WO2011060217A1 - Spiropiperidine compounds as orl-1 receptor antagonists - Google Patents
Spiropiperidine compounds as orl-1 receptor antagonists Download PDFInfo
- Publication number
- WO2011060217A1 WO2011060217A1 PCT/US2010/056449 US2010056449W WO2011060217A1 WO 2011060217 A1 WO2011060217 A1 WO 2011060217A1 US 2010056449 W US2010056449 W US 2010056449W WO 2011060217 A1 WO2011060217 A1 WO 2011060217A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- methyl
- mmol
- fluoro
- chloro
- compound
- Prior art date
Links
- UQAUXQMTUUAAIA-UHFFFAOYSA-N CC(N(C)Cc(cccc1F)c1-[n]1nc(C)c(CN(CC2)CCC2(c([s]2)c3cc2Cl)OCC3(F)F)c1)=O Chemical compound CC(N(C)Cc(cccc1F)c1-[n]1nc(C)c(CN(CC2)CCC2(c([s]2)c3cc2Cl)OCC3(F)F)c1)=O UQAUXQMTUUAAIA-UHFFFAOYSA-N 0.000 description 1
- MSGBNAWIZAUCKQ-UHFFFAOYSA-N Cc(c(C=O)c1)n[n]1-c1c(C)cccc1F Chemical compound Cc(c(C=O)c1)n[n]1-c1c(C)cccc1F MSGBNAWIZAUCKQ-UHFFFAOYSA-N 0.000 description 1
- UDRRAFLRLIHHCM-UHFFFAOYSA-N FC(COC12CCNCC1)(c1c2[s]c(F)c1)F Chemical compound FC(COC12CCNCC1)(c1c2[s]c(F)c1)F UDRRAFLRLIHHCM-UHFFFAOYSA-N 0.000 description 1
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D495/00—Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
- C07D495/12—Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains three hetero rings
- C07D495/20—Spiro-condensed systems
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/435—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
- A61K31/438—The ring being spiro-condensed with carbocyclic or heterocyclic ring systems
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
- A61P25/06—Antimigraine agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
- A61P25/24—Antidepressants
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P3/00—Drugs for disorders of the metabolism
- A61P3/04—Anorexiants; Antiobesity agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P43/00—Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
Definitions
- Orphanin FQ (OFQ)/Nociceptin is a 17 amino acid peptide that has high affinity for the ORL1 G-protein coupled receptor (GPCR).
- GPCR G-protein coupled receptor
- the ORL1 receptor is a Class A GPCR that is expressed primarily in the central nervous system and peripheral nervous system as well as in the gastrointestinal tract, smooth muscle, and immune system. While related structurally to opioid peptides/receptors the
- OFQ/Nociceptin system exhibits no significant cross reactivity to classical opioid peptides/receptors and exhibits anti-opioid activity in vivo (for example
- Nociceptin/orphanin FQ receptor (NOC/OFQ) antagonists specifically antagonists of the ORL- 1 receptor have demonstrated anti-depressant activity and anorectic activity in numerous studies with animal models for depression and feeding behavior.
- ORL-1 antagonists are deemed to be useful in the treatment of depression and/or the treatment of overweight, obesity, and/or weight maintenance post treatment for overweight or obesity.
- WO 2003/095427 describes certain spiropiperidinyl compounds as ORL-1 antagonists for use as analgesics.
- the present invention provides a family of 4',5'-dihydrospiro[piperidine-4,7'- thieno[2,3-c]pyran] compounds with high antagonist potency for the ORL-1 receptor and high in vivo ORL-1 receptor occupancy in the CNS. Additionally, certain of the compounds have a favorable cardiotoxicology profile as determined by selectivity over the hERG channel activity, as well as high selectivity over other physiologically important receptors (e.g. mu, kappa and delta opioids, serotonin, and dopamine receptors). Further, certain of the compounds of the present invention have favorable biopharmaceutical and pharmacokinetic properties (e.g. solubility, oral exposure, and CNS permeability). Certain of the compounds of the present invention exhibit reduced oxidative metabolism resulting in favorable oral bioavailability. Certain compounds have also demonstrated through animal models that the compounds of the present invention are useful for the treatment of migraine.
- the present invention provides compounds of Formula I:
- R 1 is fluoro or chloro
- R 2a and R 2b are each hydrogen or are each fluoro
- R 3 is hydrogen, methyl, hydroxymethyl, or (C1-C 3 ) alkoxymethyl
- R 4 is selected from the group consisting of fluoro, chloro, cyano, cyanomethyl, (Ci- C 3 ) alkyl, cyclopropyl, hydroxymethyl, methoxy, methoxymethyl,
- R 5 is hydrogen, C1-C4 alkyl, cyclopropyl, hydroxyethyl, methoxyethyl, -C(0)CH 3 , or -C(0)0(d-C 3 ) alkyl;
- R 6 is hydrogen or methyl
- R 7 is hydrogen, fluoro, chloro, methyl, hydroxymethyl, or methoxy; and Ar 1 is a moiety selected from the group consisting of imidizol-l-yl, imidizol-2-yl,
- a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, in combination with a pharmaceutically acceptable carrier, diluent, or excipient.
- the pharmaceutical composition further comprises at least one additional therapeutic ingredient, as for example an SSRI antidepressant, as for example fluoxetine.
- this aspect of the invention provides a pharmaceutical composition adapted for the treatment of depression comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, in combination with one or more pharmaceutically acceptable excipients, carriers, or diluents thereof.
- a pharmaceutical composition adapted for the treatment of overweight, obesity and/or weight maintenance comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, in combination with one or more pharmaceutically acceptable excipients, carriers, or diluents thereof.
- a further embodiment provides a pharmaceutical composition adapted for the treatment of migraine comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, in combination with one or more pharmaceutically acceptable excipients, carriers, or diluents thereof.
- the present invention also provides a method of treating depression in a mammal comprising administering to a mammal in need of such treatment an effective amount of a compound of Formula I or a pharmaceutically acceptable salt thereof.
- a further embodiment provides a method of treating depression in a mammal comprising administering to a mammal in need of such treatment an effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, and an effective amount of an SSRI antidepressant, or a pharmaceutically acceptable salt thereof, as for example fluoxetine.
- Other embodiments of the invention provide methods of treating overweight and/or obesity, and/or a method for weight maintenance comprising administering to a mammal in need of such treatment an effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof.
- the mammal is a human.
- This invention also provides a compound of Formula I or a pharmaceutically acceptable salt thereof for use in therapy.
- the invention provides a compound of Formula I, or a pharmaceutically acceptable salt thereof, for use in the treatment of depression in mammals, particularly humans.
- the invention also provides a compound of Formula I, or a pharmaceutically acceptable salt thereof, in combination with an SSRI antidepressant, or a pharmaceutically acceptable salt thereof, as for example fluoxetine, for use in the treatment of depression in mammals, particularly humans.
- this aspect of the invention includes any one of the following: a compound of Formula I, or a pharmaceutically acceptable salt thereof, for use in the treatment of overweight; a compound of Formula I, or a
- a compound of Formula I, or a pharmaceutically acceptable salt thereof, for use in the treatment of obesity a compound of Formula I, or a pharmaceutically acceptable salt thereof, for use in the maintenance of weight (for weight maintenance), particularly after treatment for overweight or obesity; a compound of Formula I, or a pharmaceutically acceptable salt thereof, for use in the treatment of migraine.
- Another aspect of this invention provides the use of a compound of Formula I, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of depression.
- Another embodiment of the invention provides the use of a compound of Formula I, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of overweight, obesity, and/or the maintenance of weight.
- Yet another embodiment of the invention provides the use of a compound of Formula I, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of migraine.
- Compounds of this invention are bases, and accordingly react with a number of organic and inorganic acids to form pharmaceutically acceptable salts.
- “pharmaceutically acceptable salt” refers to any salt of a compound of Formula I that is substantially non-toxic to living organisms. Such salts include those listed in Journal of Pharmaceutical Science, 66, 2-19 (1977), which are known to the skilled artisan.
- BSA bovine serum albumin
- mCPP meto-chlorophenylpiperazine, a non-selective serotonin receptor agonist.
- EDTA means ethylene diamine tetraacetic acid.
- EGTA means ethylene glycol tetraacetic acid.
- GTP means guanosine triphosphate
- HPES 4-(2-hydroxyethyl)-l-piperazineethanesulfonic acid.
- HPLC means high-pressure liquid chromatography
- IC5 0 means the concentration at which 50% of the maximum inhibition is achieved.
- LC/MS means liquid chromatography followed by mass spectroscopy.
- LC/MS/MS means liquid chromatography followed by mass spectroscopy, followed by a second ionizing mass spectroscopy.
- mouse forced swim test an animal model for antidepressant activity.
- MS means mass spectroscopy
- MS (ES+) means mass spectroscopy using electrospray ionization.
- NMR nuclear magnetic resonance
- RO Tracer means 2-[(2-Fluorophenyl)methyl]-3-(2-fluorospiro[4,5- dihydrothieno[2,3-c]pyran-7,4'-piperidine]- -yl)-N,N-dimethyl-propanamide.
- SCX column means strong cation exchange column.
- SNAr means nucleophilic aromatic substitution.
- ?Bu means a tertiary-butyl moiety
- TLC means thin layer chromatography
- XRD means X-Ray Diffraction
- Preferred compounds of the present invention are compounds wherein: ) R 1 is chloro;
- R 2a and R 2b are each fluoro
- R 1 is chloro and R 2a and R 2b are each fluoro:
- R 1 is fluoro and R 2a and R 2b are each hydrogen;
- R 3 is hydrogen, methyl, hydroxymethyl, or methoxymethyl
- R 3 is methyl
- R 3 is hydroxymethyl
- R 1 is chloro, R 2a and R 2b are each fluoro, and R 3 is methyl;
- R 1 is chloro
- R 2a and R 2b are each fluoro
- R 3 is hydroxymethyl
- R 7 is hydrogen, fluoro, or chloro
- R 7 is fluoro
- R 1 is chloro
- R 2a and R 2b are each fluoro
- R 7 is fluoro
- R 1 is chloro, R 2a and R 2b are each fluoro, R 3 is methyl, and R 7 is fluoro;4) R 1 is chloro, R 2a and R 2b are each fluoro, R 3 is hydroxymethyl, and R 7 is fluoro;
- R 4 is fluoro, hydroxymethyl, methoxymethyl, methylcarbonyl or
- R 4 is fluoro
- R 4 is hydroxymethyl
- R 4 is methoxymethyl
- R 4 is methylcarbonyl
- R 4 is 2-methylimidazol- 1-yl
- the compounds of the present invention can be prepared according to the following synthetic schemes by methods well known and appreciated in the art.
- compound III is reacted with an appropriately substituted pyrazole carbaldehyde and a reducing reagent such as sodium triacetoxyborohydride, in a suitable solvent such as tetrohydrofuran at ambient temperature to provide compound II.
- a suitable solvent such as tetrohydrofuran at ambient temperature
- compound II is coupled with compound IV, wherein substituent Y is chloro, bromo, iodo, or boronic acid, with a suitable catalyst such as copper iodide, a proper base such as potassium carbonate, in an appropriate solvent such as toluene at elevated temperature to give compound ⁇ , wherein R 4 is equal to R 4 or a precursor of R 4 .
- the compound of formula III can also react with an appropriately substituted aldehyde compound V under reductive amination conditions described above to give the desired compound of formula ⁇ . When R 4 is a precursor to R 4 , it is then converted to R 4 by known methods.
- the protecting group is a Boc (tert-Butyloxycarbonyl) group.
- Compound VII is reacted with N-bromosuccinimide in an appropriate solvent such as chlorobenzene with irradiation of a light source to give a bromide compound, which is then treated with a base solution such as aqueous sodium bicarbonate to afford a hydroxyl compound.
- the hydroxyl compound can be further oxidized under suitable oxidation conditions such as in potassium bromide, tetramethylpiperidine-N-oxide, and aqueous sodium hypochloride solution to provide the desired ketone compound VI.
- Compound VI is then reacted with (bis(2- methylethyl)amino)sulfur trifluoride in an appropriate solvent such as tetrahydrofuran at elevated temperature, the product obtained is de-protected to provide compound ma.
- compound IX can be obtained from compound XII through three-step synthesis, such as bromination, hydroxylation, and oxidation. Each intermediate can be isolated as pure compound for further reaction or reacted without isolation as described in the synthesis of compound VI.
- Compound IX is then treated with a suitable halogen-metal exchange reagent such as butyl lithium in proper solvent such as tetrahydrofuran under lowered temperature, followed by a fluorinating reagent such as N-fluoro-benzenesulfonimide to afford the desired fluorinated product, which is then de-protected appropriately to give the desired compound IIIc.
- a suitable halogen-metal exchange reagent such as butyl lithium in proper solvent such as tetrahydrofuran under lowered temperature
- a fluorinating reagent such as N-fluoro-benzenesulfonimide
- R Alkyl
- Compound of formula V can be made as illustrated in Scheme 3. Compound XVI is reacted with compound XVII to afford compound of formula XV under coupling conditions described above for the conversion of compound II to compound ⁇ .
- SNAr nucleophilic aromatic substitution
- compound XVI can react with compound XVII in an appropriate solvent such as dimethylformamide with a suitable base such as potassium carbonate at elevated temperature to provide compound XV.
- Z is hydrogen, it can be converted to an aldehyde with the Vilsmeier-Haack reaction.
- Z When Z is an ester group, it can be reduced to an alcohol first with an appropriate reducing reagent such as lithium aluminum hydride in an appropriate solvent such as tetrahydrofuran. The alcohol is then oxidized to an aldehyde with an appropriate oxidation reagent such as manganese (IV) oxide in a solvent such as dichloromethane.
- an appropriate reducing reagent such as lithium aluminum hydride in an appropriate solvent such as tetrahydrofuran.
- the alcohol is then oxidized to an aldehyde with an appropriate oxidation reagent such as manganese (IV) oxide in a solvent such as dichloromethane.
- R 4 is the precursor of R 4
- the transformation of R 4 to R 4 will include but not limited to reactions such as reductive amination to provide an desired new amine; reduction of an ester, ketone, or aldehyde to an alcohol, which can be further converted to an alkoxy compound or a carbamate; reduction of a nitrile to an amide or an amine; the transformation of an ester to a heterocycle such as oxadiazol under proper condition.
- reactions such as reductive amination to provide an desired new amine; reduction of an ester, ketone, or aldehyde to an alcohol, which can be further converted to an alkoxy compound or a carbamate; reduction of a nitrile to an amide or an amine; the transformation of an ester to a heterocycle such as oxadiazol under proper condition.
- N-bromosuccinimide 115.02 g, 639.77 mmol
- tert-butyl 2-chlorospiro[4,5-dihydrothieno[2,3-c]pyran-7,4'- piperidine]-l '-carboxylate 200 g, 581.61 mmol
- chlorobenzene 1.60 L
- the resulting suspension is irradiated with 3 X 100 w bulb lamps situated almost in contact with the external reactor wall and the reactor temperature is set to 45°C.
- N-bromosuccinimide (26.14 g, 145.40 mmol) is added and the temperature is maintained at 40°C for 15 hr.
- Reaction mixture is cooled to 0°C and methyl t-butyl ether (500 mL) is added. Solid is filtered and the solution is concentrated to about 1000 mL solution in chlorobenzene. Then, methyl t-butyl ether (1000 mL) is added, solids are filtered and filtrate is concentrated to afford 600 mL of chlorobenzene solution.
- Dimethyl sulfoxide (806.47 mL, 11.35 mol) is added and sodium bicarbonate (95.38 g, 1.14 mol) are added at room temperature.
- N-bromosuccinimide (2.2 equiv) is added to a solution of tert-butyl spiro[4,5- dihydrothieno[2,3-c]pyran-7,4'-piperidine]-l'-carboxylate (13.5 g) in chlorobenzene (108 mL). The resulting suspension is irradiated with 260 w bulb lamp overnight. More N-bromosuccinimide (1.7 g) is added to the mixture and the mixture is irradiated with 260 w bulb lamp for 3 hours.
- Potassium bromide (535.67 mg, 4.50 mmol) is added to a solution of tert-butyl 2-bromo-4-hydroxy-spiro[4,5-dihydrothieno[2,3-c]pyran-7,4'-cyclohexane]-l'- carboxylate (7.28g) and 2,2,6,6-tetramethylpiperidine-N-oxide (281.33 mg, 1.80 mmol,) in dichloromethane (70 mL) at 0°C.
- sodium bicarbonate is added to 10% in water sodium hypochlorite (22.34 mL, 36.01 mmol) until pH 9.
- methyl t-butyl ether is added (30 mL), and the reaction mixture is cautiously poured over sodium bicarbonate (saturated aqueous solution) cooled in an ice bath. C02 evolution is seen and sodium bicarbonate (saturated aqueous solution) is added until pH 8. The mixture is extracted with methyl t-butyl ether. Organic layer is decanted, washed with brine (2 x), dried over magnesium sulfate and the solvent evaporated under reduced pressure.
- the compound of Preparation 14 is essentially prepared as described in Preparation 13 by using 2,3-difluorobenzoic acid and methyl alcohol in a 96% yield:
- the compounds of Preparation 17-18 are prepared essentially as described in Preparation 16 from the corresponding 2,3-difluoro phenyl derivative.
- a capped vial is charged with ethyl l-(2-cyano-6-fluoro-phenyl)-3-methyl- pyrazole-4-carboxylate (1.97 g, 7.21 mmol) (contaminated with the other pyrazole regioisomer in a ratio of 75:25, 1,2-ethanediamine, N-methyl- (6 mL, 68.02 mmol) and phosphorus pentasulfide (229 mg, 1,01 mmol) and the mixture is stirred at 1 10°C for 30 min and then allow to reach rt.
- Solvent is evaporated in vacuo and the residue purified by normal phase Isco chromatography using dichloromethane/2M ammonia in methanol from 95/5 to 85/15 as eluent to yield 2.1 1 g of ethyl l-[2-fluoro-6-(l-methyl-4,5- dihydroimidazol-2-yl)phenyl]-3-methyl-pyrazole-4-carboxylate (contaminated with the other pyrazole regioisomer in a ratio 75:25). MS (m/z): 331 (M+1).
- Potassium permanganate (1.58 g, 10 mmmol) and montmorillonite K- 10 (3.16 g) are grounded together in a mortar until a fine homogeneous powder is obtained.
- KMn04-montmorillonite K-10 (3.2 g, 6.78 mmol) is added portionwise to a solution of ethyl l-[2-fluoro-6-(l-methyl-4,5-dihydroimidazol-2-yl)phenyl]-3-methyl-pyrazole-4- carboxylate (1.12 g, 3.39 mmol) (contaminated with the other pyrazole regioisomer in a ratio 75:25) in acetonitrile (84.76 mL, 1.62 moles). The mixture is stirred at room temperature for 6.5 hr.
- This compound is essentially prepared as described in Preparation 29 by using ethyl l-[2-fluoro-6-(l-methylimidazol-2-yl)phenyl]-3-methyl-pyrazole-4-carboxylate (contaminated with the other pyrazole regioisomer in a ratio 75:25) in 99% yield. MS (m/z): 287 (M+l).
- the reaction tube is quickly sealed (caution: build-up of pressure possible; use a safety shield) and stirred in a preheated oil bath at 100°C for 18 hr. with the aid of a magnetic stirrer.
- the mixture is diluted with water and extracted with ethyl acetate.
- the organic layer is separated, dried over magnesium sulfate, filtered and the solvent evaporated in vacuo.
- the resulting residue is purified by normal phase Isco chromatography eluting with hexane:ethanol (gradient from 2 to 15% in ethanol) to give 96 mg of the title compound. MS (m/z): 285 (M+l).
- the title compound is prepared using a method essentially as described in
- the reaction is quenched with saturated aqueous solution of sodium bicarbonate (200 mL) and the resulting phases are separated. The aqueous layer is extracted with ethyl acetate (2 x 50 mL). The organics are washed with brine (100 mL), dried over sodium sulfate and evaporated in vacuo. Crude is dissolved in methyl tert-butyl ether and a beige solid precipitated. Solid is filtered and discarded.
- triacetoxiborohydride (9.27 g, 43.75 mmoles) is added (internal temperature 25-35°) and the resulting suspension is stirred at room temperature for 2 hr. A water/ice mixture (30 mL) is added in portions with stirring. The phases are separated. The aqueous phase is extracted with tert-butyl methyl ether (50 mL).
- the tartrate salt is essentially prepared as described in Example 1. MS (m/z): 486 (M+l).
- Example 3-21 are prepared essentially as described in Example 2 from de corresponding aldehydes.
- Example 22 2'-Chloro-l-((l-(2,6-dimethylphenyl)-3-methyl-lH-pyrazol-4-yl)methyl)- 4',4'-difluoro-4',5'-dihydrospiro[piperidine-4,7'-thieno[2,3-c]pyran]
- Example 24 Methyl N-r2-r4-r(2-chloro-4,4-difluoro-spiror5H-thienor2,3-c1pyran-7,4'- piperidine]- -yl)methyl]-3-methyl-pyrazol-l-yl]-3-fluoro-phenyl]-N-methyl-carbamate
- the tartrate salt is essentially prepared as described in Example 1. MS (m z): 555 (M+l).
- the solvent is evaporated in vacuo and the residue is dissolved in 4 ⁇ hydrochloric acid in iso-propyl alcohol and stirred at room temperature for 2 hr.
- the solvent is evaporated and the residue is diluted in methanol and charged in an SCX cartridge.
- the 2 ⁇ ammonia in methanol fraction is collected and evaporated.
- the tartrate salt is essentially prepared as described in Example 1. MS (m/z): 537
- the reaction tube is quickly sealed (caution: build-up of pressure possible; use a safety shield) and immersed in a preheated oil bath at 115°C for 16 hr.
- the sample is cooled down to room temperature, diluted with ethyl acetate and filtered through celite.
- the solvent is evaporated in vacuo.
- the residue is purified by normal phase Isco chromatography using hexane/ethyl acetate (10-40% in ethyl acetate) to yield 103.9 mg of the title compound. MS (m/z): 468 (M+l).
- the tartrate salt is essentially prepared as described in Example 1. MS (m/z): 468
- Example 27-29 The compounds of Example 27-29 are prepared essentially as described in Example 26 from the corresponding phenyl halide.
- Example 30 2'-Chloro-4',4'-difluoro- 1 -(( 1 - (2-toly l)-3 -methyl- 1 H-pyrazol-4-yl)methyl)- 4',5'-dihydrospiro[piperidine-4,7'-thieno[2,3-c]pyran]
- the reaction mixture is bubbled with nitrogen for 20 minutes and then 2-iodo-toluene (272 mg, 1.25 mmol) and ?ra «s-N,N'-dimethylcyclohexane-l,2-diamine (39.31 ⁇ ⁇ , 0.25 mmol) are added.
- the reaction tube is quickly sealed (caution: build-up of pressure possible; use a safety shield) and immersed in a preheated oil bath at 110°C for 24 hours with the aid of a magnetic stirrer. Then, the mixture is poured on SCX column (25g) and eluted with methanol and then 2N solution of ammonia in methanol.
- the tartrate salt is essentially prepared as described in Example 1. MS (m/z): 464
- the reaction tube is quickly sealed (caution: build-up of pressure possible; use a safety shield) and stirred in a preheated oil bath at 110°C for 16 hr. with the aid of a magnetic stirrer.
- the mixture is diluted with water and extracted with ethyl acetate.
- the organic layer is separated, dried over magnesium sulfate, filtered and the solvent evaporated in vacuo.
- the resulting residue is purified by silica gel using normal phase Isco chromatography eluting with hexane:ethanol (gradient from 2 to 15% in ethanol) to give 17% yield of the title compound. MS (m/z): 498 (M+l).
- Example 32 l-[2-[4-[(2-Chloro-4,4-difluoro-spiro[5H-thieno[2,3-c]pyran-7,4'- piperidine]- -yl)methyl]-3-methyl-pyrazol-l-yl]-3-fluoro-phenyl]-N-methyl- methanamine (Z)-Tartrate
- Manganese (IV) oxide (13.89 g, 140.57 mmol) is added to a solution of [2-[4-[(2- chloro-4,4-difluoro-spiro[5H-thieno[2,3-c]pyran-7,4'-piperidine]-r-yl)methyl]-3-methyl- pyrazol-l-yl]-3-fluoro-phenyl]methanol (28.00 g, 56.23 mmol) in dichloromethane (224 mL) at room temperature and the resulting suspension is stirred at reflux for 2.5 hr.
- Additional manganese (IV) oxide (33.33 g, 337.37 mmol) is added and the mixture is stirred at reflux for 4h and at room temperature for 15 hr. More manganese (IV) oxide (8.33 g, 84.34 mmol) is added and stirring is continued for 7 hr. at room temperature. Reaction mixture is left stand for 1 hr. without stirring. Supernatant is decanted and filtered through a pad of celite eluting with dichloromethane.
- the tartrate salt is essentially prepared as described in Example 1. MS (m/z): 51 1
- the tartrate salt is essentially prepared as described in Example 1. MS (m/z): 551
- Example 34-35 The compounds of Example 34-35 are prepared essentially as described in Example 33 using the corresponding amine.
- Example 36 N-(2-( ' 4-( ' ( ' 2'-Chloro-4',4'-di£luoro-4',5'-dihvdrospirorpiperidine-4J'- thieno[2,3-c]pyran]-l yl)methyl)-3-methyl-lH-pyrazol-l-yl)-3-fluorobenzylidene)-2- methoxyethanamine
- the tartrate salt is essentially prepared as described in Example 1. MS (m z): 555
- Example 38 (2-(4-((2'-Chloro-4',4'-difluoro-4',5'-dihydrospiro[piperidine-4,7'-thieno[2,3- c]pyran] - 1 -yl)methyl)-3 -methyl- 1 H-pyrazol- 1 -yl)-3 -fluorophenyl)methanamine (L)- Tartrate
- Diisopropyl azodicarboxylate (0.105 mL, 0.54 mmol) is added to a solution of (2- (4-((2'-chloro-4',4'-difluoro-4',5'-dihydrospiro[piperidine-4,7'-thieno[2,3-c]pyran]- l - yl)methyl)-3 -methyl- 1 H-pyrazol- l-yl)-3-fluorophenyl)methanol (0.180 g, 0.36 mmol), phtalimide (0.079 mg, 0.54 mmol) and triphenylphosphine (0.142 g, 0.54 mmol) in 3 mL toluene at 0°C.
- Reaction mixture is refluxed for 2.5 hr. and then diluted with methanol and purified using a 2g SCX cartridge. After evaporation of the 2N ammonia in methanol fraction, the resulting product is purified by normal phase Isco chromatography eluting with ethanol and a 15% solution of ammonium hydroxide (7N in methanol) in ethanol (gradient of 25-90% of the basic eluent) yielding 75 mg of (2-(4-((2'-chloro-4',4'-difluoro-4',5'- dihydrospiro[piperidine-4,7'-thieno[2,3-c]pyran]-l-yl)methyl)-3-methyl-lH-pyrazol-l- yl)-3-fluorophenyl)methanamine. MS (m/z): 497 (M+l).
- the tartrate salt is essentially prepared as described in Example 1. MS (m z): 497 (M+l).
- Example 39 (2-(4-((2'-Chloro-4',4'-difluoro-4',5'-dihydrospiro[piperidine-4,7'-thieno[2,3- c]pyran]- 1 -yl)methyl)-3 -methyl- 1 H-pyrazol- 1 -yl)-3 -cyclopropylphenyl)methanol
- the reaction tube is sealed and stirred at 0°C for 30 min with the aid of a magnetic stirrer.
- the mixture is diluted with water (0.4 mL), stirred for 30 min and diluted with methanol.
- the product is purified using a 2g SCX cartridge and after evaporation of the basic fraction, the resulting residue is purified by normal phase Isco chromatography eluting with hexane:ethanol (2-15% in ethanol) to give 0.070 mg of the title compound.
- Example 40 l-(2-(4-((2'-Chloro-4',4'-difluoro-4',5'-dihydrospiro[piperidine-4,7'- thieno[2,3-c]pyran]-l-yl)methyl)-3-methyl-lH-pyrazol-l-yl)-3-fluorophenyl)ethanone
- the reaction tube is quickly sealed (caution: build-up of pressure possible; use a safety shield) and stirred in a preheated oil bath at 110°C for 18 hr. with the aid of a magnetic stirrer.
- the mixture is diluted with methanol and purified using a 2g SCX cartridge and the solvent is evaporated in vacuo.
- the resulting residue is purified by normal phase Isco chromatography eluting with dichloromethane/methanol (2-20% in methanol) to give 0.075 g of the title compound.
- Reaction crude is diluted with methanol and first purified using a 5g SCX cartridge. Resulting product is further purified by normal phase Isco chromatography eluting with dichloromethane/ethanol (5-30% in ethanol) to give 0.067 g of the title compound. MS (m/z): 535 (M+l).
- the tartrate salt is essentially prepared as described in Example 1. MS (m/z): 535
- Example 42 l-[2-[4-[(2-Chloro-4,4-difluoro-spiro[5H-thieno[2,3-c]pyran-7,4'- piperidine]- 1 '-yl)methyl]-3-methyl-pyrazol- 1 -yl]-3-fluoro-phenyl]cyclopropanamine (L)- Tartrate
- the tartrate salt is essentially prepared as described in Example 1. MS (m/z): 523 (M+l).
- Example 43 l-[2-[4-[(2-Chloro-4,4-difluoro-spiro[5H-thieno[2,3-c]pyran-7,4'- piperidine]-l'-yl)methyl]-3-methyl-pyrazol-l-yl]-3-fluoro-phenyl]-N,N-dimethyl- cyclopropanamine ( )-Tartrate
- the tartrate salt is essentially prepared as described in Example 1. MS (m/z): 551
- Acetic acid anhydride (0.043 mL, 0.452 mmol) is added to a solution of l-[2-[4- [(2-chloro-4,4-difluoro-spiro[5H-thieno[2,3-c]pyran-7,4'-piperidine]- -yl)methyl]-3- methyl-pyrazol-l-yl]-3-fluoro-phenyl]-N-methyl-methanamine (0.210 g, 0.41 1 mmol) in dichloromethane (4 mL) at 0°C. After 1 hr. , saturated aqueous solution of sodium bicarbonate is added to the reaction and the crude is extracted with dichloromethane.
- the tartrate salt is essentially prepared as described in Example 1. MS (m/z): 533
- Example 45 l-[2-[4-[(2-Chloro-4,4-difluoro-spiro[5H-thieno[2,3-c]pyran-7,4'- piperidine]- 1 '-yl)methyl]-3-methyl-pyrazol- 1 -yl]-3-methoxy-phenyl]-N-methyl- methanamine ( )-Tartrate
- the tartrate salt is essentially prepared as described in Example 1. MS (m/z): 523
- the title compound is prepared essentially as described in the third step of the preparation of Example 46 using 2-[4-[(2-chloro-4,4-difluoro-spiro[5H-thieno[2,3- c]pyran-7,4'-piperidine]- -yl)methyl]-3-methyl-pyrazol-l-yl]-3-fluoro-aniline(350.0 mg, 0.73 mmol) and acetaldehyde (0.163 mL, 2.90 mmol).
- the residue is purified by reverse phase HPLC to give 95.4 mg of desired compound, but further purification by normal phase HPLC is needed to yield 32 mg of the desired pure free base in 8% yield.
- the tartrate salt is essentially prepared as described in Example 1. MS (m/z): 548 (M+l).
- Example 48 2-Chloro-4,4-difluoro- 1 '- [[ 1 -[2-fluoro-6-( 1 H- 1 ,2,4-triazol-3 -yl)phenyl] -3 - methyl-pyrazol-4-yl]methyl]spiro[5H-thieno[2,3-c]pyran-7,4'-piperidine]
- the resultant solution is heated to 90°C for 2 hr.
- the reaction is cooled to room temperature.
- Acetic acid is then removed under reduced pressure and the residue is diluted with ethyl acetate and washed with saturated solution of sodium bicarbonate.
- the organic phase is evaporated under vacuo.
- the crude mixture is purified by flash chromatography on silica gel chromatography using
- reaction mixture is stirred at room temperature for 10 minutes and then sodium triacetoxyborohydride (0.53 g, 2.5 mmol) is added and stirred at room temperature for 24 hours. After completion, the reaction mixture was partitioned between dichloromethane (15 mL) and water (15 mL). The aqueous phase is extracted with dichloromethane (3 x 15 mL) and the combined organic extracts are dried over sodium sulfate and concentrated in vacuo. The crude mixture is purified by column chromatography over silica gel eluting with
- the tartrate salt is essentially prepared as described in Example 1. MS (m/z): 498
- Receptor occupancy tracer compound 2-[(2-Fluorophenyl)methyl]-3-(2-fluorospiro[4,5- dihydrothieno[2,3-c]pyran-7,4'-piperidine]- -yl)-N,N-dimethyl-propanamide ( )-Tartrate
- the mixture is stirred at room temperature for 5 hours.
- the reaction mixture is treated with aqueous saturated solution of sodium bicarbonate and extracted with dichloromethane (3 x 20 mL).
- the combined organic layers are dried over magnesium sulfate and the solvent evaporated under reduced pressure.
- the crude is purified by normal phase Isco chromatography using
- Enantiomeric resolution of racemic 2-[(2-fluorophenyl)methyl]-3-(2- fluorospiro[4,5-dihydrothieno[2,3-c]pyran-7,4'-piperidine]-r-yl)-N,N-dimethyl- propanamide (2.3 g, 5.29 mmol) ) is carried out with a Chiralpak AD column using hexane/0.2% dimethyl ethyl amine in ethanol 9/1. The desired compound is obtained in 36% yield as the first eluting enantiomer.
- the tartrate salt is essentially prepared as described in example 1. MS (m/z): 435 (M+l).
- nociceptin receptor antagonists are effective in rodent models of depression both alone and in combination with tricyclic or selective serotonergic reuptake inhibitor (SSRI) antidepressants, in rodent models of inhibiting hyperphagia, of inhibiting weight regain following previous weight loss, and in models for migraine.
- SSRI serotonergic reuptake inhibitor
- nociceptin receptor knockout mice studies conducted in nociceptin receptor knockout mice have demonstrated that the action of nociceptin antagonists in the forced swim test (measure of antidepressant activity) and in fasting-induced feeding (anti-obesity activity) is genotype dependent, supporting a specific mechanism of nociceptin antagonist action in these animal models.
- a nociceptin receptor antagonist may be particularly effective in these specific patient populations, such as patients with major depressive disorder, binge-eating disorder, overweight, obesity, and obesity with co-morbid clinical mood disorders.
- Radioligand binding assays are commonly used to determine the affinity (3 ⁇ 4) or potency of a compound to bind to a particular receptor or target protein.
- a filtration- based [3H]-OFQ/nociceptin receptor binding assay is developed based on previous assay formats (Ardati A, Henningsen RA, Higelin J, Reinscheid RK, Civelli O, Monsma FJ Jr. Mol Pharmacol. 1997 May;51(5):816-24.) with minor modifications.
- [3FFJ- OFQ/nociceptin binding assays are carried out in deep-well 96-well plates.
- [ 3 H]OFQ (final assay concentration 0.2 nM) competition studies are carried out with 5-10 ⁇ g of membrane protein (isolated from Chinese hamster ovary cells (CHO cells) expressing cloned human ORL1 receptors) in a final assay volume of 0.5 mL of buffer containing 20 mM HEPES, pH 7.4, 5 mM MgCl 2 , 1 mM EGTA, 100 mM NaCl, 0.1% bovine serum albumin. Samples are incubated for 60 min. at room temperature, which is found to be optimal for competition assays.
- membrane protein isolated from Chinese hamster ovary cells (CHO cells) expressing cloned human ORL1 receptors
- the assays are terminated by filtration through glass fiber filters (Wallac filtermat A) [pretreated with 0.3% polyethylenimine (Sigma) for 1 hr] on a Tometc cell harvester, and the filters are washed three times with 5 mL of ice-cold 50 mM Tris-HCl, pH 7.4. Filtermats are then dried and imbedded with Meltilex scintillant A and the radioactivity counted in a Wallac Microbeta scintillation counter. Specific binding is determined by displacement with 100 nM unlabeled nociceptin. Curves are plotted as the percent of specific binding and IC5 0 values are determined using a sigmoidal dose response curve with variable slope.
- 3 ⁇ 4 for the mu, kappa and delta opioids, serotonin, adrenergic, and muscarinic receptors may be determined using membranes expressing the desired receptor and appropriate corresponding radioligand competitor molecules.
- Exemplified compounds are tested essentially as described above and are found to have high affinity for the ORL-1 receptor.
- 3 ⁇ 4 's for the ORL-1 receptor for the exemplified compounds are found to be less than 0.9 nM, while the 3 ⁇ 4 for mu, kappa and delta opioids, serotonin, and dopamine receptors are found to be significantly greater.
- the compounds of Examples 2, 40, 47, and 50 are tested essentially as described above and are found to have affinities as shown in Table 1, below.
- physiologically relevant doses of the compounds of the invention are not expected to substantially interact with these sites in vivo, and thus are expected to avoid undesired effects associated with such activity.
- Agonist-mediated stimulation of G-protein coupled receptors results in the activation of membrane associated Ga y-protein heterotrimer complexes, and represents the first step in the transduction of extracellular signals to modification of intracellular pathways.
- the first step in activation of receptor-mediated activation of Ga y-proteins heterotrimer is the exchange of Ga subunit bound guanosine diphoshpate (GDP) for guanosine triphosphate (GTP).
- GDP Ga subunit bound guanosine diphoshpate
- GTP guanosine triphosphate
- Measurement of receptor- mediated G-protein activation can be measured using the non-hydrolyzable radiolabeled analog of GTP, GTP-y-[35S]. Utilizing this methodology, antagonist affinity (Kb) is measured in membranes expressing cloned human ORLl/nociceptin receptors using a GTP-y-[35S] binding assay according to previously described protocols with minor modifications (DeLapp et al, J Pharmacol Exp Ther. 1999 May;289(2):946-55; Ozaki et al, Eur J Pharmacol. 2000 Aug 18;402(l-2):45-53).
- Assays are conducted in a 200- ⁇ 1 volume with the following buffer composition: 100 mM NaCl, 20 mM HEPES, 5 mM MgCl 2 , 1 mM EDTA, 0.1% BSA, 3 ⁇ GDP, 0.5 nM [ 35 S]GTPyS.
- ORL1 receptor membrane suspension is added at a concentration of 20 ⁇ g protein per well and receptor stimulation is achieved using 300 nM nociceptin/OFQ.
- Wheat germ agglutinin coated SPA beads (Amersham, Arlington Hts., IL) are added at 1 mg per well to detect membrane-bound [ 35 S]GTPyS. Plates are sealed and incubated for 2 hr. at room temperature. Plates are then placed at 4 °C overnight to allow the SPA beads to settle and then counted in a Wallac Microbeta.
- Specific [ 35 S]GTPyS binding is determined as the difference in CPM observed in the absence and presence of 10 ⁇ unlabeled GTPyS. Data are plotted as the percent of specific [ 35 S]GTPyS bound. Curves are plotted as the percent of specific binding and IC5 0 values are determined using a sigmoidal dose response curve with variable slope.
- Antagonist affinity (3 ⁇ 4,) is estimated according to DeLapp et al, 1999 using a
- RO Receptor occupancy
- Nociceptin/ORLl receptor occupancy is measured in the hypothalamus, a structure which contains a high density of nociceptin/ORLl binding sites that are inside the blood brain barrier, using a novel proprietary nociceptin/ORLl antagonist RO tracer, 2-[(2- fluorophenyl)methyl]-3-(2-fluorospiro[4,5-dihydrothieno[2,3-c]pyran-7,4'-piperidine]-r- yl)-N,N-dimethyl-propanamide, (RO Tracer). These measurements are made without the need for a radiolabeled tracer as previously published for other receptors with
- central nociceptin/ORLl RO is measured at 6 or 24 hours following oral administration of the test compound to rats.
- Male Sprague-Dawley rats (Harlan Sprague-Dawley, Indianapolis, IN) are treated orally with a test compound, or vehicle (20% Captisol, 25 mM phosphate buffer, pH 2.0).
- test compound/vehicle All animals are administered an intravenous, 3 ⁇ g/kg dose of RO Tracer. It is at the time of RO Tracer administration that RO is considered to be measured.
- hypothalamic samples are homogenized in 4 volumes (w/v) of acetonitrile containing 0.1% formic acid and centrifuged at 14,000 RPM for 16 min. Supernatants are collected and diluted to a final volume of 0.3 mL with sterile water. Measurement of the RO Tracer is carried out using an Agilent model 1200 HPLC (Agilent Technologies, Palo Alto, CA) and an API 4000 mass spectrometer. The chromatographic separation uses a 2.1 X 50 mm CI 8 column (Agilent part number 971700-907) and a mobile phase consisting of 38% acetonitrile in water with an overall 0.1% formic acid content.
- Detection of RO Tracer is accomplished by monitoring the precursor to product ion transition with a mass to charge ratio (m/z) of 435 to 239, with levels quantified by comparison to standards prepared in brain tissue homogenates.
- m/z mass to charge ratio
- Zero percent RO is calculated as the level of RO Tracer in the hypothalamus of vehicle-pretreated animals, which represents the sum of nonspecific and specific binding (all receptors available to the tracer).
- the lower level of RO Tracer in animals pretreated with the very high intravenous dose of SB6121 11, the positive control group represents the nonspecific binding and is assigned the value of 100% occupancy (no receptors available to the tracer).
- the level of RO Tracer found in hypothalamus from the test compound treated group is linearly interpolated between these two points to calculate RO for the test compound.
- Exemplified compounds are tested essentially as described above and are found to have high receptor occupancy at the ORL- 1 receptor.
- Receptor occupancies for exemplified compounds are found to be between about 50 and about 115 % for 3 mg/kg dose after 6 hr., or between about 5 and about 108% RO for 3 mg/kg dose after 24 hr.
- the receptor occupancies for the compounds of Examples 2, 40, 47, and 50 are assayed essentially as described above for 3 mg/kg dose after 6 hr. and are found to have 76, 70, 84, and 81% RO, respectively.
- the compounds of the present invention have favorable bioavailability and penetration into the CNS to the targeted ORL-1 receptors.
- Blockade of K + -channel conductance in the heart is associated with cardiotoxicity in the form of QT-wave prolongation.
- the affinity (3 ⁇ 4) of the exemplified nociceptin receptor antagonists for the human ERG (hERG) K + channel is determined in HEK293 cells expressing cloned hERG using the hERG channel antagonist radioligand
- [3H]astemizole (2 nM final assay concentration) according to well known procedures (see for example Finlayson K, et al. (Eur J Pharmacol. 412(3):203-12, 2001)
- [3H]Astemizole binding assays are performed at the contract research company Cerep (Paris France) according to standard procedures.
- Examples 2, 40, and 50 are assayed essentially as described above and are found to have low activity, with 3 ⁇ 4 of, 2.89, 1.08, and 7.0 ⁇ , respectively.
- concentrations in vitro 3 ⁇ 4 & 3 ⁇ 4,
- physiologically relevant doses of the compounds of the invention are not expected to substantially interact with hERG sites in vivo, and thus are expected to have no substantial effect on QT prolongation.
- mFST is an established in vivo assay for antidepressant activity (Li et al. J Pharmacol Exp Ther. 319(l):254-9, 2006.). Mice treated with known clinically effective antidepressants (selective serotonin reuptake inhibitors and/or tricyclic antidepressants) exhibit the behavior of reduced time spent immobile after being placed in a water tank, a behavior associated with despair. The mFST was used to evaluate potential
- nociceptin/ORLl receptor dependent All compounds are prepared in 20% Captisol, 25 mM phosphate buffer, pH 2.0 on the day of use. Mice are placed in a cylinder (diameter: 10 cm; height: 25 cm) filled with 6 cm of water (22-25°C) for 6 min. The duration of immobility during the last 4 min. of the 6 min. period of the test was scored. A mouse is recorded as immobile when floating motionless or making only those movements necessary to keep its head above water.
- Representative compounds are tested essentially as described above and are found to significantly reduce immobilization times.
- the compounds of Examples 2 and 40 are assayed essentially as described above and are found to have ED 60 's of 23 and 23nM respectively, with maximum effects of 56% and 50% reductions in immobilization time, respectively. Therefore compounds of the present invention are expected to have antidepressant activity in vivo.
- the compounds of the present invention can be used in combination with other known antidepressants to produced enhanced efficacy.
- ORL-1 knockout mice a strain of mice engineered to lack the ORL-1 receptor
- the compounds of the present invention may be tested essentially as described above using wild type mice in one arm of the study and ORL-1 knockout mice in a second arm of the study, and are found to significantly reduce the immobilization time in the wild type mice, but showed no effect in the knockout mice.
- a norepinepherine reuptake inhibitor antidepressant, imiprimine is run as a positive control and is found to reduce immobilization times in both the wild type and knockout mice, showing that the behavioral effect mediated by the norepinepherine reuptake mechanism is intact in the knockout mouse strain.
- Blockade of fasting-induced hyperphagia in rats is an accepted model for hyperphagic eating disorders.
- All experiments are performed on naive twelve-week old male wild type and ORL knockout mice maintained on a 129S6 inbred background. Mice are individually housed a minimum of 3 days prior to the onset of testing to eliminate any effects of stress due to the change from group to individual housing.
- mice/genotype are randomly assigned to each treatment group on the test day. Pre-fast body weight measurements are taken, and food is then removed from cages overnight. Mice are fasted for approximately 15 hr. The next morning, the mice are given one of three doses of drug or vehicle via oral gavage 30 min. prior to gaining access to food. Drugs are dissolved in 20% Captisol dissolved in 25 mM phosphate buffer, pH 2.0. Measurements of body weight are taken immediately prior to drug treatment or 24 hr. after access to food is restored. It is worth noting that all mice independent of genotype lose -5-10% body weight following overnight fast. Measurements of food intake are recorded 1 hr following access to food, as indicated, by weight of food remaining at 1 hr.
- mice are typically at rest and not normally eating. Following initial testing, mice are rested for 1 week with access to unlimited chow. Following the week rest, mice are retested according to the latin-square design shown in Table 1.
- Table 2 A Latin square design is used to determine dose response curves of anorectic action of novel ORL antagonists in wild type and ORL knockout mice.
- Representative compounds are tested essentially as described above and are found to significantly reduce fasting-induced hyperphagia in mice.
- Examples 1, 2, 40, 47, and 50 are assayed essentially as described above and are found to substantially block fasting-induced hyperphagia. The effect was not observed in ORL-1 knockout mouse strain, demonstrating that the effect is mediated through the ORL-1 receptor.
- the 5-HT2 C agonist, mCPP is used as a positive control and is found to significantly reduce fasting induced hyperphagia equally in both wild type mice and the ORL-1 knockout mouse strain.
- the compounds of the present invention are useful in the treatment of overweight and/or obesity and/or for weight maintenance, as for example the treatment of binge eating.
- test compounds are prepared in a vehicle solution containing 20% Captisol in 25 mM phosphate buffer (pH 2.0).
- the positive control compound, sumatriptan is dissolved in saline.
- FITC fluoroscein isothiocyanate
- BSA bovine serum albumin
- the rats Five minutes following stimulation, the rats are killed by exsanguination with 40 mL of saline which also rinses residual FITC/BSA out of the blood vessels.
- the top of the skull is removed to collect the dural membranes.
- the membrane samples are removed from both hemispheres, rinsed with water, and spread flat on microscope slides.
- the slides are dried for 15 minutes on a slide warmer and cover-slipped with a 70% glycerol/water solution.
- a fluorescence microscope equipped with a grating monochromator and a spectrophotometer is used to quantify the amount of FITC-BSA dye in each dural sample.
- the microscope is equipped with a motorized stage interfaced with a personal computer.
- extravasation ratio that is the ratio of the amount of extravasation in the dura from the stimulated side compared to the unstimulated side
- Animals dosed with vehicle alone or an ineffective dose of the test compound have an extravasation ratio of approximately 2, while totally effective treatments result in a ratio of approximately 1.
- Results are expressed as mean values with standard errors of the mean ( ⁇ SEM). All statistical evaluations are conducting utilizing ANOVA followed by comparison to the control group by Dunnett's Method. Statistical significance is assumed when p ⁇ 0.05. Statistical analyses are performed using JMP statistical analysis software (SAS Research Institute, version 6.0.2).
- Representative compounds of the present invention are tested essentially as described above and are found to effectively block extravasation in a dose dependent manner. As a result, it is expected that the compounds of the present invention are useful in the treatment of migraine.
- compositions comprising at least one compound of Formula I, or a pharmaceutically acceptable salt thereof, as an active ingredient and at least one pharmaceutically acceptable carrier, diluent and/or excipient.
- These compositions can be administered by a variety of routes including oral, intranasal, transdermal, subcutaneous, intravenous, intramuscular, and pulmonary.
- Such pharmaceutical compositions and processes for preparing them are well known in the art. See, e.g., Remington: The Science and Practice of Pharmacy (University of the Sciences in Philadelphia, ed., 21 st ed., Lippincott Williams & Wilkins Co., 2005).
- compositions are preferably formulated in a unit dosage form, each dosage containing from about 0.1 to about 500 mg, more usually about 1.0 to about 200 mg, as for example between about 5 and 50 mg of the active ingredient.
- unit dosage form refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with at least one suitable pharmaceutically acceptable carrier, diluent and/or excipient.
- the compounds of Formula I are generally effective over a wide dosage range.
- dosages per day normally fall within the range of about 0.01 to about 50 mg/kg, more usually from about 0.05 to 5.0 mg/kg, and as for example between 0.1 and 1.0 mg/kg of body weight.
- dosage levels below the lower limit of the aforesaid range may be more than adequate, while in other cases still larger doses may be employed without causing any harmful side effect, and therefore the above dosage range is not intended to limit the scope of the invention in any way.
- the amount of the compound actually administered will be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound or compounds administered, the age, weight, and response of the individual patient, and the severity of the patient's symptoms.
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Life Sciences & Earth Sciences (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Animal Behavior & Ethology (AREA)
- Pharmacology & Pharmacy (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Engineering & Computer Science (AREA)
- Neurology (AREA)
- Neurosurgery (AREA)
- Biomedical Technology (AREA)
- Pain & Pain Management (AREA)
- Psychiatry (AREA)
- Child & Adolescent Psychology (AREA)
- Diabetes (AREA)
- Hematology (AREA)
- Obesity (AREA)
- Epidemiology (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Heterocyclic Carbon Compounds Containing A Hetero Ring Having Oxygen Or Sulfur (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Plural Heterocyclic Compounds (AREA)
Abstract
Description
Claims
Priority Applications (16)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
BR112012011710A BR112012011710A2 (en) | 2009-11-16 | 2010-11-12 | spiropiperidine compound as orl-1 receptor antagonists, their pharmaceutical composition and their use |
DK10782486.4T DK2501704T3 (en) | 2009-11-16 | 2010-11-12 | Spiropiperidine Compounds as Oral-1 Receptor Antagonists |
RS20130484A RS53017B (en) | 2009-11-16 | 2010-11-12 | Spiropiperidine compounds as oral-1 receptor antagagonisten |
AU2010319400A AU2010319400B2 (en) | 2009-11-16 | 2010-11-12 | Spiropiperidine compounds as ORL-1 receptor antagonists |
KR1020127012503A KR101368130B1 (en) | 2009-11-16 | 2010-11-12 | Spiropiperidine compounds as orl-1 receptor antagonists |
ES10782486T ES2436241T3 (en) | 2009-11-16 | 2010-11-12 | Spiropiperidine compounds as oral-1 receptor antagonists |
EA201290355A EA020391B1 (en) | 2009-11-16 | 2010-11-12 | Spiropiperidine compounds as orl-1 receptor antagonists |
CN201080051845.3A CN102666550B (en) | 2009-11-16 | 2010-11-12 | Spiropiperidine compounds as ORL-1 receptor antagonists |
PL10782486T PL2501704T3 (en) | 2009-11-16 | 2010-11-12 | Spiropiperidine compounds as oral-1 receptor antagagonisten |
SI201030389T SI2501704T1 (en) | 2009-11-16 | 2010-11-12 | Spiropiperidine compounds as oral-1 receptor antagagonisten |
US13/504,176 US20120214784A1 (en) | 2009-11-16 | 2010-11-12 | Spiropiperidine compounds as orl-1 receptor antagonists |
EP10782486.4A EP2501704B1 (en) | 2009-11-16 | 2010-11-12 | Spiropiperidine compounds as oral-1 receptor antagagonisten |
MX2012005690A MX2012005690A (en) | 2009-11-16 | 2010-11-12 | Spiropiperidine compounds as orl-1 receptor antagonists. |
JP2012538997A JP5723381B2 (en) | 2009-11-16 | 2010-11-12 | Spiropiperidine compounds as ORL-1 receptor antagonists |
CA2781041A CA2781041C (en) | 2009-11-16 | 2010-11-12 | Spiropiperidine compounds as orl-1 receptor antagonists |
HRP20130969AT HRP20130969T1 (en) | 2009-11-16 | 2013-10-11 | Spiropiperidine compounds as orl-1 receptor antagonists |
Applications Claiming Priority (4)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
EP09382245.0 | 2009-11-16 | ||
EP09382245 | 2009-11-16 | ||
US29862810P | 2010-01-27 | 2010-01-27 | |
US61/298,628 | 2010-01-27 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2011060217A1 true WO2011060217A1 (en) | 2011-05-19 |
Family
ID=41800752
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2010/056449 WO2011060217A1 (en) | 2009-11-16 | 2010-11-12 | Spiropiperidine compounds as orl-1 receptor antagonists |
Country Status (18)
Country | Link |
---|---|
US (1) | US20120214784A1 (en) |
EP (1) | EP2501704B1 (en) |
JP (1) | JP5723381B2 (en) |
KR (1) | KR101368130B1 (en) |
CN (1) | CN102666550B (en) |
AU (1) | AU2010319400B2 (en) |
BR (1) | BR112012011710A2 (en) |
CA (1) | CA2781041C (en) |
DK (1) | DK2501704T3 (en) |
EA (1) | EA020391B1 (en) |
ES (1) | ES2436241T3 (en) |
HR (1) | HRP20130969T1 (en) |
MX (1) | MX2012005690A (en) |
PL (1) | PL2501704T3 (en) |
PT (1) | PT2501704E (en) |
RS (1) | RS53017B (en) |
SI (1) | SI2501704T1 (en) |
WO (1) | WO2011060217A1 (en) |
Cited By (15)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2011069063A2 (en) | 2009-12-04 | 2011-06-09 | Sunovion Pharmaceuticals, Inc. | Multicyclic compounds and methods of use thereof |
WO2013085781A1 (en) | 2011-12-06 | 2013-06-13 | Eli Lilly And Company | Spirothienopyran- piperidine derivatives as orl-1 receptor antagonists for their use in the treatment of alcohol dependence and abuse |
WO2014022143A1 (en) | 2012-07-31 | 2014-02-06 | Eli Lilly And Company | Orl-1 receptor antagonists for the treatment of anxiety |
WO2018029102A1 (en) | 2016-08-10 | 2018-02-15 | Bayer Cropscience Aktiengesellschaft | Substituted 2-heterocyclyl imidazolyl-carboxamides as pest control agents |
US10196403B2 (en) | 2016-07-29 | 2019-02-05 | Sunovion Pharmaceuticals Inc. | Compounds and compositions and uses thereof |
US10780074B2 (en) | 2017-08-02 | 2020-09-22 | Sunovion Pharmaceuticals Inc. | Compounds and uses thereof |
US10815249B2 (en) | 2018-02-16 | 2020-10-27 | Sunovion Pharmaceuticals Inc. | Salts, crystal forms, and production methods thereof |
EP3766879A1 (en) | 2019-07-19 | 2021-01-20 | Basf Se | Pesticidal pyrazole derivatives |
US11077090B2 (en) | 2016-07-29 | 2021-08-03 | Sunovion Pharmaceuticals Inc. | Compounds and compositions and uses thereof |
US11129807B2 (en) | 2017-02-16 | 2021-09-28 | Sunovion Pharmaceuticals Inc. | Methods of treating schizophrenia |
US11136304B2 (en) | 2019-03-14 | 2021-10-05 | Sunovion Pharmaceuticals Inc. | Salts of a heterocyclic compound and crystalline forms, processes for preparing, therapeutic uses, and pharmaceutical compositions thereof |
WO2023154344A1 (en) * | 2022-02-08 | 2023-08-17 | Vertex Pharmaceuticals Incorporated | 2-methyl-4',5'-dihydrospiro[piperidine-4,7'-thieno[2,3-c]pyran] derivatives as inhibitors of apol1 and methods of using same |
US11738002B2 (en) | 2020-04-14 | 2023-08-29 | Sunovion Pharmaceuticals Inc. | Methods of treating neurological and psychiatric disorders |
US11866446B2 (en) | 2020-08-26 | 2024-01-09 | Vertex Pharmaceuticals Incorporated | Inhibitors of APOL1 and methods of using same |
US12060346B2 (en) | 2018-12-17 | 2024-08-13 | Vertex Pharmaceuticals Incorporated | Inhibitors of APOL1 and methods of using same |
Families Citing this family (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20140206667A1 (en) | 2012-11-14 | 2014-07-24 | Michela Gallagher | Methods and compositions for treating schizophrenia |
AR105821A1 (en) * | 2015-09-09 | 2017-11-15 | Lilly Co Eli | USEFUL COMPOUNDS TO INHIBIT ROR-g-T |
CN110392688B (en) * | 2017-03-02 | 2021-09-10 | 伊莱利利公司 | Compounds for inhibition of ROR-gamma-T |
DK3589638T3 (en) | 2017-03-02 | 2021-06-07 | Lilly Co Eli | Compounds useful for inhibiting ROR-Gamma-T |
Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2003095427A1 (en) | 2002-05-10 | 2003-11-20 | Taisho Pharmaceutical Co.,Ltd. | Spiro-ring compound |
WO2005016913A1 (en) * | 2003-08-19 | 2005-02-24 | Pfizer Japan, Inc. | Tetrahydroisoquinoline or isochroman compounds as orl-1 receptor ligands for the treatment of pain and cns disorders |
WO2005066183A1 (en) * | 2003-12-23 | 2005-07-21 | Grünenthal GmbH | Spirocyclic cyclohexane derivatives with affinity for the orl1-receptor |
Family Cites Families (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
BRPI0509377A (en) * | 2004-03-29 | 2007-09-11 | Pfizer | alpha aryl or heteroaryl methyl beta piperidino propanoic acid compounds as orl1 receptor antagonists |
DE102004039382A1 (en) * | 2004-08-13 | 2006-02-23 | Grünenthal GmbH | Spirocyclic cyclohexane derivatives |
AU2006252781A1 (en) * | 2005-06-02 | 2006-12-07 | Janssen Pharmaceutica, N.V. | Novel 3-spirocyclic indolyl derivatives useful as ORL-1 receptor modulators |
-
2010
- 2010-11-12 KR KR1020127012503A patent/KR101368130B1/en active IP Right Grant
- 2010-11-12 MX MX2012005690A patent/MX2012005690A/en active IP Right Grant
- 2010-11-12 BR BR112012011710A patent/BR112012011710A2/en active Search and Examination
- 2010-11-12 EA EA201290355A patent/EA020391B1/en not_active IP Right Cessation
- 2010-11-12 DK DK10782486.4T patent/DK2501704T3/en active
- 2010-11-12 CA CA2781041A patent/CA2781041C/en not_active Expired - Fee Related
- 2010-11-12 US US13/504,176 patent/US20120214784A1/en not_active Abandoned
- 2010-11-12 RS RS20130484A patent/RS53017B/en unknown
- 2010-11-12 CN CN201080051845.3A patent/CN102666550B/en not_active Expired - Fee Related
- 2010-11-12 AU AU2010319400A patent/AU2010319400B2/en not_active Ceased
- 2010-11-12 JP JP2012538997A patent/JP5723381B2/en active Active
- 2010-11-12 SI SI201030389T patent/SI2501704T1/en unknown
- 2010-11-12 WO PCT/US2010/056449 patent/WO2011060217A1/en active Application Filing
- 2010-11-12 PT PT107824864T patent/PT2501704E/en unknown
- 2010-11-12 PL PL10782486T patent/PL2501704T3/en unknown
- 2010-11-12 EP EP10782486.4A patent/EP2501704B1/en active Active
- 2010-11-12 ES ES10782486T patent/ES2436241T3/en active Active
-
2013
- 2013-10-11 HR HRP20130969AT patent/HRP20130969T1/en unknown
Patent Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2003095427A1 (en) | 2002-05-10 | 2003-11-20 | Taisho Pharmaceutical Co.,Ltd. | Spiro-ring compound |
WO2005016913A1 (en) * | 2003-08-19 | 2005-02-24 | Pfizer Japan, Inc. | Tetrahydroisoquinoline or isochroman compounds as orl-1 receptor ligands for the treatment of pain and cns disorders |
WO2005066183A1 (en) * | 2003-12-23 | 2005-07-21 | Grünenthal GmbH | Spirocyclic cyclohexane derivatives with affinity for the orl1-receptor |
Non-Patent Citations (17)
Title |
---|
ARDATI A; HENNINGSEN RA; HIGELIN J; REINSCHEID RK; CIVELLI 0; MONSMA FJ JR., MOL PHARMACOL., vol. 51, no. 5, May 1997 (1997-05-01), pages 816 - 24 |
BAILLIE; THOMAS A.: "Approaches to the Assessment of Stable and Chemically Reactive Drug Metabolites in Early Clinical Trials", CHEMICAL RESEARCH IN TOXICOLOGY, vol. 22, no. 2, 2009 |
CHENG, Y.C.; PRUSOFF, W.H., BIOCHEM. PHARMACOL., vol. 22, 1973, pages 3099 - 3108 |
CHERNET E; MARTIN LJ; LI D; NEED AB; BARTH VN; RASH KS; PHEBUS LA.: "Use of LC/MS to assess brain tracer distribution in preclinical, in vivo receptor occupancy studies: dopamine D2, serotonin 2A and NK-1 receptors as examples", LIFE SCI., vol. 78, no. 4, 2005, pages 340 - 6 |
DELAPP ET AL., J PHARMACOL EXP THER., vol. 289, no. 2, May 1999 (1999-05-01), pages 946 - 55 |
DELAPP ET AL., USING A MODIFICATION OF THE EQUATION, 1999 |
FINLAYSON K ET AL., EUR JPHARMACOL., vol. 412, no. 3, 2001, pages 203 - 12 |
GREENE; WUTS: "Protective Groups in Organic Synthesis", 1999, JOHN WILEY AND SONS INC. |
JOURNAL OF PHARMACEUTICAL SCIENCE, vol. 66, 1977, pages 2 - 19 |
LI ET AL., J PHARMACOL EXP THER., vol. 319, no. 1, 2006, pages 254 - 9 |
LI ET AL., JPHARMACOL EXP THER., vol. 319, no. 1, 2006, pages 254 - 9 |
OZAKI ET AL., EUR JPHARMACOL., vol. 402, no. 1-2, 18 August 2000 (2000-08-18), pages 45 - 53 |
REGUL PEPT., vol. 25, September 1998 (1998-09-01), pages 75 - 76,383-9 |
REMINGTON: "The Science and Practice of Pharmacy", 2005, LIPPINCOTT WILLIAMS & WILKINS CO. |
RICHARD C.; LAROCK: "Comprehensive Organic Transformations", 1999, JOHN WILEY AND SONS INC. |
YOSHIZUM TAKASHI ET AL: "Design, synthesis, and structure-activity relationship study of a novel class of ORL1 receptor antagonists based on N-biarylmethyl spiropiperidine", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS,, vol. 18, 16 May 2008 (2008-05-16), pages 3778 - 3782, XP002572595 * |
YOSHIZUMI; TAKASHI ET AL.: "Design, synthesis, and structure-activity relationship study of a novel class of ORL-1 receptor antagonists based on N-biarylmethyl spiropiperidine", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 18, 2008, pages 3778 - 3782 |
Cited By (35)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US10085968B2 (en) | 2009-12-04 | 2018-10-02 | Sunovion Pharmaceuticals Inc. | Multicyclic compounds and methods of use thereof |
US10894033B2 (en) | 2009-12-04 | 2021-01-19 | Sunovion Pharmaceuticals Inc. | Multicyclic compounds and methods of use thereof |
EP2507245A4 (en) * | 2009-12-04 | 2013-05-01 | Sunovion Pharmaceuticals Inc | MULTICYCLIC COMPOUNDS AND METHODS OF USE |
EP2507245A2 (en) * | 2009-12-04 | 2012-10-10 | Sunovion Pharmaceuticals Inc. | Multicyclic compounds and methods of use thereof |
AU2016200448C1 (en) * | 2009-12-04 | 2018-05-10 | Pgi Drug Discovery Llc. | Multicyclic compounds and methods of use thereof |
US8710245B2 (en) | 2009-12-04 | 2014-04-29 | Psychogenics Inc. | Multicyclic compounds and methods of use thereof |
EP3252057A3 (en) * | 2009-12-04 | 2018-02-21 | Sunovion Pharmaceuticals Inc. | Multicyclic compounds and methods of use thereof |
WO2011069063A2 (en) | 2009-12-04 | 2011-06-09 | Sunovion Pharmaceuticals, Inc. | Multicyclic compounds and methods of use thereof |
US9351954B2 (en) | 2009-12-04 | 2016-05-31 | Sunovion Pharmaceuticals Inc. | Multicyclic compounds and methods of use thereof |
AU2016200448B2 (en) * | 2009-12-04 | 2017-11-09 | Pgi Drug Discovery Llc. | Multicyclic compounds and methods of use thereof |
AP3880A (en) * | 2011-12-06 | 2016-10-31 | Lilly Co Eli | Spirothienopyran- piperidine derivatives as ORL-1 receptor antagonists for their use in the treatment of alcohol dependence and abuse |
TWI582096B (en) * | 2011-12-06 | 2017-05-11 | 美國禮來大藥廠 | 4',5'-dihydrospiro[piperidine-4,7'-thieno[2,3-c]pyran] compounds for the treatment of alcohol dependence and abuse |
US9371335B2 (en) | 2011-12-06 | 2016-06-21 | Eli Lilly And Company | Spirothienopyran-piperidine derivatives as ORL-1 receptor antagonists for their use in the treatment of alcohol dependence and abuse |
AU2012348140B2 (en) * | 2011-12-06 | 2015-09-17 | Eli Lilly And Company | Spirothienopyran- piperidine derivatives as ORL-1 receptor antagonists for their use in the treatment of alcohol dependence and abuse |
CN103987387A (en) * | 2011-12-06 | 2014-08-13 | 伊莱利利公司 | Spirothienopyran-piperidine derivatives as ORL-1 receptor antagonists for their use in the treatment of alcohol dependence and abuse |
WO2013085781A1 (en) | 2011-12-06 | 2013-06-13 | Eli Lilly And Company | Spirothienopyran- piperidine derivatives as orl-1 receptor antagonists for their use in the treatment of alcohol dependence and abuse |
WO2014022143A1 (en) | 2012-07-31 | 2014-02-06 | Eli Lilly And Company | Orl-1 receptor antagonists for the treatment of anxiety |
US10196403B2 (en) | 2016-07-29 | 2019-02-05 | Sunovion Pharmaceuticals Inc. | Compounds and compositions and uses thereof |
US11958862B2 (en) | 2016-07-29 | 2024-04-16 | Sumitomo Pharma America, Inc. | Compounds and compositions and uses thereof |
US10927124B2 (en) | 2016-07-29 | 2021-02-23 | Sunovion Pharmaceuticals Inc. | Compounds and compositions and uses thereof |
US11077090B2 (en) | 2016-07-29 | 2021-08-03 | Sunovion Pharmaceuticals Inc. | Compounds and compositions and uses thereof |
WO2018029102A1 (en) | 2016-08-10 | 2018-02-15 | Bayer Cropscience Aktiengesellschaft | Substituted 2-heterocyclyl imidazolyl-carboxamides as pest control agents |
US11129807B2 (en) | 2017-02-16 | 2021-09-28 | Sunovion Pharmaceuticals Inc. | Methods of treating schizophrenia |
US10780074B2 (en) | 2017-08-02 | 2020-09-22 | Sunovion Pharmaceuticals Inc. | Compounds and uses thereof |
US11491133B2 (en) | 2017-08-02 | 2022-11-08 | Sunovion Pharmaceuticals Inc. | Heteroaryl-isochroman compounds and uses thereof |
US10815249B2 (en) | 2018-02-16 | 2020-10-27 | Sunovion Pharmaceuticals Inc. | Salts, crystal forms, and production methods thereof |
US11440921B2 (en) | 2018-02-16 | 2022-09-13 | Sunovion Pharmaceuticals Inc. | Salts, crystal forms, and production methods thereof |
US11987591B2 (en) | 2018-02-16 | 2024-05-21 | Sumitomo Pharma America, Inc. | Salts, crystal forms, and production methods thereof |
US12060346B2 (en) | 2018-12-17 | 2024-08-13 | Vertex Pharmaceuticals Incorporated | Inhibitors of APOL1 and methods of using same |
US11136304B2 (en) | 2019-03-14 | 2021-10-05 | Sunovion Pharmaceuticals Inc. | Salts of a heterocyclic compound and crystalline forms, processes for preparing, therapeutic uses, and pharmaceutical compositions thereof |
WO2021013561A1 (en) | 2019-07-19 | 2021-01-28 | Basf Se | Pesticidal pyrazole and triazole derivatives |
EP3766879A1 (en) | 2019-07-19 | 2021-01-20 | Basf Se | Pesticidal pyrazole derivatives |
US11738002B2 (en) | 2020-04-14 | 2023-08-29 | Sunovion Pharmaceuticals Inc. | Methods of treating neurological and psychiatric disorders |
US11866446B2 (en) | 2020-08-26 | 2024-01-09 | Vertex Pharmaceuticals Incorporated | Inhibitors of APOL1 and methods of using same |
WO2023154344A1 (en) * | 2022-02-08 | 2023-08-17 | Vertex Pharmaceuticals Incorporated | 2-methyl-4',5'-dihydrospiro[piperidine-4,7'-thieno[2,3-c]pyran] derivatives as inhibitors of apol1 and methods of using same |
Also Published As
Publication number | Publication date |
---|---|
PL2501704T3 (en) | 2014-02-28 |
US20120214784A1 (en) | 2012-08-23 |
BR112012011710A2 (en) | 2016-03-01 |
AU2010319400B2 (en) | 2013-12-19 |
EP2501704A1 (en) | 2012-09-26 |
EA201290355A1 (en) | 2012-10-30 |
RS53017B (en) | 2014-04-30 |
JP2013510870A (en) | 2013-03-28 |
PT2501704E (en) | 2013-12-05 |
KR101368130B1 (en) | 2014-02-27 |
EA020391B1 (en) | 2014-10-30 |
KR20120082927A (en) | 2012-07-24 |
ES2436241T3 (en) | 2013-12-27 |
MX2012005690A (en) | 2012-06-13 |
HRP20130969T1 (en) | 2013-11-22 |
CN102666550B (en) | 2015-07-15 |
JP5723381B2 (en) | 2015-05-27 |
CA2781041C (en) | 2015-01-06 |
AU2010319400A1 (en) | 2012-06-07 |
CN102666550A (en) | 2012-09-12 |
SI2501704T1 (en) | 2013-11-29 |
EP2501704B1 (en) | 2013-09-18 |
CA2781041A1 (en) | 2011-05-19 |
DK2501704T3 (en) | 2013-10-14 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
CA2781041C (en) | Spiropiperidine compounds as orl-1 receptor antagonists | |
CA2796161C (en) | Spiropiperidine compounds as orl-1 receptor antagonists | |
EP2170903B1 (en) | Spiro[piperidine-4,4' -thieno[3, 2-c]pyran]derivatives and related compounds as inhibitors of the sigma receptor for the treatment of psychosis | |
WO2019121670A1 (en) | 4,5-dihydro-1h-thieno[2,3-g]indazolyl derivatives having multimodal activity against pain | |
JP6490187B2 (en) | Substituted azaspiro (4.5) decane derivatives | |
TW200524608A (en) | Therapeutic agents | |
BR112012012150A2 (en) | spiro piperidine compounds as orl-1 receptor-tor antagonists | |
JP2024523683A (en) | Piperidine derivatives and pharmaceutical compositions thereof, methods of preparation and use | |
US10183918B2 (en) | Oxa-azaspiro compounds having activity against pain |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
WWE | Wipo information: entry into national phase |
Ref document number: 201080051845.3 Country of ref document: CN |
|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 10782486 Country of ref document: EP Kind code of ref document: A1 |
|
WWE | Wipo information: entry into national phase |
Ref document number: 938/MUMNP/2012 Country of ref document: IN |
|
WWE | Wipo information: entry into national phase |
Ref document number: 13504176 Country of ref document: US |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2012538997 Country of ref document: JP |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2010319400 Country of ref document: AU |
|
ENP | Entry into the national phase |
Ref document number: 2781041 Country of ref document: CA Ref document number: 20127012503 Country of ref document: KR Kind code of ref document: A |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2010782486 Country of ref document: EP |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
WWE | Wipo information: entry into national phase |
Ref document number: MX/A/2012/005690 Country of ref document: MX |
|
ENP | Entry into the national phase |
Ref document number: 2010319400 Country of ref document: AU Date of ref document: 20101112 Kind code of ref document: A |
|
WWE | Wipo information: entry into national phase |
Ref document number: 201290355 Country of ref document: EA |
|
REG | Reference to national code |
Ref country code: BR Ref legal event code: B01A Ref document number: 112012011710 Country of ref document: BR |
|
WWE | Wipo information: entry into national phase |
Ref document number: P-2013/0484 Country of ref document: RS |
|
ENP | Entry into the national phase |
Ref document number: 112012011710 Country of ref document: BR Kind code of ref document: A2 Effective date: 20120516 |