US20110028452A1 - PYRIDO[4,3-d]PYRIMIDIN-4(3H)-ONE DERIVATIVES AS CALCIUM RECEPTOR ANTAGONISTS - Google Patents

PYRIDO[4,3-d]PYRIMIDIN-4(3H)-ONE DERIVATIVES AS CALCIUM RECEPTOR ANTAGONISTS Download PDF

Info

Publication number
US20110028452A1
US20110028452A1 US12/894,525 US89452510A US2011028452A1 US 20110028452 A1 US20110028452 A1 US 20110028452A1 US 89452510 A US89452510 A US 89452510A US 2011028452 A1 US2011028452 A1 US 2011028452A1
Authority
US
United States
Prior art keywords
pyrimidin
pyrido
trifluoromethyl
hydroxy
phenyl
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US12/894,525
Inventor
Mary T. Didiuk
Kevin K. Liu
David A. Griffith
Angel Guzman-Perez
Feng C. Bi
Daniel P. Walker
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Pfizer Inc
Original Assignee
Pfizer Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Pfizer Inc filed Critical Pfizer Inc
Priority to US12/894,525 priority Critical patent/US20110028452A1/en
Publication of US20110028452A1 publication Critical patent/US20110028452A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/12Drugs for disorders of the metabolism for electrolyte homeostasis
    • A61P3/14Drugs for disorders of the metabolism for electrolyte homeostasis for calcium homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/18Drugs for disorders of the endocrine system of the parathyroid hormones

Definitions

  • the present invention is directed toward novel pyrido[4,3-d]pyrimidin-4(3H)-one derivatives, pharmaceutical compositions containing these compounds, methods for their use and processes for their production.
  • novel pyrido[4,3-d]pyrimidin-4(3H)-one derivatives are able to inhibit calcium receptor activity and thus act as calcium receptor antagonists.
  • extracellular Ca 2+ In mammals, extracellular Ca 2+ is under rigid homeostatic control with the serum calcium concentration strictly maintained at a concentration of approximately 1.1 to 1.3 mM in a healthy mammal.
  • the extracellular Ca 2+ homeostasis depends on integrated regulation of Ca 2+ fluxes with respect to the intestine, kidneys and bone.
  • the extracellular Ca 2+ regulates various processes such as blood coagulation, nerve and muscle excitability, and normal bone homeostasis.
  • the Ca 2+ serum concentration decreases by 50% tetania occurs, and when the Ca 2+ serum concentration increases by 50% consciousness is clouded, in both instances a potentially life threatening circumstance.
  • Extracellular Ca 2+ also inhibits the secretion of parathyroid hormone (PTH) from parathyroid cells, inhibits bone resorption by osteoclasts, stimulates secretion of calcitonin from C-cells and is involved in re-absorption and excretion in the kidney.
  • PTH parathyroid hormone
  • the extracellular calcium-sensing receptor is a hormone-like receptor, more particularly a plasma membrane-bound G protein-coupled receptor (GPCR) that belongs to family 3 of the GPCR superfamily.
  • GPCR plasma membrane-bound G protein-coupled receptor
  • Family 3 of the GPCR superfamily includes metabotropic glutamate receptors (mGluRs), ⁇ -aminobutyric acid B-type receptors (GABA B Rs) as well as putative pheromone and taste receptors.
  • the CaSR has a large extracellular domain exhibiting “Venus flytrap” topology, a seven-transmembrane domain and a relatively large cytoplasmic domain.
  • Human CaSR consists of 1078 amino acids and shares 93% amino acid homology with bovine CaSR.
  • the CaSR senses and is activated by changes in extracellular Ca 2+ levels.
  • the presence of CaSR on certain specialized cells enables those Ca 2+ -sensing cells to respond to changes in extracellular Ca 2+ concentration.
  • Examples of Ca 2+ -sensing cells include the parathyroid-secreting cells of the parathyroid gland, the calcitonin-secreting C cells of the thyroid gland and certain cells in the kidney.
  • the CaSR has been found in a wide variety of other tissues including intestine, bone, bone marrow, brain, skin, pancreas, lung and heart.
  • the CaSR on the surface of parathyroid chief cells is the primary entity that regulates secretion of PTH from parathyroid cells. Activation of the CaSR on parathyroid chief cells by extracellular Ca 2+ suppresses PTH production and secretion, inhibits parathyroid cellular proliferation and likely inhibits PTH gene expression.
  • the CaSR on the surface of the calcitonin-secreting C cells of the thyroid gland mediate the stimulatory action of high extracellular Ca 2+ concentration on calcitonin secretion, thereby increasing the circulating level of the Ca 2+ -lowering hormone calcitonin.
  • the CaSR is also present in the kidney, along much of the nephrons and at the basolateral surface in the cortical thick ascending limb.
  • the CaSR In the basolateral surface in the cortical thick ascending limb the CaSR is thought to mediate high Ca 2+ -induced inhibition of the tubular re-absorption of Ca 2+ and magnesium.
  • a reduction of renal cortical synthesis of 1,25(OH) 2 vitamin D and polyuria with dilute urine are partially the result of hypercalcaemic activation of the CaSR in the nephron.
  • PTH is the primary endocrine hormone regulating Ca 2+ homeostasis in the blood and extracellular fluids.
  • PTH by acting on bone and kidney cells, increases the level of Ca 2+ in the plasma. This increase in plasma Ca 2+ concentration then acts as a negative feedback signal, thereby depressing PTH secretion.
  • the reciprocal relationship between extracellular Ca 2+ and PTH secretion forms an important mechanism for maintaining bodily Ca 2+ homeostasis.
  • PTH has been found to increase bone turnover, but the overall effect on bone is dependent on temporal changes in circulating levels of PTH. Sustained elevations in circulating plasma PTH levels, as occurs in hyperparathyroidism, have been found to result in a net catabolic effect on bone.
  • transient increases in plasma PTH levels have been found to exhibit a net anabolic effect on bone.
  • the effect of PTH on bone is likely due to PTH being able to induce a rapid release of calcium from bone and mediate other changes by acting directly on osteoblasts and indirectly on osteoclasts.
  • PTH affects cellular metabolic activity, ion transport, cell shape, gene transcriptional activity and secretion of proteases in osteoblasts.
  • PTH stimulates the production of RANKL, a protein that plays a crucial role in osteoclast differentiation and activity.
  • Calcimimetics are agents that act as allosteric modulators of the CaSR that increase the sensitivity of the CaSR to activation by extracellular Ca 2+ .
  • Calcilytics, or calcium receptor antagonists are agents that act as modulators of the CaSR that inhibit CaSR activity. This inhibition of the CaSR activity results in a decrease of one or more CaSR activities that are evoked by extracellular Ca 2+ .
  • Phenylalkylamine calcimimetic compounds include (R)-N-(1-(3-methoxyphenyl)ethyl)-3-phenylpropan-1-amine hydrochloride (NPS-467); (R)-3-(2-chlorophenyl)-N-(1-(3-methoxyphenyl)ethyl)propan-1-amine hydrochloride (NPS R-568, tecalcet hydrochloride) and (R)-( ⁇ )-N-(1-(naphthalen-1-yl)ethyl)-3-(3-(trifluoromethyl)phenyl)propan-1-amine hydrochloride (NPS-1493, cinacalcet hydrochloride).
  • Cinacalcet hydrochloride and uses thereof are disclosed in U.S. Pat. Nos. 6,011,068; 6,031,003; 6,211,244 and 6,313,146. Cinacalcet hydrochloride is marketed as Sensipar® and Minpara® in the U.S. and Europe, respectively, and is indicated for the treatment of secondary hyperparathyroidism in patients with chronic kidney disease on dialysis and for hypercalcemia in patients with parathyroid carcinoma.
  • Calcilytics or calcium receptor antagonists
  • Calcium receptor antagonists are useful in the treatment of various disease states characterized by abnormal levels of one or more components, e.g., polypeptides such as hormones, enzymes or growth factors, the expression and/or secretion of which is regulated or affected by activity at one or more CaSR.
  • Target diseases or disorders for calcium receptor antagonists include diseases involving abnormal bone and mineral homeostasis.
  • Abnormal calcium homeostasis is characterized by one or more of the following activities: an abnormal increase or decrease in serum calcium; an abnormal increase or decrease in urinary excretion of calcium; an abnormal increase or decrease in bone calcium levels (for example, as assessed by bone mineral density measurements); an abnormal absorption of dietary calcium; an abnormal increase or decrease in the production and/or release of messengers which affect serum calcium levels such as PTH and calcitonin; and an abnormal change in the response elicited by messengers which affect serum calcium levels.
  • novel calcium receptor antagonists of this invention are useful in the treatment of diseases associated with abnormal bone or mineral homeostasis.
  • these calcium receptor antagonists are useful in the treatment of hypoparathyroidism, osteoporosis, osteopenia, periodontal disease, bone fracture, osteoarthritis, rheumatoid arthritis, Paget's disease, humoral hypercalcemia associated with malignancy.
  • the present invention is directed towards calcium receptor antagonist compounds, pharmaceutical compositions containing the calcium receptor antagonist compounds and methods of treatment employing the calcium receptor antagonist compounds.
  • the present invention is directed to calcium receptor antagonists that are pyrido[4,3-d]pyrimidin-4(3H)-one derivatives of structural formula I
  • R 1 is -Q or (C 1 -C 6 )alkyl-Q
  • R 2 is (C 1 -C 6 )alkyl, (C 3 -C 7 )cycloalkyl, aryl or heteroaryl; wherein said aryl or heteroaryl is substituted with hydroxy and additionally said (C 1 -C 6 )alkyl, (C 3 -C 7 )cycloalkyl, aryl or heteroaryl within the definition of R 2 is optionally substituted with one to three substituents independently selected from halo, cyano, trifluoromethyl, trifluoromethoxy, (C 1 -C 6 )alkyl, (C 1 -C 6 )alkoxy or hydroxy;
  • R 3 is hydrogen, (C 1 -C 6 )alkyl, (C 3 -C 7 )cycloalkyl, (C 1 -C 6 )alkyl-Q, aryl, heteroaryl, OR 6 , or NR 7 R
  • Halo refers to fluoro, chloro, bromo or iodo.
  • (C 1 -C 6 )alkyl refers to a hydrocarbon group having one to six carbon atoms joined together by single carbon-carbon bonds.
  • the (C 1 -C 6 )alkyl group may be straight-chain or contain one or more branches and may be unsubstituted or substituted as specified.
  • Examples of (C 1 -C 6 )alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, tert-butyl, n-pentyl, (1-methyl)butyl, (2-methyl)butyl, (3-methyl)butyl, (1,2-dimethyl)propyl, n-hexyl, (1-methyl)pentyl, (2-methyl)pentyl, (3-methyl)pentyl, (4-methyl)pentyl, (1-ethyl)butyl, (2-ethyl)butyl, (1,2-dimethyl)butyl, (1,3-dimethyl)butyl, (2,3-dimethyl)butyl and the like.
  • (C 1 -C 6 )alkoxy refers to an oxygen joined to a (C 1 -C 6 )alkyl group.
  • the (C 1 -C 6 )alkyl group in the (C 1 -C 6 )alkoxy moiety may be straight-chain or contain one or more branches and may be unsubstituted or substituted as specified.
  • Examples of (C 1 -C 6 )alkoxy groups include methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, sec-butoxy, tert-butoxy, n-pentoxy and the like.
  • alkyl or alkoxy groups of differing length such as “(C 1 -C 3 )alkyl” or “(C 1 -C 3 )alkoxy” are defined in the same manner but limited to the number of carbons present.
  • (C 3 -C 7 )cycloalkyl refers to a saturated carbocyclic group having three to seven carbons and encompasses cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl.
  • the (C 3 -C 7 )cycloalkyl group can be unsubstituted or substituted as specified.
  • Aryl refers to a six to sixteen membered carbocyclic aromatic group having at least one ring with a conjugated pi-electron system.
  • the aryl group can have conjugated or fused rings and can be unsubstituted or substituted as specified. Examples of aryl groups include phenyl, naphthalenyl, anthracenyl, phenanthrenyl, azulenyl and biphenyl.
  • Heteroaryl refers to a five to sixteen membered aromatic group with at least one ring with a conjugated pi-electron system and containing one to four heteroatoms such as N, O or S.
  • the hetroaryl group can have conjugated or fused rings and can be unsubstituted or substituted as specified.
  • heteroaryl groups include thienyl, furanyl, pyrrolyl, pyrazolyl, imidazoyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, triazolyl, oxadiazolyl, thiadiazolyl, tetrazolyl, pyridinyl, pyridizinyl, pyrimidinyl, pyrazinyl, triazinyl, indolyl, isoindolyl, indolizinyl, benzofuranyl, benzothienyl, indazolyl, benzimidazoyl, benzthiazolyl, purinyl, quinolizinyl, quinolinyl, isoquinolinyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, naphthyridinyl, pteridinyl, carb
  • FIG. 1 is a chart depicting plasma PTH levels from the time of intravenous injection to 480 minutes following intravenous injection of 1 mg/kg of the compound of Example 1, 2-(2-Hydroxy-phenyl)-3-phenethyl-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one), in normal rats.
  • the present invention provides novel pyrido[4,3-d]pyrimidin-4(3H)-one derivatives and pharmaceutically acceptable salts thereof of structural formula I
  • the pharmaceutically acceptable salts of the compounds of formula I include the acid addition and base salts (including disalts) thereof.
  • Suitable acid addition salts are formed from acids which form non-toxic salts. Examples include the acetate, aspartate, benzoate, besylate, bicarbonate/carbonate, bisulphate/sulphate, borate, camsylate, citrate, edisylate, esylate, formate, fumarate, gluceptate, gluconate, glucuronate, hexafluorophosphate, hibenzate, hydrochloride/chloride, hydrobromide/bromide, hydroiodide/iodide, isethionate, lactate, malate, maleate, malonate, mesylate, methylsulphate, naphthylate, 2-napsylate, nicotinate, nitrate, orotate, oxalate, palmitate, pamoate, phosphate/hydrogen phosphate/dihydrogen phosphate, saccharate, stearate, succinate, tartrate, tosylate and trifluor
  • Suitable base salts are formed from bases which form non-toxic salts. Examples include the aluminium, arginine, benzathine, calcium, choline, diethylamine, diolamine, glycine, lysine, magnesium, meglumine, olamine, potassium, sodium, tromethamine and zinc salts.
  • bases which form non-toxic salts. Examples include the aluminium, arginine, benzathine, calcium, choline, diethylamine, diolamine, glycine, lysine, magnesium, meglumine, olamine, potassium, sodium, tromethamine and zinc salts.
  • a pharmaceutically acceptable salt of a compound of formula I may be readily prepared by mixing together solutions of the compound of formula I and the desired acid or base, as appropriate.
  • the salt may precipitate from solution and be collected by filtration or may be recovered by evaporation of the solvent.
  • the degree of ionization in the salt may vary from completely ionized to almost non-ionized.
  • the compounds of the invention include compounds of formula I as hereinbefore defined, polymorphs, and isomers thereof (including optical, geometric and tautomeric isomers) as hereinafter defined and isotopically-labeled compounds of formula I.
  • the compounds of the present invention may be administered as prodrugs.
  • prodrugs certain derivatives of compounds of formula I which may have little or no pharmacological activity themselves can, when administered into or onto the body, be converted into compounds of formula I having the desired activity, for example, by hydrolytic cleavage.
  • Such derivatives are referred to as ‘prodrugs’. Further information on the use of prodrugs may be found in ‘Pro-drugs as Novel Delivery Systems, Vol. 14, ACS Symposium Series (T Higuchi and W Stella) and ‘Bioreversible Carriers in Drug Design’, Pergamon Press, 1987 (ed. E B Roche, American Pharmaceutical Association).
  • Prodrugs can, for example, be produced by replacing appropriate functionalities present in the compounds of formula I with certain moieties known to those skilled in the art as ‘pro-moieties’ as described, for example, in “Design of Prodrugs” by H Bundgaard (Elsevier, 1985).
  • prodrugs include:
  • Compounds of formula I containing one or more asymmetric carbon atoms can exist as two or more stereoisomers. Where a compound of formula I contains an alkenyl or alkenylene group, geometric cis/trans (or Z/E) isomers are possible. Where the compound contains, for example, a keto or oxime group or an aromatic moiety, tautomeric isomerism (‘tautomerism’) can occur. It follows that a single compound may exhibit more than one type of isomerism.
  • racemate (or a racemic precursor) may be reacted with a suitable optically active compound, for example, an alcohol, or, in the case where the compound of formula (I) contains an acidic or basic moiety, an acid or base such as tartaric acid or 1-phenylethylamine.
  • a suitable optically active compound for example, an alcohol, or, in the case where the compound of formula (I) contains an acidic or basic moiety, an acid or base such as tartaric acid or 1-phenylethylamine.
  • the resulting diastereomeric mixture may be separated by chromatography and/or fractional crystallization and one or both of the diastereoisomers converted to the corresponding pure enantiomer(s) by means well known to a skilled person.
  • Chiral compounds of the invention may be obtained in enantiomerically-enriched form using chromatography, typically HPLC, on an asymmetric resin with a mobile phase consisting of a hydrocarbon, typically heptane or hexane, containing from 0 to 50% isopropanol, typically from 2 to 20%, and from 0 to 5% of an alkylamine, typically 0.1% diethylamine. Concentration of the eluate affords the enriched mixture. Mixtures of stereoisomers may be separated by conventional techniques known to those skilled in the art. [see, for example, “Stereochemistry of Organic Compounds” by E L Eliel (Wiley, New York, 1994).]
  • the present invention includes all pharmaceutically acceptable isotopically-labelled compounds of formula I wherein one or more atoms are replaced by atoms having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
  • isotopes suitable for inclusion in the compounds of the invention include isotopes of hydrogen, such as 2 H and 3 H, carbon, such as 11 C, 13 C and 14 C, chlorine, such as 38 Cl, fluorine, such as 18 F, iodine, such as 123 I and 125 I, nitrogen, such as 13 N and 15 N, oxygen, such as 15 O, 17 O and 18 O, phosphorus, such as 32 P, and sulfur, such as 35 S.
  • isotopically-labelled compounds of formula I for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies.
  • the radioactive isotopes tritium, i.e. 3 H, and carbon-14, i.e.
  • substitutions with heavier isotopes such as deuterium, i.e. 2 H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements, and hence may be preferred in some circumstances.
  • substitution with positron emitting isotopes, such as 11 C, 18 F, 15 O and 13 N, can be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy.
  • PET Positron Emission Topography
  • Isotopically-labeled compounds of formula (I) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the accompanying Examples and Preparations using an appropriate isotopically-labeled reagents in place of the non-labeled reagent previously employed.
  • a preferred embodiment of the present invention are compounds of Formula I as described above wherein R 2 is aryl or heteroaryl, wherein said aryl or heteroaryl is substituted with hydroxy and optionally substituted with one to three substituents independently selected from (C 1 -C 6 )alkoxy or halo; or a pharmaceutically acceptable salt thereof.
  • Another preferred embodiment of this invention is a compound of Formula I, wherein R 2 is 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl; wherein said 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl is optionally substituted with one to two substituents independently selected from (C 1 -C 6 )alkoxy or fluoro; or a pharmaceutically acceptable salt thereof.
  • Yet another preferred embodiment of the present invention is a compound of Formula I as described above wherein R 1 is (C 1 -C 6 )alkyl-Q; and Q is phenyl optionally substituted with one or two fluoro; or a pharmaceutically acceptable salt thereof.
  • Another preferred embodiment of the present invention is a compound of Formula I, wherein R 1 is phenethyl or 1-methyl-2-(phenyl)ethyl, wherein said phenethyl or 1-methyl-2-(phenyl)ethyl is optionally substituted on phenyl with one or two halo; and R 2 is 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl, wherein said 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl is optionally substituted with one or two substituents independently selected from (C 1 -C 3 )alkyl or halo; or a pharmaceutically acceptable salt thereof.
  • R 1 is phenethyl or 1-methyl-2-(phenyl)ethyl, wherein said phenethyl or 1-methyl-2-(phenyl)ethyl is optionally substituted on phenyl with one or two halo
  • R 2 is 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl, wherein said 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl is optionally substituted with one or two substituents independently selected from (C 1 -C 3 )alkyl or halo
  • R 3 is (C 1 -C 6 )alkyl, (C 1 -C 6 )alkyl-Q, aryl, heteroaryl, OR 6 , or NR 7 R 8 , wherein said (C 1 -C 6 )alkyl, aryl or heteroaryl is optionally substituted with one to three substituents independently selected from halo, cyano, triflu
  • R 1 is phenethyl or 1-methyl-2-(phenyl)ethyl, wherein said phenethyl or 1-methyl-2-(phenyl)ethyl is optionally substituted on phenyl with one or two halo
  • R 2 is 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl, wherein said 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl is optionally substituted with one or two substituents independently selected from (C 1 -C 3 )alkyl or halo
  • R 3 is trifluoromethyl
  • R 4 and R 5 are each hydrogen; or a pharmaceutically acceptable salt thereof.
  • R 1 is phenethyl or 1-methyl-2-(phenyl)ethyl, wherein said phenethyl or 1-methyl-2-(phenyl)ethyl is optionally substituted on phenyl with one or two halo; and R 2 is 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl, wherein said 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl is optionally substituted with one or two substituents independently selected from (C 1 -C 3 )alkyl or halo; R 3 is (C 1 -C 6 )alkyl; and R 4 and R 5 are each hydrogen; or a pharmaceutically acceptable salt thereof.
  • R 1 is phenethyl or 1-methyl-2-(phenyl)ethyl, wherein said phenethyl or 1-methyl-2-(phenyl)ethyl is optionally substituted on phenyl with one or two halo; and R 2 is 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl, wherein said 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl is optionally substituted with one or two substituents independently selected from (C 1 -C 3 )alkyl or halo; R 3 is (C 1 -C 6 )alkyl-Q; and R 4 and R 5 are each hydrogen; or a pharmaceutically acceptable salt thereof.
  • R 1 is phenethyl or 1-methyl-2-(phenyl)ethyl, wherein said phenethyl or 1-methyl-2-(phenyl)ethyl is optionally substituted on phenyl with one or two halo
  • R 2 is 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl, wherein said 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl is optionally substituted with one or two substituents independently selected from (C 1 -C 3 )alkyl or halo
  • R 3 is aryl, wherein said aryl is optionally substituted with one to three substituents independently selected from halo, cyano, trifluoromethyl, trifluoromethoxy, (C 1 -C 6 )alkyl, (C 1 -C 6 )alkoxy, NR 7 R 8 or hydroxy
  • R 4 and R 5 are each hydrogen; or a pharmaceutically
  • R 1 is phenethyl or 1-methyl-2-(phenyl)ethyl, wherein said phenethyl or 1-methyl-2-(phenyl)ethyl is optionally substituted on phenyl with one or two halo; and R 2 is 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl, wherein said 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl is optionally substituted with one or two substituents independently selected from (C 1 -C 3 )alkyl or halo; R 3 is heteroaryl; wherein said heteroaryl is optionally substituted with one to three substituents independently selected from halo, cyano, trifluoromethyl, trifluoromethoxy, (C 1 -C 6 )alkyl, (C 1 -C 6 )alkoxy, NR 7 R 8 or hydroxy; and R 4 and R 5 are each hydrogen; or a pharmaceutically
  • R 1 is phenethyl or 1-methyl-2-(phenyl)ethyl, wherein said phenethyl or 1-methyl-2-(phenyl)ethyl is optionally substituted on phenyl with one or two halo; and R 2 is 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl, wherein said 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl is optionally substituted with one or two substituents independently selected from (C 1 -C 3 )alkyl or halo; R 3 is OR 6 ; and R 4 and R 5 are each hydrogen; or a pharmaceutically acceptable salt thereof.
  • R 1 is phenethyl or 1-methyl-2-(phenyl)ethyl, wherein said phenethyl or 1-methyl-2-(phenyl)ethyl is optionally substituted on phenyl with one or two halo; and R 2 is 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl, wherein said 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl is optionally substituted with one or two substituents independently selected from (C 1 -C 3 )alkyl or halo; R 3 is NR 7 R 8 ; and R 4 and R 5 are each hydrogen; or a pharmaceutically acceptable salt thereof.
  • a preferred embodiment of the present invention is a compound of Formula I wherein R 3 is trifluoromethyl; or a pharmaceutically acceptable salt thereof.
  • Another preferred embodiment of the present invention is a compound of Formula I wherein R 3 is trifluoromethyl; and R 4 and R 5 are each independently hydrogen or methyl; or a pharmaceutically acceptable salt thereof.
  • Yet another preferred embodiment of the present invention is a compound of Formula I wherein R 2 is aryl or heteroaryl, wherein said aryl or heteroaryl is substituted with hydroxy and optionally substituted with one to three substituents independently selected from (C 1 -C 3 )alkyl, (C 1 -C 3 )alkoxy or fluoro; R 3 is trifluoromethyl; and R 4 and R 5 are each independently hydrogen or methyl; or a pharmaceutically acceptable salt thereof.
  • Yet another preferred embodiment of the present invention is a compound of Formula I wherein R 2 is hydroxy-phenyl or hydroxy-pyridyl, wherein said hydroxy-phenyl or hydroxy-pyridyl is optionally substituted with one to three substituents independently selected from methyl, methoxy or fluoro; R 3 is trifluoromethyl; and R 4 and R 5 are each independently hydrogen or methyl; or a pharmaceutically acceptable salt thereof.
  • R 1 is (C 1 -C 6 )alkyl-Q;
  • Q is phenyl optionally substituted with one or two substituents independently selected from fluoro, methyl or methoxy;
  • R 2 is hydroxy-phenyl or hydroxy-pyridyl, wherein said hydroxy-phenyl or hydroxy-pyridyl is optionally substituted with one to three substituents independently selected from methyl, methoxy or fluoro;
  • R 3 is trifluoromethyl; and
  • R 4 and R 5 are each independently hydrogen or methyl; or a pharmaceutically acceptable salt thereof.
  • Still another preferred embodiment of the present invention is a compound of Formula I wherein R 1 is phenethyl or 1-methyl-2-(phenyl)ethyl, wherein said phenethyl or 1-methyl-2-(phenyl)ethyl is optionally substituted on phenyl with one or two fluoro; R 3 is trifluoromethyl; and R 4 and R 5 are each independently hydrogen or methyl; or a pharmaceutically acceptable salt thereof.
  • R 1 is phenethyl or 1-methyl-2-(phenyl)ethyl, wherein said phenethyl or 1-methyl-2-(phenyl)ethyl is optionally substituted on phenyl with one or two halo
  • R 2 is hydroxy-phenyl or hydroxy-pyridyl, wherein said hydroxy-phenyl or hydroxy-pyridyl is optionally substituted with one or two substituents independently selected from (C 1 -C 3 )alkyl or halo
  • R 3 is trifluoromethyl
  • R 4 and R 5 are each independently hydrogen or methyl; or a pharmaceutically acceptable salt thereof.
  • R 1 is phenethyl or 1-methyl-2-(phenyl)ethyl, wherein said phenethyl or 1-methyl-2-(phenyl)ethyl is optionally substituted on phenyl with one or two substituents independently selected from halo, (C 1 -C 3 )alkyl or (C 1 -C 3 )alkoxy;
  • R 2 is 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl, wherein said 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl is optionally substituted with one or two substituents independently selected from (C 1 -C 3 )alkyl or halo;
  • R 3 is (C 1 -C 6 )alkyl, (C 3 -C 7 )cycloalkyl, (C 1 -C 6 )alkyl-Q, aryl, heteroaryl, or OR 6 ; wherein said (C 1 -C 6
  • Another embodiment of the present invention is a pharmaceutical composition
  • a pharmaceutical composition comprising a compound according to Formula I as described in any of the preceding embodiments hereinabove and a pharmaceutically acceptable carrier, adjuvant or diluent.
  • Another embodiment of the present invention are novel intermediate compounds 4-Amino-2-(trifluoromethyl)nicotinic acid or 4-Amino-2-chloronicotinic acid; or a pharmaceutically acceptable salt thereof.
  • Another embodiment of the present invention is a method of treating a disease or disorder characterized by abnormal bone or mineral homeostasis which comprises the administration to a patient in need of treatment thereof a therapeutically effective amount of a compound according to Formula I as described in any of the preceding embodiments hereinabove.
  • a preferred embodiment of the present invention is the method according to the preceding embodiment wherein the disease or disorder characterized by abnormal bone or mineral homeostasis is selected from the group consisting of osteoporosis, osteopenia, periodontal disease, Paget's disease, bone fracture, osteoarthritis, rheumatoid arthritis, and humoral hypercalcemia of malignancy.
  • the disease or disorder characterized by abnormal bone or mineral homeostasis is osteoporosis.
  • reaction schemes, Reaction Schemes I-IV depict methods of synthesis for compounds of formula I.
  • the variables R 1 , R 2 , R 3 , R 4 , and R 5 are as previously defined for a compound of the formula I unless otherwise stated.
  • the Reaction Schemes herein described are intended to provide a general description of the methodology employed in the preparation of many of the Examples given. However, it will be evident from the detailed descriptions given in the Experimental section that the modes of preparation employed extend further than the general procedures described herein. In particular, it is noted that the compounds prepared according to these Schemes may be modified further to provide new Examples within the scope of this invention.
  • the reagents and intermediates used in the following examples are either commercially available or can be prepared according to standard literature procedures by those skilled in the art of organic synthesis.
  • Reaction Scheme I depicts the synthesis of intermediates of formula V which are useful for preparing compounds of formula I.
  • an appropriately substituted pyridine of formula IX with an appropriate base, such as lithium diisopropylethylamide, in an appropriate solvent, such as THF, at approximately ⁇ 65° C. to ⁇ 78° C. followed by addition of CO 2 and acidification upon workup provides the corresponding nicotinic acid of formula VIII.
  • Reduction of the azide group in the compound of formula VI to the corresponding amine can be carried out using several procedures known in the art, such as catalytic hydrogenation, treatment with aqueous hydriodic acid at ambient temperature or treatment with triphenylphosphine. Saponification of the ester can then be carried out in a variety of ways such as aqueous lithium hydroxide in an appropriate solvent such as dioxane:MeOH:water (3:2:1) at a temperature of 40° C. to 100° C., preferably at approximately 85° C. for 1 to 24 hours to provide the corresponding nicotinic acid of formula V.
  • an appropriate solvent such as dioxane:MeOH:water (3:2:1
  • group R 3 in the compounds of formulae IX, VIII, VII, VI and V can have the same meaning as defined for group R 3 in compounds of formula I or alternatively can be a functionality that can be converted to a group R 3 as defined for compounds of formula I.
  • R 3 in formulae IX, VIII, VII, VI and V can be a halo group, such as chloro, which can then be further converted, for example, as shown in Reaction Scheme VI, below to a group R 3 as defined for a compound of formula I.
  • Reaction Scheme II depicts the preparation of compounds of Formula I.
  • Formula IV compounds are prepared by reacting the corresponding acid of Formula V with a variety of primary amines of general formula R 1 NH 2 under standard amide coupling conditions for a period between 4 to 24 hours.
  • Formula III compounds are prepared by reaction of Formula IV compounds with an appropriate aldehyde, R 2′ CHO, in the presence of a catalytic amount of an acid, such as p-toluenesulfonic acid, in a suitable reaction inert solvent such as acetonitrile or toluene at a temperature between 55° C. to 100° C., typically 80° C., for a period between 1 to 24 hours.
  • an acid such as p-toluenesulfonic acid
  • R 2′ moiety in the aldehyde of formula R 2′ CHO is one in which a protected hydroxy group typically exists, such as a methoxy or benzyloxy, which can subsequently be deprotected to the free hydroxy present in group R 2 in the compound of Formula I.
  • Compounds of Formula III are then treated with an oxidizing agent such as potassium permanganate in a polar solvent such as acetone at a temperature between 20° C. to 50° C., typically ambient temperature, to provide compounds of Formula II after a period of 1 to 24 hours.
  • Compounds of Formula II which contain a protected hydroxy group such as a suitable methyl ether moiety or benzyloxy moiety, may be converted to the free hydroxy (such as a phenolic hydroxy group) by standard deprotection conditions.
  • hydroxy groups in intermediates useful for preparing compounds of Formula I may be protected by other conventional protecting groups known to those skilled in the art.
  • intermediates containing a hydroxy group may be protected as the corresponding benzyloxy ether and subsequently deprotected by hydrogenation to provide the free hydroxy derivative.
  • Suitable protecting groups and methods for their removal are illustrated in Protective Groups in Organic Synthesis, 3 rd Ed., Theodora W. Greene, and Peter G. M. Wuts (John Wiley & Sons, 1999).
  • the compound of formula II contains a suitable methyl ether moiety
  • the compound of Formula II is dissolved in a suitable solvent such as methylene chloride at a temperature between ⁇ 78° C. to 10° C. followed by the addition of boron trichloride or boron tribromide.
  • the reaction mixture is stirred at 10° C. to 50° C. for 1 to 24 hours to provide the desired deprotected compound of Formula I.
  • compounds of formula II containing a suitable benzyloxy moiety can be deprotected to provide compounds of formula I by hydrogenation, such as H-cube hydrogenation (Pd/C cartridge) carried out at ambient temperature using methanol as solvent.
  • Reaction Scheme III depicts Formula IVa compounds prepared by reacting the corresponding acid of Formula Va with a variety of primary amines of general formula R 1 NH 2 under standard amide coupling conditions for a period between 4 to 24 hours.
  • Formula IIIa compounds may be prepared by reaction of Formula IVa compounds with an appropriate aldehyde, R 2′ CHO, in the presence of a catalytic amount of an acid, such as p-toluenesulfonic acid, in a suitable reaction inert solvent such as acetonitrile or toluene at a temperature between 55° C. to 100° C., typically 80° C., for a period between 1 to 24 hours.
  • an acid such as p-toluenesulfonic acid
  • R 2′ moiety in the aldehyde of formula R 2′ CHO is one in which a protected hydroxy group exists, such as a methoxy or benzyloxy, which can subsequently be deprotected to the free hydroxy present in group R 2 in the compound of formula I.
  • Compounds of Formula IIIa are then treated with an oxidizing agent such as potassium permanganate in a polar solvent such as acetone at a temperature between 20° C. to 50° C., typically ambient to provide compounds of Formula IIa after a period of 1 to 24 hours.
  • Reaction Scheme IV depicts the conversion of functional groups at the R 3 position of various intermediates useful in preparing compounds of Formula I.
  • Formula IIa compounds, wherein R 3 is chloro are converted to Formula IIb compounds, wherein R 3 is iodo, by reaction with a reagent such as sodium iodide and trimethylsilyl chloride in the dark in an inert solvent such as acetonitrile or propionitrile at a temperature between 55° C. to 120° C., typically 100° C., for a period between 1 to 24 hours.
  • the compounds of Formula IIc are prepared by reacting the compounds of Formula IIb with methyl-2,2-difluoro-2-(fluoro-sulfonyl)acetate and copper iodide in a mixture of solvent such as HMPA and DMF at a temperature between 55° C. to 120° C., typically 80° C., for a period between 1 to 24 hours.
  • the R 2′ group in the compounds of formulae IIa, IIb and IIc is one in which a protected hydroxy group exists, such as a methoxy or benzyloxy, which can subsequently be deprotected to the free hydroxy present in group R 2 of the corresponding compound within formula I.
  • Reaction Scheme V depicts the conversion of a methyl ether moiety in certain intermediates of formulae VI or VI′ wherein R 2 is 2-methoxyphenyl, to a free phenolic hydroxy moiety in compounds of Formula Ia or Ia′.
  • Compounds of Formula VI (or VI′ in which R 3 is CF 3 ) which contain a methyl ether moiety may be converted to the free phenol by dissolving the compounds of Formula VI or VI′ in a suitable solvent such as methylene chloride at a temperature between ⁇ 78° C. to 10° C. followed by the addition of boron trichloride or boron tribromide. The reaction mixture is stirred at 10° C. to 50° C. for 1 to 24 hours to provide the desired compounds, such as those of Formula Ia or Ia′.
  • Reaction Scheme VI depicts three reactions showing the conversion of chloro derivative IIa or IIa′ to alkoxy (Ib, wherein R 3 is OR 6 ), amino (Ic, wherein R 3 is NR 7 R 8 ) and carbon-linked derivatives (Id, wherein R 3 results from an appropriate boronic acid derivative such as an aryl, heteroaryl or (C 1 -C 6 )alkyl boronic acid), respectively.
  • Compounds of Formula IIa may be converted to compounds of Formula Ib by treatment of the corresponding alcohol, R 6 OH, in the presence of a suitable base such as sodium hydride in an inert solvent such as DMF or THF at 20° C. to 80° C. for 6 to 24 hours, followed by deprotection as necessary to provide compounds of Formula Ib.
  • Compounds of Formula IIa′ in which the hydroxy moiety in R 2 is in deprotected form, may be reacted with an appropriate amine of formula R 7 R 8 NH in a solvent such as methanol or ethanol at 20° C. to 80° C. for 1 to 24 hours to provide compounds of Formula Ic.
  • Compounds of Formula IIa may also be treated with an appropriate corresponding boronic acid derivative of formula R 3 B(OH) 2 in the presence of a catalyst such as palladium tetrakis(triphenylphosphine) in a mixture of aqueous sodium carbonate and a polar solvent such as dioxane in a microwave for 1 to 25 minutes at 50° C. to 150° C., followed by deprotection as necessary to provide compounds of Formula Id.
  • a catalyst such as palladium tetrakis(triphenylphosphine
  • the term “patient in need of treatment thereof” means humans and other animals who have or are at risk of having a disease or disorder characterized by abnormal bone or mineral homeostasis.
  • the “patient in need of treatment thereof” may have or be at risk of having a disease or disorder characterized by abnormal bone or mineral homeostasis selected from the group consisting of osteoporosis, osteopenia, periodontal disease, Paget's disease, bone fracture, osteoarthritis, rheumatoid arthritis, and humoral hypercalcemia of malignancy.
  • a preferred patient is a female, and particularly a postmenopausal female human.
  • treating includes preventative (e.g., prophylactic), palliative, adjuvant and curative treatment.
  • the treatment of osteoporosis means that a patient having osteoporosis or at risk of having osteoporosis can be treated according to the methods described herein.
  • a resulting reduction in the incidence of the disease state being preventively treated is the measurable outcome of the preventative treatment.
  • the present invention provides methods of treating osteopenia and osteoporosis by administering to a patient in need thereof a therapeutically effective amount of a compound of formula I.
  • Osteopenia is a thinning of the bones, but less than is seen with osteoporosis and is the stage before true osteoporosis.
  • the World Health Organization has developed diagnostic categories based on bone mass density (BMD) to indicate if a person has normal bones, has osteopenia or has osteoporosis. Normal bone density is within one standard deviation (+1 or ⁇ 1) of the young adult mean bone density.
  • Osteopenia (low bone mass) is defined as bone density of 1 to 2.5 standard deviations below the young adult mean ( ⁇ 1 to ⁇ 2.5), and osteoporosis is defined as a bone density that is 2.5 standard deviations or more below the young adult mean (> ⁇ 2.5).
  • Bone fractures can be a fracture to any bone in the body, and hip fracture being of particular concern. Hip fracture has a significant impact on medical resources and patient morbidity and mortality. Few patients admitted with a hip fracture are considered for prophylactic measures aimed at the reduction of further fracture risk. Currently, 10-13% of patients will later sustain a second hip fracture. Of patients who suffered a second hip fracture, fewer patients maintained their ability to walk independently after the second fracture than did so after the first (53 and 91% respectively, P ⁇ 0.0005). Pearse E. O. et al., Injury, 2003, 34(7), 518-521.
  • Second hip fracture has a significant further impact on patients' mobility and social independence. It is therefore desirable to have new methods for the treatment of bone fractures including hip fracture.
  • the compounds of Formula I can be administered together with additional agents which are useful for treating a disease or disorder characterized by abnormal bone or mineral homeostasis.
  • additional agents include calcium receptor antagonists other than those of Formula I, selective estrogen receptor modulators (SERMs), bisphosphonates, parathyroid hormone (PTH) and fragments and analogues thereof, estrogens, calcitonins, synthetic steroids, synthetic isoflavones, vitamin D analogues, vitamin K analogues, strontium salts, cathepsin K inhibitors, ⁇ v ⁇ 3 integrin (vitronectin) antagonists, prostaglandin (PGE2) receptor agonists and receptor activator of nuclear factor ⁇ B ligand (RANKL) inhibitors.
  • SERMs selective estrogen receptor modulators
  • PTH parathyroid hormone
  • PTH parathyroid hormone
  • PTH parathyroid hormone
  • PTH parathyroid hormone
  • synthetic steroids synthetic isoflavones
  • vitamin D analogues vitamin K analogues
  • Additional calcium receptor antagonists that can be used together with compounds of Formula I in the methods and compositions of this invention include those described in PCT International Publication Nos. WO 93/04373; WO 94/18959; WO 95/11211; WO 97/37967; WO 98/44925; WO 98/45255; WO 99/51241; WO 99/51569; WO 00/45816; WO 02/14259; WO 02/38106; WO 2004/041755; and WO 2005/030746; Nemeth, E. F.; Journal of Molecular Endocrinology (2002) 29, 15-21; Kessler, A. et al.; ChemBioChem (2004) 5, 1131; Steddon, S. J.
  • SERMs that can be used together with compounds of Formula I in the methods and compositions of this invention include, but are not limited to, lasofoxifene (Oporia®), raloxifene (Evista®), arzoxifene, ospemifene, Chiesi's CHF-4227 and Prostrakan's PSK-3471.
  • Bisphosphonates that can be used together with compounds of Formula I in the methods and compositions of this invention include, but are not limited to, tiludronate (Skelid®), clondronate (Bonefos®), etidronate (Didronel®), alendronate (Fosamax®), risedronate (Actonel®), ibandronate (Boniva®), zoledronate (Zometa®), minodronate (Onobis®), neridronate and pamidronate.
  • PTH is an 84 amino acid polypeptide produced by the parathyroid gland that controls serum calcium levels through its action on various cells.
  • PTH-related proteins PTH-related proteins
  • Parathyroid hormone (PTH) and fragments and analogues thereof that can be used together with compounds of Formula I in the methods and compositions of this invention include, but are not limited to, the full length PTH (such as PTH 1-84, Preos®/Preotact®, Unigene's 768974, Bone Medical's BN-003), the 1-31 (such as Zelos Therapeutics' Ostabolin-C), 1-34 (such as teriparatide, Forteo®, or Ipsen's BIM-44058) or 1-38 fragments.
  • PTH Parathyroid hormone
  • the full length PTH such as PTH 1-84, Preos®/Preotact®, Unigene's 768974, Bone Medical's BN-003
  • the 1-31 such as Zelos Therapeutics' Ostabolin-C
  • 1-34 such as teriparatide, Forteo®, or Ipsen's BIM-44058
  • Calcitonin is a 32 amino-acid peptide hormone produced by the thyroid gland which inhibits osteoclast activity by binding to calcitonin receptors on the surface of those cells.
  • Calcitonins that can be used together with compounds of Formula I in the methods and compositions of this invention include, but are not limited to, human calcitonin or salmon or eel calcitonins.
  • the calcitonins may be used as injectable or intranasal formulations such as Miacalcin®, Miacalcic®, Calcitonia®, Fortical® or Elcitonin® or as oral formulations such as Novartis' SMC-021, Bone Medical's BN-002 (Capsitonin®) or Nobex's NCT-025 (Oratonin®).
  • Synthetic steroids that can be used together with compounds of Formula I in the methods and compositions of this invention include, but are not limited to, mixed estrogen and progesterone agonists such as tibolone which is marketed as Livial®.
  • Synthetic isoflavones are chemically synthesized derivatives of plant isoflavones, such as phytoestrogens extracted from soy products.
  • a synthetic isoflavone that can be used together with compounds of Formula I in the methods and compositions of this invention includes, but is not limited to, ipraflavone which is marketed by Takeda as Iprosten® and Osten®.
  • Vitamin D analogues are compounds that act by binding to the nuclear vitamin D receptor in osteoblasts.
  • Vitamin D analogues that can be used together with compounds of Formula I in the methods and compositions of this invention include, but are not limited to, Chugai's ED-71 and Deltanoid's 2MD.
  • a strontium salt that can be used together with compounds of Formula I in the methods and compositions of this invention includes, but is not limited to, strontium ranelate (Servier's Protelos®).
  • Cathepsin K inhibitors that can be used together with compounds of Formula I in the methods and compositions of this invention include, but are not limited to, Novartis's AAE-581, balicatib, GlaxoSmithKline's SB-462795 and Merck's c-3578.
  • An ⁇ v ⁇ 3 integrin (vitronectin) antagonist that can be used together with compounds of Formula I in the methods and compositions of this invention includes, but is not limited to, Merck's MRL-123.
  • Prostaglandin E2 (PGE2) receptor agonists that can be used together with compounds of Formula I in the methods and compositions of this invention include, but are not limited to, PGE2 subtype 2 (EP2) receptor agonists, such as (3- ⁇ [4-Tert-butyl-benzyl)-(pyridine-3-sulfonyl)-amino]-methyl ⁇ -phenoxy)-acetic acid, or a pharmaceutically acceptable salt thereof or PGE2 subtype 4 (EP4) receptor agonists, such as ONO-4819.
  • a receptor activator of nuclear factor ⁇ B ligand (RANKL) inhibitor that can be used together with compounds of Formula I in the methods and compositions of this invention includes, but is not limited to, Amgen's RANKL antibody AMG-162.
  • Specific combinations of particular interest include compounds of Formula I and lasofoxifene or compounds of Formula I and (3- ⁇ [4-Tert-butyl-benzyl)-(pyridine-3-sulfonyl)-amino]-methyl ⁇ -phenoxy)-acetic acid, or a pharmaceutically acceptable salt thereof.
  • Compounds of the invention intended for pharmaceutical use may be administered as crystalline or amorphous products.
  • the compounds may be obtained, for example, as solid plugs, powders, or films by methods such as precipitation, crystallization, freeze drying, spray drying, or evaporative drying. Microwave or radio frequency drying may be used for this purpose.
  • the dosage administered will, of course, vary with the compound employed, the mode of administration, the treatment desired and the disorder indicated.
  • the total daily dosage of the compound of formula I/salt/solvate (active ingredient) will, generally, be in the range from 1 mg to 1 gram, preferably 1 mg to 250 mg, more preferably 10 mg to 100 mg.
  • the total daily dose may be administered in single or divided doses.
  • the present invention also encompasses sustained release compositions.
  • the pharmaceutical composition may, for example, be in a form suitable for oral administration as a tablet, capsule, pill, powder, sustained release formulations, solution, suspension, for parenteral injection as a sterile solution, suspension or emulsion, for topical administration as an ointment or cream or for rectal administration as a suppository.
  • the pharmaceutical composition may be in unit dosage forms suitable for single administration of precise dosages.
  • the pharmaceutical composition will include a conventional pharmaceutical carrier or excipient and a compound according to the invention as an active ingredient. In addition, it may include other medicinal or pharmaceutical agents, carriers, adjuvants, etc.
  • the dissolution rate of poorly water-soluble compounds may be enhanced by the use of a spray-dried dispersion, such as those described by Takeuchi, H., et al. in “Enhancement of the dissolution rate of a poorly water-soluble drug (tolbutamide) by a spray-drying solvent depostion method and disintegrants” J. Pharm. Pharmacol., 39, 769-773 (1987).
  • a spray-dried dispersion such as those described by Takeuchi, H., et al. in “Enhancement of the dissolution rate of a poorly water-soluble drug (tolbutamide) by a spray-drying solvent depostion method and disintegrants” J. Pharm. Pharmacol., 39, 769-773 (1987).
  • Exemplary parenteral administration forms include solutions or suspensions of active compounds in sterile aqueous solutions, for example, aqueous propylene glycol or dextrose solutions. Such dosage forms can be suitably buffered, if desired.
  • Suitable pharmaceutical carriers include inert diluents or fillers, water and various organic solvents.
  • the pharmaceutical compositions may, if desired, contain additional ingredients such as flavorings, binders, excipients and the like.
  • excipients such as citric acid
  • disintegrants such as starch, alginic acid and certain complex silicates
  • binding agents such as sucrose, gelatin and acacia.
  • lubricating agents such as magnesium stearate, sodium lauryl sulfate and talc are often useful for tableting purposes.
  • Solid compositions of a similar type may also be employed in soft and hard filled gelatin capsules.
  • Preferred materials, therefor, include lactose or milk sugar and high molecular weight polyethylene glycols.
  • the active compound therein may be combined with various sweetening or flavoring agents, coloring matters or dyes and, if desired, emulsifying agents or suspending agents, together with diluents such as water, ethanol, propylene glycol, glycerin, or combinations thereof.
  • diluents such as water, ethanol, propylene glycol, glycerin, or combinations thereof.
  • Dosage regimens may be adjusted to provide the optimum desired response. For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the specification for the dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the chemotherapeutic agent and the particular therapeutic or prophylactic effect to be achieved, and (b) the limitations inherent in the art of compounding such an active compound for the treatment of sensitivity in individuals.
  • the dose and dosing regimen is adjusted in accordance with methods well-known in the therapeutic arts. That is, the maximum tolerable dose can be readily established, and the effective amount providing a detectable therapeutic benefit to a patient may also be determined, as can the temporal requirements for administering each agent to provide a detectable therapeutic benefit to the patient. Accordingly, while certain dose and administration regimens are exemplified herein, these examples in no way limit the dose and administration regimen that may be provided to a patient in practicing the present invention.
  • dosage values may vary with the type and severity of the condition to be alleviated, and may include single or multiple doses. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and that dosage ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed composition. For example, doses may be adjusted based on pharmacokinetic or pharmacodynamic parameters, which may include clinical effects such as toxic effects and/or laboratory values. Thus, the present invention encompasses intra-patient dose-escalation as determined by the skilled artisan. Determining appropriate dosages and regiments for administration of the chemotherapeutic agent are well-known in the relevant art and would be understood to be encompassed by the skilled artisan once provided the teachings disclosed herein.
  • a pharmaceutical composition of the invention may be prepared, packaged, or sold in bulk, as a single unit dose, or as a plurality of single unit doses.
  • a “unit dose” is discrete amount of the pharmaceutical composition comprising a predetermined amount of the active ingredient.
  • the amount of the active ingredient is generally equal to the dosage of the active ingredient which would be administered to a subject or a convenient fraction of such a dosage such as, for example, one-half or one-third of such a dosage.
  • compositions of the invention will vary, depending upon the identity, size, and condition of the subject treated and further depending upon the route by which the composition is to be administered.
  • the composition may comprise between 0.1% and 100% (w/w) active ingredient.
  • a pharmaceutical composition of the invention may further comprise one or more additional pharmaceutically active agents.
  • additional agents include selective estrogen receptor modulators (SERMs), bisphosphonates, parathyroid hormone (PTH) and fragments and analogues thereof, estrogens, calcitonins, synthetic steroids, synthetic isoflavones, vitamin D analogues, vitamin K analogues, strontium salts, cathepsin K inhibitors, ⁇ v ⁇ 3 integrin (vitronectin) antagonists, prostaglandin (PGE2) receptor agonists and receptor activator of nuclear factor ⁇ B ligand (RANKL) inhibitors, such as those described hereinabove.
  • SERMs selective estrogen receptor modulators
  • PTH parathyroid hormone
  • PTH parathyroid hormone
  • RTKL nuclear factor ⁇ B ligand
  • Controlled- or sustained-release formulations of a pharmaceutical composition of the invention may be made using conventional technology.
  • parenteral administration of a pharmaceutical composition includes any route of administration characterized by physical breaching of a tissue of a subject and administration of the pharmaceutical composition through the breach in the tissue.
  • Parenteral administration thus includes, but is not limited to, administration of a pharmaceutical composition by injection of the composition, by application of the composition through a surgical incision, by application of the composition through a tissue-penetrating non-surgical wound, and the like.
  • parenteral administration is contemplated to include, but is not limited to, subcutaneous, intraperitoneal, intramuscular, intrasternal injection, and kidney dialytic infusion techniques.
  • Formulations of a pharmaceutical composition suitable for parenteral administration comprise the active ingredient combined with a pharmaceutically acceptable carrier, such as sterile water or sterile isotonic saline. Such formulations may be prepared, packaged, or sold in a form suitable for bolus administration or for continuous administration. Injectable formulations may be prepared, packaged, or sold in unit dosage form, such as in ampules or in multi-dose containers containing a preservative. Formulations for parenteral administration include, but are not limited to, suspensions, solutions, emulsions in oily or aqueous vehicles, pastes, and implantable sustained-release or biodegradable formulations as discussed below.
  • Such formulations may further comprise one or more additional ingredients including, but not limited to, suspending, stabilizing, or dispersing agents.
  • the active ingredient is provided in dry (i.e. powder or granular) form for reconstitution with a suitable vehicle (e.g. sterile pyrogen-free water) prior to parenteral administration of the reconstituted composition.
  • a composition of the present invention can be administered by a variety of methods known in the art.
  • the route and/or mode of administration vary depending upon the desired results.
  • the active compounds can be prepared with carriers that protect the compound against rapid release, such as a controlled release formulation, including implants, transdermal patches, and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Many methods for the preparation of such formulations are described by e.g., Sustained and Controlled Release Drug Delivery Systems, J. R. Robinson, ed., Marcel Dekker, Inc., New York, (1978).
  • Pharmaceutical compositions are preferably manufactured under GMP conditions.
  • compositions may be prepared, packaged, or sold in the form of a sterile injectable aqueous or oily suspension or solution.
  • This suspension or solution may be formulated according to the known art, and may comprise, in addition to the active ingredient, additional ingredients such as the dispersing agents, wetting agents, or suspending agents described herein.
  • Such sterile injectable formulations may be prepared using a non-toxic parenterally-acceptable diluent or solvent, such as water or 1,3-butane diol, for example.
  • Other acceptable diluents and solvents include, but are not limited to, Ringer's solution, isotonic sodium chloride solution, and fixed oils such as synthetic mono- or di-glycerides.
  • compositions for sustained release or implantation may comprise pharmaceutically acceptable polymeric or hydrophobic materials such as an emulsion, an ion exchange resin, a sparingly soluble polymer, or a sparingly soluble salt.
  • each active ingredient will vary depending upon any number of factors, including but not limited to, the type of animal and type of disease state being treated, the age of the animal and the route(s) of administration.
  • the amounts of various CaR antagonist compounds of formula I to be administered can be determined by standard procedures taking into account factors such as the compound IC 50 , EC 50 , the biological half-life of the compound, the age, size and weight of the patient, and the type of condition or symptom associated with the patient. The importance of these and other factors to be considered are known to those of ordinary skill in the art.
  • PTH secretion can be measured using techniques known in the art (see, e.g., U.S. Pat. No. 6,031,003, hereby incorporated by reference). For example, PTH secretion can be measured by first suspending cells in parathyroid cell buffer containing 0.5 mM CaCl 2 and 0.1% bovine serum albumin. Incubations can be performed in plastic tubes (Falcon 2058) containing 0.3 mL of the cell suspension with or without small volumes of CaCl 2 and/or organic polycations. After incubation at 37° C., typically 30 minutes, the tubes can then be placed on ice and the cells pelleted at 2° C.
  • parathyroid cell buffer containing 0.5 mM CaCl 2 and 0.1% bovine serum albumin.
  • Incubations can be performed in plastic tubes (Falcon 2058) containing 0.3 mL of the cell suspension with or without small volumes of CaCl 2 and/or organic polycations. After incubation at 37° C., typically 30 minutes, the tubes
  • Samples of the supernatant should then be brought to pH 4.5 with acetic acid and, if needed, stored at ⁇ 70° C.
  • the amount of PTH in bovine cell supernatants can be determined by a homologous radioimmunoassay using GW-1 antibody or its equivalent at a final dilution of 1/45,000.
  • 1251-PTH 65-84; INCSTAR, Stillwater, Minn.
  • Counting of samples and data reduction can be performed on a Packard Cobra 5005 gamma counter.
  • specific assays useful for evaluating the compounds of Formula I include the FLIPR Assay for Evaluating the Potency and Selectivity of Test Compounds; Assay for Evaluating the Effects of Test Compounds on Endogenous PTH Secretion; Evaluation of Effects of Test Compounds on PTH Secretion In Vivo; Effect of Calcium Receptor Antagonist Compound of Formula I on Body Weight, Body Composition and Bone Density in the Aged Intact and Ovariectomized Female Rat; and Fracture healing Assays as described below.
  • Human kidney cell HEK 293 expressing the calcium receptor (CasR) are used to detect antagonists of the receptor using Fluorometric imaging plate reader (FLIPR, Molecular Devices, Sunnyvale Calif.). Receptor activation by extracellular calcium results in the release of calcium from intracellular stores into the cytosol.
  • a fluorescent indicator (Fluo-4) is internalized by the cells from growth media and interacts with calcium released into the cytosol to provide a means of quantifying intracellular Ca 2+ levels and receptor agonism/antagonism. Fluorescence intensity is detected by the FLIPR CCD camera and traced as a function of time. Potential antagonists are identified by their ability to decrease this fluorescent response.
  • IC 50 values are loaded with Fluo4 (2.05 mM Fluo-4, 0.04% pluronic acid, 2.6 mM probenecid in 90% DMEM high glucose, 10% dialyzed Fetal Bovine Serum, 1 ⁇ Pen Strep, 1 ⁇ L-Glutamine, 3 ug/ml Puromycin, 27.5 nM Methotrexate) for 1 hour at 37° C.
  • Prior to the addition of test compound cells are washed with a 10 mM HEPES buffer solution.
  • the test compound for example the compound of Example 1 is added at various doses (from 1 ⁇ M to 3 nM) and pre-incubated with cells for 30 minutes followed by stimulation of the CasR by the addition of 1.7 mM Ca 2+ .
  • IC 50 values are based on the ability of the cells to inhibit the Ca 2+ induced increase in intracellular Ca 2+ . Fluorescence signal is read 42 seconds after the stimulation of the CasR by the addition of 1.7 mM Ca 2+ .
  • Test compounds are given to the animals by various routes of administration including subcutaneous injection, or intraveneous injection. Serum or plasma PTH concentrations are examined before and after dosing at various times using a commercially available rat intact PTH ELISA kit (Immutopics, Inc. San Clemente, Calif. Cat. #60-2500).
  • test compositions comprising compounds of Formula I in aged intact or ovariectomized (OVX) female rat model.
  • the compound of Formula I can be administered as a pharmaceutically acceptable salt or prodrug thereof.
  • Sprague-Dawley female rats are sham-operated or OVX at 18 months of age, while a group of rats is necropsied at day 0 to serve as baseline controls.
  • the rats are treated with either vehicle or test compound of Formula I, or a combination of test compound of Formula I and other active agent test compound for 59 days.
  • the vehicle or test compound of Formula I is administered either orally, by oral gavage, or by subcutaneous injection (s.c.), with the test compound being administered at a therapeutically effective dose.
  • mice All rats are given s.c. injection of 10 mg/kg of calcein (Sigma, St. Louis, Mo.) for fluorescent bone label 2 and 12 days before necropsy. On the day of necropsy, all rats under ketamine/xylazine anesthesia are weighed and undergoe dual-energy X-ray absorptiometry (DXA, QDR-4500/W, Hologic Inc., Waltham, Mass.) equipped with Rat Whole Body Scan software for lean and fat body mass determination. The rats are necropsied, then autopsied and blood is obtained by cardiac puncture. The distal femoral metaphysis and femoral shafts from each rat are analyzed by peripheral quantitative computerized tomography (pQCT), and volumetric total, trabecular and cortical bone mineral content and density are determined.
  • pQCT peripheral quantitative computerized tomography
  • pQCT Peripheral Quantitative Computerized Tomography
  • Excised femurs are scanned by a pQCT X-ray machine (Stratec XCT Research M, Norland Medical Systems, Fort Atkinson, Wis.) with software version 5.40.
  • a 1 millimeter (mm) thick cross section of the femur metaphysis is taken at 5.0 mm (proximal femoral metaphysis, a primary cancellous bone site) and 13 mm (femoral shafts, a cortical bone site) proximal from the distal end with a voxel size of 0.10 mm.
  • Cortical bone is defined and analyzed using contour mode 2 and cortical mode 4.
  • An outer threshold setting of 340 mg/cm 3 is used to distinguish the cortical shell from soft tissue and an inner threshold of 529 mg/cm 3 to distinguish cortical bone along the endocortical surface.
  • Trabecular bone is determined using peel mode 4 with a threshold of 655 mg/cm 3 to distinguish (sub)cortical from cancellous bone.
  • An additional concentric peel of 1% of the defined cancellous bone is used to ensure that (sub)cortical bone was eliminated from the analysis.
  • Volumetric content, density, and area are determined for both trabecular and cortical bone (Jamsa T. et al., Bone 23:155-161, 1998; Ke, H. Z. et al., Journal of Bone and Mineral Research, 16:765-773, 2001).
  • Fracture Technique Sprague-Dawley rats at 3 months of age are anesthetized with Ketamine. A 1 cm incision is made on the anteromedial aspect of the proximal part of the right tibia or femur. The following describes the tibial surgical technique. The incision is carried through to the bone, and a 1 mm hole is drilled 4 mm proximal to the distal aspect of the tibial tuberosity 2 mm medial to the anterior ridge. Intramedullary nailing is performed with a 0.8 mm stainless steel tube (maximum load 36.3 N, maximum stiffness 61.8 N/mm, tested under the same conditions as the bones). No reaming of the medullary canal is performed.
  • a standardized closed fracture is produced 2 mm above the tibiofibular junction by three-point bending using specially designed adjustable forceps with blunt jaws. To minimize soft tissue damage, care is taken not to displace the fracture.
  • the skin is closed with monofilament nylon sutures. The operation is performed under sterile conditions. Radiographs of all fractures are taken immediately after nailing, and rats with fractures outside the specified diaphyseal area or with displaced nails are excluded. The remaining animals are divided randomly into the following groups with 10-12 animals per each subgroup per time point for testing the fracture healing.
  • 10-12 rats from each group are anesthetized with Ketamine and sacrificed by exsanguination. Both tibiofibular bones are removed by dissection and all soft tissue is stripped. Bones from 5-6 rats for each group are stored in 70% ethanol for histological analysis, and bones from another 5-6 rats for each group are stored in a buffered Ringer's solution (+4° C., pH 7.4) for radiographs and biomechanical testing which is performed.
  • Sirius red stained sections are used to demonstrate the characteristics of the callus structure and to differentiate between woven bone and lamellar bone at the fracture site. The following measurements are performed: (1) fracture gap—measured as the shortest distance between the cortical bone ends in the fracture, (2) callus length and callus diameter, (3) total bone volume area of callus, (4) bony tissue per tissue area inside the callus area, (5) fibrous tissue in the callus, and (6) cartilage area in the callus.
  • Biomechanical Analysis The methods for biomechanical analysis have been previously published by Bak and Andreassen (The Effects of Aging on Fracture Healing in Rats. Calcif Tissue Int 45:292-297, 1989). Briefly, radiographs of all fractures are taken prior to the biomechanical test. The mechanical properties of the healing fractures are analyzed by a destructive three- or four-point bending procedure. Maximum load, stiffness, energy at maximum load, deflection at maximum load, and maximum stress are determined.
  • a calcium receptor antagonist may be usefully combined with another pharmacologically active compound, or with two or more other pharmacologically active compounds, particularly in the treatment of osteoporosis.
  • a calcium receptor antagonist particularly a compound of the formula I, or a pharmaceutically acceptable salt or solvate thereof, as defined above, may be administered simultaneously, sequentially or separately in combination with one or more agents selected from: selective estrogen receptor modulators (SERMs), bisphosphonates, parathyroid hormone (PTH) and fragments and analogues thereof, estrogens, calcitonins, synthetic steroids, synthetic isoflavones, vitamin D analogues, vitamin K analogues, strontium salts, cathepsin K inhibitors, ⁇ v ⁇ 3 integrin (vitronectin) antagonists, prostaglandin (PGE2) receptor agonists and receptor activator of nuclear factor ⁇ B ligand (RANKL) inhibitors as described hereinabove.
  • SERMs selective estrogen receptor modulators
  • PTH parat
  • Ammonia refers to a concentrated solution of ammonia in water possessing a specific gravity of 0.88. Where thin layer chromatography (TLC) has been used it refers to silica gel TLC using silica gel 60 F 254 plates, R f is the distance traveled by a compound divided by the distance traveled by the solvent front on a TLC plate. HPLC refers to high performance liquid chromatography.
  • the solvent was evaporated under reduced pressure and dissolved in a minimum volume of water.
  • the aqueous layer was washed with water and acidified to pH 4 with conc. HCl. It was then extracted with ethyl acetate, the organic layer was washed with brine and dried over sodium sulfate.
  • the organic solvent was removed under reduced pressure to provide 2,4-dichloronicotinic acid (10.6 g, 82%) as off white solid.
  • Methyl 4-azido-2-chloronicotinate (15 g, 70.75 mmol) produced methyl 4-amino-2-chloronicotinate (11.2 g, 85%) as light yellow solid following the literature procedure, Tetrahedron letters, 2002; 43, 6629-6631 (reduction of the azido group in methyl 4-azido-2-chloronicotinate to the corresponding amine by stirring methyl 4-azido-2-chloronicotinate in aqueous HI at room temperature).
  • Methyl 4-amino-2-chloronicotinate (21.0 g, 112.9 mmol) and LiOH (10.3 g, 247.5 mmol) was taken in a mixture of dioxane: MeOH:water (3:2:1, ml) and the reaction mixture was heated to 85° C. for 2 hours. The solvent was evaporated under reduced pressure, the residue was dissolved in minimum volume of water and acidified (up to pH 4) with saturated citric acid. The aqueous solution was concentrated until precipitation just started. The mixture was allowed to stand overnight at 10° C. for complete precipitation.
  • hexachloroethane (29.4 g, 124 mmol) is dissolved in THF (200 mL) and cooled to ⁇ 15° C. and stirred rapidly.
  • the solution containing the lithiated pyridine is added to the hexachloroethane solution via canula. After complete addition, the mixture is allowed to ward to room temperature. Water (ca 200 mL) is added, followed by removal of the tetrathydrofuran in vacuo. The remaining aqueous residue is extracted with ether-pentane (1:1, 100 mL). The organic layer was discarded.
  • the aqueous layer is acidified with hydrochloric acid (ca.
  • 2-(2-Hydroxy-phenyl)-3-phenethyl-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one) can be prepared using 4-amino-2-(trifluoromethyl)nicotinic acid as the starting material.
  • the product can be converted to 2-(2-Hydroxy-phenyl)-3-phenethyl-5-trifluoromethyl-3H-pyrido[4,3-d]pyri-mid in-4-one) by using the title compound above, Preparation 1f, and following the procedures described in Preparation 1b (condensation with an appropriate aldehyde R 2′ CHO), Preparation 1c (oxidation with KMnO 4 ) and the deprotection step using BCl 3 (when converting a methoxy moiety in R 2′ to the hydroxy in R 2 ) as described above.
  • Example 6 The racemic compound of Example 6 can be separated into its single enantiomer components (Examples 6a and 6b) by preparative chromatography on a ChiralpakTM AS (10 cm ⁇ 50 cm) column (Daicel (U.S.A.) Chemical Industries, Ltd, Fort Lee, N.J. 07024, U.S.A.) using Heptane/EtOH (90/10) as mobile phase at a flow rate of 475 mL/min.
  • Enantiomer 1 2-(3-Hydroxy-pyridin-2-yl)-3-(1-methyl-2-(2-fluorophenyl)ethyl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Enantiomer 2 2-(3-Hydroxy-pyridin-2-yl)-3-(1-methyl-2-(2-fluorophenyl)ethyl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Examples 17-23 may be prepared in a manner analogous to Example 16 using procedures from Example 16 with the corresponding amine followed by deprotection conditions employing BCl 3 found in the final step of Example 1.
  • Examples 25-29 may be prepared in a manner analogous to Example 24 using procedures from Example 24 with the corresponding boronic acid followed by deprotection conditions employing BCl 3 and isolation conditions found in the final step of Example 1.
  • Examples 31-36 may be prepared in a manner analogous to Example 30 using procedures from Example 30 with the corresponding alcohol followed by H-cube hydrogenation.
  • Example 37 The racemic compound of Example 37 can be separated into its single enantiomer components (Examples 37a and 37b) by preparative chromatography on a Chiralcel OD-H (2.1 cm ⁇ 25 cm) column (Daicel (U.S.A.) Chemical Industries, Ltd, Fort Lee, N.J. 07024, U.S.A.) using MeOH/CHCL 3 (3/1) as mobile phase at a flow rate of 65 g/min.
  • Chiralcel OD-H (2.1 cm ⁇ 25 cm) column Daicel (U.S.A.) Chemical Industries, Ltd, Fort Lee, N.J. 07024, U.S.A.

Abstract

The present invention is directed to novel pyrido[4,3-d]pyrimidin-4(3H)-one derivatives and pharmaceutically acceptable salts thereof of structural formula I
Figure US20110028452A1-20110203-C00001
wherein the variables R1, R2, R3, R4 and R5 are as described herein. Also provided are pharmaceutical compositions comprising the compounds of formula I as well as methods of treatment employing compounds of formula I to treat a disease or disorder characterized by abnormal bone or mineral homeostasis such as hypoparathyroidism, osteoporosis, osteopenia, periodontal disease, Paget's disease, bone fracture, osteoarthritis, rheumatoid arthritis, and humoral hypercalcemia of malignancy.

Description

  • This application is a divisional of U.S. patent application Ser. No. 11/867,255 filed Oct. 4, 2007, which claims the benefit of Provisional Patent Application Nos. 60/828,162 filed Oct. 4, 2006 and 60/969,083 filed Aug. 30, 2007.
  • FIELD OF INVENTION
  • The present invention is directed toward novel pyrido[4,3-d]pyrimidin-4(3H)-one derivatives, pharmaceutical compositions containing these compounds, methods for their use and processes for their production. These novel pyrido[4,3-d]pyrimidin-4(3H)-one derivatives are able to inhibit calcium receptor activity and thus act as calcium receptor antagonists.
  • BACKGROUND OF THE INVENTION
  • In mammals, extracellular Ca2+ is under rigid homeostatic control with the serum calcium concentration strictly maintained at a concentration of approximately 1.1 to 1.3 mM in a healthy mammal. The extracellular Ca2+ homeostasis depends on integrated regulation of Ca2+ fluxes with respect to the intestine, kidneys and bone. The extracellular Ca2+ regulates various processes such as blood coagulation, nerve and muscle excitability, and normal bone homeostasis. When the Ca2+ serum concentration decreases by 50% tetania occurs, and when the Ca2+ serum concentration increases by 50% consciousness is clouded, in both instances a potentially life threatening circumstance. Extracellular Ca2+ also inhibits the secretion of parathyroid hormone (PTH) from parathyroid cells, inhibits bone resorption by osteoclasts, stimulates secretion of calcitonin from C-cells and is involved in re-absorption and excretion in the kidney.
  • The extracellular calcium-sensing receptor (CaSR) is a hormone-like receptor, more particularly a plasma membrane-bound G protein-coupled receptor (GPCR) that belongs to family 3 of the GPCR superfamily. Family 3 of the GPCR superfamily includes metabotropic glutamate receptors (mGluRs), γ-aminobutyric acid B-type receptors (GABABRs) as well as putative pheromone and taste receptors. The CaSR has a large extracellular domain exhibiting “Venus flytrap” topology, a seven-transmembrane domain and a relatively large cytoplasmic domain. Human CaSR consists of 1078 amino acids and shares 93% amino acid homology with bovine CaSR. The CaSR senses and is activated by changes in extracellular Ca2+ levels. The presence of CaSR on certain specialized cells enables those Ca2+-sensing cells to respond to changes in extracellular Ca2+ concentration. Examples of Ca2+-sensing cells include the parathyroid-secreting cells of the parathyroid gland, the calcitonin-secreting C cells of the thyroid gland and certain cells in the kidney. In addition, the CaSR has been found in a wide variety of other tissues including intestine, bone, bone marrow, brain, skin, pancreas, lung and heart.
  • The CaSR on the surface of parathyroid chief cells is the primary entity that regulates secretion of PTH from parathyroid cells. Activation of the CaSR on parathyroid chief cells by extracellular Ca2+ suppresses PTH production and secretion, inhibits parathyroid cellular proliferation and likely inhibits PTH gene expression. The CaSR on the surface of the calcitonin-secreting C cells of the thyroid gland mediate the stimulatory action of high extracellular Ca2+ concentration on calcitonin secretion, thereby increasing the circulating level of the Ca2+-lowering hormone calcitonin. The CaSR is also present in the kidney, along much of the nephrons and at the basolateral surface in the cortical thick ascending limb. In the basolateral surface in the cortical thick ascending limb the CaSR is thought to mediate high Ca2+-induced inhibition of the tubular re-absorption of Ca2+ and magnesium. A reduction of renal cortical synthesis of 1,25(OH)2 vitamin D and polyuria with dilute urine are partially the result of hypercalcaemic activation of the CaSR in the nephron.
  • PTH is the primary endocrine hormone regulating Ca2+ homeostasis in the blood and extracellular fluids. PTH, by acting on bone and kidney cells, increases the level of Ca2+ in the plasma. This increase in plasma Ca2+ concentration then acts as a negative feedback signal, thereby depressing PTH secretion. The reciprocal relationship between extracellular Ca2+ and PTH secretion forms an important mechanism for maintaining bodily Ca2+ homeostasis. PTH has been found to increase bone turnover, but the overall effect on bone is dependent on temporal changes in circulating levels of PTH. Sustained elevations in circulating plasma PTH levels, as occurs in hyperparathyroidism, have been found to result in a net catabolic effect on bone. By contrast, transient increases in plasma PTH levels, achieved by daily or near daily injection of exogenous hormone, have been found to exhibit a net anabolic effect on bone. The effect of PTH on bone is likely due to PTH being able to induce a rapid release of calcium from bone and mediate other changes by acting directly on osteoblasts and indirectly on osteoclasts. PTH affects cellular metabolic activity, ion transport, cell shape, gene transcriptional activity and secretion of proteases in osteoblasts. Also, PTH stimulates the production of RANKL, a protein that plays a crucial role in osteoclast differentiation and activity.
  • Various compounds are known to modulate the effects of extracellular Ca2+ on the CaSR. Calcimimetics are agents that act as allosteric modulators of the CaSR that increase the sensitivity of the CaSR to activation by extracellular Ca2+. Calcilytics, or calcium receptor antagonists, are agents that act as modulators of the CaSR that inhibit CaSR activity. This inhibition of the CaSR activity results in a decrease of one or more CaSR activities that are evoked by extracellular Ca2+.
  • Certain urea derivatives, such as those disclosed in PCT International Publication WO 02/059102, are described as having calcimimetic activity. In addition, certain phenylalkylamine derivatives have been identified as calcimimetics. Phenylalkylamine calcimimetic compounds include (R)-N-(1-(3-methoxyphenyl)ethyl)-3-phenylpropan-1-amine hydrochloride (NPS-467); (R)-3-(2-chlorophenyl)-N-(1-(3-methoxyphenyl)ethyl)propan-1-amine hydrochloride (NPS R-568, tecalcet hydrochloride) and (R)-(−)-N-(1-(naphthalen-1-yl)ethyl)-3-(3-(trifluoromethyl)phenyl)propan-1-amine hydrochloride (NPS-1493, cinacalcet hydrochloride). Cinacalcet hydrochloride and uses thereof are disclosed in U.S. Pat. Nos. 6,011,068; 6,031,003; 6,211,244 and 6,313,146. Cinacalcet hydrochloride is marketed as Sensipar® and Minpara® in the U.S. and Europe, respectively, and is indicated for the treatment of secondary hyperparathyroidism in patients with chronic kidney disease on dialysis and for hypercalcemia in patients with parathyroid carcinoma.
  • Calcilytics, or calcium receptor antagonists, have been described in various publications such as PCT International Publication Nos. WO 93/04373; WO 94/18959; WO 95/11211; WO 97/37967; WO 98/44925; WO 98/45255; WO 99/51241; WO 99/51569; WO 00/45816; WO 02/14259; WO 02/38106; WO 2004/041755; and WO 2005/030746; Nemeth, E. F.; Journal of Molecular Endocrinology (2002) 29, 15-21; Kessler, A. et al.; ChemBioChem (2004) 5, 1131; Steddon, S. J. et al.; Lancet (2005) 365, 2237-2239; and Shcherbakova, I.; et al.; Bioorganic & Medicinal Chemistry Letters (2005) 15, 1557-1560.
  • Calcium receptor antagonists are useful in the treatment of various disease states characterized by abnormal levels of one or more components, e.g., polypeptides such as hormones, enzymes or growth factors, the expression and/or secretion of which is regulated or affected by activity at one or more CaSR. Target diseases or disorders for calcium receptor antagonists include diseases involving abnormal bone and mineral homeostasis. Abnormal calcium homeostasis is characterized by one or more of the following activities: an abnormal increase or decrease in serum calcium; an abnormal increase or decrease in urinary excretion of calcium; an abnormal increase or decrease in bone calcium levels (for example, as assessed by bone mineral density measurements); an abnormal absorption of dietary calcium; an abnormal increase or decrease in the production and/or release of messengers which affect serum calcium levels such as PTH and calcitonin; and an abnormal change in the response elicited by messengers which affect serum calcium levels.
  • The novel calcium receptor antagonists of this invention are useful in the treatment of diseases associated with abnormal bone or mineral homeostasis. Thus, these calcium receptor antagonists are useful in the treatment of hypoparathyroidism, osteoporosis, osteopenia, periodontal disease, bone fracture, osteoarthritis, rheumatoid arthritis, Paget's disease, humoral hypercalcemia associated with malignancy.
  • SUMMARY OF THE INVENTION
  • The present invention is directed towards calcium receptor antagonist compounds, pharmaceutical compositions containing the calcium receptor antagonist compounds and methods of treatment employing the calcium receptor antagonist compounds.
  • More specifically, the present invention is directed to calcium receptor antagonists that are pyrido[4,3-d]pyrimidin-4(3H)-one derivatives of structural formula I
  • Figure US20110028452A1-20110203-C00002
  • wherein
    R1 is -Q or (C1-C6)alkyl-Q;
    R2 is (C1-C6)alkyl, (C3-C7)cycloalkyl, aryl or heteroaryl; wherein said aryl or heteroaryl is substituted with hydroxy and additionally said (C1-C6)alkyl, (C3-C7)cycloalkyl, aryl or heteroaryl within the definition of R2 is optionally substituted with one to three substituents independently selected from halo, cyano, trifluoromethyl, trifluoromethoxy, (C1-C6)alkyl, (C1-C6)alkoxy or hydroxy;
    R3 is hydrogen, (C1-C6)alkyl, (C3-C7)cycloalkyl, (C1-C6)alkyl-Q, aryl, heteroaryl, OR6, or NR7R8; wherein said (C1-C6)alkyl, (C3-C7)cycloalkyl, aryl or heteroaryl is optionally substituted with one to three substituents independently selected from halo, cyano, trifluoromethyl, trifluoromethoxy, (C1-C6)alkyl; (C1-C6)alkoxy, NR7R8 or hydroxy;
    R4 and R5 are each independently hydrogen, halo, cyano, (C1-C6)alkyl optionally substituted with one to three fluoro, aryl, heteroaryl, or OR6;
    R6 at each occurrence is independently (C1-C6)alkyl, (C3-C7)cycloalkyl, (C3-C7)cycloalkyl-(C1-C6)alkyl, aryl, heteroaryl, (C1-C6)alkylaryl, or (C1-C6)alkylheteroaryl; each of said (C1-C6)alkyl, (C3-C7)cycloalkyl, (C3-C7)cycloalkyl-(C1-C6)alkyl, aryl, heteroaryl, (C1-C6)alkylaryl, or (C1-C6)alkylheteroaryl optionally substituted with one to three substituents independently selected from halo, hydroxy or (C1-C3)alkyl;
    R7 and R8, at each occurrence, are independently hydrogen, (C1-C6)alkyl, or (C3-C7)cycloalkyl; or R7 and R8 taken together with the nitrogen to which they are attached form a 3 to 7 membered fully saturated, partially saturated or fully unsaturated ring optionally containing one to two additional heteroatoms independently selected from N(R9)n; O or S(O)p;
    n is 0 or 1;
    p is 0, 1 or 2;
    R9 is hydrogen or (C1-C6)alkyl;
    Q, at each occurrence, is independently aryl or heteroaryl; wherein said aryl or heteroaryl is optionally substituted with one to three substituents independently selected from halo, cyano, trifluoromethyl, trifluoromethoxy, (C1-C6)alkyl, (C1-C6)alkoxy or hydroxy;
    or a pharmaceutically acceptable salt thereof.
  • “Halo” refers to fluoro, chloro, bromo or iodo.
  • “(C1-C6)alkyl” refers to a hydrocarbon group having one to six carbon atoms joined together by single carbon-carbon bonds. The (C1-C6)alkyl group may be straight-chain or contain one or more branches and may be unsubstituted or substituted as specified. Examples of (C1-C6)alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, tert-butyl, n-pentyl, (1-methyl)butyl, (2-methyl)butyl, (3-methyl)butyl, (1,2-dimethyl)propyl, n-hexyl, (1-methyl)pentyl, (2-methyl)pentyl, (3-methyl)pentyl, (4-methyl)pentyl, (1-ethyl)butyl, (2-ethyl)butyl, (1,2-dimethyl)butyl, (1,3-dimethyl)butyl, (2,3-dimethyl)butyl and the like.
  • “(C1-C6)alkoxy” refers to an oxygen joined to a (C1-C6)alkyl group. The (C1-C6)alkyl group in the (C1-C6)alkoxy moiety may be straight-chain or contain one or more branches and may be unsubstituted or substituted as specified. Examples of (C1-C6)alkoxy groups include methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, sec-butoxy, tert-butoxy, n-pentoxy and the like. Likewise, alkyl or alkoxy groups of differing length such as “(C1-C3)alkyl” or “(C1-C3)alkoxy” are defined in the same manner but limited to the number of carbons present.
  • “(C3-C7)cycloalkyl” refers to a saturated carbocyclic group having three to seven carbons and encompasses cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl. The (C3-C7)cycloalkyl group can be unsubstituted or substituted as specified.
  • “Aryl” refers to a six to sixteen membered carbocyclic aromatic group having at least one ring with a conjugated pi-electron system. The aryl group can have conjugated or fused rings and can be unsubstituted or substituted as specified. Examples of aryl groups include phenyl, naphthalenyl, anthracenyl, phenanthrenyl, azulenyl and biphenyl.
  • “Heteroaryl” refers to a five to sixteen membered aromatic group with at least one ring with a conjugated pi-electron system and containing one to four heteroatoms such as N, O or S. The hetroaryl group can have conjugated or fused rings and can be unsubstituted or substituted as specified. Examples of heteroaryl groups include thienyl, furanyl, pyrrolyl, pyrazolyl, imidazoyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, triazolyl, oxadiazolyl, thiadiazolyl, tetrazolyl, pyridinyl, pyridizinyl, pyrimidinyl, pyrazinyl, triazinyl, indolyl, isoindolyl, indolizinyl, benzofuranyl, benzothienyl, indazolyl, benzimidazoyl, benzthiazolyl, purinyl, quinolizinyl, quinolinyl, isoquinolinyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, naphthyridinyl, pteridinyl, carbazolyl, acridinyl, phenazinyl, phenothiazinyl or phenoxazinyl.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 is a chart depicting plasma PTH levels from the time of intravenous injection to 480 minutes following intravenous injection of 1 mg/kg of the compound of Example 1, 2-(2-Hydroxy-phenyl)-3-phenethyl-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one), in normal rats.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention provides novel pyrido[4,3-d]pyrimidin-4(3H)-one derivatives and pharmaceutically acceptable salts thereof of structural formula I
  • Figure US20110028452A1-20110203-C00003
  • wherein the variables R1, R2, R3, R4 and R5 are described hereinabove. The pharmaceutically acceptable salts of the compounds of formula I include the acid addition and base salts (including disalts) thereof.
  • Suitable acid addition salts are formed from acids which form non-toxic salts. Examples include the acetate, aspartate, benzoate, besylate, bicarbonate/carbonate, bisulphate/sulphate, borate, camsylate, citrate, edisylate, esylate, formate, fumarate, gluceptate, gluconate, glucuronate, hexafluorophosphate, hibenzate, hydrochloride/chloride, hydrobromide/bromide, hydroiodide/iodide, isethionate, lactate, malate, maleate, malonate, mesylate, methylsulphate, naphthylate, 2-napsylate, nicotinate, nitrate, orotate, oxalate, palmitate, pamoate, phosphate/hydrogen phosphate/dihydrogen phosphate, saccharate, stearate, succinate, tartrate, tosylate and trifluoroacetate salts.
  • Suitable base salts are formed from bases which form non-toxic salts. Examples include the aluminium, arginine, benzathine, calcium, choline, diethylamine, diolamine, glycine, lysine, magnesium, meglumine, olamine, potassium, sodium, tromethamine and zinc salts. For a review on suitable salts, see “Handbook of Pharmaceutical Salts: Properties, Selection, and Use” by Stahl and Wermuth (Wiley-VCH, Weinheim, Germany, 2002).
  • A pharmaceutically acceptable salt of a compound of formula I may be readily prepared by mixing together solutions of the compound of formula I and the desired acid or base, as appropriate. The salt may precipitate from solution and be collected by filtration or may be recovered by evaporation of the solvent. The degree of ionization in the salt may vary from completely ionized to almost non-ionized.
  • The compounds of the invention include compounds of formula I as hereinbefore defined, polymorphs, and isomers thereof (including optical, geometric and tautomeric isomers) as hereinafter defined and isotopically-labeled compounds of formula I.
  • The compounds of the present invention may be administered as prodrugs. Thus certain derivatives of compounds of formula I which may have little or no pharmacological activity themselves can, when administered into or onto the body, be converted into compounds of formula I having the desired activity, for example, by hydrolytic cleavage. Such derivatives are referred to as ‘prodrugs’. Further information on the use of prodrugs may be found in ‘Pro-drugs as Novel Delivery Systems, Vol. 14, ACS Symposium Series (T Higuchi and W Stella) and ‘Bioreversible Carriers in Drug Design’, Pergamon Press, 1987 (ed. E B Roche, American Pharmaceutical Association).
  • Prodrugs can, for example, be produced by replacing appropriate functionalities present in the compounds of formula I with certain moieties known to those skilled in the art as ‘pro-moieties’ as described, for example, in “Design of Prodrugs” by H Bundgaard (Elsevier, 1985).
  • Some examples of such prodrugs include:
  • (i) where the compound of formula I contains a carboxylic acid functionality (—COOH), an ester thereof, for example, replacement of the hydrogen with (C1-C8)alkyl;
    (ii) where the compound of formula I contains an alcohol functionality (—OH), an ether thereof, for example, replacement of the hydrogen with (C1-C6)alkanoyloxymethyl; and
    (iii) where the compound of formula I contains a primary or secondary amino functionality (—NH2 or —NHR where R is not H), an amide thereof, for example, replacement of one or both hydrogens with (C1-C10)alkanoyl. Further examples of replacement groups in accordance with the foregoing examples and examples of other prodrug types may be found in the aforementioned references. Finally, certain compounds of formula I may themselves act as prodrugs of other compounds of formula I.
  • Compounds of formula I containing one or more asymmetric carbon atoms can exist as two or more stereoisomers. Where a compound of formula I contains an alkenyl or alkenylene group, geometric cis/trans (or Z/E) isomers are possible. Where the compound contains, for example, a keto or oxime group or an aromatic moiety, tautomeric isomerism (‘tautomerism’) can occur. It follows that a single compound may exhibit more than one type of isomerism.
  • Included within the scope of the claimed compounds of formula I are all stereoisomers, geometric isomers and tautomeric forms of the compounds of formula I, including compounds exhibiting more than one type of isomerism, and mixtures of one or more thereof. Also included are acid addition or base salts wherein the counterion is optically active, for example, D-lactate or L-lysine, or racemic, for example, DL-tartrate or DL-arginine. Cis/trans isomers may be separated by conventional techniques well known to those skilled in the art, for example, chromatography and fractional crystallisation. Conventional techniques for the preparation/isolation of individual enantiomers include chiral synthesis from a suitable optically pure precursor or resolution of the racemate (or the racemate of a salt or derivative) using, for example, chiral high pressure liquid chromatography (HPLC).
  • Alternatively, the racemate (or a racemic precursor) may be reacted with a suitable optically active compound, for example, an alcohol, or, in the case where the compound of formula (I) contains an acidic or basic moiety, an acid or base such as tartaric acid or 1-phenylethylamine. The resulting diastereomeric mixture may be separated by chromatography and/or fractional crystallization and one or both of the diastereoisomers converted to the corresponding pure enantiomer(s) by means well known to a skilled person. Chiral compounds of the invention (and chiral precursors thereof) may be obtained in enantiomerically-enriched form using chromatography, typically HPLC, on an asymmetric resin with a mobile phase consisting of a hydrocarbon, typically heptane or hexane, containing from 0 to 50% isopropanol, typically from 2 to 20%, and from 0 to 5% of an alkylamine, typically 0.1% diethylamine. Concentration of the eluate affords the enriched mixture. Mixtures of stereoisomers may be separated by conventional techniques known to those skilled in the art. [see, for example, “Stereochemistry of Organic Compounds” by E L Eliel (Wiley, New York, 1994).]
  • The present invention includes all pharmaceutically acceptable isotopically-labelled compounds of formula I wherein one or more atoms are replaced by atoms having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
  • Examples of isotopes suitable for inclusion in the compounds of the invention include isotopes of hydrogen, such as 2H and 3H, carbon, such as 11C, 13C and 14C, chlorine, such as 38Cl, fluorine, such as 18F, iodine, such as 123I and 125I, nitrogen, such as 13N and 15N, oxygen, such as 15O, 17O and 18O, phosphorus, such as 32P, and sulfur, such as 35S. Certain isotopically-labelled compounds of formula I, for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies. The radioactive isotopes tritium, i.e. 3H, and carbon-14, i.e. 14C, are particularly useful for this purpose in view of their ease of incorporation and ready means of detection. Substitution with heavier isotopes such as deuterium, i.e. 2H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements, and hence may be preferred in some circumstances. Substitution with positron emitting isotopes, such as 11C, 18F, 15O and 13N, can be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy. Isotopically-labeled compounds of formula (I) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the accompanying Examples and Preparations using an appropriate isotopically-labeled reagents in place of the non-labeled reagent previously employed.
  • A preferred embodiment of the present invention are compounds of Formula I as described above wherein R2 is aryl or heteroaryl, wherein said aryl or heteroaryl is substituted with hydroxy and optionally substituted with one to three substituents independently selected from (C1-C6)alkoxy or halo; or a pharmaceutically acceptable salt thereof. Another preferred embodiment of this invention is a compound of Formula I, wherein R2 is 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl; wherein said 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl is optionally substituted with one to two substituents independently selected from (C1-C6)alkoxy or fluoro; or a pharmaceutically acceptable salt thereof. Yet another preferred embodiment of the present invention is a compound of Formula I as described above wherein R1 is (C1-C6)alkyl-Q; and Q is phenyl optionally substituted with one or two fluoro; or a pharmaceutically acceptable salt thereof.
  • Another preferred embodiment of the present invention is a compound of Formula I, wherein R1 is phenethyl or 1-methyl-2-(phenyl)ethyl, wherein said phenethyl or 1-methyl-2-(phenyl)ethyl is optionally substituted on phenyl with one or two halo; and R2 is 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl, wherein said 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl is optionally substituted with one or two substituents independently selected from (C1-C3)alkyl or halo; or a pharmaceutically acceptable salt thereof.
  • Another preferred embodiment of the present invention is a compound of Formula I, wherein R1 is phenethyl or 1-methyl-2-(phenyl)ethyl, wherein said phenethyl or 1-methyl-2-(phenyl)ethyl is optionally substituted on phenyl with one or two halo; and R2 is 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl, wherein said 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl is optionally substituted with one or two substituents independently selected from (C1-C3)alkyl or halo; R3 is (C1-C6)alkyl, (C1-C6)alkyl-Q, aryl, heteroaryl, OR6, or NR7R8, wherein said (C1-C6)alkyl, aryl or heteroaryl is optionally substituted with one to three substituents independently selected from halo, cyano, trifluoromethyl, trifluoromethoxy, (C1-C6)alkyl, (C1-C6)alkoxy, NR7R8 or hydroxy; and R4 and R5 are each hydrogen; or a pharmaceutically acceptable salt thereof.
  • Another preferred embodiment of the present invention is a compound of Formula I, wherein R1 is phenethyl or 1-methyl-2-(phenyl)ethyl, wherein said phenethyl or 1-methyl-2-(phenyl)ethyl is optionally substituted on phenyl with one or two halo; R2 is 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl, wherein said 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl is optionally substituted with one or two substituents independently selected from (C1-C3)alkyl or halo; R3 is trifluoromethyl; and R4 and R5 are each hydrogen; or a pharmaceutically acceptable salt thereof.
  • Another preferred embodiment of the present invention is a compound of Formula I, wherein R1 is phenethyl or 1-methyl-2-(phenyl)ethyl, wherein said phenethyl or 1-methyl-2-(phenyl)ethyl is optionally substituted on phenyl with one or two halo; and R2 is 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl, wherein said 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl is optionally substituted with one or two substituents independently selected from (C1-C3)alkyl or halo; R3 is (C1-C6)alkyl; and R4 and R5 are each hydrogen; or a pharmaceutically acceptable salt thereof.
  • Another preferred embodiment of the present invention is a compound of Formula I, wherein R1 is phenethyl or 1-methyl-2-(phenyl)ethyl, wherein said phenethyl or 1-methyl-2-(phenyl)ethyl is optionally substituted on phenyl with one or two halo; and R2 is 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl, wherein said 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl is optionally substituted with one or two substituents independently selected from (C1-C3)alkyl or halo; R3 is (C1-C6)alkyl-Q; and R4 and R5 are each hydrogen; or a pharmaceutically acceptable salt thereof.
  • Another preferred embodiment of the present invention is a compound of Formula I, wherein R1 is phenethyl or 1-methyl-2-(phenyl)ethyl, wherein said phenethyl or 1-methyl-2-(phenyl)ethyl is optionally substituted on phenyl with one or two halo; and R2 is 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl, wherein said 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl is optionally substituted with one or two substituents independently selected from (C1-C3)alkyl or halo; R3 is aryl, wherein said aryl is optionally substituted with one to three substituents independently selected from halo, cyano, trifluoromethyl, trifluoromethoxy, (C1-C6)alkyl, (C1-C6)alkoxy, NR7R8 or hydroxy; and R4 and R5 are each hydrogen; or a pharmaceutically acceptable salt thereof.
  • Another preferred embodiment of the present invention is a compound of Formula I, wherein R1 is phenethyl or 1-methyl-2-(phenyl)ethyl, wherein said phenethyl or 1-methyl-2-(phenyl)ethyl is optionally substituted on phenyl with one or two halo; and R2 is 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl, wherein said 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl is optionally substituted with one or two substituents independently selected from (C1-C3)alkyl or halo; R3 is heteroaryl; wherein said heteroaryl is optionally substituted with one to three substituents independently selected from halo, cyano, trifluoromethyl, trifluoromethoxy, (C1-C6)alkyl, (C1-C6)alkoxy, NR7R8 or hydroxy; and R4 and R5 are each hydrogen; or a pharmaceutically acceptable salt thereof.
  • Another preferred embodiment of the present invention is a compound of Formula I, wherein R1 is phenethyl or 1-methyl-2-(phenyl)ethyl, wherein said phenethyl or 1-methyl-2-(phenyl)ethyl is optionally substituted on phenyl with one or two halo; and R2 is 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl, wherein said 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl is optionally substituted with one or two substituents independently selected from (C1-C3)alkyl or halo; R3 is OR6; and R4 and R5 are each hydrogen; or a pharmaceutically acceptable salt thereof.
  • Another preferred embodiment of the present invention is a compound of Formula I, wherein R1 is phenethyl or 1-methyl-2-(phenyl)ethyl, wherein said phenethyl or 1-methyl-2-(phenyl)ethyl is optionally substituted on phenyl with one or two halo; and R2 is 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl, wherein said 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl is optionally substituted with one or two substituents independently selected from (C1-C3)alkyl or halo; R3 is NR7R8; and R4 and R5 are each hydrogen; or a pharmaceutically acceptable salt thereof.
  • A preferred embodiment of the present invention is a compound of Formula I wherein R3 is trifluoromethyl; or a pharmaceutically acceptable salt thereof. Another preferred embodiment of the present invention is a compound of Formula I wherein R3 is trifluoromethyl; and R4 and R5 are each independently hydrogen or methyl; or a pharmaceutically acceptable salt thereof. Yet another preferred embodiment of the present invention is a compound of Formula I wherein R2 is aryl or heteroaryl, wherein said aryl or heteroaryl is substituted with hydroxy and optionally substituted with one to three substituents independently selected from (C1-C3)alkyl, (C1-C3)alkoxy or fluoro; R3 is trifluoromethyl; and R4 and R5 are each independently hydrogen or methyl; or a pharmaceutically acceptable salt thereof. Yet another preferred embodiment of the present invention is a compound of Formula I wherein R2 is hydroxy-phenyl or hydroxy-pyridyl, wherein said hydroxy-phenyl or hydroxy-pyridyl is optionally substituted with one to three substituents independently selected from methyl, methoxy or fluoro; R3 is trifluoromethyl; and R4 and R5 are each independently hydrogen or methyl; or a pharmaceutically acceptable salt thereof.
  • Another preferred embodiment of the present invention is a compound of Formula I wherein R1 is (C1-C6)alkyl-Q; Q is phenyl optionally substituted with one or two substituents independently selected from fluoro, methyl or methoxy; R2 is hydroxy-phenyl or hydroxy-pyridyl, wherein said hydroxy-phenyl or hydroxy-pyridyl is optionally substituted with one to three substituents independently selected from methyl, methoxy or fluoro; R3 is trifluoromethyl; and R4 and R5 are each independently hydrogen or methyl; or a pharmaceutically acceptable salt thereof. Still another preferred embodiment of the present invention is a compound of Formula I wherein R1 is phenethyl or 1-methyl-2-(phenyl)ethyl, wherein said phenethyl or 1-methyl-2-(phenyl)ethyl is optionally substituted on phenyl with one or two fluoro; R3 is trifluoromethyl; and R4 and R5 are each independently hydrogen or methyl; or a pharmaceutically acceptable salt thereof.
  • Another preferred embodiment of the present invention is a compound of Formula I wherein R1 is phenethyl or 1-methyl-2-(phenyl)ethyl, wherein said phenethyl or 1-methyl-2-(phenyl)ethyl is optionally substituted on phenyl with one or two halo; R2 is hydroxy-phenyl or hydroxy-pyridyl, wherein said hydroxy-phenyl or hydroxy-pyridyl is optionally substituted with one or two substituents independently selected from (C1-C3)alkyl or halo; R3 is trifluoromethyl; and R4 and R5 are each independently hydrogen or methyl; or a pharmaceutically acceptable salt thereof.
  • Another preferred embodiment of the present invention is a compound of Formula I wherein R1 is phenethyl or 1-methyl-2-(phenyl)ethyl, wherein said phenethyl or 1-methyl-2-(phenyl)ethyl is optionally substituted on phenyl with one or two substituents independently selected from halo, (C1-C3)alkyl or (C1-C3)alkoxy; R2 is 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl, wherein said 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl is optionally substituted with one or two substituents independently selected from (C1-C3)alkyl or halo; R3 is (C1-C6)alkyl, (C3-C7)cycloalkyl, (C1-C6)alkyl-Q, aryl, heteroaryl, or OR6; wherein said (C1-C6)alkyl, (C3-C7)cycloalkyl, aryl or heteroaryl is optionally substituted with one to three substituents independently selected from fluoro, cyano, trifluoromethyl, trifluoromethoxy, (C1-C3)alkyl; (C1-C3)alkoxy, NR7R8 or hydroxy; and R4 and R5 are each independently hydrogen or methyl; or a pharmaceutically acceptable salt thereof.
  • Another preferred embodiment of the present invention is a compound selected from the group consisting of:
    • 2-(2-Hydroxy-phenyl)-3-phenethyl-5-trifluoromethyl-3H-pyrido[4,3-d]pyri-mid in-4-one;
    • 2-(3-Fluoro-2-hydroxy-phenyl)-3-phenethyl-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
    • 2-(3-Hydroxy-pyridin-2-yl)-3-phenethyl-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
    • (R)-2-(2-Hydroxy-phenyl)-3-(1-methyl-2-phenyl-ethyl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
    • (S)-2-(2-Hydroxy-phenyl)-3-(1-methyl-2-phenyl-ethyl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
    • (R)-2-(3-Hydroxy-pyridin-2-yl)-3-(1-methyl-2-phenyl-ethyl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
    • (R,S)-2-(3-Hydroxy-pyridin-2-yl)-3-(1-methyl-2-(2-fluorophenyl)ethyl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
    • (R)-2-(3-Hydroxy-pyridin-2-yl)-3-(1-methyl-2-(2-fluorophenyl)ethyl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
    • (S)-2-(3-Hydroxy-pyridin-2-yl)-3-(1-methyl-2-(2-fluorophenyl)ethyl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
    • 2-(2-Hydroxyphenyl)-3-(2-fluorophenyl)ethyl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
    • 2-(2-Hydroxyphenyl)-3-(3-fluorophenyl)ethyl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
    • 2-(3-Fluoro-2-hydroxy-phenyl)-3-[2-(2-fluoro-phenyl)-ethyl]-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
    • 2-(2-Hydroxy-phenyl)-7-methyl-3-(phenyl-ethyl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
    • 2-(3-Fluoro-2-hydroxy-phenyl)-3-[2-(3-fluoro-phenyl)-ethyl]-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
    • 3-[2-(2-Fluoro-phenyl)-ethyl]-2-(3-hydroxy-pyridin-2-yl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
    • 3-[2-(3,4-Difluoro-phenyl)-ethyl]-2-(3-hydroxy-pyridin-2-yl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
    • 3-[2-(2,4-Difluoro-phenyl)-ethyl]-2-(3-hydroxy-pyridin-2-yl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
    • 3-[2-(3,4-Difluoro-phenyl)-ethyl]-2-(3-hydroxy-pyridin-2-yl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
    • 2-(2-Hydroxy-phenyl)-5-methylamino-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
    • 2-(2-Hydroxy-phenyl)-5-isopropylamino-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
    • 2-(2-Hydroxy-phenyl)-3-phenethyl-5-pyrrolidin-1-yl-3H-pyrido[4,3-d]pyrimidin-4-one;
    • 2-(2-Hydroxy-phenyl)-5-(4-methyl-piperazin-1-yl)-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
    • 2-(2-Hydroxy-phenyl)-3-phenethyl-5-piperazin-1-yl-3H-pyrido[4,3-d]pyrimidin-4-one;
    • 5-Dimethylamino-2-(2-hydroxy-phenyl)-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
    • 2-(2-Hydroxy-phenyl)-5-morpholin-4-yl-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
    • 5-Azetidin-1-yl-2-(2-hydroxy-phenyl)-3-phenethyl-3H-pyrido[4,3-d-pyrimidin-4-one;
    • 2-(2-Hydroxy-phenyl)-3-phenethyl-5-phenyl-3H-pyrido[4,3-d]pyrimidin-4-one;
    • 5-Benzyl-2-(2-hydroxy-phenyl)-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
    • 2-(2-Hydroxy-phenyl)-5-methyl-3-phenethyl-3H-pyrido[4,3-d]pyrimidi-4-one;
    • 5-(6-Dimethylamino-pyridin-3-yl)-2-(2-hydroxy-phenyl)-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
    • 5-(6-Dimethylamino-5-methyl-pyridin-3-yl)-2-(2-hydroxy-phenyl)-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
    • 5-(6-pyrrolidine-5-pyridin-3-yl)-2-(2-hydroxy-phenyl)-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
    • 2-(2-Hydroxy-phenyl)-5-methoxy-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
    • 2-(2-Hydroxy-phenyl)-5-(1-methyl-cyclopropylmethoxy)-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
    • 2-(2-Hydroxy-phenyl)-3-phenethyl-5-propoxy-3H-pyrido[4,3-d]pyrimidin-4-one;
    • 5-Cyclobutyloxy-2-(2-hydroxy-phenyl)-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
    • 2-(2-Hydroxy-phenyl)-5-isobutoxy-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
    • 2-(2-Hydroxy-phenyl)-5-isopropoxy-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
    • 2-(2-Hydroxy-phenyl)-3-phenethyl-5-(2,2,2-trifluoro-ethoxy)-3H-pyrido[4,3-d]pyrimidin-4-one;
    • 3-(1-(3,4-difluorophenyl)propan-2-yl)-2-(3-hydroxypyridin-2-yl) 5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • (R)-3-(1-(3,4-difluorophenyl)propan-2-yl)-2-(3-hydroxypyridin-2-yl) 5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • (S)-3-(1-(3,4-difluorophenyl)propan-2-yl)-2-(3-hydroxypyridin-2-yl) 5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • 3-(1-(2,4-difluorophenyl)propan-2-yl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • 3-(1-(2,4-difluorophenyl)propan-2-yl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • 3-(2-methoxyphenethyl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • 3-(2,3-difluorophenethyl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • 3-(5-fluoro-2-methoxyphenethyl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • 3-(2-fluoro-6-methoxyphenethyl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • 2-(3-hydroxypyridin-2-yl)-3-(1-(2-methoxyphenyl)propan-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • 2-(3-hydroxypyridin-2-yl)-3-(1-(2-methoxyphenyl)propan-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • 2-(3-hydroxypyridin-2-yl)-3-(1-(2-methoxyphenyl)propan-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • 3-(1-(2-fluorophenyl)butan-2-yl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • 3-(3-fluoro-2-methoxyphenethyl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • 3-(2-cyclopentylethyl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • 3-(2-cyclohexylethyl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • (R)-3-(1-cyclohexylpropan-2-yl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • (S)-3-(1-cyclohexylpropan-2-yl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • (R,S)-2-(3-hydroxypyridin-2-yl)-3-(2-(tetrahydro-2H-pyran-2-yl)ethyl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • (R)-2-(3-fluoro-2-hydroxyphenyl)-3-(1-phenylpropan-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • (R)-2-(1H-imidazol-2-yl)-3-(1-phenylpropan-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • N-(2-(3-(2-fluorophenethyl)-4-oxo-5-(trifluoromethyl)-3,4-dihydropyrido[4,3-d]pyrimidin-2-yl)pyridin-3-yl)acetamide;
    • (R)-3-(1-phenylpropan-2-yl)-2-(thiazol-4-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • 3-phenethyl-2-(thiazol-4-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • 3-(2-methoxyphenethyl)-2-(2-hydroxyphenyl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • 3-(2-fluorophenethyl)-2-(thiazol-4-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • (R)-2-cyclopentyl-3-(1-phenylpropan-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • 2-isopropyl-3-phenethyl-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
    • 2-cyclopentyl-3-phenethyl-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • 3-(2-methoxyphenethyl)-2-(thiazol-4-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • 3-(2-cyclohexylethyl)-2-(thiazol-4-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • 2-(3-(2-cyclohexylethyl)-4-oxo-5-(trifluoromethyl)-3,4-dihydropyrido[4,3-d]pyrimidin-2-yl)benzonitrile;
    • 2-(3-aminopyridin-2-yl)-3-phenethyl-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • N-(2-(4-oxo-3-phenethyl-5-(trifluoromethyl)-3,4-dihydropyrido[4,3-d]pyrimidin-2-yl)pyridin-3-yl)methanesulfonamide;
    • 3-(1-(2-fluorophenyl)propan-2-yl)-2-(thiazol-4-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • 2-(3-hydroxypyridin-2-yl)-3-isopentyl-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • 2-(3-hydroxypyridin-2-yl)-3-((tetrahydro-2H-pyran-4-yl)methyl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • 2-(3-hydroxypyridin-2-yl)-3-(2-(tetrahydro-2H-pyran-4-yl)ethyl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • 3-(2-fluoro-5-methoxyphenethyl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • 3-(2-fluoro-3-methoxyphenethyl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • 2-(3-(2-fluorophenethyl)-4-oxo-5-(trifluoromethyl)-3,4-dihydroquinazolin-2-yl)benzonitrile
    • N-(2-(3-(2-fluorophenethyl)-4-oxo-5-(trifluoromethyl)-3,4-dihydropyrido[4,3-d]pyrimidin-2-yl)pyridin-3-yl)-2,2,2-trifluoroacetamide;
    • (R)-2,2,2-trifluoro-N-(2-(4-oxo-3-(1-phenylpropan-2-yl)-5-(trifluoromethyl)-3,4-dihydropyrido[4,3-d]pyrimidin-2-yl)pyridin-3-yl)acetamide;
    • 3-cyclohexyl-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • 3-(cyclohexylmethyl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • 3-Phenethyl-5-trifluoromethyl-2-(3-trifluoromethyl-pyridin-2-yl)-3H-pyrido[4,3-d]pyrimidin-4-one;
    • 2-(5-aminothiazol-4-yl)-3-phenethyl-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • 2-(2-(difluoromethyl)phenyl)-3-phenethyl-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • 2-(3-(difluoromethyl)pyridin-2-yl)-3-phenethyl-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
    • 3-(2-cyclohexylethyl)-5-(trifluoromethyl)-2-(2-(trifluoromethyl)phenyl)pyrido[4,3-d]pyrimidin-4(3H)-one; and
    • 3-(2-cyclohexylethyl)-5-(trifluoromethyl)-2-(3-(trifluoromethyl)pyridin-2-yl)pyrido[4,3-d]pyrimidin-4(3H)-one; or a pharmaceutically acceptable salt thereof.
  • Another embodiment of the present invention is a pharmaceutical composition comprising a compound according to Formula I as described in any of the preceding embodiments hereinabove and a pharmaceutically acceptable carrier, adjuvant or diluent.
  • Another embodiment of the present invention are novel intermediate compounds 4-Amino-2-(trifluoromethyl)nicotinic acid or 4-Amino-2-chloronicotinic acid; or a pharmaceutically acceptable salt thereof.
  • Another embodiment of the present invention is a method of treating a disease or disorder characterized by abnormal bone or mineral homeostasis which comprises the administration to a patient in need of treatment thereof a therapeutically effective amount of a compound according to Formula I as described in any of the preceding embodiments hereinabove. A preferred embodiment of the present invention is the method according to the preceding embodiment wherein the disease or disorder characterized by abnormal bone or mineral homeostasis is selected from the group consisting of osteoporosis, osteopenia, periodontal disease, Paget's disease, bone fracture, osteoarthritis, rheumatoid arthritis, and humoral hypercalcemia of malignancy. Yet another preferred embodiment is the method according to the preceding embodiment wherein the disease or disorder characterized by abnormal bone or mineral homeostasis is osteoporosis.
  • The following reaction schemes, Reaction Schemes I-IV, depict methods of synthesis for compounds of formula I. In the following general methods for preparation of the compounds of formula I the variables R1, R2, R3, R4, and R5, are as previously defined for a compound of the formula I unless otherwise stated. The Reaction Schemes herein described are intended to provide a general description of the methodology employed in the preparation of many of the Examples given. However, it will be evident from the detailed descriptions given in the Experimental section that the modes of preparation employed extend further than the general procedures described herein. In particular, it is noted that the compounds prepared according to these Schemes may be modified further to provide new Examples within the scope of this invention. The reagents and intermediates used in the following examples are either commercially available or can be prepared according to standard literature procedures by those skilled in the art of organic synthesis.
  • Reaction Scheme I, below, depicts the synthesis of intermediates of formula V which are useful for preparing compounds of formula I. Treatment of an appropriately substituted pyridine of formula IX with an appropriate base, such as lithium diisopropylethylamide, in an appropriate solvent, such as THF, at approximately −65° C. to −78° C. followed by addition of CO2 and acidification upon workup provides the corresponding nicotinic acid of formula VIII. Treatment of the appropriately substituted nicotinic acid of formula VIII with an appropriate base, such as n-butyl lithium and lithium 2,2,6,6-tetramethylpiperidide (LiTMP), followed by trapping with an appropriate halogen source, such as hexachloroethane, and followed by esterification, with diazomethane for example, provides the corresponding chlorinated pyridine ester of formula VII. The chlorinated pyridine ester VII is then treated with an appropriate nitrogen source, such as azide anion (i.e. sodium azide) in an appropriate solvent, such as DMF, typically at 40° C. to 50° C. for 1 to 24 hours, to provide the azido pyridine ester of formula VI [Cf: Kiyama, Chem. Pharm. Bull. 1995, 43, 450.]. Reduction of the azide group in the compound of formula VI to the corresponding amine can be carried out using several procedures known in the art, such as catalytic hydrogenation, treatment with aqueous hydriodic acid at ambient temperature or treatment with triphenylphosphine. Saponification of the ester can then be carried out in a variety of ways such as aqueous lithium hydroxide in an appropriate solvent such as dioxane:MeOH:water (3:2:1) at a temperature of 40° C. to 100° C., preferably at approximately 85° C. for 1 to 24 hours to provide the corresponding nicotinic acid of formula V. It is to be appreciated that the group R3 in the compounds of formulae IX, VIII, VII, VI and V can have the same meaning as defined for group R3 in compounds of formula I or alternatively can be a functionality that can be converted to a group R3 as defined for compounds of formula I. For example, R3 in formulae IX, VIII, VII, VI and V can be a halo group, such as chloro, which can then be further converted, for example, as shown in Reaction Scheme VI, below to a group R3 as defined for a compound of formula I.
  • Figure US20110028452A1-20110203-C00004
  • Reaction Scheme II, below, depicts the preparation of compounds of Formula I. Formula IV compounds are prepared by reacting the corresponding acid of Formula V with a variety of primary amines of general formula R1NH2 under standard amide coupling conditions for a period between 4 to 24 hours. Formula III compounds are prepared by reaction of Formula IV compounds with an appropriate aldehyde, R2′CHO, in the presence of a catalytic amount of an acid, such as p-toluenesulfonic acid, in a suitable reaction inert solvent such as acetonitrile or toluene at a temperature between 55° C. to 100° C., typically 80° C., for a period between 1 to 24 hours. The R2′ moiety in the aldehyde of formula R2′CHO is one in which a protected hydroxy group typically exists, such as a methoxy or benzyloxy, which can subsequently be deprotected to the free hydroxy present in group R2 in the compound of Formula I. Compounds of Formula III are then treated with an oxidizing agent such as potassium permanganate in a polar solvent such as acetone at a temperature between 20° C. to 50° C., typically ambient temperature, to provide compounds of Formula II after a period of 1 to 24 hours. Compounds of Formula II which contain a protected hydroxy group such as a suitable methyl ether moiety or benzyloxy moiety, may be converted to the free hydroxy (such as a phenolic hydroxy group) by standard deprotection conditions.
  • Figure US20110028452A1-20110203-C00005
  • In the reaction schemes described herein it is to be understood that hydroxy groups in intermediates useful for preparing compounds of Formula I may be protected by other conventional protecting groups known to those skilled in the art. For example, intermediates containing a hydroxy group may be protected as the corresponding benzyloxy ether and subsequently deprotected by hydrogenation to provide the free hydroxy derivative. Suitable protecting groups and methods for their removal are illustrated in Protective Groups in Organic Synthesis, 3rd Ed., Theodora W. Greene, and Peter G. M. Wuts (John Wiley & Sons, 1999). For example, in cases where the compound of formula II contains a suitable methyl ether moiety, the compound of Formula II is dissolved in a suitable solvent such as methylene chloride at a temperature between −78° C. to 10° C. followed by the addition of boron trichloride or boron tribromide. The reaction mixture is stirred at 10° C. to 50° C. for 1 to 24 hours to provide the desired deprotected compound of Formula I. Similarly, compounds of formula II containing a suitable benzyloxy moiety can be deprotected to provide compounds of formula I by hydrogenation, such as H-cube hydrogenation (Pd/C cartridge) carried out at ambient temperature using methanol as solvent.
  • Figure US20110028452A1-20110203-C00006
  • Reaction Scheme III depicts Formula IVa compounds prepared by reacting the corresponding acid of Formula Va with a variety of primary amines of general formula R1NH2 under standard amide coupling conditions for a period between 4 to 24 hours. Formula IIIa compounds may be prepared by reaction of Formula IVa compounds with an appropriate aldehyde, R2′CHO, in the presence of a catalytic amount of an acid, such as p-toluenesulfonic acid, in a suitable reaction inert solvent such as acetonitrile or toluene at a temperature between 55° C. to 100° C., typically 80° C., for a period between 1 to 24 hours. The R2′ moiety in the aldehyde of formula R2′CHO is one in which a protected hydroxy group exists, such as a methoxy or benzyloxy, which can subsequently be deprotected to the free hydroxy present in group R2 in the compound of formula I. Compounds of Formula IIIa are then treated with an oxidizing agent such as potassium permanganate in a polar solvent such as acetone at a temperature between 20° C. to 50° C., typically ambient to provide compounds of Formula IIa after a period of 1 to 24 hours.
  • Figure US20110028452A1-20110203-C00007
  • Reaction Scheme IV depicts the conversion of functional groups at the R3 position of various intermediates useful in preparing compounds of Formula I. Formula IIa compounds, wherein R3 is chloro, are converted to Formula IIb compounds, wherein R3 is iodo, by reaction with a reagent such as sodium iodide and trimethylsilyl chloride in the dark in an inert solvent such as acetonitrile or propionitrile at a temperature between 55° C. to 120° C., typically 100° C., for a period between 1 to 24 hours. The compounds of Formula IIc are prepared by reacting the compounds of Formula IIb with methyl-2,2-difluoro-2-(fluoro-sulfonyl)acetate and copper iodide in a mixture of solvent such as HMPA and DMF at a temperature between 55° C. to 120° C., typically 80° C., for a period between 1 to 24 hours. The R2′ group in the compounds of formulae IIa, IIb and IIc is one in which a protected hydroxy group exists, such as a methoxy or benzyloxy, which can subsequently be deprotected to the free hydroxy present in group R2 of the corresponding compound within formula I.
  • Figure US20110028452A1-20110203-C00008
  • Reaction Scheme V depicts the conversion of a methyl ether moiety in certain intermediates of formulae VI or VI′ wherein R2 is 2-methoxyphenyl, to a free phenolic hydroxy moiety in compounds of Formula Ia or Ia′. Compounds of Formula VI (or VI′ in which R3 is CF3) which contain a methyl ether moiety may be converted to the free phenol by dissolving the compounds of Formula VI or VI′ in a suitable solvent such as methylene chloride at a temperature between −78° C. to 10° C. followed by the addition of boron trichloride or boron tribromide. The reaction mixture is stirred at 10° C. to 50° C. for 1 to 24 hours to provide the desired compounds, such as those of Formula Ia or Ia′.
  • Reaction Scheme VI, below, depicts three reactions showing the conversion of chloro derivative IIa or IIa′ to alkoxy (Ib, wherein R3 is OR6), amino (Ic, wherein R3 is NR7R8) and carbon-linked derivatives (Id, wherein R3 results from an appropriate boronic acid derivative such as an aryl, heteroaryl or (C1-C6)alkyl boronic acid), respectively. Compounds of Formula IIa may be converted to compounds of Formula Ib by treatment of the corresponding alcohol, R6OH, in the presence of a suitable base such as sodium hydride in an inert solvent such as DMF or THF at 20° C. to 80° C. for 6 to 24 hours, followed by deprotection as necessary to provide compounds of Formula Ib.
  • Compounds of Formula IIa′, in which the hydroxy moiety in R2 is in deprotected form, may be reacted with an appropriate amine of formula R7R8NH in a solvent such as methanol or ethanol at 20° C. to 80° C. for 1 to 24 hours to provide compounds of Formula Ic. Compounds of Formula IIa may also be treated with an appropriate corresponding boronic acid derivative of formula R3B(OH)2 in the presence of a catalyst such as palladium tetrakis(triphenylphosphine) in a mixture of aqueous sodium carbonate and a polar solvent such as dioxane in a microwave for 1 to 25 minutes at 50° C. to 150° C., followed by deprotection as necessary to provide compounds of Formula Id.
  • Figure US20110028452A1-20110203-C00009
  • The term “patient in need of treatment thereof” means humans and other animals who have or are at risk of having a disease or disorder characterized by abnormal bone or mineral homeostasis. The “patient in need of treatment thereof” may have or be at risk of having a disease or disorder characterized by abnormal bone or mineral homeostasis selected from the group consisting of osteoporosis, osteopenia, periodontal disease, Paget's disease, bone fracture, osteoarthritis, rheumatoid arthritis, and humoral hypercalcemia of malignancy. As certain of the conditions being treated have a higher incidence in females a preferred patient is a female, and particularly a postmenopausal female human.
  • The term “treating”, “treat” or “treatment” as used herein includes preventative (e.g., prophylactic), palliative, adjuvant and curative treatment. For example, the treatment of osteoporosis, as used herein means that a patient having osteoporosis or at risk of having osteoporosis can be treated according to the methods described herein. For patients undergoing preventative treatment, a resulting reduction in the incidence of the disease state being preventively treated is the measurable outcome of the preventative treatment.
  • The present invention provides methods of treating osteopenia and osteoporosis by administering to a patient in need thereof a therapeutically effective amount of a compound of formula I. Osteopenia is a thinning of the bones, but less than is seen with osteoporosis and is the stage before true osteoporosis. The World Health Organization has developed diagnostic categories based on bone mass density (BMD) to indicate if a person has normal bones, has osteopenia or has osteoporosis. Normal bone density is within one standard deviation (+1 or −1) of the young adult mean bone density. Osteopenia (low bone mass) is defined as bone density of 1 to 2.5 standard deviations below the young adult mean (−1 to −2.5), and osteoporosis is defined as a bone density that is 2.5 standard deviations or more below the young adult mean (>−2.5).
  • The present invention provides methods of treating bone fractures by administering to a patient in need thereof a therapeutically effective amount of a compound of formula I. Bone fractures can be a fracture to any bone in the body, and hip fracture being of particular concern. Hip fracture has a significant impact on medical resources and patient morbidity and mortality. Few patients admitted with a hip fracture are considered for prophylactic measures aimed at the reduction of further fracture risk. Currently, 10-13% of patients will later sustain a second hip fracture. Of patients who suffered a second hip fracture, fewer patients maintained their ability to walk independently after the second fracture than did so after the first (53 and 91% respectively, P<0.0005). Pearse E. O. et al., Injury, 2003, 34(7), 518-521. Following second hip fracture, patients' level of mobility determined their future social independence. Older patients and those with a history of multiple falls had a shorter time interval between fractures. Second hip fracture has a significant further impact on patients' mobility and social independence. It is therefore desirable to have new methods for the treatment of bone fractures including hip fracture.
  • The compounds of Formula I can be administered together with additional agents which are useful for treating a disease or disorder characterized by abnormal bone or mineral homeostasis. Particularly contemplated additional agents include calcium receptor antagonists other than those of Formula I, selective estrogen receptor modulators (SERMs), bisphosphonates, parathyroid hormone (PTH) and fragments and analogues thereof, estrogens, calcitonins, synthetic steroids, synthetic isoflavones, vitamin D analogues, vitamin K analogues, strontium salts, cathepsin K inhibitors, αvβ3 integrin (vitronectin) antagonists, prostaglandin (PGE2) receptor agonists and receptor activator of nuclear factor κB ligand (RANKL) inhibitors.
  • Additional calcium receptor antagonists that can be used together with compounds of Formula I in the methods and compositions of this invention include those described in PCT International Publication Nos. WO 93/04373; WO 94/18959; WO 95/11211; WO 97/37967; WO 98/44925; WO 98/45255; WO 99/51241; WO 99/51569; WO 00/45816; WO 02/14259; WO 02/38106; WO 2004/041755; and WO 2005/030746; Nemeth, E. F.; Journal of Molecular Endocrinology (2002) 29, 15-21; Kessler, A. et al.; ChemBioChem (2004) 5, 1131; Steddon, S. J. et al.; Lancet (2005) 365, 2237-2239; and Shcherbakova, I.; et al.; Bioorganic & Medicinal Chemistry Letters (2005) 15, 1557-1560. Specific calcilytic compounds that can be used together with compounds of Formula I in the methods and compositions of this invention include NPS-2143 and 423562.
  • SERMs that can be used together with compounds of Formula I in the methods and compositions of this invention include, but are not limited to, lasofoxifene (Oporia®), raloxifene (Evista®), arzoxifene, bazedoxifene, ospemifene, Chiesi's CHF-4227 and Prostrakan's PSK-3471. Bisphosphonates that can be used together with compounds of Formula I in the methods and compositions of this invention include, but are not limited to, tiludronate (Skelid®), clondronate (Bonefos®), etidronate (Didronel®), alendronate (Fosamax®), risedronate (Actonel®), ibandronate (Boniva®), zoledronate (Zometa®), minodronate (Onobis®), neridronate and pamidronate.
  • In humans, PTH is an 84 amino acid polypeptide produced by the parathyroid gland that controls serum calcium levels through its action on various cells. Several N-terminal amino acids fragments of PTH, including the 1-31, 1-34 and 1-38 fragments (PTH-related proteins; “PTHrP”) are considered biologically equivalent to the full length hormone. Parathyroid hormone (PTH) and fragments and analogues thereof that can be used together with compounds of Formula I in the methods and compositions of this invention include, but are not limited to, the full length PTH (such as PTH 1-84, Preos®/Preotact®, Unigene's 768974, Bone Medical's BN-003), the 1-31 (such as Zelos Therapeutics' Ostabolin-C), 1-34 (such as teriparatide, Forteo®, or Ipsen's BIM-44058) or 1-38 fragments.
  • Estrogens that can be used together with compounds of Formula I in the methods and compositions of this invention include, but are not limited to, estradiol, conjugated equine estrogens (Wyeth's Premarin®) or other estrogens.
  • Calcitonin is a 32 amino-acid peptide hormone produced by the thyroid gland which inhibits osteoclast activity by binding to calcitonin receptors on the surface of those cells. Calcitonins that can be used together with compounds of Formula I in the methods and compositions of this invention include, but are not limited to, human calcitonin or salmon or eel calcitonins. The calcitonins may be used as injectable or intranasal formulations such as Miacalcin®, Miacalcic®, Calcitonia®, Fortical® or Elcitonin® or as oral formulations such as Novartis' SMC-021, Bone Medical's BN-002 (Capsitonin®) or Nobex's NCT-025 (Oratonin®).
  • Synthetic steroids that can be used together with compounds of Formula I in the methods and compositions of this invention include, but are not limited to, mixed estrogen and progesterone agonists such as tibolone which is marketed as Livial®. Synthetic isoflavones are chemically synthesized derivatives of plant isoflavones, such as phytoestrogens extracted from soy products. A synthetic isoflavone that can be used together with compounds of Formula I in the methods and compositions of this invention includes, but is not limited to, ipraflavone which is marketed by Takeda as Iprosten® and Osten®.
  • Vitamin D analogues are compounds that act by binding to the nuclear vitamin D receptor in osteoblasts. Vitamin D analogues that can be used together with compounds of Formula I in the methods and compositions of this invention include, but are not limited to, Chugai's ED-71 and Deltanoid's 2MD.
  • A strontium salt that can be used together with compounds of Formula I in the methods and compositions of this invention includes, but is not limited to, strontium ranelate (Servier's Protelos®). Cathepsin K inhibitors that can be used together with compounds of Formula I in the methods and compositions of this invention include, but are not limited to, Novartis's AAE-581, balicatib, GlaxoSmithKline's SB-462795 and Merck's c-3578. An αvβ3 integrin (vitronectin) antagonist that can be used together with compounds of Formula I in the methods and compositions of this invention includes, but is not limited to, Merck's MRL-123.
  • Prostaglandin E2 (PGE2) receptor agonists that can be used together with compounds of Formula I in the methods and compositions of this invention include, but are not limited to, PGE2 subtype 2 (EP2) receptor agonists, such as (3-{[4-Tert-butyl-benzyl)-(pyridine-3-sulfonyl)-amino]-methyl}-phenoxy)-acetic acid, or a pharmaceutically acceptable salt thereof or PGE2 subtype 4 (EP4) receptor agonists, such as ONO-4819. A receptor activator of nuclear factor κB ligand (RANKL) inhibitor that can be used together with compounds of Formula I in the methods and compositions of this invention includes, but is not limited to, Amgen's RANKL antibody AMG-162.
  • Specific combinations of particular interest include compounds of Formula I and lasofoxifene or compounds of Formula I and (3-{[4-Tert-butyl-benzyl)-(pyridine-3-sulfonyl)-amino]-methyl}-phenoxy)-acetic acid, or a pharmaceutically acceptable salt thereof.
  • Compounds of the invention intended for pharmaceutical use may be administered as crystalline or amorphous products. The compounds may be obtained, for example, as solid plugs, powders, or films by methods such as precipitation, crystallization, freeze drying, spray drying, or evaporative drying. Microwave or radio frequency drying may be used for this purpose.
  • For the above-mentioned therapeutic uses, the dosage administered will, of course, vary with the compound employed, the mode of administration, the treatment desired and the disorder indicated. The total daily dosage of the compound of formula I/salt/solvate (active ingredient) will, generally, be in the range from 1 mg to 1 gram, preferably 1 mg to 250 mg, more preferably 10 mg to 100 mg. The total daily dose may be administered in single or divided doses. The present invention also encompasses sustained release compositions.
  • The pharmaceutical composition may, for example, be in a form suitable for oral administration as a tablet, capsule, pill, powder, sustained release formulations, solution, suspension, for parenteral injection as a sterile solution, suspension or emulsion, for topical administration as an ointment or cream or for rectal administration as a suppository. The pharmaceutical composition may be in unit dosage forms suitable for single administration of precise dosages. The pharmaceutical composition will include a conventional pharmaceutical carrier or excipient and a compound according to the invention as an active ingredient. In addition, it may include other medicinal or pharmaceutical agents, carriers, adjuvants, etc. The dissolution rate of poorly water-soluble compounds may be enhanced by the use of a spray-dried dispersion, such as those described by Takeuchi, H., et al. in “Enhancement of the dissolution rate of a poorly water-soluble drug (tolbutamide) by a spray-drying solvent depostion method and disintegrants” J. Pharm. Pharmacol., 39, 769-773 (1987).
  • Exemplary parenteral administration forms include solutions or suspensions of active compounds in sterile aqueous solutions, for example, aqueous propylene glycol or dextrose solutions. Such dosage forms can be suitably buffered, if desired.
  • Suitable pharmaceutical carriers include inert diluents or fillers, water and various organic solvents. The pharmaceutical compositions may, if desired, contain additional ingredients such as flavorings, binders, excipients and the like. Thus for oral administration, tablets containing various excipients, such as citric acid may be employed together with various disintegrants such as starch, alginic acid and certain complex silicates and with binding agents such as sucrose, gelatin and acacia. Additionally, lubricating agents such as magnesium stearate, sodium lauryl sulfate and talc are often useful for tableting purposes. Solid compositions of a similar type may also be employed in soft and hard filled gelatin capsules. Preferred materials, therefor, include lactose or milk sugar and high molecular weight polyethylene glycols. When aqueous suspensions or elixirs are desired for oral administration the active compound therein may be combined with various sweetening or flavoring agents, coloring matters or dyes and, if desired, emulsifying agents or suspending agents, together with diluents such as water, ethanol, propylene glycol, glycerin, or combinations thereof. Methods of preparing various pharmaceutical compositions with a specific amount of active compound are known, or will be apparent, to those skilled in this art. For examples, see Remington's Pharmaceutical Sciences, Mack Publishing Company, Easter, Pa., 15th Edition (1975).
  • Dosage regimens may be adjusted to provide the optimum desired response. For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form, as used herein, refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the chemotherapeutic agent and the particular therapeutic or prophylactic effect to be achieved, and (b) the limitations inherent in the art of compounding such an active compound for the treatment of sensitivity in individuals. Thus, the skilled artisan would appreciate, based upon the disclosure provided herein, that the dose and dosing regimen is adjusted in accordance with methods well-known in the therapeutic arts. That is, the maximum tolerable dose can be readily established, and the effective amount providing a detectable therapeutic benefit to a patient may also be determined, as can the temporal requirements for administering each agent to provide a detectable therapeutic benefit to the patient. Accordingly, while certain dose and administration regimens are exemplified herein, these examples in no way limit the dose and administration regimen that may be provided to a patient in practicing the present invention.
  • It is to be noted that dosage values may vary with the type and severity of the condition to be alleviated, and may include single or multiple doses. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and that dosage ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed composition. For example, doses may be adjusted based on pharmacokinetic or pharmacodynamic parameters, which may include clinical effects such as toxic effects and/or laboratory values. Thus, the present invention encompasses intra-patient dose-escalation as determined by the skilled artisan. Determining appropriate dosages and regiments for administration of the chemotherapeutic agent are well-known in the relevant art and would be understood to be encompassed by the skilled artisan once provided the teachings disclosed herein.
  • A pharmaceutical composition of the invention may be prepared, packaged, or sold in bulk, as a single unit dose, or as a plurality of single unit doses. As used herein, a “unit dose” is discrete amount of the pharmaceutical composition comprising a predetermined amount of the active ingredient. The amount of the active ingredient is generally equal to the dosage of the active ingredient which would be administered to a subject or a convenient fraction of such a dosage such as, for example, one-half or one-third of such a dosage.
  • The relative amounts of the active ingredient, the pharmaceutically acceptable carrier, and any additional ingredients in a pharmaceutical composition of the invention will vary, depending upon the identity, size, and condition of the subject treated and further depending upon the route by which the composition is to be administered. By way of example, the composition may comprise between 0.1% and 100% (w/w) active ingredient.
  • In addition to the active ingredient, a pharmaceutical composition of the invention may further comprise one or more additional pharmaceutically active agents. Particularly contemplated additional agents include selective estrogen receptor modulators (SERMs), bisphosphonates, parathyroid hormone (PTH) and fragments and analogues thereof, estrogens, calcitonins, synthetic steroids, synthetic isoflavones, vitamin D analogues, vitamin K analogues, strontium salts, cathepsin K inhibitors, αvβ3 integrin (vitronectin) antagonists, prostaglandin (PGE2) receptor agonists and receptor activator of nuclear factor κB ligand (RANKL) inhibitors, such as those described hereinabove.
  • Controlled- or sustained-release formulations of a pharmaceutical composition of the invention may be made using conventional technology.
  • As used herein, “parenteral administration” of a pharmaceutical composition includes any route of administration characterized by physical breaching of a tissue of a subject and administration of the pharmaceutical composition through the breach in the tissue. Parenteral administration thus includes, but is not limited to, administration of a pharmaceutical composition by injection of the composition, by application of the composition through a surgical incision, by application of the composition through a tissue-penetrating non-surgical wound, and the like. In particular, parenteral administration is contemplated to include, but is not limited to, subcutaneous, intraperitoneal, intramuscular, intrasternal injection, and kidney dialytic infusion techniques.
  • Formulations of a pharmaceutical composition suitable for parenteral administration comprise the active ingredient combined with a pharmaceutically acceptable carrier, such as sterile water or sterile isotonic saline. Such formulations may be prepared, packaged, or sold in a form suitable for bolus administration or for continuous administration. Injectable formulations may be prepared, packaged, or sold in unit dosage form, such as in ampules or in multi-dose containers containing a preservative. Formulations for parenteral administration include, but are not limited to, suspensions, solutions, emulsions in oily or aqueous vehicles, pastes, and implantable sustained-release or biodegradable formulations as discussed below. Such formulations may further comprise one or more additional ingredients including, but not limited to, suspending, stabilizing, or dispersing agents. In one embodiment of a formulation for parenteral administration, the active ingredient is provided in dry (i.e. powder or granular) form for reconstitution with a suitable vehicle (e.g. sterile pyrogen-free water) prior to parenteral administration of the reconstituted composition.
  • A composition of the present invention can be administered by a variety of methods known in the art. The route and/or mode of administration vary depending upon the desired results. The active compounds can be prepared with carriers that protect the compound against rapid release, such as a controlled release formulation, including implants, transdermal patches, and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Many methods for the preparation of such formulations are described by e.g., Sustained and Controlled Release Drug Delivery Systems, J. R. Robinson, ed., Marcel Dekker, Inc., New York, (1978). Pharmaceutical compositions are preferably manufactured under GMP conditions.
  • The pharmaceutical compositions may be prepared, packaged, or sold in the form of a sterile injectable aqueous or oily suspension or solution. This suspension or solution may be formulated according to the known art, and may comprise, in addition to the active ingredient, additional ingredients such as the dispersing agents, wetting agents, or suspending agents described herein. Such sterile injectable formulations may be prepared using a non-toxic parenterally-acceptable diluent or solvent, such as water or 1,3-butane diol, for example. Other acceptable diluents and solvents include, but are not limited to, Ringer's solution, isotonic sodium chloride solution, and fixed oils such as synthetic mono- or di-glycerides. Other parentally-administrable formulations which are useful include those which comprise the active ingredient in microcrystalline form, in a liposomal preparation, or as a component of a biodegradable polymer systems. Compositions for sustained release or implantation may comprise pharmaceutically acceptable polymeric or hydrophobic materials such as an emulsion, an ion exchange resin, a sparingly soluble polymer, or a sparingly soluble salt.
  • While the precise dosage administered of each active ingredient will vary depending upon any number of factors, including but not limited to, the type of animal and type of disease state being treated, the age of the animal and the route(s) of administration. The amounts of various CaR antagonist compounds of formula I to be administered can be determined by standard procedures taking into account factors such as the compound IC50, EC50, the biological half-life of the compound, the age, size and weight of the patient, and the type of condition or symptom associated with the patient. The importance of these and other factors to be considered are known to those of ordinary skill in the art.
  • PTH secretion can be measured using techniques known in the art (see, e.g., U.S. Pat. No. 6,031,003, hereby incorporated by reference). For example, PTH secretion can be measured by first suspending cells in parathyroid cell buffer containing 0.5 mM CaCl2 and 0.1% bovine serum albumin. Incubations can be performed in plastic tubes (Falcon 2058) containing 0.3 mL of the cell suspension with or without small volumes of CaCl2 and/or organic polycations. After incubation at 37° C., typically 30 minutes, the tubes can then be placed on ice and the cells pelleted at 2° C. Samples of the supernatant should then be brought to pH 4.5 with acetic acid and, if needed, stored at −70° C. The amount of PTH in bovine cell supernatants can be determined by a homologous radioimmunoassay using GW-1 antibody or its equivalent at a final dilution of 1/45,000. 1251-PTH (65-84; INCSTAR, Stillwater, Minn.) can be used as tracer and fractions separated by dextran-activated charcoal. Counting of samples and data reduction can be performed on a Packard Cobra 5005 gamma counter. For testing PTH levels in human cell supernatants, a commercially available radioimmunoassay kit (INS-PTH; Nichols Institute, Los Angeles, Calif.) which recognizes intact and N-terminal human PTH is preferable because GW-1 antibody recognizes human PTH poorly.
  • In addition, specific assays useful for evaluating the compounds of Formula I include the FLIPR Assay for Evaluating the Potency and Selectivity of Test Compounds; Assay for Evaluating the Effects of Test Compounds on Endogenous PTH Secretion; Evaluation of Effects of Test Compounds on PTH Secretion In Vivo; Effect of Calcium Receptor Antagonist Compound of Formula I on Body Weight, Body Composition and Bone Density in the Aged Intact and Ovariectomized Female Rat; and Fracture Healing Assays as described below.
  • FLIPR Assay for Evaluating the Potency and Selectivity of Test Compounds
  • Human kidney cell (HEK 293) expressing the calcium receptor (CasR) are used to detect antagonists of the receptor using Fluorometric imaging plate reader (FLIPR, Molecular Devices, Sunnyvale Calif.). Receptor activation by extracellular calcium results in the release of calcium from intracellular stores into the cytosol. A fluorescent indicator (Fluo-4) is internalized by the cells from growth media and interacts with calcium released into the cytosol to provide a means of quantifying intracellular Ca2+ levels and receptor agonism/antagonism. Fluorescence intensity is detected by the FLIPR CCD camera and traced as a function of time. Potential antagonists are identified by their ability to decrease this fluorescent response.
  • To determine the IC50 values cells are loaded with Fluo4 (2.05 mM Fluo-4, 0.04% pluronic acid, 2.6 mM probenecid in 90% DMEM high glucose, 10% dialyzed Fetal Bovine Serum, 1× Pen Strep, 1× L-Glutamine, 3 ug/ml Puromycin, 27.5 nM Methotrexate) for 1 hour at 37° C. Prior to the addition of test compound cells are washed with a 10 mM HEPES buffer solution. The test compound, for example the compound of Example 1, is added at various doses (from 1 □M to 3 nM) and pre-incubated with cells for 30 minutes followed by stimulation of the CasR by the addition of 1.7 mM Ca2+. IC50 values are based on the ability of the cells to inhibit the Ca2+ induced increase in intracellular Ca2+. Fluorescence signal is read 42 seconds after the stimulation of the CasR by the addition of 1.7 mM Ca2+.
  • Assay for Evaluating the Effects of Test Compounds on Endogenous PTH Secretion
  • Adult male or female Sprague-Dawley rats (Charles River Laboratories, Wilmington, Mass.) with jugular vein catheter are used in this assay. The test compounds at various doses are given to the animals by various routes of administration including subcutaneous injection, or intraveneous injection. Serum or plasma PTH concentrations are examined before and after dosing at various times using a commercially available rat intact PTH ELISA kit (Immutopics, Inc. San Clemente, Calif. Cat. #60-2500).
  • Evaluation of Effects of Test Compounds on PTH Secretion In Vivo
  • Overnight fasted male Sprague-Dawley rats (250 g) with jugular vein catheter are used in this study. Whole blood sample is collected from each animal prior to compound treatment for measuring baseline PTH concentrations. The test animals are then given a single dose of the tested compound at 1 mg/kg in glycerol formal: 2% DMSO by intravenous administration via jugular vein. Whole blood samples are collected at 2, 5, 15, 30 and 45 minutes, and 1, 2, 3, 4, 6 and 8 hours after dosing. Plasma samples are obtained by centrifugation and PTH concentrations are determined using a commercially available rat intact PTH ELISA kit (Immutopics, Inc. San Clemente, Calif. Cat. #60-2500). A significant burst of PTH was seen following the treatment with the tested compound. The elevated PTH secretion induced by the tested compound was peak at 2 minutes and returned to baseline level at 30 minutes after dosing (FIG. 1).
  • Effect of Calcium Receptor Antagonist Compound of Formula I on Bad Weight, Body Composition and Bone Density in the Aged Intact and Ovariectomized Female Rat
  • The purpose of this study is to test the effects of test compositions comprising compounds of Formula I in aged intact or ovariectomized (OVX) female rat model. In the following protocol the compound of Formula I can be administered as a pharmaceutically acceptable salt or prodrug thereof.
  • Study Protocol
  • Sprague-Dawley female rats are sham-operated or OVX at 18 months of age, while a group of rats is necropsied at day 0 to serve as baseline controls. One day post-surgery, the rats are treated with either vehicle or test compound of Formula I, or a combination of test compound of Formula I and other active agent test compound for 59 days. The vehicle or test compound of Formula I is administered either orally, by oral gavage, or by subcutaneous injection (s.c.), with the test compound being administered at a therapeutically effective dose.
  • All rats are given s.c. injection of 10 mg/kg of calcein (Sigma, St. Louis, Mo.) for fluorescent bone label 2 and 12 days before necropsy. On the day of necropsy, all rats under ketamine/xylazine anesthesia are weighed and undergoe dual-energy X-ray absorptiometry (DXA, QDR-4500/W, Hologic Inc., Waltham, Mass.) equipped with Rat Whole Body Scan software for lean and fat body mass determination. The rats are necropsied, then autopsied and blood is obtained by cardiac puncture. The distal femoral metaphysis and femoral shafts from each rat are analyzed by peripheral quantitative computerized tomography (pQCT), and volumetric total, trabecular and cortical bone mineral content and density are determined.
  • Peripheral Quantitative Computerized Tomography (pQCT) Analysis: Excised femurs are scanned by a pQCT X-ray machine (Stratec XCT Research M, Norland Medical Systems, Fort Atkinson, Wis.) with software version 5.40. A 1 millimeter (mm) thick cross section of the femur metaphysis is taken at 5.0 mm (proximal femoral metaphysis, a primary cancellous bone site) and 13 mm (femoral shafts, a cortical bone site) proximal from the distal end with a voxel size of 0.10 mm. Cortical bone is defined and analyzed using contour mode 2 and cortical mode 4. An outer threshold setting of 340 mg/cm3 is used to distinguish the cortical shell from soft tissue and an inner threshold of 529 mg/cm3 to distinguish cortical bone along the endocortical surface. Trabecular bone is determined using peel mode 4 with a threshold of 655 mg/cm3 to distinguish (sub)cortical from cancellous bone. An additional concentric peel of 1% of the defined cancellous bone is used to ensure that (sub)cortical bone was eliminated from the analysis. Volumetric content, density, and area are determined for both trabecular and cortical bone (Jamsa T. et al., Bone 23:155-161, 1998; Ke, H. Z. et al., Journal of Bone and Mineral Research, 16:765-773, 2001).
  • The experimental groups for the protocol are as follows:
  • Group I: Baseline controls
  • Group II: Sham+Vehicle Group III: OVX+Vehicle Group IV: OVX+Test Compound of Formula I (in Vehicle) Group V: OVX+Test Compound of Formula I and Additional Active Agent
      • Note: Group V only employed when it is desired to test a combination of a compound of Formula I and an additional active agent.
    Fracture Healing Assays
  • Assay for Effects on Fracture Healing after Systemic Administration
  • Fracture Technique: Sprague-Dawley rats at 3 months of age are anesthetized with Ketamine. A 1 cm incision is made on the anteromedial aspect of the proximal part of the right tibia or femur. The following describes the tibial surgical technique. The incision is carried through to the bone, and a 1 mm hole is drilled 4 mm proximal to the distal aspect of the tibial tuberosity 2 mm medial to the anterior ridge. Intramedullary nailing is performed with a 0.8 mm stainless steel tube (maximum load 36.3 N, maximum stiffness 61.8 N/mm, tested under the same conditions as the bones). No reaming of the medullary canal is performed. A standardized closed fracture is produced 2 mm above the tibiofibular junction by three-point bending using specially designed adjustable forceps with blunt jaws. To minimize soft tissue damage, care is taken not to displace the fracture. The skin is closed with monofilament nylon sutures. The operation is performed under sterile conditions. Radiographs of all fractures are taken immediately after nailing, and rats with fractures outside the specified diaphyseal area or with displaced nails are excluded. The remaining animals are divided randomly into the following groups with 10-12 animals per each subgroup per time point for testing the fracture healing. The first group receives daily gavage of vehicle (water: 100% Ethanol=95:5) at 1 ml/rat, while the others receive daily gavage from 0.01 to 100 mg/kg/day of the compound of Formula I to be tested (1 ml/rat) for 10, 20, 40 and 80 days.
  • At 10, 20, 40 and 80 days, 10-12 rats from each group are anesthetized with Ketamine and sacrificed by exsanguination. Both tibiofibular bones are removed by dissection and all soft tissue is stripped. Bones from 5-6 rats for each group are stored in 70% ethanol for histological analysis, and bones from another 5-6 rats for each group are stored in a buffered Ringer's solution (+4° C., pH 7.4) for radiographs and biomechanical testing which is performed.
  • Histological Analysis: The methods for histologic analysis of fractured bone have been previously published by Mosekilde and Bak (The Effects of Growth Hormone on Fracture Healing in Rats: A Histological Description. Bone, 14:19-27, 1993). Briefly, the fracture site is sawed 8 mm to each side of the fracture line, embedded undecalcified in methymethacrylate, and cut frontals sections on a Reichert-Jung Polycut microtome in 8 μm thick. Masson-Trichrome stained mid-frontal sections (including both tibia and fibula) are used for visualization of the cellular and tissue response to fracture healing with and without treatment. Sirius red stained sections are used to demonstrate the characteristics of the callus structure and to differentiate between woven bone and lamellar bone at the fracture site. The following measurements are performed: (1) fracture gap—measured as the shortest distance between the cortical bone ends in the fracture, (2) callus length and callus diameter, (3) total bone volume area of callus, (4) bony tissue per tissue area inside the callus area, (5) fibrous tissue in the callus, and (6) cartilage area in the callus.
  • Biomechanical Analysis: The methods for biomechanical analysis have been previously published by Bak and Andreassen (The Effects of Aging on Fracture Healing in Rats. Calcif Tissue Int 45:292-297, 1989). Briefly, radiographs of all fractures are taken prior to the biomechanical test. The mechanical properties of the healing fractures are analyzed by a destructive three- or four-point bending procedure. Maximum load, stiffness, energy at maximum load, deflection at maximum load, and maximum stress are determined.
  • A calcium receptor antagonist may be usefully combined with another pharmacologically active compound, or with two or more other pharmacologically active compounds, particularly in the treatment of osteoporosis. For example, a calcium receptor antagonist, particularly a compound of the formula I, or a pharmaceutically acceptable salt or solvate thereof, as defined above, may be administered simultaneously, sequentially or separately in combination with one or more agents selected from: selective estrogen receptor modulators (SERMs), bisphosphonates, parathyroid hormone (PTH) and fragments and analogues thereof, estrogens, calcitonins, synthetic steroids, synthetic isoflavones, vitamin D analogues, vitamin K analogues, strontium salts, cathepsin K inhibitors, αvβ3 integrin (vitronectin) antagonists, prostaglandin (PGE2) receptor agonists and receptor activator of nuclear factor κB ligand (RANKL) inhibitors as described hereinabove.
  • The following non-limiting Preparations and Examples illustrate the preparation of compounds of the present invention.
  • 1H Nuclear magnetic resonance (NMR) spectra were in all cases consistent with the proposed structures. Characteristic chemical shifts (δ) are given in parts-per-million downfield from tetramethylsilane using conventional abbreviations for designation of major peaks: e.g. s, singlet; d, doublet; t, triplet; q, quartet; m, multiplet; br, broad. The mass spectra (m/z) were recorded using either electrospray ionisation (ESI) or atmospheric pressure chemical ionisation (APCI). The following abbreviations have been used for common solvents and various reagents: CDCl3, deuterochloroform; DIEA, diisopropylethylamine; DMF, dimethylformamide; CD3OD, deuteromethanol; D6-DMSO, deuterodimethylsulfoxide; HBTU, O-(Benzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate; HCl, hydrochloric acid; HI, hydroiodic acid; HMPA, hexamethylphosphoramide; KMnO4, potassium permanganate; MgSO4, magnesium sulfate; NaHCO3, sodium bicarbonate; NaHSO3, sodium bisulfite; NaOH, sodium hydroxide; Na2SO4, sodium sulfate; NH4Cl, ammonium chloride; TEA, triethylamine; THF, tetrahydrofuran. ‘Ammonia’ refers to a concentrated solution of ammonia in water possessing a specific gravity of 0.88. Where thin layer chromatography (TLC) has been used it refers to silica gel TLC using silica gel 60 F254 plates, Rf is the distance traveled by a compound divided by the distance traveled by the solvent front on a TLC plate. HPLC refers to high performance liquid chromatography.
  • The following specific examples are included for illustrative purposes and are not to be construed as a limitation to this disclosure.
  • Preparation of Intermediates Preparation of 2,4-Dichloronicotinic acid
  • Figure US20110028452A1-20110203-C00010
  • To a stirring solution of diisopropyl ethyl amine (11.1 ml, 81.08 mmol) in THF (50 ml) was added dropwise a solution of BuLi (1.46 M, 43.3 ml, 73.65 mmol) in hexane below −65° C. and the mixture was stirred for 40 minutes. To this solution was added dropwise 2,4-dichloropyridine (10 g, 67.57 mmol) in THF (15 mL) at −78° C. and stirred for 30 minutes. Carbon dioxide generated from freshly crushed dry ice was passed through CaCl2 guard tube and then charged into the reaction mixture for 10 minutes and the reaction mixture was slowly allowed to come to room temperature. The solvent was evaporated under reduced pressure and dissolved in a minimum volume of water. The aqueous layer was washed with water and acidified to pH 4 with conc. HCl. It was then extracted with ethyl acetate, the organic layer was washed with brine and dried over sodium sulfate. The organic solvent was removed under reduced pressure to provide 2,4-dichloronicotinic acid (10.6 g, 82%) as off white solid.
  • 1H NMR (400 MHz, DMSO-d6): δ 14.88-14.54 (br s, 1H), 8.46 (d, J=5.3 Hz, 1H), 7.74 (d, J=5.5 Hz, 1H). FIA MS [M+H]: 191.8
  • Preparation of Methyl 2,4-dichloronicotinate
  • Figure US20110028452A1-20110203-C00011
  • Nitrosomethyl urea (8 g, 78.16 mmol), taken in diethyl ether (30 ml) was cooled to 0° C. and 25% aqueous KOH solution was added slowly under cooling. The ether layer was collected, dried over KOH and added dropwise to a stirring solution of 2,4-Dichloronicotinic acid (3 g, 15.62 mmol) in methanol (5 mL) at 0° C. The reaction mixture was allowed to come to room temperature within 1 hour. The organic solvent was removed under reduced pressure and the crude residue was purified by column chromatography (5-10% EtOAc in hexane) to obtain the pure methyl 2,4-dichloronicotinate (3.00 g, 99%) as colorless oil. 1H NMR (400 MHz, CDCl3): δ 8.34 (d, J=5.3 Hz, 1H), 7.33 (d, J=5.4 Hz, 1H), 3.99 (s, 3H).
  • Preparation of Methyl 4-azido-2-chloronicotinate
  • Figure US20110028452A1-20110203-C00012
  • Methyl 2,4-dichloronicotinate (20 g, 96.67 mmol) afforded methyl 4-azido-2-chloronicotinate (15 g, 73%) as cream colored solid following the literature procedure, J. Prakt. Chem, 2000; 342, 33-39 (reaction of Methyl 2,4-dichloronicotinate with NaN3 in DMF at 50° C. followed by aqueous workup). 1H NMR (400 MHz, DMSO-d6): δ 8.37 (d, J=5.52 Hz, 1H), 7.06 (d, J=5.6 Hz, 1H), 3.96 (s, 3H). FIA MS [M+H]: 213.1 (small peak). IR (KBr): 2126, 1741 cm−1
  • Preparation of Methyl 4-amino-2-chloronicotinate
  • Figure US20110028452A1-20110203-C00013
  • Methyl 4-azido-2-chloronicotinate (15 g, 70.75 mmol) produced methyl 4-amino-2-chloronicotinate (11.2 g, 85%) as light yellow solid following the literature procedure, Tetrahedron letters, 2002; 43, 6629-6631 (reduction of the azido group in methyl 4-azido-2-chloronicotinate to the corresponding amine by stirring methyl 4-azido-2-chloronicotinate in aqueous HI at room temperature). 1H NMR (400 MHz, DMSO-d6): δ 7.81 (d, J=5.79 Hz, 1H), 6.76 (br s, 2H), 6.62 (d, J=5.81 Hz, 1H), 3.82 (s, 3H). FIA MS [M+H]: 186.9
  • Preparation of 4-amino-2-chloronicotinic acid
  • Figure US20110028452A1-20110203-C00014
  • Methyl 4-amino-2-chloronicotinate (21.0 g, 112.9 mmol) and LiOH (10.3 g, 247.5 mmol) was taken in a mixture of dioxane: MeOH:water (3:2:1, ml) and the reaction mixture was heated to 85° C. for 2 hours. The solvent was evaporated under reduced pressure, the residue was dissolved in minimum volume of water and acidified (up to pH 4) with saturated citric acid. The aqueous solution was concentrated until precipitation just started. The mixture was allowed to stand overnight at 10° C. for complete precipitation. The resultant white solid was collected by filtration and recrystallized from isopropyl alcohol-hexane to afford 4-amino-2-chloronicotinic acid (13 g, 66% yield). 1H NMR (400 MHz, DMSO-d6): δ 7.59 (d, 1H), 6.47 (d, 1H), 6.37 (brs, 2H).
  • Preparation of 4-Chloro-2-(trifluoromethyl)nicotinic acid
  • Figure US20110028452A1-20110203-C00015
  • To a stirred solution of 2,2,6,6-tetramethylpiperidine (21.0 mL, 124 mmol) in dry THF (200 mL) in a −78° C. bath is added n-Butyl lithium (66.2 mL of a 2.5M solution in hexanes, 166 mmol). The solution is stirred at −78° C. for 30 minutes, followed by the addition of a solution of 2-(trifluoromethyl)nicotinic acid (7.90 g, 41.4 mmol) in THF (35 mL) via canula. The solution is stirred at −78° C. for 20 minutes, followed by warming to −50° C. for 1 hour (to provide the corresponding lithiated pyridine). In a separate flask, hexachloroethane (29.4 g, 124 mmol) is dissolved in THF (200 mL) and cooled to −15° C. and stirred rapidly. The solution containing the lithiated pyridine is added to the hexachloroethane solution via canula. After complete addition, the mixture is allowed to ward to room temperature. Water (ca 200 mL) is added, followed by removal of the tetrathydrofuran in vacuo. The remaining aqueous residue is extracted with ether-pentane (1:1, 100 mL). The organic layer was discarded. The aqueous layer is acidified with hydrochloric acid (ca. 50 mL of a 1.0 M solution). The aqueous layer is extracted with ethyl acetate. The organic layer is dried over anhydrous magnesium sulfate, filtered and concentrated in vacuo to afford 8.75 g (94%) of the title compound as a tan solid: 1H NMR (400 MHz, CD3OD), 8.63 (d, 1H), 7.79 (d, 1H). LC/MS (M+1)=226.1; [see: Schlosser et al., Eur. J. Org. Chem. 2003, 1559].
  • Preparation of Methyl 4-chloro-2-(trifluoromethyl)nicotinate
  • Figure US20110028452A1-20110203-C00016
  • To a stirred solution of 4-chloro-2-(trifluoromethyl)nicotinic acid (8.75 g, 38.8 mmol) in ethyl acetate (100 mL) in an ice bath is added a solution of diazomethane [generated from 12 g (116 mmol) of N-nitroso-N-methylurea] in diethyl ether (200 mL). The solution is stirred for 15 minutes, followed by the addition of acetic acid (until effervescence stops). The solution is concentrated in vacuo to afford 9.75 g (99%) of the title compound as a brown oil: 1H NMR (400 MHz, CD3OD), 8.686 (d, 1H), 7.828 (d, 1H), 3.937 (s, 3H). LC/MS (M+1)=240.1
  • Preparation of Methyl 4-azido-2-(trifluoromethyl)nicotinate
  • Figure US20110028452A1-20110203-C00017
  • To a stirred solution of methyl 4-chloro-2-(trifluoromethyl)nicotinate (9.75 g, 40.7 mmol) in DMF (300 mL) is added sodium azide (21.2 g, 326 mmol). The solution is warmed to 45° C. for 3 hours and then to 50° C. for 1 hour. The reaction is diluted with water (300 mL) and extracted with ether-pentane (1:1, ca. 1 L). The organic layer is back extracted with water (3×). The organic layer is dried over anhydrous magnesium sulfate, filtered and concentrated in vacuo to afford 9.50 g (95%) of the title compound as a brown oil: 1H NMR (400 MHz, CD3OD), 8.651 (d, 1H), 7.603 (d, 1H), 3.878 (s, 3H).
  • Preparation of Methyl 4-amino-2-(trifluoromethyl)nicotinate and 4-amino-2-(trifluoromethyl)nicotinic acid
  • Figure US20110028452A1-20110203-C00018
  • Cold (0° C. ice bath) concentrated hydriodic acid (182 mL of a 57% aqueous solution) is added to methyl 4-azido-2-(trifluoromethyl)nicotinate (7.50 g, 30.5 mmol) in a flask. The mixture is stirred at 0° C. until TLC (ethyl acetate-hexanes, 75:25) shows complete consumption of starting material. The mixture is poured over an ice cold solution of potassium hydroxide (95 g of solid KOH dissolved in 300 mL of water). The mixture is extracted with diethyl ether. The ethereal layer is washed with water, dried over anhydrous magnesium sulfate, filtered and concentrated in vacuo to afford a yellow solid. The solid is dissolved in a minimal amount of dichloromethane and passed through a silica gel plug, eluting with ethyl acetate-hexanes (10:90). The eluant is concentrated to afford 3.42 g (51%) of methyl 4-amino-2-(trifluoromethyl)nicotinate as an off-white solid. 1HNMR (400 MHz, CD3OD), 8.11 (d, 1H), 6.93 (d, 1H), 3.878 (s, 3H). LC/MS (M+1)=221.2
  • The remaining aqueous layer from above is acidified with concentrated hydrochloric acid to pH 2. The aqueous layer is extracted with ethyl acetate (16×). The organic layer is washed with water, dried over anhydrous magnesium sulfate, filtered and concentrated in vacuo to provide an orange solid. The orange solid is triturated in diethyl ether to afford 2.5 g (40%) of 4-amino-2-(trifluoromethyl)nicotinic acid as a yellow solid. 1H NMR (400 MHz, CD3OD), 8.09 (d, 1H), 6.93 (d, 1H), LC/MS (M+1)=207.1.
  • Preparation of 4-Amino-2-(trifluoromethyl)nicotinic acid
  • Figure US20110028452A1-20110203-C00019
  • To a stirred solution of methyl 4-amino-2-(trifluoromethyl)nicotinate (1.97 g, 8.97 mmol) in dioxane/methanol/water (3:2:1 ratio by volume) is added lithium hydroxide (0.365 g, 15.2 mmol). The mixture was heated at 85° C. for 8 hours. The reaction mixture was cooled to room temperature and conc. HCl was added dropwise to adjust the pH to 2-3. Saturated NaCl solution was added. The aqueous layer is extracted with ethyl acetate (12×). The organic layer is washed with water, dried over anhydrous magnesium sulfate, filtered and concentrated in vacuo to afford 1.75 g (95%) of 4-amino-2-(trifluoromethyl)nicotinic acid as a yellow solid. 1H NMR (400 MHz, CD3OD), 8.09 (d, 1H), 6.93 (d, 1H), LC/MS (M+1)=207.1.
  • Preparation of ethyl 2-amino-2-cyanoacetate
  • Figure US20110028452A1-20110203-C00020
  • To a stirred solution of the ethyl (hydroxyimino)cyanoacetate (10 g, 70.4 mmol) in water and sodium bicarbonate saturated solution (80 mL) was added sodium dithionite (34.3 g, 197 mmol). The reaction was heated up to 35° C. for 3 h. The solution was then saturated with sodium chloride and extracted with ethyl acetate (3×200 mL). The combined organic phases were dried over sodium sulfate and concentrated, giving an oil of ethyl 2-amino-2-cyanoacetate (1.6 g). 1H NMR (400 MHz, CDCl3) δ 4.42 (s, 1H), 4.32 (q, 2H), 1.33 (t, 3H).
  • Preparation of ethyl 2-cyano-2-formamidoacetate
  • Figure US20110028452A1-20110203-C00021
  • To ethyl 2-amino-2-cyanoacetate (800 mg, 6.24 mmol) in dichloromethane (5 mL) was added cyanomethyl formate (531 mg, 6.24 mmol) in dichloromethane (5 mL) dropwise at 0° C. The reaction was warmed up to room temperature for twelve hours. The reaction was concentrated and purified by silica gel chromatography to give an oil of ethyl 2-cyano-2-formamidoacetate (500 mg). NMR spectrum shows a mixture of starting material and desired product in a 1:1 ratio. The product was dissolved in methylene chloride (10 mL). To this solution was added cyanomethyl formate (797 mg, 9.37 mmol) and the reaction was stirred for twelve more hours. The reaction was concentrated and purified with silica gel chromatography to give an oil of ethyl 2-cyano-2-formamidoacetate (460 mg). 1H NMR (400 MHz, CDCl3) δ 8.30 (s, 1H), 6.61 (s, 1H), 5.55 (d, 2H), 4.36 (q, 2H), 1.35 (t, 3H).
  • Preparation of ethyl 5-aminothiazole-4-carboxylate
  • Figure US20110028452A1-20110203-C00022
  • A solution of ethyl 2-cyano-2-formamidoacetate (480 mg, 3.07 mmol) and Lawesson's reagent (746 mg, 1.84 mmol) in toluene (5 mL) was heated up to 80° C. for overnight. The reaction was then diluted with sodium carbonate aqueous solution (20 mL) and methylene chloride (50 mL). Organic phase was washed with brine (20 mL), dried over sodium sulfate and concentrated. Purified with silica gel chromatography to give a solid material of ethyl 5-aminothiazole-4-carboxylate (120 mg). 1H NMR (400 MHz, CDCl3) δ 7.86 (s, 1H), 5.98 (s, br, 2H), 4.38 (q, 2H), 1.41 (t, 3H). MS m/z 173.2 (M+H)+.
  • Preparation of ethyl 5-(2,5-dimethyl-1H-pyrrol-1-yl)thiazole-4-carboxylate
  • Figure US20110028452A1-20110203-C00023
  • A mixture of ethyl 5-aminothiazole-4-carboxylate (91.3 mg, 0.53 mmol), scandium (III) triflate (2.61 mg, 0.005 mmol) and 2,5-hexanedione (1.5 mL) was heated up to 180° C. for 1 h by microwave. The reaction was then concentrated and purified with silica gel chromatography to give a tan solid of ethyl 5-(2,5-dimethyl-1H-pyrrol-1-yl)thiazole-4-carboxylate (104 mg). 1H NMR (400 MHz, CDCl3) δ 8.82 (s, 1H), 5.92 (s, 2H), 4.25 (q, 2H), 2.01 (s, 6H), 1.20 (t, 3H). MS m/z 251.3 (M+H)+.
  • Preparation of ethyl 5-(2,5-dimethyl-1H-pyrrol-1-yl)thiazole-4-carboxylate
  • Figure US20110028452A1-20110203-C00024
  • To ethyl 5-(2,5-dimethyl-1H-pyrrol-1-yl)thiazole-4-carboxylate (104 mg, 0.415 mmol) in THF was added 1M DIBAL/THF (1.24 mL, 1.24 mmol) dropwise slowly at −78° C. and stirred for 1 h. The reaction mixture was warmed up to room temperature slowly and stirred for twelve hours. The reaction was then quenched with ice water (5 mL) and diluted with ethyl acetate (100 mL) and 2N NaOH (20 mL). The organic phase was then washed with brine (10 mL), dried over sodium sulfate and concentrated, giving a semi-solid of ethyl 5-(2,5-dimethyl-1H-pyrrol-1-yl)thiazole-4-carboxylate (86 mg). 1H NMR (400 MHz, CDCl3) δ 8.81 (s, 1H), 5.90 (s, 2H), 4.43 (s, 2H), 2.00 (s, 6H). MS m/z 209.3 (M+H)+.
  • Preparation of 5-(2,5-dimethyl-1H-pyrrol-1-yl)thiazole-4-carbaldehyde
  • Figure US20110028452A1-20110203-C00025
  • To a solution of (5-(2,5-dimethyl-1H-pyrrol-1-yl)thiazol-4-yl)methanol (86 mg, 0.41 mmol) in methylene chloride was added manganese dioxide (359 mg, 4.13 mmol). The reaction was warmed up to reflux for 3 h. The crude mixture was purified with silica gel chromatography to give an oil of 5-(2,5-dimethyl-1H-pyrrol-1-yl)thiazole-4-carbaldehyde (17 mg). 1H NMR (400 MHz, CDCl3) δ 9.72 (s, 1H), 8.87 (s, 1H), 5.98 (s, 2H), 2.05 (s, 6H). MS m/z 207.2 (M+H)+.
  • 2-(5-(2,5-dimethyl-1H-pyrrol-1-yl)thiazol-4-yl)-3-phenethyl-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00026
  • A solution of 4-amino-N-phenethyl-2-(trifluoromethyl)nicotinamide (25.5 mg, 0.082 mmol), 5-(2,5-dimethyl-1H-pyrrol-1-yl)thiazole-4-carbaldehyde (17 mg, 0.082 mmol) and TsOH (1.4 mg, 0.008 mmol) in toluene was heated at reflux overnight. The reaction was then filtered and to the crude mixture was added manganese oxide. The reaction was heated at 110° C. for 4 h. The reaction mixture was filtered, concentrated and purified with silica gel chromatography providing a solid, 2-(5-(2,5-dimethyl-1H-pyrrol-1-yl)thiazol-4-yl)-3-phenethyl-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one (7 mg). 1H NMR (400 MHz, CDCl3) δ 9.02 (s, 1H), 8.76 (d, 1H), 7.69 (m, 1H), 7.51 (m, 1H), 7.48 (d, 1H), 7.32 (m, 5H), 4.20 (m, 2H), 3.32 (m, 2H), 2.07 (s, 6H). MS m/z 496.5 (M+H)+.
  • Preparation of methyl 2-(difluoromethyl)benzoate
  • Figure US20110028452A1-20110203-C00027
  • A solution of methyl 2-formylbenzoate (1.0 g, 6.09 mmol) and bis(2-methoxyethyl)amino-sulfur trifluoride (4.04 g, 18.3 mmol) in methylene chloride (10 mL) was heated at reflux for twelve hours. The reaction was concentrated, diluted with ethyl acetate (200 mL) and water (100 mL). Sodium bicarbonate solid was used to neutralize the mixture slowly to pH of 8. The organic phase was then separated and washed with brine (50 mL), dried over sodium sulfate and concentrated. The residue was purified with silica gel chromatography to give methyl 2-(difluoromethyl)benzoate as an oil (700 mg). 1H NMR (400 MHz, CDCl3) δ 8.02 (d, 1H), 7.82 (d, 1H), 7.64 (t, 1H), 7.52 (m, 1H). 7.39 (t, 1H), 3.93 (s, 3H).
  • Preparation of (2-(difluoromethyl)phenyl)methanol
  • Figure US20110028452A1-20110203-C00028
  • To a solution of methyl 2-(difluoromethyl)benzoate (700 mg, 3.76 mmol) in THF was added 1M lithium aluminum hydride in THF (5.64 mL, 11.3 mmol) dropwise at room temperature for 1 h. The reaction was then poured into ice water (100 mL) and diluted with ethyl acetate (200 mL) and 2N sodium hydroxide aqueous solution (100 mL). The organic phase was then separated and washed with brine (50 mL), dried over sodium sulfate and concentrated, to give (2-(difluoromethyl)phenyl)methanol as an oil (480 mg).
  • 1H NMR (400 MHz, CDCl3) δ 7.56 (d, 1H), 7.41 (m, 3H), 6.93 (t, 1H), 4.82 (s, 2H).
  • Preparation of 2-(difluoromethyl)benzaldehyde
  • Figure US20110028452A1-20110203-C00029
  • A mixture of (2-(difluoromethyl)phenyl)methanol (100 mg, 0.632 mmol) and manganese oxide (275 mg, 3.16 mmol) in dichloromethane was stirred at room temperature for twelve hours and then at 45° C. for 1 h. The reaction was filtered through celite and concentrated, to give 2-(difluoromethyl)benzaldehyde as an oil (99 mg). 1H NMR (400 MHz, CDCl3) δ 10.2 (s, 1H), 7.94 (d, 1H), 7.81 (d, 1H), 7.70 (m, 2H), 6.93 (t, 1H).
  • Preparation of 2-methylnicotinaldehyde
  • Figure US20110028452A1-20110203-C00030
  • A mixture of (2-methylpyridin-3-yl)methanol (1.0 g, 8.12 mmol) in dichloromethane (10 mL) and manganese oxide (7.06 g, 81.2 mmol) was stirred at room temperature for sixty hours and then heated at 45° C. for 4 h. The reaction was filtered through celite and a plug of silica gel (5×7 cm) using ethyl acetate as eluent. The filtrate was then concentrated to give 2-methyl nicotinaldehyde as an oil (680 mg).
  • 1H NMR (400 MHz, CDCl3) δ 10.32 (s, 1H), 8.67 (dd, 1H), 8.09 (dd, 1H), 7.31 (dd, 1H), 2.88 (s, 3H).
  • Preparation of 3-(difluoromethyl)-2-methylpyridine
  • Figure US20110028452A1-20110203-C00031
  • A solution of 2-methylnicotinaldehyde (680 mg, 5.61 mmol) and bis(2-methoxyethyl)amino-sulfur trifluoride (3.72 g, 16.8 mmol) in dichloroethane (10 mL) was warmed at reflux overnight. The reaction was concentrated, diluted with ethyl acetate (200 mL) and water (100 mL). Sodium bicarbonate solid was used to neutralize the mixture slowly to pH of 8. The organic phase was then separated and washed with brine (50 mL), dried over sodium sulfate and concentrated. The residue was purified with flash chromatography (25+S Biotage, heptane/ethyl acetate=100:0 to 80:20), to give 3-(difluoromethyl)-2-methylpyridine as an oil (110 mg). 1H NMR (400 MHz, CDCl3) δ 7.58 (d, 1H), 7.81 (d, 1H), 7.22 (m, 1H), 6.77 (t, 1H), 2.64 (s, 3H).
  • Preparation of 3-(difluoromethyl)picolinaldehyde
  • Figure US20110028452A1-20110203-C00032
  • A mixture of 3-(difluoromethyl)-2-methylpyridine (110 mg, 0.768 mmol) and selenium oxide (102 mg, 0.922 mmol) in 1,4-dioxane (3 mL) was warmed at reflux for twelve hours. The reaction mixture was then concentrated and purified with flash chromatography (12+S Biotage, heptane/ethyl acetate=1:0 to 1:1), to give 3-(difluoromethyl)picolinaldehyde as an oil (11 mg). 1H NMR (400 MHz, CDCl3) δ 10.1 (s, 1H), 8.90 (d, 1H), 8.22 (d, 1H), 7.64 (m, 1H), 7.59 (t, 1H).
  • Preparation of 1,2-difluoro-4-(2-nitroprop-1-enyl)benzene
  • Figure US20110028452A1-20110203-C00033
  • 3,4-difluorobenzaldehyde (54 g, 380 mmol), ammonium acetate (19 g, 247 mmol), and 4A molecular sieves (54 g) were combined in nitroethane (810 mL) and heated at reflux for 24 hours. The reaction was filtered, and concentrated in vacuo. The resultant mixture was partitioned between ethyl acetate (500 mL) and water (300 mL). The organic phase was separated, washed with brine, dried over sodium sulfate, filtered and concentrated in vacuo. The residue was cooled to 4° C. for 2 hours then ethanol (100 mL) was added to give yellow crystals which were collected by filtration. (57.2 g 75.6% yield). 1H NMR (400 MHz, CDCl3) δ 8.0 (s, 1H), 7.15-7.25 (m, 2H), 3.7 (t, 1H), 2.4 (s, 3H).
  • Preparation of 1-(3,4-difluorophenyl)propan-2-amine hydrochloride
  • Figure US20110028452A1-20110203-C00034
  • (E,Z)-1,2-difluoro-4-(2-nitroprop-1-enyl)benzene (2.52 g, 12.65 mmol) was dissolved in anhydrous THF (30 mL) and cooled in an ice bath. To this was added a 1.0M THF solution of lithium aluminum hydride (38 mL, 38 mmol). The reaction was stirred at room temperature for 15 minutes then heated at reflux for 2 hours. The reaction mixture was cooled to room temperature, and worked up by the successive addition of water (1.5 mL), 15% NaOH (1.5 mL), and water (4.5 mL). The reaction mixture was stirred to granulate a white precipitate which was filtered. The filtrate was diluted with water (50 mL), extracted with ethyl acetate (2×100 mL). The organic phase was dried over sodium sulfate, filtered, and concentrated in vacuo. The residue was redissolved in ethyl acetate (25 mL), to which was added 4N HCl/dioxane solution (7 mL), and the resulting mixture was stirred at room temperature for 10 minutes. This mixture was concentrated in vacuo and triturated with hexanes to afford the product as a colorless solid (2.32 g, 88% yield). 1H NMR (400 MHz, CD3OD) δ 7.2 (m, 2H), 7.05 (m, 1H), 3.5 (m, 1H), 2.8-3.0 (dd, 2H), 1.25 (d, 3H). MS m/z 172.2 (M+H)+.
  • Preparation of 4-amino-N-(1-(3,4-difluorophenyl)propan-2-yl)-2-(trifluoromethyl)nicotinamide
  • Figure US20110028452A1-20110203-C00035
  • 4-amino-2-(trifluoromethyl)nicotinic acid (4.4 g, 18.14 mmol) was suspended in a mixture of anhydrous DMF (100 mL) and anhydrous dichloromethane (200 mL). To this was added triethylamine (6 g, 60 mmol), followed by 1-(3,4-difluorophenyl)propan-2-amine (3.75 g, 21.90 mmol). HBTU (9.63 g, 25.4 mmol) was added and the mixture was allowed to stir at room temperature overnight. The reaction mixture was concentrated in vacuo to remove dichloromethane. The resultant DMF solution was poured into water and extracted twice with ethyl acetate. The organic layer was washed successively with 1N NaOH and water, then dried over magnesium sulfate, filtered and concentrated in vacuo to give an oil. This oil was purified by silica gel column chromatography using 20-40% acetone/hexanes as eluant to provide the product as a colorless solid (6.57 g, 55% yield). 1H NMR (400 MHz, CDCl3) δ 8.2 (d, 1H), 7.1 (m, 2H), 6.95 (m, 1H), 6.7 (d, 1H), 4.07 (m, 1H), 4.9 (bd, 2H), 4.4 (m, 1H), 2.7-2.9 (dd, 2H), 1.2 (d, 2H). MS m/z 360.2 (M+H)+.
  • Preparation of (3-(benzyloxy)pyridin-2-yl)methanol
  • Figure US20110028452A1-20110203-C00036
  • 2-hydroxymethyl-3-hydroxypyridine (100.3 g, 620.7 mmol) was dissolved in acetone (1 L). Potassium carbonate (180 g, 1.3 mol) was added followed by benzyl bromide (127 g, 745 mmol). The mixture was heated at reflux for 48 hours. The resultant mixture was cooled to room temperature and filtered through celite. The filter cake was washed with acetone (1 L). The combined filtrate was concentrated in vacuo to provide an orange oil. This oil was purified by silica gel column chromatography using 10-40% ethyl acetate/hexanes as eluant. The resultant tan solid was recrystallized from hexanes to provide the product (88.2 g, 66% yield). 1H NMR (400 MHz, CDCl3) δ 8.2 (t, 1H), 7.4 (m, 5H), 7.15 (d, 2H), 5.1 (s, 2H), 4.8 (d, 2H), 4.3 (t, 1H).
  • Preparation of 3-(benzyloxy)picolinaldehyde
  • Figure US20110028452A1-20110203-C00037
  • (3-(benzyloxy)pyridin-2-yl)methanol (87 g, 400 mmol) was dissolved in anhydrous dioxane (80 mL). Manganese dioxide (351 g) was added and the mixture was heated for 2 hours at 80° C.
  • The reaction mixture was filtered through celite and the filter cake was washed with ethyl acetate (300 mL). The filtrate was concentrated in vacuo to yield a brown oil which solidified upon standing. (84.12 g, 98% yield). 1H NMR (400 MHz, CDCl3) δ 8.4 (d, 1H), 7.25-7.45 (m, 8H), 5.25 (s, 2H).
  • Preparation of 2-(3-(benzyloxy)pyridin-2-yl)-3-(1-(3,4-difluorophenyl)propan-2-yl)-5-(trifluoromethyl)-2,3-dihydropyrido[4,3-d]pyrimidin-4(1H)-one
  • Figure US20110028452A1-20110203-C00038
  • 4-amino-N-(1-(3,4-difluorophenyl)propan-2-yl)-2-(trifluoromethyl)nicotinamide (3.61 g, 10.05 mmol) and 3-(benzyloxy)picolinaldehyde (2.57 g, 12.1 mmol) were combined along with a catalytic amount of 4-toluenesulfonic acid (0.03 g, 0.20 mmol) in anhydrous toluene (150 mL). This was heated at reflux overnight in a round bottom flask fitted with a Dean-Stark trap. The resultant dark solution was cooled to room temperature, concentrated in vacuo to a dark brown oil. This oil was purified by silica gel column chromatography using 30% acetone/hexanes as eluant to provide the product diastereomers as an oil (4.61 g, 82% yield).
  • 1H NMR (400 MHz, CDCl3) δ 8.2 (m, 1H), 8.0 (m, 1H), 7.3-7.6 (m, 7H), 5.15 (d, 2H), 3.0 (dd, 1H), 2.8 (m, 1H), 1.25 (d, 3H). MS m/z 555.2 (M+H)+.
  • Preparation of 3-(1-(3,4-difluorophenyl)propan-2-yl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)-2,3-dihydropyrido[4,3-d]pyrimidin-4(1H)-one
  • Figure US20110028452A1-20110203-C00039
  • 2-(3-(benzyloxy)pyridin-2-yl)-3-(1-(3,4-difluorophenyl)propan-2-yl)-5-(trifluoromethyl)-2,3-dihydropyrido[4,3-d]pyrimidin-4(1H)-one (4.61 g, 8.31 mmol) was dissolved in absolute ethanol (150 mL). To this was added 10% palladium on carbon (0.50 g) and the mixture was hydrogenated on a Parr shaker at 45 PSI for 2.5 hours. The reaction was filtered through celite, and the filter cake washed with ethanol. The combined filtrates were concentrated in vacuo to an oil. This oil was purified by silica gel column chromatography using 20%-40% acetone/hexanes as eluant to yield the product diastereomers as a pale yellow foam (3.86 g, 77% yield).
  • 1H NMR (400 MHz, CD3OD) δ 8.0 (t, 1H), 7.8 (t, 1H), 7-7.2 (m, 5H), 4.8 (m, 1H), 4.5 (m, 1H), 2.8-3.1 (dd, 2H), 1.3 (d, 3H), 1.0 (d, 3H). MS m/z 465.3 (M+H)+.
  • Preparation of 3-(1-(3,4-difluorophenyl)propan-2-yl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00040
  • 3-(1-(3,4-difluorophenyl)propan-2-yl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)-2,3-dihydropyrido[4,3-d]pyrimidin-4(1H)-one (27.3 g, 58.8 mmol) was dissolved in 4-methyl-2-pentanone (500 mL). To this was added manganese dioxide (76.7 g, 882 mmol) and the mixture was heated to 90° C. for 1 hour. The reaction mixture was cooled and filtered through celite. The filter cake was washed with ethyl acetate. Combined filtrates were concentrated in vacuo to a pale yellow solid (8.35 g). The solid was slurried in hexanes and the resultant colorless solid was collected. The filtrate was concentrated and purified by silica gel column chromatography using 20-30% acetone/hexanes as eluant to yield additional product (3.47 g, 43.5% total yield).
  • Preparation of (R)-4-amino-N-(1-phenylpropan-2-yl)-2-(trifluoromethyl)nicotinamide
  • Figure US20110028452A1-20110203-C00041
  • 4-amino-2-(trifluoromethyl)nicotinic acid (15 g, 61.83 mmol) was suspended in a mixture of anhydrous DMF (300 mL) and anhydrous dichloromethane (500 mL). To this was added triethylamine (30.4 mL, 216 mmol), followed by L-amphetamine hydrochloride (12.7 g, 74.2 mmol). HBTU (30.5 g, 80.4 mmol) was added and the mixture was allowed to stir at room temperature overnight. The reaction mixture was concentrated in vacuo to remove dichloromethane. The resultant DMF solution was poured into water, extracted with ethyl acetate (1 L). The organic layer was washed successively with 1N NaOH, saturated sodium bicarbonate, water, dried over sodium sulfate, filtered and concentrated in vacuo to give an oil. This oil was purified by silica gel column chromatography using 40-70% ethyl acetate/heptanes as eluant to provide the product as a colorless foam (18.3 g, 92% yield). 1H NMR (400 MHz, CDCl3) δ 8.2 (d, 1H), 7.2-7.4 (m, 5H), 6.6 (d, 1H), 5.8 (d, 1H), 4.7 (bs, 2H), 4.5 (m, 1H), 2.9 (m, 2H), 1.25 (d, 3H). MS m/z 324.3 (M+H)+.
  • Preparation of 2-(2-methoxyphenyl)-3-((R)-1-phenylpropan-2-yl)-5-(trifluoromethyl)-2,3-dihydropyrido[4,3-d]pyrimidin-4(1H)-one
  • Figure US20110028452A1-20110203-C00042
  • (R)-4-amino-N-(1-phenylpropan-2-yl)-2-(trifluoromethyl)nicotinamide (16.23 g, 50.2 mmol) and ortho-anisaldehyde (8.2 g, 60.2 mmol) were combined along with a catalytic amount of 4-toluenesulfonic acid (0.174 g, 1.0 mmol) in anhydrous toluene (550 mL). This mixture was heated at reflux overnight in a round bottom flask fitted with a Dean-Stark trap. The resultant dark solution was cooled to room temperature, then concentrated in vacuo to a dark brown paste. This paste was suspended in ethyl acetate, and the resultant solids were collected by filtration to provide the product (6.94 g). The filtrate was concentrated in vacuo then purified by silica gel column chromatography using 40-60% ethyl acetate/hexanes as eluant to give 12.1 g of additional product diastereomers (84% total yield). 1H NMR (400 MHz, CDCl3) δ 8.19 (d, 1H), 7.1-7.3 (m, 7H), 6.9 (d, 1H), 6.8 (t, 1H), 6.5 (d, 1H), 5.95 (d, 1H), 5.7 (d, 1H), 4.8 (m, 1H), 3.95 (s, 3H), 3.0 (dd, 1H), 2.4 (dd, 1H), 1.3 (d, 3H). MS m/z 442.4 (M+H)+.
  • Preparation of (R)-2-(2-methoxyphenyl)-3-(1-phenylpropan-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00043
  • 2-(2-methoxyphenyl)-3-((R)-1-phenylpropan-2-yl)-5-(trifluoromethyl)-2,3-dihydropyrido[4,3-d]pyrimidin-4(1H)-one (19.04 g, 43.13 mmol) was dissolved in acetone (800 mL). To this was added a 5% aqueous solution of potassium permanganate (34.1 g KMnO4 in 700 mL water). The resultant mixture was stirred at room temperature overnight. The reaction was quenched by pouring it into a 10% aqueous solution of sodium bisulfite (3 L). The resulting aqueous solution was extracted with ethyl acetate (3×700 mL). The organic phase was washed with brine, dried over magnesium sulfate, filtered and concentrated in vacuo to provide an oil. This oil was purified by flash chromatography using 25-50% ethyl acetate/heptanes as eluant to provide the product as a solid. This solid was recrystallized from ethyl acetate/hexanes to yield a colorless solid (12.5 g). 1H NMR (400 MHz, CDCl3) δ 8.8 (d, 1H), 7.65 (d, 1H), 7.4 (t, 1H), 7.15 (m, 7H), 6.95 (t, 1H), 6.8 (d, 1H), 6.35 (d, 1H), 4.2 (m, 1H), 3.8 (s, 3H), 3.6 (dd, 1H), 2.9 (dd, 1H), 1.75 (d, 3H). MS m/z 440.3 (M+H)+.
  • Example 1 Preparation of 2-(2-Hydroxy-phenyl)-3-phenethyl-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one) Preparation 1a: 4-Amino-2-chloro-N-phenethyl-nicotinamide
  • Figure US20110028452A1-20110203-C00044
  • 4-Amino-2-chloro-nicotinic acid (1 g, 5.795 mmol) was dissolved in DMF (30 mL) and dichloromethane (50 mL). Phenethylamine (0.87 mL, 6.954 mmol, 1.2 equiv.) was added followed by TEA (1.21 mL, 8.693 mmol, 1.5 equiv.) and HBTU (2.64 g, 6.954 mmol, 1.2 equiv.) and the reaction stirred at 20° C. for 12 hours. The reaction mixture was then diluted with dichloromethane and the aqueous layer was extracted with 20% isopropyl alcohol/dichloromethane. The combined organic portions were washed with 0.5N NaOH solution, NH4Cl solution, water, brine, dried over anhydrous MgSO4, filtered and concentrated. The crude material was purified by column chromatography using 2:1 ethyl acetate:hexane to provide 1.23 g (77%) of product.
  • Preparation 1b: 5-Chloro-2-(2-methoxy-phenyl)-3-phenethyl-2,3-dihydro-1H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00045
  • To a solution of 4-amino-2-chloro-N-phenethyl-nicotinamide (808 mg, 2.93 mmol) in 8 mL of acetonitrile at 20° C. was added o-anisaldehyde (399 mg, 2.93 mmol) and a catalytic amount of p-toluenesulfonic acid (10 mg, 0.059 mmol). The resulting solution was then heated to reflux under Dean-Stark trap for 12 hours. The reaction mixture was cooled to 20° C. and diluted with ethyl acetate. The mixture was then partitioned between ethyl acetate and aqueous sodium bicarbonate. The organic phase was washed 3× with NaHCO3, 1× with water, 1× with brine and then dried over anhydrous Na2SO4, filtered and evaporated to dryness. 1.08 g of pale yellow sticky solid was obtained. The resulting crude material was used without further purification.
  • Preparation 1c: 5-Chloro-2-(2-methoxy-phenyl)-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00046
  • 5-Chloro-2-(2-methoxy-phenyl)-3-phenethyl-2,3-dihydro-1H-pyrido[4,3-d]pyrimidin-4-one (1.0 g, 2.54 mmol) was placed in a flask, acetone (10 mL) was added and the mixture stirred vigorously at 20° C. A 5% aqueous solution of potassium permanganate (KMnO4) was added (50.8 mmol, 2 equiv.). The reaction progress was monitored by HPLC. The reaction was stirred for 12 hours. Another 2 equiv. of KMnO4 was added and reaction stirred at ambient temperature for 4 hours until reaction was complete according to HPLC. The reaction was then quenched with 10% aqueous NaHSO3, filtered by gravity filtration and partitioned between ethyl acetate and water. The aqueous layer was extracted 3× with ethyl acetate, the combined organic layers washed 1× with water, 1× brine and then dried over anhydrous Na2SO4. Filtration and evaporation of the solvent gave 398 mg of tan solid. The original filter cake was suspended in dichloromethane and sonicated for 5 minutes, then repeated two more times. The suspensions were filtered through a nylon filter, combined and evaporated to dryness to give additional 282 mg of product for combined total crude yield of 680 mg (68%) of a tan solid. The product was used further without purification.
  • Preparation 1d: 5-Iodo-2-(2-methoxy-phenyl-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00047
  • A mixture of 5-chloro-2-(2-methoxy-phenyl)-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one (300 mg, 0.766 mmol), trimethylsilyl chloride (0.097 mL, 0.766 mmol) and sodium iodide (344 mg, 2.297 mmol, 3 equiv.) in propionitrile (7.66 mL) was heated to reflux under nitrogen in the dark for 4 hours. TLC showed no starting material remaining. The reaction mixture was cooled to ambient temperature and poured into 1N NaOH (30 mL). The resulting suspension was extracted with ethyl acetate (3×20 mL), the combined extracts were washed with water, brine and dried, filtered and concentrated in vacuo to give a brown oil. The crude product was purified by column chromatography using 10% ethyl acetate in dichloromethane to provide 235 mg (64%) of product.
  • Preparation 1e: 2-(2-Methoxy-phenyl)-3-phenethyl-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00048
  • In a flame dried 2-dram vial, a mixture of 5-iodo-2-(2-methoxy-phenyl)-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one (67 mg, 0.138 mmol), methyl 2,2-difluoro-2-(fluoro-sulfonyl)acetate (0.035 mL, 0.277 mmol, 2 equiv.), copper iodide (32 mg, 0.167 mmol, 1.2 equiv), HMPA (0.048 mL, 0.277 mmol, 2 equiv) in DMF (2 mL) was stirred under nitrogen at 80° C. for 12 hrs. The reaction mixture was then cooled to ambient temperature, diluted with dichloromethane and washed 3× with water. The organic phase was then dried over anhydrous MgSO4, filtered and concentrated. The crude material was purified on preparative TLC in 50:50 ethyl acetate:hexane to provide 16.8 mg of product (29%).
  • 2-(2-Hydroxy-phenyl)-3-phenethyl-5-trifluoromethyl-3H-pyrido[4,3-d]pyri-midin-4-one)
  • Figure US20110028452A1-20110203-C00049
  • 2-(2-Methoxy-phenyl)-3-phenethyl-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one (8.4 mg, 0.0197 mmol) was dissolved in dichloromethane (0.5 mL), the mixture cooled to 0° C. and a solution of boron trichloride was added (1M in hexane, 0.16 mL, 8 equiv.). The reaction mixture was then heated at 40° C. overnight. The reaction mixture was then cooled to ambient temperature, diluted with methanol (2 mL) and then heated to 70° C. for 1 hour. The solvent was evaporated and the residue partitioned between methylene chloride and saturated NaHCO3 solution. The aqueous layer was extracted 3× with dichloromethane. The pooled organic extracts were washed with water, brine, dried, filtered and concentrated. The crude product was purified using preparative TLC in 50:50 ethyl acetate:hexane to provide 5 mg (62%) of final product.
  • 1H NMR (400 MHz, CDCl3) δ 8.82 (d, 1H), 7.67 (d, 1H), 7.41 (t, 1H), 7.27 (m, 1H), 7.15 (m, 3H), 7.05 (m, 2H), 6.86 (m, 2H), 4.38 (t, 2H), 2.94 (t, 2H). MS m/z 412.3 (M+H)+.
  • Alternatively, 2-(2-Hydroxy-phenyl)-3-phenethyl-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one) can be prepared using 4-amino-2-(trifluoromethyl)nicotinic acid as the starting material.
  • Preparation 1f: 4-Amino-N-phenethyl-2-(trifluoromethyl)nicotinamide
  • Figure US20110028452A1-20110203-C00050
  • 4-Amino-2-(trifluoromethyl)nicotinic acid (150 mg, 0.78 mmol) was dissolved in DMF (4 mL) and dichloromethane (6 mL). Phenethylamine (106 mg, 0.87 mmol, 1.2 equiv.) was added followed by TEA (0.15 mL, 1.1 mmol, 1.5 equiv.) and HBTU (331 mg, 0.87 mmol, 1.2 equiv.) and the reaction stirred at 20° C. for 12 hours. The reaction mixture was then diluted with dichloromethane and the aqueous layer was extracted with dichloromethane. The combined organic portions were washed with 0.5N NaOH solution, NH4Cl solution, water, brine, dried over anhydrous MgSO4, filtered and concentrated. The crude material was purified by column chromatography using 2:1 ethyl acetate:hexane to provide 79 mg (36%) of product. The product can be converted to 2-(2-Hydroxy-phenyl)-3-phenethyl-5-trifluoromethyl-3H-pyrido[4,3-d]pyri-mid in-4-one) by using the title compound above, Preparation 1f, and following the procedures described in Preparation 1b (condensation with an appropriate aldehyde R2′CHO), Preparation 1c (oxidation with KMnO4) and the deprotection step using BCl3 (when converting a methoxy moiety in R2′ to the hydroxy in R2) as described above.
  • The following title compounds may be prepared in a manner analogous to Example 1 using Preparation 1a, 4-Amino-2-chloro-N-phenethyl-nicotinamide or 4-amino-2-chloro-6-methylnicotinic acid or Preparation 1f, 4-Amino-N-phenethyl-2-(trifluoromethyl)nicotinamide, or other appropriate nicotinamide derivative with the corresponding aldehyde of formula R2′CHO (it is to be understood that a methoxy group or benzyloxy group in the R2′ moiety can be deprotected to the corresponding hydroxy group to provide the R2 moiety in the compound of formula I).
  • Example 2 2-(3-Fluoro-2-hydroxy-phenyl)-3-phenethyl-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00051
  • 1H NMR (400 MHz, CDCl3) δ 8.90 (d, 1H), 7.75 (d, 1H), 7.25 (t, 3H), 7.05 (m, 3H), 6.90 (m, 2H), 4.39 (m, 2H), 2.94 (m, 2H). MS m/z 430.3 (M+H)+.
  • Example 3 2-(3-Hydroxy-pyridin-2-yl)-3-phenethyl-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00052
  • 1H NMR (400 MHz, CDCl3) δ 8.95 (d, 1H), 8.30 (d, 1H), 7.75 (d, 1H), 7.45 (m, 3H), 7.25 (m, 5H), 4.99 (m, 2H), 3.33 (m, 2H). MS m/z 413.2 (M+H)+.
  • Example 4a (R)-2-(2-Hydroxy-phenyl)-3-(1-methyl-2-phenyl-ethyl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00053
  • (R)-2-(2-methoxyphenyl)-3-(1-phenylpropan-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one (6.6 g, 15.02 mmol) was dissolved in anhydrous dichloromethane (75 mL) and cooled to 0° C. in an ice bath. To this was added a 1M dichloromethane solution of boron trichloride (31.5 mL, 31.5 mmol) slowly to maintain temperature. After addition, the reaction mixture was stirred for 2-5 minutes. The reaction mixture was transferred via canulla to a 0° C. aqueous solution of diethanolamine (10.3 g, 97.6 mmol in 150 mL water) with stirring. The reaction mixture was allowed to come to room temperature and stir for 1 hour. The reaction mixture was diluted with dichloromethane, and the layers separated. The organic layer was washed with brine, dried over magnesium sulfate, filtered, and concentrated in vacuo to provide a foam. This foam was purified by silica gel column chromatography using 20-40% ethyl acetate/hexanes as eluant to give a colorless foam which was then crystallized from ethyl acetate/hexanes to provide the product (4.2 g). 1H NMR (400 MHz, CDCl3) δ 8.79 (d, 1H), 7.6 (d, 1H), 7.45 (m, 1H), 7.00 (m, 6H), 6.90 (m, 2H), 3.60 (m, 2H), 3.10 (m, 2H), 1.99 (t, 3H). MS m/z 426.3 (M+H)+.
  • Example 4b (S)-2-(2-Hydroxy-phenyl)-3-(1-methyl-2-phenyl-ethyl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00054
  • Prepared analogous to Example 4a starting from (S)-2-(2-methoxyphenyl)-3-(1-phenylpropan-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one. 1H NMR (400 MHz, CDCl3) δ 8.79 (d, 1H), 7.6 (d, 1H), 7.45 (m, 1H), 7.00 (m, 6H), 6.90 (m, 2H), 3.60 (m, 2H), 3.10 (m, 2H), 1.99 (t, 3H). MS m/z 426.3 (M+H)+.
  • Example 5 (R)-2-(3-Hydroxy-pyridin-2-yl)-3-(1-methyl-2-phenyl-ethyl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00055
  • 1H NMR (400 MHz, CDCl3) δ 8.78 (d, 1H), 8.31 (dd, 1H), 7.49 (d, 1H), 7.41 (m, 2H), 7.02 (m, 2H), 6.88 (m, 3H), 5.31 (m, 1H), 3.62 (dd, 1H), 3.18 (dd, 1H), 1.89 (d, 3H). MS m/z 427.4 (M+1).
  • Example 6 (R,S)-2-(3-Hydroxy-pyridin-2-yl)-3-(1-methyl-2-(2-fluorophenyl)ethyl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00056
  • 1H NMR (400 MHz, CD3OD), 8.784 (d, 1H), 8.10 (dd, 1H), 7.701 (d, 1H), 7.40 (m, 2H), 7.14 (m, 1H), 6.97 (t, 1H), 6.890 (m 2H), 4.23 (m, 1H), 3.42 (dd, 2H), 3.29 (dd, 2H), 1.59 (d, 3H)
  • MS m/z (M+1) 445.4
  • The racemic compound of Example 6 can be separated into its single enantiomer components (Examples 6a and 6b) by preparative chromatography on a Chiralpak™ AS (10 cm×50 cm) column (Daicel (U.S.A.) Chemical Industries, Ltd, Fort Lee, N.J. 07024, U.S.A.) using Heptane/EtOH (90/10) as mobile phase at a flow rate of 475 mL/min.
  • Example 6a Enantiomer 1: 2-(3-Hydroxy-pyridin-2-yl)-3-(1-methyl-2-(2-fluorophenyl)ethyl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00057
  • 1H NMR (400 MHz, CD3OD), 8.78 (d, 1H), 8.10 (dd, 1H), 7.70 (d, 1H), 7.40 (m, 2H), 7.12 (m, 1H), 6.96 (dt, 1H), 6.87 (m, 2H), 4.28 (m, 1H), 3.34 (m, 1H), 3.30 (m, 1H), 1.59 (d, 2H)
  • LC/MS (M+1)=445.3
  • Example 6b Enantiomer 2: 2-(3-Hydroxy-pyridin-2-yl)-3-(1-methyl-2-(2-fluorophenyl)ethyl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00058
  • 1H NMR (400 MHz, CD3OD), 8.78 (d, 1H), 8.10 (dd, 1H), 7.70 (d, 1H), 7.40 (m, 2H), 7.12 (m, 1H), 6.96 (dt, 1H), 6.87 (m, 2H), 4.28 (m, 1H), 3.34 (m, 1H), 3.30 (m, 1H), 1.59 (d, 2H)
  • LC/MS (M+1)=445.3.
  • Example 7 2-(2-Hydroxyphenyl)-3-(2-fluorophenyl)ethyl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00059
  • 1H NMR (400 MHz, CD3OD) δ 8.8 (d, 1H), 7.75 (d, 1H), 7.4 (m, 1H), 7.2 (m, 1H), 6.8-7 (m, 6H), 4.2 (t, 2H), 2.95 (t, 2H). MS m/z 430.1 (M+H)+.
  • Example 8 2-(2-Hydroxyphenyl)-3-(3-fluorophenyl)ethyl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00060
  • 1H NMR (400 MHz, CD3OD) δ 8.8 (d, 1H), 7.75 (d, 1H), 7.45 (t, 1H), 7.15 (m, 2H), 7 (m, 2H), 6.9 (m, 1H), 6.65 (d, 1H), 6.55 (d, 1H), 4.15 (t, 2H), 2.9 (t, 2H). MS m/z 430.1 (M+H)+.
  • Example 9 2-(3-Fluoro-2-hydroxy-phenyl)-3-[2-(2-fluoro-phenyl)-ethyl]-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00061
  • 1H NMR (400 MHz, CD3OD) δ 8.85 (d, 1H), 7.8 (d, 1H), 7.35 (m, 1H), 7.2 (m, 1H), 7 (m, 2H), 6.9 (m, 1H), 6.65 (d, 1H), 6.6 (d, 1H), 4.15 (t, 2H), 2.9 (t, 2H). MS m/z 448.2 (M+H)+.
  • Example 10 2-(2-Hydroxy-phenyl)-7-methyl-3-(phenyl-ethyl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00062
  • 1H NMR (400 MHz, CDCl3) δ 7.6 (s, 1H), 7.45 (m, 1H), 7.30 (m, 1H), 7.12 (m, 5H), 6.90 (m, 2H), 4.44 (m, 2H), 2.99 (m, 2H), 2.80 (s, 3H). MS m/z 426.3 (M+H)+.
  • Example 11 2-(3-Fluoro-2-hydroxy-phenyl)-3-[2-(3-fluoro-phenyl)-ethyl]-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00063
  • 1H NMR (400 MHz, CD3OD) δ 8.8 (d, 1H), 7.75 (d, 1H), 7.25 (m, 1H), 7.2 (m, 1H), 6.8-7 (m, 4H), 6.75 (d, 1H), 4.2 (t, 2H), 2.95 (t, 2H). MS m/z 448.2 (M+H)+.
  • Example 12 3-[2-(2-Fluoro-phenyl)-ethyl]-2-(3-hydroxy-pyridin-2-yl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00064
  • 1H NMR (400 MHz, (CD3)2SO) δ 9.89 (s, 1H), 8.95 (d, 1H), 8.25 (d, 1H), 7.95 (d, 1H), 7.50 (m, 2H), 7.25 (m, 1H), 7.00 (m, 3H), 4.11 (m, 2H), 2.99 (m, 2H). MS m/z 431.1 (M+H)+.
  • Example 13 3-[2-(3,4-Difluoro-phenyl)-ethyl]-2-(3-hydroxy-pyridin-2-yl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00065
  • 1H NMR (400 MHz, CDCl3) δ 12.66 (brs, 1H), 8.86 (d, 1H), 8.30 (d, 1H), 7.66 (d, 1H), 7.44 (m, 2H), 7.10-6.95 (m, 3H), 4.84 (t, 2H), 3.24 (t, 2H). MS (LC-MS) 449.2 (M+H)+.
  • Example 14 3-[2-(2,4-Difluoro-phenyl)-ethyl]-2-(3-hydroxy-pyridin-2-yl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00066
  • 1H NMR (400 MHz, CDCl3) rotomeric mixture δ 12.40 (brs), 8.85 (d), 8.82 (d), 8.25 (m), 7.68 (d), 7.65 (d), 7.37 (m), 7.15 (q), 7.06 (m), 6.71 (m), 4.98 (t), 4.20 (t), 3.23 (t), 3.13 (t).
  • MS (LC-MS) 449.2 (M+H)+.
  • Example 15 3-[2-(3,4-Difluoro-phenyl)-ethyl]-2-(3-hydroxy-pyridin-2-yl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00067
  • 1H NMR (400 MHz, CDCl3) δ 8.83 (d, 1H), 7.84 (brs, 1H), 7.67 (d, 1H), 7.44 (dt, 1H), 7.28 (dd, 1H), 7.04 (t, 1H), 7.02 (d, 1H), 6.91 (q, 1H), 6.59 (m, 2H), 4.34 (t, 2H), 2.88 (t, 2H).
  • MS (LC-MS) 448.2 (M+H)+.
  • Example 16 2-(2-Hydroxy-phenyl)-5-methylamino-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00068
  • 5-Chloro-2-(2-hydroxy-phenyl)-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one, (obtained from Example 1, Preparation 1c and deprotection 1f), (0.0048 mmol, 1 equiv.) was dissolved in methanol (0.5 mL) then 500 uL of methylamine (2M solution in methanol) was added. The reaction mixture was then heated to 50° C. for 48 hour. The solvent was removed and crude products purified by preparative HPLC or preparative TLC (80% ethyl acetate in hexane).
  • 1H NMR (400 MHz, CD3OD) δ 8.17 (d, 1H), 7.40 (t, 1H), 7.15 (m, 3H), 7.05 (d, 1H), 6.94 (m, 2H), 6.80 (m, 2H), 6.59 (d, 1H), 4.10 (t, 2H), 3.08 (s, 3H), 2.85 (t, 2H). MS m/z 373.2 (M+H)+.
  • The following title compounds (Examples 17-23) may be prepared in a manner analogous to Example 16 using procedures from Example 16 with the corresponding amine followed by deprotection conditions employing BCl3 found in the final step of Example 1.
  • Example 17 2-(2-Hydroxy-phenyl)-5-isopropylamino-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00069
  • 1H NMR (400 MHz, CD3OD) δ 9.00 (m, 1H), 8.17 (d, 1H), 7.40 (t, 1H), 7.15 (m, 3H), 7.05 (d, 1H), 6.94 (m, 2H), 6.80 (m, 2H), 6.59 (d, 1H), 4.30 (m, 1H), 4.09 (t, 2H), 2.85 (t, 2H), 1.32 (d, 6H). MS m/z 401.3 (M+H)+.
  • Example 18 2-(2-Hydroxy-phenyl)-3-phenethyl-5-pyrrolidin-1-yl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00070
  • 1H NMR (400 MHz, CD3OD) δ 8.12 (d, 1H), 7.41 (t, 1H), 7.14 (m, 4H), 6.95 (m, 2H), 6.85 (m, 2H), 6.70 (d, 1H), 4.12 (t, 2H), 3.51 (t, 4H), 2.81 (t, 2H), 1.97 (m, 4H). MS m/z 413.2 (M+H)+.
  • Example 19 2-(2-Hydroxy-phenyl)-5-(4-methyl-piperazin-1-yl)-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00071
  • 1H NMR (400 MHz, CD3OD) δ 8.27 (d, 1H), 7.4 (m, 1H), 7.11-7.15 (m, 4H), 6.94 (m, 2H), 6.88 (d, 1H), 6.82 (m, 2H), 4.15 (t, 2H), 3.46 (bs, 4H), 2.80 (t, 2H), 2.65 (t, 4H), 2.36 (S, 3H). MS m/z 440.1 (M+H)+.
  • Example 20 2-(2-Hydroxy-phenyl)-3-phenethyl-5-piperazin-1-yl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00072
  • 1H NMR (400 MHz, CD3OD) δ8.26 (d, 1H), 7.41 (t, 1H), 7.14-7.10 (m, 4H), 6.92-6.96 (m, 2H), 6.89 (d, 1H), 6.83-6.81 (m, 2H), 4.16 (t, 2H), 3.42 (t, 4H), 3.04 (t, 4H), 2.79-2.83 (m, 2H). MS m/z 428.2 (M+H)+.
  • Example 21 5-Dimethylamino-2-(2-hydroxy-phenyl)-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00073
  • 1H NMR (400 MHz, CD3OD) δ 8.19 (d, 1H), 7.39-7.43 (m, 1H), 7.15-7.09 (m, 4H), 6.98-6.94 (m, 2H), 6.83-6.80 (m, 2H), 6.75 (d, 1H), 4.11 (t, 2H), 3.07 (s, 6H), 2.82 (t, 2H). MS m/z 387.1 (M+H)+.
  • Example 22 2-(2-Hydroxy-phenyl)-5-morpholin-4-yl-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00074
  • 1H NMR (400 MHz, CD3OD) δ 8.28 (d, 1H), 7.44-7.39 (m, 1H), 7.15-7.11 (m, 4H), 6.99-6.95 (m, 2H), 6.89 (d, 1H), 6.83-6.81 (m, 2H), 4.14 (t, 2H), 3.86 (t, 4H), 3.41 (t, 4H), 2.81 (t, 2H). MS m/z 429.2 (M+H)+.
  • Example 23 5-Azetidin-1-yl-2-(2-hydroxy-phenyl)-3-phenethyl-3H-pyrido[4,3-d-pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00075
  • 1H NMR (400 MHz, CD3OD) δ 8.19 (d, 1H), 7.4 (t, 1H), 7.15-7.13 (m, 3H), 7.07-7.04 (m, 1H), 6.95-6.93 (m, 2H), 6.82-6.8 (m, 2H), 6.71 (d, 1H), 4.26 (t, 4H), 4.09 (t, 2H), 2.82 (t, 2H), 2.37 (q, 2H). MS m/z 399.1 (M+H)+.
  • Example 24 Preparation of 2-(2-Hydroxy-phenyl)-3-phenethyl-5-phenyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00076
  • 5-Chloro-2-(2-methoxy-phenyl)-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one, phenylboronic acid (1.1 equiv.), palladium tetrakis(triphenylphosphine) (7 mol %) and 2N aqueous Na2CO3 (4 equiv.) were combined in dioxane (0.5 mL) and heated in a microwave for 15 minutes to 140° C. The reaction mixture was cooled to room temperature, diluted with water and extracted (3×) with ethyl acetate. The combined organic extracts were washed with brine, dried over anhydrous MgSO4 and filtered. The crude products were purified on preparative TLC using 1:1 ethyl acetate:hexane. The desired product can be obtained by using the methyl ether deprotection conditions employing BCl3 and isolation conditions found in the final step of Example 1. 1H NMR (400 MHz, CDCl3) δ 8.90 (s, 1H), 8.78 (d, 1H), 7.53 (m, 2H), 7.46 (m, 3H), 7.37 (t, 1H), 7.29 (d, 1H), 7.13 (m, 3H), 6.97 (m, 2H), 6.77 (m, 2H), 4.30 (t, 2H), 2.79 (t, 2H). MS m/z 420.4 (M+H)+.
  • The following title compounds (Examples 25-29) may be prepared in a manner analogous to Example 24 using procedures from Example 24 with the corresponding boronic acid followed by deprotection conditions employing BCl3 and isolation conditions found in the final step of Example 1.
  • Example 25 5-Benzyl-2-(2-hydroxy-phenyl)-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00077
  • 1H NMR (400 MHz, CDCl3) δ 9.40 (s, 1H), 7.58 (d, 1H), 7.40-7.26 (m, 4H), 7.24 (m, 2H), 7.12 (m, 5H), 6.96 (m, 2H), 6.74 (m, 2H), 4.87 (s, 2H), 4.29 (t, 2H), 2.81 (t, 2H).
  • MS m/z 434.3 (M+H)+.
  • Example 26 2-(2-Hydroxy-phenyl)-5-methyl-3-phenethyl-3H-pyrido[4,3-d]pyrimidi-4-one
  • Figure US20110028452A1-20110203-C00078
  • 1H NMR (400 MHz, CDCl3) δ 8.44 (d, 1H), 7.42 (m, 2H), 7.30-7.10 (m, 5H), 7.00 (m, 1H); 6.82 (m, 2H), 4.30 (t, 2H), 3.16 (s, 3H), 2.89 (t, 2H). MS m/z 358.3 (M+H)+.
  • Example 27 5-(6-Dimethylamino-pyridin-3-yl)-2-(2-hydroxy-phenyl)-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00079
  • 1H NMR (400 MHz, CD3OD) δ 8.72 (d, 1H), 8.30 (s, 1H), 7.72 (d, 1H), 7.46 (m, 2H), 7.15 (m, 4H), 7.00 (m, 2H), 6.80 (m, 2H), 6.73 (d, 1H), 4.08 (t, 2H), 3.19 (s, 6H), 2.80 (t, 2H). MS m/z 464.4 (M+H)+.
  • Example 28 5-(6-Dimethylamino-5-methyl-pyridin-3-yl)-2-(2-hydroxy-phenyl)-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00080
  • 1H NMR (400 MHz, CD3OD) δ 8.77 (d, 1H), 8.20 (s, 1H), 7.66 (s, 1H), 7.54 (d, 1H), 7.45 (m, 1H), 7.19 (d, 1H), 7.12 (m, 3H), 6.99 (m, 2H), 6.78 (m, 2H), 4.09 (t, 2H), 2.87 (s, 6H), 2.80 (t, 2H), 2.40 (s, 3H). MS m/z 478.4 (M+H)+.
  • Example 29 5-(6-pyrrolidine-5-pyridin-3-yl)-2-(2-hydroxy-phenyl)-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00081
  • 1H NMR (400 MHz, CD3OD) δ 8.72 (d, 1H), 8.28 (s, 1H), 7.72 (d, 1H), 7.45 (m, 2H), 7.19 (d, 1H), 7.15 (m, 3H), 6.99 (m, 2H), 6.79 (m, 2H), 6.58 (d, 1H), 4.09 (t, 2H), 3.55 (t, 4H), 2.80 (t, 2H), 2.07 (m, 4H). MS m/z 490.4 (M+H)+.
  • Example 30 Preparation of 2-(2-Hydroxy-phenyl)-5-methoxy-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one Preparation of 2-(2-Benzyloxy-phenyl)-5-chloro-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00082
  • 5-Chloro-2-(2-methoxy-phenyl)-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one was subjected to the same deprotection conditions found in the final step of Example 1 to provide the desired 5-chloro-2-(2-hydroxy-phenyl)-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one. A mixture of 5-chloro-2-(2-hydroxy-phenyl)-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one (88 mg, 0.233 mmol), potassium carbonate (64 mg, 0.466 mmol, 2 equiv.) and benzyl bromide (0.029 mL, 0.245 mmol, 1.05 equiv.) in 2 mL of acetone was heated to reflux overnight. The reaction mixture was cooled to RT, filtered through celite, concentrated and the residue purified on preparative TLC plate using 1:1 ethyl acetate:heptane to give 95 mg (87%) of the desired product.
  • Preparation of 2-(2-Benzyloxy-phenyl)-5-methoxy-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00083
  • A mixture of 2-(2-benzyloxy-phenyl)-5-chloro-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one, sodium hydride (60% suspension in mineral oil) (1.5 equiv.), and corresponding alcohol (1.5 equiv.) in THF was heated at 70° C. overnight. The reaction was cooled to RT, solvent removed, water and ethyl acetate were added and phases separated. The aqueous phase was washed with ethyl acetate (2×), organic portions combined, washed with water, brine, dried over anhydrous MgSO4, filtered and concentrated. The residue was purified on preparative TLC plate using 1:1 ethyl acetate:heptane.
  • 2-(2-Hydroxy-phenyl-methoxy-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00084
  • 2-(2-Benzyloxy-phenyl)-5-methoxy-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one dissolved in methanol (0.05M) was subjected to H-cube hydrogenation (room temperature, Pd/C cartridge). Solvent was then removed and product purified on preparative TLC plate to afford the desired 2-(2-Hydroxy-phenyl)-5-methoxy-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one.
  • 1H NMR (400 MHz, CD3OD) δ 8.33 (d, 1H), 7.42 (t, 1H), 7.16 (m, 3H), 7.05 (m, 2H), 6.95 (m, 2H), 6.80 (m, 2H), 4.10 (m, 5H), 2.86 (t, 2H). MS m/z 374.2 (M+H)+.
  • The following title compounds (Examples 31-36) may be prepared in a manner analogous to Example 30 using procedures from Example 30 with the corresponding alcohol followed by H-cube hydrogenation.
  • Example 31 2-(2-Hydroxy-phenyl)-5-(1-methyl-cyclopropylmethoxy)-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00085
  • 1H NMR (400 MHz, CD3OD) δ 8.33 (d, 1H), 7.45 (t, 1H), 7.19 (m, 3H), 7.12 (d, 1H), 7.05 (d, 1H), 7.00 (m, 2H), 6.83 (m, 2H), 4.39 (s, 2H), 4.12 (t, 2H), 2.91 (t, 2H), 1.32 (s, 3H), 0.70 (t, 2H), 0.47 (t, 2H). MS m/z 428.2 (M+H)+.
  • Example 32 2-(2-Hydroxy-phenyl)-3-phenethyl-5-propoxy-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00086
  • 1H NMR (400 MHz, CD3OD) δ 8.33 (d, 1H), 7.45 (t, 1H), 7.19 (m, 3H), 7.12 (d, 1H), 7.05 (d, 1H), 7.00 (m, 2H), 6.83 (m, 2H), 4.52 (t, 2H), 4.12 (t, 2H), 2.91 (t, 2H), 1.93 (m, 2H), 1.12 (t, 3H). MS m/z 402.2 (M+H)+.
  • Example 33 5-Cyclobutyloxy-2-(2-hydroxy-phenyl)-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00087
  • 1H NMR (400 MHz, CD3OD) δ 8.33 (d, 1H), 7.45 (t, 1H), 7.19 (m, 3H), 7.12 (d, 1H), 7.05 (d, 1H), 7.00 (m, 2H), 6.83 (m, 2H), 5.41 (m, 1H), 4.12 (t, 2H), 2.91 (t, 2H), 2.55 (m, 2H), 2.36 (m, 2H), 1.93 (m, 1H), 1.76 (m, 1H). MS m/z 414.2 (M+H)+.
  • Example 34 2-(2-Hydroxy-phenyl)-5-isobutoxy-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00088
  • 1H NMR (400 MHz, CD3OD) δ 8.33 (d, 1H), 7.45 (t, 1H), 7.19 (m, 3H), 7.12 (d, 1H), 7.05 (d, 1H), 7.00 (m, 2H), 6.83 (m, 2H), 4.33 (d, 2H), 4.12 (t, 2H), 2.91 (t, 2H), 2.28 (m, 1H), 1.12 (d, 6H). MS m/z 416.3 (M+H)+.
  • Example 35 2-(2-Hydroxy-phenyl)-5-isopropoxy-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00089
  • 1H NMR (400 MHz, CD3OD) δ 8.34 (d, 1H), 7.46 (t, 1H), 7.19 (m, 3H), 7.10 (d, 1H), 7.04 (d, 1H), 7.00 (m, 2H), 6.82 (m, 2H), 5.61 (m, 1H), 4.12 (t, 2H), 2.90 (t, 2H), 1.50 (d, 6H). MS m/z 402.3 (M+H)+.
  • Example 36 2-(2-Hydroxy-phenyl)-3-phenethyl-5-(2,2,2-trifluoro-ethoxy)-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00090
  • 1H NMR (400 MHz, CD3OD) δ 8.36 (d, 1H), 7.45 (t, 1H), 7.15 (m, 5H), 7.00 (m, 2H), 6.83 (m, 2H), 5.10 (q, 2H), 4.15 (m, 2H), 2.90 (t, 2H). MS m/z 442.2 (M+H)+.
  • Example 37 Preparation of 3-(1-(3,4-difluorophenyl)propan-2-yl)-2-(3-hydroxypyridin-2-yl) 5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00091
  • 2-(3-(benzyloxy)pyridin-2-yl)-3-(1-(3,4-difluorophenyl)propan-2-yl)-5-(trifluoromethyl)-2,3-dihydropyrido[4,3-d]pyrimidin-4(1H)-one (12.1 grams, 21.8 mmol) prepared by the same reactions conditions described above was dissolved in 300 mL of ethanol and added to a parr bottle that contained 10% Pd(C). The mixture was subjected to hydrogenation condition (45 psi) for two hours. At this time, the reaction mixture was filtered through Celite and washed with ethanol. The fitrate was concentrated in vacuo and purified by silica gel chromatography to afford a pale yellow foam of 3-(1-(3,4-difluorophenyl)propan-2-yl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)-2,3-dihydropyrido[4,3-d]pyrimidin-4(1H)-one.
  • Figure US20110028452A1-20110203-C00092
  • 3-(1-(3,4-difluorophenyl)propan-2-yl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)-2,3-dihydropyrido[4,3-d]pyrimidin-4(1H)-one (5.90 g, 12.7 mmol) was dissolved in 500 mL of 4-methyl-2-pentanone. To this solution, manganese dioxide was added (16.6 g, 191 mmol) and the mixture was heated to 90° C. for 1 hour. The reaction was cooled, filtered through Celite and washed with CH2CL2. The filtrate was collected and concentrated in vacuo as a yellow solid which was purified by silica gel chromatography to afford the racemic mixture 3-(1-(3,4-difluorophenyl)propan-2-yl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one.
  • The racemic compound of Example 37 can be separated into its single enantiomer components (Examples 37a and 37b) by preparative chromatography on a Chiralcel OD-H (2.1 cm×25 cm) column (Daicel (U.S.A.) Chemical Industries, Ltd, Fort Lee, N.J. 07024, U.S.A.) using MeOH/CHCL3 (3/1) as mobile phase at a flow rate of 65 g/min.
  • Example 37a (R)-3-(1-(3,4-difluorophenyl)propan-2-yl)-2-(3-hydroxypyridin-2-yl) 5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00093
  • 1H NMR (400 MHz, CDCl3) δ 11.4 (s, 1H), 8.85 (d, 1H), 8.39 (d, 1H), 7.60 (d, 1H), 7.42 (m, 2H), 6.99 (m, 2H), 6.67 (brs, 1H), 5.24 (m, 1H), 3.63 (m, 1H), 3.24 (m, 1H), 1.90 (d, 3H).
  • MS m/z 463.3 (M+H)+.
  • Example 37b (S)-3-(1-(3,4-difluorophenyl)propan-2-yl)-2-(3-hydroxypyridin-2-yl) 5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00094
  • 1H NMR (400 MHz, CDCl3) δ 11.4 (s, 1H), 8.85 (d, 1H), 8.39 (d, 1H), 7.60 (d, 1H), 7.42 (m, 2H), 6.99 (m, 2H), 6.67 (brs, 1H), 5.24 (m, 1H), 3.63 (m, 1H), 3.24 (m, 1H), 1.90 (d, 3H).
  • MS m/z 463.3 (M+H)+.
  • Example 38a 3-(1-(2,4-difluorophenyl)propan-2-yl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00095
  • 1H NMR (400 MHz, CDCl3) δ 8.90 (d, 1H), 8.30 (d, 1H), 7.60 (d, 1H), 7.42 (m, 2H), 6.99 (m, 1H), 6.66 (m, 2H), 5.65 (m, 1H), 3.63 (m, 1H), 3.20 (m, 1H), 1.90 (d, 3H).
  • MS m/z 463.2 (M+H)+.
  • Example 38b 3-(1-(2,4-difluorophenyl)propan-2-yl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00096
  • 1H NMR (400 MHz, CDCl3) δ 8.90 (d, 1H), 8.30 (d, 1H), 7.60 (d, 1H), 7.42 (m, 2H), 6.99 (m, 1H), 6.66 (m, 2H), 5.65 (m, 1H), 3.63 (m, 1H), 3.20 (m, 1H), 1.90 (d, 3H).
  • MS m/z 463.2 (M+H)+.
  • Example 39 3-(2-methoxyphenethyl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00097
  • 1H NMR (400 MHz, CDCl3) δ 10.64 (s, 1H), 8.87 (d, 1H), 8.17 (dd, 1H), 7.87 (d, 1H), 7.46 (dq, 2H), 7.10 (dt, 1H), 6.81 (t, 1H), 6.79 (t, 1H), 6.721 (t, 1H), 3.97 (t, 2H), 3.45 (s, 3H), 2.75 (t, 2H). MS m/z 443.1 (M+H)+.
  • Example 40 3-(2,3-difluorophenethyl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00098
  • 1H NMR (400 MHz, CDCl3) δ 8.85 (d, 1H), 8.28 (m, 1H), 7.65 (d, 1H), 7.37 (m, 2H), 6.95 (m, 4H), 5.01 (t, 2H), 3.31 (t, 2H). MS m/z 449.1 (M+H)+.
  • Example 41 3-(5-fluoro-2-methoxyphenethyl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00099
  • 1H NMR (400 MHz, CDCl3) δ 8.90 (d, 1H), 8.20 (d, 1H), 7.70 (d, 1H), 7.38 (m, 2H), 6.80 (m, 1H), 6.56 (m, 2H), 3.40 (s, 3H), 3.01 (m, 2H). MS m/z 461.2 (M+H)+.
  • Example 42 3-(2-fluoro-6-methoxyphenethyl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00100
  • MS m/z 461 (M+H)+.
  • Example 43 2-(3-hydroxypyridin-2-yl)-3-(1-(2-methoxyphenyl)propan-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00101
  • 1H NMR (400 MHz, CD3OD) δ 8.90 (d, 1H), 8.10 (m, 1H), 7.70 (d, 1H), 7.40 (m, 2H), 7.10 (t, 1H), 6.90 (d, 1H), 6.70 (d, 1H), 6.61 (t, 1H), 4.45 (m, 1H), 3.40 (s, 3H), 3.39 (m, 1H) 3.01 (m, 2H), 1.75 (d, 3H). MS m/z 457.3 (M+H)+.
  • Example 43a 2-(3-hydroxypyridin-2-yl)-3-(1-(2-methoxyphenyl)propan-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00102
  • 1H NMR (400 MHz, CD3OD) δ 8.90 (d, 1H), 8.10 (m, 1H), 7.70 (d, 1H), 7.40 (m, 2H), 7.10 (t, 1H), 6.90 (d, 1H), 6.70 (d, 1H), 6.61 (t, 1H), 4.45 (m, 1H), 3.40 (s, 3H), 3.39 (m, 1H) 3.01 (m, 2H), 1.75 (d, 3H). MS m/z 457.3 (M+H)+.
  • Example 43b 2-(3-hydroxypyridin-2-yl)-3-(1-(2-methoxyphenyl)propan-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00103
  • 1H NMR (400 MHz, CD3OD) δ 8.90 (d, 1H), 8.10 (m, 1H), 7.70 (d, 1H), 7.40 (m, 2H), 7.10 (t, 1H), 6.90 (d, 1H), 6.70 (d, 1H), 6.61 (t, 1H), 4.45 (m, 1H), 3.40 (s, 3H), 3.39 (m, 1H) 3.01 (m, 2H), 1.75 (d, 3H). MS m/z 457.3 (M+H)+.
  • Example 44 3-(1-(2-fluorophenyl)butan-2-yl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00104
  • 1H NMR (400 MHz, CDCl3) δ 8.75 (d, 1H), 8.07 (brs, 1H), 7.51 (d, 1H), 7.36 (t, 2H), 7.07 (m, 2H), 6.95 (m, 2H), 6.84 (m, 2H), 6.76 (t, 1H), 4.48 (m, 1H), 3.48 (dd, 1H), 3.24 (dd, 1H), 2.41 (m, 1H), 2.29 (m, 1H), 0.91 (t, 3H). MS m/z 458.3 (M+H)+.
  • Example 45 3-(3-fluoro-2-methoxyphenethyl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00105
  • 1H NMR (400 MHz, CD3OD) δ 8.90 (d, 1H), 8.18 (d, 1H), 7.80 (d, 1H), 7.45 (m, 2H), 6.99 (m, 1H), 6.85 (m, 1H), 6.79 (d, 1H), 4.25 (m, 2H), 3.60 (s, 3H), 2.99 (m, 2H). MS m/z 461.2 (M+H)+.
  • Example 46 3-(2-cyclopentylethyl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00106
  • 1H NMR (400 MHz, dmso-d6) δ 10.64 (s, 1H), 8.87 (d, 1H), 8.15 (dd, 1H), 7.86 (d, 1H), 7.43 (m, 2H), 3.77 (brm, 2H), 1.48 (brm, 5H), 1.34 (brm, 4H), 0.69 (brm, 2H). MS m/z 405.2 (M+H)+.
  • Example 47 3-(2-cyclohexylethyl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00107
  • 1H NMR (400 MHz, dmso-d6) δ 10.62 (s, 1H), 8.87 (d, 1H), 8.15 (dd, 1H), 7.86 (d, 1H), 7.43 (m, 2H), 3.81 (m, 2H), 1.48 (brm, 5H), 1.50-1.25 (m, 7H), 1.00 (m, 4H), 0.58 (m, 2H). MS m/z 419.2 (M+H)+.
  • Example 48a (R)-3-(1-cyclohexylpropan-2-yl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00108
  • 1H NMR (400 MHz, dmso-d6) δ 10.61 (s, 1H), 8.85 (d, 1H), 8.12 (dd, 1H), 7.83 (d, 1H), 7.39 (m, 2H), 3.97 (brs, 1H), 1.82 (m, 1H), 1.72 (m, 1H), 1.46 (m, 2H), 1.42 (d, 3H), 1.35 (m, 2H), 1.17-0.93 (m, 5H), 0.66 (m, 1H), 0.56 (m, 1H). MS m/z 433.3 (M+H)+.
  • Example 48b (S)-3-(1-cyclohexylpropan-2-yl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00109
  • 1H NMR (400 MHz, dmso-d6) δ 10.61 (s, 1H), 8.85 (d, 1H), 8.12 (dd, 1H), 7.83 (d, 1H), 7.39 (m, 2H), 3.97 (brs, 1H), 1.82 (m, 1H), 1.72 (m, 1H), 1.46 (m, 2H), 1.42 (d, 3H), 1.35 (m, 2H), 1.17-0.93 (m, 5H), 0.66 (m, 1H), 0.56 (m, 1H). MS m/z 433.3 (M+H)+.
  • Example 49 (R,S)-2-(3-hydroxypyridin-2-yl)-3-(2-(tetrahydro-2H-pyran-2-yl)ethyl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00110
  • 1H NMR (400 MHz, dmso-d6) δ 10.61 (s, 1H), 8.86 (d, 1H), 8.15 (dd, 1H), 7.85 (d, 1H), 7.41 (m, 2H), 3.94 (m, 1H), 3.79 (m, 1H), 3.56 (d, 1H), 3.10 (t, 1H), 3.02 (m, 1H), 1.57 (m, 3H), 1.27 (m, 3H), 1.07 (m, 4H), 0.93 (m, 1H). MS m/z 421.3 (M+H)+.
  • Example 50 (R)-2-(3-fluoro-2-hydroxyphenyl)-3-(1-phenylpropan-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00111
  • 1H NMR (400 MHz, CDCl3) δ 8.80 (d, 1H), 7.60 (d, 1H), 7.2 (m, 5H), 6.90 (m, 1H), 6.80 (m, 1H), 6.67 (brs, 1H), 4.44 (m, 1H), 3.63 (m, 1H), 3.10 (m, 1H), 1.90 (d, 3H). MS m/z 444.3 (M+H)+.
  • Example 51 (R)-2-(1H-imidazol-2-yl)-3-(1-phenylpropan-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00112
  • 1H NMR (400 MHz, CDCl3) δ 8.79 (d, 1H), 7.58 (d, 1H), 7.4 (s, 1H), 7.30 (d, 1H), 7.10 (m, 4H), 6.98 (m, 1H), 3.70 (m, 1H), 3.30 (m, 1H), 1.90 (d, 3H). MS m/z 400.2 (M+H)+.
  • Example 52 N-(2-(3-(2-fluorophenethyl)-4-oxo-5-(trifluoromethyl)-3,4-dihydropyrido[4,3-d]pyrimidin-2-yl)pyridin-3-yl)acetamide
  • Figure US20110028452A1-20110203-C00113
  • 1H NMR (400 MHz, CDCl3) δ 9.40 (brs, 1H), 8.98 (d, 1H), 8.80 (d, 1H), 8.45 (m, 1H), 7.67 (d, 1H), 7.44 (m, 1H), 7.10 (m, 1H), 7.00 (m, 1H), 6.99 (m, 1H), 6.85 (m, 1H), 4.60 (m, 2H), 3.05 (m, 2H), 2.10 (s, 3H). MS m/z 472.1 (M+H)+.
  • Example 53 (R)-3-(1-phenylpropan-2-yl)-2-(thiazol-4-yl)-5(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00114
  • 1H NMR (400 MHz, CDCl3) δ 8.99 (s, 1H), 8.89 (d, 1H), 7.60 (d, 1H), 7.59 (s, 1H), 7.10 (m, 3H), 6.98 (m, 2H), 4.61 (m, 1H), 3.61 (m, 2H), 3.10 (m, 2H), 1.90 (d, 3H). MS m/z 417.2 (M+H)+.
  • Example 54 3-phenethyl-2-(thiazol-4-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00115
  • 1H NMR (400 MHz, CDCl3) δ 8.99 (s, 1H), 8.89 (d, 1H), 8.01 (s, 1H), 7.70 (d, 1H), 7.22 (m, 5H), 4.60 (m, 2H), 3.15 (m, 2H), 3.24 (m, 1H).
  • Example 55 3-(2-methoxyphenethyl)-2-(2-hydroxyphenyl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00116
  • 1H NMR (400 MHz, CDCl3) δ 8.82 (d, 1H), 7.65 (d, 1H), 7.40 (m, 1H), 7.20 (m, 2H), 6.90 (m, 2H), 6.79 (d, 1H), 6.65 (m, 2H), 4.60 (t, 2H), 3.40 (s, 3H), 3.01 (t, 2H). MS m/z 442.3 (M+H)+.
  • Example 56 3-(2-fluorophenethyl)-2-(thiazol-4-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00117
  • 1H NMR (400 MHz, CDCl3) δ 8.99 (s, 1H), 8.89 (d, 1H), 7.90 (s, 1H), 7.79 (d, 1H), 7.22 (m, 5H), 4.68 (m, 2H), 3.15 (m, 2H). MS m/z 421.3 (M+H)+.
  • Example 57 (R)-2-cyclopentyl-3-(1-phenylpropan-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00118
  • 1H NMR (400 MHz, CDCl3) δ 8.79 (d, 1H), 7.59 (d, 1H), 7.2 (m, 5H), 4.67 (brd, 1H), 3.69 (m, 1H), 3.30 (m, 1H), 3.0 (brm, 1H), 1.90 (m, 8H), 1.89 (d, 3H). MS m/z 402.3 (M+H)+.
  • Example 58 2-isopropyl-3-phenethyl-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00119
  • 1H NMR (400 MHz, CDCl3) δ 8.80 (d, 1H), 8.65 (d, 1H), 7.35 (m, 5H), 4.42 (m, 2H), 3.13 (m, 2H), 2.99 (m, 1H), 1.90 (d, 6H). MS m/z 362.3 (M+H)+.
  • Example 59 2-cyclopentyl-3-phenethyl-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00120
  • 1H NMR (400 MHz, CDCl3) δ 8.79 (d, 1H), 8.65 (d, 1H), 7.35 (m, 5H), 4.40 (m, 2H), 3.10 (m, 2H), 2.00-1.6 (m, 8H). MS m/z 388.3 (M+H)+.
  • Example 60 3-(2-methoxyphenethyl)-2-(thiazol-4-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00121
  • 1H NMR (400 MHz, CDCl3) δ 8.89 (t, 2H), 8.70 (d, 1H), 7.45 (s, 1H), 7.10 (m, 1H), 6.79 (m, 1H), 6.70 (m, 1H), 6.60 (d, 1H), 4.80 (m, 2H), 3.40 (s, 3H), 3.0 (t, 2H), MS m/z 433.4 (M+H)+.
  • Example 61 3-(2-cyclohexylethyl)-2-(thiazol-4-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00122
  • 1H NMR (500 MHz, CDCl3) □ ppm 0.78-0.95 (m, 2H), 1.07-1.25 (m, 3H), 1.24-1.34 (m, 1H), 1.56-1.74 (m, 7H), 4.35-4.52 (m, 2H), 7.75 (d, 1H), 8.22 (d, 1H), 8.86 (d, 1H), 8.98 (d, 1H). MS m/z 409.5 (M+H)+.
  • Example 62 2-(3-(2-cyclohexylethyl)-4-oxo-5-(trifluoromethyl)-3,4-dihydropyrido[4,3-d]pyrimidin-2-yl)benzonitrile
  • Figure US20110028452A1-20110203-C00123
  • 1H NMR (500 MHz, CDCl3) □ (ppm) 0.67-0.80 (m, 2H), 1.03-1.20 (m, 4H), 1.44 (d, 2H), 1.50-1.57 (m, 2H), 1.57-1.65 (m, 3H), 3.87-4.02 (m, 2H), 7.64 (d, 1H), 7.70-7.81 (m, 2H), 7.85 (t, 1H), 7.93 (d, 1H), 8.91 (d, 1H). MS m/z 427.5 (M+H)+.
  • Example 63 2-(3-aminopyridin-2-yl)-3-phenethyl-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00124
  • 1H NMR (400 MHz, CDCl3) δ 8.84 (d, 1H), 8.17 (q, 1H), 7.69 (d, 1H), 7.27 (m, 1H), 7.18 (m, 4H), 7.06 (m, 2H), 4.68 (s, br, 2H), 4.38 (m, 2H), 3.11 (m, 2H). MS m/z 412.4 (M+H)+.
  • Example 64 N-(2-(4-oxo-3-phenethyl-5-(trifluoromethyl)-3,4-dihydropyrido[4,3-d]pyrimidin-2-yl)pyridin-3-yl)methanesulfonamide
  • Figure US20110028452A1-20110203-C00125
  • 1H NMR (400 MHz, CDCl3) δ 8.94 (s, 1H), 8.90 (d, 1H), 8.55 (dd, 1H), 8.12 (dd, 1H), 7.70 (d, 1H), 7.52 (dd, 1H), 7.22 (m, 3H), 7.07 (m, 2H), 4.44 (m, 2H), 3.19 (m, 2H), 3.11 (s, 3H). MS m/z 490.5 (M+H)+.
  • Example 65 3-(1-(2-fluorophenyl)propan-2-yl)-2-(thiazol-4-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00126
  • 1H NMR (400 MHz, CDCl3) δ 8.99 (s, 1H), 8.89 (d, 1H), 7.65 (d, 1H), 7.59 (s, 1H), 7.18 (m, 1H), 6.99 (m, 1H), 6.86 (m, 2H), 4.88 (m, 1H), 3.60 (m, 1H), 3.05 (m, 1H), 1.90 (d, 3H).
  • MS m/z 435.2 (M+H)+.
  • Example 66 2-(3-hydroxypyridin-2-yl)-3-isopentyl-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00127
  • 1H NMR (400 MHz, dmso-d6) δ 10.63 (s, 1H), 8.87 (d, 1H), 8.16 (m, 1H), 7.86 (d, 1H), 7.42 (m, 2H), 3.79 (t, 2H), 1.40-1.28 (m, 3H), 0.58 (d, 6H). MS m/z 379.2 (M+H)+.
  • Example 67 2-(3-hydroxypyridin-2-yl)-3-((tetrahydro-2H-pyran-4-yl)methyl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00128
  • 1H NMR (400 MHz, dmso-d6) δ 10.66 (s, 1H), 8.87 (d, 1H), 8.16 (dd, 1H), 7.86 (d, 1H), 7.42 (m, 2H), 3.79 (d, 2H), 3.64 (dd, 2H), 3.03 (dt, 2H), 1.76 (m, 1H), 1.24 (d, 2H), 0.94 (dq, 2H). MS m/z 407.3 (M+H)+.
  • Example 68 2-(3-hydroxypyridin-2-yl)-3-(2-(tetrahydro-2H-pyran-4-yl)ethyl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00129
  • 1H NMR (400 MHz, dmso-d6) δ 10.68 (s, 1H), 8.91 (d, 1H), 8.19 (dd, 1H), 7.80 (d, 1H), 7.47 (m, 2H), 3.86 (t, 2H), 3.66 (dd, 2H), 3.12 (t, 2H), 1.46 (q, 2H), 1.26 (m, 3H), 0.86 (dq, 2H). MS m/z 421.2 (M+H)+.
  • Example 69 3-(2-fluoro-5-methoxyphenethyl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00130
  • MS m/z 461 (M+H)+.
  • Example 70 3-(2-fluoro-3-methoxyphenethyl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00131
  • 1H NMR (400 MHz, CD3OD) δ 8.90 (d, 1H), 8.20 (d, 1H), 7.89 (d, 1H), 7.56 (m, 2H), 6.90 (t, 1H), 6.76 (m, 1H), 6.56 (d, 1H), 4.30 (m, 2H), 3.64 (s, 3H), 2.99 (m, 2H). MS m/z 461.2 (M+H)+.
  • Example 71 2-(3-(2-fluorophenethyl)-4-oxo-5-(trifluoromethyl)-3,4-dihydroquinazolin-2-yl)benzonitrile
  • Figure US20110028452A1-20110203-C00132
  • 1H NMR (400 MHz, CDCl3) δ 7.68 (t, 1H), 7.64 (t, 1H), 7.25 (m, 6H), 7.14 (m, 1H), 7.01 (t, 1H), 6.96 (t, 1H), 4.32 (t, 2H), 3.06 (t, 2H). MS m/z 438.0 (M+H)+.
  • Example 72 N-(2-(3-(2-fluorophenethyl)-4-oxo-5-(trifluoromethyl)-3,4-dihydropyrido[4,3-d]pyrimidin-2-yl)pyridin-3-yl)-2,2,2-trifluoroacetamide
  • Figure US20110028452A1-20110203-C00133
  • 1H NMR (400 MHz, CDCl3) δ 11.45 (s, 1H), 9.99 (d, 1H), 8.79 (d, 1H), 8.55 (d, 1H), 7.76 (d, 1H), 7.50 (m, 1H), 7.15 (m, 1H), 6.90 (m, 1H), 6.70 (m, 1H), 4.99 (m, 2H), 3.05 (m, 2H). MS m/z 526.3 (M+H)+.
  • Example 73 (R)-2,2,2-trifluoro-N-(2-(4-oxo-3-(1-phenylpropan-2-yl)-5-(trifluoromethyl)-3,4-dihydropyrido[4,3-d]pyrimidin-2-yl)pyridin-3-yl)acetamide
  • Figure US20110028452A1-20110203-C00134
  • 1H NMR (400 MHz, CDCl3) δ 10.65 (s, 1H), 9.99 (d, 1H), 8.79 (d, 1H), 8.60 (d, 1H), 7.66 (m, 1H), 7.44 (d, 1H), 7.00 (m, 2H), 6.79 (m, 2H), 4.69 (m, 1H), 3.70 (m, 1H), 2.95 (m, 1H), 2.01 (d, 3H). MS m/z 520.3 (M+H)+.
  • Example 74 3-cyclohexyl-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00135
  • 1H NMR (400 MHz, CDCl3) δ 8.99 (d, 1H), 8.25 (d, 1H), 7.77 (d, 1H), 7.43 (m, 2H), 4.80 (m, 1H), 2.65 (m, 2H),) 1.50-1.25 (m, 8H). MS m/z 391.3 (M+H)+.
  • Example 75 3-(cyclohexylmethyl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00136
  • 1H NMR (400 MHz, CDCl3) δ 12.39 (brs, 1H), 8.99 (d, 1H), 8.30 (d, 1H), 7.70 (d, 1H), 7.43 (m, 2H), 5.0 (brs, 2H), 1.50-1.25 (m, 10H). MS m/z 405.4 (M+H)+.
  • Example 76 3-Phenethyl-5-trifluoromethyl-2-(3-trifluoromethyl-pyridin-2-yl)-3H-pyrido[4,3-d]pyrimidin-4-one
  • Figure US20110028452A1-20110203-C00137
  • 1H NMR (400 MHz, CD3OD)□ 9.02 (d, 1H); 8.91-8.92 (d, 1H); 8.25-8.27 (d, 1H); 7.78-7.79, (m, 2H); 7.22-7.23 (m, 3H); 7.0-7.02 (d, 2H); 3.95 (br, 2H), 3.10-3.14 (t, 2H). MS m/z 465.5 (M+H)+.
  • Example 77
  • Figure US20110028452A1-20110203-C00138
  • 1H NMR (400 MHz, CD3OD) □ 8.88 (d, 1H); 8.87 (s, 1H); 8.22-8.24 (d, 1H); 7.73, 7.72, 7.69 (t, 2H); 7.20, 7.19 (d, 3H); 6.98, 6.96 (d, 2H); 3.88 (m, 1H); 3.64 (br, 1H), 3.36 (br, 1H); 1.59 (s, 3H); 1.55 (br, 1H). LC/MS 479.5 (M+H)+.
  • Example 78 2-(5-aminothiazol-4-yl)-3-phenethyl-5-(trifluoromethyl)pyrido[4,3-d]pyridimin-4(3H)-one
  • Figure US20110028452A1-20110203-C00139
  • To a solution of 2-(5-(2,5-dimethyl-1H-pyrrol-1-yl)thiazol-4-yl)-3-phenethyl-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one (7 mg, 0.014 mmol) in ethanol was added hydroxylamine hydrochloride (9.82 mg, 0.14 mmol) and the mixture was heated up to 80° C. for 60 hours. The reaction was then concentrated and purified with flash chromatography providing an off-white solid (1.5 mg). 1H NMR (400 MHz, CDCl3) δ 8.72 (d, 1H), 8.02 (s, 1H), 7.52 (d, 1H), 7.30 (m, 5H), 6.93 (s, br, 2H), 4.89 (t, 2H), 3.17 (t, 2H). MS m/z 418.3 (M+H)+.
  • Example 79 2-(2-(difluoromethyl)phenyl)-3-phenethyl-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00140
  • Prepared analogous to Example 1 starting from 2-(difluoromethyl)benzaldehyde and 4-Amino-N-phenethyl-2-(trifluoromethyl)nicotinamide. 1H NMR (400 MHz, CDCl3) δ 8.88 (d, 1H), 7.77 (d, 1H), 7.72 (d, 1H), 7.69 (t, 1H), 7.61 (t, 1H), 7.19 (m, 4H), 6.89 (m, 2H), 6.77 (t, 1H), 4.28 (m, 1H), 3.78 (m, 1H), 2.95 (m, 2H). MS m/z 446.4 (M+H)+.
  • Example 80 2-(3-(difluoromethyl)pyridin-2-yl)-3-phenethyl-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00141
  • Prepared analogous to Example 1 starting from 2-(difluoromethyl)picolinaldehyde and 4-Amino-N-phenethyl-2-(trifluoromethyl)nicotinamide. 1H NMR (400 MHz, CDCl3) δ 8.88 (d, 1H), 8.19 (d, 1H), 7.72 (d, 1H), 7.67 (m, 1H), 7.19 (m, 4H), 7.02 (m, 2H), 4.13 (m, 2H), 3.12 (m, 2H). MS m/z 447.5 (M+H)+.
  • Example 81 3-(2-cyclohexylethyl)-5-(trifluoromethyl)-2-(2-(trifluoromethyl)phenyl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00142
  • 1H NMR (500 MHz, CDCl3) □ ppm 0.55-0.71 (m, 1H) 0.73-0.86 (m, 1H) 1.03-1.19 (m, 3H) 1.24-1.41 (m, 2H) 1.46-1.67 (m, 5H) 1.68-1.83 (m, 1H) 3.38-3.52 (m, 1H) 4.07-4.25 (m, 1H) 7.54 (d, 1H) 7.69-7.84 (m, 3H) 7.90 (d, 1H) 8.89 (d, 1H). MS m/z 470.5 (M+H)+.
  • Example 82 3-(2-cyclohexylethyl)-5-(trifluoromethyl)-2-(3-(trifluoromethyl)pyridin-2-yl)pyrido[4,3-d]pyrimidin-4(3H)-one
  • Figure US20110028452A1-20110203-C00143
  • 1H NMR (500 MHz, CDCl3) □ ppm 0.66-0.82 (m, 2H), 1.02-1.20 (m, 4H), 1.46 (d, 2H), 1.54-1.71 (m, 5H), 3.86 (br. s., 2H), 7.72 (dd, 1H), 7.76 (d, 1H), 8.25 (d, 1H), 8.89 (d, 1H), 8.96 (d, 1H). MS m/z 471.5 (M+H)+.
  • The following table provides FLIPR IC50 data for the specified Examples. The IC50s are reported as micromolar concentration with n being the number of times the particular compound was assayed.
  • TABLE of FLIPR assay data
    FLIPR IC50
    Example n= (uM)
     1 6 0.05
     2 6 0.62
     3 4 0.39
     4a 11 0.06
     4b 3 0.16
     5 4 0.02
     6 4 0.05
     6a 5 0.05
     6b 3 1.40
     7 6 0.13
     8 5 0.09
     9 6 0.29
    10 6 0.12
    11 5 0.46
    12 12 0.25
    13 8 0.07
    14 8 0.17
    15 4 0.16
    16 3 10.8
    17 2 11.0
    18 5 12.3
    19 1 >100
    20 1 >100
    21 2 13.8
    22 1 >100
    23 3 13.7
    24 1 2.60
    25 1 3.01
    26 1 2.99
    27 1 2.26
    28 1 4.83
    29 4 0.49
    30 6 1.17
    31 3 1.93
    32 5 0.57
    33 5 0.52
    34 3 1.47
    35 6 0.38
    36 2 0.60
    37a 11 .008
    37b 3 2.12
    38a 3 0.02
    38b 3 4.69
    39 8 0.11
    40 3 0.46
    41 7 0.25
    42 3 1.12
    43 2 1.12
    43a 2 0.53
    43b 4 61.3
    44 4 2.44
    45 2 1.87
    46 3 0.29
    47 6 0.06
    48a 2 0.16
    48b 2 1.69
    49 2 0.48
    50 8 0.03
    51 4 0.11
    52 2 0.23
    53 6 0.26
    54 4 0.71
    55 5 0.72
    56 6 1.54
    57 2 1.67
    58 4 4.68
    59 4 7.79
    60 4 23.2
    61 2 0.50
    62 2 4.63
    63 2 0.77
    64 2 61.8
    65 4 10.5
    65a 4 0.32
    66 2 36.2
    67 1 >1.00
    68 1 >1.00
    69 2 66.9
    70 2 61.9
    71 4 0.42
    72 4 0.94
    73 4 0.20
    74 2 47.8
    75 2 10000
    76 3 6.30
    77 7 0.70
    78 2 9.83
    79 3 0.39
    80 2 0.30
    81 2 7.89
    82 2 2.53
  • All documents cited in this application, including scientific publications, patents and patent applications, are hereby incorporated by reference in their entirety.

Claims (23)

1. A compound of formula I
Figure US20110028452A1-20110203-C00144
wherein
R1 is -Q or (C1-C6)alkyl-Q;
R2 is (C1-C6)alkyl, (C3-C7)cycloalkyl, aryl or heteroaryl; wherein said aryl or heteroaryl is substituted with hydroxy and additionally said (C1-C6)alkyl, (C3-C7)cycloalkyl, aryl or heteroaryl within the definition of R2 is optionally substituted with one to three substituents independently selected from halo, cyano, trifluoromethyl, trifluoromethoxy, (C1-C6)alkyl, (C1-C6)alkoxy or hydroxy;
R3 is hydrogen, (C1-C6)alkyl, (C3-C7)cycloalkyl, (C1-C6)alkyl-Q, aryl, heteroaryl, OR6, or NR7R8; wherein said (C1-C6)alkyl, (C3-C7)cycloalkyl, aryl or heteroaryl is optionally substituted with one to three substituents independently selected from halo, cyano, trifluoromethyl, trifluoromethoxy, (C1-C6)alkyl; (C1-C6)alkoxy, NR7R8 or hydroxy;
R4 and R5 are each independently hydrogen, halo, cyano, (C1-C6)alkyl optionally substituted with one to three fluoro, aryl, heteroaryl, or OR6;
R6 at each occurrence is independently (C1-C6)alkyl, (C3-C7)cycloalkyl, (C3-C7)cycloalkyl-(C1-C6)alkyl, aryl, heteroaryl, (C1-C6)alkylaryl, or (C1-C6)alkylheteroaryl; each of said (C1-C6)alkyl, (C3-C7)cycloalkyl, (C3-C7)cycloalkyl-(C1-C6)alkyl, aryl, heteroaryl, (C1-C6)alkylaryl, or (C1-C6)alkylheteroaryl optionally substituted with one to three substituents independently selected from halo, hydroxy or (C1-C3)alkyl;
R7 and R8, at each occurrence, are independently hydrogen, (C1-C6)alkyl, or (C3-C7)cycloalkyl; or R7 and R8 taken together with the nitrogen to which they are attached form a 3 to 7 membered fully saturated, partially saturated or fully unsaturated ring optionally containing one to two additional heteroatoms independently selected from N(R9)n; O or S(O)p;
n is 0 or 1;
p is 0, 1 or 2;
R9 is hydrogen or (C1-C6)alkyl;
Q, at each occurrence, is independently aryl or heteroaryl; wherein said aryl or heteroaryl is optionally substituted with one to three substituents independently selected from halo, cyano, trifluoromethyl, trifluoromethoxy, (C1-C6)alkyl, (C1-C6)alkoxy or hydroxy;
or a pharmaceutically acceptable salt thereof.
2. A compound according to claim 1, wherein R2 is aryl or heteroaryl, wherein said aryl or heteroaryl is substituted with hydroxy and optionally substituted with one to three substituents independently selected from (C1-C6)alkoxy or halo; or a pharmaceutically acceptable salt thereof.
3. A compound according to claim 2, wherein R2 is aryl, wherein said aryl is substituted with hydroxy and optionally substituted with one to three substituents independently selected from (C1-C6)alkoxy or halo; or a pharmaceutically acceptable salt thereof.
4. A compound according to claim 1, wherein R2 is heteroaryl, wherein said heteroaryl is substituted with hydroxy and optionally substituted with one to three substituents independently selected from (C1-C6)alkoxy or halo; or a pharmaceutically acceptable salt thereof.
5. A compound according to claim 2, wherein R2 is 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl; wherein said 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl is optionally substituted with one to two substituents independently selected from (C1-C6)alkoxy or fluoro; or a pharmaceutically acceptable salt thereof.
6. A compound according to claim 5, wherein R1 is (C1-C6)alkyl-Q; and Q is phenyl optionally substituted with one or two fluoro; or a pharmaceutically acceptable salt thereof.
7. A compound according to claim 1, wherein R1 is phenethyl or 1-methyl-2-(phenyl)ethyl, wherein said phenethyl or 1-methyl-2-(phenyl)ethyl is optionally substituted on phenyl with one or two halo; R2 is 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl, wherein said 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl is optionally substituted with one or two substituents independently selected from (C1-C3)alkyl or halo; or a pharmaceutically acceptable salt thereof.
8. A compound according to claim 7 wherein R3 is (C1-C6)alkyl, (C1-C6)alkyl-Q, aryl, heteroaryl, OR6, or NR7R8, wherein said (C1-C6)alkyl, aryl or heteroaryl is optionally substituted with one to three substituents independently selected from halo, cyano, trifluoromethyl, trifluoromethoxy, (C1-C6)alkyl, (C1-C6)alkoxy, NR7R8 or hydroxy; and
R4 and R5 are each hydrogen; or a pharmaceutically acceptable salt thereof.
9. A compound according to claim 8, wherein R3 is trifluoromethyl; or a pharmaceutically acceptable salt thereof.
10. A compound according to claim 1, wherein R3 is trifluoromethyl; or a pharmaceutically acceptable salt thereof.
11. A compound according to claim 10, wherein R4 and R5 are each independently hydrogen or methyl; or a pharmaceutically acceptable salt thereof.
12. A compound according to claim 11, wherein R2 is aryl or heteroaryl, wherein said aryl or heteroaryl is substituted with hydroxy and optionally substituted with one to three substituents independently selected from (C1-C6)alkoxy or halo; or a pharmaceutically acceptable salt thereof.
13. A compound according to claim 12, wherein R2 is hydroxy-phenyl or hydroxy-pyridyl; wherein said hydroxy-phenyl or hydroxy-pyridyl is optionally substituted with one to two substituents independently selected from (C1-C6)alkoxy or fluoro; or a pharmaceutically acceptable salt thereof.
14. A compound according to claim 13, wherein R1 is (C1-C6)alkyl-Q; and Q is phenyl optionally substituted with one or two substituents independently selected from fluoro, methyl or methoxy; or a pharmaceutically acceptable salt thereof.
15. A compound according to claim 11, wherein R1 is phenethyl or 1-methyl-2-(phenyl)ethyl, wherein said phenethyl or 1-methyl-2-(phenyl)ethyl is optionally substituted on phenyl with one or two fluoro; or a pharmaceutically acceptable salt thereof.
16. A compound according to claim 11, wherein R2 is 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl, wherein said 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl is optionally substituted with one or two substituents independently selected from (C1-C3)alkyl or halo; or a pharmaceutically acceptable salt thereof.
17. A compound according to claim 1 wherein R1 is phenethyl or 1-methyl-2-(phenyl)ethyl, wherein said phenethyl or 1-methyl-2-(phenyl)ethyl is optionally substituted on phenyl with one or two substituents independently selected from halo or (C1-C3)alkyl or (C1-C3)alkoxy;
R2 is 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl, wherein said 2-hydroxy-phenyl or 3-hydroxy-pyridin-2-yl is optionally substituted with one or two substituents independently selected from (C1-C3)alkyl or halo;
R3 is (C1-C6)alkyl, (C3-C7)cycloalkyl, (C1-C6)alkyl-Q, aryl, heteroaryl, or OR6; wherein said (C1-C6)alkyl, (C3-C7)cycloalkyl, aryl or heteroaryl is optionally substituted with one to three substituents independently selected from fluoro, cyano, trifluoromethyl, trifluoromethoxy, (C1-C3)alkyl; (C1-C3)alkoxy, NR7R8 or hydroxy; and
R4 and R5 are each independently hydrogen or methyl; or a pharmaceutically acceptable salt thereof.
18. A compound selected from the group consisting of:
2-(2-Hydroxy-phenyl)-3-phenethyl-5-trifluoromethyl-3H-pyrido[4,3-d]pyri-midin-4-one;
2-(3-Fluoro-2-hydroxy-phenyl)-3-phenethyl-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
2-(3-Hydroxy-pyridin-2-yl)-3-phenethyl-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
(R)-2-(2-Hydroxy-phenyl)-3-(1-methyl-2-phenyl-ethyl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
(S)-2-(2-Hydroxy-phenyl)-3-(1-methyl-2-phenyl-ethyl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
(R)-2-(3-Hydroxy-pyridin-2-yl)-3-(1-methyl-2-phenyl-ethyl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
(R,S)-2-(3-Hydroxy-pyridin-2-yl)-3-(1-methyl-2-(2-fluorophenyl)ethyl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
(R)-2-(3-Hydroxy-pyridin-2-yl)-3-(1-methyl-2-(2-fluorophenyl)ethyl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
(S)-2-(3-Hydroxy-pyridin-2-yl)-3-(1-methyl-2-(2-fluorophenyl)ethyl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
2-(2-Hydroxyphenyl)-3-(2-fluorophenyl)ethyl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
2-(2-Hydroxyphenyl)-3-(3-fluorophenyl)ethyl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
2-(3-Fluoro-2-hydroxy-phenyl)-3-[2-(2-fluoro-phenyl)-ethyl]-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
2-(2-Hydroxy-phenyl)-7-methyl-3-(phenyl-ethyl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
2-(3-Fluoro-2-hydroxy-phenyl)-3-[2-(3-fluoro-phenyl)-ethyl]-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
3-[2-(2-Fluoro-phenyl)-ethyl]-2-(3-hydroxy-pyridin-2-yl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
3-[2-(3,4-Difluoro-phenyl)-ethyl]-2-(3-hydroxy-pyridin-2-yl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
3-[2-(2,4-Difluoro-phenyl)-ethyl]-2-(3-hydroxy-pyridin-2-yl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
3-[2-(3,4-Difluoro-phenyl)-ethyl]-2-(3-hydroxy-pyridin-2-yl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
2-(2-Hydroxy-phenyl)-5-methylamino-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
2-(2-Hydroxy-phenyl)-5-isopropylamino-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
2-(2-Hydroxy-phenyl)-3-phenethyl-5-pyrrolidin-1-yl-3H-pyrido[4,3-d]pyrimidin-4-one;
2-(2-Hydroxy-phenyl)-5-(4-methyl-piperazin-1-yl)-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
2-(2-Hydroxy-phenyl)-3-phenethyl-5-piperazin-1-yl-3H-pyrido[4,3-d]pyrimidin-4-one;
5-Dimethylamino-2-(2-hydroxy-phenyl)-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
2-(2-Hydroxy-phenyl)-5-morpholin-4-yl-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
5-Azetidin-1-yl-2-(2-hydroxy-phenyl)-3-phenethyl-3H-pyrido[4,3-d-pyrimidin-4-one;
2-(2-Hydroxy-phenyl)-3-phenethyl-5-phenyl-3H-pyrido[4,3-d]pyrimidin-4-one;
5-Benzyl-2-(2-hydroxy-phenyl)-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
2-(2-Hydroxy-phenyl)-5-methyl-3-phenethyl-3H-pyrido[4,3-d]pyrimidi-4-one;
5-(6-Dimethylamino-pyridin-3-yl)-2-(2-hydroxy-phenyl)-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
5-(6-Dimethylamino-5-methyl-pyridin-3-yl)-2-(2-hydroxy-phenyl)-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
5-(6-pyrrolidine-5-pyridin-3-yl)-2-(2-hydroxy-phenyl)-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
2-(2-Hydroxy-phenyl)-5-methoxy-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
2-(2-Hydroxy-phenyl)-5-(1-methyl-cyclopropylmethoxy)-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
2-(2-Hydroxy-phenyl)-3-phenethyl-5-propoxy-3H-pyrido[4,3-d]pyrimidin-4-one;
5-Cyclobutyloxy-2-(2-hydroxy-phenyl)-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
2-(2-Hydroxy-phenyl)-5-isobutoxy-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
2-(2-Hydroxy-phenyl)-5-isopropoxy-3-phenethyl-3H-pyrido[4,3-d]pyrimidin-4-one;
2-(2-Hydroxy-phenyl)-3-phenethyl-5-(2,2,2-trifluoro-ethoxy)-3H-pyrido[4,3-d]pyrimidin-4-one;
3-(1-(3,4-difluorophenyl)propan-2-yl)-2-(3-hydroxypyridin-2-yl) 5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
(R)-3-(1-(3,4-difluorophenyl)propan-2-yl)-2-(3-hydroxypyridin-2-yl) 5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
(S)-3-(1-(3,4-difluorophenyl)propan-2-yl)-2-(3-hydroxypyridin-2-yl) 5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
3-(1-(2,4-difluorophenyl)propan-2-yl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
3-(1-(2,4-difluorophenyl)propan-2-yl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
3-(2-methoxyphenethyl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
3-(2,3-difluorophenethyl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
3-(5-fluoro-2-methoxyphenethyl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
3-(2-fluoro-6-methoxyphenethyl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
2-(3-hydroxypyridin-2-yl)-3-(1-(2-methoxyphenyl)propan-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
2-(3-hydroxypyridin-2-yl)-3-(1-(2-methoxyphenyl)propan-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
2-(3-hydroxypyridin-2-yl)-3-(1-(2-methoxyphenyl)propan-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
3-(1-(2-fluorophenyl)butan-2-yl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
3-(3-fluoro-2-methoxyphenethyl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
3-(2-cyclopentylethyl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
3-(2-cyclohexylethyl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
(R)-3-(1-cyclohexylpropan-2-yl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
(S)-3-(1-cyclohexylpropan-2-yl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
(R,S)-2-(3-hydroxypyridin-2-yl)-3-(2-(tetrahydro-2H-pyran-2-yl)ethyl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
(R)-2-(3-fluoro-2-hydroxyphenyl)-3-(1-phenylpropan-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
(R)-2-(1H-imidazol-2-yl)-3-(1-phenylpropan-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
N-(2-(3-(2-fluorophenethyl)-4-oxo-5-(trifluoromethyl)-3,4-dihydropyrido[4,3-d]pyrimidin-2-yl)pyridin-3-yl)acetamide;
(R)-3-(1-phenylpropan-2-yl)-2-(thiazol-4-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
3-phenethyl-2-(thiazol-4-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
3-(2-methoxyphenethyl)-2-(2-hydroxyphenyl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
3-(2-fluorophenethyl)-2-(thiazol-4-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
(R)-2-cyclopentyl-3-(1-phenylpropan-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
2-isopropyl-3-phenethyl-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one
2-cyclopentyl-3-phenethyl-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
3-(2-methoxyphenethyl)-2-(thiazol-4-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
3-(2-cyclohexylethyl)-2-(thiazol-4-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
2-(3-(2-cyclohexylethyl)-4-oxo-5-(trifluoromethyl)-3,4-dihydropyrido[4,3-d]pyrimidin-2-yl)benzonitrile;
2-(3-aminopyridin-2-yl)-3-phenethyl-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
N-(2-(4-oxo-3-phenethyl-5-(trifluoromethyl)-3,4-dihydropyrido[4,3-d]pyrimidin-2-yl)pyridin-3-yl)methanesulfonamide;
3-(1-(2-fluorophenyl)propan-2-yl)-2-(thiazol-4-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
2-(3-hydroxypyridin-2-yl)-3-isopentyl-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
2-(3-hydroxypyridin-2-yl)-3-((tetrahydro-2H-pyran-4-yl)methyl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
2-(3-hydroxypyridin-2-yl)-3-(2-(tetrahydro-2H-pyran-4-yl)ethyl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
3-(2-fluoro-5-methoxyphenethyl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
3-(2-fluoro-3-methoxyphenethyl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
2-(3-(2-fluorophenethyl)-4-oxo-5-(trifluoromethyl)-3,4-dihydroquinazolin-2-yl)benzonitrile;
N-(2-(3-(2-fluorophenethyl)-4-oxo-5-(trifluoromethyl)-3,4-dihydropyrido[4,3-d]pyrimidin-2-yl)pyridin-3-yl)-2,2,2-trifluoroacetamide;
(R)-2,2,2-trifluoro-N-(2-(4-oxo-3-(1-phenylpropan-2-yl)-5-(trifluoromethyl)-3,4-dihydropyrido[4,3-d]pyrimidin-2-yl)pyridin-3-yl)acetamide;
3-cyclohexyl-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
3-(cyclohexylmethyl)-2-(3-hydroxypyridin-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
3-Phenethyl-5-trifluoromethyl-2-(3-trifluoromethyl-pyridin-2-yl)-3H-pyrido[4,3-d]pyrimidin-4-one;
2-(5-aminothiazol-4-yl)-3-phenethyl-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
2-(2-(difluoromethyl)phenyl)-3-phenethyl-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
2-(3-(difluoromethyl)pyridin-2-yl)-3-phenethyl-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one;
3-(2-cyclohexylethyl)-5-(trifluoromethyl)-2-(2-(trifluoromethyl)phenyl)pyrido[4,3-d]pyrimidin-4(3H)-one; and
3-(2-cyclohexylethyl)-5-(trifluoromethyl)-2-(3-(trifluoromethyl)pyridin-2-yl)pyrido[4,3-d]pyrimidin-4(3H)-one; or a pharmaceutically acceptable salt thereof.
19. A compound according to claim 1 which is (R)-2-(2-Hydroxy-phenyl)-3-(1-methyl-2-phenyl-ethyl)-5-trifluoromethyl-3H-pyrido[4,3-d]pyrimidin-4-one or a pharmaceutically acceptable salt thereof.
20. A compound according to claim 1 which is (R)-3-(1-(3,4-difluorophenyl) propan-2-yl)-2-(3-hydroxypyridin-2-yl) 5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one or a pharmaceutically acceptable salt thereof.
21. A pharmaceutical composition comprising a compound according to claim 1 and a pharmaceutically acceptable carrier, adjuvant or diluent.
22. A method of treating a disease or disorder selected from hypoparathyroidism, osteoporosis, osteopenia, periodontal disease, Paget's disease, bone fracture, osteoarthritis, rheumatoid arthritis and humoral hypercalcemia of malignancy, the method comprising administration to a patient in need of treatment thereof a therapeutically effective amount of a compound according to claim 1.
23. The method according to claim 22 wherein the disease or disorder is osteoporosis.
US12/894,525 2006-10-04 2010-09-30 PYRIDO[4,3-d]PYRIMIDIN-4(3H)-ONE DERIVATIVES AS CALCIUM RECEPTOR ANTAGONISTS Abandoned US20110028452A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/894,525 US20110028452A1 (en) 2006-10-04 2010-09-30 PYRIDO[4,3-d]PYRIMIDIN-4(3H)-ONE DERIVATIVES AS CALCIUM RECEPTOR ANTAGONISTS

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US82816206P 2006-10-04 2006-10-04
US96908307P 2007-08-30 2007-08-30
US11/867,255 US7829572B2 (en) 2006-10-04 2007-10-04 Pyrido[4,3-d]pyrimidin-4(3H)-one derivatives as calcium receptor antagonists
US12/894,525 US20110028452A1 (en) 2006-10-04 2010-09-30 PYRIDO[4,3-d]PYRIMIDIN-4(3H)-ONE DERIVATIVES AS CALCIUM RECEPTOR ANTAGONISTS

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/867,255 Division US7829572B2 (en) 2006-10-04 2007-10-04 Pyrido[4,3-d]pyrimidin-4(3H)-one derivatives as calcium receptor antagonists

Publications (1)

Publication Number Publication Date
US20110028452A1 true US20110028452A1 (en) 2011-02-03

Family

ID=39183043

Family Applications (2)

Application Number Title Priority Date Filing Date
US11/867,255 Expired - Fee Related US7829572B2 (en) 2006-10-04 2007-10-04 Pyrido[4,3-d]pyrimidin-4(3H)-one derivatives as calcium receptor antagonists
US12/894,525 Abandoned US20110028452A1 (en) 2006-10-04 2010-09-30 PYRIDO[4,3-d]PYRIMIDIN-4(3H)-ONE DERIVATIVES AS CALCIUM RECEPTOR ANTAGONISTS

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US11/867,255 Expired - Fee Related US7829572B2 (en) 2006-10-04 2007-10-04 Pyrido[4,3-d]pyrimidin-4(3H)-one derivatives as calcium receptor antagonists

Country Status (17)

Country Link
US (2) US7829572B2 (en)
EP (1) EP2079739A2 (en)
JP (1) JP2010505811A (en)
KR (1) KR101129868B1 (en)
CN (1) CN101547922B (en)
AR (1) AR063122A1 (en)
AU (1) AU2007303846B2 (en)
CA (1) CA2663436A1 (en)
CL (1) CL2007002864A1 (en)
HK (1) HK1137753A1 (en)
IL (1) IL197536A0 (en)
MX (1) MX2009003673A (en)
NO (1) NO20091328L (en)
PE (1) PE20081455A1 (en)
TW (1) TW200822929A (en)
UY (1) UY30620A1 (en)
WO (1) WO2008041118A2 (en)

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012120476A1 (en) 2011-03-10 2012-09-13 Lupin Limited Substituted morpholines as modulators for the calcium sensing receptor
WO2012127385A1 (en) 2011-03-18 2012-09-27 Lupin Limited Benzo [b] [1, 4] oxazin derivatives as calcium sensing receptor modulators
WO2013124828A1 (en) 2012-02-24 2013-08-29 Lupin Limited Substituted chroman compounds as calcium sensing receptor modulators
WO2013136288A1 (en) 2012-03-16 2013-09-19 Lupin Limited Substituted 3,4-dihydro-2h-benzo[b] [1,4]oxazine compounds as calcium sensing receptor modulators
WO2014033604A1 (en) 2012-08-27 2014-03-06 Lupin Limited Arylalkylamine compounds as calcium sensing receptor modulators
WO2015022631A1 (en) 2013-08-12 2015-02-19 Lupin Limited Substituted biphenyl compounds as calcium sensing receptor modulators
WO2015028938A1 (en) 2013-08-28 2015-03-05 Lupin Limited Substituted naphthalene compounds as calcium sensing receptor modulators
US9023858B2 (en) 2010-07-23 2015-05-05 Takeda Pharmaceutical Company Limited Substituted pyrido[2,3-d]pyrimidines as delta-5-desaturase inhibitors
WO2015162538A1 (en) 2014-04-21 2015-10-29 Lupin Limited Heterocyclic compounds as calcium sensing receptor modulators for the treatment of hyperparathyroidism, chronic renal failure and chronic kidney disease
US20160073452A1 (en) * 2013-05-28 2016-03-10 Illinois Tool Works Inc. Induction pre-heating and butt welding device for adjacent edges of at least one element to be welded
US20160105933A1 (en) * 2014-10-14 2016-04-14 Illinois Tool Works Inc. Reduced-distortion hybrid induction heating/welding assembly
WO2017037616A1 (en) 2015-08-31 2017-03-09 Lupin Limited Arylalkylamine compounds as calcium sensing receptor modulators
WO2021130779A1 (en) 2019-12-27 2021-07-01 Lupin Limited Pharmaceutical composition of casr modulators and methods and uses thereof
WO2021144814A1 (en) 2020-01-17 2021-07-22 Lupin Limited Methods, processes and intermediates for preparing chroman compounds

Families Citing this family (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8845672B2 (en) 2002-05-09 2014-09-30 Reshape Medical, Inc. Balloon system and methods for treating obesity
US20070100368A1 (en) 2005-10-31 2007-05-03 Quijano Rodolfo C Intragastric space filler
US8226602B2 (en) * 2007-03-30 2012-07-24 Reshape Medical, Inc. Intragastric balloon system and therapeutic processes and products
US8142469B2 (en) 2007-06-25 2012-03-27 Reshape Medical, Inc. Gastric space filler device, delivery system, and related methods
US9174031B2 (en) * 2009-03-13 2015-11-03 Reshape Medical, Inc. Device and method for deflation and removal of implantable and inflatable devices
US8683881B2 (en) * 2009-04-03 2014-04-01 Reshape Medical, Inc. Intragastric space fillers and methods of manufacturing including in vitro testing
CN101619063B (en) * 2009-06-02 2011-08-10 华中师范大学 3,7,8-polysubstituted pyridine-[4,3-d] pyrimidine derivant with antitumor activity and preparation
WO2011011629A2 (en) 2009-07-22 2011-01-27 Reshape Medical, Inc. Retrieval mechanisms for implantable medical devices
US9050174B2 (en) 2009-07-23 2015-06-09 Reshape Medical, Inc. Deflation and removal of implantable medical devices
WO2011011741A2 (en) 2009-07-23 2011-01-27 Reshape Medical, Inc. Inflation and deflation mechanisms for inflatable medical devices
EP2533845A4 (en) 2010-02-08 2016-04-06 Reshape Medical Inc Improved and enhanced aspiration processes and mechanisms for intragastric devices
EP2533846B1 (en) 2010-02-08 2018-08-22 ReShape Medical LLC Materials and methods for improved intragastric balloon devices
EP2539011A4 (en) 2010-02-25 2014-03-26 Reshape Medical Inc Improved and enhanced explant processes and mechanisms for intragastric devices
EP2555705A4 (en) 2010-04-06 2014-01-15 Reshape Medical Inc Inflation devices for intragastric devices with improved attachment and detachment and associated systems and methods
EP2413139B1 (en) 2010-07-30 2015-05-20 Universität Leipzig Method and compositions for the identification of agents that have a potential effect against chronic inflammatory diseases
LT3141542T (en) 2011-12-28 2020-09-25 Global Blood Therapeutics, Inc. Substituted benzaldehyde compounds and methods for their use in increasing tissue oxygenation
ES2790358T3 (en) 2011-12-28 2020-10-27 Global Blood Therapeutics Inc Substituted Heteroaryl Aldehyde Compounds and Methods for Their Use in Increasing Tissue Oxygenation
US10100043B2 (en) 2013-03-15 2018-10-16 Global Blood Therapeutics, Inc. Substituted aldehyde compounds and methods for their use in increasing tissue oxygenation
KR102293060B1 (en) 2013-03-15 2021-08-23 글로벌 블러드 테라퓨틱스, 인크. Compounds and uses thereof for the modulation of hemoglobin
KR20150132146A (en) 2013-03-15 2015-11-25 글로벌 블러드 테라퓨틱스, 인크. Compounds and uses thereof for the modulation of hemoglobin
US9802900B2 (en) 2013-03-15 2017-10-31 Global Blood Therapeutics, Inc. Bicyclic heteroaryl compounds and uses thereof for the modulation of hemoglobin
US9422279B2 (en) 2013-03-15 2016-08-23 Global Blood Therapeutics, Inc. Compounds and uses thereof for the modulation of hemoglobin
AP2015008721A0 (en) 2013-03-15 2015-09-30 Global Blood Therapeutics Inc Compounds and uses thereof for the modulation of hemoglobin
US9458139B2 (en) 2013-03-15 2016-10-04 Global Blood Therapeutics, Inc. Compounds and uses thereof for the modulation of hemoglobin
US8952171B2 (en) 2013-03-15 2015-02-10 Global Blood Therapeutics, Inc. Compounds and uses thereof for the modulation of hemoglobin
US20140274961A1 (en) 2013-03-15 2014-09-18 Global Blood Therapeutics, Inc. Compounds and uses thereof for the modulation of hemoglobin
US9604999B2 (en) 2013-03-15 2017-03-28 Global Blood Therapeutics, Inc. Compounds and uses thereof for the modulation of hemoglobin
US10266551B2 (en) 2013-03-15 2019-04-23 Global Blood Therapeutics, Inc. Compounds and uses thereof for the modulation of hemoglobin
EA201992707A1 (en) 2013-11-18 2020-06-30 Глобал Блад Терапьютикс, Инк. COMPOUNDS AND THEIR APPLICATIONS FOR HEMOGLOBIN MODULATION
CN114181194A (en) 2014-02-07 2022-03-15 全球血液疗法股份有限公司 Crystalline polymorph of a compound
MA41841A (en) 2015-03-30 2018-02-06 Global Blood Therapeutics Inc ALDEHYDE COMPOUNDS FOR THE TREATMENT OF PULMONARY FIBROSIS, HYPOXIA, AND AUTOIMMUNE AND CONNECTIVE TISSUE DISEASES
EP3383392A1 (en) 2015-12-04 2018-10-10 Global Blood Therapeutics, Inc. Dosing regimens for 2-hydroxy-6-((2-(1-isopropyl-1h-pyrazol-5-yl)pyridin-3-yl)methoxy)benzaldehyde
TWI752307B (en) 2016-05-12 2022-01-11 美商全球血液治療公司 Novel compound and method of preparing compound
TW202332423A (en) 2016-10-12 2023-08-16 美商全球血液治療公司 Tablets comprising 2-hydroxy-6-((2-(1-isopropyl-1h-pyrazol-5-yl)pyridin-3-yl)methoxy)benzaldehyde
CN108164455A (en) * 2017-03-23 2018-06-15 广东众生药业股份有限公司 The synthetic method of pheniramine maleate
CN107903209B (en) * 2017-12-29 2021-05-14 宁夏忠同生物科技有限公司 Synthetic method of 2-amino-5-fluoropyridine-3-methyl formate
EP3860975B1 (en) 2018-10-01 2023-10-18 Global Blood Therapeutics, Inc. Modulators of hemoglobin for the treatment of sickle cell disease
KR102338609B1 (en) * 2019-05-20 2021-12-14 보령제약 주식회사 Pyrido-pyrimidin compound and pharmaceutical composition for use in preventing or treating PI3 kinase related diseases
CN113956268B (en) * 2021-11-19 2022-07-15 西安欧得光电材料有限公司 6-bromo-1-chlorobenzothiophene [2,3-c ] pyridine and synthetic method

Citations (99)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US646572A (en) * 1899-03-15 1900-04-03 Archibald C Floyd Bath-cabinet.
US4140789A (en) * 1976-01-08 1979-02-20 Ciba-Geigy Corporation Etherified hydroxy-benzodiheterocyclic compounds
US4379788A (en) * 1980-12-12 1983-04-12 Dr. Karl Thomae Gesellschaft Mit Beschrankter Haftung 2-Phenyl-pyrimidones
US4431440A (en) * 1981-02-20 1984-02-14 American Cyanamid Company Method to alter or control the development and/or the life cycle of various plant species
US4588812A (en) * 1984-05-30 1986-05-13 Pennwalt Corporation Substituted 2,3-dihydro-5H-thiazolo[2,3,-b]quinazoline derivatives
US4731106A (en) * 1984-11-24 1988-03-15 Schering Agrochemicals Limited Fungicidal azole compounds
US4994495A (en) * 1988-05-13 1991-02-19 Imperial Chemical Industries Plc Fungicides
US4999381A (en) * 1989-02-02 1991-03-12 Imperial Chemical Industries Plc Fungicides
US5100886A (en) * 1989-12-18 1992-03-31 Imperial Chemical Industries Plc Metal complexes of pyridyl cyclopropane carboxamide compounds which are useful as fungicides
US5110471A (en) * 1990-08-30 1992-05-05 Conoco Specialty Products Inc. High efficiency liquid/liquid hydrocyclone
US5124329A (en) * 1989-06-28 1992-06-23 Imperial Chemical Industries Plc Fungicides
US5126338A (en) * 1990-01-04 1992-06-30 Imperial Chemical Industries Plc Fungicides which are n-pyridyl-cyclopropane carboxamides or derivatives thereof
US5185339A (en) * 1990-05-30 1993-02-09 Imperial Chemical Industries Plc Fungicidal compounds
US5187779A (en) * 1989-08-11 1993-02-16 Micral, Inc. Memory controller with synchronous processor bus and asynchronous i/o bus interfaces
US5206245A (en) * 1990-07-27 1993-04-27 Imperial Chemical Industries Plc Fungicidal aromatic pyrimidinyl oxime ethers
US5225318A (en) * 1990-07-02 1993-07-06 Ocg Microelectronic Materials, Inc. Selected photoactive methylolated cyclohexanol compounds and their use in forming positive resist image patterns
US5290780A (en) * 1991-01-30 1994-03-01 American Cyanamid Co. Angiotensin II receptor blocking 2,3,6 substituted quinazolinones
US5304565A (en) * 1990-03-07 1994-04-19 Takeda Chemical Industries, Ltd. Nitrogen containing heterocyclic compounds, their production and use
US5401745A (en) * 1993-03-19 1995-03-28 Merck & Co., Inc. Quinazolinones substituted with phenoxyphenylacetic acid derivatives
US5409930A (en) * 1991-05-10 1995-04-25 Rhone-Poulenc Rorer Pharmaceuticals Inc. Bis mono- and bicyclic aryl and heteroaryl compounds which inhibit EGF and/or PDGF receptor tyrosine kinase
US5420133A (en) * 1993-03-19 1995-05-30 Merck & Co., Inc. Quinazolinones substituted with phenoxyphenylacetic acid derivatives
US5482941A (en) * 1991-12-11 1996-01-09 Pfizer Inc. Quinazolinone antianginal agents
US5710346A (en) * 1993-10-22 1998-01-20 Ihara Chemical Industry Co., Ltd. 2,2-bis(3,5-disubstituted-4-hydroxyphenyl) propane derivative, process for production thereof, and process for producing pyrogallol using said derivative
US5719098A (en) * 1992-12-21 1998-02-17 Sud-Chemie A.G. Sorbent based on smectitic clay minerals reacted with alkaline ion exchanger
US5721236A (en) * 1993-10-15 1998-02-24 Schering Corporation Tricyclic carbamate compounds useful for inhibition of G-protein function and for treatment of proliferative diseases
US5753851A (en) * 1997-06-16 1998-05-19 The United States Of America As Represented By The Secretary Of The Army Spinning mine with concentrated projectiles
US5756502A (en) * 1994-08-08 1998-05-26 Warner-Lambert Company Quinazolinone derivatives as cholyecystokinin (CCK) ligands
US6011068A (en) * 1991-08-23 2000-01-04 Nps Pharmaceuticals, Inc. Calcium receptor-active molecules
US6031003A (en) * 1991-08-23 2000-02-29 Nps Pharmaceuticals, Inc. Calcium receptor-active molecules
US6060479A (en) * 1997-06-09 2000-05-09 Pfizer Inc Quinazoline-4-one AMPA antagonists
US6211236B1 (en) * 1996-04-26 2001-04-03 Basf Aktiengesellschaft Fungicide mixtures
US6211244B1 (en) * 1994-10-21 2001-04-03 Nps Pharmaceuticals, Inc. Calcium receptor-active compounds
US6225315B1 (en) * 1998-11-30 2001-05-01 Pfizer Inc Method of treating nitrate-induced tolerance
US6235730B1 (en) * 1997-12-12 2001-05-22 Japan Tobacco, Inc. 3-piperidyl-4-oxoquinazoline derivatives and pharmaceutical compositions comprising the same
US6239130B1 (en) * 1997-04-30 2001-05-29 Warner-Lambert Company Phosphodiesterase 4-inhibiting diazepinoindolones
US6245792B1 (en) * 1996-04-26 2001-06-12 Basf Aktiengesellschaft Fungicidal mixture
US6255352B1 (en) * 1997-05-28 2001-07-03 Basf Aktiengesellschaft Method for combating harmful fungi
US6337332B1 (en) * 1998-09-17 2002-01-08 Pfizer Inc. Neuropeptide Y receptor antagonists
US6350785B2 (en) * 1999-01-08 2002-02-26 Maxim Pharmaceuticals, Inc. Methods and compositions for topical treatment of damaged tissue using reactive oxygen metabolite production or release inhibitors
US6350765B1 (en) * 1997-12-18 2002-02-26 Basf Aktiengesellschaft Fungicidal mixtures based on amide compounds and azoles
US6369090B1 (en) * 1997-06-04 2002-04-09 Basf Aktiengesellschaft Fungicidal mixture
US6380204B1 (en) * 1997-02-28 2002-04-30 Pfizer Inc Atropisomers of 3-heteroaryl-4(3H)-quinazolinones for the treatment of neurodegenerative and CNS-trauma related conditions
US6391874B1 (en) * 1996-07-13 2002-05-21 Smithkline Beecham Corporation Fused heterocyclic compounds as protein tyrosine kinase inhibitors
US20020094326A1 (en) * 2000-10-13 2002-07-18 Johns Hopkins University Methods and compositions for nucleic acid delivery
US20030013733A1 (en) * 2000-09-22 2003-01-16 Richard Apodaca Octahydro-indolizine and quinolizine and hexahydro-pyrrolizine
US6518277B1 (en) * 2000-04-25 2003-02-11 Icos Corporation Inhibitors of human phosphatidylinositol 3-kinase delta
US20030060626A1 (en) * 1990-07-27 2003-03-27 Clough John Martin Fungicides
US6545004B1 (en) * 1999-10-27 2003-04-08 Cytokinetics, Inc. Methods and compositions utilizing quinazolinones
US20030069230A1 (en) * 2001-01-02 2003-04-10 Becker Cyrus Kephra Quinazolone derivatives as alpha 1A/B adrenergic receptor antagonists
US6559160B1 (en) * 1999-08-27 2003-05-06 Chemocentryx, Inc. Compounds and methods for modulating cxcr3 function
US6562830B1 (en) * 1999-11-09 2003-05-13 Cell Pathways, Inc. Method of treating a patient having precancerous lesions with phenyl quinazolinone derivatives
US20040006130A1 (en) * 2000-08-11 2004-01-08 Yuko Shinagawa Calcium receptor antagonist
US20040009980A1 (en) * 2000-10-25 2004-01-15 Pradip Bhatnagar Calcilytic compounds
US20040019049A1 (en) * 2002-05-23 2004-01-29 Chiron Corporation Substituted quinazolinone compounds
US20040048853A1 (en) * 2002-08-21 2004-03-11 Gustave Bergnes Compounds, compositions, and methods
US6713485B2 (en) * 1998-01-12 2004-03-30 Smithkline Beecham Corporation Heterocyclic compounds
US20040067969A1 (en) * 2002-02-15 2004-04-08 Gustave Bergnes Syntheses of quinazolinones
US20040077668A1 (en) * 2002-05-09 2004-04-22 Cytokinetics, Inc. Compounds, compositins, and methods
US20040102450A1 (en) * 1998-01-27 2004-05-27 Aventis Pharmaceuticals Inc. Substituted oxoazaheterocyclyl compounds
US20040106616A1 (en) * 2002-01-17 2004-06-03 Rajagopal Bakthavatchalam Substituted quinazolin-4-ylamine analogues
US20040110777A1 (en) * 2001-12-03 2004-06-10 Annis Gary David Quinazolinones and pyridinylpyrimidinones for controlling invertebrate pests
US20040132732A1 (en) * 2002-10-21 2004-07-08 Wei Han Quinazolinones and derivatives thereof as factor Xa inhibitors
US20040142958A1 (en) * 2002-12-13 2004-07-22 Neurogen Corporation Combination therapy for the treatment of pain
US20040142949A1 (en) * 2002-07-23 2004-07-22 Gustave Bergnes Compounds, compositions, and methods
US20050004177A1 (en) * 2003-07-02 2005-01-06 Warner-Lambert Company Llc Combination of an allosteric inhibitor of matrix metalloproteinase-13 and a ligand to an alpha-2-delta receptor
US20050038051A1 (en) * 2003-06-09 2005-02-17 Jodi Nunnari Novel molecules for regulating cell death
US20050043239A1 (en) * 2003-08-14 2005-02-24 Jason Douangpanya Methods of inhibiting immune responses stimulated by an endogenous factor
US20050054651A1 (en) * 2003-08-22 2005-03-10 Dendreon Corporation Compositions and methods for the treatment of disease associated with Trp-p8 expression
US20050059862A1 (en) * 2003-09-12 2005-03-17 Scimed Life Systems, Inc. Cannula with integrated imaging and optical capability
US20050059851A1 (en) * 2003-09-17 2005-03-17 Vincent Ardizzone Flexible magnetic pad with multi-directional constantly alternating polarity zones
US6890930B1 (en) * 1999-09-28 2005-05-10 3-Dimensional Pharmaceuticals, Inc. Quinazolinones
US6897213B1 (en) * 1995-11-21 2005-05-24 Warner-Lambert Company Heterocycles as cholecystokinin (CCK) ligands
US20050130954A1 (en) * 2003-11-21 2005-06-16 Mitchell Ian S. AKT protein kinase inhibitors
US20050136085A1 (en) * 2003-12-19 2005-06-23 David Bellamy Panthenol and natural organic extracts for reducing skin irritation
US20050137213A1 (en) * 2003-07-03 2005-06-23 Myriad Genetics, Incorporated Compounds and therapeutical use thereof
US20060009455A1 (en) * 2004-06-15 2006-01-12 Corte James R Six-membered heterocycles useful as serine protease inhibitors
US20060019974A1 (en) * 2000-09-20 2006-01-26 Werner Mederski 4-Amino-quinazolines
US20060017428A1 (en) * 2004-07-20 2006-01-26 Chin-Yuan Lin Electrical magnetic interference test system
US20060025420A1 (en) * 2004-07-30 2006-02-02 Boehringer Ingelheimn International GmbH Pharmaceutical compositions for the treatment of female sexual disorders
US20060052345A1 (en) * 2002-11-04 2006-03-09 Nps Pharmaceuticals, Inc. Quinazolinone compounds as calcilytics
US20060063707A1 (en) * 2004-09-17 2006-03-23 Lifelike Biomatic, Inc. Compositions for enhancing memory and methods therefor
US20060069099A1 (en) * 2004-06-28 2006-03-30 Zice Fu Compounds, compositions and methods for prevention and treatment of inflammatory and immunoregulatory disorders and diseases
US20060079538A1 (en) * 2004-05-13 2006-04-13 Dennis Hallahan Methods for inhibiting angiogenesis
US20060079685A1 (en) * 2002-12-23 2006-04-13 Eva Altmann Aryl-quinazoline/aryl-2amino-phenyl methanone derivatives
US20060094723A1 (en) * 2003-05-22 2006-05-04 Torsten Dunkern Composition comprising a pde4 inhibitor and a pde5 inhibitor
US20060108038A1 (en) * 2004-11-25 2006-05-25 Sumitomo Rubber Industries, Ltd. Low noise tire support ring
US20060107448A1 (en) * 2004-11-17 2006-05-25 Interspiro, Inc. Protective seal mechanism
US7060706B1 (en) * 1999-09-28 2006-06-13 3-Dimensional Pharmaceuticals, Inc. Quinazolinones
US20060148693A1 (en) * 2003-06-16 2006-07-06 Altana Pharma Ag Composition comprising a pulmonary surfactant and a pde5 inhibitor for the treatment of lung diseases
US20060154935A1 (en) * 2004-09-02 2006-07-13 Dean Wilson Quinazolines useful as modulators of ion channels
US7157476B2 (en) * 2003-08-20 2007-01-02 Vertex Pharmaceuticals Incorporated Aminofurazan compounds useful as protein kinase inhibitors
US20070032506A1 (en) * 2005-07-02 2007-02-08 Peter Giannousis Crystalline forms of (2r-trans)-6-chloro-5[[4-[(4-fluorophenyl)methyl]-2,5-dimethyl-1-piperazinyl]carbonyl]-n,n, 1-trimethyl-alpha-oxo-1h-indole-3-acetamide monohydrochloride
US20070060601A1 (en) * 2003-12-19 2007-03-15 Arrington Kenneth L Mitotic kinesin inhibitors
US20070082873A1 (en) * 2003-10-28 2007-04-12 Lingenhoehl Kurt Combinations comprising ampa receptor antagonists for the treatment of schizophrenia
US20070149548A1 (en) * 2005-12-22 2007-06-28 Alcon Manufacturing, Ltd. (indazol-5-yl)-pyrazines and (1,3-dihydro-indol-2-one)-pyrazines for treating rho kinase-mediated diseases and conditions
US20070149553A1 (en) * 2003-12-19 2007-06-28 Arrington Kenneth L Mitotic kinesin inhibitors
US20080154942A1 (en) * 2006-12-22 2008-06-26 Cheng-Fa Tsai Method for Grid-Based Data Clustering
US7488727B2 (en) * 2001-06-01 2009-02-10 Vertex Pharmaceuticals Incorporated Thiazole compounds useful as inhibitors of protein kinase
WO2010079431A2 (en) * 2009-01-08 2010-07-15 Resverlogix Corp. Compounds for the prevention and treatment of cardiovascular disease

Family Cites Families (102)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4343940A (en) 1979-02-13 1982-08-10 Mead Johnson & Company Anti-tumor quinazoline compounds
US4422711A (en) 1981-12-21 1983-12-27 Amp Incorporated Active pin contact
GB8513754D0 (en) 1985-05-31 1985-07-03 Jones T R Anti-cancer quinazoline derivatives
ES2118769T3 (en) 1988-11-21 1998-10-01 Zeneca Ltd FUNGICIDES.
GB8827988D0 (en) 1988-11-30 1989-01-05 Smith Kline French Lab Chemical compounds
US5145856A (en) 1989-02-10 1992-09-08 Imperial Chemical Industries Plc Fungicides
BE1003002A6 (en) 1989-03-21 1991-10-22 Coppe Jan Formulation to be applied for the treatment of baldness and methods makinguse thereof
US5240928A (en) 1989-07-03 1993-08-31 Merck & Co., Inc. Substituted quinazolinones as angiotensin II antagonists
GB9016783D0 (en) 1989-09-01 1990-09-12 Ici Plc Agrochemical compositions
HU206680B (en) 1989-12-22 1992-12-28 Ici Plc Fungicide compositions containing pyridyl-cyclopropane-amides as active components and process for producing the active components
US5264439A (en) 1990-02-13 1993-11-23 Merck & Co., Inc. Quinazolinone, triazolinone and pyrimidinone angiotensin II antagonists incorporating a substituted benzyl element
GB9013615D0 (en) 1990-06-19 1990-08-08 Wellcome Found Pharmaceutical compounds
GB9016578D0 (en) 1990-07-27 1990-09-12 Ici Plc Fungicides
GB9018408D0 (en) 1990-08-22 1990-10-03 Ici Plc Fungicides
GB9023294D0 (en) 1990-10-25 1990-12-05 Ici Plc Fungicides
US5385894A (en) 1991-03-06 1995-01-31 Merck & Co., Inc. Disubstituted 6-aminoquinazolinones
NZ242290A (en) 1991-04-15 1994-12-22 Zeneca Ltd Pyridyl and pyrimidinyl substituted oxime-o-benzyl ether derivatives; preparatory processes, fungicidal compositions and an intermediate
US5252574A (en) 1991-04-26 1993-10-12 Merck & Co., Inc. Angiotensin II antagonists incorporating a substituted thiophene or furan
US5162325A (en) 1991-05-07 1992-11-10 Merck & Co., Inc. Angiotensin ii antagonists incorporating a substituted benzyl element
US5158953A (en) 1991-08-13 1992-10-27 National Science Council 2-substituted methyl-2,3-dihydroimidazo[1,2-c]quinazolin-5(6H)-ones (thiones), the preparation and use thereof
US6313146B1 (en) 1991-08-23 2001-11-06 Nps Pharmaceuticals, Inc. Calcium receptor-active molecules
DE4130298A1 (en) 1991-09-12 1993-03-18 Basf Ag FUNGICIDAL MIXTURES
EP0645091B1 (en) 1993-09-24 1996-04-17 BASF Aktiengesellschaft Fungicidal mixtures
US5464854A (en) 1993-11-11 1995-11-07 Depadova; Anathony S. Method of modifying ovarian hormone-regulated AT1 receptor activity as treatment of incapacitating symptom(s) of P.M.S.
GB2295387A (en) 1994-11-23 1996-05-29 Glaxo Inc Quinazoline antagonists of alpha 1c adrenergic receptors
GB9423911D0 (en) 1994-11-26 1995-01-11 Pfizer Ltd Therapeutic agents
DE19501481A1 (en) 1995-01-19 1996-07-25 Bayer Ag 2,8-disubstituted quinazolinones
DK0817775T3 (en) 1995-03-30 2001-11-19 Pfizer quinazoline
MY115814A (en) 1995-06-16 2003-09-30 Bayer Ip Gmbh Crop protection compositions
BR9609930A (en) 1995-08-17 1999-06-08 Basf Ag Fungicidal mixing process to control harmful fungi and use of compounds
JPH09241110A (en) 1996-03-08 1997-09-16 Sumitomo Chem Co Ltd Pesticide composition
PL330042A1 (en) 1996-05-15 1999-04-26 Pfizer Novel 2,3-disubstituted 4(3h)-quinazolynones
DE19629825A1 (en) 1996-07-24 1998-01-29 Bayer Ag Dihydrofuran carboxamides
WO1998010765A1 (en) 1996-09-13 1998-03-19 The Board Of Trustees Of The Leland Stanford Junior University Non-hormonal method of contraception
US6225318B1 (en) 1996-10-17 2001-05-01 Pfizer Inc 4-aminoquinazolone derivatives
US6331543B1 (en) 1996-11-01 2001-12-18 Nitromed, Inc. Nitrosated and nitrosylated phosphodiesterase inhibitors, compositions and methods of use
SK113299A3 (en) 1997-02-28 2001-05-10 Pfizer Prod Inc Atropisomers of 3-aryl-4(3h)-quinazolinones and their use as ampa-receptor antagonists
US5948775A (en) * 1997-03-19 1999-09-07 American Home Products Corporation 2- or 3-(substitutedaminoalkoxyphenyl)quinazolin-4-ones
US6627755B1 (en) 1997-06-09 2003-09-30 Pfizer Inc Quinazolin-4-one AMPA antagonists
EP0900567A3 (en) 1997-09-05 2001-05-02 Pfizer Products Inc. Quinazoline-4-one AMPA antagonists for the treatment of dyskinesias associated with dopamine agonist therapy
IL125950A0 (en) 1997-09-05 1999-04-11 Pfizer Prod Inc Methods of administering ampa receptor antagonists to treat dyskinesias associated with dopamine agonist therapy
US20030069231A1 (en) 1999-10-12 2003-04-10 Klaus Rudolf Modified aminoacids, pharmaceuticals containing these compounds and method for their production
JPH11279158A (en) 1998-02-09 1999-10-12 Pfizer Prod Inc Production of quinazoline-4-one derivative
JPH11228643A (en) * 1998-02-17 1999-08-24 Menicon Co Ltd Material for eye lens and its production
WO1999044612A1 (en) 1998-03-02 1999-09-10 Cocensys, Inc. Substituted quinazolines and analogs and the use thereof
US6187779B1 (en) 1998-11-20 2001-02-13 Cell Pathways, Inc. Method for inhibiting neoplastic cells and related conditions by exposure to 2,8-disubstituted quinazoline derivatives
GB9828340D0 (en) 1998-12-22 1999-02-17 Novartis Ag Organic compounds
DE19911509A1 (en) 1999-03-15 2000-09-21 Boehringer Ingelheim Pharma Bicyclic heterocycles, medicaments containing these compounds, their use and processes for their preparation
FR2792938B1 (en) 1999-04-28 2001-07-06 Warner Lambert Co NEWS 1-AMINO TRIAZOLO [4,3-a] QUINAZOLINE-5-ONES PHOSPHODIESTERASE IV INHIBITORS
US6664390B2 (en) 2000-02-02 2003-12-16 Warner-Lambert Company Llc Method for the simplified production of (3-chloro-4-fluorophenyl)-[7-(3-morpholin-4-yl-propoxy)-6-nitro-quinazoline-4-yl]-amine or (3-chloro-4-fluorophenyl)-[7-(3-morpholin-4-yl-propoxy)-6-amino-quinazoline-4-yl]-amine
US6697213B2 (en) * 2000-03-31 2004-02-24 Seagate Technology Llc Cover including multiple cover plates with damped layers
AU2001257146B2 (en) 2000-04-19 2006-11-30 Johns Hopkins University Methods for prevention and treatment of gastrointestinal disorders
US6667300B2 (en) 2000-04-25 2003-12-23 Icos Corporation Inhibitors of human phosphatidylinositol 3-kinase delta
EP1188438A1 (en) 2000-09-15 2002-03-20 Warner-Lambert Company Pharmaceutical composition for preventing or treating a disease associated with an excess of Il-12 production
EA007538B1 (en) 2000-12-11 2006-10-27 Туларик Инк. Cxcr3 antagonists
HUP0303249A3 (en) 2001-02-22 2007-03-28 Sankyo Co Water-soluble triazole fungicide compounds and pharmaceutical compositions containing them
EP1396488A1 (en) 2001-05-23 2004-03-10 Mitsubishi Pharma Corporation Fused heterocyclic compound and medicinal use thereof
US7220777B2 (en) 2001-10-15 2007-05-22 Smithkline Beecham P.L.C. Lactam derivatives as antagonists for human 11cby receptors
WO2003039460A2 (en) 2001-11-07 2003-05-15 Merck & Co., Inc. Mitotic kinesin inhibitors
US6559293B1 (en) 2002-02-15 2003-05-06 Transform Pharmaceuticals, Inc. Topiramate sodium trihydrate
US20050181066A1 (en) 2002-04-26 2005-08-18 Altana Pharma Ag Novel use of guanylate cyclase activators for the treatment of respiratory insufficiency
MXPA04011048A (en) 2002-05-06 2005-02-17 Vertex Pharmaceutivals Inc Thiadiazoles or oxadiazoles and their use as inhibitors of jak protein kinase.
CA2485343A1 (en) 2002-05-23 2004-05-13 Merck & Co., Inc. Mitotic kinesin inhibitors
WO2003103575A2 (en) * 2002-05-23 2003-12-18 Cytokinetics, Inc. Compounds, compositions, and methods
US20040242572A1 (en) 2002-08-24 2004-12-02 Boehringer Ingelheim International Gmbh New carboxamide compounds having melanin concentrating hormone antagonistic activity, pharmaceutical preparations comprising these compounds and process for their manufacture
US7557115B2 (en) 2002-09-30 2009-07-07 Cytokinetics, Inc. Compounds, compositions, and methods
KR20110050745A (en) 2002-10-03 2011-05-16 탈자진 인코포레이티드 Vasculostatic agents and methods of use thereof
GB0223730D0 (en) 2002-10-11 2002-11-20 Novartis Ag Organic compounds
FR2846657B1 (en) * 2002-11-05 2004-12-24 Servier Lab NOVEL PYRIDOPYRIMIDINONE COMPOUNDS, PROCESS FOR THEIR PREPARATION AND THE PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
US7407962B2 (en) 2003-02-07 2008-08-05 Vertex Pharmaceuticals Incorporated Heteroaryl compounds useful as inhibitors or protein kinases
TW200508224A (en) 2003-02-12 2005-03-01 Bristol Myers Squibb Co Cyclic derivatives as modulators of chemokine receptor activity
CL2004000409A1 (en) 2003-03-03 2005-01-07 Vertex Pharma COMPOUNDS DERIVED FROM 2- (REPLACED CILO) -1- (AMINO OR REPLACED OXI) -CHINAZOLINE, INHIBITORS OF IONIC SODIUM AND CALCIUM VOLTAGE DEPENDENTS; PHARMACEUTICAL COMPOSITION; AND USE OF THE COMPOUND IN THE TREATMENT OF ACUTE PAIN, CHRONIC, NEU
EP1603908B1 (en) 2003-03-12 2008-06-11 Millennium Pharmaceuticals, Inc. Quinazoline derivatives as tgf-beta inhibitors
WO2004084973A2 (en) * 2003-03-24 2004-10-07 Becton, Dickinson And Company Invisible antimicrobial glove and hand antiseptic
US20040242568A1 (en) 2003-03-25 2004-12-02 Syrrx, Inc. Dipeptidyl peptidase inhibitors
US8134015B2 (en) 2003-03-27 2012-03-13 Kyowa Hakko Kirin Co., Ltd. Compound inhibiting in vivo phosphorous transport and medicine containing the same
ES2423800T3 (en) 2003-03-28 2013-09-24 Novartis Vaccines And Diagnostics, Inc. Use of organic compounds for immunopotentiation
WO2004092120A2 (en) * 2003-04-07 2004-10-28 Nps Pharmaceuticals, Inc. Pyrimidinone compounds as calcilytics
ES2359451T3 (en) * 2003-04-07 2011-05-23 Nps Pharmaceuticals, Inc. PIRIMIDONE COMPOUNDS IN QUALITY CALCILITICS.
US20070161792A1 (en) 2003-04-07 2007-07-12 Nps Pharmacueticals, Inc Methods for preparing 2,3,5,6-substituted 3h-pyrimidin-4-ones
GB0309009D0 (en) 2003-04-22 2003-05-28 Astrazeneca Ab Quinazoline derivatives
KR20060018217A (en) 2003-05-23 2006-02-28 카이론 코포레이션 Guanidino-substituted quinazolinone compounds as mc4-r agonists
EP1636195A1 (en) 2003-05-27 2006-03-22 Pfizer Products Inc. Quinazolines and pyrido[3,4-d]pyrimidines as receptor tyrosine kinase inhibitors
PT1631295E (en) 2003-06-06 2010-05-24 Arexis Ab Use of condensed heterocyclic compounds as scce inhibitors for the treatment of skin diseases
KR100802856B1 (en) 2003-07-02 2008-02-12 에프. 호프만-라 로슈 아게 Arylamine-substituted quinazolinone compounds
AU2004272350A1 (en) 2003-09-16 2005-03-24 Astrazeneca Ab Quinazoline derivatives as tyrosine kinase inhibitors
AR045799A1 (en) 2003-09-24 2005-11-16 Glaxo Group Ltd COMPOUND 6- OXO-2,5- DIOXABICICLIC AND ITS USE TO PREPARE A PHARMACEUTICAL COMPOSITION
GB0326459D0 (en) 2003-11-13 2003-12-17 Astrazeneca Ab Quinazoline derivatives
KR20060105785A (en) 2003-11-19 2006-10-11 카이론 코포레이션 Quinazolinone compounds with reduced bioaccumulation
WO2005053662A1 (en) 2003-12-01 2005-06-16 Kudos Pharmaceuticals Limited Dna damage repair inhibitors for treatment of cancer
US20050239809A1 (en) 2004-01-08 2005-10-27 Watts Stephanie W Methods for treating and preventing hypertension and hypertension-related disorders
WO2005071096A2 (en) 2004-01-21 2005-08-04 Molecular Probes, Inc. Derivatives of cephalosporin and clavulanic acid for detecting beta-lacamase in a sample
RU2358969C2 (en) 2004-02-13 2009-06-20 Баниу Фармасьютикал Ко., Лтд. Condensed 4-oxopyrimidine derivative
DE602005017135D1 (en) 2004-03-05 2009-11-26 Nycomed Gmbh NEW USE OF PDE5 INHIBITORS
EP1730134A1 (en) 2004-03-26 2006-12-13 Vertex Pharmaceuticals Incorporated Pyridine inhibitors of erk2 and uses thereof
MXPA06012059A (en) 2004-04-22 2007-01-25 Boehringer Ingelheim Int New pharmaceutical compositions for the treatment of sexual disorders ii.
JP2007536239A (en) * 2004-05-06 2007-12-13 スミスクライン・ビーチャム・コーポレイション Calcium receptor antagonist compound
JP2008500338A (en) 2004-05-25 2008-01-10 イコス・コーポレイション Method for treating and / or preventing abnormal proliferation of hematopoietic cells
KR20070038500A (en) 2004-06-04 2007-04-10 아스트라제네카 아베 Quinazoline derivatives as erbb receptor tyrosine kinases
US7203079B2 (en) 2004-07-23 2007-04-10 System General Corp. Switching controller having frequency hopping for power supplies
US7466645B2 (en) * 2004-12-21 2008-12-16 Panasonic Corporation OFDM signal receiving apparatus
EP1964548A1 (en) 2007-03-02 2008-09-03 Novartis AG Pharmaceutical compositions comprising a calcilytic agent

Patent Citations (100)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US646572A (en) * 1899-03-15 1900-04-03 Archibald C Floyd Bath-cabinet.
US4140789A (en) * 1976-01-08 1979-02-20 Ciba-Geigy Corporation Etherified hydroxy-benzodiheterocyclic compounds
US4379788A (en) * 1980-12-12 1983-04-12 Dr. Karl Thomae Gesellschaft Mit Beschrankter Haftung 2-Phenyl-pyrimidones
US4431440A (en) * 1981-02-20 1984-02-14 American Cyanamid Company Method to alter or control the development and/or the life cycle of various plant species
US4588812A (en) * 1984-05-30 1986-05-13 Pennwalt Corporation Substituted 2,3-dihydro-5H-thiazolo[2,3,-b]quinazoline derivatives
US4731106A (en) * 1984-11-24 1988-03-15 Schering Agrochemicals Limited Fungicidal azole compounds
US4994495A (en) * 1988-05-13 1991-02-19 Imperial Chemical Industries Plc Fungicides
US4999381A (en) * 1989-02-02 1991-03-12 Imperial Chemical Industries Plc Fungicides
US5124329A (en) * 1989-06-28 1992-06-23 Imperial Chemical Industries Plc Fungicides
US5187779A (en) * 1989-08-11 1993-02-16 Micral, Inc. Memory controller with synchronous processor bus and asynchronous i/o bus interfaces
US5100886A (en) * 1989-12-18 1992-03-31 Imperial Chemical Industries Plc Metal complexes of pyridyl cyclopropane carboxamide compounds which are useful as fungicides
US5126338A (en) * 1990-01-04 1992-06-30 Imperial Chemical Industries Plc Fungicides which are n-pyridyl-cyclopropane carboxamides or derivatives thereof
US5304565A (en) * 1990-03-07 1994-04-19 Takeda Chemical Industries, Ltd. Nitrogen containing heterocyclic compounds, their production and use
US5185339A (en) * 1990-05-30 1993-02-09 Imperial Chemical Industries Plc Fungicidal compounds
US5225318A (en) * 1990-07-02 1993-07-06 Ocg Microelectronic Materials, Inc. Selected photoactive methylolated cyclohexanol compounds and their use in forming positive resist image patterns
US5206245A (en) * 1990-07-27 1993-04-27 Imperial Chemical Industries Plc Fungicidal aromatic pyrimidinyl oxime ethers
US20030060626A1 (en) * 1990-07-27 2003-03-27 Clough John Martin Fungicides
US5110471A (en) * 1990-08-30 1992-05-05 Conoco Specialty Products Inc. High efficiency liquid/liquid hydrocyclone
US5290780A (en) * 1991-01-30 1994-03-01 American Cyanamid Co. Angiotensin II receptor blocking 2,3,6 substituted quinazolinones
US5409930A (en) * 1991-05-10 1995-04-25 Rhone-Poulenc Rorer Pharmaceuticals Inc. Bis mono- and bicyclic aryl and heteroaryl compounds which inhibit EGF and/or PDGF receptor tyrosine kinase
US6011068A (en) * 1991-08-23 2000-01-04 Nps Pharmaceuticals, Inc. Calcium receptor-active molecules
US6031003A (en) * 1991-08-23 2000-02-29 Nps Pharmaceuticals, Inc. Calcium receptor-active molecules
US5482941A (en) * 1991-12-11 1996-01-09 Pfizer Inc. Quinazolinone antianginal agents
US5719098A (en) * 1992-12-21 1998-02-17 Sud-Chemie A.G. Sorbent based on smectitic clay minerals reacted with alkaline ion exchanger
US5401745A (en) * 1993-03-19 1995-03-28 Merck & Co., Inc. Quinazolinones substituted with phenoxyphenylacetic acid derivatives
US5420133A (en) * 1993-03-19 1995-05-30 Merck & Co., Inc. Quinazolinones substituted with phenoxyphenylacetic acid derivatives
US5721236A (en) * 1993-10-15 1998-02-24 Schering Corporation Tricyclic carbamate compounds useful for inhibition of G-protein function and for treatment of proliferative diseases
US5710346A (en) * 1993-10-22 1998-01-20 Ihara Chemical Industry Co., Ltd. 2,2-bis(3,5-disubstituted-4-hydroxyphenyl) propane derivative, process for production thereof, and process for producing pyrogallol using said derivative
US5756502A (en) * 1994-08-08 1998-05-26 Warner-Lambert Company Quinazolinone derivatives as cholyecystokinin (CCK) ligands
US6211244B1 (en) * 1994-10-21 2001-04-03 Nps Pharmaceuticals, Inc. Calcium receptor-active compounds
US6897213B1 (en) * 1995-11-21 2005-05-24 Warner-Lambert Company Heterocycles as cholecystokinin (CCK) ligands
US6211236B1 (en) * 1996-04-26 2001-04-03 Basf Aktiengesellschaft Fungicide mixtures
US6245792B1 (en) * 1996-04-26 2001-06-12 Basf Aktiengesellschaft Fungicidal mixture
US6391874B1 (en) * 1996-07-13 2002-05-21 Smithkline Beecham Corporation Fused heterocyclic compounds as protein tyrosine kinase inhibitors
US6380204B1 (en) * 1997-02-28 2002-04-30 Pfizer Inc Atropisomers of 3-heteroaryl-4(3H)-quinazolinones for the treatment of neurodegenerative and CNS-trauma related conditions
US6239130B1 (en) * 1997-04-30 2001-05-29 Warner-Lambert Company Phosphodiesterase 4-inhibiting diazepinoindolones
US6255352B1 (en) * 1997-05-28 2001-07-03 Basf Aktiengesellschaft Method for combating harmful fungi
US6369090B1 (en) * 1997-06-04 2002-04-09 Basf Aktiengesellschaft Fungicidal mixture
US6060479A (en) * 1997-06-09 2000-05-09 Pfizer Inc Quinazoline-4-one AMPA antagonists
US5753851A (en) * 1997-06-16 1998-05-19 The United States Of America As Represented By The Secretary Of The Army Spinning mine with concentrated projectiles
US6235730B1 (en) * 1997-12-12 2001-05-22 Japan Tobacco, Inc. 3-piperidyl-4-oxoquinazoline derivatives and pharmaceutical compositions comprising the same
US6350765B1 (en) * 1997-12-18 2002-02-26 Basf Aktiengesellschaft Fungicidal mixtures based on amide compounds and azoles
US6713485B2 (en) * 1998-01-12 2004-03-30 Smithkline Beecham Corporation Heterocyclic compounds
US20040102450A1 (en) * 1998-01-27 2004-05-27 Aventis Pharmaceuticals Inc. Substituted oxoazaheterocyclyl compounds
US6337332B1 (en) * 1998-09-17 2002-01-08 Pfizer Inc. Neuropeptide Y receptor antagonists
US6225315B1 (en) * 1998-11-30 2001-05-01 Pfizer Inc Method of treating nitrate-induced tolerance
US6350785B2 (en) * 1999-01-08 2002-02-26 Maxim Pharmaceuticals, Inc. Methods and compositions for topical treatment of damaged tissue using reactive oxygen metabolite production or release inhibitors
US6559160B1 (en) * 1999-08-27 2003-05-06 Chemocentryx, Inc. Compounds and methods for modulating cxcr3 function
US7060706B1 (en) * 1999-09-28 2006-06-13 3-Dimensional Pharmaceuticals, Inc. Quinazolinones
US6890930B1 (en) * 1999-09-28 2005-05-10 3-Dimensional Pharmaceuticals, Inc. Quinazolinones
US6545004B1 (en) * 1999-10-27 2003-04-08 Cytokinetics, Inc. Methods and compositions utilizing quinazolinones
US6562830B1 (en) * 1999-11-09 2003-05-13 Cell Pathways, Inc. Method of treating a patient having precancerous lesions with phenyl quinazolinone derivatives
US6518277B1 (en) * 2000-04-25 2003-02-11 Icos Corporation Inhibitors of human phosphatidylinositol 3-kinase delta
US6916956B2 (en) * 2000-08-11 2005-07-12 Japan Tobacco, Inc. Calcium receptor antagonist
US20040006130A1 (en) * 2000-08-11 2004-01-08 Yuko Shinagawa Calcium receptor antagonist
US20060019974A1 (en) * 2000-09-20 2006-01-26 Werner Mederski 4-Amino-quinazolines
US20030013733A1 (en) * 2000-09-22 2003-01-16 Richard Apodaca Octahydro-indolizine and quinolizine and hexahydro-pyrrolizine
US20020094326A1 (en) * 2000-10-13 2002-07-18 Johns Hopkins University Methods and compositions for nucleic acid delivery
US20040009980A1 (en) * 2000-10-25 2004-01-15 Pradip Bhatnagar Calcilytic compounds
US20030069230A1 (en) * 2001-01-02 2003-04-10 Becker Cyrus Kephra Quinazolone derivatives as alpha 1A/B adrenergic receptor antagonists
US7488727B2 (en) * 2001-06-01 2009-02-10 Vertex Pharmaceuticals Incorporated Thiazole compounds useful as inhibitors of protein kinase
US20040110777A1 (en) * 2001-12-03 2004-06-10 Annis Gary David Quinazolinones and pyridinylpyrimidinones for controlling invertebrate pests
US20040106616A1 (en) * 2002-01-17 2004-06-03 Rajagopal Bakthavatchalam Substituted quinazolin-4-ylamine analogues
US20040067969A1 (en) * 2002-02-15 2004-04-08 Gustave Bergnes Syntheses of quinazolinones
US20040077668A1 (en) * 2002-05-09 2004-04-22 Cytokinetics, Inc. Compounds, compositins, and methods
US20040019049A1 (en) * 2002-05-23 2004-01-29 Chiron Corporation Substituted quinazolinone compounds
US20040142949A1 (en) * 2002-07-23 2004-07-22 Gustave Bergnes Compounds, compositions, and methods
US20040048853A1 (en) * 2002-08-21 2004-03-11 Gustave Bergnes Compounds, compositions, and methods
US20040132732A1 (en) * 2002-10-21 2004-07-08 Wei Han Quinazolinones and derivatives thereof as factor Xa inhibitors
US20060052345A1 (en) * 2002-11-04 2006-03-09 Nps Pharmaceuticals, Inc. Quinazolinone compounds as calcilytics
US20040142958A1 (en) * 2002-12-13 2004-07-22 Neurogen Corporation Combination therapy for the treatment of pain
US20060079685A1 (en) * 2002-12-23 2006-04-13 Eva Altmann Aryl-quinazoline/aryl-2amino-phenyl methanone derivatives
US20060094723A1 (en) * 2003-05-22 2006-05-04 Torsten Dunkern Composition comprising a pde4 inhibitor and a pde5 inhibitor
US20050038051A1 (en) * 2003-06-09 2005-02-17 Jodi Nunnari Novel molecules for regulating cell death
US20060148693A1 (en) * 2003-06-16 2006-07-06 Altana Pharma Ag Composition comprising a pulmonary surfactant and a pde5 inhibitor for the treatment of lung diseases
US20050004177A1 (en) * 2003-07-02 2005-01-06 Warner-Lambert Company Llc Combination of an allosteric inhibitor of matrix metalloproteinase-13 and a ligand to an alpha-2-delta receptor
US20050137213A1 (en) * 2003-07-03 2005-06-23 Myriad Genetics, Incorporated Compounds and therapeutical use thereof
US20050043239A1 (en) * 2003-08-14 2005-02-24 Jason Douangpanya Methods of inhibiting immune responses stimulated by an endogenous factor
US7157476B2 (en) * 2003-08-20 2007-01-02 Vertex Pharmaceuticals Incorporated Aminofurazan compounds useful as protein kinase inhibitors
US20050054651A1 (en) * 2003-08-22 2005-03-10 Dendreon Corporation Compositions and methods for the treatment of disease associated with Trp-p8 expression
US20050059862A1 (en) * 2003-09-12 2005-03-17 Scimed Life Systems, Inc. Cannula with integrated imaging and optical capability
US20050059851A1 (en) * 2003-09-17 2005-03-17 Vincent Ardizzone Flexible magnetic pad with multi-directional constantly alternating polarity zones
US20070082873A1 (en) * 2003-10-28 2007-04-12 Lingenhoehl Kurt Combinations comprising ampa receptor antagonists for the treatment of schizophrenia
US20050130954A1 (en) * 2003-11-21 2005-06-16 Mitchell Ian S. AKT protein kinase inhibitors
US20050136085A1 (en) * 2003-12-19 2005-06-23 David Bellamy Panthenol and natural organic extracts for reducing skin irritation
US20070149553A1 (en) * 2003-12-19 2007-06-28 Arrington Kenneth L Mitotic kinesin inhibitors
US20070060601A1 (en) * 2003-12-19 2007-03-15 Arrington Kenneth L Mitotic kinesin inhibitors
US20060079538A1 (en) * 2004-05-13 2006-04-13 Dennis Hallahan Methods for inhibiting angiogenesis
US20060009455A1 (en) * 2004-06-15 2006-01-12 Corte James R Six-membered heterocycles useful as serine protease inhibitors
US20060069099A1 (en) * 2004-06-28 2006-03-30 Zice Fu Compounds, compositions and methods for prevention and treatment of inflammatory and immunoregulatory disorders and diseases
US20060017428A1 (en) * 2004-07-20 2006-01-26 Chin-Yuan Lin Electrical magnetic interference test system
US20060025420A1 (en) * 2004-07-30 2006-02-02 Boehringer Ingelheimn International GmbH Pharmaceutical compositions for the treatment of female sexual disorders
US20060154935A1 (en) * 2004-09-02 2006-07-13 Dean Wilson Quinazolines useful as modulators of ion channels
US20060063707A1 (en) * 2004-09-17 2006-03-23 Lifelike Biomatic, Inc. Compositions for enhancing memory and methods therefor
US20060107448A1 (en) * 2004-11-17 2006-05-25 Interspiro, Inc. Protective seal mechanism
US20060108038A1 (en) * 2004-11-25 2006-05-25 Sumitomo Rubber Industries, Ltd. Low noise tire support ring
US20070032506A1 (en) * 2005-07-02 2007-02-08 Peter Giannousis Crystalline forms of (2r-trans)-6-chloro-5[[4-[(4-fluorophenyl)methyl]-2,5-dimethyl-1-piperazinyl]carbonyl]-n,n, 1-trimethyl-alpha-oxo-1h-indole-3-acetamide monohydrochloride
US20070149548A1 (en) * 2005-12-22 2007-06-28 Alcon Manufacturing, Ltd. (indazol-5-yl)-pyrazines and (1,3-dihydro-indol-2-one)-pyrazines for treating rho kinase-mediated diseases and conditions
US20080154942A1 (en) * 2006-12-22 2008-06-26 Cheng-Fa Tsai Method for Grid-Based Data Clustering
WO2010079431A2 (en) * 2009-01-08 2010-07-15 Resverlogix Corp. Compounds for the prevention and treatment of cardiovascular disease

Cited By (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9023858B2 (en) 2010-07-23 2015-05-05 Takeda Pharmaceutical Company Limited Substituted pyrido[2,3-d]pyrimidines as delta-5-desaturase inhibitors
US9382216B2 (en) 2011-03-10 2016-07-05 Lupin Limited Substituted morpholines as modulators for the calcium sensing receptor
WO2012120476A1 (en) 2011-03-10 2012-09-13 Lupin Limited Substituted morpholines as modulators for the calcium sensing receptor
WO2012127385A1 (en) 2011-03-18 2012-09-27 Lupin Limited Benzo [b] [1, 4] oxazin derivatives as calcium sensing receptor modulators
WO2012127388A1 (en) 2011-03-18 2012-09-27 Lupin Limited Benzo [b] [1, 4] oxazin derivatives as calcium sensing receptor modulators
US9464063B2 (en) 2011-03-18 2016-10-11 Lupin Atlantis Holdings Sa Benzo [B] [1,4] oxazin derivatives as calcium sensing receptor modulators
US9163001B2 (en) 2012-02-24 2015-10-20 Lupin Limited Substituted chroman compounds as calcium sensing receptor modulators
US9987249B2 (en) 2012-02-24 2018-06-05 Lupin Limited Substituted chroman compounds as calcium sensing receptor modulators
US9598391B2 (en) 2012-02-24 2017-03-21 Lupin Atlantis Holdings Sa Substituted chroman compounds as calcium sensing receptor modulators
WO2013124828A1 (en) 2012-02-24 2013-08-29 Lupin Limited Substituted chroman compounds as calcium sensing receptor modulators
WO2013136288A1 (en) 2012-03-16 2013-09-19 Lupin Limited Substituted 3,4-dihydro-2h-benzo[b] [1,4]oxazine compounds as calcium sensing receptor modulators
WO2014033604A1 (en) 2012-08-27 2014-03-06 Lupin Limited Arylalkylamine compounds as calcium sensing receptor modulators
US20160073452A1 (en) * 2013-05-28 2016-03-10 Illinois Tool Works Inc. Induction pre-heating and butt welding device for adjacent edges of at least one element to be welded
WO2015022631A1 (en) 2013-08-12 2015-02-19 Lupin Limited Substituted biphenyl compounds as calcium sensing receptor modulators
US9493396B2 (en) 2013-08-28 2016-11-15 Lupin Atlantis Holdings Sa Substituted naphthalene compounds as calcium sensing receptor modulators
WO2015028938A1 (en) 2013-08-28 2015-03-05 Lupin Limited Substituted naphthalene compounds as calcium sensing receptor modulators
WO2015162538A1 (en) 2014-04-21 2015-10-29 Lupin Limited Heterocyclic compounds as calcium sensing receptor modulators for the treatment of hyperparathyroidism, chronic renal failure and chronic kidney disease
US20160105933A1 (en) * 2014-10-14 2016-04-14 Illinois Tool Works Inc. Reduced-distortion hybrid induction heating/welding assembly
WO2017037616A1 (en) 2015-08-31 2017-03-09 Lupin Limited Arylalkylamine compounds as calcium sensing receptor modulators
WO2021130779A1 (en) 2019-12-27 2021-07-01 Lupin Limited Pharmaceutical composition of casr modulators and methods and uses thereof
WO2021144814A1 (en) 2020-01-17 2021-07-22 Lupin Limited Methods, processes and intermediates for preparing chroman compounds

Also Published As

Publication number Publication date
US20080085887A1 (en) 2008-04-10
CA2663436A1 (en) 2008-04-10
CN101547922A (en) 2009-09-30
MX2009003673A (en) 2009-04-22
HK1137753A1 (en) 2010-08-06
AU2007303846B2 (en) 2011-03-10
UY30620A1 (en) 2008-05-31
WO2008041118A3 (en) 2008-10-23
US7829572B2 (en) 2010-11-09
EP2079739A2 (en) 2009-07-22
WO2008041118A2 (en) 2008-04-10
TW200822929A (en) 2008-06-01
AR063122A1 (en) 2008-12-30
CN101547922B (en) 2012-06-20
IL197536A0 (en) 2009-12-24
KR20090074066A (en) 2009-07-03
KR101129868B1 (en) 2012-04-12
PE20081455A1 (en) 2008-10-18
CL2007002864A1 (en) 2008-04-04
JP2010505811A (en) 2010-02-25
NO20091328L (en) 2009-05-04
AU2007303846A1 (en) 2008-04-10

Similar Documents

Publication Publication Date Title
US7829572B2 (en) Pyrido[4,3-d]pyrimidin-4(3H)-one derivatives as calcium receptor antagonists
WO2009001214A2 (en) Thieno[2,3-d]pyrimidin-4(3h)-one, isoxazolo[5,4-d]pyrimidin-4(5h)-one and isothiazolo[5,4-d]pyrimidin-4(5h)-one derivatives as calcium receptor antagonists
TWI751585B (en) Glucagon-like peptide 1 receptor agonists
JP6905530B2 (en) Farnesoid X receptor agonist and its use
RU2734261C2 (en) Optionally condensed heterocyclyl-substituted pyrimidine derivatives suitable for treating inflammatory, metabolic, oncological and autoimmune diseases
EP3025716B1 (en) Substance for treatment or relief of pain
US8076345B2 (en) 2-cyanophenyl-7,8-dihydro-5H-pyrido[4,3-d]pyrimidine compounds, compositions and uses thereof
US11560373B2 (en) Compounds and their use as PDE4 activators
TW201206944A (en) Morpholine compounds
US8592590B2 (en) Tetrahydrotriazolopyridine compounds as selective MGLU5 receptor potentiators useful for the treatment of schizophrenia
CA2566053A1 (en) Phenyl carboxamide compounds useful as beta-secretase inhibitors for the treatment of alzheimer&#39;s disease
HRP20030141A2 (en) Phenoxybenzylamine derivatives as selective serotonin re-uptake inhibitors
US20140200215A1 (en) Lysophosphatidic acid receptor antagonists
US11160797B2 (en) Pyridinone derivatives and their use as selective ALK-2 inhibitors
WO2010103429A1 (en) 1,1-(Dimethyl-Ethylamino)-2-Hydroxy-Propoxy]-Ethyl}-3-Methyl-Biphenyl-4- Carboxylic Acid Derivatives As Calcium Receptor Antagonists
EA045067B1 (en) GLUCAGON-LIKE PEPTIDE 1 RECEPTOR AGONISTS
JP2023531097A (en) Salt of Tetrahydroisoquinoline Derivatives, Method for Producing the Same, and Pharmaceutical Application Thereof

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO PAY ISSUE FEE