.52 (m, IH), 6.39 (m, IH), 3.92 (m, 3H).
Pyrazole 15-2 (Route-2)
[0417] The intermediate iodopyrazole 5-1 (17.4 gm, 83.5 mmol) was dissolved in DME (200 mL) and H20 (1 mL). To this solution was added 3-nifrophenylboronic acid (14.0 gm, 83.5 mmol), Cs2C03 (27.4 gm, 83.5 mmol) and Pd2(dba)3 (1.37 gm, 14 mmol). The mixture was degassed with argon and heated to 80°C for 12 hours. After cooling to room temperature, brine (100 mL) was added and the mixture extracted with EtOAc (2 x 100 mL). The combined organic layers were dried over Na2S0 , filtered and the solvent removed under reduced pressure. The residue was purified by flash chromatography (Si02, Hexanes/EtOAc gradient elution) to provide 17.1 gm (67%) of Pyrazole 15-2 as a pale yellow solid: LCMS m/z 204 MH+. Η NMR (400MHz. CDC13) σ 8.25 (s, IH), 8.22 (dd, J=7.8, 1.0 Hz, IH), 7.74 (d, J=7.6 Hz, IH), 7.64 (dd, J=8.0, 8.0 Hz, IH ), 7.52 (m, IH), 6.39 (m, IH), 3.92 (m, 3H).
Experiment 15.3
Preparation and Analysis of Compound Method A Bromopyrazole 15-3
[0418] Pyrazole 15-2 (5.79 g, 28.5 mmol) was dissolved in 150 mL of CH2C12 and cooled to
0°C. Bromine (5.40 g, 34.2 mmol) in CH2C12 (55 mL) was added drop wise under argon over 30 minutes. After the addition was complete, the reaction mixture stirred for 3 hours at room temp. The reaction mixture was quenched with a saturated solution of Na2S203 and IN NaOH. The layers were separated and the aqueous layer was exfracted with CH2C12 (2x200 ml). The organic layers were combined, dried over Na2S0 , and the solvent removed under reduced pressure. The residue was purified by flash chromatography (Si02, Hexanes/EtOAc gradient elution) to provide 5.4 gm (68%) of Bromopyrazole 15-3 and 1.1 gm (14%) of unreacted starting material. LCMS m/z (%) = 284 (M+H81Br, 100), 282 (M+H79Br, 89). __ NMR (400MHz. CDC13) σ 8.33 (d, J=6.0 Hz, IH), 8.29 (s, IH), 7.77 (d, J=8.0 Hz, IH), 7.71 (dd, J=8.0 Hz, IH), 7.56 (s, IH), 3.87 (s, 3H).
Intermediate 15-4:
[0419] A slurry of pyrazole 15-3 (2.05 g, 7.26 mmol) in a 1:1 mixture of acetic acid and ethanol (30 ml) was treated with iron powder (1.62 g, 29.0 mol) and heated to 70°C for 4 hours. The reaction mixture was allowed to cool to room temperature and IN NaOH (100 ml) was added to form white precipitate (FeOH). EtOAc (100 mL) was added and filtered through celite. The organic layer was separated and the aqueous layer was extracted with EtOAc (2x100 ml). The organic layers were combined and dried over Na2S04, filtered, and concenfrated to afford 1.46 g of 15-4, in 80% yield as a white solid. LCMS m/z (%) = 254 (M+H81Br, 100), 252 (M+H79Br, 96). __ NMR (400MHz. CDC13) σ 7.50 (s, 2H), 7.26 (m, 2H), 6.75 (m, 2H), 6.68 (s, IH), 3.81 (s, 3H).
Experiment 15.4 Preparation and Analysis of Compound
Compound 173
[0420] Intermediate 4 (209 mg, 0.83 mmol, see Experiment 15.3) was dissolved in 3 mL of
CH2C12, freated with 2,4-difluorophenylisocyanate (385 mg, 2.49 mmol), diisopropylethylamine (168 mg, 1.66 mmol), and stirred at room temperature overnight. The solvent was removed under reduced pressure, dissolved in DMSO (5 ml), and purified by preparative HPLC to afford Compound 173 as a white solid, 325 mg, 97% yield: LCMS m/z (%) = 409 (M+H81Br, 100), 407 (M+H79Br, 97). Η NMR (400 MHz. DMSO-ds) σ 9.20 (s, IH), 8.55 (s, IH), 8.04 (m, IH), 7.64 (m, IH), 7.59 (s, IH), 7.51 (d, J=8.4 Hz, IH), 7.44 (dd, J=7.8 Hz, IH), 7.29 (m, IH), 7.09 (d, J=7.2 Hz, IH), 7.03 (dd, J=8.8 Hz, IH), 3.77 (s, 3H). Experiment 15.5
Preparation and Analysis of Compound
Compound 165
[0421] Intermediate 4 was freated with 3,4-difluorophenylisocyanate, in a similar manner to Compound 173, providing a 99% yield of Compound 165: LCMS m/z (%) = 409 (M+H81Br, 100), 407 (M+H79Br, 97). Η NMR (400 MHz. DMSO-d6) σ 8.98 (s, 2H), 7.57 (m, 3H), 7.44 (dd, J=8.0 Hz, IH), 7.36 (d, J=9.2 Hz, IH), 7.31 (d, J=9.6 Hz, IH), 7.10 (m, 2H), 3.78 (s, 3H).
Experiment 15.6 Preparation and Analysis of Compound
Compound 166
[0422] Intermediate 4 was freated with 2,5-difluorophenylisocyanate, in a similar manner to
Compound 173, providing a 76% yield of Compound 166: LCMS m/z (%) = 409 (M+H81Br, 100), 407 (M+H79Br, 97). Η NMR (400 MHz. CDCL3) σ 7.93 (m, IH), 7.79 (s, IH), 7.73 (s, IH), 7.57 (s, IH), 7.50 (s, IH), 7.24 (m, 2H), 7.10 (d, J=6.8 Hz, IH), 6.96 (m, IH), 6.64 (m, IH), 3.82 (s, 3H).
Experiment 15.7
Preparation and Analysis of Compound
Compound 167
[0423] Intermediate 4 was freated with 3,5-difluoroρhenylisocyanate, in a similar manner to
Compound 173, after chromatography on silica (Chromatatron, Hex/EtOAc gradient), providing a 45% yield of Compound 167: LCMS m/z (%) = 409 (M+H81Br, 100), 407 (M+H79Br, 96). Η NMR (400 MHz. Acetone- d_) σ 8.59 (s, IH), 8.47 (s, IH), 7.68 (s, IH), 7.64 (d, J=8.0 Hz, IH), 7.52 (s, IH), 7.47 (dd, J=7.8 Hz, IH), 7.24 (J=9.6 Hz, 2H), 7.14 (d, J=7.6 Hz, IH), 6.61 (dd, J=8.6 Hz, IH), 3.83 (s, 3H).
Experiment 15.8
Preparation and Analysis of Compound Compound 168
[0424] Intermediate 4 was freated with 3-chloro-4-fluorophenylisocyanate, in a similar manner to Compound 173, providing a 91% yield of Compound 168: LCMS m/z (%) = 427 (M+H81Br, 37C1, 33), 425 (M+H81Br, 35C1 & 79Br, 37C1, 100), 423 (M+H79Br, 35C1, 63). >H NMR (400 MHz. Acetone- d6) σ 8.41 (s,lH), 8.37 (s, IH), 7.87 (m,lH),7.69 ( , IH), 7.62 (d, J=2.4 Hz, IH), 7.52 (s, IH), 7.45 (dd, J=7.8 Hz, IH), 7.38 (m, IH), 7.21 (dd, J=9.0 Hz, IH), 7.12 (d, J=6.8 Hz, IH), 3.83 (s, 3H).
Experiment 15.9
Preparation and Analysis of Compound Compound 169
[0425] Intermediate 4 was freated with 2-frifluoromethyl-4-fluorophenylisocyanate, in a similar manner to Compound 173, providing a 47% yield of Compound 169: LCMS m/z (%) = 459 (M+H81Br, 100), 457 (M+H79Br, 78). _ϊ NMR (400 MHz. DMSO-ds) σ 9.47 (s, IH), 8.64 (s, IH), 8.16 (s, IH), 7.86 (m, IH), 7.76 (m, IH), 7.64 (s, IH), 7.54 (m, 2H), 7.45 (dd, J=7.8 Hz, IH), 7.09 (d, J=7.2 Hz, IH), 3.77 (s, 3H).
Experiment 15.10 Preparation and Analysis of Compound
Compound 170
[0426] Intermediate 4 was freated with 3-frifluoromethyl-4-fluorophenylisocyanate, in a similar manner to Compound 173, providing a 56% yield of Compound 170: LCMS m/z (%) = 459 (M+H81Br, 100), 457 (M+H79Br, 82). Η NMR (400 MHz. DMSO-d6) σ 9.13 (s, IH), 9.04 (s, IH), 7.98 (m, IH), 7.64 (m, 2H), 7.58 (s, IH), 7.55 (d, J=8.4 Hz, IH), 7.45 (m, 2H), 7.09 (d, J=8.8 Hz, IH), 3.77 (s, 3H).
Experiment 15.11 Preparation and Analysis of Compound
Compound 174
[0427] Intermediate 4 was freated with 2,3,4-frifhιorophenylisocyanate, in a similar manner to Compound 173, providing a 22% yield of Compound 174: LCMS m/z (%) = 427 (M+H81Br, 100), 425 (M+H79Br, 78). _ϊ NMR (400 MHz. Acetone- d_) σ 8.74 (s, IH), 8.22 (s, IH), 8.01 (m, IH), 7.71 (m, IH), 7.64 (d, J=8.0 Hz, IH), 7.52 (s, IH), 7.48 (dd, J=7.8 Hz, IH), 7.15 (m, 2H), 3.84 (s, 3H). Experiment 15.12
Preparation and Analysis of Compound
Compound 180
[0428] Intermediate 4 was freated with 2-frifluoromethyl-4-chlorophenylisocyanate, in a similar manner to Compound 173, after chromatography on silica (Biotage, Hex/EtOAc gradient) provided Compound 180 in a 73% yield: LCMS m/z (%) = 477 (M+H81Br, 37C1, 40), 475 (M+H81Br, 35C1 & 79Br, 37C1, 100), 473 (M+H79Br, 35C1, 72). Η NMR (400 MHz. Acetone-d6) σ 9.04 (s, IH), 8.19 (d, J=9.2 Hz, IH), 7.77 (s, IH), 7.66 (m, 4H), 7.52 (s, IH), 7.48 (dd, J=8.0 Hz, IH), 7.14 (d, J=7.6, lH), 3.83 (s, 3H).
Experiment 15.13
Preparation and Analysis of Compound
Compound 181
[0429] Intermediate 4 was freated with 3-frifluoromethyl-4-chlorophenylisocyanate, in a similar manner to Compound 173, providing a 56% yield of Compound 181: LCMS m/z (%) = 477
(M+H81Br, 37C1, 34), 475 (M+H81Br, 35C1 & 79Br, 37C1, 100), 473 (M+H79Br, 35C1, 62). Η NMR (400
MHz. Acetone-ds) σ 8.85 (s, IH), 8.72 (s, IH), 8.16 (d, J=2.8 Hz, IH), 7.77 (s, J=8.8Hz, IH), 7.71 (s,
IH), 7.67 (d, J=8.4 Hz, IH), 7.55 (d, J=8.8 Hz, IH), 7.52 (s, IH), 7.48 (dd, J=7.8 Hz, IH), 7.14 (d,
J=8.8, lH), 3.84 (s, 3H).
Experiment 15.14
Preparation and Analysis of Compound
Method B:
Chloropyrazole 15-5
[0430] Pyrazole 15-2 (2.51 g, 12.3 mmol) was dissolved in 70 ml of CC and heated to 85°C under reflux. N-Chlorosuccinimide (1.81 g, 13.6 mmol) and 1,1-Azobis (cyclohexanecarbonifrile) were added and stirred overnight. The reaction mixture was allowed to cool to room temperature then to 0°C and the white precipitate filtered off. The solvent was removed under reduced pressure and purified by flash chromatography (Si02, Hexanes/EtOAc gradient elution) to provide 2.54 g, 87% yield of chloropyrazole 15-5. LCMS m/z (%) = 240 (M+H37C1, 54), 238 (M+H35C1, 100). *H NMR (400 MHz. CDC13) σ 8.33 (d, J=8.4 Hz, IH), 8.31 (s, IH), 7.78 (d, J=7.6 Hz, IH), 7.72 (dd, J=7.8 Hz, IH), 7.55 (s, IH), 3.87 (s, 3H). Intermediate 15-6:
[0431] A slurry of the chloropyrazole 15-5 (973 mg, 4.09 mmol) was reduced at 55 psi H2 /
Pd-C for 4 hours to provide 749 mg of intermediate 15-6 in 88% yield. LCMS m/z (%) = 210 (M+H37C1, 68), 208 (M+H35C1, 100). _ϊ NMR (400 MHz. CDC13 ) σ 7.47 (s, 2H), 7.29 (s, IH), 7.25 (dd, J=3.8 Hz, IH), 6.77 (d, J=2.0 Hz, IH), 6.75 (d, J=2.0 Hz, IH), 6.7 (m, IH), 3.81 (s, 3H).
Experiment 15.15
Preparation and Analysis of Compound
Compound 171
[0432] Intermediate 15-6 (200 mg, 0.96 mmol) was dissolved in 3 mL of CH2C12, freated with 3,4-difluorophenylisocyanate ( 4.49 mg, 2.89 mmol), diisopropylethylamine (194 mg, 1.92 mmol), and stirred at room temperature overnight. The solvent was removed under reduced pressure, dissolved in DMSO (5 ml), and purified by preparative HPLC to afford Compound 171 as a white solid, 298 mg, 85% yield: LCMS m/z (%) = 365 (M+H37C1, 27), 363 (M+H35C1, 100). __ NMR (400 MHz. DMSO-ds) σ 8.95 (m, 2H), 7.62 (m, 3H), 7.52 (d, J=8.0 Hz, IH), 7.45 (dd, J=7.8 Hz, IH), 7.32 (m, IH), 7.10 (m, 2H), 3.77 (s, 3H).
Experiment 15.16
Preparation and Analysis of Compound Compound 172
[0433] Intennediate 6 was freated with 2,5-difluorophenylisocyanate, in a similar manner to
Compound 171, after cliromatography on silica (Biotage, Hex/EtOAc gradient) providing a 57% yield of Compound 172: LCMS m/z (%) = 365 (M+H37C1, 32), 363 (M+H35C1, 100). Η NMR (400 MHz. CD3OD) σ 7.99 (m, IH), 7.63 (s, IH), 7.49 (m, 2H), 7.44 (dd, J=8.0 Hz, IH), 7.10 (m, 3H), 6.69 (m, 2H), 3.81 (s, 3H).
Experiment 15.17
Preparation and Analysis of Compound
Compound 175
[0434] Intermediate 6 was treated with 3,5-difluorophenylisocyanate, in a similar manner to
Compound 171, after chromatography on silica (Biotage, Hex/EtOAc gradient) providing a 20% yield of Compound 175: LCMS m/z (%) = 365 (M+H37C1, 27), 363 (M+H35C1, 100). Η NMR (400 MHz. CDC13) σ 7.50 (s, IH), 7.44 (s, IH), 7.43 (s, IH), 7.36 (s, IH), 7.25 (s, 2H), 7.14 (m, IH), 6.98 (d, J=6.8 Hz, 2H), 6.49 (m, IH), 3.82 (s, 3H).
Experiment 15.18 Preparation and Analysis of Compound
Compound 176
[0435] Intermediate 6 was treated with 3-chloro-4-fluorophenylisocyanate, in a similar manner to Compound 171, after chromatography on silica (Biotage, Hex/EtOAc gradient) providing a 21% yield of Compound 176: LCMS m/z (%) = 383 (M+H37C1,37C1, 19), 381 (M+H37C1,35C1, 73), 379 (M+H35C1,35C1, 100). Η NMR (400 MHz. CDC13) σ 7.50 (m, 2H), 7.43 (m, 2H), 7.25 (s, IH), 7.19 (m, IH), 7.13 (m, IH), 7.06 (dd, J=8.6 Hz, IH), 6.99 (s, IH), 6.95 (s, IH), 3.82 (s, 3H).
Experiment 15.19
Preparation and Analysis of Compound Compound 177
[0436] Intermediate 6 was freated with 2,4-difluorophenylisocyanate, in a similar manner to
Compound 171, after chromatography on silica (Biotage, Hex/EtOAc gradient) providing a 37% yield of Compound 177: LCMS m/z (%) = 365 (M+H37C1, 32), 363 (M+H35C1, 100). Η NMR (400 MHz. DMSO-ds) σ 9.20 (s, IH), 8.55 (s, IH), 8.04 (m, IH), 7.64 (s, IH), 7.59(s, IH), 7.50 (d, J=5.6 Hz, IH), 7.44 (dd, J=7.8, IH), 7.29 (m, IH), 7.09 (d, J=6.0, IH), 7.03 (dd, J=8.8, IH), 3.77 (s, 3H).
Experiment 15.20
Preparation and Analysis of Compound
Compound 178
[0437] Intermediate 6 was freated with 2,3,4-frifluorophenylisocyanate, in a similar manner to Compound 171, after chromatography on silica (Biotage, Hex/EtOAc gradient) providing a 45% yield of Compound 178: LCMS m/z (%) = 383 (M+H37C1, 52), 381 (M+H35C1, 100). Η NMR (400 MHz. Acetone-d6) σ 8.68 (s, IH), 8.17 (s, IH), 8.02 (m, 2H), 7.72 (s, IH), 7.63 (d, J=7.2 Hz, IH), 7.51 (s, IH), 7.48 (d, J=8.4 Hz, IH), 7.16 (m, IH), 3.83 (s, 3H). Experiment 15.21
Preparation and Analysis of Compound
Compound 179
[0438] • Intermediate 6 was treated with 2-trifluoromethyl-4-fluorophenylisocyanate, in a similar manner to Compound 171, after cliromatography on silica (Biotage, Hex/EtOAc gradient) providing a 32% yield of Compound 179: LCMS (%) = 415 (M+H37C1, 48), 413 (M+H35C1, 100). Η NMR (400 MHz. Acetone-ds) σ 8.92 (s, IH), 8.15 (s, IH), 8.07 (dd, J=6.8 Hz, IH), 7.96 (m, IH), 7.72 (m, IH), 7.62 (d, J=7.6 Hz, IH), 7.49 (m, 3H), 7.14 (d, J=7.6 Hz, IH), 3.82 (s, 3H).
Experiment 15.22
Preparation and Analysis of Compound '
Compound 182
[0439] Intermediate 4 was treated with 3-trifluoromethyl-4-fluorophenylisocyanate, in a similar manner to Compound 171, providing a 71% yield of Compound 182: LCMS m/z (%) = 415 (M+H37C1, 79), 413 (M+H35C1, 100). !H NMR (400 MHz. Acetone-d6) σ 8.50 (s, IH), 8.45 (s, IH), 8.04 (d, J=8.8 Hz, IH), 7.76 (m, IH), 7.71 (s, IH), 7.62 (d, J=8.0 Hz, IH), 7.51 (s, IH), 7.48 (dd, J=7.8 Hz, IH), 7.33 (dd, J=9.6 Hz, IH), 7.15 (d, J=7.2 Hz, IH), 3.83 (s, 3H).
EXAMPLE 16 Experiments 16.1 - 16.5
General Synthetic Strategies, Preparation and Analysis of Compounds
[0440] Compounds of certain embodiments of the invention were prepared according to the procedures below.
General Synthetic Strategies
[0441] Various aryl functionalities were introduced on the phenyl ring. By way of example, dinifro 14-15 (see Schemel4-2) was reduced with NaSH in ethanol under reflux conditions, followed by bromination under Sandmyer's conditions to afford Aryl bromide 16-1 in 56 % yield (2 steps). Aryl bromide 16-1 was coupled with p -methoxyphenyl boronic acid with Pd(PPh3)4 and K2C03 as a base in dioxane to furnish the coupled product 16-2 (Scheme 16-1). The reaction was very clean and the desired product was purified by recrystallization in ethanol. Coupled product 16-2 was subsequently brominated with Br2 in dichloromethane at room temperature (i.e., 16-3) followed by reduction with SnCl2*2H20 in Ethanol to furnish the aniline precursor 16-4. 4-Fluorophenyl functionality was also introduced on the ring in a similar manner (Scheme 16-2).
Figure imgf000175_0001
Scheme 16-1
Figure imgf000175_0002
16-2 16-3 16-4
Figure imgf000175_0003
16-5
Scheme 16-2 [0442] Anilines 16-4 and 16-5 were coupled with various isocyanates, acyl halides and sulfonyl halides to form urea, amide and sulfonamide type molecules respectively in good yields. The tables ofthe new compounds are shown below (TABLES 16-1 and 16-2).
TABLE 16-1 Compounds ofthe invention containing p-methoxyphenyl functionality.
Figure imgf000176_0001
Compound 191 Compound 192 l-[5-(4-Bromo-2-methyl-2H-pyrazol-3- 1 -[5-(4-Bromo-2-mefhyl-2H- yl)-4'-methoxy-biphenyl-3-yl]-3-(4- pyrazol-3-yl)-4'-methoxy-biphenyl- chloro-phenyl)-urea 3-yl]-3-(4-fluoro-phenyl)-urea
Figure imgf000176_0002
Compound 186 Compound 187 l-[5-(4-Bromo-2-methyl-2H-pyrazol-3- 1 - [5 -(4-Bromo-2-methyl-2H-pyrazol- yl)-4'-methoxy-biphenyl-3-yl]-3-(4- 3 -yl)-4'-methoxy-biphenyl-3 -yl] -3 - trifluoromethyl-phenyl)-urea (2,4-dichloro-phenyl)-urea
Figure imgf000176_0003
Compound 200 Compound 198
1 -[5-(4-Bromo-2-mefhyl-2H- 1 -[5-(4-Bromo-2-methyl-2H- pyrazol-3-yl)-4'-methoxy-biphenyl- pyrazol-3-yl)-4'-methoxy-biphenyl-
3-yl]-3-(4-bromo-phenyl)-urea 3 -yl]-3 -(4-methoxy-phenyl)-urea
Figure imgf000177_0001
Compound 196 Compound 201
1 -[5-(4-Bromo-2-methyl-2H- 1 -[5-(4-Bromo-2-methyl-2H- pyrazol-3-yl)-4'-methoxy-biphenyl- pyrazol-3-yl)-4'-methoxy-biphenyl-
3 -yl] -3 -(4-cyano-phenyl)-urea 3-yl]-3-(4-isopropyl-phenyl)-urea
Figure imgf000177_0002
Compound 185 Compound 202 l-[5-(4-Bromo-2-methyl-2H-pyrazol- l-[5-(4-Bromo-2-methyl-2H-pyrazol-
3-yl)-4'-methoxy-biphenyl-3-yl]-3- 3 -yl)-4'-methoxy-biphenyl-3 -yl]-3 -
(4-dimethylamino-phenyl)-urea (2-phenyl-cyclopropyl)-urea
TABLE 16-1 Compounds ofthe invention containing ^fluorophenyl functionality.
Figure imgf000177_0003
Compound 199 Compound 204
1 -[5-(4-Bromo-2-methyl-2H- 1 -[5-(4-Bromo-2-methyl-2H- pyrazol-3-yl)-4'-fluoro-biphenyl-3- pyrazol-3 -yl)-4'-fluoro-biphenyl-3 - yl]-3-(4-chloro-phenyl)-urea yl]-3-(4-fluoro-phenyl)-urea
Figure imgf000177_0004
Compound 190
1 -[5-(4-Bromo-2-methyl-2H- l-[5-(4-Bromo-2-methyl-2H-pyrazol- pyrazol-3 -yl)-4'-fluoro-biphenyl-3 -
3-yl)-4'-fluoro-biρhenyl-3-yl]-3-(4- yl]-3-(2,4-dichloro-phenyl)-urea trifluoromethyl-phenyl)-urea
Figure imgf000178_0001
Compound 197 Compound 188 l-[5-(4-Bromo-2-methyl-2H- 1 -[5-(4-Bromo-2-methyl-2H- pyrazol-3-yl)-4'-fluoro-biphenyl-3- pyrazol-3-yl)-4'-fluoro-biphenyl-3- yl]-3-(4-bromo-phenyl)-urea yl]-3 -(4-methoxy-phenyl)-urea
Figure imgf000178_0002
Compound 193 Compound 195
1 -[5-(4-Bromo-2-methyl-2H- 1 -[5-(4-Bromo-2-methyl-27J- pyrazol-3 -yl)-4'-fluoro-biphenyl- pyrazol-3-yl)-4'-fluoro-biphenyl-3-
3-yl]-3-(4-cyano-phenyl)-urea yl]-3-(4-isopropyl-phenyl)-urea
Figure imgf000178_0003
Compound 189 Compound 203 l-[5-(4-Bromo-2-methyl-2H-pyrazol- l-[5-(4-Bromo-2-methyl-2H-pyrazol-
3-yl)-4'-fluoro-biphenyl-3-yl]-3-(4- 3-yl)-4'-fluoro-biphenyl-3-yl]-3-(2- dimethylamino-phenyl)-urea phenyl-cyclopropyl)-urea
Experiment 16.1
Preparation and Analysis of Compound
Aryl bromide 16-1:
Figure imgf000178_0004
[0443] Aniline 14-17 (9.89 g, 39.9 mmol, Experiment 14.4 and Scheme 14-3) was dissolved in MeOΗ (150 mL) and toluene (50 mL) and refluxed to dissolve all solids. NaSΗ (6.0 g, 107.7 mrnol) in MeOH (50 mL) was added into the solution for 45 min. The reaction mixture was stirred for 30 min at the same temperature and cooled to room temperature. The reaction mixture was concenfrated under vaccum and dissolved in EtOAc (250 mL). The EtOAc was washed with H20 (100 mL x 2), dried over MgS04 and concenfrated to afford crude Aniline 14-17. The crude compound was used for the next step without further purification. The crade Aniline 14-17 was added into 45% HBr (50 mL) at 0 °C and warmed to room temperature. After stirring for 30 min, the solution was cooled to -30 °C and NaN02 (2.75 g, 39.9 mmol) added portionwise for 10 min. The reaction mixture was maintained at the same temperature for 30 min. The mixture was heated to 100 °C and stirred for 10 liiin. The reaction mixture was cooled to room temperature and quenched with crushed ice (250 g). The crude product was extracted with EtOAc (250 mL), dried over MgS0 and concenfrated under vaccum. The crude material was purified by column chromatograph (EtOAc/Hex = 1/1, Rf = 0.85) to afford the desired product as a white crystal Aryl bromide 16-1 (5.8 g, 52.1 % in 2 steps).
Experiment 16.2
Preparation and Analysis of Compound Biaryl 16-2:
Figure imgf000179_0001
[0444] Aryl bromide 16-1 (700 mg, 2.5 mmol), ^-methoxyphenyl boronic acid (380 mg, 2.5 mmol) and Pd(PPh3)4 (289 mg, 0.25mmol) were directly weighed into a 25 mL round-bottom flask and dissolved in dioxane (10 mL). The solution was treated with K2C03 (760 mg, 5.5 mmol) in H20 (2.75 mL) and heated to 80 °C. After stirring for 4 hrs, the reaction was poured into H20 (50 mL) and exfracted with EtOAc (20 mL x 2). The EtOAc was dried over MgS0 and concenfrated under vaccum. The residue was purified over silica gel (EtOAc/Hex = 1/2, Rf = 0.64) to afford Biaryl 16-2 as a white crystal (649 mg, 84.2%).: Experiment 16.3
Preparation and Analysis of Compound
Amino biaryl 16-4:
Figure imgf000180_0001
[0445] To the solution of Biaryl 16-2 (618 mg, 2.0 mmol) in dichloromethane (5 mL), was added dropwise Br2 (128 μL, 2.5 mmol) in dichloromethane (1 mL) for 5 min at 0 °C. After addition of Br2, the reaction mixture was warmed to room temperature and stirred for 2 hrs. The reaction mixture was washed with H20 (10 mL), concentrated under vacuum and friturated in EtOH to afford yellowish crystals. A 5 mL round-bottom flask was charged with the solids and SnCl2*2H20 (2.61 g, 10.0 mmol) and dissolved in EtOH (5 mL). The mixture was refluxed for 1.5 hrs and cooled to room temperature. The reaction was concenfrated under vaccum and exfracted with EtOAc (20 mL x 2) and H20 (20 mL). The EtOAc was dried over MgS04, concenfrated under vaccum and purified over silica gel (EtOAc/Hex = 1/2, Rf = 0.25) to afford the desire Amino biaryl 16-4 as white crystals (583 mg, 82% in 2 steps).
Experiment 16.4
Preparation and Analysis of Compound
Amino biaryl 16-5:
Figure imgf000180_0002
[0446] Aryl bromide 16-1 (1.28 g, 4.57 mmol), ^-fluorophenyl boronic acid (700 mg, 5.0 mmol) and Pd(PPh3)4 (578 mg, 0.5 mmol) were directly weighed into a 50 mL round-bottom flask and dissolved in dioxane (30 mL). The solution was treated with K2C03 (1.38 g, 10.0 mmol) in H20 (5.43 mL) and heated to 80 °C. After stirring for 4 hrs, the reaction was poured into H20 (50 mL) and exfracted with EtOAc (30 mL x 2). The EtOAc was dried over MgS04 and concentrated under vaccum. The residue was dissolved in dichloromethane (25 mL), and Br2 (282 μL, 5.5 mmol) in dichloromethane (3 mL) was added dropwise for 5 min at 0 °C. After addition of Br2, the reaction mixture was warmed to room temperature and stirred for 2 hrs. The reaction mixture was washed with H20 (20 mL), concenfrated under vaccum and friturated in EtOH to afford yellowish crystals. A 5 mL round-bottom flask was charged with the solids and SnCl2*2H20 (4.12 g, 18.3 mmol) and dissolved in EtOH (20 mL). The mixture was refluxed for 1.5 hrs and cooled to room temperature. The reaction was concenfrated under vaccum and exfracted with EtOAc (20 mL x 2) and H20 (20 mL). The EtOAc was dried over MgS04, concenfrated under vaccum and purified over silica gel (EtOAc/Hex = 1/2, Rf = 0.31) to afford Amino biaryl 16-5 as white crystals (1.31 g, 83 % in 3 steps).
[0447] In general, the coupling procedures disclosed herein between an amine, such as,
Amino biaryls 16-5, 16-5 and the like, with isocyanates, acid halides and sulfonyl halides can be utilized to achieve the desired urea, amide or sulfonamide products respectively. Analytical data for certain representative compounds, such as those compounds in TABLES 16-1 and 16-2, can be seen in TABLE 16-3 below:
TABLE 16-3
Analytical Data for Representative Compounds ofthe Invention
Figure imgf000181_0001
[0448] An important point that can be derived from the foregoing data is that by using a constitutively activated form of the receptor in the direct identification of candidate compounds, the selectivity of the compounds is exceptional: as those in the art appreciate, the homology between the human 5-HT2A and 5-HT2C receptors is about 95%, and even with such homology, certain of the directly identified compounds, e.g., Compounds 8 and 9 evidence a 100-fold difference in selectivity preference (as measured by IC50 values) for the 5-HT2A receptor compared with the 5-HT2C receptor. This is important for pharmaceutical compositions in that such selectivity can help to reduce side- effects associated with interaction of a drug with a non-target receptor.
[0449] Different embodiments ofthe invention will consist of different constitutively activated receptors, different expression systems, different assays, and different compounds. Those skilled in the art will understand which receptors to use with which expression systems and assay methods. All are considered within the scope ofthe teaching of this invention.
[0450] In some embodiments of each of the genera disclosed in this specification, the following compounds (a) tiirough (k), and combinations or subcombinations thereof, are included therein: (a) N-(3-(4-bromo-2-methylpyι-azol-3-yl)phenyl)(2-chlorophenyl)carboxamide;
(b)
Figure imgf000182_0001
(c) N-(3 -(4-bromo-2 )carboxamide;
Figure imgf000182_0002
(d) N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)((3-carboxy-prop-l-yl)carboxamide;
Figure imgf000182_0003
(e) N-(3-(4-bromo-2-metiιylpyrazol-3-yl)phenyl)((2-carboxy-eth-l-yl)carboxarnide;
Figure imgf000183_0001
(f) N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)((2-chloro-3-pyridyl)carboxamide;
Figure imgf000183_0002
(g) N-(3 -(4-bromo-2-methylpyrazol-3 -yl)phenyl)((3 -carboxy-pyrazin-2-yl)carboxarnide;
Figure imgf000183_0003
(h) N-(3 -(4-bromo-2-methylpyrazol-3 -yl)phenyl)((2-methyl- 1 -carboxy-prop-2- yl)carboxamide;
Figure imgf000183_0004
(i) N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)((5-chlorothiophen-2-yl)sulfonamide;
Figure imgf000183_0005
(j) N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)(((4-trifluorome1hoxy)phenyl) amino) carboxamide;
Figure imgf000183_0006
(k) N-(3-(4-bromo-2-methylpyrazol-3-yl)ρhenyl)(frichloromethyl)carboxamide.
Figure imgf000184_0001
[0451] In some embodiments of each of the general disclosed in this specification, compounds (a) through (k) above, and combinations or subcombinations thereof, are not included therein.
[0452] Those skilled in the art will recognize that various modifications, additions, substitutions, and variations to the illusfrative examples set forth herein can be made without departing from the spirit of the invention and are, therefore, considered within the scope of the invention. All documents referenced above, including, but not limited to, printed publications, and provisional and regular patent applications, are incoφorated herein by reference in their entirety. :

Claims

We claim:
A compound of Formula (I):
Figure imgf000185_0001
(I) wherein: i) Ri is H, halogens, NR5R6, OH or OR7, wherein
R5 and R are independently H, or Cι_6 alkyl, or C2.6 alkenyl, or cycloalkyl, or aryl, or CH2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, NO2, OH, OMe, OEt, CONR8R9, NR8R9, NHCOCH3, OCF3, SMe, COOR10, S03R8, SO2NR8R9, COMe, COEt, CO-lower alkyl, SCF3, CN, C2.6 alkenyl, H, halogens, C1-4 alkoxy, C3.6 cycloalkyl, C].6 alkyl, and aryl wherein each ofthe C3.6 cycloalkyl, C_ 6 alkyl, or aryl groups maybe further optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, NO2, OH, OMe, OEt, CONR8R9, NR8R9, NHCOCH3, OCF3, SMe, COOR10, S02NR8R9, SO3R10, COMe, COEt, CO-lower alkyl, SCF3, CN, C2. 6 alkenyl, H, halogens, CM alkoxy, C3.6 cycloalkyl, CI-6 alkyl, and aryl; or
R5 and Rg may form part of a 5, 6 or 7 membered cyclic structure which may be either saturated or unsaturated and that may contain up to four heteroatoms selected from O, N or S and said cyclic structure may be optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02, OH, OMe, OEt, OCF3, SMe, COOR10, SO2NR8R9, SO3R10, NHCOCH3, COEt, COMe, or halogen;
R8 and R9 are independently a H, or Cι.6 alkyl, or C2-6 alkenyl, or cycloalkyl, or aryl, or CH2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from halogen, CF3, OCF3, OEt, CC13, Me, N02, OH, OMe, SMe, COMe, CN, COOR10, SO3R10, COEt, NHCOCH3, or aryl; Rio is H or Cι.6 alkyl; R7 is H or d-6 alkyl; ii) R2 is H, sfraight chain or branched Cι.6 alkyl, C2-6 alkenyl, or cycloalkyl; iii) R3 is halogen, carboxy, CN, alkoxycarbonyl, straight chain or branched Cι-6 alkyl, C2-6 alkenyl, C2.6 alkynyl, cycloalkyl, aryl or heteroaryl, wherein each of said alkoxycarbonyl, straight chain or branched C 6 alkyl, C2.6 alkenyl, C2.s alkynyl, cycloalkyl, aryl or heteroaryl groups can be optionally substituted with up to four substituents in any position selected from OH, ORio, NR8R9, halogen, -C(p)3j or -0-C(p)3 where p is halogen, and said cycloalkyl, aryl or heteroaryl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position; iv) R4 is C1.6 alkyl, C2.6 alkenyl, or cycloalkyl; v) A is C(=O), C(=S) or S02;
Figure imgf000186_0001
L, is:
Figure imgf000186_0002
q is O or l; m is 0 or 1; n is O or l; Ri 1 and R]2 are each independently H, straight chain or branched Cι.6 alkyl, C2-6 alkenyl, or cycloalkyl;
Figure imgf000186_0003
wherein: Ri3, R14, R]5, Ri6 and Rn are each independently H, halogen, CN, NR8R9, COORio, SR10, sfraight chain or branched C].6 alkyl, C2.6 alkenyl, C2-6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl, wherein each of said alkoxycarbonyl, straight chain or branched Cι.6 alkyl, C2.6 alkenyl, C2.6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl groups can be optionally substituted with up to four substituents in any position selected from N02, OR10, NR8R9, halogen, -C(p)3, or -0-C(p)3 where p is halogen, and said cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position;
L2 is -O-Q2 wherein Q2 is straight chain or branched Cι-6 alkyl, C2-6 alkenyl, C2.6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl, wherein each of said cycloalkyl, aryl, alkylaryl, or arylalkyl groups can be optionally substituted with up to four substituents in any position selected from N02, OR7, halogen, -C(p) or -O-C(p)3 where p is halogen, or an aliphatic or aromatic heterocycle, and said cycloalkyl, aryl, alkylaryl, and arylalkyl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position; or a pharmaceutically acceptable salt.
2. The compound of claim 1 wherein B is L] , q is 1 , m is 0, and n is 0.
3. The compound of claim 1 wherein B is L q is 1, m is 1, and n is 0.
4. The compound of claim 1 wherein B is Li, q is 1, m is 0, and n is 1.
5. The compound of claim 1 wherein B is Li, q is 0, m is 0, and n is 0.
6. The compound of claim 1 wherein B is L_.
7. The compound of claim 2 wherein A is C(=0).
8. The compound of claim 3 wherein A is C(=0).
9. The compound of claim 4 wherein A is C(=0).
10. The compound of claim 5 wherein A is C(=0).
11. The compound of claim 2 wherein A is C(=S).
12. The compound of claim 3 wherein A is C(=S).
13. The compound of claim 4 wherein A is C(=S).
14. The compound of claim 5 wherein A is C(=S).
15. The compound of claim 5 wherein A is SO2.
16. The compound of claim 1 wherein Ri is H, Cl, F, dimethylamino, pyrrolidin-l-yl, moφholin-1- yl, 4-methylpiperazin-l-yl, OH or OCH3.
17. The compound of claim 1 wherem R3 is Cl, Br, I, -COOCH3, 2-hydroxyethyl, 2- (dimethylamino)ethyl, vinyl, ethyl, phenyl, 4-methoxyphenyl, 3-methoxyphenyl, 4-fluorophenyl, 4-trifluoromethoxyphenyl, thiophenyl, carboxy, cyclopropyl, -CCH, -CH=CH-CCH, or CN.
18. The compound of claim 7 wherein Ri is H, Cl, F, dimethylamino, pyrrolidin-l-yl, moφholin-1- yl, 4-methylpiperazin-l-yl, OH or OCH3.
19. The compound of claim 7 wherein R3 is Cl, Br, I, -COOCH3, 2-hydroxyethyl, 2- (dimethylamino)ethyl, vinyl, ethyl, phenyl, 4-methoxyphenyl, 3-methoxyphenyl, 4-fluorophenyl, 4-trifluoromethoxyphenyl, thiophenyl, carboxy, cyclopropyl, -CCH, -CH=CH-CCH, or CN.
20. The compound of claim 8 wherein Ri is H, Cl, F, dimethylamino, pyrrolidin-l-yl, moφholin-1- yl, 4-methylpiperazin-l-yl, OH or OCH3.
21. The compound of claim 8 wherein R3 is Cl, Br, I, -COOCH3, 2-hydroxyethyl, 2- (dimethylamino)ethyl, vinyl, ethyl, phenyl, 4-methoxyphenyl, 3-methoxyphenyl, 4-fluorophenyl, 4-trifluoromethoxyphenyl, thiophenyl, carboxy, cyclopropyl, -CCH, -CH=CH-CCH, or CN.
22. The compound of claim 9 wherein R] is H, Cl, F, dimethylamino, pyrrolidin-l-yl, moφholin-1- yl, 4-methylpiperazin- 1 -yl, OH or OCH3.
23. The compound of claim 9 wherein R3 is Cl, Br, I, -COOCH3, 2-hydroxyethyl, 2- (dimethylamino)ethyl, vinyl, ethyl, phenyl, 4-methoxyphenyl, 3-methoxyphenyl, 4-fluorophenyl, 4-trifluoromethoxyphenyl, thiophenyl, carboxy, cyclopropyl, -CCH, -CH=CH-CCH, or CN.
24. The compound of claim 1 wherein R2 is H and R4 is methyl.
25. The compound of claim 16 wherein R2 is H and R4 is methyl.
26. The compound of claim 17 wherein R2 is H and R4 is methyl.
27. The compound of claim 18 wherein R2 is H and R4 is methyl.
28. The compound of claim 19 wherein R2 is H and R4 is methyl.
29. The compound of claim 20 wherein R2 is H and R4 is methyl.
30. The compound of claim 21 wherein R2 is H and R4 is methyl.
31. The compound of claim 22 wherein R2 is H and R4 is methyl.
32. The compound of claim 23 wherein R2 is H and R4 is methyl.
33. The compound of any of claims 25 through 32 wherein R13, Rj4, Rι5, R16 and R17 are each independently H, F, Cl, Br, CN, dimethylamino, ethoxycarbonyl, methylthio, methyl, ethyl, isopropyl, trifluoromethyl, trifluoromethoxy, methoxy, NH2 or NO2.
34. The compound of claim 15 wherein R2 is H and R4 is methyl.
35. The compound of claim 34 wherein R3 is Cl, Br, I, -COOCH3, 2-hydroxyethyl, 2- (dimethylamino)ethyl, vinyl, ethyl, phenyl, 4-methoxyphenyl, 3-methoxyphenyl, 4-fluorophenyl, 4-frifluoromethoxyphenyl, thiophenyl, carboxy, cyclopropyl, -CCH, -CH=CH-CCH, or CN.
36. The compound of claim 6 wherein A is (C=0).
37. The compound of claim 36 wherein R is methyl.
38. The compound of claim 37 wherein R2 is H.
39. The compound of claim 38 wherein Qi is ethyl, 4-nitrophenyl, allyl, 4-methylphenyl, isopropyl, butyl, 2-isopropyl-5-methylcyclohexyl, benzyl, 3-bromophenyl, 4-fluorophenyl, 2- methoxyphenyl, 2-chlorophenyl, -C(CH3)=CH, l-(N-pyridyl)ethyl, or 9-fluoreneylmethyl.
40. The compound of claim 39 wherein Ri is H, Cl, F, dimethylamino, pyrrolidin-l-yl, moφholin-1- yl, 4-methylpiperazin-l-yl, OH or OCH3; and R3 is Cl, Br, I, -COOCH3, 2-hydroxyethyl, 2- (dimethylamino)ethyl, vinyl, ethyl, phenyl, 4-methoxyphenyl, 3-methoxyphenyl, 4-fluorophenyl, 4-frifluoromethoxyphenyl, thiophenyl, carboxy, cyclopropyl, -CCH, -CH=CH-CCH, or CN.
41. A compound of claim 1 selected from the group consisting of l-[3-(4-Chloro-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(4-chloro-phenyl)-urea; l-[3-Chloro-5-(4-chloro-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(4-chloro-phenyl)-urea; l-[3-(4-Chloro-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(2,4-dichloro-phenyl)-urea; l-[3-(4-Chloro-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(l-phenyl-ethyl)-urea; l-[3-(4-Chloro-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(4-cyano-phenyl)-urea; l-[3-(4-Chloro-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(4-dimethylamino-phenyl)-urea; 4-{3-[3-(4-Chloro-2-methyl-2H-pyrazol-3-yl)-phenyl]-ureido}-benzoic acid ethyl ester; l-[3-(4-Chloro-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(4-methylsulfanyl-phenyl)-urea; l-[3-(4-Chloro-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(4-isopropyl-phenyl)-urea; l-(4-Chloro-phenyl)-3-[3-(4-iodo-2-methyl-2H-pyrazol-3-yl)-phenyl]-urea; [3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]-carbamic acid ethyl ester; [3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]-carbamic acid 4-nitro-phenyl ester; [3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]-carbamic acid 9H-fluoren-9-ylmethyl ester; [3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]-carbamic acid allyl ester; [3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]-carbamic acid p-tolyl ester; [3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]-carbamic acid butyl ester; [3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]-carbamic acid 2-isopropyl-5-methyl- cyclohexyl ester; [3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]-carbamic acid benzyl ester; [3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]-carbamic acid 3-frifluoromethyl-phenyl ester; [3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]-carbamic acid 4-bromo-phenyl ester; [3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]-carbamic acid 4-fluoro-phenyl ester; [3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]-carbamic acid 2-methoxy-phenyl ester; [3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]-carbamic acid 2-chloro-phenyl ester; [3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]-carbamic acid isopropenyl ester; [3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]-carbamic acid 1-pyridin-l-yl-ethyl ester; [3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]-carbamic acid isopropyl ester; 5-{3-[3-(4-Chloro-phenyl)-ureido]-phenyl}-l-methyl-lH-pyrazole-4-carboxylic acid methyl ester; l-(4-Fluoro-phenyl)-3-{3-[4-(2-hydroxy-ethyl)-2-methyl-2H-pyrazol-3-yl]-phenyl}-urea; l-{3-[4-(2-Dimethylamino-ethyl)-2-methyl-2H-pyrazol-3-yl]-phenyl}-3-(4-fluoro- phenyl)-urea; l-(4-Chloro-phenyl)-3-[3-(2-methyl-4-vinyl-2H-pyrazol-3-yl)-phenyl]-urea; l-(4-Chloro-phenyl)-3-[3-(4-ethyl-2-methyl-2H-pyrazol-3-yl)-phenyl]-urea; l-(4-Fluoro-phenyl)-3-[3-(2-methyl-4-vinyl-2H-pyrazol-3-yl)-phenyl]-urea; l-(4-Chloro-phenyl)-3-[3-(2-methyl-4-phenyl-2H-pyrazol-3-yl)-phenyl]-urea; l-(4-Chloro-phenyl)-3-{3-[4-(4-methoxy-phenyl)-2-methyl-2H-pyrazol-3-yl]-phenyl}- urea; l-(4-Chloro-phenyl)-3-{3-[4-(3-methoxy-phenyl)-2-methyl-2H-pyrazol-3-yl]-phenyl}- urea; l-(4-Chloro-phenyl)-3-{3-[4-(4-fluoro-phenyl)-2-methyl-2H-pyrazol-3-yl]-phenyl}-urea; l-(4-Chloro-phenyl)-3-{3-[2-methyl-4-(4-frifluoromethoxy-phenyl)-2H-pyrazol-3-yl]- phenyl} -urea; l-(4-CWoro-phenyl)-3-[3-(2-methyl-4-thiophen-2-yl-2H-pyrazol-3-yl)-phenyl]-urea; 5-{3-[3-(4-Chloro-phenyl)-ureido]-phenyl}-l-methyl-lH-pyrazole-4-carboxylic acid; l-[3-(4-Cyclopropyl-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(3-trifluoromethyl-phenyl)- urea; l-(3-Chloro-phenyl)-3-[3-(4-cyclopropyl-2-methyl-2H-pyrazol-3-yl)-phenyl]-urea; 1 -[3-(4-Cyclopropyl-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(2-methoxy-phenyl)-urea; l-(4-Chloro-phenyl)-3-[3-(4-ethynyl-2-methyl-2H-pyrazol-3-yl)-phenyl]-urea; 1 -[3 -(4-But- 1 -en-3 -ynyl-2-methyl-2H-pyrazol-3 -yl)-phenyl] -3 -(4-chloro-phenyl)-urea; l-[3-(4-But-l-en-3-ynyl-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-p-tolyl-urea; l-(4-Chloro-phenyl)-3-[3-(4-cyano-2-methyl-2H-pyrazol-3-yl)-phenyl]-urea; 1 -[3 -(4-Cyano-2-methyl-2H-pyrazol-3 -yl)-phenyl] -3 -(4-isopropyl-phenyl)-urea; l-[3-(4-Cyclopropyl-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(4-fluoro-phenyl)-urea; l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-fluoro-phenyl]-3-(4-chloro-phenyl)-urea; N-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-fluoro-phenyl]-4-fluoro- benzenesulfonamide; l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-fluoro-phenyl]-3-(4-bromo-phenyl)-urea; l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-fluoro-phenyl]-3-(4-isopropyl-phenyl)-urea; l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-fluoro-phenyl]-3-(4-fluoro-phenyl)-urea; l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-fluoro-phenyl]-3-(4-methoxy-phenyl)-urea; l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-fluoro-phenyl]-3-(4-cyano-phenyl)-urea; N-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-fluoro-phenyl]-4-chloro- benzenesulfonamide; l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-fluoro-phenyl]-3-(2,4-dichloro-phenyl)- urea; l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-dimethylamino-phenyl]-3-(4-bromo- phenyl)-urea; l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-pyrrolidin-l-yl-phenyl]-3-(l-phenyl-ethyl)- urea; l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-moφholin-4-yl-phenyl]-3-(2,4-dichloro- phenyl)-urea; N-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-pyrrolidin-l-yl-phenyl]-benzamide; l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-pyrrolidin-l-yl-phenyl]-3-(4-bromo- phenyl)-urea; l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-pyrrolidin-l-yl-phenyl]-3-(4- dimethylamino-phenyl)-urea; l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-moφholin-4-yl-phenyl]-3-(4-isopropyl- phenyl)-urea; 85 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-(4-methyl-piperazin-l-yl)-phenyl]-3-(4-
86 bromo-phenyl)-urea;
87 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-dimethylamino-phenyl]-3-(4-chloro-
88 phenyl)-urea;
89 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-(4-methyl-piperazin-l-yl)-phenyl]-3-(2-
90 trifluoromethoxy-phenyl)-urea;
91 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-pyrrolidin-l-yl-phenyl]-3-(4-methoxy-
92 phenyl)-thiourea;
93 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-(4-methyl-piperazin-l-yl)-phenyl]-3-(4-
94 isopropyl-phenyl)-urea;
95 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-(4-methyl-piperazin-l-yl)-phenyl]-3-(4-
96 chloro-phenyl)-urea;
97 N-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-pyrrolidin-l-yl-phenyl]-4-fluoro-
98 benzenesulfonamide;
99 N-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-dimethylamino-phenyl]-4-chloro-
100 benzenesulfonamide;
101 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-moφholin-4-yl-phenyl]-3-(4-fluoro-
102 phenyl)-urea;
103 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-moφholin-4-yl-phenyl]-3-(4-chloro-
104 phenyl)-urea;
105 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-pyrrolidin-l-yl-phenyl]-3-(2,4-dichloro-
106 phenyl)-urea;
107 1 -[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-dimethylamino-phenyl]-3-(4-
108 dimethylamino-phenyl)-urea;
109 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-(4-methyl-piperazin-l-yl)-phenyl]-3-(4-
110 fluoro-phenyl)-urea;
111 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-pyrrolidin-l-yl-phenyl]-3-(4-methoxy-
112 phenyl)-urea;
113 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-moφholin-4-yl-phenyl]-3-(2-phenyl-
114 cyclopropyl)-urea;
115 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-(4-methyl-piperazin-l-yl)-phenyl]-3-(4-
116 cyano-phenyl)-urea; 117 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-moφholin-4-yl-phenyl]-3-(4-cyano-phenyl)-
118 urea;
119 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-dimethylamino-phenyl]-3-(4-fluoro-phenyl)-
120 urea;
121 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-dimethylamino-phenyl]-3-(4-isopropyl-
122 phenyl)-urea;
123 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-pyrrolidin-l-yl-phenyl]-3-(4-fluoro-phenyl)-
124 urea;
125 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-(4-methyl-piperazin-l-yl)-phenyl]-3-(2-
126 phenyl-cyclopropyl)-urea;
127 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-(4-methyl-piperazin-l-yl)-phenyl]-3-(2,4-
128 dichloro-phenyl)-urea;
129 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-moφholin-4-yl-phenyl]-3-(4-
130 trifluoromethyl-phenyl)-urea;
131 1 -(3 -Amino-4-fluoro-phenyl)-3 - [3 -(4-bromo-2-methyl-2H-pyrazol-3 -yl)-5-pyrrolidin- 1 -
132 yl-phenyl]-urea;
133 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-dimethylamino-phenyl]-3-(4-methoxy-
134 phenyl)-urea;
135 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-pyrrolidin-l-yl-phenyl]-3-(4-fluoro-3-nitro-
136 phenyl)-urea;
137 1 -[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-dimethylamino-phenyl]-3-(4-cyano-phenyl)-
138 urea;
139 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-pyrrolidin-l-yl-phenyl]-3-(l-phenyl-ethyl)-
140 urea;
141 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-pyrrolidin-l-yl-phenyl]-3-(2-phenyl-
142 cyclopropyl)-urea;
143 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-pyrrolidin-l-yl-phenyl]-3-(4-chloro-phenyl)-
144 urea;
145 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-moφholin-4-yl-phenyl]-3-(4-bromo-
146 phenyl)-urea;
147 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-pyrrolidin-l-yl-phenyl]-3-(4-cyano-phenyl)-
148 urea; 149 N-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-dimethylamino-phenyl]-4-fluoro-
150 benzenesulfonamide;
151 N-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-pyrrolidin-l-yl-phenyl]-
152 benzenesulfonamide;
153 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-moφholin-4-yl-phenyl]-3-(4-methoxy-
154 phenyl)-urea;
155 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-(4-methyl-piperazin-l-yl)-phenyl]-3-(4-
156 dimethylamino-phenyl)-urea;
157 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-pyrrolidin-l-yl-phenyl]-3-(4-isopropyl-
158 phenyl)-urea;
159 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-methoxy-phenyl]-3-(4-fluoro-phenyl)-urea;
160 1 -[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-hydiOxy-phenyl]-3-(3,5-dichloro-phenyl)-
161 urea;
162 1 -[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-hydroxy-phenyl]-3-(4-fluoro-phenyl)-urea;
163 N-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-methoxy-phenyl]-4-chloro-
164 benzenesulfonamide;
165 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-methoxy-phenyl]-3-(4-dimethylamino-
166 phenyl)-urea;
167 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-methoxy-phenyl]-3-(4-bromo-phenyl)-urea;
168 N-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-hydroxy-phenyl]-4-chloro-
169 benzenesulfonamide;
170 1 -[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-methoxy-phenyl]-3-(3,5-dichloro-phenyl)-
171 urea;
172 1 -[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-hydroxy-phenyl]-3-(4-chloro-phenyl)-urea;
173 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-methoxy-phenyl]-3-(4-chloro-phenyl)-urea;
174 1 -[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-methoxy-phenyl]-3-(2,4-dichloro-phenyl)-
175 urea;
176 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-pyrrolidin-l-yl-phenyl]-3-(4-
177 trifluoromethyl-phenyl)-urea;
178 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-hydroxy-phenyl]-3-(4-methoxy-phenyl)-
179 urea;
180 1 -[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-methoxy-phenyl]-3-(4-methoxy-phenyl)-
181 urea; 182 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-hydroxy-phenyl]-3-(4-cyano-phenyl)-urea 183 and 184 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(4-chloro-phenyl)-thiourea.
1 42. A compound of claim 1 selected from the group consisting of:
2 1 -[3 -(4-Bromo-2-methyl-2H-pyrazo: 3-yl)-phenyl]-3-(3,4-difluoro-phenyl)-urea;
3 l-[3-(4-Bromo-2-methyl-2H-pyrazo: 3-yl)-phenyl]-3-(2,5-difluoro-phenyl)-urea;
4 1 -[3 -(4-Bromo-2-methyl-2H-pyrazo: 3-yl)-phenyl]-3-(3,5-difluoro-phenyl)-urea;
5 l-[3-(4-Bromo-2-methyl-2H-pyrazoJ -3-yl)-phenyl]-3-(3-chloro-4-fluoro-phenyl)-urea;
6 l-[3-(4-Bromo-2-methyl-2H-pyrazo: •3-yl)-phenyl]-3-(4-fluoro-2-trifluoromethyl-
7 phenyl)-urea;
8 l-[3-(4-Bromo-2-methyl-2H-pyrazo; -3 -yl)-phenyl]-3 -(4-fluoro-3 -trifluoromethyl-
9 phenyl)-urea;
10 l-[3-(4-Chloro-2-methyl-2H-pyrazα -3-yl)-phenyl]-3-(2,5-difluoro-phenyl)-urea;
11 l-[3-(4-Bromo-2-methyl-2H-pyrazo: -3-yl)-phenyl]-3-(2,3,4-trifluoro-phenyl)-urea;
12 l-[3-(4-Chloro-2-methyl-2H-pyrazo: -3-yl)-phenyl]-3-(3,5-difluoro-phenyl)-urea;
13 l-(3-Chloro-4-fluoro-phenyl)-3-[3-(4-chloro-2-methyl-2H-pyrazol-3-yl)-phenyl]-urea;
14 l-[3-(4-Chloro-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(2,4-difluoro-phenyl)-urea;
15 l-[3-(4-Chloro-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(2,3,4-trifluoro-phenyl)-urea;
16 l-[3-(4-Chloro-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(4-fluoro-2-trifluoromethyl-
17 phenyl)-urea;
18 l-[3-(4-Chloro-2-methyl-2H-pyrazol •3-yl)-phenyl]-3-(4-chloro-2-trifluoromethyl-
19 phenyl)-urea;
20 l-[3-(4-Bromo-2-methyl-2H-pyrazol ■3-yl)-phenyl]-3-(4-chloro-3-frifluoromethyl-
21 phenyl)-urea
22 and
23 l-[3-(4-Chloro-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(4-fluoro-3-trifluoromethyl-phenyl)-urea.
1 43. The compound of claim 1 selected from the group consisting of:
2 N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)(4-methoxyphenoxy)carboxamide;
3 N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)(2-thienyl)carboxamide;
4 N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)(((4-trifluoromethoxy)
5 phenyl)amino)carboxamide; N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)(((4-trifluoromethoxy) phenyl)methyl)amino)carboxamide; ((3 -(4-bromo-2-methylpyrazol-3 -yl)phenyl)amino)-N-( 1 , 1 -dimethylethoxy)carboxamide; N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)(4-trifluoromethoxyphenyl) carboxamide; N-(3 -(4-bromo-2-methylpyrazol-3 -yl)phenyl)(4-chlorophenyl) carboxamide; N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)-2-(4-(frifluoromethoxy) phenyl)acetamide; N-(3 ~(4-bromo-2-methylpyrazol-3 -yl)phenyl)-2-(3 -fluorophenyl) acetamide; N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)-2-(3-methoxyphenyl) acetamide; N-(3 -(4-bromo-2-methylpyrazol-3-yl)phenyl)-2-(2-fluorophenyl) acetamide; N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)-2-(4-nitrophenyl)acetamide; N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)-2-(2-methoxyphenyl) acetamide; N-(3 -(4-bromo-2-methylpyrazol-3 -yl)phenyl)((4-methylthiophenyl)amino) carboxamide; N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)((4-chlorophenyl)amino) carboxamide; ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(4-fluorophenyl) carboxamide; ((3 -(4-bromo-2-methylpyrazol-3 -yl)phenyl)amino)-N-(2- (trifluoromethoxy)phenyl)carboxamide; ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(2-nitrophenyl) carboxamide; ((3 -(4-bromo-2-methylpyrazol-3 -yl)phenyl)amino)-N-(4-methoxyphenyl) carboxamide; ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(2-methylphenyl) carboxamide; ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(4-(trifluoromethyl) phenyl)carboxamide; ((3 -(4-bromo-2-methylpyrazol-3 -yl)phenyl)amino)-N-(3 -chlorophenyl) carboxamide; ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(2-chlorophenyl) carboxamide; ((3 -(4-bromo-2-methylpyrazol-3 -yl)phenyl)amino)-N-(4-(methylethyl) phenyl)carboxamide; ((3 -(4-bromo-2-methylpyrazol-3 -yl)phenyl)amino)-N-(3 -methoxyphenyl) carboxamide; ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(3-methylphenyl) carboxamide; ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-methyl-N-(4- (trifluoromethoxy)phenyl)-carboxamide; N-(4-(tert-butyl)phenyl)((3-(4-bromo-2-methylpyrazol-3-yl)phenyl) amino)carboxamide; N-(4-(dimethylamino)phenyl)((3-(4-bromo-2-methylpyrazol-3- yl)phenyl)amino)carboxamide; N-(3,5-dichloro-4-methylphenyl)((3-(4-bromo-2-methylpyrazol-3- yl)phenyl)amino)carboxamide; ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(4- (trifluoromethylthio)phenyl)carboxamide; ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(cyclohexyl) carboxamide; ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(phenylmethyl) carboxamide; ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(2-fluorophenyl) carboxamide; 2-(((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)-amino)carbonylamino) benzamide; ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(4-cyanophenyl) carboxamide; ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(2-cyanophenyl) carboxamide; ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(4- fluorophenylmethyl)carboxamide; ((3 -(4-bromo-2-methylpyrazol-3 -yl)phenyl)amino)-N-(3 ,4- dimethoxyphenylmethyl)carboxamide; ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(3,4,5- trimethoxyphenylmethyl)carboxamide; N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)-(((2-methylphenyl)methyl) amino)carboxamide; ((3 -(4-bromo-2-methylpyrazol-3 -yl)phenyl)amino)-N-(4- methoxyphenylmethyl)carboxamide and ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(2-(4-methoxy)phenylethyl)carboxamide.
44. The compound of claim 1 selected from the group consisting of: l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(2-chloro-4-trifluoromethyl- phenyl)-urea; l-(3,4-Bis-trifluoromethyl-phenyl)-3-[3-(4-bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]- urea; 3 -[3 -(4-Bromo-2-methyl-2H-pyrazol-3 -yl)-phenyl]- 1 -(4-fluoro-phenyl)- 1 -methyl-urea; l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(3-fluoro-4-trifluoromethyl- phenyl)-urea; 3-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]-l-(4-chloro-phenyl)-l-methyl-urea; 3-[3-(4-Chloro-2-methyl-2H-pyrazol-3-yl)-phenyl]-l-(4-chloro-phenyl)-l-methyl-urea and
3-[3-(4-Chloro-2-methyl-2H-pyrazol-3-yl)-phenyl]-l-(4-fluoro-phenyl)-l-methyl-urea.
45. A compound of Formula (I) :
Figure imgf000199_0001
(I) wherein: i) Ri is H, halogens, NR5Rs, OH or OR7, wherein
R5 and R$ are independently H, or Cι.β alkyl, or C2.6 alkenyl, or cycloalkyl, or aryl, or CH2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02, OH, OMe, OEt, CONR8R9, NR8R9, NHCOCH3, OCF3, SMe, COOR10, S03R8, S02NR8R9, COMe, COEt, CO-lower alkyl, SCF3, CN, C2.6 alkenyl, H, halogens, CM alkoxy, C3.6 cycloalkyl, C]-6 alkyl, and aryl wherein each ofthe C3.6 cycloalkyl, C1-6 alkyl, or aryl groups maybe further optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02, OH, OMe, OEt, CONR8R9, NR8R9, NHCOCH3, OCF3, SMe, COOR10, SO2NR8R9, SO3R10, COMe, COEt, CO-lower alkyl, SCF3, CN, C2. 6 alkenyl, H, halogens, CM alkoxy, C3.6 cycloalkyl, C1-6 alkyl, and aryl; or
R5 and R^ may form part of a 5, 6 or 7 membered cyclic structure which may be either saturated or unsaturated and that may contain up to four heteroatoms selected from O, N or S and said cyclic structure may be optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, NO2, OH, OMe, OEt, OCF3, SMe, COOR10) S02NR8R9, SO3R,0, NHCOCH3, COEt, COMe, or halogen;
R8 and R9 are independently a H, or Cι.6 alkyl, or C2.6 alkenyl, or cycloalkyl, or aryl, or CH2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from halogen, CF3, OCF3, OEt, CC13, Me, NO2, OH, OMe, SMe, COMe, CN, COOR10, SO3R10, COEt, NHCOCH3, or aryl;
Rio is H or Cι-6 alkyl; R7 is H or Cw alkyl; ii) R2 is H, straight chain or branched Cι.6 alkyl, C2-6 alkenyl, or cycloalkyl; iii) R3 is halogen, carboxy, CN, alkoxycarbonyl, straight chain or branched Cι.β alkyl, C2-6 alkenyl, C2-6 alkynyl, cycloalkyl, aryl or heteroaryl, wherein each of said alkoxycarbonyl, straight chain or branched Cι.6 alkyl, C2.6 alkenyl, C2-s alkynyl, cycloalkyl, aryl or heteroaryl groups can be optionally substituted with up to four substituents in any position selected from OH, ORio, NR8R9, halogen, -C(p)3> or -0-C(p)3 where p is halogen, and said cycloalkyl, aryl or heteroaryl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position; iv) R4 is Cι-6 alkyl, C2.6 alkenyl, or cycloalkyl; v) A is C(=O), C(=S) or SO2;
Figure imgf000200_0001
q is O or l; m is O or l; n is O or l; Rn and Rι2 are each independently H, straight chain or branched C1.6 alkyl, C2-6 alkenyl, or cycloalkyl;
Figure imgf000200_0002
wherein: R13, Rι , R15, Ri6 and R17 are each independently H, halogen, CN, NR8R9, COORio, SRio, sfraight chain or branched Cι.6 alkyl, C2.6 alkenyl, C2.6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl, wherein each of said alkoxycarbonyl, straight chain or branched C].6 alkyl, C2.s alkenyl, C2-6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl groups can be optionally substituted with up to four substituents in any position selected from N02, ORι0, R8R9, halogen, -C(p)3, or -O-C(p)3 where p is halogen, and said cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position; I^ is -O-Q2 wherein Q2 is straight chain or branched C].6 alkyl, C2.6 alkenyl, C2.6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl, wherein each of said cycloalkyl, aryl, alkylaryl, or arylalkyl groups can be optionally substituted with up to four substituents in any position selected from N02, OR7, halogen, -C(p)3> or -0-C(p)3 where p is halogen, or an aliphatic or aromatic heterocycle, and said cycloalkyl, aryl, alkylaryl, and arylalkyl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position; or a pharmaceutically acceptable salt; provided that the compound is not any ofthe following 43 compounds: N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)(4-methoxyphenoxy)carboxamide; N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)(2-thienyl)carboxamide; N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)(((4-trifluoromethoxy) phenyl)amino)carboxamide; N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)(((4-trifluoromethoxy) phenyl)methyl)amino)carboxamide; ((3 -(4-bromo-2-methylpyrazol-3 -yl)phenyl)amino)-N-( 1 , 1 -dimethylethoxy)carboxamide; N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)(4-trifluoromethoxyphenyl) carboxamide; N-(3 -(4-bromo-2-methylpyrazol-3 -yl)phenyl)(4-chlorophenyl) carboxamide; N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)-2-(4-(frifluoromethoxy) phenyl)acetamide; N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)-2-(3-fluorophenyl) acetamide; N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)-2-(3-methoxyphenyl) acetamide; N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)-2-(2-fluorophenyl) acetamide; N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)-2-(4-nitrophenyl)acetamide; N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)-2-(2-methoxyphenyl) acetamide; N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)((4-methylthiophenyl)amino) carboxamide; N-(3 -(4-bromo-2-methylpyrazol-3-yl)phenyl)((4-chlorophenyl)amino) carboxamide; ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(4-fluorophenyl) carboxamide; ((3 -(4-bromo-2-methylpyrazol-3 -yl)phenyl)amino)-N-(2- (trifluoromethoxy)phenyl)carboxamide; ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(2-nitrophenyl) carboxamide; ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(4-methoxyphenyl) carboxamide; 88 ((3 -(4-bromo-2-methylpyr azol-3 -yl)phenyl)amino)-N-(2-methylphenyl) carboxamide;
89 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(4-(trifluoromethyl)
90 phenyl)carboxamide;
91 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(3-chlorophenyl) carboxamide;
92 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(2-chlorophenyl) carboxamide;
93 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(4-(methylethyl)
94 ρhenyl)carboxamide;
95 ((3 -(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(3 -methoxyphenyl) carboxamide;
96 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(3-methylphenyl) carboxamide;
97 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-methyl-N-(4-
98 (trifluoromethoxy)phenyl)-carboxamide;
99 N-(4-(tert-butyl)phenyl)((3-(4-bromo-2-methylpyrazol-3-yl)phenyl) amino)carboxamide;
100 N-(4-(dimethylamino)phenyl)((3-(4-bromo-2-methylpyrazol-3-
101 yl)phenyl)amino)carboxamide;
102 N-(3,5-dichloro-4-methylphenyl)((3-(4-bromo-2-methylpyrazol-3-
103 yl)phenyl)amino)carboxamide;
104 ((3 -(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(4-
105 (trifluoromethylthio)phenyl)carboxamide;
106 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(cyclohexyl) carboxamide;
107 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(phenylmethyl) carboxamide;
108 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(2-fluorophenyl) carboxamide;
109 2-(((3 -(4-bromo-2-methylpyrazol-3 -yl)phenyl)-amino)carbonylamino) benzamide;
110 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(4-cyanophenyl) carboxamide;
111 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(2-cyanophenyl) carboxamide;
112 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(4-
113 fluorophenylmethyl)carboxamide;
114 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(3,4-
115 dimethoxyphenylmethyl)carboxamide;
116 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(3,4,5-
117 trimethoxyphenylmethyl)carboxamide;
118 N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)-(((2-methylphenyl)methyl)
119 amino)carboxamide; 120 ((3 -(4-bromo-2-methylpyrazol-3 -yl)phenyl)amino)-N-(4-
121 methoxyphenylmethyl)carboxamide
122 and
123 ((3 -(4-bromo-2-methylpyrazol-3 -yl)phenyl)amino)-N-(2-(4-methoxy)phenylethyl)carboxamide.
46. The compound of claim 45 wherein B is Lh q is 1, m is 0, and n is 0.
47. The compound of claim 45 wherein B is L^ q is 1, m is 1, and n is 0.
48. The compound of claim 45 wherein B is Li, q is 1, m is 0, and n is 1.
49. The compound of claim 45 wherein B is Lj, q is 0, m is 0, and n is 0.
50. The compound of claim 45 wherein B is L_.
51. The compound of claim 46 wherein A is C(=0.
52. The compound of claim 47 wherem A is C(=0).
53. The compound of claim 48 wherein A is C(=0).
54. The compound of claim 49 wherein A is C(=0).
55. The compound of claim 46 wherein A is C(=S).
56. The compound of claim 47 wherein A is C(=S).
57. The compound of claim 48 wherein A is C(=S).
58. The compound of claim 49 wherein A is C(=S).
59. The compound of claim 49 wherein A is S02.
60. The compound of claim 45 wherein Ri is H, Cl, F, dimethylamino, pyrrolidin-l-yl, moφholin-1- yl, 4-methylpiperazin-l-yl, OH or OCH3.
61. The compound of claim 45 wherein R3 is Cl, Br, I, -COOCH3, 2-hydroxyethyl, 2- (dimethylamino)ethyl, vinyl, ethyl, phenyl, 4-methoxyphenyl, 3-methoxyphenyl, 4-fluorophenyl, 4-trifluoromethoxyphenyl, thiophenyl, carboxy, cyclopropyl, -CCH, -CH=CH-CCH, or CN.
62. The compound of claim 51 wherein Ri is H, Cl, F, dimethylamino, pyrrolidin-l-yl, moφholin-1- yl, 4-methylpiperazin-l-yl, OH or OCH3.
63. The compound of claim 51 wherein R3 is Cl, Br, I, -COOCH3, 2-hydroxyethyl, 2- (dimethylamino)ethyl, vinyl, ethyl, phenyl, 4-methoxyphenyl, 3-methoxyphenyl, 4-fluorophenyl, 4-trifluoromethoxyphenyl, thiophenyl, carboxy, cyclopropyl, -CCH, -CH=CH-CCH, or CN.
64. The compound of claim 52 wherein Ri is H, Cl, F, dimethylamino, pyrrolidin-l-yl, moφholin-1- yl, 4-methylpiperazin-l-yl, OH or OCH3.
65. The compound of claim 52 wherein R3 is Cl, Br, I, -COOCH3, 2-hydroxyethyl, 2- (dimethylamino)ethyl, vinyl, ethyl, phenyl, 4-methoxyphenyl, 3-methoxyphenyl, 4-fluorophenyl, 4-trifluoromethoxyphenyl, thiophenyl, carboxy, cyclopropyl, -CCH, -CH=CH-CCH, or CN.
66. The compound of claim 53 wherein Ri is H, Cl, F, dimethylamino, pyrrolidin-l-yl, moφholin-1- yl, 4-methylpiperazin-l-yl, OH or OCH3.
67. The compound of claim 53 wherein R3 is Cl, Br, I, -COOCH3, 2-hydroxyethyl, 2- (dimethylamino)ethyl, vinyl, ethyl, phenyl, 4-methoxyphenyl, 3-methoxyphenyl, 4-fluorophenyl, 4-trifluoromethoxyphenyl, thiophenyl, carboxy, cyclopropyl, -CCH, -CH=CH-CCH, or CN.
68. The compound of claim 45 wherein R2 is H and R4 is methyl.
69. The compound of claim 60 wherein R2 is H and R4 is methyl.
70. The compound of claim 61 wherein R2 is H and R4 is methyl.
71. The compound of claim 62 wherein R2is H and R is methyl.
72. The compound of claim 63 wherein R2 is H and R4 is methyl.
73. The compound of claim 64 wherein R2 is H and R4 is methyl.
74. The compound of claim 65 wherein R2 is H and R4 is methyl.
75. The compound of claim 66 wherein R2 is H and R4 is methyl.
76. The compound of claim 67 wherein R2 is H and R4 is methyl.
77. The compound of any of claims 69 through 76 wherein Rio, Rn, Rι2, Ri3 and R]4are each independently H, F, Cl, Br, CN, dimethylamino, ethoxycarbonyl, methylthio, methyl, ethyl, isopropyl, trifluoromethyl, trifluoromethoxy, methoxy, NH2 or N02.
78. The compound of claim 59 wherein R2 is H and R4 is methyl.
79. The compound of claim 78 wherein R3 is Cl, Br, I, -COOCH3, 2-hydroxyethyl, 2- (dimethylamino)ethyl, vinyl, ethyl, phenyl, 4-methoxyphenyl, 3-methoxyphenyl, 4-fluorophenyl, 4-trifluoromethoxyphenyl, thiophenyl, carboxy, cyclopropyl, -CCH, -CH=CH-CCH, or CN.
80. The compound of claim 50 wherein A is (C=0).
81. The compound of claim 80 wherein 4 is methyl.
82. The compound of claim 81 wherein R2 is H.
83. The compound of claim 82 wherein Q is ethyl, 4-nitrophenyl, allyl, 4-methylphenyl, isopropyl, butyl, 2-isopropyl-5-methylcyclohexyl, benzyl, 3-bromophenyl, 4-fluorophenyl, 2- methoxyphenyl, 2-chlorophenyl, -C(CH3)=CH, l-(N-pyridyl)ethyl, or 9-fluoreneylmethyl.
84. The compound of claim 83 wherein Ri is H, Cl, F, dimethylamino, pyrrolidin-l-yl, moφholin-1- yl, 4-methylpiperazin-l-yl, OH or OCH3; and R3 is Cl, Br, I, -COOCH3, 2-hydroxyethyl, 2- (dimethylamino)ethyl, vinyl, ethyl, phenyl, 4-methoxyphenyl, 3-methoxyphenyl, 4-fluorophenyl, 4-frifluoromethoxyphenyl, thiophenyl, carboxy, cyclopropyl, -CCH, -CH=CH-CCH, or CN.
85. A compound of claim 45 selected from the group consisting of: l-[3-(4-Chloro-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(4-chloro-phenyl)-urea; l-[3-Chloro-5-(4-chloro-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(4-chloro-phenyl)-urea; l-[3-(4-Chloro-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(2,4-dichloro-phenyl)-urea; l-[3-(4-Chloro-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(l-phenyl-ethyl)-urea; l-[3-(4-Chloro-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(4-cyano-phenyl)-urea; l-[3-(4-Chloro-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(4-dimethylamino-phenyl)-urea;
4-{3-[3-(4-Chloro-2-methyl-2H-pyrazol-3-yl)-phenyl]-ureido}-benzoic acid ethyl ester; l-[3-(4-Chloro-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(4-methylsulfanyl-phenyl)-urea; l-[3-(4-Chloro-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(4-isopropyl-phenyl)-urea; l-(4-Chloro-phenyl)-3-[3-(4-iodo-2-methyl-2H-pyrazol-3-yl)-phenyl]-urea;
[3 -(4-Bromo-2-methyl-2H-pyrazo •3-yl)-phenyf -carbamic acid ethyl ester; [3 -(4-Bromo-2-methyl-2H-pyrazo •3-yl)-phenyl -carbamic acid 4-nitro-phenyl ester; [3 -(4-Bromo-2-methyl-2H-pyrazo: 3-yl)-phenyl -carbamic acid 9/J-fluoren-9-ylmethyl ester;
[3 -(4-Bromo-2-methyl-2H-pyrazo: •3-yl)-phenyI -carbamic acid allyl ester; [3 -(4-Bromo-2-methyl-2H-pyrazo: ■3-yl)-phenyl -carbamic acid p-tolyl ester; [3-(4-Bromo-2-methyl-2H-pyrazo: 3-yl)-phenyl -carbamic acid butyl ester; [3 -(4-Bromo-2-methyl-2H-pyrazo: 3-yl)-phenyI -carbamic acid 2-isopropyl-5-methyl- cyclohexyl ester;
[3 -(4-Bromo-2-methyl-2H-pyrazo: ■3-yl)-phenyl -carbamic acid benzyl ester; [3 -(4-Bromo-2-methyl-2H-pyrazo: •3-yl)-phenyl -carbamic acid 3-frifluoromethyl-phenyl ester;
[3 -(4-Bromo-2-methyl-2H-pyrazo: ■3-yl)-phenyI -carbamic acid 4-bromo-phenyl ester; [3 -(4-Bromo-2-methyl-2H-pyrazo' •3-yl)-phenyI -carbamic acid 4-fluoro-phenyl ester; [3-(4-Bromo-2-methyl-2H-pyrazo: ■3-yl)-phenyl -carbamic acid 2-methoxy-phenyl ester; [3 -(4-Bromo-2-methyl-2H-pyrazo: 3-yl)-phenyI •carbamic acid 2-chloro-phenyl ester; [3 -(4-Bromo-2-methyl-2H-pyrazo: 3-yl)-phenyI -carbamic acid isopropenyl ester; [3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]-carbamic acid 1-pyridin-l-yl-ethyl ester; [3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]-carbamic acid isopropyl ester; 5-{3-[3-(4-Chloro-phenyl)-ureido]-phenyl}-l-methyl-lH-pyrazole-4-carboxylic acid methyl ester; 1 -(4-Fluoro-phenyl)-3- {3-[4-(2-hydroxy-ethyl)-2-methyl-2H-pyrazol-3-yl]-phenyl} -urea; l-{3-[4-(2-Dimethylamino-ethyl)-2-methyl-2H-pyrazol-3-yl]-phenyl}-3-(4-fluoro- phenyl)-urea; l-(4-Chloro-phenyl)-3-[3-(2-methyl-4-vinyl-2H-pyrazol-3-yl)-phenyl]-urea; l-(4-Chloro-phenyl)-3-[3-(4-ethyl-2-methyl-2H-pyrazol-3-yl)-phenyl]-urea; l-(4-Fluoro-phenyl)-3-[3-(2-methyl-4-vinyl-2H-pyrazol-3-yl)-phenyl]-urea; l-(4-Chloro-phenyl)-3-[3-(2-methyl-4-phenyl-2H-pyrazol-3-yl)-phenyl]-urea; l-(4-Chloro-phenyl)-3-{3-[4-(4-methoxy-phenyl)-2-methyl-2H-pyrazol-3-yl]-phenyl}- urea; 1 -(4-Chloro-phenyl)-3- {3-[4-(3-methoxy-phenyl)-2-methyl-2H-pyrazol-3-yl]-phenyl} - urea; l-(4-Chloro-phenyl)-3-{3-[4-(4-fluoro-phenyl)-2-methyl-2H-pyrazol-3-yl]-phenyl}-urea; 1 -(4-Chloro-phenyl)-3- {3-[2-methyl-4-(4-trifluoromethoxy-phenyl)-2H-pyrazol-3-yl]- phenyl} -urea; l-(4-Chloro-phenyl)-3-[3-(2-methyl-4-thiophen-2-yl-2H-pyrazol-3-yl)-phenyl]-urea; 5-{3-[3-(4-Chloro-phenyl)-ureido]-phenyl}-l-methyl-lH-pyrazole-4-carboxylic acid; 1 -[3 -(4-Cyclopropyl-2-methyl-2H-pyrazol-3 -yl)-phenyl]-3 -(3 -trifluoromethyl-phenyl)- urea; l-(3-Chloro-phenyl)-3-[3-(4-cyclopropyl-2-methyl-2H-pyrazol-3-yl)-phenyl]-urea; l-[3-(4-Cyclopropyl-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(2-methoxy-phenyl)-urea; l-(4-Chloro-phenyl)-3-[3-(4-ethynyl-2-methyl-2H-pyrazol-3-yl)-phenyl]-urea; l-[3-(4-But-l-en-3-ynyl-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(4-chloro-phenyl)-urea; l-[3-(4-But-l-en-3-ynyl-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-p-tolyl-urea; l-(4-Chloro-phenyl)-3-[3-(4-cyano-2-methyl-2H-pyrazol-3-yl)-phenyl]-urea; l-[3-(4-Cyano-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(4-isopropyl-phenyl)-urea; l-[3-(4-Cyclopropyl-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(4-fluoro-phenyl)-urea; l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-fluoro-phenyl]-3-(4-chloro-phenyl)-urea; N-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-fluoro-phenyl]-4-fluoro- benzenesulfonamide; l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-fluoro-phenyl]-3-(4-bromo-phenyl)-urea; l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-fluoro-phenyl]-3-(4-isopropyl-phenyl)-urea; l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-fluoro-phenyl]-3-(4-fluoro-phenyl)-urea; l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-fluoro-phenyl]-3-(4-methoxy-phenyl)-urea; l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-fluoro-phenyl]-3-(4-cyano-phenyl)-urea; N-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-fluoro-phenyl]-4-chloro- benzenesulfonamide; l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-fluoro-phenyl]-3-(2,4-dichloro-phenyl)- urea; l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-dimethylamino-phenyl]-3-(4-bromo- phenyl)-urea; l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-pyrrolidin-l-yl-phenyl]-3-(l-phenyl-ethyl)- urea; l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-moφholin-4-yl-phenyl]-3-(2,4-dichloro- phenyl)-urea; N-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-pyrrolidin-l-yl-phenyl]-benzamide; l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-pyrrolidin-l-yl-phenyl]-3-(4-bromo- phenyl)-urea; l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-pyrrolidin-l-yl-phenyl]-3-(4- dimethylamino-phenyl)-urea; l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-moφholin-4-yl-phenyl]-3-(4-isopropyl- phenyl)-urea; l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-(4-methyl-piperazin-l-yl)-phenyl]-3-(4- bromo-phenyl)-urea; l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-dimethylamino-phenyl]-3-(4-chloro- phenyl)-urea; l-[3-(4-Bromo-2-me%l-2H-pyrazol-3-yl)-5-(4-methyl-piperazin-l-yl)-phenyl]-3-(2- trifluoromethoxy-phenyl)-urea; l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-pyrrolidin-l-yl-phenyl]-3-(4-methoxy- phenyl)-thiourea; 93 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-(4-methyl-piperazin-l-yl)-phenyl]-3-(4-
94 isopropyl-phenyl)-urea;
95 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-(4-methyl-piperazin-l-yl)-phenyl]-3-(4-
96 chloro-phenyl)-urea;
97 N-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-pyrrolidin-l-yl-phenyl]-4-fluoro-
98 benzenesulfonamide;
99 N-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-dimethylamino-phenyl]-4-chloro-
100 benzenesulfonamide;
101 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-moφholin-4-yl-phenyl]-3-(4-fluoro-
102 phenyl)-urea;
103 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-moφholin-4-yl-phenyl]-3-(4-chloro-
104 phenyl)-urea;
105 1 -[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-pyrrolidin- 1 -yl-phenyl]-3-(2,4-dichloro-
106 phenyl)-urea;
107 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-dimethylamino-phenyl]-3-(4-
108 dimethylamino-phenyl)-urea;
109 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-(4-methyl-piperazin-l-yl)-phenyl]-3-(4-
110 fluoro-phenyl)-urea;
111 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-pyrrolidin-l-yl-phenyl]-3-(4-methoxy-
112 phenyl)-urea;
113 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-moφholin-4-yl-phenyl]-3-(2-phenyl-
114 cyclopropyl)-urea;
115 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-(4-methyl-piperazin-l-yl)-phenyl]-3-(4-
116 cyano-phenyl)-urea;
117 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-moφholin-4-yl-phenyl]-3-(4-cyano-phenyl)-
118 urea;
119 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-dimethylamino-phenyl]-3-(4-fluoro-phenyl)-
120 urea;
121 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-dimethylamino-phenyl]-3-(4-isopropyl-
122 phenyl)-urea;
123 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-pyrrolidin-l-yl-phenyl]-3-(4-fluoro-phenyl)-
124 urea; 125 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-(4-methyl-piperazin-l-yl)-phenyl]-3-(2-
126 phenyl-cyclopropyl)-urea;
127 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-(4-methyl-piperazin-l-yl)-phenyl]-3-(2,4-
128 dichloro-phenyl)-urea;
129 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-moφholin-4-yl-phenyl]-3-(4-
130 trifluoromethyl-phenyl)-urea;
131 l-(3-Amino-4-fluoro-phenyl)-3-[3-(4-bromo-2-methyl-2H-pyrazol-3-yl)-5-pyrrolidin-l-
132 yl-phenyl]-urea;
133 1 -[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-dimethylamino-phenyl]-3-(4-methoxy-
134 phenyl)-urea;
135 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-pyrrolidin-l-yl-phenyl]-3-(4-fluoro-3-nitro-
136 phenyl)-urea;
137 1 -[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-dimethylamino-phenyl]-3-(4-cyano-phenyl)-
138 urea;
139 1 -[3 -(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-pyrrolidin- 1 -yl-phenyl]-3-( 1 -phenyl-ethyl)-
140 urea;
141 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-pyrrolidin-l-yl-phenyl]-3-(2-phenyl-
142 cyclopropyl)-urea;
143 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-pyrrolidin-l-yl-phenyl]-3-(4-chloro-phenyl)-
144 urea;
145 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-moφholin-4-yl-phenyl]-3-(4-bromo-
146 phenyl)-urea;
147 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-pyrrolidin-l-yl-phenyl]-3-(4-cyano-phenyl)-
148 urea;
149 N-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-dimethylamino-phenyl]-4-fluoro-
150 benzenesulfonamide;
151 N-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-pyrrolidin- 1-yl-phenyl]-
152 benzenesulfonamide;
153 1 -[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-moφholin-4-yl-phenyl]-3-(4-methoxy-
154 phenyl)-urea;
155 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-(4-methyl-piperazin-l-yl)-phenyl]-3-(4-
156 dimethylamino-phenyl)-urea; 157 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-pyrrolidin-l-yl-phenyl]-3-(4-isopropyl-
158 phenyl)-urea;
159 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-methoxy-phenyl]-3-(4-fluoro-phenyl)-urea;
160 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-hydroxy-phenyl]-3-(3,5-dichloro-phenyl)-
161 urea;
162 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-hydroxy-phenyl]-3-(4-fluoro-phenyl)-urea;
163 N-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-methoxy-phenyl]-4-chloro-
164 benzenesulfonamide;
165 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-methoxy-phenyl]-3-(4-dimethylamino-
166 phenyl)-urea;
167 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-methoxy-phenyl]-3-(4-bromo-phenyl)-urea;
168 N-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-hydroxy-phenyl]-4-chloro-
169 benzenesulfonamide;
170 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-methoxy-phenyl]-3-(3,5-dichloro-phenyl)-
171 urea;
172 1 -[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-hydroxy-phenyl]-3-(4-chloro-phenyl)-urea;
173 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-methoxy-phenyl]-3-(4-chloro-phenyl)-urea;
174 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-methoxy-phenyl]-3-(2,4-dichloro-phenyl)-
175 urea;
176 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-pyrrolidin-l-yl-phenyl]-3-(4-
177 trifluoromethyl-phenyl)-urea;
178 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-hydroxy-phenyl]-3-(4-methoxy-phenyl)-
179 urea;
180 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-methoxy-phenyl]-3-(4-methoxy-phenyl)-
181 urea;
182 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-5-hydroxy-phenyl]-3-(4-cyano-phenyl)-urea
183 and
184 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(4-chloro-phenyl)-thiourea.
1 86. A compound of claim 45 selected from the group consiting of:
2 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(3,4-difluoro-phenyl)-urea;
3 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(2,5-difluoro-phenyl)-urea;
4 l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(3,5-difluoro-phenyl)-urea; l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(3-chloro-4-fluoro-phenyl)-urea; l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(4-fluoro-2-trifluoromethyl- phenyl)-urea; l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(4-fluoro-3-trifluoromethyl- phenyl)-urea; l-[3-(4-Chloro-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(2,5-difluoro-phenyl)-urea; 1 -[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(2,3,4-trifluoro-phenyl)-urea; l-[3-(4-Chloro-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(3,5-difluoro-phenyl)-urea; 1 -(3-Chloro-4-fluoro-phenyl)-3-[3-(4-chloro-2-methyl-2H-pyrazol-3-yl)-phenyl]-urea; l-[3-(4-Chloro-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(2,4-difluoro-phenyl)-urea; 1 -[3-(4-Chloro-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(2,3,4-frifluoro-phenyl)-urea; 1 -[3-(4-Chloro-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(4-fluoro-2-trifluoromethyl- phenyl)-urea; 1 -[3-(4-Chloro-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(4-chloro-2-trifluoromethyl- phenyl)-urea; l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(4-chloro-3-trifluoromethyl- phenyl)-urea and l-[3-(4-Chloro-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(4-fluoro-3-trifluoromethyl-phenyl)-urea.
87. A compound of claim 45 selected from the group consisting of: l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(2-chloro-4-trifluoromethyl- phenyl)-urea; l-(3,4-Bis-trifluoromethyl-phenyl)-3-[3-(4-bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]- urea; 3-[3 -(4-Bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]- 1 -(4-fluoro-phenyl)- 1 -methyl-urea; l-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]-3-(3-fluoro-4-trifluoromethyl- phenyl)-urea; 3-[3-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-phenyl]-l-(4-chloro-phenyl)-l-methyl-urea; 3-[3-(4-Chloro-2-methyl-2H-pyrazol-3-yl)-phenyl]-l-(4-chloro-phenyl)-l-methyl-urea and 3 -[3 -(4-Chloro-2-methyl-2H-pyrazol-3 -yl)-phenyl]- 1 -(4-fluoro-phenyl)- 1 -methyl-urea. A compound of Formula (XV):
Figure imgf000213_0001
(XV) wherein: i) Ar is a phenyl ring optionally substituted with up to five groups selected from the group consisting of halogen, OR7, OH, NR8R9, carboxy, CN, alkoxycarbonyl, sfraight chain or branched .6 alkyl -C(p)3j or -0-C(p)3 where p is halogen; R8 and R9 are independently a H, or Cι._ alkyl, or C2.6 alkenyl, or cycloalkyl, or aryl, or CH2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from halogen, CF3, OCF3, OEt, CC13, Me, N02, OH, OMe, SMe, COMe, CN, COOR]0, SO3R10, COEt, NHCOCH3, or aryl; or R8 and R9 may form part of a 5, 6 or 7 membered cyclic structure which may be either saturated or unsaturated and that may contain up to four heteroatoms selected from O, N or S and said cyclic structure may be optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02, OH, OMe, OEt, OCF3, SMe, COOR!0, SO2NR8R9, SO3R,o, NHCOCH3, COEt, COMe, or halogen; R7 is H or C,.6 alkyl; Rio is H or C1.6 alkyl; ii) R2 is H, straight chain or branched C].6 alkyl, C2.6 alkenyl, or cycloalkyl; iii) R3 is halogen, carboxy, CN, alkoxycarbonyl, straight chain or branched C1.6 alkyl, C2-6 alkenyl, C2.6 alkynyl, cycloalkyl, aryl or heteroaryl, wherein each of said alkoxycarbonyl, straight chain or branched Cι.6 alkyl, C2.6 alkenyl, C2.6 alkynyl, cycloalkyl, aryl or heteroaryl groups can be optionally substituted with up to four substituents in any position selected from OH, OR10, NR8R9, halogen, -C(p)3, or -O-C(p)3 where p is halogen, and said cycloalkyl, aryl or heteroaryl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position; iv) R is Ci„6 alkyl, C2.6 alkenyl, or cycloalkyl; v) A is C(=O), C(=S) or S02; vi) B is L, or I__;
Figure imgf000214_0001
q is O or l; m is 0 or 1; n is O or l; Ri i and Rj2 are each independently H, straight chain or branched Cι.6 alkyl, C2-6 alkenyl, or cycloalkyl; Q, is:
Figure imgf000214_0002
wherein: Rn, Rι , R15, Ri6 and Rn are each independently H, halogen, CN, NR8R9, COORio, SRio, sfraight chain or branched Cι.6 alkyl, C2-6 alkenyl, C2.6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl, wherein each of said alkoxycarbonyl, straight chain or branched .6 alkyl, C2.6 alkenyl, C2.6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl groups can be optionally substituted with up to four substituents in any position selected from NO2, ORio, NR8R9, halogen, -C(p)3j or -0-C(p)3 where p is halogen, and said cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position;
L_ is -O-Q2 wherem Q2 is straight chain or branched -β alkyl, C2-6 alkenyl, C2.6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl, wherein each of said cycloalkyl, aryl, alkylaryl, or arylalkyl groups can be optionally substituted with up to four substituents in any position selected from NO2, OR7, halogen, -C(p)3> or -O-C(p)3 where p is halogen, or an aliphatic or aromatic heterocycle, and said cycloalkyl, aryl, alkylaryl, and arylalkyl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position; or a pharmaceutically acceptable salt.
89. The compound of claim 88 wherein B is Lb q is 1, m is 0, and n is 0.
90. The compound of claim 88 wherein B is Lh q is 1, m is 1, and n is 0.
91. The compound of claim 88 wherein B is Li , q is 1 , m is 0, and n is \,
92. The compound of claim 88 wherein B is Lj, q is 0, m is 0, and n is 0.
93. The compound of claim 88 wherein B is L2.
94. The compound of claim 89 wherein A is C(=0).
95. The compound of claim 90 wherein A is C(=0).
96. The compound of claim 91 wherein A is C(=0).
97. The compound of claim 92 wherein A is C(=0).
98. The compound of claim 89 wherein A is C(=S).
99. The compound of claim 90 wherein A is C(=S).
100. The compound of claim 91 wherein A is C(=S).
101. The compound of claim 92 wherein A is C(=S) .
102. The compound of claim 92 wherem A is SO2.
103. The compound of claim 88 wherein Ar is 4-methoxyphenyl or 4-fluorophenyl.
104. The compound of claim 88 wherein R3 is Cl, Br, I, -COOCH3, 2-hydroxyethyl, 2- (dimethylamino)ethyl, vinyl, ethyl, phenyl, 4-methoxyphenyl, 3-methoxyphenyl, 4-fluorophenyl, 4-trifluoromethoxyphenyl, thiophenyl, carboxy, cyclopropyl, -CCH, -CH=CH-CCH, or CN.
105. The compound of claim 94 wherein Ar is 4-methoxyphenyl or 4-fluorophenyl.
106. The compound of claim 94 wherein R3 is Cl, Br, I, -COOCH3, 2-hydroxyethyl, 2- (dimethylamino)ethyl, vinyl, ethyl, phenyl, 4-methoxyphenyl, 3-methoxyphenyl, 4-fluorophenyl, 4-trifluoromethoxyphenyl, thiophenyl, carboxy, cyclopropyl, -CCH, -CH=CH-CCH, or CN.
107. The compound of claim 95 wherein Ar is 4-methoxyphenyl or 4-fluorophenyl.
108. The compound of claim 95 wherein R3 is Cl, Br, I, -COOCH3, 2-hydroxyethyl, 2- (dimethylamino)ethyl, vinyl, ethyl, phenyl, 4-methoxyphenyl, 3-methoxyphenyl, 4-fluorophenyl, 4-trifluoromethoxyphenyl, thiophenyl, carboxy, cyclopropyl, -CCH, -CH=CH-CCH, or CN.
109. The compound of claim 96 wherein Ar is 4-methoxyphenyl or 4-fluorophenyl.
110. The compound of claim 96 wherein R3 is Cl, Br, I, -COOCH3, 2-hydroxyethyl, 2- (dimethylamino)ethyl, vinyl, ethyl, phenyl, 4-methoxyphenyl, 3-methoxyphenyl, 4-fluorophenyl, 4-trifluoromethoxyphenyl, thiophenyl, carboxy, cyclopropyl, -CCH, -CH=CH-CCH, or CN.
111. The compound of claim 88 wherein R2 is H and R4 is methyl.
112. The compound of claim 103 wherein R2 is H and R4 is methyl.
113. The compound of claim 104 wherein R2 is H and R4 is methyl.
114. The compound of claim 105 wherein R2is H and R4 is methyl.
115. The compound of claim 106 wherein R2 is H and R4 is methyl.
116. The compound of claim 107 wherem R2 is H and R4 is methyl.
117. The compound of claim 108 wherein R2 is H and R4 is methyl.
118. The compound of claim 109 wherein R2 is H and R4 is methyl.
119. The compound of claim 110 wherein R2 is H and R4 is methyl.
120. The compound of any of claims 112 through 119 wherein R[3, R] , Rι5, Rj6 and R are each independently H, F, Cl, Br, CN, dimethylamino, ethoxycarbonyl, methylthio, methyl, ethyl, isopropyl, trifluoromethyl, trifluoromethoxy, methoxy, NH2 or N02.
121. The compound of claim 102 wherein R2 is H and R4 is methyl.
122. The compound of claim 121 wherein R3 is Cl, Br, I, -COOCH3, 2-hydroxyethyl, 2- (dimethylamino)ethyl, vinyl, ethyl, phenyl, 4-methoxyphenyl, 3-methoxyphenyl, 4-fluorophenyl, 4-frifluoromethoxyphenyl, thiophenyl, carboxy, cyclopropyl, -CCH, -CH=CH-CCH, or CN.
123. The compound of claim 93 wherein A is (C=0).
124. The compound of claim 123 wherein R4 is methyl.
125. The compound of claim 124 wherein R2 is H.
126. The compound of claim 125 wherein Qi is ethyl, 4-nifrophenyl, allyl, 4-methylphenyl, isopropyl, butyl, 2-isopropyl-5-methylcyclohexyl, benzyl, 3-bromophenyl, 4-fluorophenyl, 2- methoxyphenyl, 2-chlorophenyl, -C(CH3)=CH, l-(N-pyridyl)ethyl, or 9-fluoreneylmethyl.
127. The compound of claim 126 wherein Ar is 4-methoxyphenyl or 4-fluorophenyl; and R3 is Cl, Br, I, -COOCH3, 2-hydroxyethyl, 2-(dimethylamino)ethyl, vinyl, ethyl, phenyl, 4-methoxyphenyl, 3- methoxyphenyl, 4-fluorophenyl, 4-trifluoromethoxyphenyl, thiophenyl, carboxy, cyclopropyl, - CCH, -CH=CH-CCH, or CN.
128. The compound of claim 88 selected from the group consiting of: l-[5-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-4'-methoxy-biphenyl-3-yl]-3-(4-chloro- phenyl)-urea; l-[5-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-4'-methoxy-biphenyl-3-yl]-3-(4-fluoro- phenyl)-urea; l-[5-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-4'-methoxy-biphenyl-3-yl]-3-(4- trifluoromethyl-phenyl)-urea; l-[5-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-4'-methoxy-biphenyl-3-yl]-3-(2,4-dichloro- phenyl)-urea; l-[5-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-4'-methoxy-biphenyl-3-yl]-3-(4-bromo- phenyl)-urea; l-[5-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-4'-methoxy-biphenyl-3-yl]-3-(4-methoxy- phenyl)-urea; l-[5-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-4'-methoxy-biphenyl-3-yl]-3-(4-cyano- phenyl)-urea; 1 -[5-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-4'-methoxy-biphenyl-3-yl]-3-(4-isopropyl- phenyl)-urea; l-[5-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-4'-methoxy-biphenyl-3-yl]-3-(4- dimethylamino-phenyl)-urea; l-[5-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-4'-methoxy-biphenyl-3-yl]-3-(2-phenyl- cyclopropyl)-urea; l-[5-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-4'-fluoro-biphenyl-3-yl]-3-(4-chloro-phenyl)- urea; l-[5-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-4'-fluoro-biphenyl-3-yl]-3-(4-fluoro-phenyl)- urea; l-[5-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-4'-fluoro-biphenyl-3-yl]-3-(4-trifluoromethyl- phenyl)-urea; l-[5-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-4'-fluoro-biphenyl-3-yl]-3-(2,4-dichloro- phenyl)-urea; l-[5-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-4'-fluoro-biphenyl-3-yl]-3-(4-bromo-phenyl)- urea. l-[5-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-4'-fluoro-biphenyl-3-yl]-3-(4-methoxy- phenyl)-urea; l-[5-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-4'-fluoro-biphenyl-3-yl]-3-(4-cyano-phenyl)- urea; l-[5-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-4'-fluoro-biphenyl-3-yl]-3-(4-isopropyl- phenyl)-urea; l-[5-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-4'-fluoro-biphenyl-3-yl]-3-(4-dimethylamino- phenyl)-urea and l-[5-(4-Bromo-2-methyl-2H-pyrazol-3-yl)-4'-fluoro-biphenyl-3-yl]-3-(2-phenyl-cyclopropyl)- urea.
A method for modulating the activity of a human 5-HT2A serotonm receptor by contacting the receptor with a compound of Formula (I):
Figure imgf000219_0001
(I) wherein: i) Ri is H, halogens, NR5R6, OH or OR7, wherein R5 and R^ are independently H, or Cι.6 alkyl, or C2.6 alkenyl, or cycloalkyl, or aryl, or CH2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from CF3, CCI3, Me, NO2, OH, OMe, OEt, CONR8R9, NR8R9, NHCOCH3, OCF3, SMe, COORio, S03R8, SO2NR8R9, COMe, COEt, CO-lower alkyl, SCF3, CN, C2.6 alkenyl, H, halogens, CM alkoxy, C3-6 cycloalkyl, C1.6 alkyl, and aryl wherein each ofthe C3.6 cycloalkyl, Cι.6 alkyl, or aryl groups may be further optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02, OH, OMe, OEt, CONR8R9, NR8R9, NHCOCH3, OCF3, SMe, COORio, S02NR8R9, SO3R10, COMe, COEt, CO-lower alkyl, SCF3, CN, C2- 6 alkenyl, H, halogens, C alkoxy, C3.6 cycloalkyl, Cι-6 alkyl, and aryl; or R5 and Rg may form part of a 5, 6 or 7 membered cyclic structure wliich may be either saturated or unsaturated and that may contain up to four heteroatoms selected from O, N or S and said cyclic structure may be optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02, OH, OMe, OEt, OCF3, SMe, COORio, S02NR8R9, SO3R10, NHCOCH3, COEt, COMe, or halogen;
R8 and R9 are independently a H, or C_._ alkyl, or C2-6 alkenyl, or cycloalkyl, or aryl, or CH2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from halogen, CF3, OCF3, OEt, CC13, Me, NO2) OH, OMe, SMe, COMe, CN, COORio, SO30, COEt, NHCOCH3, or aryl;
Rio is H or Cι.6 alkyl;
R7 is H or C1.6 alkyl; ϋ) R2 is H, straight chain or branched Cι.6 alkyl, C2.6 alkenyl, or cycloalkyl; ϋi) R3 is halogen, carboxy, CN, alkoxycarbonyl, straight chain or branched Cι.s alkyl, C2.6 alkenyl, C2.6 alkynyl, cycloalkyl, aryl or heteroaryl, wherein each of said alkoxycarbonyl, straight chain or branched Cι-6 alkyl, C2.6 alkenyl, C2.6 alkynyl, cycloalkyl, aryl or heteroaryl groups can be optionally substituted with up to four substituents in any position selected from OH, OR]0, NR8R9, halogen, -C(p)3, or -O-C(p)3 where p is halogen, and said cycloalkyl, aryl or heteroaryl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position; iv) R4 is C1.6 alkyl, C2.6 alkenyl, or cycloalkyl;
V) A is C(=O), C(=S) or SO2;
Figure imgf000220_0001
q is O or l; m is O or l; n is O or l; Ri 1 and Rι2 are each independently H, straight chain or branched Cj.6 alkyl, C2.6 alkenyl, or cycloalkyl;
Figure imgf000220_0002
wherein: R13, R14, Ris, R-16 and R are each independently H, halogen, CN, NR8R9, COORio, SRio, straight chain or branched Cι.6 alkyl, C2.6 alkenyl, C2-6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl, wherein each of said alkoxycarbonyl, straight chain or branched Cι-6 alkyl, C2.6 alkenyl, C2-6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl groups can be optionally substituted with up to four substituents in any position selected from N02, ORio, NR8R9, halogen, -C(p)3> or -O-C(p)3 where p is halogen, and said cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position;
L2 is -0-Q2 wherein Q2 is sfraight chain or branched .6 alkyl, C2-6 alkenyl, C2.6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl, wherein each of said cycloalkyl, aryl, alkylaryl, or arylalkyl groups can be optionally substituted with up to four substituents in any position selected from N02, OR7, halogen, -C(p) or -0-C(p)3 where p is halogen, or an aliphatic or aromatic heterocycle, and said cycloalkyl, aryl, alkylaryl, and arylalkyl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position; or a pharmaceutically acceptable salt.
A method for modulating the activity of a human 5-HT2A serotonin receptor by contacting the receptor with a compound of Formula (I) :
Figure imgf000221_0001
(I) wherein: i) Ri is H, halogens, NR5R6, OH or OR7, wherein ' R5 and R^ are independently H, or Cι.6 alkyl, or C2.β alkenyl, or cycloalkyl, or aryl, or CH2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02, OH, OMe, OEt, C0NR8R9, NR8R9, NHCOCH3, OCF3, SMe, COORio, S03R8, S02NR8R9, COMe, COEt, CO-lower alkyl, SCF3, CN, C2.6 alkenyl, H, halogens, C alkoxy, C3.6 cycloalkyl, Cι.6 alkyl, and aryl wherem each ofthe C3.6 cycloalkyl, Cι_6 alkyl, or aryl groups may be further optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, NO2, OH, OMe, OEt, CONR8R9, NR8R9, NHCOCH3, OCF3, SMe, COORio, S02NR8R9, SO30) COMe, COEt, CO-lower alkyl, SCF3, CN, C2- 6 alkenyl, H, halogens, CM alkoxy, C3-6 cycloalkyl, Cι-6 alkyl, and aryl; or R5 and Re may form part of a 5, 6 or 7 membered cyclic structure which may be either saturated or unsaturated and that may contain up to four heteroatoms selected from O, N or S and said cyclic structure may be optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02, OH, OMe, OEt, OCF3, SMe, COOR]0, S02NR8R9, SO3R10, NHCOCH3, COEt, COMe, or halogen; R8 and R9 are independently a H, or Ci .5 alkyl, or C2.6 alkenyl, or cycloalkyl, or aryl, or CH2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from halogen, CF3, OCF3, OEt, CC13, Me, N02, OH, OMe, SMe, COMe, CN, COORio, SO3R10, COEt, NHCOCH3, or aryl; R10 is H or C].6 alkyl; R7 is H or Cι-6 alkyl; ii) R2 is H, straight chain or branched Cι-6 alkyl, C2.6 alkenyl, or cycloalkyl; iii) R3 is halogen, carboxy, CN, alkoxycarbonyl, straight chain or branched .6 alkyl, C2.6 alkenyl, C2.6 alkynyl, cycloalkyl, aryl or heteroaryl, wherein each of said alkoxycarbonyl, sfraight chain or branched Cι-6 alkyl, C2.6 alkenyl, C2.β alkynyl, cycloalkyl, aryl or heteroaryl groups can be optionally substituted with up to four substituents in any position selected from OH, ORι0, NR8R9, halogen, -C(p)3, or -O-C(p)3 where p is halogen, and said cycloalkyl, aryl or heteroaryl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position; iv) R4 is Cι-6 alkyl, C2-6 alkenyl, or cycloalkyl; v) A is C(=O), C(=S) or S02;
Figure imgf000222_0001
L, is:
Figure imgf000222_0002
q is O or 1; m is O or l; n is O or l; Rn and Rj2 are each independently H, straight chain or branched Cι-6 alkyl, C2.β alkenyl, or cycloalkyl; Q, is:
Figure imgf000223_0001
wherein: Ri3, Rn, R15, Ris and Rn are each independently H, halogen, CN, NR8R9, COORio, Rio, sfraight chain or branched d-β alkyl, C2.6 alkenyl, C2.6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl, wherein each of said alkoxycarbonyl, straight chain or branched Cι-6 alkyl, C2.6 alkenyl, C2.6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl groups can be optionally substituted with up to four substituents in any position selected from NO2, ORio, NR8R9, halogen, -C(p) or -0-C(p)3 where p is halogen, and said cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position; L_ is -O-Q2 wherein Q2 is sfraight chain or branched Cι.6 alkyl, C2_6 alkenyl, C2-6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl, wherein each of said cycloalkyl, aryl, alkylaryl, or arylalkyl groups can be optionally substituted with up to four substituents in any position selected from N02, OR7, halogen, -C(p)3i or -0-C(p)3 where p is halogen, or an aliphatic or aromatic heterocycle, and said cycloalkyl, aryl, alkylaryl, and arylalkyl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position; or a pharmaceutically acceptable salt; provided that the compound is not any ofthe following 43 compounds: N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)(4-methoxyphenoxy)carboxamide; N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)(2-thienyl)carboxamide; N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)(((4-trifluoromethoxy) phenyl)amino)carboxamide; 71 N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)(((4-trifluoromethoxy)
72 phenyl)methyl)amino)carboxamide;
73 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(l,l-dimethylethoxy)carboxamide;
74 N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)(4-trifluoromethoxyphenyl) carboxamide;
75 N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)(4-chlorophenyl) carboxamide;
76 N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)-2-(4-(trifluoromethoxy) phenyl)acetamide;
77 N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)-2-(3-fluorophenyl) acetamide;
78 N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)-2-(3-methoxyphenyl) acetamide;
79 N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)-2-(2-fluorophenyl) acetamide;
80 N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)-2-(4-nitrophenyl)acetamide;
81 N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)-2-(2-methoxyphenyl) acetamide;
82 N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)((4-methylthiophenyl)amino) carboxamide;
83 N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)((4-chlorophenyl)amino) carboxamide;
84 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(4-fluorophenyl) carboxamide;
85 ((3 -(4-bromo-2-methylpyrazol-3 -yl)phenyl)amino)-N-(2-
86 (frifluoromethoxy)phenyl)carboxamide;
87 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(2-nitrophenyl) carboxamide;
88 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(4-methoxyphenyl) carboxamide;
89 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(2-methylphenyl) carboxamide;
90 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(4-(trifluoromethyl)
91 phenyl)carboxamide;
92 ((3 -(4-bromo-2-methylpyrazol-3 -yl)phenyl)amino)-N-(3 -chlorophenyl) carboxamide;
93 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(2-chlorophenyl) carboxamide;
94 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(4-(methylethyl)
95 phenyl)carboxamide;
96 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(3-methoxyphenyl) carboxamide;
97 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(3-methylphenyl) carboxamide;
98 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-methyl-N-(4-
99 (frifluoromethoxy)phenyl)-carboxamide;
100 N-(4-(tert-butyl)phenyl)((3-(4-bromo-2-methylpyrazol-3-yl)phenyl) amino)carboxamide;
101 N-(4-(dimethylamino)phenyl)((3 -(4-bromo-2-methylpyrazol-3 -
102 yl)phenyl)amino)carboxamide; 103 N-( ,5-dichloro-4-methylphenyl)((3-(4-bromo-2-methylpyrazol-3-
104 yl)phenyl)amino)carboxamide;
105 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(4-
106 (trifluoromethylthio)phenyl)carboxamide;
107 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(cyclohexyl) carboxamide;
108 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(phenylmethyl) carboxamide;
109 ((3 -(4-bromo-2-methylpyrazol-3 -yl)phenyl)amino)-N-(2-fluorophenyl) carboxamide;
110 2-(((3 -(4-bromo-2-methylpyrazol-3 -yl)phenyl)-amino)carbonylamino) benzamide;
111 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(4-cyanophenyl) carboxamide;
112 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(2-cyanophenyl) carboxamide;
113 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(4-
114 fluorophenylmethyl)carboxamide;
115 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(3,4-
116 dimethoxyphenylmethyl)carboxamide;
117 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(3,4,5-
118 trimethoxyphenylmethyl)carboxamide;
119 N-(3-(4-bromo-2-methylpyrazol-3-yl)phenyl)-(((2-methylphenyl)methyl)
120 amino)carboxamide;
121 ((3 -(4-bromo-2-methylpyrazol-3 -yl)phenyl)amino)-N-(4-
122 methoxyphenylmethyl)carboxamide
123 and
124 ((3-(4-bromo-2-methylpyrazol-3-yl)phenyl)amino)-N-(2-(4-
125 methoxy)phenylethyl)carboxamide.
1 131. A method for modulating the activity of a human 5-HT2A serotonin receptor by contacting the
2 receptor with a compound of Formula (XV) :
Figure imgf000225_0001
3 . (XV)
4 wherein: i) Ar is a phenyl ring optionally substituted with up to five groups selected from the group consisting of halogen, OR7, OH, NR8R9, carboxy, CN, alkoxycarbonyl, straight chain or branched Cι_6 alkyl -C(p)3> or -O-C(p)3 where p is halogen;
R8 and R9 are independently a H, or Cι-6 alkyl, or C2.6 alkenyl, or cycloalkyl, or aryl, or CH2 aryl group and each said group may be optionally substituted by up to four substituents in any position independently selected from halogen, CF3, OCF3, OEt, CC13, Me, N02, OH, OMe, SMe, COMe, CN, COOR,0, SO3R10, COEt, NHCOCH3, or aryl; or
R8 and R9 may form part of a 5, 6 or 7 membered cyclic structure which may be either saturated or unsaturated and that may contain up to four heteroatoms selected from O, N or S and said cyclic structure may be optionally substituted by up to four substituents in any position independently selected from CF3, CC13, Me, N02, OH, OMe, OEt, OCF3, SMe, COOR,0, S02NRsR9, SO30, NHCOCH3, COEt, COMe, or halogen;
R7 is H or Cι-6 alkyl;
Rio is H or Cι-6 alkyl; ϋ) R2 is H, straight chain or branched Cι-6 alkyl, C2.6 alkenyl, or cycloalkyl; iϋ) R3 is halogen, carboxy, CN, alkoxycarbonyl, straight chain or branched Cι.6 alkyl, C2.6 alkenyl, C2_6 alkynyl, cycloalkyl, aryl or heteroaryl, wherein each of said alkoxycarbonyl, straight chain or branched Cι-6 alkyl, C2.6 alkenyl, C2.6 alkynyl, cycloalkyl, aryl or heteroaryl groups can be optionally substituted with up to four substituents in any position selected from OH, ORio, NR8R9, halogen, -C(p)3) or -O-C(p)3 where p is halogen, and said cycloalkyl, aryl or heteroaryl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position; iv) Ri is Ci.6 alkyl, C2-6 alkenyl, or cycloalkyl;
" v) A is C(=O), C(=S) or S02;
Figure imgf000226_0001
q is O or l; m is O or l; n is O or l; Ri i and Rι2 are each independently H, straight chain or branched Cι.6 alkyl, C2.β alkenyl, or cycloalkyl;
Figure imgf000227_0001
wherein: R]3, R]4, R15, Ri and Rn are each independently H, halogen, CN, NR8R9, COORio, SRJO, straight chain or branched Cι-6 alkyl, C2.6 alkenyl, C2.β alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl, wherein each of said alkoxycarbonyl, straight chain or branched C].6 alkyl, C2.6 alkenyl, C2.6 alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl groups can be optionally substituted with up to four substituents in any position selected from NO2, ORio, NR8R9, halogen, -C(p) or -0-C(p)3 where p is halogen, and said cycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position; L2 is -O-Q2 wherein Q2 is straight chain or branched Cι.6 alkyl, C2-6 alkenyl, C2.β alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl, wherein each of said cycloalkyl, aryl, alkylaryl, or arylalkyl groups can be optionally substituted with up to four substituents in any position selected from N02, OR7, halogen, -C(p)3j or -0-C(p)3 where p is halogen, or an aliphatic or aromatic heterocycle, and said cycloalkyl, aryl, alkylaryl, and arylalkyl groups can alternatively or additionally be optionally substituted with up to four alkyl substituents in any position; or a pharmaceutically acceptable salt.
132. A pharmaceutical composition comprising a compound of any one ofthe claims 1, 41-45, 88 and 128.
133. A method for the prophylaxis or treatment of reducing platelet aggregation in a patient in need of said prophylaxis or treatment comprising administering a pharmaceutically effective amount of a compound according to any one of claims 1, 41 to 45 and 88 or a pharmaceutical composition according to claim 132.
134. A method for the prophylaxis or freatment of any one of the group of indications consisting of coronary artery disease, myocardial infarction, transient ischemic attack, angina, stroke and atrial fibrillation in a patient in need of said prophylaxis or freatment comprising administering a pharmaceutically effective amount of a compound according to any one of claims 1, 41 to 45 and 88 or a pharmaceutical composition according to claim 132.
135. A method for the prophylaxis or treatment of reducing risk of blood clot formation in an angioplasty or coronary bypass in a surgery patient in need of such prophylaxis or freatment comprising administering a pharmaceutically effective amount of a compound according to any one of claims 1, 41 to 45 and 88 or a pharmaceutical composition according to claims 132.
136. A method for the prophylaxis or treatment of reducing risk of blood clot formation in a patient suffering from atrial fibrillation comprising administering to said patient in need thereof a pharmaceutically effective amount of a compound according to any one of claims 1, 41 to 45 and 88 or a pharmaceutical composition according to claim 132.
137. A method for the prophylaxis or treatment of asthma in a patient in need of said prophylaxis or freatment comprising administering a pharmaceutically effective amount of a compound according to any one of claims 1, 41 to 45 and 88 or a pharmaceutical composition according to claim 132.
138. A method for the prophylaxis or freatment of a symptom of asthma in a patient in need of said prophylaxis or treatment comprising administering a pharmaceutically effective amount of a compound according to any one of claims 1, 41 to 45 and 88 or a pharmaceutical composition according to claim 132.
139. A method for the prophylaxis or freatment of agitation or a symptom thereof in a patient in need of said prophylaxis or treatment comprising administering a pharmaceutically effective amount of a compound according to any one of claims 1, 41 to 45 and 88 or a pharmaceutical composition according to claim 132.
140. The method of claim 139 wherein said patient is a cognitively intact elderly patient.
141. A method for the prophylaxis or freatment of agitation or a symptom thereof in a patient suffering from dementia, comprising administering to said patient in need thereof a pharmaceutically effective amount of a compound according to any one of claims 1, 41 to 45 and 88 or a pharmaceutical composition according to claim 132.
142. The method of claim 141 wherein said dementia is due to a degenerative disease ofthe nervous system.
143. The method of claim 141 wherein said dementia is Alzheimers disease, Lewy Body, Parkinson's disease, or Huntington's disease.
144. The method of claim 141 wherein said dementia is due to diseases that affect blood vessels.
145. The method of claim 141 wherein said dementia is due to stroke or multi-infarct dementia.
146. A method for the prophylaxis or treatment of a behavioral disorder, drug induced psychosis, excitative psychosis, Gilles de la Tourette's syndrome, manic disorder, organic or NOS psychosis, psychotic disorder, psychosis, acute schizophrenia, chronic schizophrenia, or NOS schizophrenia comprising administering to said patient in need thereof a dopamine D2 receptor antagonist and a according to any one of claims 1, 41 to 45 and 88 or a pharmaceutical composition according to claim 132.
147. The method of claim 146 wherein said dopamine D2 receptor antagonist is haloperidol.
148. A method for the prophylaxis or treatment of infantile autism, huntington's chorea, or nausea and vomiting from chemotherapy or chemotherapeutic antibodies comprising administering to said patient in need thereof a dopamine D2 receptor antagonist and a compound according to any one of claims 1, 41 to 45 and 88 or a pharmaceutical composition according to claim 132.
149. The method of claim 148 wherein said dopamine D2 receptor antagonist is haloperidol.
150. A method for the prophylaxis or freatment of schizophrenia in a patient, comprising administering to said patient in need thereof a dopamine D2 receptor antagonist and a compound according to any one of claims 1, 41 to 45 and 88 or a pharmaceutical composition according to claim 132.
151. The method of claim 150 wherein said dopamine D2 receptor antagonist is haloperidol.
152. A method for the prophylaxis or treatment of alleviating negative symptoms of schizophrenia induced by the adminisfration of haloperidol to a patient suffering from said schizophrenia, comprising administering to said patient in need thereof a compound according to any one of claims 1, 41 to 45 and 88 or a pharmaceutical composition according to claim 132.
153. The method of any one of claims 147, 149, 151 and 152 wherein said haloperidol and said compound are administered in separate dosage forms.
154. The method of any one of claims 147, 149, 151 and 152 wherein said haloperidol and said compound are administered in a single dosage form.
155. Use of a compound according to any one of claims 1, 41 to 45, 88 and 128 for production of a medicament for use in prophylaxis or freatment of a 5-HT2A related disorder.
156. The use according to claim 155 wherein the 5-HT2A related disorder is reducing platelet aggregation in a patient in need of said prophylaxis or treatment.
157. The use according to claim 155 wherein the 5-HT2A related disorder is any one ofthe group of indications consisting of coronary artery disease, myocardial infarction, transient ischemic attack, angina, stroke and atrial fibrillation in a patient in need of said prophylaxis or treatment.
158. The use according to claim 155 wherein the 5-HT2A related disorder is reducing risk of blood clot formation in an angioplasty or coronary bypass in a surgery patient in need of such prophylaxis or treatment.
159. The use according to claim 155 wherein the 5-HT2A related disorder is reducing risk of blood clot formation in a patient suffering from atrial fibrillation.
160. The use according to claim 155 wherein the 5-HT2A related disorder is asthma.
161. The use according to claim 155 wherein the 5-HT2A related disorder is agitation.
162. The use according to claim 155 wherein the 5-HT2A related disorder is dementia.
163. The use according to claim 155 wherein the 5-HT2A related disorder is Alzheimers disease, Lewy Body, Parkinson's disease, or Huntington's disease.
164. The use according to claim 155 wherein the 5-HT2A related disorder is a behavioral disorder, drug induced psychosis, excitative psychosis, Gilles de la Tourette's syndrome, manic disorder, organic or NOS psychosis, psychotic disorder, psychosis, acute schizophrenia, chronic schizophrenia, or NOS schizophrenia.
165. The use according to claim 155 wherein the 5-HT2A related disorder is infantile autism, huntington's chorea, or nausea and vomiting from chemotherapy or chemotherapeutic antibodies.
166. The use according to claim 155 wherein the 5-HT2A related disorder is schizophrenia.
PCT/US2003/002059 2002-01-23 2003-01-23 Small molecule modulators of the 5-ht2a serotonin receptor useful for the prophylaxis and treatment of disorders related thereto WO2003062206A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP03705889A EP1509505A2 (en) 2002-01-23 2003-01-23 SMALL MOLECULE MODULATORS OF THE 5−HT2A SEROTONIN RECEPTOR USEFUL FOR THE PROPHYLAXIS AND TREATMENT OF DISORDERS RELATED THERETO

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US38619802P 2002-01-23 2002-01-23
US60/386,198 2002-01-23
US38638402P 2002-06-05 2002-06-05
US60/386,384 2002-06-05
US40146702P 2002-08-05 2002-08-05
US60/401,467 2002-08-05

Publications (2)

Publication Number Publication Date
WO2003062206A2 true WO2003062206A2 (en) 2003-07-31
WO2003062206A3 WO2003062206A3 (en) 2004-01-08

Family

ID=27617596

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2003/002059 WO2003062206A2 (en) 2002-01-23 2003-01-23 Small molecule modulators of the 5-ht2a serotonin receptor useful for the prophylaxis and treatment of disorders related thereto

Country Status (2)

Country Link
EP (1) EP1509505A2 (en)
WO (1) WO2003062206A2 (en)

Cited By (48)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004056782A1 (en) * 2002-12-19 2004-07-08 Vernalis (Cambridge) Limited Pyrazole compounds
WO2005012254A1 (en) * 2003-07-22 2005-02-10 Arena Pharmaceuticals, Inc. Diaryl and arylheteroaryl urea derivatives as modulators of the 5-ht2a serotonin receptor useful for the prophylaxis and treatment of disorders related therto
WO2005103011A1 (en) * 2004-03-23 2005-11-03 Arena Pharmaceuticals, Inc. Processes for preparing substituted n-aryl-n’-[3-(1h-pyrazol-5-yl) phenyl] ureas and intermediates thereof
EP1615895A1 (en) * 2003-04-03 2006-01-18 Merck & Co., Inc. Biaryl substituted pyrazoles as sodium channel blockers
WO2006055734A2 (en) * 2004-11-19 2006-05-26 Arena Pharmaceuticals, Inc. 3-phenyl-pyrazole derivatives as modulators of the 5-ht2a serotonin receptor useful for the treatment of disorders related thereto
WO2006060762A2 (en) * 2004-12-03 2006-06-08 Arena Pharmaceuticals, Inc. Pyrazole derivatives as modulators of the 5-ht2a serotonin receptor useful for the treatment of disorders related thereto
WO2006078610A1 (en) * 2005-01-19 2006-07-27 Arena Pharmaceuticals, Inc. Diaryl and arylheteroaryl urea derivatives as modulators of the 5-ht2a serotonin receptor useful for the prophylaxis or treatment of progressive multifocal leukoencephalopathy
WO2006081335A2 (en) * 2005-01-26 2006-08-03 Arena Pharmaceuticals, Inc. Process for preparing substituted 1- [3- ( pyrazolyl) phenyl] -3-phenyl ureas
EP1695966A1 (en) * 2003-07-22 2006-08-30 Arena Pharmaceuticals, Inc. Diaryl and arylheteroaryl urea derivatives as modulators of the 5-ht2a serotonin receptor useful for the prophylaxis and treatment of disorders related thereto
US7217719B2 (en) 2001-12-28 2007-05-15 Acadia Pharmaceuticals Inc. Spiroazacyclic compounds as monoamine receptor modulators
US7253186B2 (en) 2002-06-24 2007-08-07 Carl-Magnus Andersson N-substituted piperidine derivatives as serotonin receptor agents
WO2007136680A2 (en) * 2006-05-18 2007-11-29 Arena Pharmaceuticals, Inc. 3-pyraz0lyl-benzamide-4-ethers, secondary amines and derivatives thereof as modulators of the 5-ht2a serotonin receptor useful for the treatment of disorders related thereto
US7476682B2 (en) 2002-06-24 2009-01-13 Acadia Pharmaceuticals, Inc. N-substituted piperidine derivatives as serotonin receptor agents
EP2019093A1 (en) * 2006-05-19 2009-01-28 Eisai R&D Management Co., Ltd. Urea type cinnamide derivative
US7538222B2 (en) 2002-06-24 2009-05-26 Acadia Pharmaceuticals, Inc. N-substituted piperidine derivatives as serotonin receptor agents
US7601740B2 (en) 2003-01-16 2009-10-13 Acadia Pharmaceuticals, Inc. Selective serotonin 2A/2C receptor inverse agonists as therapeutics for neurodegenerative diseases
JP2009537545A (en) * 2006-05-18 2009-10-29 アリーナ ファーマシューティカルズ, インコーポレイテッド Primary amines as modulators of 5-HT2A serotonin receptors useful for the treatment of disorders associated with 5-HT2A serotonin receptors, and derivatives thereof
US7732615B2 (en) 2004-09-27 2010-06-08 Acadia Pharmaceuticals Inc. N-(4-fluorobenzyl)-N-(1-methylpiperidin-4-yl)-N′-(4-(2-methylpropyloxy)phenylmethyl)carbamide and its tartrate salt and crystalline forms
US7790899B2 (en) 2004-09-27 2010-09-07 Acadia Pharmaceuticals, Inc. Synthesis of N-(4-fluorobenzyl)-N-(1-methylpiperidin-4-yl)-N′-(4-(2-methylpropyloxy)phenylmethyl)carbamide and its tartrate salt and crystalline forms
US7820695B2 (en) 2004-05-21 2010-10-26 Acadia Pharmaceuticals, Inc. Selective serotonin receptor inverse agonists as therapeutics for disease
US7935815B2 (en) 2007-08-31 2011-05-03 Eisai R&D Management Co., Ltd. Imidazoyl pyridine compounds and salts thereof
US7973033B2 (en) 2006-03-09 2011-07-05 Eisai R&D Management Co., Ltd. Multi-cyclic cinnamide derivatives
US8008293B2 (en) 2007-02-28 2011-08-30 Eisai R&D Management Co., Ltd. Bicyclic oxomorpholine derivative
US8048878B2 (en) 2005-11-24 2011-11-01 Eisai R&D Management Co., Ltd. Two cyclic cinnamide compound
US8481535B2 (en) 2006-05-18 2013-07-09 Arena Pharmaceuticals, Inc. Crystalline forms and processes for the preparation of phenyl-pyrazoles useful as modulators of the 5-HT2A serotonin receptor
WO2014149139A2 (en) 2013-03-15 2014-09-25 Verseon, Inc. Halogenopyrazoles as inhibitors of thrombin
US8980891B2 (en) 2009-12-18 2015-03-17 Arena Pharmaceuticals, Inc. Crystalline forms of certain 3-phenyl-pyrazole derivatives as modulators of the 5-HT2A serotonin receptor useful for the treatment of disorders related thereto
US9034911B2 (en) 2008-10-28 2015-05-19 Arena Pharmaceuticals, Inc. Composition of a 5-HT2A serotonin receptor modulator useful for the treatment of disorders related thereto
US9126946B2 (en) 2008-10-28 2015-09-08 Arena Pharmaceuticals, Inc. Processes useful for the preparation of 1-[3-(4-bromo-2-methyl-2H-pyrazol-3-yl)-4-methoxy-phenyl]-3-(2,4-difluoro-phenyl)urea and crystalline forms related thereto
US9156837B2 (en) 2011-07-29 2015-10-13 Takeda Pharmaceutical Company Limited Heterocyclic compound
US9296694B2 (en) 2000-03-06 2016-03-29 Acadia Pharmaceuticals Inc. Azacyclic compounds
US9434692B2 (en) 2006-10-03 2016-09-06 Arena Pharmaceuticals, Inc. Pyrazole derivatives as modulators of the 5-HT2A serotonin receptor useful for the treatment of disorders related thereto
US9453000B2 (en) 2007-08-31 2016-09-27 Eisai R&D Management Co., Ltd. Polycyclic compound
US9533970B2 (en) 2013-03-15 2017-01-03 Verseon Corporation Multisubstituted aromatic compounds as serine protease inhibitors
US9533967B2 (en) 2010-03-30 2017-01-03 Verseon Corporation Multisubstituted aromatic compounds as inhibitors of thrombin
US9556149B2 (en) 2008-04-02 2017-01-31 Arena Pharmaceuticals, Inc. Processes for the preparation of pyrazole derivatives useful as modulators of the 5-HT2A serotonin receptor
US9567327B2 (en) 2007-08-15 2017-02-14 Arena Pharmaceuticals, Inc. Imidazo[1,2-a]pyridine derivatives as modulators of the 5-HT2A serotonin receptor useful for the treatment of disorders related thereto
US10022355B2 (en) 2015-06-12 2018-07-17 Axovant Sciences Gmbh Diaryl and arylheteroaryl urea derivatives as modulators of the 5-HT2A serotonin receptor useful for the prophylaxis and treatment of REM sleep behavior disorder
US10034859B2 (en) 2015-07-15 2018-07-31 Axovant Sciences Gmbh Diaryl and arylheteroaryl urea derivatives as modulators of the 5-HT2A serotonin receptor useful for the prophylaxis and treatment of hallucinations associated with a neurodegenerative disease
US10189810B2 (en) 2014-09-17 2019-01-29 Verseon Corporation Pyrazolyl-substituted pyridone compounds as serine protease inhibitors
US10449185B2 (en) 2017-08-30 2019-10-22 Acadia Pharmaceuticals Inc. Formulations of pimavanserin
US10517860B2 (en) 2016-03-25 2019-12-31 Acadia Pharmaceuticals Inc. Combination of pimavanserin and cytochrome P450 modulators
US10532995B2 (en) 2015-02-27 2020-01-14 Verseon Corporation Substituted pyrazole compounds as serine protease inhibitors
US10953000B2 (en) 2016-03-25 2021-03-23 Acadia Pharmaceuticals Inc. Combination of pimavanserin and cytochrome P450 modulators
US10981870B2 (en) 2015-07-20 2021-04-20 Acadia Pharmaceuticals Inc. Methods for preparing N-(4-fluorobenzyl)-N-(1-methylpiperidin-4-yl)-N′-(4-(2-methylpropyloxy)phenylmethyl)carbamide and its tartrate salt and polymorphic form
US11135211B2 (en) 2017-04-28 2021-10-05 Acadia Pharmaceuticals Inc. Pimavanserin for treating impulse control disorder
US11464768B2 (en) 2016-12-20 2022-10-11 Acadia Pharmaceuticals Inc. Pimavanserin alone or in combination for use in the treatment of Alzheimer's disease psychosis
WO2023236920A1 (en) * 2022-06-07 2023-12-14 Sironax Ltd. Sarm1 modulators, preparations, and uses thereof

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2010522198A (en) 2007-03-19 2010-07-01 アカドイア プハルマセウチカルス インコーポレーテッド Combination of 5-HT2A inverse agonists and antagonists with antipsychotics

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5281571A (en) * 1990-10-18 1994-01-25 Monsanto Company Herbicidal benzoxazinone- and benzothiazinone-substituted pyrazoles
WO1998024785A1 (en) * 1996-12-02 1998-06-11 Fujisawa Pharmaceutical Co., Ltd. Indole-urea derivatives with 5-ht antagonist properties
WO1999052927A1 (en) * 1998-04-14 1999-10-21 Arena Pharmaceuticals, Inc. Non-endogenous, constitutively activated human serotonin receptors and small molecule modulators thereof
WO2002076464A1 (en) * 2001-03-22 2002-10-03 Arena Pharmaceuticals, Inc. Anti-psychosis combination

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5281571A (en) * 1990-10-18 1994-01-25 Monsanto Company Herbicidal benzoxazinone- and benzothiazinone-substituted pyrazoles
WO1998024785A1 (en) * 1996-12-02 1998-06-11 Fujisawa Pharmaceutical Co., Ltd. Indole-urea derivatives with 5-ht antagonist properties
WO1999052927A1 (en) * 1998-04-14 1999-10-21 Arena Pharmaceuticals, Inc. Non-endogenous, constitutively activated human serotonin receptors and small molecule modulators thereof
WO2002076464A1 (en) * 2001-03-22 2002-10-03 Arena Pharmaceuticals, Inc. Anti-psychosis combination

Cited By (136)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9296694B2 (en) 2000-03-06 2016-03-29 Acadia Pharmaceuticals Inc. Azacyclic compounds
US9765053B2 (en) 2000-03-06 2017-09-19 Acadia Pharmaceuticals Inc. Methods of treatment using selective 5-HT2A inverse agonists
US7351707B2 (en) 2001-12-28 2008-04-01 Acadia Pharmaceuticals, Inc. Spiroazacyclic compounds as monoamine receptor modulators
US7511053B2 (en) 2001-12-28 2009-03-31 Acadia Pharmaceuticals, Inc. Spiroazacyclic compounds as monoamine receptor modulators
US7727999B2 (en) 2001-12-28 2010-06-01 Acadia Pharmaceuticals Inc. Spiroazacyclic compounds as monoamine receptor modulators
US7402590B2 (en) 2001-12-28 2008-07-22 Acadia Pharmaceuticals Inc. Spiroazacyclic compounds as monoamine receptor modulators
US7217719B2 (en) 2001-12-28 2007-05-15 Acadia Pharmaceuticals Inc. Spiroazacyclic compounds as monoamine receptor modulators
US7538222B2 (en) 2002-06-24 2009-05-26 Acadia Pharmaceuticals, Inc. N-substituted piperidine derivatives as serotonin receptor agents
US7253186B2 (en) 2002-06-24 2007-08-07 Carl-Magnus Andersson N-substituted piperidine derivatives as serotonin receptor agents
US7476682B2 (en) 2002-06-24 2009-01-13 Acadia Pharmaceuticals, Inc. N-substituted piperidine derivatives as serotonin receptor agents
US7612201B2 (en) 2002-12-19 2009-11-03 Vernalis (Cambridge) Limited Pyrazole compounds
WO2004056782A1 (en) * 2002-12-19 2004-07-08 Vernalis (Cambridge) Limited Pyrazole compounds
US10525046B2 (en) 2003-01-16 2020-01-07 Acadia Pharmaceuticals Inc. Selective serotonin 2A/2C receptor inverse agonists as therapeutics for neurodegenerative diseases
US8921393B2 (en) 2003-01-16 2014-12-30 Acadia Pharmaceuticals Inc. Selective serotonin 2A/2C receptor inverse agonists as therapeutics for neurodegenerative diseases
US8008323B2 (en) 2003-01-16 2011-08-30 Acadia Pharmaceuticals Inc. Selective serotonin 2A/2C receptor inverse agonists as therapeutics for neurodegenerative diseases
US7659285B2 (en) 2003-01-16 2010-02-09 Acadia Pharmaceuticals, Inc. Selective serotonin 2A/2C receptor inverse agonists as therapeutics for neurodegenerative diseases
US7994193B2 (en) 2003-01-16 2011-08-09 Acadia Pharmaceuticals Inc. Selective serotonin 2A/2C receptor inverse agonists as therapeutics for neurodegenerative diseases
US8377959B2 (en) 2003-01-16 2013-02-19 Acadia Pharmaceuticals, Inc. Selective serotonin 2A/2C receptor inverse agonists as therapeutics for neurodegenerative diseases
US7713995B2 (en) 2003-01-16 2010-05-11 Acadia Pharmaceuticals, Inc. Selective serotonin 2A/2C receptor inverse agonists as therapeutics for neurodegenerative diseases
US8618130B2 (en) 2003-01-16 2013-12-31 Acadia Pharmaceuticals Inc. Selective serotonin 2A/2C receptor inverse agonists as therapeutics for neurodegenerative diseases
US9211289B2 (en) 2003-01-16 2015-12-15 Acadia Pharmaceuticals Inc. Selective serotonin 2A/2C receptor inverse agonists as therapeutics for neurodegenerative diseases
US7601740B2 (en) 2003-01-16 2009-10-13 Acadia Pharmaceuticals, Inc. Selective serotonin 2A/2C receptor inverse agonists as therapeutics for neurodegenerative diseases
US7732462B2 (en) 2003-01-16 2010-06-08 Acadia Pharmaceuticals Inc. Selective serotonin 2A/2C receptor inverse agonists as therapeutics for neurodegenerative diseases
US10028944B2 (en) 2003-01-16 2018-07-24 Acadia Pharmaceuticals Inc. Selective serotonin 2A/2C receptor inverse agonists as therapeutics for neurodegenerative diseases
US9566271B2 (en) 2003-01-16 2017-02-14 Acadia Pharmaceuticals Inc. Selective serotonin 2A/2C receptor inverse agonists as therapeutics for neurodegenerative diseases
EP1615895A4 (en) * 2003-04-03 2007-11-07 Merck & Co Inc Biaryl substituted pyrazoles as sodium channel blockers
EP1615895A1 (en) * 2003-04-03 2006-01-18 Merck & Co., Inc. Biaryl substituted pyrazoles as sodium channel blockers
US7589116B2 (en) 2003-04-03 2009-09-15 Merck & Co. Inc. Biaryl substituted pyrazoles as sodium channel blockers
US8754238B2 (en) 2003-07-22 2014-06-17 Arena Pharmaceuticals, Inc. Diaryl and arylheteroaryl urea derivatives as modulators of the 5-HT2A serotonin receptor useful for the prophylaxis and treatment of disorders related thereto
WO2005012254A1 (en) * 2003-07-22 2005-02-10 Arena Pharmaceuticals, Inc. Diaryl and arylheteroaryl urea derivatives as modulators of the 5-ht2a serotonin receptor useful for the prophylaxis and treatment of disorders related therto
US8871797B2 (en) 2003-07-22 2014-10-28 Arena Pharmaceuticals, Inc. Diaryl and arylheteroaryl urea derivatives as modulators of the 5-HT2A serotonin receptor useful for the prophylaxis and treatment of disorders related thereto
EA009732B1 (en) * 2003-07-22 2008-02-28 Арена Фармасьютикалз, Инк. Diaryl and arylheteroaryl urea derivatives as modulators of the 5-htserotonin receptor useful for the prophylaxis and treatment of disorders related thereto
AU2004261582C1 (en) * 2003-07-22 2010-01-07 Arena Pharmaceuticals, Inc. Diaryl and arylheteroaryl urea derivatives as modulators of the 5-HT2A serotonin receptor useful for the prophylaxis and treatment of disorders related thereto
US9273035B2 (en) 2003-07-22 2016-03-01 Arena Pharmaceuticals, Inc. Diaryl and arylheteroaryl urea derivatives as modulators of the 5-HT2A serotonin receptor useful for the prophylaxis and treatment of disorders related thereto
EP1695966A1 (en) * 2003-07-22 2006-08-30 Arena Pharmaceuticals, Inc. Diaryl and arylheteroaryl urea derivatives as modulators of the 5-ht2a serotonin receptor useful for the prophylaxis and treatment of disorders related thereto
US9775829B2 (en) 2003-07-22 2017-10-03 Arena Pharmaceuticals, Inc. Diaryl and arylheteroaryl urea derivatives as modulators of the 5-HT2A serotonin receptor useful for the prophylaxis and treatment of disorders related thereto
US7812176B2 (en) 2004-03-23 2010-10-12 Arena Pharmaceuticals, Inc. Processes for preparing substituted N-aryl-N′-[3-(1H-pyrazol-5-YL) phenyl] ureas and intermediates thereof
WO2005103011A1 (en) * 2004-03-23 2005-11-03 Arena Pharmaceuticals, Inc. Processes for preparing substituted n-aryl-n’-[3-(1h-pyrazol-5-yl) phenyl] ureas and intermediates thereof
US7820695B2 (en) 2004-05-21 2010-10-26 Acadia Pharmaceuticals, Inc. Selective serotonin receptor inverse agonists as therapeutics for disease
US7732615B2 (en) 2004-09-27 2010-06-08 Acadia Pharmaceuticals Inc. N-(4-fluorobenzyl)-N-(1-methylpiperidin-4-yl)-N′-(4-(2-methylpropyloxy)phenylmethyl)carbamide and its tartrate salt and crystalline forms
US7923564B2 (en) 2004-09-27 2011-04-12 Acadia Pharmaceuticals, Inc. Synthesis of N-(4-fluorobenzyl)-N-(1-methylpiperidin-4-yl)-N′-(4-(2-methylpropyloxy) phenylmethyl)carbamide and its tartrate salt and crystalline forms
US7790899B2 (en) 2004-09-27 2010-09-07 Acadia Pharmaceuticals, Inc. Synthesis of N-(4-fluorobenzyl)-N-(1-methylpiperidin-4-yl)-N′-(4-(2-methylpropyloxy)phenylmethyl)carbamide and its tartrate salt and crystalline forms
US7868176B2 (en) 2004-09-27 2011-01-11 Acadia Pharmaceuticals, Inc. Salts of N-(4-fluorobenzyl)-N-(1-methylpiperidin-4-y1)-N′-(4-(2-methylpropyloxy)phenylmethyl)carbamide and their preparation
US7884101B2 (en) 2004-11-19 2011-02-08 Arena Pharmaceuticals, Inc. 3-phenyl-pyrazole derivatives as modulators of the 5-HT2A serotonin receptor useful for the treatment of disorders related thereto
WO2006055734A2 (en) * 2004-11-19 2006-05-26 Arena Pharmaceuticals, Inc. 3-phenyl-pyrazole derivatives as modulators of the 5-ht2a serotonin receptor useful for the treatment of disorders related thereto
WO2006055734A3 (en) * 2004-11-19 2006-08-17 Arena Pharm Inc 3-phenyl-pyrazole derivatives as modulators of the 5-ht2a serotonin receptor useful for the treatment of disorders related thereto
US8785441B2 (en) 2004-11-19 2014-07-22 Arena Pharmaceuticals, Inc. 3-phenyl-pyrazole derivatives as modulators of the 5-HT2A serotonin receptor useful for the treatment of disorders related thereto
EP1978019A1 (en) * 2004-11-19 2008-10-08 Arena Pharmaceuticals, Inc. 3-phenyl-pyrazole derivatives as modulators of the 5-HT2A serotonin receptor useful for the treatment of disorders related thereto
JP2008520695A (en) * 2004-11-19 2008-06-19 アリーナ ファーマシューティカルズ, インコーポレイテッド Use of 3-phenyl-pyrazole derivatives as modulators of 5-HT2A serotonin receptors useful for the treatment of 5-HT2A serotonin receptor mediated disorders
US10781180B2 (en) 2004-11-19 2020-09-22 Arena Pharmaceuticals, Inc. 3-phenyl-pyrazole derivatives as modulators of the 5-HT2A serotonin receptor useful for the treatment of disorders related thereto
CN102977029A (en) * 2004-11-19 2013-03-20 艾尼纳制药公司 3-phenyl-pyrazole derivatives as modulators of the 5-HT2A serotonin receptor useful for the treatment of disorders related thereto
EA011935B1 (en) * 2004-11-19 2009-06-30 Арена Фармасьютикалз, Инк. 3-phenyl-pyrazole derivatives as modulators ot the 5-htserotonin receptor useful for the treatment of disorders related thereto
NO340656B1 (en) * 2004-11-19 2017-05-22 Arena Pharm Inc 3-Phenylpyrazole Derivatives, Pharmaceutical Compositions Containing Such, Process for Preparation and Use as Modulators of the 5HT2A Serotonin Receptor for the Treatment of Related Disorders
WO2006060762A2 (en) * 2004-12-03 2006-06-08 Arena Pharmaceuticals, Inc. Pyrazole derivatives as modulators of the 5-ht2a serotonin receptor useful for the treatment of disorders related thereto
WO2006060762A3 (en) * 2004-12-03 2006-07-20 Arena Pharm Inc Pyrazole derivatives as modulators of the 5-ht2a serotonin receptor useful for the treatment of disorders related thereto
WO2006078610A1 (en) * 2005-01-19 2006-07-27 Arena Pharmaceuticals, Inc. Diaryl and arylheteroaryl urea derivatives as modulators of the 5-ht2a serotonin receptor useful for the prophylaxis or treatment of progressive multifocal leukoencephalopathy
WO2006081335A3 (en) * 2005-01-26 2007-05-24 Arena Pharm Inc Process for preparing substituted 1- [3- ( pyrazolyl) phenyl] -3-phenyl ureas
WO2006081335A2 (en) * 2005-01-26 2006-08-03 Arena Pharmaceuticals, Inc. Process for preparing substituted 1- [3- ( pyrazolyl) phenyl] -3-phenyl ureas
US8048878B2 (en) 2005-11-24 2011-11-01 Eisai R&D Management Co., Ltd. Two cyclic cinnamide compound
US7973033B2 (en) 2006-03-09 2011-07-05 Eisai R&D Management Co., Ltd. Multi-cyclic cinnamide derivatives
USRE45336E1 (en) 2006-05-18 2015-01-13 Arena Pharmaceuticals, Inc. Primary amines and derivatives thereof as modulators of the 5-HT2A serotonin receptor useful for the treatment of disorders related thereto
CN104761498B (en) * 2006-05-18 2017-12-26 艾尼纳制药公司 The conditioning agent of 5 HT2A serotonin receptors
US8664258B2 (en) 2006-05-18 2014-03-04 Arena Pharmaceuticals, Inc. Primary amines and derivatives thereof as modulators of the 5-HT2A serotonin receptor useful for the treatment of disorders related thereto
AU2007254244C1 (en) * 2006-05-18 2014-07-31 Arena Pharmaceuticals, Inc. 3-pyrazolyl-benzamide-4-ethers, secondary amines and derivatives thereof as modulators of the 5-HT2A serotonin receptor useful for the treatment of disorders related thereto
WO2007136680A2 (en) * 2006-05-18 2007-11-29 Arena Pharmaceuticals, Inc. 3-pyraz0lyl-benzamide-4-ethers, secondary amines and derivatives thereof as modulators of the 5-ht2a serotonin receptor useful for the treatment of disorders related thereto
CN103396412A (en) * 2006-05-18 2013-11-20 艾尼纳制药公司 Modifier of 5-HT2A serotonin receptor
US8481535B2 (en) 2006-05-18 2013-07-09 Arena Pharmaceuticals, Inc. Crystalline forms and processes for the preparation of phenyl-pyrazoles useful as modulators of the 5-HT2A serotonin receptor
USRE45337E1 (en) 2006-05-18 2015-01-13 Arena Pharmaceuticals, Inc. Ethers, secondary amines and derivatives thereof as modulators of the 5-HT2A serotonin receptor useful for the treatment of disorders related thereto
AU2007254244B2 (en) * 2006-05-18 2013-05-02 Arena Pharmaceuticals, Inc. 3-pyrazolyl-benzamide-4-ethers, secondary amines and derivatives thereof as modulators of the 5-HT2A serotonin receptor useful for the treatment of disorders related thereto
WO2007136680A3 (en) * 2006-05-18 2008-01-10 Arena Pharm Inc 3-pyraz0lyl-benzamide-4-ethers, secondary amines and derivatives thereof as modulators of the 5-ht2a serotonin receptor useful for the treatment of disorders related thereto
US10450276B2 (en) 2006-05-18 2019-10-22 Arena Pharmaceuticals, Inc. Ethers, secondary amines and derivatives thereof as modulators of the 5-HT2A serotonin receptor useful for the treatment of disorders related thereto
CN104761498A (en) * 2006-05-18 2015-07-08 艾尼纳制药公司 Modifier of 5-HT2A serotonin receptor
EP3395816A1 (en) * 2006-05-18 2018-10-31 Arena Pharmaceuticals, Inc. Ethers, secondary amines and derivatives thereof as modulators of the 5-ht2a serotonin receptor useful for the treatment of disorders related thereto
US9987252B2 (en) 2006-05-18 2018-06-05 Arena Pharmaceuticals, Inc. Primary amines and derivitves thereof as modulators of the 5-HT2A serotonin receptor useful for the treatment of disorders related thereto
US9199940B2 (en) 2006-05-18 2015-12-01 Arena Pharmaceuticals, Inc. Crystalline forms and processes for the preparation of phenyl-pyrazoles useful as modulators of the 5-HT2A serotonin receptor
US8148417B2 (en) 2006-05-18 2012-04-03 Arena Pharmaceuticals, Inc. Primary amines and derivatives thereof as modulators of the 5-HT2A serotonin receptor useful for the treatment of disorders related thereto
US9221755B2 (en) 2006-05-18 2015-12-29 Arena Pharmaceuticals, Inc. Ethers, secondary amines and derivatives thereof as modulators of the 5-HT2A serotonin receptor useful for the treatment of disorders related thereto
US8680119B2 (en) 2006-05-18 2014-03-25 Arena Pharmaceuticals, Inc. Ethers, secondary amines and derivatives thereof as modulators of the 5-HT2A serotonin receptor useful for the treatment of disorders related thereto
US8148418B2 (en) 2006-05-18 2012-04-03 Arena Pharmaceuticals, Inc. Ethers, secondary amines and derivatives thereof as modulators of the 5-HT2A serotonin receptor useful for the treatment of disorders related thereto
US9783502B2 (en) 2006-05-18 2017-10-10 Arena Pharmaceuticals, Inc. Crystalline forms and processes for the preparation of phenyl-pyrazoles useful as modulators of the 5-HT2A serotonin receptor
US9328107B2 (en) 2006-05-18 2016-05-03 Arena Pharmaceuticals, Inc. Primary amines and derivatives thereof as modulators of the 5-HT2A serotonin receptor useful for the treatment of disorders related thereto
JP2009537545A (en) * 2006-05-18 2009-10-29 アリーナ ファーマシューティカルズ, インコーポレイテッド Primary amines as modulators of 5-HT2A serotonin receptors useful for the treatment of disorders associated with 5-HT2A serotonin receptors, and derivatives thereof
JP2009537543A (en) * 2006-05-18 2009-10-29 アリーナ ファーマシューティカルズ, インコーポレイテッド 3-pyrazolyl-benzamido-4-ethers, secondary amines and derivatives thereof as modulators of 5-HT2A serotonin receptors useful for the treatment of disorders associated with 5-HT2A serotonin receptors
EP2019093A4 (en) * 2006-05-19 2011-02-23 Eisai R&D Man Co Ltd Urea type cinnamide derivative
EP2019093A1 (en) * 2006-05-19 2009-01-28 Eisai R&D Management Co., Ltd. Urea type cinnamide derivative
US10351531B2 (en) 2006-10-03 2019-07-16 Arena Pharmaceuticals, Inc. Pyrazole derivatives as modulators of the 5-HT2A serotonin receptor useful for the treatment of disorders related thereto
US9434692B2 (en) 2006-10-03 2016-09-06 Arena Pharmaceuticals, Inc. Pyrazole derivatives as modulators of the 5-HT2A serotonin receptor useful for the treatment of disorders related thereto
US9732039B2 (en) 2006-10-03 2017-08-15 Arena Pharmeceuticals, Inc. Pyrazole derivatives as modulators of the 5-HT2A serotonin receptor useful for the treatment of disorders related thereto
US8008293B2 (en) 2007-02-28 2011-08-30 Eisai R&D Management Co., Ltd. Bicyclic oxomorpholine derivative
US9567327B2 (en) 2007-08-15 2017-02-14 Arena Pharmaceuticals, Inc. Imidazo[1,2-a]pyridine derivatives as modulators of the 5-HT2A serotonin receptor useful for the treatment of disorders related thereto
US10058549B2 (en) 2007-08-15 2018-08-28 Arena Pharmaceuticals, Inc. Imidazo[1,2-α]pyridine derivatives as modulators of the 5-HT2A serotonin receptor useful for the treatment of disorders related thereto
US9453000B2 (en) 2007-08-31 2016-09-27 Eisai R&D Management Co., Ltd. Polycyclic compound
US7935815B2 (en) 2007-08-31 2011-05-03 Eisai R&D Management Co., Ltd. Imidazoyl pyridine compounds and salts thereof
US10787437B2 (en) 2008-04-02 2020-09-29 Arena Pharmaceuticals, Inc. Processes for the preparation of pyrazole derivatives useful as modulators of the 5-HT2A serotonin receptor
US9556149B2 (en) 2008-04-02 2017-01-31 Arena Pharmaceuticals, Inc. Processes for the preparation of pyrazole derivatives useful as modulators of the 5-HT2A serotonin receptor
US10117851B2 (en) 2008-10-28 2018-11-06 Arena Pharmaceuticals, Inc. Composition of a 5-HT2A serotonin receptor modulator useful for the treatment of disorders related thereto
US10543193B2 (en) 2008-10-28 2020-01-28 Arena Pharmaceuticals, Inc. Processes useful for the preparation of 1-[3-(4-bromo-2-methyl-2H-pyrazol-3-yl)-4-methoxy-phenyl]-3-(2,4-difluoro-phenyl)-urea and crystalline forms related thereto
US9801856B2 (en) 2008-10-28 2017-10-31 Arena Pharmaceuticals, Inc. Composition of a 5-HT2A serotonin receptor modulator useful for the treatment of disorders related thereto
US9126946B2 (en) 2008-10-28 2015-09-08 Arena Pharmaceuticals, Inc. Processes useful for the preparation of 1-[3-(4-bromo-2-methyl-2H-pyrazol-3-yl)-4-methoxy-phenyl]-3-(2,4-difluoro-phenyl)urea and crystalline forms related thereto
US10583122B2 (en) 2008-10-28 2020-03-10 Arena Pharmaceuticals, Inc. Composition of a 5-HT2A serotonin receptor modulator useful for the treatment of disorders related thereto
US10071075B2 (en) 2008-10-28 2018-09-11 Arena Pharmaceuticals, Inc. Processes useful for the preparation of 1-[3-(4-bromo-2-methyl-2H-pyrazol-3-yl)-4-methoxy-phenyl]-3-(2,4-difluoro-phenyl)-urea and crystalline forms related thereto
US9745270B2 (en) 2008-10-28 2017-08-29 Arena Pharmaceuticals, Inc. Processes useful for the preparation of 1-[3-(4-bromo-2-methyl-2H-pyrazol-3-yl)-4-methoxy-phenyl]-3-(2,4-difluoro-phenyl)-urea and crystalline forms related thereto
US9034911B2 (en) 2008-10-28 2015-05-19 Arena Pharmaceuticals, Inc. Composition of a 5-HT2A serotonin receptor modulator useful for the treatment of disorders related thereto
US9353064B2 (en) 2008-10-28 2016-05-31 Arena Pharmaceuticals, Inc. Processes useful for the preparation of 1-[3-(4-bromo-2-methyl-2H-pyrazol-3-yl)-4-methoxy-phenyl]-3-(2,4-difluoro-phenyl)-urea and crystalline forms related thereto
US8980891B2 (en) 2009-12-18 2015-03-17 Arena Pharmaceuticals, Inc. Crystalline forms of certain 3-phenyl-pyrazole derivatives as modulators of the 5-HT2A serotonin receptor useful for the treatment of disorders related thereto
US9533967B2 (en) 2010-03-30 2017-01-03 Verseon Corporation Multisubstituted aromatic compounds as inhibitors of thrombin
US9963440B2 (en) 2010-03-30 2018-05-08 Verseon Corporation Multisubstituted aromatic compounds as inhibitors of thrombin
US10653674B2 (en) 2010-03-30 2020-05-19 Verseon Corporation Multisubstituted aromatic compounds as inhibitors of thrombin
US9156837B2 (en) 2011-07-29 2015-10-13 Takeda Pharmaceutical Company Limited Heterocyclic compound
EP2970141A4 (en) * 2013-03-15 2016-08-31 Verseon Corp Halogenopyrazoles as inhibitors of thrombin
CN105209440A (en) * 2013-03-15 2015-12-30 维颂公司 Halogenopyrazoles as inhibitors of thrombin
US10251872B2 (en) 2013-03-15 2019-04-09 Verseon Corporation Multisubstituted aromatic compounds as serine protease inhibitors
US9687479B2 (en) 2013-03-15 2017-06-27 Verseon Corporation Multisubstituted aromatic compounds as serine protease inhibitors
US9533970B2 (en) 2013-03-15 2017-01-03 Verseon Corporation Multisubstituted aromatic compounds as serine protease inhibitors
US10058541B2 (en) 2013-03-15 2018-08-28 Verseon Corporation Multisubstituted aromatic compounds as serine protease inhibitors
CN110423221A (en) * 2013-03-15 2019-11-08 维颂公司 Halo-pyrazole as thrombin inhibitor
WO2014149139A2 (en) 2013-03-15 2014-09-25 Verseon, Inc. Halogenopyrazoles as inhibitors of thrombin
US9951025B2 (en) 2013-03-15 2018-04-24 Verseon Corporation Halogenopyrazoles as inhibitors of thrombin
US10189810B2 (en) 2014-09-17 2019-01-29 Verseon Corporation Pyrazolyl-substituted pyridone compounds as serine protease inhibitors
US10532995B2 (en) 2015-02-27 2020-01-14 Verseon Corporation Substituted pyrazole compounds as serine protease inhibitors
US10022355B2 (en) 2015-06-12 2018-07-17 Axovant Sciences Gmbh Diaryl and arylheteroaryl urea derivatives as modulators of the 5-HT2A serotonin receptor useful for the prophylaxis and treatment of REM sleep behavior disorder
US11304932B2 (en) 2015-07-15 2022-04-19 Axovant Sciences Gmbh Diaryl and arylheteroaryl urea derivatives as modulators of the 5-HT2A serotonin receptor useful for the prophylaxis and treatment of hallucinations associated with a neurodegenerative disease
US10034859B2 (en) 2015-07-15 2018-07-31 Axovant Sciences Gmbh Diaryl and arylheteroaryl urea derivatives as modulators of the 5-HT2A serotonin receptor useful for the prophylaxis and treatment of hallucinations associated with a neurodegenerative disease
US10981870B2 (en) 2015-07-20 2021-04-20 Acadia Pharmaceuticals Inc. Methods for preparing N-(4-fluorobenzyl)-N-(1-methylpiperidin-4-yl)-N′-(4-(2-methylpropyloxy)phenylmethyl)carbamide and its tartrate salt and polymorphic form
US11840515B2 (en) 2015-07-20 2023-12-12 Acadia Pharmaceuticals Inc. Methods for preparing N-(4-fluorobenzyl)-N-(1-methylpiperidin-4-yl)-N′-(4-(2-methylpropyloxy)phenylmethyl)carbamide and its tartrate salt and polymorphic form c
US10981871B2 (en) 2015-07-20 2021-04-20 Acadia Pharmaceuticals Inc. Methods for preparing N-(4-fluorobenzyl)-N-(1-methylpiperidin-4-yl)-N′-(4-(2-methylpropyloxy)phenylmethyl)carbamide and its tartrate salt and polymorphic form C
US10517860B2 (en) 2016-03-25 2019-12-31 Acadia Pharmaceuticals Inc. Combination of pimavanserin and cytochrome P450 modulators
US10953000B2 (en) 2016-03-25 2021-03-23 Acadia Pharmaceuticals Inc. Combination of pimavanserin and cytochrome P450 modulators
US11191757B2 (en) 2016-03-25 2021-12-07 Acadia Pharmaceuticals Inc. Combination of pimavanserin and cytochrome P450 modulators
US11464768B2 (en) 2016-12-20 2022-10-11 Acadia Pharmaceuticals Inc. Pimavanserin alone or in combination for use in the treatment of Alzheimer's disease psychosis
US11135211B2 (en) 2017-04-28 2021-10-05 Acadia Pharmaceuticals Inc. Pimavanserin for treating impulse control disorder
US10849891B2 (en) 2017-08-30 2020-12-01 Acadia Pharmaceuticals Inc. Formulations of pimavanserin
US10646480B2 (en) 2017-08-30 2020-05-12 Acadia Pharmaceuticals Inc. Formulations of pimavanserin
US11452721B2 (en) 2017-08-30 2022-09-27 Acadia Pharmaceuticals Inc. Formulations of pimavanserin
US10449185B2 (en) 2017-08-30 2019-10-22 Acadia Pharmaceuticals Inc. Formulations of pimavanserin
WO2023236920A1 (en) * 2022-06-07 2023-12-14 Sironax Ltd. Sarm1 modulators, preparations, and uses thereof

Also Published As

Publication number Publication date
WO2003062206A3 (en) 2004-01-08
EP1509505A2 (en) 2005-03-02

Similar Documents

Publication Publication Date Title
WO2003062206A2 (en) Small molecule modulators of the 5-ht2a serotonin receptor useful for the prophylaxis and treatment of disorders related thereto
US6107324A (en) 5-HT2A receptor inverse agonists
US6140509A (en) Non-endogenous, constitutively activated human serotonin receptors and small molecule modulators thereof
US20230277505A1 (en) Diaryl and arylheteroaryl urea derivatives as modulators of the 5-ht2a serotonin receptor useful for the prophylaxis and treatment of disorders related thereto
US7754866B2 (en) Non-endogenous, constitutively activated human serotonin receptors and small molecule modulators thereof
US6150393A (en) Small molecule modulators of non-endogenous, constitutively activated human serotonin receptors
AU2004261582B2 (en) Diaryl and arylheteroaryl urea derivatives as modulators of the 5-HT2A serotonin receptor useful for the prophylaxis and treatment of disorders related thereto
US6541209B1 (en) Non-endogenous, constitutively activated human serotonin receptors and small molecule modulators thereof
WO2004028450A2 (en) Process of making phenylpyrazoles useful as selective 5ht2a modulators and intermediates thereof
EP1695966A1 (en) Diaryl and arylheteroaryl urea derivatives as modulators of the 5-ht2a serotonin receptor useful for the prophylaxis and treatment of disorders related thereto
US20030224442A1 (en) Non-endogeneous, constitutively activated human serotonin receptors and small molecule modualtors thereof
ZA200600596B (en) Diaryl and arylheteroaryl urea derivatives as modulators of the 5-HT2A serotonin receptor useful for the prophylaxis and treatment of disorders related thereto
AU2003257531A1 (en) Non-endogenous,. constitutively activated human serotonin receptors and small molecule modulators thereof

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

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 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 NO NZ OM PH PL PT RO RU SC SD SE SG SK SL TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): 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 SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

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

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP