Aminodihydrothiazine derivatives substituted with a cyclic group

Abstract
This invention provides a compound of the formula (I):
Description
TECHNICAL FIELD

This invention relates to a compound having an inhibitory activity against production of amyloid β protein and useful for treating diseases induced by production, secretion and/or deposition of amyloid β protein.


BACKGROUND ART

In the brain of Alzheimer's patient many insoluble spots (senile plaque) are found, which is formed by extraneuronal accumulation of a peptide called amyloid β protein comprised of about 40 amino acids. Neuronal death caused by the senile plaque is considered to develop Alzheimer's Disease and an enhancer of amyloid β protein decomposition or vaccine of amyloid β protein etc. are extensively studied as a remedy for Alzheimer's Disease.


Secretase is an enzyme producing amyloid β protein by intracellular cleavage of a protein called amyloid β protein precursor (APP). An enzyme playing a role for producing an N-terminal of the amyloid β protein is called BACE1 (beta-site APP-cleaving enzyme) and an inhibitor of the BACE1, which will reduce production of amyloid β protein, could be a remedy for treating Alzheimers disease.


Patent literature 1 discloses a compound with a chemical structure similar to that of the compound of the present invention, having an inhibitory activity of NO synthetase and effective for treating dementia.


Patent literature 2-5 and non-patent literature 1-2 disclose compounds with a chemical structure similar to those of the compound of the present invention, and describe that each compound is useful as an anti-hypotensive agent, morphine-like analgesic or tranquilizer, intermediate of a therapeutic agent. NPYY5 antagonist, analgesic and the like.


Patent literatures 6-14 disclose BACE-1 inhibitors having a chemical structure different from that of the compound of the present invention. Also, patent literature 15 discloses a BACE-1 inhibitor,

  • Patent literature 1: WO 96/014842 Pamphlet
  • Patent literature 2: U.S. Pat. No. 3,235,551
  • Patent literature 3: U.S. Pat. No. 3,227,713
  • Patent literature 4: JP H09-067355
  • Patent literature 5: WO 2005/111031 Pamphlet
  • Patent literature 6: WO 02/96897 Pamphlet
  • Patent literature 7: WO 04/043916 Pamphlet
  • Patent literature 8: WO 2005/058311 Pamphlet
  • Patent literature 9: WO 2005/097767 Pamphlet
  • Patent literature 10: WO 2006/041404 Pamphlet
  • Patent literature 11: WO 2006/041405 Pamphlet
  • Patent literature 12: US 2007/0004786A
  • Patent literature 13: US 2007/0004730A
  • Patent literature 14: US 2007/27199A
  • Patent literature 15: WO 2007/049532 Pamphlet
  • Non-patent literature 1: Journal of Heterocyclic Chemistry, 14, 717-723 (1977)
  • Non-patent literature 2: Journal of Organic Chemistry, 33(8), 3126-3132 (1968).


DISCLOSURE OF INVENTION
Problem to be Solved

This invention provides with a compound having an inhibitory activity against BACE-1 and useful for treating diseases induced by production, secretion and/or deposition of amyloid β protein.


Means to Solve the Problem

The present invention provides with


1) a compound of the formula (I):




embedded image



wherein the ring A is an optionally substituted carbocyclic group or an optionally substituted heterocyclic group,


R1 is optionally substituted lower alkyl, optionally substituted lower alkenyl, optionally substituted lower alkynyl, an optionally substituted carbocyclic group or an optionally substituted heterocyclic group,


R2a and R2b are each independently hydrogen, optionally substituted lower alkyl or optionally substituted acyl,


R3a, R3b, R3c and R3d are each independently hydrogen, halogen, hydroxy, optionally substituted lower alkyl, optionally substituted lower alkenyl, optionally substituted acyl, optionally substituted lower alkoxy, optionally substituted carbocyclyl lower alkyl, optionally substituted heterocyclyl lower alkyl, optionally substituted carbocyclyl lower alkoxy, optionally substituted heterocyclyl lower alkoxy, optionally substituted aralkyl, optionally substituted heteroaralkyl, optionally substituted aralkyloxy, optionally substituted heteroaralkyloxy, optionally substituted lower alkylthio, carboxy, optionally substituted lower alkoxycarbonyl, optionally substituted amino, optionally substituted carbamoyl, an optionally substituted carbocyclic group or an optionally substituted heterocyclic group, or R3a and R3b or R3c and R3d may form a carbocyclic ring together with a linked carbon atom or may form oxo,


provided the following compounds i) and ii) are excluded;


i) a compound in which R2a is hydrogen, R2b is hydrogen, acetyl or phenyl, R1 is methyl, and the ring A is phenyl or 4-methoxyphenyl;


ii) a compound in which R2a is hydrogen, R2b is hydrogen, acetyl or phenyl, R1 is ethyl and the ring A is 3,4-dimethoxyphenyl,


a pharmaceutically acceptable salt or solvate thereof;


1′) a compound of the formula (I):




embedded image



wherein the ring A is an optionally substituted carbocyclic group or an optionally substituted heterocyclic group,


R1 is optionally substituted lower alkyl, optionally substituted lower alkenyl, optionally substituted lower alkynyl, an optionally substituted carbocyclic group or an optionally substituted heterocyclic group,


R2a and R2b are each independently hydrogen, optionally substituted lower alkyl or optionally substituted acyl,


R3a, R3b, R3c and R3d are each independently hydrogen, halogen, hydroxy, optionally substituted lower alkyl, optionally substituted lower alkenyl, optionally substituted acyl, optionally substituted lower alkoxy, optionally substituted lower alkylthio, carboxy, optionally substituted lower alkoxycarbonyl, optionally substituted amino, optionally substituted carbarnoyl, an optionally substituted carbocyclic group or an optionally substituted heterocyclic group, or


R3a and R3b or R3c and R3d may form a carbocyclic ring together with a linked carbon atom, provided the following compounds i) and ii) are excluded:


i) a compound in which R2a is hydrogen, R2b is hydrogen, acetyl or phenyl, R1 is methyl, and the ring A is phenyl or 4-methoxyphenyl;


ii) a compound in which R2a is hydrogen, R2b is hydrogen, acetyl or phenyl, R1 is ethyl and the ring A is 3,4-dimethoxyphenyl,


a pharmaceutically acceptable salt or solvate thereof;


2) the compound of I) or 1′) described above,


wherein the ring A is




embedded image



wherein the ring A′ is a carbocyclic group or a heterocyclic group,


G is




embedded image



wherein R5 is hydrogen, lower alkyl or acyl,


R6 is optionally substituted lower alkyl, optionally substituted lower alkenyl or optionally substituted lower alkynyl,


W1 is O or S,


W2 is O, S or NR5,


Ak is optionally substituted lower alkylene, optionally substituted lower alkenylene or optionally substituted lower alkynylene,


the ring B is an optionally substituted carbocyclic group or an optionally substituted heterocyclic group and each le may be independent:


R4 is halogen, hydroxyl, mercapto, halogeno lower alkyl, lower alkoxy, amino, lower alkylamino, acylamino or lower alkylthio and each R4 may be independent;


A pharmaceutically acceptable salt or solvate thereof;


2′) the compound of 1) or 1′) described above


wherein the ring A is




embedded image



wherein the ring A′ is a carbocyclic group or a heterocyclic group,


G is




embedded image



wherein R5 is hydrogen, lower alkyl or acyl,


R6 is optionally substituted lower alkyl, optionally substituted lower alkenyl or optionally substituted lower alkynyl,


W1 is O or S,


W2 is O, S or NR5,


Ak is optionally substituted lower alkylene, optionally substituted lower alkenylene or optionally substituted lower alkynylene,


the ring B is an optionally substituted carbocyclic group or an optionally substituted heterocyclic group and each R5 may be independent:


R4 is halogen, hydroxyl, mercapto, halogeno lower alkyl, lower alkyl, lower alkoxy, amino, lower alkylamino, acylamino or lower alkylthio and each R4 may be independent;


A pharmaceutically acceptable salt or solvate thereof;


3) the compound of 2) or 2′) described above wherein the ring A′ is phenyl or a nitrogen-containing aromatic heterocyclic group, a pharmaceutically acceptable salt or solvate thereof;


3′) the compound of 2) or 2′) described above wherein the ring A′ is phenyl, a pharmaceutically acceptable salt or solvate thereof;


3″) the compound of 2) or 2′) described above wherein the ring A′ is a nitrogen-containing aromatic monocyclic heterocyclic group, a pharmaceutically acceptable salt or solvate thereof;


3′″) the compound of 2) or 2′) described above wherein the ring A′ is pyridyl, a pharmaceutically acceptable salt or solvate thereof;


4) the compound of 1)-3), 1′), 2′), 3″), 3″) or 3′″) described above wherein R1 is C1-C3 alkyl, a pharmaceutically acceptable salt or solvate thereof;


4′) the compound of 1)-3), 1′), 2′), 3′), 3″) or 3′″) described above wherein R1 is optionally substituted lower alkynyl, a pharmaceutically acceptable salt or solvate thereof;


5) the compound of 1)-4), 1′), 2′), 3′), 3″), 3′″), 4′) described above wherein R2a and R2b are both hydrogen, a pharmaceutically acceptable salt or solvate thereof;


6) the compound of 1)-5), 1′), 2′), 3′), 3″), 3′″) or 4′) described above wherein all of R3a, R3b, R3c and R3d are hydrogen, a pharmaceutically acceptable salt or solvate thereof;


6′) the compound of 1)-5), 1′), 2′), 3′), 3″), 3′″) or 4′) described above wherein R3a and R3b are the same substituent selected from halogen and optionally substituted lower alkyl, a pharmaceutically acceptable salt or solvate thereof;


6″) the compound of 1)-5), 1′), 2′), 3′), 3″), 3′″) or 4′) described above wherein R3c and R3d are the same substituent selected from halogen and optionally substituted lower alkyl, a pharmaceutically acceptable salt or solvate thereof;


6′″) the compound of 1)-5), 1′), 2′), 3′), 3″), 3′″) or 4′) described above wherein R3a and R3b or R3c and R3d form a carbocyclic ring together with a linked carbon atom a pharmaceutically acceptable salt or solvate thereof;


7) the compound of 1)-5), 1′), 2′), 3′), 3″), 3′″) or 4′) described above wherein R3c or R3d is optionally substituted carbocyclic ring lower alkoxy or optionally substituted heterocyclyl lower alkoxy, a pharmaceutically acceptable salt or solvate thereof;


7′) the compound of 1)-5), 1′), 2′), 3′), 3″), 3′″) or 4′) described above wherein R3c and R3d form oxo together with a linked carbon atom, a pharmaceutically acceptable salt or solvate thereof;


8) a pharmaceutical composition comprising a compound of 1) 7), 1′), 2′), 3′), 3′), 3′″), 4′) 6′), 6″), 6′″) or 7′) described above, a pharmaceutically acceptable salt or solvate thereof as an active ingredient; and


9) a pharmaceutical composition having a BACE 1 inhibitory activity comprising a compound of 1) 7), 1′), 2′), 3′), 3″), 3′″), 4′) 6′), 6′″) or 7′) described above, a pharmaceutically acceptable salt or solvate thereof as an active ingredient;


The present invention also provides with


10) the pharmaceutical composition having a BACE 1 inhibitory activity of 9) described above, which is a composition having inhibitory activity of amyloid β protein production;


11) the pharmaceutical composition having a BACE 1 inhibitory activity of 9) described above, which is a medicine for treating diseases induced by production, secretion and/or deposition of amyloid 0 protein;


12) the pharmaceutical composition having a BACE 1 inhibitory activity of 9) described above, which is a medicine for treating Alzheimer's disease;


13) a method for treating diseases induced by production, secretion and/or deposition of amyloid β protein, characterized in administering a compound of the formula (I) described in 1) above, a pharmaceutically acceptable salt or solvate thereof;


14) use of a compound of the formula (I) described in 1) above, a pharmaceutically acceptable salt or solvate thereof described in 1) above, in manufacturing a medicine for treating diseases induced by production, secretion and/or deposition of amyloid 3 protein;


15) a method for treating diseases induced by BACE 1 characterized in administering a compound of the formula (I) described in 1) above, a pharmaceutically acceptable salt or solvate thereof;


16) use of a compound of the formula (I) described in 1) above, a pharmaceutically acceptable salt or solvate thereof, in manufacturing a medicine for treating diseases induced by BACE 1;


17) a method for treating Alzheimer's disease characterized in administering a compound of the formula (I) described in 1) above, a pharmaceutically acceptable salt or solvate thereof; and


18) use of a compound of the formula (I) described in 1) above, a pharmaceutically acceptable salt or solvate thereof in manufacturing a medicine for treating Alzheimer's disease.


Effect of Invention

A compound of the present invention is useful for treating diseases induced by production, secretion and/or deposition of amyloid β protein (Alzheimer's disease etc.).







BEST MODE FOR CARRYING OUT THE INVENTION

In this description, “halogen” includes fluorine, chlorine, bromine and iodine.


A moiety of halogen in “halogeno lower alkyl” and “halogen lower alkoxycarbonyl” is the same as “halogen” above.


“Lower alkyl” includes C1-C15, preferably C1-C10, more preferably C1-C6 and further more preferably C1-C3 straight or branched alkyl, and for example, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, isopentyl, neopentyl, hexyl, isohexyl, n-heptyl, isoheptyl, n-octyl, isooctyl, n-nonyl and n-decyl are exemplified.


A moiety of alkyl in “lower alkoxy”, “halogeno lower alkyl”, “hydroxyl lower alkyl”, “hydroxyl lower alkoxy”, “lower alkoxycarbonyl”, “halogeno lower alkoxycarbonyl”, “lower alkoxycarbortyl lower alkyl”, “lower alkylamino”, “lower alkoxy lower alkyl”, “hydroxyimino lower alkyl”, “lower alkoxyitnino lower alkyl”, “amino lower alkyl” “lower alkoxy lower alkoxy”, “lower alkoxy lower alkenyl”, “lower alkoxycarbonyl lower alkenyl”, “lower alkoxy lower alkynyl”, “lower alkoxycarbonyl lower alkynyl”, “lower alkyl carbamoyl”, “lower alkyl carbamoyl”, “lower alkoxyimino”, “lower alkylthio”, “lower alkylsulfonyl”, “lower alkylsulfonyloxy”, “lower alkyl sulfamoyl”, “lower alkyl sulfinyl”, “carbocyclyl lower alkyl”, “carbocyclyl lower alkyl”, “carbocyclyl lower alkoxy”, “carbocyclyl lower alkoxycarbonyl”, “carbocyclyl lower alkylamino”, “carbocyclyl lower alkyl carbamoyl”, “cycloalkyl lower alkyl”, “cycloalkyl lower alkoxy”, “cycloalkyl lower alkylamino”, “cycloalkyl lower alkoxycarbonyl”, “cycloalkyl lower alkylcarbamoyl”, “aryl lower alkyl”, “aryl lower alkoxy”, “aryl lower alkylamino”, “lower alkoxycarbonyl”, “aryl lower alkoxycarbarnoyl”, “heterocyclyl lower alkyl”, “heterocyclyl lower alkoxy”, “heterocyclyl lower alkylamino”, “heterocyclyl lower alkoxycarbonyl” and “heterocyclyl lower alkylcarbamoyl” is the same as “alkyl” above.


“Optionally substituted lower alkyl” may be substituted with one or more of substituent(s) selected from a substituent group α.


Group α is a group consisting of halogen, hydroxy, lower alkoxy, hydroxy lower alkoxy, lower alkoxy lower alkoxy, acyl, acyloxy, carboxy, lower alkoxycarbonyl, amino, acylamino, lower alkylamino, imino, hydroxyimino, lower alkoxyimino, lower alkylthio, carbamoyl, lower alkylcarbamoyl, hydroxy lower alkylcarbarnoyl, sulfamoyl, lower alkylsulfamoyl, lower alkylsulfonyl, cyano, nitro, a carbocyclic group and a heterocyclic group.


One or more of substituent(s) selected from the substituent group α is exemplified as a substituent of “optionally substituted lower alkoxy”, “optionally substituted lower alkoxycarbonyl” and “optionally substituted lower alkylthio”


“Lower alkylidene” includes a divalent group derived from the “lower alkyl” above, and methylidene, ethylidene, propylidene, isopropylidene, butylidene, pentylidene and hexylidene etc. are exemplified.


“Lower alkenyl” includes C2-C15, preferably C2-C10, more preferably C2-C6 and further more preferably C2-C4 straight or branched alkenyl having one or more double bond(s) at any position thereof. Examples of lower alkenyl include vinyl, allyl, propenyl, isopropenyl, butenyl, isobutenyl, prenyl, butadienyl, pentenyl, isopentenyl, pentadienyl, hexenyl, isohexenyl, hexadienyl, heptenyl, octenyl, nonenyl, decenyl, undecenyl, dodecenyl, tridecenyl, tetradecenyl, pentadecenyl and the like.


“Lower alkynyl” includes C2-C10, preferably C2-C8, more preferably C3-C6 straight or branched alkynyl haying one or more triple bond(s) at any position thereof. Examples of lower alkynyl include ethynyl, propynyl, butynyl, pentynyl, hexynyl, heptynyl, octynyl, nonynyl, decynyl and the like. Lower alkynyl may additionally have a double bond at any position thereof.


One or more of subsistent(s) selected from the substituent group α is exemplified as a substituent of “optionally substituted lower alkenyl” and “optionally substituted lower alkynyl”.


A moiety of lower alkenyl in “hydroxyl lower alkenyl”, “lower alkoxy lower alkenyl”, “lower alkoxycarbonyl lower alkenyl”, “carbocyclyl lower alkenyl”, “lower alkenyloxy”, “lower alkenylthio” and “lower alkenylamino” is the same as that of “lower alkenyl”.


A moiety of lower alkynyl in “hydroxyl lower alkynyl”, “lower alkoxy lower alkynyl”, “lower alkoxycarbonyl lower alkynyl”, “carbocyclyl lower alkynyl”, “lower alkynyloxy”, “lower alkenylamino” and “lower alkynylamino” is the same as that of “lower alkynyl” above


One or more substituents selected from lower alkyl, acyl, hydroxyl, lower alkoxy, lower alkoxycarbonyl, a carbocyclic group and a heterocyclic group etc. is exemplified as a substituent of “optionally substituted amino” and “optionally substituted carbamoyl”.


“Acyl” includes C1-C10 aliphatic acyl, carbocyclyl carbonyl and heterocyclic carbonyl, and examples of acyl include formyl, acetyl, propionyl, butyryl, isobutyryl, valeryl, pivaloyl, hexanoyl, acryloyl, propioloyl, methacryloyl, crotonoyl, benzoyl, cyclohexanecarbonyl, pyridinecarbonyl, furancarbonyl, thiopheriecarbonyl, benzothiazolecarbonyl, pyrazinecarbonyl, piperidinecarbonyl, thiomorpholino and the like.


A moiety of acyl in “acylamino” and “acyloxy” is the some as described above.


One or more substituents selected from the substituent group α is exemplified as a substituent in “optionally substituted acyl” and a moiety of the ring in carbocyclyl carbonyl and heterocyclylcarbonyl is optionally substituted with one or more substituent(s) selected from lower alkyl, the substituent group α and lower alkyl substituted with one or more substituent(s) selected from the substituent group α.


“A carbocyclic group” includes cycloalkyl, cycloalkenyl, aryl, and non-aromatic fused carbocyclic group etc.


“Cycloalkyl” includes C3-C10, preferably C3-C8 and more preferably C4-C8 carbocyclic group and examples of cycloalkyl include cyclopropyl, cyclobutyl, cyclopentyl cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl and the like.


A moiety of cycloalkyl in “cycloalkyl lower alkyl”, “cycloalkyloxy”, “cycloalkyl lower alkoxy”, “cycloalkylthio”, “cycloalkylamio”, “cycloalky lower alkylamino”, “cycloalkylsulfamoyl”, “cycloalkylsulfonyl”, “cycloalkylcaxbamoyl”, “cycloalkyl lower alkylcarbamoyl”, “cycloalkyl lower alkoxycarbonyl” and “cycloalkylcarbonyl” is the same as “cycloalkyl” described above


“Cycloalkenyl” includes the above cycloalkyl having one or more double bond(s) at any position on the ring, and examples of the cycloalkenyl include cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, cycloheptynyl, cyclooctynyl, and cyclohexadienyl etc.


Examples of “aryl” include phenyl, naphthyl, anthryl and phenanthryl etc, and especially phenyl is preferable.


“Non-aromatic fused carbocyclic group” includes a group in which two or more cyclic groups selected from “cycloalkyl”, “cycloalkenyl” and “aryl” described above fused, and examples of “Non-aromatic fused carbocyclic group” include indanyl, indenyl, tetrahydronaphthyl and fluorenyl etc.


“Forming a carbocyclic ring together with a linked carbon atom” means that two substituents jointly form “cycloalkyl” above.


A moiety of the carbocyclic ring in “carbocyclyloxy”, “carbocyclyl lower alkyl”, “carbocyclyl lower alkenyl”, “carbocyclyl lower alkynyl”, “carbocyclyl lower alkoxy”, “carbocyclyl lower alkoxycarbonyl”, “carbocyclylthio”, “carbocyclyl amino”, “carbocyclyl lower alkylamino”, “carbocyclylcarbonyl”, “carbocyclylsulfamoyl”, “carbocyclysulfonyl”, “carbocyclylcarbamoyl”, “carbocyclyl lower alkyl carbamoyl”, “carbocyclyloxycarbonyl” is the same as the “carbocyclic group”.


A moiety of aryl in “aryl lower alkyl”, “aryloxy”, “aryloxycarbonyl”, “aryloxycarbonyloxy”, “aryl lower alkoxycarbonyl”, “arylthio”, “arylamino”, “aryl lower alkoxy”, “aryl lower alkylamino”, “arylsulfonyl”, “arylsulfonyloxy”, “arylsulfinyl”, “arylsulfamoyl”, “arylcarbamoyl” and “aryl lower alkylcarbamoyl” is the same as the “aryl” above,


“Heterocyclic group” includes a heterocyclic group containing one or more heteroatom(s) each independently selected from O, S and N, and examples of “heterocyclic group” include 5- or 6-membered heteroaryl such as pyrrolyl, pyrazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazolyl, triazinyl, tetrazolyl, furyl, thienyl, isoxazolyl, oxazolyl, oxadiazolyl, isothiazolyl, thiazolyl and thiadiazolyl etc.; a non-aromatic heterocyclic group such as dioxanyl, thiiranyl, oxiranyl, oxetanyl, oxathiolanyl, azetidinyl, thianyl, thiazolidinyl, pyrrolidinyl, pyrrolinyl, imidazolinyl, pyrazolidinyl, pirazolinyl, piperidyl, piperazinyl, morpholinyl, morpholino, thiomorpholinyl, thiomorpholino, dihydropyridyl, tetrahydropyridyl, tetrahydrofuryl, tetrahydropyranyl, dihydrothiazolyl, tetrahydrothiazolyl, tetrahydroisothiazolyl, dihydrooxazinyl, hexahydroazepinyl, tetrahydrodiazepinyl and tetrahydropyridazinyl etc.;


a fused bicyclic heterocyclic group such as indolyl, isoindolyl, indazolyl, indolidinyl, quinolyl, isoquinolyl, cinnolinyl, phthaladinyl, quinazolinyl, naphthilidinyl, quinoxalinyl, purinyl, pteridinyl, benzopyranyl, benzimidazolyl, benzotriazolyl, benzisoxazolyl, benzoxazolyl, benzoxadiazolyl, benzoisothiazolyl, benzothiazolyl, benzothiadiazolyl, benzofuryl, isobenzofuryl, benzothienyl, benzotriazolyl, thienopyridyl, thienopyrrolyl, thienopyrazolyl thienopyrazinyl, furopyrrolyl, thienothienyl, imidazopyridyl, pyrazolopyridyl, thiazolopyridyl, pyrazolopyrimidinyl, pyrazolotriazinyl, pyridazolopyridyl, triazolopyridyl, imidazothiazolyl, pyrazinopyridazinyl, quinazolinyl, dthydrothiazolopyrimidinyl, tetrahydroquinolyl, tetrahydroisoquinolyl, dihydrobenzofuryl, dihydrobenzoxazinyl, dihydrobenzjmidazolyl, tetrahydrobenzothienyl, tetrahydrobenzofuryl, benzodioxolyl, benzodioxonyl, chromanyl, chromenyl, octahydrochromenyl, dihydrobenzodioxinyl, dihydrobenzooxedinyl, ditiydrobenzooxepinyl and dihydrothienodioxinyl etc.; and a fused tricyclic heterocyclic group such as carbazolyl, xanthenyl, phenothiazinyl, phenoxathiinyl, pherioxaclinyl, dibenzofuryl, imidazoquinolyl and tetrahydrocarbazolyl etc.; and preferably includes 5- or 6-membered heteroaryl and a non-aromatic heterocyclic group.


A moiety of the heterocyclic group “heterocyclyl lower alkyl”, “heterocyclyloxy”, “heterocyclylthio”, “heterocyclylcarbonyl”, “heterocyclyl lower alkoxy”, “heterocyclyl amino”, “heterocyclyl carbonylamino”, “heterocyclyl sulfamoyl”, “heterocyclylsulfonyl”, “heterocyclylcarbamoyl”, “heterocyclyloxycarbonyl”, “heterocyclyl lower alkylamino”, “heterocyclyl lower alkoxycarbonyl” and “heterocyclyl lower alkylcarbarnoyl” is the same as the “heterocyclic group” above.


“A nitrogen-containing aromatic heterocyclic group” means a group of the “heterocyclic group” above containing at least one nitrogen atom, and examples of the “nitrogen-containing aromatic heterocyclic group” include 5- or 6-membered heteroaryl such as pyrrolyl, imidazolyl, pyrazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazolyl, triazinyl, tetrazolyl, isoxazolyl, oxazolyl, oxadiazolyl, isothiazolyl, thiazolyl and thiadiazolyl etc.;


a fused bicyclic heterocyclo group such as indolyl, isoindolyl, indazolyl, indolidinyl, isoindolinyl, quinolyl, isoquinolyl, cinnolinyl, phthaladinyl, quinazolinyl, naphthilidinyl, quinoxalinyl, purinyl, pteridinyl, benzopyranyl, benzimidazolyl, benzotriazolyl, benzisoxazolyl, benzoxazolyl, benzoxadiazolyl, benzoisothiazolyl, benzothiazolyl, benzothiadiazolyl, benzotriazolyl, imidazopyridyl, pyrazolopyridine, triazolopyridyl, imidazothiazolyl, pyrazinopyridazinyl, quinazolinyl, quinolyl, isoquinolyl, naphthylidinyl, dihydrobenzofuryl, tetrahydroquinolyl, tetrahydroisoquinolyl, dihydrobenzoxazine etc.; and


a fused tricyclic heterocyclo group such as carbazolyl, acridinyl, xanthenyl and imidazoquinolyl etc.; and pyrrolidinyl, pyrrolinyl, imidazolidinyl, imidazolinyl, pyrazolidinyl, pyrazolinyl, piperidyl, piperazinyl, morpholinyl, morpholino, thiomorpholinyl, thiomorpholino, dihydropiridyl, dihydrobenzimidazolyl, tetrahydropyridyl, tetrahydrothiazolyl and tetrahydroisothiazolyl etc.


“The heterocyclic group” or “nitrogen-containing aromatic heterocyclic group” above may be linked to other group at any position on the ring,


“Nitrogen-containing aromatic monocyclic heterocyclic group” means a monocyclic group in the “nitrogen-containing aromatic heterocyclic group” and examples of the “Nitrogen-containing aromatic monocyclic heterocyclic group” include 5- or 6-membered heteroaryl such as pyrrolyl, pyrazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazolyl, triazinyl, tetrazolyl, isoxazolyl, oxazolyl, oxadiazolyl, isothiazolyl, thiazolyl and thiadiazolyl etc.


“The nitrogen-containing aromatic monocyclic heterocyclic group” above may be linked to other group at any carbon atom on the ring.


Examples of a substituent in the “optionally substituted carbocyclic group” and “optionally substituted heterocyclic group” of the ring A and B include the substituent group α (preferably halogen, hydroxyl, acyl, acyloxy, carboxy, lower alkoxycarbonyl, carbamoyl, amino, cyano, lower alkylamino, lower alkylthio etc.); lower alkyl optionally substituted with one or more substituent(s) selected from the substituent group α, hydroxyimino and lower alkoxyimino, wherein examples of preferable substituents include halogen, hydroxyl, lower alkoxy, lower alkoxycarbonyl etc.; amino lower alkyl substituted with one or more substituent(s) selected from the substituent group α, wherein examples of preferable substituents include acyl, lower alkyl and/or lower alkoxy etc.;


hydroxyimino lower alkyl, lower alkoxyimino lower alkyl;


lower alkenyl optionally substituted with one or more substituent(s) selected from the substituent group α, wherein examples of preferable substituents include lower alkoxycarbonyl, halogen and/or halogeno lower alkoxycarbonyl;


lower alkynyl optionally substituted with one or more substituent(s) selected from the substituent group α, wherein examples of preferable substituents include lower alkoxycarbonyl etc;


lower alkoxy optionally substituted with one or more substituent(s) selected from the substituent group α, wherein examples of preferable substituents include halogenocarbamoyl, oxetane, lower alkylcarbarnoyl, hydroxyl lower alkylcarbamoyl;


lower alkoxy lower alkoxy optionally substituted with one or more substituent(s) selected from the substituent group α;


lower alkenyloxy optionally substituted with one or more substituent(s) selected from the substitute group α wherein examples of preferable substituents include halogen, hydroxyl, amino, lower alkyl etc.;


lower alkoxy lower alkenyloxy optionally substituted with one or more substituent(s) selected from the substituent group α;


lower alkynyloxy optionally substituted with one or more substituent(s) selected from the substituent group α, wherein examples of preferable substituents include halogen, hydroxyl etc.;


lower alkoxy lower alkynyloxy optionally substituted with one or more substituent(s) selected from the substituent group α;


lower alkylthio optionally substituted with one or more substituent(s) selected from the substituent group α;


lower alkenylthio optionally substituted with one or more substituent(s) selected from the substituent group α;


lower alkynylthio optionally substituted with one or more substituent(s) selected from the substituent group α;


lower alkynylthio substituted with one or more substituent(s) selected from the substituent group α;


lower alkenylamino substituted with one or more substituent(s) selected from the substituent group α;


lower alkynylamino substituted with one or more substituent(s) selected from the substituent group α;


aminooxy optionally substituted with one or more substituent(s) selected from lower alkylidene and the substituent group α;


acyl substituted with one or more substituent(s) selected from the substituent group α;


lower alkylsulfonyl optionally substituted with one or more substituent(s) selected from the substituent group α;


lower alkylsulfinyl optionally substituted with one or more substituent(s) selected from the substituent group α;


sulfamoyl;


lower alkylsulfamoyl optionally substituted with one or more substituent(s) selected from the substituent group α;


a carbocyclic group (preferably cycloalkyl, aryl etc.) optionally substituted with one or more substituent(s) selected from the substituent group α, azide, lower alkyl and halogeno lower alkyl;


a heterocyclic group optionally substituted with one or more substituent(s) selected from the substituent group α, azide, lower alkyl and halogeno lower alkyl;


carbocyclyl lower alkyl (preferably cycloalkyl lower alkyl, aryl lower alkyl etc.) optionally substituted with one or more substituent(s) selected from the substituent group α, azide, lower alkyl and halogeno lower alkyl;


heterocyclyl lower alkyl optionally substituted with one or more substituent(s) selected from the substituent group α, azide, lower alkyl and halogeno lower alkyl;


carbocyclyloxy (preferably cycloalkyloxy, aryloxy etc.) optionally substituted with one or more substituent(s) selected from the substituent group α, azide, lower alkyl and halogeno lower alkyl;


heterocyclyloxy optionally substituted with one or more substituent(s) selected from the substituent group α, azide, lower alkyl and halogeno lower alkyl;


carbocyclyl lower alkoxy (preferably cycloalkyl lower alkoxy, aryl lower alkoxy, etc) optionally substituted with one or more substituent(s) selected from the substituent group α, azide, lower alkyl and halogeno lower alkyl;


heterocyclyl lower alkoxy (preferably cycloalkyl lower alkoxycarbonyl, aryl lower alkoxycarbonyl etc.) optionally substituted with one or more substituent(s) selected from the substituent group α, azide, lower alkyl and halogeno lower alkyl,


carbocyclyl lower alkoxycarbonyl optionally substituted with one or more substituent(s) selected from the substituent group α, azide, lower alkyl and halogeno lower alkyl;


heterocyclyl lower alkoxycarbonyl optionally substituted with one or more substituent(s) selected from the substituent group α, azide, lower alkyl and halogeno lower alkyl;


carbocyclylthio (preferably cycloalkylthio, arylthio etc.) optionally substituted with one or more substituent(s) selected from the substituent group α, azide, lower alkyl and halogeno lower alkyl;


heterocyclylthio optionally substituted with one or more substituent(s) selected from the substituent group α, azide, lower alkyl and halogeno lower alkyl;


carbocyclyl amino (preferably cycloalkylamino, arylamino etc.) optionally substituted with one or more substituent(s) selected from the substituent group α, azide, lower alkyl and halogeno lower alkyl;


heterocyclylamino optionally substituted with one or more substituent(s) selected from the substituent group α, azide, lower alkyl and halogeno lower alkyl;


carbocyclyl lower alkylamino (preferably cycloalkyl lower alkylamino, aryl lower alkylamino etc.) optionally substituted with one or more substituent(s) selected from the substituent group α, azide, lower alkyl and halogeno lower alkyl;


heterocyclyl lower alkylamino optionally substituted with one or more substituent(s) selected from the substituent group α, azide, lower alkyl and halogeno lower alkyl;


lower alkylsulfamoyl optionally substituted with one or more substituent(s) selected from the substituent group α;


carbocyclylsulfamoyl (preferably cycloalkyl sulfamoyl, arylsulfamoyl etc.) optionally substituted with one or more substituent(s) selected from the substituent group α, azide, lower alkyl and halogeno lower alkyl;


heterocyclylsulfamoyl optionally substituted with one or more substituent(s) selected from the substituent group α, azide, lower alkyl and halogeno lower alkyl;


carbocyclylsulfonyl (preferably cycloalkyl sulfonyl, arylsulfonyl etc.) optionally substituted with one or more substituent(s) selected from the substituent group α, azide, lower alkyl and halogeno lower alkyl;


heterocyclylsulfonyl optionally substituted with one or more substituent(s) selected from the substituent group α, azide, lower alkyl and halogeno lower alkyl;


carbocyclylcarbamoyl (preferably cycloalkyl carbamoyl, aryl carbamoyl etc.) optionally substituted with one or more substituent(s) selected from the substituent group α, azide, lower alkyl and halogeno lower alkyl;


heterocyclyl carbamoyl optionally substituted with one or more substituent(s) selected from the substituent group α, azide, lower alkyl and halogeno lower alkyl;


carbocyclyl lower alkylcarbamoyl (preferably cycloalkyl lower alkylcarbamoyl, aryl lower alkylcarbamoyl etc.) optionally substituted with one or more substituent(s) selected from the substituent group α, azide, lower alkyl and halogeno lower alkyl;


heterocyclyl lower alkylcarbamoyl optionally substituted with one or more substituent(s) selected from the substituent group α, azide, lower alkyl and halogeno lower alkyl, carbocyclyloxycarbonyl (preferably cycloalkyloxycarbonyl, aryloxycarbonyl etc.) optionally substituted with one or more substituent(s) selected from the substituent group α, azide, lower alkyl and halogeno lower alkyl;


heterocyclyloxycarbonyl optionally substituted with one or more substituent(s) selected from the substituent group et, azide, lower alkyl and halogeno lower alkyl;


lower alkylenedioxy optionally substituted with halogen;


oxo, azide and the like.


These may be substituted with one or more substituents selected from these groups.


Also the ring A may be substituted with one or more group(s) selected from




embedded image


embedded image


embedded image




    • wherein Ak1, Ak2 and Ak3 are each independently a single bond, optionally substituted lower alkylene, optionally substituted lower alkenylene or optionally substituted lower alkynylene;

    • Ak4 is optionally substituted lower alkylene, optionally substituted lower alkenylene or optionally substituted lower alkynylene;

    • W1 and W3 are each independently O or S,

    • W2 is O, S or NR5,

    • R5 and R6 are each independently hydrogen, lower alkyl, hydroxy lower alkyl, lower alkoxy lower alkyl, lower alkoxycarbonyl lower alkyl, carbocyclyl lower alkyl, lower alkenyl, hydroxyl lower alkenyl, lower alkoxy lower alkenyl, lower alkoxycarbonyl lower alkenyl, carbocyclyl lower alkenyl, lower alkynyl, hydroxyl lower alkynyl, lower alkoxy lower alkynyl, lower alkoxycarbonyl lower alkynyl, carbocyclyl lower alkynyl or acyl;

    • R7 is hydrogen or lower alkyl;

    • the ring B is an optionally substituted carbocyclic group or an optionally substituted heterocyclic group; and

    • p is 1 or 2; W1, W3 or W5 may be independent when it is pluralized. Additionally the oxygen atom of (xii) may be cis or trans to the substituent R7.





Preferable examples of (i) to (xixi) above include




embedded image



wherein Ak is optionally substituted lower alkylene, optionally substituted lower alkenylene or optionally substituted lower alkynylene, and the other symbols are the same as described above.


In other cases of “an optionally substituted carbocyclic group” and “an optionally substituted heterocyclic group”, one or more substituent(s) selected from a group of lower alkyl and the substituent group α may be exemplified as a substituent of “an optionally substituted carbocyclic group” and “an optionally substituted heterocyclic group”


“Heteroaryl” includes an aromatic cyclic group among the “heterocyclic group” above.


“Lower alkylene” includes C1-C10, preferably C1-C6, more preferably C1-C3 straight or branched divalent carbon chain, and for example, methylene, climethylene, trimethylene, tetramethylene and methyl trimethylene are exemplified.


A moiety of lower alkylene in “lower alkylenedioxy” is the same as the “lower alkylene” described above,


“Lower alkenylene” includes C2-C10, preferably C2-C6, more preferably C2-C4 straight or branched divalent carbon chain having a double bond at any arbitrary position thereof, and vinylene, propenylene, butenylene, butadienylene, methyl propenylene, pentenylene and hexenylene are exemplified.


“Lower alkynylene” includes C2-C10, preferably C2-C6, more preferably C2-C4 straight or branched divalent carbon chain having a triple bond and also a double bond at any arbitrary position thereof, and for example, ethynylene, propynylene, butynylene, pentynylene and hexynylene are exemplified.


Examples of a substituent in “optionally substituted lower alkylene”, “optionally substituted lower alkenylene” and “optionally substituted lower alkynylene” include the substituent group α, and preferably halogen and hydroxyl etc. are exemplified.


Examples of a substituent in “optionally substituted carbocyclyl lower alkyl”, “optionally substituted heterocyclyl lower alkyl”, “optionally substituted carbocyclyl lower alkoxy”, and “optionally substituted heterocyclyl lower alkoxy” include one or more substituent(s) selected from lower alkyl and the substituent group α.


In this specification, “solvate” includes a solvate with an organic solvent and a hydrate etc. and hydrate may be coordinated with optional number of water molecule.


The compound (1) includes pharmaceutical acceptable salt thereof. Examples of the pharmaceutical acceptable salt include a salt with an alkali metal such as lithium, sodium and potassium etc., an alkali earth metal such as magnesium, calcium etc., ammonium, an organic base and an amino acid; a salt with an inorganic acid such as hydrochloric acid, sulfuric acid, nitric acid, hydrobromic acid, phosphoric acid or hydroiodic acid etc., and an organic acid such as acetic acid, trifluoroacetic acid, citric acid, lactic acid, tartaric acid, oxalic acid, maleic acid, fumaric acid, mandelic acid, glutaric acid, malic acid, benzoic acid, phthalic acid, benzensulfonic acid, p-toluenesulfonic acid, methanesulfonic acid, ethanesulfonic acid, etc. Especially hydrochloric acid, phosphoric acid, tartaric acid or methane sulfonic acid is preferable. These salts can be prepared by a method usually carried out.


The compound (I) is not construed to be limited to a specific isomer but to include all possible isomers such as a keto-enol isomer, an imine-enamine isomer, a diastereoisomer, an optical isomer and a rotational isomer etc. For example, a compound (I) in which R2a is hydrogen includes a tautomer as follows;




embedded image


The compound (I) of the present invention can be prepared, for example, according to the non-patent literature 1 or a method described below; Preparation of an aminodihydrothiazine ring (1-1) or (1-2):




embedded image



(In the scheme above, at least one of R2b and R2c is hydrogen, R3b and R3d are each independently hydrogen, halogen, hydroxyl, optionally substituted lower alkyl, optionally substituted lower alkenyl, optionally substituted acyl, optionally substituted lower alkoxy, optionally substituted lower alkylthio, carboxy, optionally substituted lower alkoxycarbonyl, optionally substituted amino, optionally substituted carbamoyl, an optionally substituted carbocyclic group, or an optionally substituted heterocyclic group, and the other symbols are the same as described above.)


The 1st step: A Grignard reagent having a corresponding substituent of the objective compound such as vinyl magnesium chloride, vinyl magnesium bromide and propenyl magnesium bromide etc. is added to a compound a, which is commercially available or can be prepared by a known method, in a solvent such as ether, tetrahydrofuran etc. or a mixed solvent of ether-tetrahydrofuran etc., at −100° C. to 50° C., preferably −80° C. to 0° C. and the mixture is stirred for 0.2 to 24 hours, preferably 0.2 to 5 hours to give a compound b.


The 2nd step: To a compound b in an acid such as acetic acid, trifluoroacetic acid, hydrochloric acid, sulfuric acid etc. or a mixture thereof under the presence of a solvent such as toluene etc. or without a solvent, is added a substituted thiourea having a corresponding substituent of the objective compound such as thiourea, N-methylthiourea, N,N′-dimethylthiourea etc., and the mixture is stirred at −20° C. to 100° C., preferably 0° C. to 80° C., for 0.5 hours to 120 hours, preferably 1 hour to 72 hours to give a compound c,


The 3rd step: To a compound c in a solvent such as toluene etc. or without a solvent, is added an acid such as trifluoroacetic acid, methanesulfonic acid, trifluoromethanesulfonic acid, sulfuric acid etc. or a mixture thereof and reacted at −20° C. to 100° C., preferably 0° C. to 50° C., for 0.5 hours to 120 hours, preferably 1 hour to 72 hours to give a compound (I-2) when R2b is hydrogen or a compound (I-1) when R2c is hydrogen.


Preparation of an Aminodihydrothiazine Ring (I-3)



embedded image



(In the scheme above, L is a leaving group such as halogen or lower alkylsulfonyl etc. and the other symbols are the same as described above.)


The 1st step: Thiocyanate such as sodium thiocyanate or ammonium thiocyanate etc. is reacted with a compound d, which is commercially available or can be prepared by a known method, in a solvent such as toluene, chloroform, tetrahydrofuran etc. under the presence of water and an acid such as hydrochloric acid or sulfuric acid etc. at 0° C. to 150° C., preferably at 20° C. to 100° C. for 0.5 to 24 hours, preferably 1 to 12 hours to give a compound e.


The 2nd step: A reducing agent such as sodium borohydride etc. is added to and reacted with a compound e in a solvent such as tetrahydrofuran, methanol, ethanol, water etc. or a mixture of ethanol-water etc. under the presence of buffering agent such as sodium dihydrogen phosphate at −80° C. to 50° C., preferably at −20° C. to 20° C. for 0.1 to 24 hours, preferably 0.5 to 12 hours to give a compound f.


The 3rd step: A compound f is reacted with a halogenating agent such as thionyl chloride, phosphoryl chloride, carbon tetrachloride-triphenylphosphine etc, in a solvent such as toluene, dichloromethane etc. or without a solvent at −80° C. to 50° C., preferably at −20° C. to 20° C. for 0.1 to 24 hours, preferably 0.5 to 12 hours; or it is reacted with a sulfonating agent such as methanesulfonyl chloride, p-toluenesulfonyl chloride etc. in a solvent such as toluene, dichloromethane etc. under the presence of a base such as triethylamine etc. at −80° C. to 50° C., preferably at −20° C. to 20° C. for 0.1 to 24 hours, preferably 0.5 to 12 hours to give a compound g.


The 4th step: A compound g is reacted with ammonia or a primary amine such as methylamine etc. in a solvent such as methanol, ethanol, water etc. or a mixture of methanol-water etc. at −20° C. to 80° C., preferably at 0° C. to 40° C. for 0.5 to 48 hours, preferably 1 to 24 hours to give the compound (I-3).


Preparation of an Aminodihydrothiazine Ring (I-6) or an Aminotetrahydrothiazine Ring (I-7)



embedded image



(In the scheme above, at least one of R2b and R2c is hydrogen and the other symbols are the same as described above.)


The step: Thiourea or a substituted thiourea corresponding to the objective compound such as N-methyl thiourea, N,N-dimethylthiouers, N,N′-dimethylthiouera etc. is reacted with a compound o, which is commercially available or can be prepared by a known method, in a solvent such as ethanol, methanol, tetrahydrofuran, toluene etc. at −20° C. to 200° C., preferably at 0° C. to 150° C. for 0.5 to 200 hours, preferably 1 to 120 hours to give a compound p,


The 2nd step: A Grignard reagent corresponding to the objective compound such as methyl magnesium chloride, ethyl magnesium bromide and benzyl magnesium bromide etc, is added to a compound p in a solvent such as ether, tetrahydrofuran etc. or a mixed solvent thereof at −100° C. to 50° C., preferably −80° C. to 30° C. and the mixture is stirred for 0.2 to 24 hours, preferably 0.5 to 5 hours to give a compound q.


The step: To a compound q in a solvent such as toluene etc. or without a solvent, is added an acid such as trifluoroacetic acid, methanesulfonic acid, trifluoromethanesulfonic acid, sulfuric acid etc. or a mixture thereof and reacted at −20° C. to 100° C., preferably 0° C. to 50° C. for 0.5 hours to 200 hours, preferably 1 hour to 150 hours to give a compound (I-6) (R2c═H) or a compound (I-7) (R2b═H).


Preparation of an Aminodihydrothiazine Ring (I-8)




embedded image



(In the scheme, each symbol is the same as described above)


The 1st step: Ammonium chloride is added to a compound r which can be prepared by a known method in a solvent such as acetic acid etc. at 0° C. to 200° C., preferably 10° C. to 100° C. for 0.1 hours to 100 hours, preferably 0.5 hour to 24 hours to give a compound.


The 2nd step: A reducing agent such as lithium aluminium hydride; diisobutyl aluminium hydride etc. is reacted with a compound s in a solvent such as tetrahydrofuran, diethyl ether etc. at −80° C. to 150° C., preferably 0° C. to 100° C. for 0.1 hours to 24 hours, preferably 0.5 hour to 12 hours to give a compound t.


The 3rd step: Isothiocyanate corresponding to the objective compound such as 4-methoxybenzyl isothiocyanate, t-butyl isothiocyanate etc, or carbamoyl halide corresponding to the objective compound such as N,N-dimethyl thiocarbamoyl chloride, N,N-diethyl thiocarbamoyl chloride etc. is reacted with a compound t in a solvent such as toluene, chloroform, tetrahydrofuran etc. under the presence of a base such as diisopropylethylamine, triethylamine, pyridine, sodium hydroxide etc. or without a base at 0° C. to 150° C., preferably 20° C. to 100° C. for 0.5 hours to 120 hours, preferably 1 hour to 72 hours to give a compound u.


The 4th step: A halogenating agent such as thionyl chloride, phosphoryl oxychloride, carbon tetrachloride-triphenyl phosphine etc. is reacted with a compound u in a solvent such as acetonitrile, toluene, dichloromethane etc. at −80° C. to 50° C., preferably −20° C. to 20° C. for 0.1 hours to 24 hours, preferably 0.5 hour to 12 hours, or a sulfonylating agent such as methanesulfonyl chloride, p-toluenesulfonyl chloride is reacted with a compound u in a solvent such as toluene, dichloromethane etc. under the presence of a base such as triethylamine at −80° C. to 50° C., preferably −20° C. to 20° C. for 0.1 hours to 24 hours, preferably 0.5 hour to 12 hours. The resulting halogenated compound or sulfonate ester derivative is reacted with a base such as diisopropylethylamine, potassium carbonate, sodium bicarbonate, sodium hydride, sodium hydroxide etc. at 0° C. to 150° C., preferably 20° C. to 100° C. for 0.5 hours to 120 hours, preferably 1 hour to 72 hours to give a compound (I-8).


Preparation of an Acylamino Derivative (I-13) and/or (I-14)



embedded image



(In the scheme, R17 is optionally substituted lower alkyl, an optionally substituted carbocyclic group or an optionally substituted heterocyclic group and other symbols are the same as described above)


An acylating agent corresponding to the objective compound such as benzoyl chloride, 2-furoyl chloride, acetic anhydride etc, is reacted with a compound (I-12) in which R2b is hydrogen under the presence a solvent such as tetrahydrofuran, dichloromethane etc. or without a solvent and under the presence of a base such as pyridine or triethylamine etc. or without a solvent at −80° C. to 100° C. preferably −20° C. to 40° C. for 0.1 hours to 24 hours, preferably 1 hour to 12 hours, or a compound (I-12) is reacted with a carboxylic acid having a substituent corresponding to the objective compound such as amino acid or glycolic acid etc. in a solvent such as dimethylformamide, tetrahydrofuran, dichloromethane etc. under the presence of a condensation agent such as dicyclohexylcarbodiimide, carbonyldiimidazole etc. at −80° C. to 100° C., preferably −20° C. to 40° C. for 0.1 hours to 24 hours, preferably 1 hour to 12 hours to give a compound (I-13) and/or (I-14)(when R2a is hydrogen).


Preparation of a Carbamoyl Derivative (I-17)



embedded image



(In the scheme above, CONR18R19 is optionally substituted carbamoyl and the other symbols are the same as described above)


A compound (I-16) having a carboxyl group as a substituent on the ring A is reacted with a primary or secondary amine having a substituent corresponding to the objective compound (e.g., aniline, 2-aminopyridine, dimethylamine etc.) in a solvent such as dimethylformamide, tetrahydrofuran, dichloromethane etc. under the presence of a condensation agent such as dicyclohexylcarbodiimide, carbonyldiimidazole, dicyclohexylcarbodiimide-N-hydroxybenzotriazole etc. at −80° C. to 100° C., preferably −20° C. to 40° C. for 0.1 hours to 24 hours, preferably 1 hour to 12 hours to give a compound (I-17).


Preparation of an Acylamino Derivative (I-19)



embedded image



(In the scheme, NHR20 is optionally substituted amino, NR20COR21 is optionally substituted acylamino, optionally substituted ureido or carboxyamino having a substituent on the oxygen atom and the other symbols are the same as described above.)


A compound (I-18) having an optionally substituted amino group on the ring A is reacted with a reagent having a substituent corresponding to the objective compound such as acid chlorides, acid anhydrides, chlorocarbonate esters, isocyanates etc. under the presence of a solvent such as tetrahydrofuran, dichloromethane etc. or without a solvent under the presence of a base such as pyridine, triethylamine etc. or without a base at −80° C. to 100° C., preferably −20° C. to 40° C. for 0.1 hours to 24 hours, preferably 1 hour to 12 hours, or a compound (I-18) is reacted with a carboxylic acid having a substituent corresponding to the objective compound such as benzoic acid, 2-pyridinecarboxylic acid etc. in a solvent such as dimethylformamide, tetrahydrofuran, dichloromethane etc. under the presence of a condensation agent such as dicyclohexylcarbodiimide, carbonyldiimidazole, dicyclohexylcarbodiimide-N-hydroxybenzotriazote etc. at −80° C. to 100° C., preferably −20° C. to 40° C. for 0.1 hours to 24 hours, preferably 1 hour to 12 hours to give a compound (I-19).


Preparation of an Alkylamino Derivative (I-20)



embedded image



(In the scheme, NHR20 is optionally substituted amino and R22 is lower alkyl.)


A compound (I-18) having an amino group on the ring A is reacted with an aldehyde having a substituent corresponding to the objective compound such as benzaldehyde, pyridine-2-carboaldehyde etc. and a reducing agent such as sodium cyanoborohydride, sodium triacetoxyborohydride etc. in a solvent such as dichloromethane, tetrahydrofuran etc. under the presence of an acid such as acetic add etc. or without an acid at −80° C. to 100° C., preferably 0° C. to 40° C. for 0.5 hours to 150 hours, preferably 1 hour to 24 hours to give a compound (I-20),


Preparation of a Substituted Alkoxy Derivative (I-22)



embedded image



(In the scheme above, R23 is optionally substituted lower alkyl, an optionally substituted carbocyclic group or an optionally substituted heterocyclic group and the other symbols are the same as described above.)


A compound (I-21) having a hydroxy group on the ring A is reacted with an alkylating agent having a substituent corresponding to the objective compound such as benzyl chloride, methyl iodide etc. in a solvent such as dimethylformamide, tetrahydrofuran etc. under the presence of a base such as potassium carbonate, sodium hydroxide, sodium hydride etc. at −80° C. to 100° C., preferably 0° C. to 40° C. for 0.5 hours to 150 hours, preferably 1 hour to 24 hours, or a compound (I-18) is reacted with an alcohol such as 2-aminoethanol etc. in a solvent such as dimethylformamide, tetrahydrofuran etc. under the presence of a Mitsunobu reagent such as triphenylphosphine-azodicarboxylic acid diethyl ester etc. at −80° C. to 100° C., preferably 0° C. to 40° C. for 0.5 hours to 72 hours, preferably 1 hour to 24 hours to Rive a compound (I-22).


Introduction of a Substituent by Palladium Coupling



embedded image



(In the scheme above, Hal is halogen, G is optionally substituted lower alkenyl, optionally substituted alkynyl, optionally substituted alkoxycarbonyl, an optionally substituted carbocyclic, group or an optionally substituted heterocyclic group etc. and the other symbols are the same as described above)


A compound (I-23) having halogen as a substituent on the ring A is reacted with a compound having substituent corresponding to the objective compound (e.g., styrene, propargyl alcohol, aryl boronic acid, carbon monoxide etc.) in a solvent such as tetrahydrofuran, dimethylformamide, 1,2-dimethoxyethane, methanol etc. under the presence of a base such as triethylamine, sodium carbonate etc., a palladium catalyst such as palladium acetate, palladium chloride etc. and a ligand such as triphenylphosphine etc. and under irradiation of microwave or without the irradiation, at −80° C. to 150° C., preferably 0° C. to 100° C. for 0.5 hours to 72 hours, preferably 1 hour to 24 hours to give a compound (I-24).


Preparation of an Oxime Derivative (I-26)



embedded image



(In the scheme above. R24 is hydrogen, optionally substituted lower alkyl etc., R25 is hydrogen, optionally substituted lower alkyl, optionally substituted lower alkenyl, an optionally substituted carbocyclic group or an optionally substituted heterocyclic group etc., and the other symbols are the same as described above.)


A compound (I-25) having an acyl group as a substituent of the ring A is reacted with a hydroxylamine having a substituent corresponding to the objective compound such as hydroxylamine, methoxylamine, O-benzylhydroxylamine etc. or a salt thereof in a solvent such as methanol, ethanol etc. under the presence of an additive such as potassium acetate etc. or without an additive at −80° C. to 100° C., preferably 0° C. to 40° C. for 0.5 hours to 150 hours, preferably 1 hour to 72 hours to give a compound (I-26).


Coupling Reaction




embedded image



(In the scheme above. R26 is a substituent corresponding to each objective compound)


The 1st step:


A compound v is reacted with a reagent having a substituent corresponding to the objective compound such as acyl halide, acid anhydride, chlorocarbonate ester, isocyanate etc. (e.g., benzoyl chloride, 2-furoyl chloride, acetic anhydride, benzyl chloroformate, di-ten-butyl dicarbonate, phenyl isocyanate etc.) in a solvent such as tetrahydrofuran, dichloromethane, dimethylformamide etc. or without a solvent under the presence of a base such as pyridine, triethylamine etc. or without a base at −80° C. to 100° C., preferably −20° C. to 40° C. for 0.1 hours to 24 hours, preferably 1 hour to 12 hours, or a compound A is reacted with a carboxylic acid having a substituent corresponding to the objective compound such as benzoic acid, 2-pyridinecarboxylic acid etc. in a solvent such as dimethylformamide, tetrahydrofuran, dichloromethane, methanol etc. under the presence of a condensation agent such as dicyclohexylcarbodiimide, carbonyl diimidazole, dicyclohexylcarbodiimide-N-hydroxybenzotriazole, 4-(4,6-dimethoxy-1,3,5-triazine-2-yl)-4-methylmorpholinium chloride, 2-(7-aza-1H-benzotriazole-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate etc. at −80° C. to 100° C., preferably −20° C. to 40° C. for 0.1 hours to 24 hours, preferably 1 hour to 12 hours to give a compound w.


When the substituent R has a functional group which disturb the said reaction, it can be carried out by protecting the functional group with a suitable protecting group and then deprotecting it at a subsequent appropriate step.


The 2nd Step:


A compound w is reacted in a solvent such as methanol, ethanol, ether, tetrahydro furan, 1,4-dioxane, dichloromethane, ethyl acetate etc. containing trifluoroacetic acid etc. or in neat, or in neat trifluoroacetic acid at −30° C. to 100° C., preferably 0° C. to 90° C. for 0.5 to 12 hours to give a compound (I-27). Alternatively, the objective compound can be synthesized according to the method described in Protective Groups Organic Synthesis, Theodora W Green (John Wiley & Sons) etc.


Preparation of an Optically Active Isomer


1) Preparation of an Optically Active Isomer ae


For example, an optically active isomer ae, one embodiment of the compounds of the present invention, can be prepared according to the following scheme:




embedded image



(In the scheme above, R1 is optionally substituted lower alkyl, optionally substituted lower alkenyl or optionally substituted lower alkynyl; R27 is a chiral sulfoxide having optionally substituted lower alkyl, optionally substituted lower alkenyl, an optionally substituted carbocyclic group or an optionally substituted heterocyclic group, or a chiral auxiliary group such as α-methyl benzyl etc.; R3a, R3b, R3c, and R3d are each independently hydrogen, halogen, hydroxyl, optionally substituted lower alkyl, optionally substituted lower alkenyl, optionally substituted acyl, carboxy, optionally substituted lower alkoxycarbonyl, optionally substituted amino, optionally substituted carbamoyl, an optionally substituted carbocyclic group or an optionally substituted heterocyclic group; R28 is optionally substituted lower alkyl or optionally substituted lower alkenyl; R2a and R2b are each independently hydrogen, optionally substituted lower alkyl or optionally substituted acyl; and the other symbols are the same as described above.)


The compounds y and z above can be prepared by a method described in

  • (1) T. Fujisawa et al., Tetrahedron Lett., 37, 3881-3884 (1996),
  • (2) D. H. Hua et al, Sulfur Reports, vol. 21, pp. 211-239 (1999)
  • (3) Y. Koriyama et al., Tetrahedron, 58, 9621-9628 (2002), or
  • (4) T. Vilavan et al, Cuuent Organic Chemistry, 9, 1315-1392 (2005).


Alternatively, these compounds can be prepared by optical resolution of each intermediate or the final product, or according to methods described below. Examples of the optical resolution method include a separation of optical isomers using an optically active column, kinetic resolution by an enzyme reaction etc., crystallization of diastereomers by salt formulation using a chiral acid or chiral base, and a preferential crystallization etc,


The 1st step: Compound y can be obtained by reacting Compound x, which is commercially available or can be prepared by a known method, with a chiral reagent having a substituent corresponding to the objective compound such as α-methylbenzylamine, para-toluene, tert-butylsulfine amide etc, at 60° C. to 120° C., preferably 80° C. to 100° C. in a solvent such as ether, tetrahydrofuran, toluene, benzene etc. or a mixed solvent such as ether-tetrahydrofuran etc. for 0.5 to 24 hours, preferably 0.5 to 5 hours, in the presence of molecular sieves or magnesium sulfate etc., under continuous evaporation by Dean-Stark apparatus, or according to the method described in the above literatures.


The 2nd step: A compound z can be diastereo-selectively obtained by reacting an enolate of lithium, aluminium, zinc, titan etc, prepared Gonna reagent having a substituent corresponding to the objective compound such as acetate ester etc., which is commercially available or can be prepared by a known method, or ketenesilyl acetate prepared from a reagent having a substituent corresponding to the objective compound such as ethyl acetate etc. with a compound a in a solvent such as ether, tetrahydrofuran toluene, dichloromethane etc. or a mixed solvent such as ether-tetrahydrofuran etc. under the presence of a Lewis acid such as titanium tetrachloride, ether-trifluoroborane complex etc. or without a Lewis acid at −100° C. to 50° C., preferably −80° C. to −30° C. for 0.5 to 24 hours, preferably 0.5 to 5 hours. Alternatively, the compound z can be diastereo-selectively prepared by the method described in the literature (1) or (3).


The 3rd step: A compound z is reacted with a compound c in a solvent such as methanol, ethanol, ether, tetrahydrofuran, 1,4-dioxane, dichloromethane, ethyl acetate etc. containing hydrogen chloride, trifluoroacetic acid etc. or in neat trifluoroacetic acid at −30° C. to 100° C., preferably −10° C. to 90° C. for 0.5 to 12 hours, preferably 0.5 to 5 hours to give a compound aa.


The 4th step: A reducing agent such as borane-tetrahydrofuran complex, borane-dimethyl sulfoxide complex, borane-triethylamine complex, borane-pyridine complex etc. or ether- or tetrahydrofuran-solution thereof is reacted with a compound aa in a solvent such as ether, tetrahydrofuran, toluene etc. or a mixed solvent such as ether-tetrahydrofuran etc. at −30° C. to 30° C., preferably −10° C. to 20° C. for 0.5 to 12 hours, preferably 0.5 to 5 hours to give a compound ab.


The 5th step: Calcium carbonate or potassium carbonate etc. is added to a compound ab in a solvent such as dichloromethane, toluene etc. or a mixed solvent such as dichloromethane-water etc. and thiophosgene is added at −30° C. to 50° C., preferably −10° C. to 25° C. and the mixture is reacted for 0.5 to 12 hours, preferably 0.5 to 5 hours to give a compound ac.


The 6th step: Oxalyl chloride or thionyl chloride etc, and a catalytic amount of N,N-dimethylformamide are added to a compound ac in a solvent such as dichloromethane, tetrahydrofuran, toluene etc. at −30° C. to 50° C., preferably −10° C. to 20° C. and the mixture is reacted at 0° C. to 100° C., preferably 20° C. to 90° C. for 0.5 to 12 hours, preferably 0.5 to 5 hours to give a compound ad. Alternatively, it is obtained by a method described in Comprehensive Organic Transformations, Richard C Larock (Mcgraw-Hill).


The 7th step: 15% to 30% Ammonia water or a reagent having a substituent corresponding to the objective compound such as tert-butylamine etc. is added to a compound ad in a solvent such as ethyl acetate, dichloromethane, tetrahydrofuran, toluene etc. at −30° C. to 50° C., preferably −10° C. to 30° C. and the mixture is reacted at −10″C to 30° C., preferably 0° C. to 30° C. for 0.5 to 72 hours to give a compound ae-i or a compound ae-ii.


When R2a and/or R2b is hydrogen in the resulting compound ae-i or ae-ii, a substituent of the objective compound, R2a and/or R2b, may be further introduced by a conventional method if it is necessary.


1′) Method for Preparing an Optically Active Isomer Method B


An optically active compound ah of the present invention can be also prepared by a method below:




embedded image



(In the scheme, the symbols are the same as described above.)


The 1st step to the 4th step: the same as described in 1) above.


The 5th step: Isothiocyanate having a protecting group which is commercially available or can be prepared by a known method is added to a compound ab in a solvent such as dichloromethane, toluene, acetone etc, or a mixed solvent at −30° C. to 50° C., preferably −10° C. to 2.5° C. and the mixture is reacted for 0.5 to 12 hours, preferably 0.5 to 5 hours to give a compound ag.


The 6th step: Oxalyl chloride or thionyl chloride etc. and a catalytic amount of N,N-dimethylformamide are added to a compound ag in a solvent such as dichloromethane, tetrahydrofuran, toluene etc. at −30° C. to 50° C., preferably −10° C. to 25° C., or 1-chloro-N,N-2-trimethyl-1-propenenylamine is added to a compound ag, and reacted at 0° C. to 100° C., preferably 20° C. to 90° C. for 0.5 to 72 hours to give a compound ah-i or ah-ii.


2) Introduction of R3a and R3b


An optically active compound ae-iii or ae-iv of the present invention can be also prepared by introducing R3a and R3b as shown below:




embedded image



(In the scheme above, each symbol is the same as described above)


When preparing a compound ae-iii ae-iv in which R3a and R3b are substituted on the carbon atom next to S atom, a compound z is processed through the 3rd and 4th steps in place of the 3rd and 4th steps of 1) described above, and R3a and R3b are introduced in advance.


The 3rd step: A Grignard reagent having a substituent corresponding to the objective compound such as methyl magnesium chloride, ethyl magnesium bromide etc. is added to a compound z in a solvent such as ether, tetrahydrofuran etc. or a mixed solvent such as ether-tetrahydrofuran etc. at −100° C. to 50° C., preferably −80° C. to 30° C., or a compound z is convened to Weinreb Amide and reacted with a Grignard reagent having a substituent corresponding to the objective compound such as R3aMgBr, R3bMgBr. The reaction mixture is reacted for 0.2 to 24 hours, preferably 0.2 to 5 hours to give a compound aa′.


The 4th Step: A compound aa′ is reacted in a solvent such as methanol, ethanol, ether, tetrahydrofuran, 1,4-dioxane, dichloromethane, ethyl acetate etc. containing hydrogen chloride, trifluoroacetic acid etc. or n neat trifluoroacetic acid at −30° C. to 100° C., preferably −10° C. to 90° C. for 0.5 to 12 hours, preferably 0.5 to 5 hours to give a compound ab′.


The compound ab′ is processed in the same reactions as the 5th to 7th steps of 1) above to give the objective compound ae-iii or ae-iv.


When the substituent L of a compound ad′ is eliminated to give a compound ad″ shown below, the objective compound ae′-iii or ae′-iv is obtained by processing the compound ad″ it through the 7th step in place of the 7th step described in 1) above.




embedded image



The 7th step: A compound ad″ is dissolved in conc. sulfuric acid, trifluoroacetic acid, trifluoromethanesulfonic acid etc. and reacted at −30° C. to 100° C., preferably −10° C. to 40° C. for 0.1 to 12 hours, preferably 0.5 to 5 hours to give a compound ae′.


3) Conversion of a Substituent (1)


A preparation of a compound af-1 by conversion of the substituent is illustrated below:




embedded image



(In the scheme, R8a and R8b are an amino-protecting group, and the other symbols are the same as described above.)


Trisdibenzylideneacetonedipalladium, palladium acetate, palladium(0) prepared in situ etc. and a phosphine ligand such as tri-tert-butylphosphine, dicyclohexylbiphenylphosphine etc. are added to a compound ae-1 in a solvent such as tetrahydrofuran, toluene, xylene etc. and further a reagent having a substituent corresponding to the objective compound such as lithium hexamethylenedisilazide, benzophenonimine etc. is added thereto at −10° C. to 30° C., then the reaction mixture is reacted at 30° C. to 120° C., preferably 50° C. to 100° C. for 0.5 to 48 hours, preferably 3 to 20 hours to give a compound af-1.


Any amino-protecting group which is deprotected by a method described in Protective Groups in Organic Synthesis, Theodora W Green (John Wiley & Sons) etc. can be used and examples of the protecting group include lower alkoxycarbonyl, lower alkenyloxycarbonyl, acyl, methane-sulfonyl), trifluoromethanesulfonyl and toluenesulfonyl etc.


4) Conversion of a Substituent (2)


A preparation of a compound af-2 by conversion of the substituent is illustrated below:




embedded image



(In the scheme, each symbol is the same as described above.)


A catalyst of catalytic reduction such as 10% palladium-carbon etc, is added to a compound ae-2 in a solvent such as tetrahydrofuran, ethyl acetate, methanol etc. and it is reacted under the pressure of normal to 5 atom, preferably normal to 2 atom of hydrogen atmosphere at 30° C. to 120° C., preferably 50° C. to 80° C. for 0.5 to 48 hours, preferably 6 to 20 hours to give a compound af-2. Alternatively, the compound af-2 is obtained by a method described in Comprehensive Organic Transformations, Richard C Larock (Mcgraw-Hill).


5) Conversion of a Substituent (3)


A preparation of a compound af-3 by conversion of the substituent is illustrated below:




embedded image



(In the scheme, R9 is hydroxyl, optionally substituted lower alkyl, optionally substituted lower alkoxy, optionally substituted lower alkylthio, optionally substituted lower alkyl amino, optionally substituted aromatic carbocyclyloxy, optionally substituted heterocyclyloxy, optionally substituted aromatic carbocyclylthio, optionally substituted heterocyclylthio, optionally substituted carbocyclylamino, optionally substituted heterocyclylamino, cyano azide, an optionally substituted carbocyclic group, an optionally substituted heterocyclic group, optionally substituted carbamoyl etc, and the other symbols are the same as described above.)


A reagent having a substituent corresponding to the objective compound such as ethanol, methanthiol, dimethylamine etc. is added to a compound ae-3 in a solvent such as tetrahydrofuran, ethanol etc, under the presence of a base such as sodium methoxide, potassium tert-butoxide, sodium hydroxide, sodium hydride etc. or without a base at −10° C. to 50° C. and it is reacted for 0.5 to 12 hours, preferably 1 to 8 hours to give a compound af-3. If necessary, a coupling reaction may be carried out in the same manner as the method for preparing a compound (I-19) described above.


In every step described above, f a starting compound has a functional group which disturb the reaction (e.g., hydroxyl, mercapto, amino, formyl, carbonyl, carboxyl etc.), it is recommended to protect the functional group and deprotect it at a subsequent appropriate step with a method described in Protective Groups in Organic Synthesis, Theodora W Green (John Wiley & Sons) etc.


Further the order of steps may be changed and each reaction intermediate may be isolated and used in the subsequent step.


Examples of a preferable compound in the present invention include the followings:


In a formula (I′)




embedded image



1) a compound in which the ring A′ is phenyl or a nitrogen-containing aromatic heterocyclic group (hereinafter called a compound in which the ring A′ is A′1),


a compound in which the ring A′ is benzene, pyridine, indole, benzisoxazole, benzopyrazole, benzofuran, benzothiophene, benzodioxole, or dihydrobenzodioxolane (hereinafter called a compound in which the ring A′ is A′2),


a compound in which the ring A′ is benzene (hereinafter called a compound in which the ring A′ is A′3),


a compound in which the ring A′ is pyridine (hereinafter called a compound in which the ring A′ is A′4)


2) a compound in which R1 is optionally substituted lower alkyl (hereinafter called a compound in which R1 is R1-1),


a compound in which R1 is methyl (hereinafter called a compound in which R1 is R1-2),


3) a compound in which R2a and R2-b are each independently hydrogen, lower alkyl or acyl(hereinafter called a compound in which R2a and R2b are R2-1),


a compound in which both of R2a and R2b are hydrogens (hereinafter called a compound in which R2a and R2b are R2-2),


4) a compound in which R3a, R3b, R3c and R3d are each independently hydrogen, halogen, hydroxyl, lower alkyl or amino (hereinafter called a compound in which R3a, R3b, R3c and R3d are R3-1),


a compound in which R3a and R3b, or R3c and R3d taken together foil cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl together (hereinafter called a compound in R3a, R3b, R3c and R3d are R3-2)


a compound in which R3a and R3b, or R3c and R3d are the same substituent selected compound in which R3a, R3b, R3c and R3d from halogen and lower alkyl (hereinafter called a compound in which R3a, R3b, R3c and R3d are R3-3),


a compound in which all of R3a, R3b, R3c and R3d are hydrogens (hereinafter called a compound in which R3a, R3b, R3c and R3d are R3-4),


5) a compound in which n is 0 to 2, R4 is each independently halogen, lower alkoxy, lower alkylamino, lower alkylthio, oxo or lower alkylenedioxy (hereinafter called a compound in which R4 is R4-1),


a compound in which n is 0 to 2, R4 is each independently halogen (hereinafter called a compound in which R4 is R4-2),


6) a compound in which G is (ii), (iv), (v), (x), (xiii) or (xiv) above (hereinafter called a compound in which G is G1),


a compound in which G is (ii′), (ii″), (iv′), (v′), (x′), (xiii′ (xiv′) above (hereinafter called a compound in which G is G2),


a compound in which G is (ii′), (ii′), (iv′), (v′), (x′), (xiii′) or (xiv′) above, and the ring B is optionally substituted pyridyl, optionally substituted pyrazinyl, optionally substituted thiazolyl, optionally substituted isoxazolyl, optionally substituted benzothiazolyl, optionally substituted thiazolopyridyl, optionally substituted quinolyl, optionally substituted isoquinolyl or optionally substituted naphthylidinyl, optionally substituted quinazolinyl, or optionally substituted pyridopyrimidinyl (hereinafter called a compound in which G is G3),


a compound in which G is (ii′) above (hereinafter called a compound in which G is G4),


a compound in which a combination of the ring A′, R1, R2a and R2b, R3a, and R3d, n and R4, and G is as follows;


(A′1, R1-1, R2-1, R3-1, R4-1, G1), (A′1, R1-1, R2-1, R3-1, R4-1, G2), (A′1, R1-1, R2-1, R3-1, R4-1, G3), (A′1, R-1, R2-1, R3-1, R4-1, G4), (A′1, R1-1, R2-1, R3-1, R4-2, G1), (A′1, R1-1, R2-1, R3-1, R 4-2, G2), (A′1, R1-1, R2-1, R3-1, R4-2, G3), (A′1, R1-1, R2-1, R3-1, R4-2, G4), (A′1, R1-1, R2-1, R3-2, R4-1, G1), (A′1, R1-1, R2-1, R3-2, R4-1, G2), (A′1, R1-1, R2-1, R3-2, R4-1, G3), (A′1, R1-1, R2-1, R3-2, R4-1, G4), (A′1, R1-1, R2-1, R3-2, R4-2, G1), (A′1, R1-1, R2-1, R3-2, R4-2, G2), (A′1, R1-1, R 2-1, R3-2, R4-2, G3), (A′1, R1-1, R2-1, R3-2, R4-2, G4), (A′1, R1-1, R2-1, R3-3, R4-1, G1), (A′1, R1-1, R2-1, R3-3, R4-1, G2), (A′1, R1-1, R2-1, R3-3, R4-1, G3), (A′1, R1-1, R2-1, R3-3, R4-1, G4), (A′1, R1-1, R2-1, R3-3, R4-2, G1), (A′1, R1-1, R2-1, R3-3, R4-2, G2), (A′1, R1-1, R2-1, R3-3, R4-2, G3), (A′1, R1-1, R2-1, R3-3, R4-2, G4), (A′1, R1-1, R2-1, R3-4, R4-1, G1), (A′1, R1-1, R2-1, R3-4, R4-1, G 2), (A′1, R1-1, R2-1, R3-4, R4-1, G3), (A′1, R1-1, R2-1, R3-4, R4-1, G4), (A′1, R1-1, R2-1, R3-4, R4-2, G1), (A′1, R1-1, R2-1, R3-4, R4-2, G2), (A′1, R1-1, R2-1, R3-4, R4-2, G3), (A′1, R1-1, R2-1, R3-4, R4-2, G4), (A′1, R1-1, R2-2, R3-1, R4-1, G1), (A′1, R1-1, R2-2, R3-1, R4-1, G2), (A′1, R1-1, R2-2, R 3-1, R4-1, G3), (A′1, R1-1, R2-2, R3-1, R4-1, G4), (A′1, R1-1, R2-2, R3-1, R4-2, G1), (A′1, R1-1, R2-2, R3-1, R4-2, G2), (A′1, R1-1, R2-2, R3-1, R4-2, G3), (A′1, R1-1, R2-2, R3-1, R4-2, G4), (A′1, R1-1, R2-2, R3-2, R4-1, G1), (A′1, R1-1, R2-2, R3-2, R4-1, G2), (A′1, R1-1, R2-2, R3-2, R4-1, G3), (A′1, R 1-1, R2-2, R3-2, R4-1, G4), (A′1, R1-1, R2-2, R3-2, R4-2, G1), (A′1, R1-1, R2-2, R3-2, R4-2, G2), (A′1, R1-1, R2-2, R3-2, R4-2, G3), (A′1, R1-1, R2-2, R3-2, R4-2, G4), (A′1, R1-1, R2-2, R3-3, R4-1, G1), (A′1, R1-1, R2-2, R3-3, R4-1, G2), (A′1, R1-1, R2-2, R3-3, R4-1, G3), (A′1, R1-1, R2-2, R3-3, R4-1, G4), (A′1, R1-1, R2-2, R3-3, R4-2, G1), (A′1, R1-1, R2-2, R3-3, R4-2, G2), (A′1, R1-1, R2-2, R3-3, R4-2, G3), (A′1, R1-1, R2-2, R3-3, R4-2, G4), (A′1, R1-1, R2-2, R3-4, R4-1, G1), (A′1, R1-1, R2-2, R3-4, R4-1, G2), (A′1, R1-1, R2-2, R3-4, R4-1, G3), (A′1, R1-1, R2-2, R3-4, R4-1, G4), (A′1, R1-1, R2-2, R3-4, R4-2, G1), (A′1, R1-1, R2-2, R3-4, R4-2, G2), (A′1, R1-1, R2-2, R3-4, R4-2, G3), (A′1, R1-1, R 2-2, R3-4, R4-2, G4), (A′1, R1-2, R2-1, R3-1, R4-1, G1), (A′1, R1-2, R2-1, R3-1, R4-1, G2), (A′1, R1-2, R2-1, R3-1, R4-1, G3), (A′1, R1-2, R2-1, R3-1, R4-1, G4), (A′1, R1-2, R2-1, R3-1, R4-2, G1), (A′1, R1-2, R2-1, R3-1, R4-2, G2), (A′1, R1-2, R2-1, R3-1, R4-2, G3), (A′1, R1-2, R2-1, R3-1, R4-2, G4), (A′1, R1-2, R2-11R3-2, R4-1, G1), (A′1, R1-2, R2-1, R3-2, R4-1, G2), (A′1, R1-2, R2-1, R3-2, R4-1, G3), (A′1, R1-2, R2-1, R3-2, R4-1, G4), (A′1, R1-2, R2-1, R3-2, R4-2, G1), (A′1, R1-2, R2-1, R3-2, R4-2, G2), (A′1, R1-2, R2-1, R3-2, R4-2, G3), (A′1, R1-2, R2-1, R3-2, R4-2, G4), (A′1, R1-2, R2-1, R3-3, R4-1, G1), (A′1, R1-2, R2-1, R3-3, R4-1, G2), (A′1, R1-2, R2-1, R3-3, R4-1, G3), (A′1, R1-2, R2-1, R 3-3, R4-1, G4), (A′1, R1-2, R2-1, R3-3, R4-2, G1), (A′1, R1-2, R2-1, R3-3, R4-2, G2), (A′1, R1-2, R2-1, R3-3, R4-2, G3), (A′1, R1-2, R2-1, R3-3, R4-2, G4), (A′1, R1-2, R2-1, R3-4, R4-1, G1), (A′1, R1-2, R2-1, R3-4, R4-1, G2), (A′1, R1-2, R2-1, R3-4, R4-1, G3), (A′1, R1-2, R2-1, R3-4, R4-1, G4), (A′1, R 1-2, R2-1, R3-4, R4-2, G1), (A′1, R1-2, R2-1, R3-4, R4-2, G2), (A′1, R1-2, R2-1, R3-4, R4-2, G3), (A′1, R1-2, R2-1, R3-4, R4-2, G4), (A′1, R1-2, R2-2, R3-1, R4-1, G1), (A′1, R1-2, R2-2, R3-1, R4-1, G2), (A′1, R1-2, R2-2, R3-1, R4-1, G3), (A′1, R1-2, R2-2, R3-1, R4-1, G4), (A′1, R1-2, R2-2, R3-1, R4-2, G1), (A′1, R1-2, R2-2, R3-1, R4-2, G2), (A′1, R1-2, R2-2, R3-1, R4-2, G3), (A′1, R1-2, R2-2, R3-1, R 4-2, G4), (A′1, R1-2, R2-2, R3-2, R4-1, G1), (A′1, R1-2, R2-2, R3-2, R4-1, G2), (A′1, R1-2, R2-2, R3-2, R4-1, G3), (A′1, R1-2, R2-2, R3-2, R4-1, G4), (A′1, R1-2, R2-2, R3-2, R4-2, G1), (A′1, R1-2, R2-2, R3-2, R4-2, G2), (A′1, R1-2, R2-2, R3-2, R4-2, G3), (A′1, R1-2, R2-2, R3-2, R4-2, G4), (A′1, R1-2, R2-2, R3-3, R4-1, G1), (A′1, R1-2, R2-2, R3-3, R4-1, G2), (A′1, R1-2, R2-2, R3-3, R4-1, G3), (A′1, R1-2, R2-2, R3-3, R4-1, G4), (A′1, R1-2, R2-2, R3-3, R4-2, G1), (A′1, R1-2, R2-2, R3-3, R4-2, G2), (A′1, R1-2, R2-2, R3-3, R4-2, G3), (A′1, R1-2, R2-2, R3-3, R4-2, G4), (A′1, R1-2, R2-2, R3-4, R4-1, G1), (A′1, R1-2, R2-2, R3-4, R4-1, G2), (A′1, R1-2, R2-2, R3-4, R4-1, G3), (A′1, R1-2, R2-2, R3-4, R4-1, G4), (A′1, R1-2, R2-2, R3-4, R4-2, G1), (A′1, R1-2, R2-2, R3-4, R4-2, G2), (A′1, R1-2, R2-2, R3-4, R4-2, G3), (A′1, R1-2, R2-2, R3-4, R4-2, G4),


(A′2, R1-1, R2-1, R3-1, R4-1, G1), (A′2, R1-1, R2-1, R3-1, R4-1, G2), (A′2, R1-1, R2-1, R3-1, R4-1, G3), (A′2, R1-1, R2-1, R3-1, R4-1, G4), (A′2, R1-1, R2-1, R3-1, R4-2, G1), (A′2, R1-1R2-1, R3-1, R 4-2, G2), (A′2, R1-1, R2-1, R3-1, R4-2, G3), (A′2, R1-1, R2-1, R3-1, R4-2, G4), (A′2, R1-1, R2-1, R3-2, R4-1, G11), (A′2, R1-1, R2-1, R3-2, R4-1, G2), (A′2, R1-1, R2-1, R3-2, R4-1, G3), (A′2, R1-1, R2-1, R3-2, R4-1, G4), (A′2, R1-1, R2-1, R3-2, R4-2, G1), (A′2, R1-1, R2-1, R3-2, R4-2, G2), (A′2, R1-1, R 2-1, R3-2, R4-2, G3), (A′2, R1-1, R2-1, R3-2, R4-2, G4), (A′2, R1-1, R2-1, R3-3, R4-1, G1), (A′2, R1-1, R2-1, R3-3, R4-1, G2), (A′2, R1-1, R2-1, R3-3, R4-1, G3), (A2, R1-1, R2-11, R3-3, R4-1, G4), (A′2, R1-1, R2-1, R3-3, R4-2, G1), (A′2, R1-1, R2-1, R3-3, R4-2, G2), (A′2, R1-1, R2-1, R3-3, R4-2, G3), (A′2, R1-1, R1-1, R3-3, R4-2, G4), (A′2, R1-1, R2-1, R3-4, R4-1, G11), (A′2, R1-1, R2-1, R3-4, R4-1, G 2), (A′2, R1-1, R2-1, R3-4, R4-1, G3), (A′2, R1-1, R2-1, R3-4, R4-1, G4), (A′2, R1-1, R2-1, R3-4, R4-2, G1), (A′2, R1-1, R2-1, R3-4, R4-2, G2), (A′2, R1-1, R2-1, R3-4, R4-2, G3), (A′2, R-1, R2-1, R3-4, R4-2, G4), (A′2, R1-1, R2-2, R3-1, R4-1, G1), (A′2, R1-1, R2-2, R3-1, R4-1, G2), (A′2, R1-1, R2-2, R3-1, R4-1 G3), (A′2, R1-1, R2-2, R3-1, R4-1, G4), (A′2, R1-1, R2-2, R3-1, R4-2, G1), (A′2, R1-1, R2-2, R3-1, R4-2, G2), (A′2, R1-1, R2-2, R3-1, R4-2, G3), (A′2, R1-1, R2-2, R3-1, R4-2, G4), (A′2, R1-1, R2-2, R3-2, R4-1, G 1), (A′2, R1-1, R2-2, R3-2, R4-1, G2), (A′2, R1-1, R2-2, R3-2, R4-1, G3), (A′2, R 1-1, R2-2, R3-2, R4-1, G4), (A′2, R1-1, R2-2, R3-2, R4-2, G1), (A′2, R1-1, R2-2, R3-2, R4-2, G2), (A′2, R1-1, R2-2, R3-2, R4-2, G3), (A′2, R1-1, R2-2, R3-2, R4-2, G4), (A′2, R1-1, R2-2, R3-3, R4-1, G1), (A′2, R1-1, R2-2, R3-3, R4-1, G2), (A′2, R1-1, R2-2, R3-3, R4-1, G3), (A′2, R1-1, R2-2, R3-3, R4-1, G4), (A′2, R1-1, R2-2, R3-3, R4-2, G1), (A′2, R1-1, R2-2, R3-3, R4-2, G2), (A′2, R1-1, R2-2, R3-3, R 4-2, G3), (A′2, R1-1, R2-2, R3-3, R4-2, G4)(A′2, R1-1, R2-2, R3-4, R4-1, G1), (A′2, R1-1, R2-2, R3-4, R4-1, G2), (A′2, R1-1, R2-2, R3-4, R4-1, G3), (A2, R1-1, R2-2, R3-4, R4-1, G4), (A′2, R1-1, R2-2, R3-4, R4-2, G1), (A′2, R1-1, R2-2, R3-4, R4-2, G2), (A′2, R1-1, R2-2, R3-4, R4-2, G3), (A2, R1-1, R 2-2, R3-4, R4-2, G4), (A′2, R1-2, R2-1, R3-1, R4-1, G1), (A′2, R1-2, R2-1, R3-1, R4-1, G2), (A′2, R1-2, R2-1, R3-1, R4-1, G3), (A′2, R1-2, R2-1, R3-1, R4-1, G4), (A′2, R1-2, R2-1, R3-1, R4-2, G1), (A′2, R1-2, R2-1, R3-1, R4-2, G2), (A′2, R1-2, R2-1, R3-1, R4-2, G3), (A′2, R1-2, R2-1, R3-1, R4-2, G4), (A′2, R1-2, R2-1, R3-2, R4-1, G1), (A′2, R1-2, R2-1, R3-2, R4-1, G2), (A′2, R1-2, R2-1, R3-2, R4-1, G3), (A′2, R1-2, R2-1, R3-2, R4-1, G4), (A′2, R1-2, R2-1, R3-2, R4-2, G1), (A′2, R1-2, R2-1, R3-2, R4-2, G2), (A′2, R1-2, R2-1, R3-2, R4-2, G3), (A′2, R1-2, R2-1, R3-2, R4-2, G4), (A′2, R1-2, R2-1, R3-3, R-4-1, G1), (A′2, R1-2, R2-1, R3-3, R4-1, G2), (A′2, R1-2, R2-1, R3-3, R4-1, G3), (A′2, R1-2, R2-1, R 3-3, R4-1, G4), (A′2, R1-2, R2-1, R3-3, R4-2, G1), (A′2, R1-2, R2-1, R3-3, R4-2, G2), (A′2, R1-2, R2-1, R3-3, R4-2, G3), (A′2, R1-2, R2-1, R3-3, R4-2, G4), (A′2, R1-2, R2-1, R3-4, R4-1, G1), (A′2, R1-2, R2-1, R3-4, R4-1, G2), (A′2, R1-2, R2-1, R3-1, R4-1, G3), (A′2, R1-2, R2-1, R3-4, R4-1, G4), (A′2, R 1-2, R2-1, R3-4, R4-2, G1), (A′2, R1-2, R2-1, R3-4, R4-2, G2), (A′2, R1-2, R2-1, R3-4, R4-2, G3), (A′2, R1-2, R2-1, R3-4, R4-2, G4), (A′2, R1-2, R2-2, R3-1, R4-1, G1), (A′2, R1-2, R2-2, R3-1, R4-1, G2), (A′2, R1-2, R2-2, R3-1, R4-1, G3), (A′2, R1-2, R2-2, R3-1, R4-1, G4), (A′2, R1-2, R2-2, R3-1, R4-2, G1), (A′2, R1-2, R2-2, R3-1, R4-2, G2), (A′2, R1-2, R2-2, R3-1, R4-2, G3), (A′2, R1-2, R2-2, R3-1, R 4-2, G4), (A′2, R1-2, R2-2, R3-2, R4-1, G1), (A′2, R1-2, R2-2, R3-2, R4-1, G2), (A′2, R1-2, R2-2, R3-2, R4-1, G3), (A′2, R1-2, R2-2, R3-2, R4-1, G4), (A′2, R1-2, R2-2, R3-2, R4-2, G1), (A′2, R1-2, R2-2, R3-2, R4-2, G2), (A′2, R1-2, R2-2, R3-2, R4-2, G3), (A′2, R1-2, R2-2, R3-2, R4-2, G4), (A′2, R1-2, R 2-2, R3-3, R4-1, G1), (A′2, R1-2, R2-2, R3-3, R4-1, G2), (A′2, R1-2, R2-2, R3-3, R4-1, G3), (A′2, R1-2, R2-2, R3-3, R4-1, G4), (A′2, R1-2, R2-2, R3-3, R4-2, G1), (A′2, R1-2, R2-2, R3-3, R4-2, G2), (A′2, R1-2, R2-2, R3-3, R4-2, G3), (A′2, R1-2, R2-2, R3-3, R4-2, G4), (A2, R1-2, R2-2, R3-4, R4-1, G1), (A′2, R1-2, R2-2, R3-4, R4-1, G2), (A′2, R1-2, R2-2, R3-4, R4-1, G3), (A′2, R1-2, R2-2, R3-4, R4-1, G 4), (A′2, R1-2, R2-2, R3-4, R4-2, G1), (A2, R1-2, R2-2, R3-4, R4-2, G2), (A′2, R1-2, R2-2, R3-4, R4-2, G3), (A′2, R1-2, R2-2, R3-4, R4-2, G4),


(A′3, R1-1, R2-1, R3-1, R4-1, G 1), (A′3, R1-1, R2-1, R3-1, R4-1, G2), (A′3, R1-1, R2-1, R3-1, R4-1, G3), (A′3, R1-1, R2-1, R3-1, R4-1, G4), (A′3, R1-1, R2-1, R3-1, R4-2, G1), (A′3, R1-1, R2-1, R3-1, R 4-2, G2), (A′3, R1-1, R2-1, R3-1, R4-2, G3), (A′3, R1-1, R2-1, R3-1, R4-2, G4), (A′3, R1-1, R2-1, R3-2, R4-1, G1), (A′3, R1-1R2-1, R3-2, R4-1, G2), (A′3, R1-1, R2-1, R3-2, R4-1, G3), (A′3, R1-1, R2-1, R3-2, R4-1, G4), (A′3, R1-1, R2-1, R3-2, R4-2, G1), (A′3, R1-1, R2-1, R3-2, R4-2, G2), (A′3, R1-1, R 2-1, R3-2, R4-2, G3), (A′3, R1-1, R2-1, R3-2, R4-2, G4)(A′3, R1-1, R2-1, R3-3, R4-1, G1), (A′3, R1-1, R2-1, R3-3, R4-1, G2), (A′3, R1-1, R2-1, R3-3, R4-1, G3), (A′3, R1-1, R2-1, R3-3, R4-1, G4), (A′3, R1-1, R2-1, R3-3, R4-2, G1), (A′3, R1-1, R2-1, R3-3, R4-2, G2), (A′3, R1-1, R2-1, R3-3, R4-2, G3), (A′3, R1-1, R2-1, R3-3, R4-2, G4), (A′3, R1-1, R2-1, R3-4, R4-1, G1)(A′3, R1-1, R2-1, R3-4, R4-1, G 2), (A′3, R1-1, R2-1, R3-4, R4-1, G3), (A′3, R1-1, R2-1, R3-4, R4-1, G4), (A′3, R1-1, R2-1, R3-4, R4-2, G1), (A′3, R1-1, R2-1, R3-4, R4-2, G2), (A′3, R1-1, R2-1, R3-4, R4-2, G3), (A′3, R1-1, R2-1, R3-4, R4-2, G4), (A′3, R1-1, R2-2, R3-1, R4-1, G1), (A′3, R1-1, R2-2, R3-1, R4-1, G2), (A′3, R1-1, R2-2, R 3-1, R4-1, G3), (A′3, R1-1, R2-2, R3-1, R4-1, G4), (A′3, R1-1, R2-2, R3-1, R4-2, G1), (A′3, R1-1, R2-2, R3-1, R4-2, G2), (A′3, R1-1, R2-2, R3-1, R4-2, G3), (A′3, R1-1, R2-2, R3-1, R4-2, G4), (A′3, R1-1, R2-2, R3-2, R4-1, G1), (A′3, R1-1, R2-2, R3-2, R4-1, G2), (A′3, R1-1, R2-2, R3-2, R4-1, G3), (A′3, R 1-1, R2-2, R3-2, R4-1, G4), (A′3, R1-1, R2-2, R3-2, R4-2, G1), (A′3, R1-1, R2-2, R3-2, R4-2, G2), (A′3, R1-1, R2-2, R3-2, R4-2, G3), (A′3, R1-1, R2-2, R3-2, R4-2, G4), (A′3, R1-1, R2-2, R3-3, R4-1, G1), (A′3, R1-1, R2-2, R3-3, R4-1, G2), (A′3, R1-1, R2-2, R3-3, R4-1, G3), (A′3, R1-1, R2-2, R3-3, R4-1, G4), (A′3, R1-1, R2-2, R3-3, R4-2, G1), (A′3, R1-1, R2-2, R3-3, R4-2, G2), (A′3, R1-1, R2-2, R3-3, R 4-2, G3), (A′3, R1-1, R2-2, R3-3, R4-2, G4), (A′3, R1-1, R2-2, R3-4, R4-1, G1), (A′3, R1-1, R2-2, R3-4, R4-1, G2), (A′3, R1-1, R2-2, R3-4, R4-1, G3), (A′3, R1-1, R2-2, R3-4, R4-1, G4), (A′3, R1-1, R2-2, R3-4, R4-2, G1), (A′3, R1-1, R2-2, R3-4, R4-2, G2), (A′3, R1-1, R2-2, R3-4, R4-2, G3)(A′3, R1-1, R 2-2, R3-4, R4-2, G4), (A′3, R1-2, R2-1, R3-1, R4-1, G1), (A′3, R1-2, R2-1, R3-1, R4-1, G2), (A′3, R1-2, R2-1, R3-1, R4-1, G3), (A′3, R1-2, R2-1, R3-1, R4-1, G4), (A′3, R1-2, R2-1, R3-1, R4-2, G1), (A′3, R1-2, R2-1, R3-1, R4-2, G2), (A′3, R1-2, R2-1, R3-1, R4-2, G3), (A′3, R1-2, R2-1, R3-1, R4-2, G4), (A′3, R1-2, R2-1, R3-2, R4-1, G1), (A′3, R1-2, R2-1, R3-2, R4-1, G2), (A′3, R1-2, R2-1, R3-2, R4-1, G 3), (A′3, R1-2, R2-1, R3-2, R4-1, G4), (A′3, R1-2, R2-1, R3-2, R4-2, G1), (A′3, R1-2, R2-1R3-2, R4-2, G2), (A′3, R1-2, R2-1, R3-2, R4-2, G3), (A′3, R1-2, R2-1, R3-2, R4-2, G4), (A′3, R1-2, R2-1, R3-3, R4-1, G1), (A′3, R1-2, R2-1, R3-3, R4-1, G2), (A′3, R1-2, R2-1, R3-3, R4-1, G3), (A′3, R1-2, R2-1, R 3-3, R4-1, G4), (A′3, R1-2, R2-1, R3-3, R4-2, G1), (A′3, R1-2, R2-1, R3-3, R4-2G2), (A′3, R1-2, R2-1, R3-3, R4-2, G3), (A′3, R1-2, R2-1, R3-3, R4-2, G4), (A′3, R1-2, R2-1, R3-4, R4-1, G1), (A′3, R1-2, R2-1, R3-4, R4-1, G2), (A′3, R1-2, R2-1, R3-4, R4-1, G3), (A′3, R1-2, R2-1, R3-4, R4-1, G4), (A′3, R 1-2, R2-1, R3-4, R4-2, G1), (A′3, R1-2, R2-1, R3-4, R4-2, G2), (A′3, R1-2, R2-1, R3-4, R4-2, G3), (A′3, R1-2, R2-1, R3-4, R4-2, G4), (A′3, R1-2, R2-2, R3-1, R4-1, G1), (A′3, R1-2, R2-2, R3-1, R4-1, G2), (A′3, R1-2, R2-2, R3-1, R4-1, G3), (A′3, R1-2, R2-2, R3-1, R4-1, G4), (A′3, R1-2, R2-2, R3-1, R4-2, G1), (A′3, R1-2, R2-2, R3-1, R4-2, G2), (A′3, R1-2, R2-2, R3-1, R4-2, G3), (A′3, R1-2, R2-2, R3-1, R 4-2, G4), (A′3, R1-2, R2-2, R3-2, R4-G1)(A′3, R1-2, R2-2, R3-2, R4-1, G2), (A′3, R1-2, R2-2, R3-2, R4-1, G3), (A′3, R1-2, R2-2, R3-2, R4-1, G4), (A′3, R1-2, R2-2, R3-2, R4-2, G1), (A′3, R1-2, R2-2, R3-2, R4-2, G2), (A′3, R1-2, R2-2, R3-2, R4-2, G3), (A′3, R1-2, R2-2, R3-2, R4-2, G4), (A′3, R1-2, R 2-2, R3-3, R4-1, G1), (A′3, R1-2, R2-2, R3-3, R4-1, G2), (A′3, R1-2, R2-2, R3-3, R4-1 G3), (A′3, R1-2, R2-2, R3-3, R4-1, G4), (A′3, R1-2, R2-2, R3-3, R4-2, G1), (A′3, R1-2, R2-2, R3-3, R4-2, G2), (A3, R1-2, R2-2, R3-3, R4-2, G3), (A′3, R1-2, R2-2, R3-3, R4-2, G4), (A′3, R1-2, R2-2, R3-4, R4-1, G1), (A′3, R1-2, R2-2, R3-4, R4-1, G2), (A′3, R1-2, R2-2, R3-4, R4-1, G3), (A′3, R1-2, R2-2, R3-4, R4-1, G4), (A′3, R1-2, R2-2, R3-4, R4-2, G1), (A′3, R1-2, R2-2, R3-4, R4-2, G2), (A′3, R1-2, R2-2, R3-4, R4-2, G3), (A′3, R1-2, R2-2, R3-4, R4-2, G4),


(A′4, R1-1, R2-1, R3-1, R4-1, G1), (A′4, R1-1, R2-1, R3-1, R4-1, G2), (A′4, R1-1, R2-1, R3-1, R4-1, G3), (A′4, R1-1, R2-1, R3-1, R4-1, G4), (A′4, R1-1, R2-1, R3-1, R4-2, G1), (A′4, R1-1, R2-1, R3-1, R 4-2, G2), (A′4, R1-1, R2-1, R3-1, R4-2, G3), (A′4, R1-1, R2-1, R3-1, R4-2, G4), (A′4, R1-1, R2-1, R3-2, R4-1, G1), (A′4, R1-1, R2-1, R3-2, R4-1, G2), (A′4, R1-1, R2-1, R3-2, R4-1, G3), (A′4, R1-1, R2-1, R3-2, R4-1, G4), (A′4, R1-1, R2-1, R3-2, R4-2, G1), (A′4, R1-1, R2-1, R3-2, R4-2, G2), (A′4, R1-1, R 2-1, R3-2, R4-2, G3), (A′4, R1-1, R2-1, R3-2, R4-2, G4), (A′4, R1-1, R2-1, R3-3, R4-1, G1), (A′4, R1-1, R2-1, R3-3, R4-1, G2), (A′4, R1-1, R2-1, R3-3, R4-1, G3), (A′4, R1-1, R2-1, R3-3, R4-1, G4), (A′4, R1-1, R2-1, R3-3, R4-2, G1), (A′4, R1-1, R2-1, R3-3, R4-2, G2), (A′4, R1-1, R2-1, R3-3, R4-2, G3), (A′4, R1-1, R2-1, R3-3, R4-2, G4), (A′4, R1-1, R2-1, R3-4, R4-1, G1), (A′4, R1-1, R2-1, R3-4, R4-1, G 2), (A′4, R1-1, R2-1, R3-4, R4-1, G3), (A′4, R1-1, R2-1, R3-4, R4-1, G4), (A′4, R1-1, R2-1, R3-4, R4-2, G1), (A′4, R1-1, R2-1, R3-4, R4-2, G2), (A′4, R1-1, R2-1, R3-4, R4-2, G3), (A′4, R1-1, R2-1, R3-4, R4-2, G4), (A′4, R1-1, R2-2, R3-1, R4-1, G1), (A′4, R1-1, R2-2, R3-1, R4-1, G2), (A4, R1-1, R2-2, R 3-1, R4-1, G3), (A′4, R1-1, R2-2, R3-1, R4-1, G4), (A′4, R1-1, R2-2, R3-1, R4-2, G1), (A′4, R1-1, R2-2, R3-1, R4-2, G2), (A′4, R1-1, R2-2, R3-1, R4-2, G3), (A′4, R1-1, R2-2, R3-1, R4-2, G4), (A′4, R1-1. R2-2, R3-2, R4-1, G1), (A′4, R1-1, R2-2, R3-2, R4-11 G2), (A′4, R1-1, R2-2, R3-2, R4-1, G3), (A′4, R 1-1, R2-2, R3-2, R4-1, G4), (A′4, R1-1, R2-2, R3-2, R4-2, G1), (A′4, R1-1, R2-2, R3-2, R4-2, G2), (A′4, R1-1, R2-2, R3-2, R4-2, G3), (A′4, R1-1, R2-2, R3-2, R4-2, G4), (A′4, R1-1, R2-2, R3-3, R4-1, G1), (A′4, R1-1, R2-2, R3-3, R4-1, G2), (A′4, R1-1, R2-2, R3-3, R4-1, G3), (A′4, R1-1, R2-2, R3-3, R4-1, G4), (A′4, R1-1, R2-2, R3-3, R4-2, G1), (A′4, R1-1, R2-2, R3-3, R4-2, G2), (A′4, R1-1, R2-2, R3-3, R 4-2, G3), (A′4, R1-1, R2-2, R3-3, R4-2, G4), (A′4, R1-1, R2-2, R3-4, R4-1, G1), (A′4, R1-1, R2-2, R3-4, R4-1, G2), (A′4, R1-1, R2-2, R3-4, R4-1, G3), (A′4, R1-1, R2-2, R3-4, R4-1, G4), (A′4, R1-1, R2-2, R3-4, R4-2, G1), (A′4, R1-1, R2-2, R3-4, R4-2, G2), (A′4, R1-1, R2-2, R3-4, R4-2, G3), (A′4, R1-1, R 2-2, R3-4, R4-2, G4), (A′4, R1-2, R2-1, R3-1, R4-1, G1), (A′4, R1-2, R2-1, R3-1, R4-1, G2), (A′4, R1-2, R2-1, R3-1, R4-1, G3), (A′4, R1-2, R2-1, R3-1, R4-1, G4), (A′4, R1-2, R2-1, R3-1, R4-2, G1), (A′4, R1-2, R2-1, R3-1, R4-2, G2), (A′4, R1-2, R2-1, R3-1, R4-2, G3), (A′4, R1-2, R2-1, R3-1, R4-2, G4), (A′4, R1-2, R2-1, R3-2, R4-1, G1), (A′4, R1-2, R2-1, R3-2, R4-1, G2), (A′4, R1-2, R2-1, R3-2, R4-1, G 3), (A′4, R1-2, R2-1, R3-2, R4-1, G4), (A′4, R1-2, R2-1, R3-2, R4-2, G1), (A′4, R1-2, R2-1, R3-2, R4-2, G2), (A′4, R1-2, R2-1, R3-2, R4-2, G3), (A′4, R1-2, R2-1, R3-2, R4-2, G4), (A′4, R1-2, R2-1, R3-3, R4-1, G1), (A′4, R1-2, R2-1, R3-3, R4-1, G2), (A′4, R1-2, R2-1, R3-3, R4-1, G3), (A′4, R1-2, R2-1, R 3-3, R4-1, G4), (A′4, R1-2, R2-1, R3-3, R4-2, G1), (A′4, R1-2, R2-1, R3-3, R4-2, G2), (A′4, R1-2, R2-1, R3-3, R4-2, G3), (A′4, R1-2, R2-1, R3-3, R4-2, G4), (A′4, R1-2, R2-1, R3-4, R4-1, G1), (A′4, R1-2, R2-1, R3-4, R4-1, G2), (A′4, R1-2, R2-1, R3-4, R4-1, G3), (A′4, R1-2, R2-1, R3-4, R4-1, G4), (A′4, R 1-2, R2-1, R3-4, R4-2, G1), (A′4, R1-2, R2-1, R3-4, R4-2, G2), (A′4, R1-2, R2-1, R3-4, R4-2, G3), (A′4, R1-2, R2-1, R3-4, R4-2, G4), (A′4, R1-2, R2-2, R3-1, R4-1, G1), (A′4, R1-2, R2-2, R3-1, R4-1, G2), (A′4, R1-2, R2-2, R3-1, R4-1, G3), (A′4, R1-2, R2-2, R3-1, R4-1, G4), (A′4, R1-2, R2-2, R3-1, R4-2, G1), (A′4, R1-2, R2-2, R3-1, R4-2, G2), (A′4, R1-2, R2-2, R3-1, R4-2, G3), (A′4, R1-2, R2-2, R3-1, R4-2, G4), (A′4, R1-2, R2-2, R3-2, R4-1, G1), (A′4, R1-2, R2-2, R3-2, R4-1, G2), (A′4, R1-2, R2-2, R3-2, R4-1, G3), (A′4, R1-2, R2-2, R3-2, R4-1, G4), (A′4, R1-2, R2-2, R3-2, R4-2, G1), (A′4, R1-2, R2-2, R3-2, R4-2, G2), (A′4, R1-2, R2-2, R3-2, R4-2, G3), (A′4, R1-2, R2-2, R3-2, R4-2, G4), (A′4, R1-2, R 2-2, R3-3, R4-1, G1), (A′4, R1-2, R2-2, R3-3, R4-1, G2), (A′4, R1-2, R2-2, R3-3, R4-1, G3), (A′4, R1-2, R2-2, R3-3, R4-1, G4), (A′4, R1-2, R2-2, R3-3, R4-2, G1), (A′4, R1-2, R2-2, R3-3, R4-2, G2), (A′4, R1-2, R2-2, R3-3, R4-2, G3), (A′4, R1-2, R2-2, R3-3, R4-2, G4), (A′4, R1-2, R2-2, R3-4, R4-1, G1), (A′4, R1-2, R2-2, R3-4, R4-1, G2), (A′4, R1-2, R2-2, R3-4, R4-1, G3), (A′4, R1-2, R2-2, R3-4, R4-1, G 4), (A′4, R1-2, R2-2, R3-4, R4-2, G1), (A′4, R1-2, R2-2, R3-4, R4-2, G2), (A′4, R1-2, R2-2, R3-4, R4-2, G3) or (A′4, R1-2, R2-2, R3-4, R4-2, G4).


Compounds of the present invention are useful for treating diseases induced by production, secretion or deposition of amyloid β protein, and effective for the treatment and/or prophylaxis, or improvement of conditions for Alzheimer's dementia (Alzheimer's disease, senile dementia of Alzheimer type etc.), Down's disease, disturbance of memory, prion disease (Creutzfeldt-Jakob disease etc.), mild cognitive impairment (MCI), Dutch-type hereditary cerebral hemorrhage with amyloidosis, cerebral amyloid angiopathy, other degenerated dementia, vascular degenerated mixed dementia, dementia associated with Parkinson's disease, dementia associated with progressive supranuclear paralysis, dementia associated with corticobasal degeneration, diffuse Lewy Bodies Alzheimer's disease, age-related macular degeneration, Parkinson's disease, or amyloid angiopathy etc.


Since compounds of the present invention have several efficacies such as having a potent inhibitory activity against BACE-1 having a high selectivity against other enzymes etc., they can be a drug with less side effects. Moreover they can be a drug having a wide margin of safety by choosing an optically active isomer of appropriate stereochemistry. Further they have a lot of merits such as good metabolic stability, high solubility, high absorbability of oral administration, high bioavailability, good clearance and high transitivity to brain, long half-life, high ratio of non protein binding, lower inhibition of hERG channel and CYP, and/or negative result of Ames Test, and, therefore, they can be superior drugs.


A compound of the present invention may be administrated together with other agent (e.g., other agent for treating Alzheimer's disease such as acetylcholine esterase etc.). The compound can be given in combination with an antidementia drug such as donepezil hydrochloride, tacrine, galantamine, rivastigmine, zanapezil, memantine or vinpocetine, for example.


A compound of the present invention may be orally administrated as powder, granule, tablet, capsule, pill or liquid formulation, or parentally administrated as injection, suppository, formulation of transdermal absorption or inhalation. Also, an effective amount of the compound may be formulated together with medicinal additives suitable for the formulation such as an excipient, binder, moistening agent, disintegrant and/or lubricant etc.


Dose of a compounds of the present invention depends on condition of diseases, route of administration, age and body weight of a patient, but in the case of oral administration to an adult, the dose range is usually 0.1 μg to 1 g/day, preferably 0.01 to 200 mg/day and in the case of parenteral administration the dose range is usually 1 μg to 10 g/day, preferably 0.1 to 2 g/day.


EXAMPLES

The present invention is illustrated in details by examples and test examples but the present invention is not limited to these examples.


In EXAMPLES, each abbreviation has the following meaning:


Me: methyl


Et: ethyl


iPr, Pri isopropyl


tBu: t-butyl


Ph: phenyl


Bn: benzyl


Boc: tert-butoxycarbonyl


TFA: trifluoroacetic acid


THF: tetrahydrofuran


DMT-MM: 4-(4,6-dimethoxy-1,3,5-triazin-2-yl)-4-methylmorpholinium chloride n-hydrate


DMF: N,N-dimethylformamide


Reference Example 1



embedded image


The 1st step: Compound (1) (101.5 g) was cooled to −18° C. and conc. sulfuric acid (400 ml) was added dropwise in 65 minutes while the inner temperature was kept at −15° C. or below. Separately fuming nitric acid (60 ml) was added to conc. sulfuric acid (180 ml) chilled to 4° C. in 45 minutes while the temperature was kept at 10° C. or below and the resulted mixed acid was added dropwise to the solution of (1) prepared before in an hour while the temperature was kept at −30° C. or below. The mixture was stirred at −20° C. or below for 1.5 hours, poured into 2.5 kg of ice-water and stirred for an hour. The precipitated crystals were filtered to give compound (2) (121.5 g).



1H-NMR (CDCl3): 2.71 (3H, d, J=5.1 Hz), 7.35 (1H, dd, J=9.3, 9.0 Hz), 8.41 (1H, ddd, J=9.0, 3.9, 3.0 Hz), 8.78 (1H, dd, J=6.3, 3.0 Hz).


The 2nd step: Compound (2) (20 g) was dissolved in ethanol (400 ml), Pd—C (10% dry)(2.0 g) was added thereto and the mixture was stirred in a hydrogen atmosphere at room temperature for 2 hours. Then Pd—C (10% dry) (1.0 g) was added and the mixture was stirred in a hydrogen atmosphere at room temperature for 1.5 hours, and further Pd—C (10% (dry) (1.0 g) was added and the mixture was stirred in a hydrogen atmosphere at room temperature for 15 minutes. Pd—C was filtered off, the solvent was evaporated under reduced pressure and the residue of compound (3) (15.9 g) was obtained.



1H-NMR (DMSO-d6): 2.50 (3H, d, J=4.8 Hz), 5.21 (2H, brs, 1H), 6.78 (1H, ddd, J=8.7, 4.2, 3.0 Hz), 6.94 (1H, dd, J=6.3, 3.0 Hz), 6.99 (1H, dd, J=1.4, 8.7 Hz).


The 3rd step: Compound (3) (15.8 g) was dissolved in THF (79 ml), anhydrous trifluoroacetic acid (16.1 ml) and triethylamine (20.2 ml) were added under ice cooling and the mixture was stirred for 20 minutes. After the addition of water (30 ml), it was stirred under ice cooling for 20 minutes and the precipitated crystals were filtered. The filtrate was extracted with ethyl acetate (80 ml) and 50 ml, the organic layer was washed with water (60 ml), saturated brine. The crystals filtered previously were dissolved in the organic layer and dried over sodium sulfate. Sodium sulfate was filtered, the filtrate was concentrated under reduced pressure and the residue was dissolved in ethyl acetate under warming. After the addition of hexane (50 ml) and stirring under ice cooling for 20 minutes, the precipitated crystals were filtered. The mother liquid was concentrated again under reduced pressure, crystallized by the addition of ethyl acetate (8 ml) and hexane (12 ml) and compound (4) (totally 20.4 g) was obtained.



1H-NMR (CDCl3): 2.70 (3H, d, J=5.1 Hz), 7.24 (1H, dd, J=10.5, 9.3 Hz), 8.00 (1H, dd, J=6.2, 2.9 Hz), 8.21 (1H, m), 8.78 (1H, brs).


The 4th step: 1.6 M Vinyl magnesium chloride-THF solution (122 mil) was dissolved in THF (161 ml), cooled to −40° C. in a nitrogen atmosphere and a THF (81 ml) solution of compound (4) (16.1 g) was added dropwise thereto. The reaction solution was stirred at −40° C. for 20 minutes, 1.6 M vinyl magnesium chloride-THF solution (20 mil) was further added and the mixture was stirred at −40° C. for 15 minutes. The reaction solution was poured into a mixture of chilled ethyl acetate (480 ml), a saturated aqueous solution of ammonium chloride (80 ml) and water (80 ml) with stirring, and the organic layer was separated. The aqueous layer was further extracted with ethyl acetate (200 ml), the organic layers were combined, washed with water (80 ml), and saturated brine successively, and dried over sodium sulfate. Sodium sulfate was filtered, the filtrate was concentrated under reduced pressure and the residue of compound (5) (22.4 g) was obtained.



1H-NMR (CDCl3): 1.74 (3H, d, J=1.2 Hz), 5.16 (1H, dd, J=10.5, 0.9 Hz), 5.27 (1H, d, J=17.3, Hz), 6.26 (1H, ddd, J=17.3, 10.5, 1.7 Hz), 7.07 (1H, dd, J=11.1, 9.6 Hz), 7.64-7.69 (2H, m), 7.94 (1H, brs).


The 5th step: The residue of compound (5) (22.3 g) and thiourea (5.17 g) were dissolved in acetic acid (112 ml), 1 M HCl-ethyl acetate (97 ml) was added thereto and the mixture was stirred at 40° C. for 18 hours. The solvent was evaporated under reduced pressure, toluene (150 ml) was added and again concentrated under reduced pressure. After repeating the same procedure, crystals were precipitated. Ethyl acetate (100 ml) was added to the crystalline residue, the mixture was stirred under ice cooling for an hour and the crystals were filtered to give compound (6) (15.1 g).



1H-NMR (DMSO-d6): 2.08 (3H, s), 4.10 (2H, d, J=7.8 Hz), 5.72 (1H, t, J=7.8 Hz), 7.23-7.32 (1H, m), 7.60-7.69 (2H, m), 9.25 (3H, brs), 11.39 (1H, brs).


The 6th step: Compound (6) (10.0 g) was dissolved in THF (50 ml), conc. sulfuric acid (5.74 ml) was added thereto and stirred at 60° C. for 2 hours. After evaporation of TFA under reduced pressure, ice-water (100 ml) was added. The mixture was stirred under ice-cooling for an hour, and the precipitated crystals were filtered to give compound (7) (11.2 g).



1H-NMR (CDCl3): 1.72 (3H, s), 2.02-2.18 (1H, m), 2.54-2.76 (2H, m), 3.14-3.28 (1H, m), 7.37 (1H, dd, J=1.9, 8.8 Hz), 7.62 (1H, dd, J=7.5, 3.0 Hz), 7.80 (1H, ddd, J=8.8, 3.9, 3.0 Hz), 8.77 (1H, brs), 9.38 (H, brs), 10.66 (1H, brs), 11.50 (1H, brs).


The 7th step: MeOH (28 ml), THF (35 ml) and 5 N NaOH (10.9 ml) were added to compound (7) (7.00 g) and stirred at 50° C. for 4 hours. Toluene (50 ml) was added and extracted, and the aqueous layer was further extracted with toluene (50 ml) and ethyl acetate (60 ml). All the organic layers were combined, washed with water and saturated brine, and dried over sodium sulfate. The solvent was concentrated under reduced pressure, the resulted crystalline residue was washed with hexane (20 ml) to give compound (8) (3.45 g).



1H-NMR (CDCl3): 1.60 (3H, d, J=1.5 Hz), 1.76-1.87 (1H, m), 2.44-2.54 (1H, m), 2.66-2.76 (1H, m), 2.86-2.94 (1H, m), 6.50 (1H, ddd, J=8.7, 3.6, 3.0 Hz), 6.66 (1H, dd, J=7.1, 3.0 Hz), 6.81 (1H, dd, J=12.0, 8.7 Hz).


Reference Example 2



embedded image


The 1st step: Compound (3) (15.6 g) was dissolved in ethyl acetate (78 ml), acetic anhydride (10.6 ml) and pyridine (9.07 ml) were added under ice cooling, and the mixture was stirred for 15 minutes. Ethyl acetate (100 ml) and water (50 ml) were added, extracted, and the aqueous layer was extracted with ethyl acetate (50 ml). The organic layers were combined, washed with 2 M HCl (50 ml), a saturated solution of sodium bicarbonate (50 ml) and saturated brine, dried over sodium sulfate. Sodium sulfate was filtered, the filtrate was concentrated under reduced pressure, and ethyl acetate (50 ml) and hexane (50 mil) were added to the residue. The mixture was stirred under ice cooling for 30 minutes and the precipitated crystals were filtered to give compound (9) (total 14.9 g).



1H-NMR (CDCl3): 2.20 (3H, s), 2.66 (3H, d, J=5.1 Hz), 7.13 (H, dd, J=10.5, 9.0 Hz), 7.70 (1H, dd, J=6.3, 3.0 Hz), 7.79 (1H, brs), 8.11 (1H, ddd, J=9.0, 4.1, 3.0 Hz).


The 2nd step: Compound (9) (10.0 g) was dissolved in THF (50 ml), cooled in ice and sodium hydride (2.25 g) was added in a nitrogen atmosphere. After stirring for 15 minutes, the resulted mixture was added dropwise to a solution of 1.6 M vinyl magnesium chloride (86 ml)/THF (70 ml) cooled to −40° C. After stirring at −40° C. for 15 minutes and then 0° C. for 20 minutes, a saturated aqueous solution of ammonium chloride (50 ml)/water (50 ml) was chilled and added. The layers were separated and the aqueous layer was extracted with ethyl acetate (100 ml). Organic layers were combined, washed with water and saturated brine, dried over sodium sulfate. Sodium sulfate was filtered, the filtrate was concentrated under reduced pressure to give a residue of compound (10) (13.7 g).



1H-NMR (CDCl3): 1.69 (3H, s), 2.16 (3H, s), 5.12 (1H, d, J=10.5 Hz), 5.24 (1H, d, J=17.4 Hz), 6.26 (1H, ddd, J=17.4, 10.5, 1.5 Hz), 6.98 (1H, dd, J=11.1, 8.7 Hz), 7.33 (1H, brs), 7.50-7.59 (2H, m).


The 3rd step: The residue of compound (10) (6.56 g) and thiourea (1.88 g) were dissolved in acetic acid (33 ml), 1 M HCl-acetic acid (37 ml) was added and stirred at 40° C. for 7 hours. The solvent was evaporated under reduced pressure, toluene (50 ml) was added and concentrated again under reduced pressure. The same procedure was repeated again, ethyl acetate (30 ml) was added to the residue and stirred at room temperature overnight. The precipitate was filtered to give compound (11) (5.77 g).



1H-NMR (DMSO-d6): 2.03 (3H, s), 2.06 (3H, s), 4.09 (2H, d, J=7.5 Hz), 5.67 (1H, t, J=7.5 Hz), 7.12 (1H, dd, J=10.7, 8.9 Hz), 7.46-7.59 (2H, m), 9.24 (4H, brs), 10.11 (1H, s).


The 4th step: compound (11) (5.16 g) was dissolved in conc. sulfuric acid (15.5 ml) and stirred at room temperature for an hour. It was poured into ice-water (100 ml), adjusted to pH 10 by the addition of an aqueous solution of potassium hydroxide and extracted with ethyl acetate (200 ml) and a little amount of MeOH. The organic layer was washed with water and saturated brine, dried over sodium sulfate. Sodium sulfate was filtered, the filtrate was concentrated under reduced pressure, ethyl acetate (20 ml) and hexane (15 ml) were added to the residue and the precipitate was filtered. The filtrate was concentrated, ethyl acetate (5 ml) and hexane (5 ml) were added and the precipitate was filtered to give compound (12) (total 3.16 g).



1H-NMR (CDCl3): 1.62 (3H, d, J=0.9 Hz), 1.80-1.91 (1H, m), 2.16 (3H, s), 2.47-2.58 (1H, m), 2.62-2.73 (1H, m), 2.87-2.98 (1H, m), 4.36 (2H, brs), 6.99 (1H, dd, J=11.7, 8.7 Hz), 7.14 (1H, dd, J=7.1, 3.0 Hz), 7.80 (1H, ddd, J=8.7, 4.2, 3.0 Hz), 7.97 (1H, brs),


The 5th step: Compound (12) (2.50 g) was suspended in ethanol (25 ml), 6 M HCl (10.2 ml) was added and the mixture was stirred at 90° C. for 3 hours. 2 M NaOH (35 ml) was added, the organic solvent was evaporated and the residue was extracted with ethyl acetate (70 ml). The aqueous layer was further extracted with ethyl acetate (30 ml), organic layers were combined, washed with water and saturated brine, and dried over sodium sulfate.


Sodium sulfate was filtered, the filtrate was concentrated under reduced pressure, and the crystalline residue was washed with ethyl acetate (3 ml) and hexane (10 ml). The crystals were filtered to give compound (8) (total 1.22 g).


Reference Example 3



embedded image



The 1st step: A ethanol solution of 20% sodium ethoxide (5.12 ml, 16.2 mmol, 40 eq.) was added to compound (13) (150 mg, 406 μmol) and stirred at room temperature for 6 hours. The reaction solvent was evaporated under reduced pressure, 2 M hydrochloric acid (8.12 ml, 16.2 mmol, 40 eq.) was added to the resulted residue and extracted with chloroform. The extracting solution was washed with water and dried over anhydrous sodium sulfate. The crude product (189 mg) was obtained by evaporation of the solvent under reduced pressure, to which 4 M HCl-ethyl acetate solution (1.89 ml) was added and the mixture was stirred at room temperature for 14 hours. A saturated aqueous solution of sodium bicarbonate was added to the reaction solution, extracted with ethyl acetate and dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure to give compound (14) (90.8 mg, 76% yield) as a yellow powder.



1H NMR (CDCl3) δ1.52 (3H, t, J=6.8 Hz), 1.67 (3H, s), 1.93-2.00 (1H, m), 2.60-2.67 (2H, m), 2.94-3.00 (1H, m), 4.19 (2H, q, J=6.8 Hz), 6.93 (1H, d, J=9.3 Hz), 8.14 (1H, dd, J=8.7, 2.4 Hz), 8.31 (1H, d, J=2.5 Hz).


The 2nd step: A powder of 10% palladium-carbon (45.4 mg) was added to a methanol (908 μl) solution of compound (14) (90.8 mg, 307 mmol) and the mixture was stirred in a hydrogen atmosphere for 22 hours. The reaction mixture was filtered through a Celite pad and the filtrate was evaporated under reduced pressure. The residue was washed with ethyl acetate to give compound (15) (65.8 mg, 81% yield) as an yellow powder.



1H NMR (DMSO-d6) δ 1.29 (3H, t, J=6.9 Hz), 1.45 (3H, s), 1.51-1.58 (1H, m), 2.46-2.48 (1H, m), 2.61-2.64 (1H, m), 2.80-2.83 (1H, m), 3.85-3.91 (2H, m), 6.38 (1H, dd, J=8.3, 2.5 Hz), 6.52 (1H, d, J=2.4 Hz), 6.67 (1H, d, J=8.6 Hz)


Reference Example 4



embedded image


embedded image


The 1st step: 3′-Bromoacetophenone (15.0 g) and compound (16) (9.13 g) were dissolved in tetrahydrofuran (250 ml), and tetraethoxytitanium (39.5 ml) was added thereto at room temperature with stirring. Then the reaction mixture was stirred at 75° C. for 5 hours, and saturated brine was added after disappearance of compound (1) was confirmed. Titanium oxide formed in the reaction was filtered off, the filtrate was extracted with ethyl acetate, the organic layer was dried over anhydrous magnesium sulfate and the solvent was evaporated under reduced pressure. The residue was purified with a column chromatography to give compound (17) (20.1 g).



1H-NMR (CDCl3): 1.33 (9H, s), 2.75 (3H, s), 7.30 (1H, t. J=7.8) 7.59-7.63 (1H, m), 7.79 (1H, d, J=7.8) 8.0 (1H, s)


The 2nd step: A 2.64 M hexane solution of n-butyllithium (79.5 ml) is added dropwise to a tetrahydrofuran (100 ml) solution of diisopropylamine (42.1 ml) in a nitrogen atmosphere at −78° C. After stirring at 0° C. for 30 minutes, the reaction solution was again cooled to −78° C. and tert-butyl acetate (26.9 ml) dissolved in tetrahydrofuran (100 ml) is added dropwise. After stirring at −78° C. for 30 minutes, chlorotriisopropoxytitanium dissolved in tetrahydrofuran (150 ml) is added dropwise. After stirring at the same temperature for 70 minutes, compound (2) (20.1 g) dissolved in tetrahydrofuran (100 ml) was added dropwise. After then, the reaction solution was stirred at −78° C. for 3 hours, and an aqueous solution of ammonium chloride was added after disappearance of compound (2) was confirmed. Titanium oxide formed in the reaction was filtered off, the filtrate was extracted with ethyl acetate, the organic layer was washed with saturated brine and dried over anhydrous magnesium sulfate. The solvent was evaporated under reduced pressure to give a crude product of compound (18) (26.4 g).


The 3rd step: The crude product of compound (18) (26.4 g) was dissolved in toluene (80 ml), the solution was added dropwise to a 1 M solution of aluminium diisobutyl hydride in toluene (253 ml) with stirring at 0° C. The reaction solution was stirred at room temperature for 1.5 hours and 1 N hydrochloric acid solution was added after disappearance of compound (3) was confirmed. The mixture was extracted with ethyl acetate, the organic layer was washed with saturated brine and dried over anhydrous magnesium sulfate. The solvent was evaporated under reduced pressure and the residue was purified by crystallization to give compound (19) (18.1 g).



1H-NMR (CDCl3): 1.28 (9H, s), 1.71 (3H, s), 2.19-2.24 (2H, m), 3.27-3.32 (1H, m), 3.54-3.66 (1H, m), 3.87-3.97 (1H, m), 5.10-5.11 (1H, m), 7.22 (1H, t. J=8.1) 7.32-7.41 (2H, m), 7.56-7.58 (1H, m)


The 4th step: Compound (19) (18. g) was dissolved in methanol (30 ml), and 10% hydrochloric acid in methanol (130 ml) was added dropwise therein at room temperature. The reaction solution was stirred at room temperature for 4 hours and 1 N hydrochloric acid was added after disappearance of compound (4) was confirmed. The mixture was extracted with ethyl acetate, the aqueous layer was neutralized with a 2 N aqueous solution of sodium hydroxide and extracted with ethyl acetate. The organic layer was dried over anhydrous magnesium sulfate and the solvent was evaporated under reduced pressure to give a crude product of compound (20) (14.1 g).


The 5th step: The crude product of compound (20) (32.8 g) and potassium carbonate (37.1 g) were dissolved in a mixed solvent of toluene (450 nm) and water (225 ml) and thiopbosgene (15.3 ml) was added dropwise cooled at 0° C. with stirring. After then, the reaction solution was stirred at 0° C. for an hour and water was added when disappearance of compound (5) was confirmed. The mixture was extracted with ethyl acetate and the organic layer was washed with brine and dried over anhydrous magnesium sulfate. The solvent was evaporated under reduced pressure to give a crude product of compound (21) (38.4 g).


The 6st step: The crude product of (21) (38.4 g) was dissolved in toluene (384 ml), and thionyl chloride (29.4 ml) and N,N-dimethylformamide (1.04 ml) were added dropwise at 0° C. with stirring. After then, the reaction solution was stirred at 80° C. for 5 hours, and after disappearance of compound (6) was confirmed, the reaction solution was evaporated under reduced pressure to give a crude product of compound (22) (40.9 g).


The 7th step: The crude product of compound (22) (40.9 g) was dissolved in tetrahydrofuran (250 ml) and 25% ammonia-water (250 ml) was added with stirring at 0° C. After then the reaction solution was stirred at room temperature for 16 hours, and a saturated aqueous solution of sodium bicarbonate was added after disappearance of compound (21) was confirmed. The organic layer was separated and the aqueous solution was extracted with dichloromethane. The organic layers were combined, dried over anhydrous magnesium sulfate and evaporated under reduced pressure to give a crude product of compound (23) (38.3 g).


The 8th step: The crude product of compound (23) (38.3 g) is dissolved in tetrahydrofuran (383 ml), di-tert-butyl dicarbonate (61.5 g) and N,N-dimethylaminopyridine (1.64 g) are added and the mixture was stirred at room temperature for 72 hours. After disappearance of compound (23) was confirmed, the solvent was evaporated under reduced pressure. The residue was purified with a silicagel column chromatography to give compound (24) (45.3 g)



1H-NMR (CDCl3): 1.54 (9H, s), 1.57 (3H, s), 1.96 (2H, t, J=6.0), 2.80-2.92 (1H, m), 3.00-3.13 (1H, m), 7.21 (1H, J=8.1) 7.28-7.41 (2H, m), 7.52-7.55 (1H, m)


The 9th step: In a nitrogen atmosphere, compound (24) (12.1 g), trisdibenzylideneacetonedipalladium (1.14 g) and dicyclohexylbiphenylpbosphine (0.88 g) were dissolved in toluene (125 ml), and a 1.6 M solution of lithium hexamethyldisilazide in tetrahydrofuran (46.9 ml) was added with stirring at room temperature. The reaction solution was warmed up to 80° C. and stirred for 16 hours. After disappearance of compound (21) was confirmed, the reaction solution was cooled at 0° C. and diethyl ether and 1 N hydrochloric acid were added. After stirring at 0° C. for 10 minutes, the solution was neutralized with a saturated aqueous solution of sodium carbonate. It was extracted with ethyl acetate, the organic layer was washed with saturated brine and dried over anhydrous magnesium sulfate. The solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give compound (25) (6.84 g).



1H-NMR (CDCl3): 1.51 (9H, s), 1.69 (3H, s), 2.01-2.12 (1H, m), 2.40-2.51 (1H, m), 2.67-2.76 (2H, m), 6.55-6.67 (3H, m), 7.15 (1H, t. J=8.1).


Reference Example 5



embedded image


embedded image


The 1st step: After the addition of compound (1) (70.00 g) to conc. sulfuric acid (279 ml) cooled in an acetonitrile/dry ice bath with stirring, a mixture of fuming nitric acid (42 ml) and conc. sulfuric acid (98 ml) were added dropwise. After stirring for 16 minutes, the mixture was gradually poured into ice, the precipitated crystals were filtered and dried to give compound (2) (77.79 g).



1H-NMR (CDCl3) δ: 2.71 (3H, d, J=4.9 Hz), 7.34 (1H, t, J=9.3 Hz), 8.40 (H, ddd, J=9.3, 6.2, 3.0 Hz), 8.78 (1H, dd, J=6.2, 3.0 Hz).


The 2nd step: A solution of compound (2) (73.94 g), (R)-(+)-2-methyl-2-propanesulfinamide (53.82 g) and tetraethyl orthotitanate (230.20 g) in tetrahydrofuran (500 ml) were reacted for 2.5 hours under heating to reflux, and then the reaction mixture was gradually poured into ice and the resulted insoluble materials were filtered. It was extracted with ethyl acetate, the organic layer was dried over anhydrous magnesium sulfate and evaporated under reduced pressure. The residue was crystallized from ethyl acetate/n-hexane to give compound (26) (85.44 g).



1H-NMR (CDCl3) δ: 1.34 (9H, s), 2.81 (3H, d, J=3.5 Hz), 7.29 (1H, t, J=8.9 Hz), 8.31 (1H, dt, J=8.9, 2.9 Hz), 8.55 (1H, dd, J=6.3, 2.9 Hz).


The 3rd step: A solution of tert-butyl acetate (6.08 g) in tetrahydrofuran (10 ml) was added dropwise to a 2 M solution of lithium diisopropylamide/tetrahydrofuran/n-heptane/ethylbenzene (27.9 ml) cooled in an acetone/dry ice bath with stirring. After stirring for 20 minutes, a solution of chlorotitanium isopropoxide (17.5 ml) in tetrahydrofuran (30 ml) was added dropwise, the mixture was stirred for an hour and a solution of compound (26) (5.00 g) in tetrahydrofuran (10 ml) was added dropwise. After reacting for an hour, the reaction solution was gradually poured into an aqueous solution of ammonium chloride cooled in ice with stirring and the resulted insoluble materials were filtered. It was extracted with ethyl acetate, the organic layer was dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give compound (27) (5.49 g).



1H-NMR (CDCl3) δ: 1.30 (9H, s), 1.35 (9H, s), 1.86 (3H, s), 3.11 (1H, dd, J=16.2, 2.1 Hz), 3.26 (1H, dd, J=16.2, 2.1 Hz), 5.55 (1H, s), 7.18 (1H, dd, J=11.1, 8.9 Hz), 8.18 (1H, ddd, J=8.9, 4.0, 2.9 Hz), 8.53 (1H, dd, J=7.0, 2.9 Hz).


Ratio of diastereomers (3S:3R=97:3) HPLC Column: CHIRALPAK AS-RH, Detection: 254 nm: Column temp.: 25° C., Mobile phase: 40% MeCNaq., Flow rate: 0.5 ml/min.


Note: As to the stereochemistry of compound (27) obtained above, it is known that 3S-isomer is preferentially prepared as written in the literature A etc. and it is also possible to prepare each diastereomer selectively by choosing appropriate metal species and/or a reaction condition.


Literature A: (I) T. Fujisawa et al., Terahedron Lett., 37, 3881-3884 (1996), (2) D. H. Hua et al, Sulfur Reports, vol. 21, pp. 211-239 (1999), (3) Y. Koriyama et al., Tetrahedron, 58, 9621-9628 (2002), (4) Yong Qin et al., J. Org. Chem., 71, 1588-1591 (2006).


The 4th step: A solution of 4 M HCl/1,4-dioxane (50 ml) was added to compound (27) (12.74 g) and the mixture was stirred at 80° C. for an hour, diethyl ether (50 ml) was added, the precipitated crystals were filtered and dried to give compound (28) (7.67 g).



1H-NMR (DMSO-d6) δ: 1.76 (3H, s), 3.25 (2H, s), 7.62 (1H, dd, J=119., 4 Hz), 8.33-8.48 (2H, m).


The 5th step: A solution of 1 M tetrahydrofuran-borane in tetrahydrofuan (2029 mil) was added dropwise to a solution of compound (28) (141.32 g) in tetrahydrofuran (707 ml) cooled in ice with stirring and it was reacted for 3 hours and 6 minutes. The reaction mixture was poured into a mixture of sodium bicarbonate (511 g), ice (1500 g) and ethyl acetate (3000 ml) stirred at room temperature, extracted with ethyl acetate and the organic layer was dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure to give compound (29) (115.46 g) as a crude product.


The 6th step: Toluene (25 ml) and water (12.5 ml) were added to the compound (29) (3.76 g) obtained in the 5th step and stirred under ice cooling. After the addition of potassium carbonate (7.97 g), thiophosgene (2.85 g) was added dropwise. After reacting for 3 hours, water was added, extracted with toluene and the organic layer was dried over anhydrous magnesium sulfate. A part of the solvent was evaporated under reduced pressure to give compound (30) as a crude product.


The 7th step: Compound (30) obtained in the 6th step was dissolved in toluene (17.4 ml) and thionyl chloride (6.67 g) and N,N-dimethylformamide (0.128 ml) were added with stirring at room temperature. The mixture was stirred at 80° C. for 2 hours, water was added, extracted with toluene and concentrated under reduced pressure to give compound (31) (4.03 g) as a crude product.


The 8 step: Compound (31) (4.03 g) obtained in the 7th step was dissolved in tetrahydrofuran (23.8 ml) and 28% ammonia-water (23.8 ml) was added under ice cooling with stirring. The mixture was stirred at room temperature for 3 days, the reaction solution was concentrated under reduced pressure and ethyl acetate was added therein. Conc. hydrochloric acid (6 ml) was added under ice cooling with stirring, the precipitated crystals were washed with ethyl acetate and water and dried to give compound (32) (2.14 g).



1H-NMR (DMSO-d6) δ: 1.76 (3H, s), 2.13-2.24 (1H, m), 2.68-2.74 (2H, m), 3.19-3.25 (1H, m), 7.63 (1H, dd, J=11.4, 8.9 Hz), 8.07 (1H, dd, J=7.0, 3.5 Hz), 8.36 (1H, dt, J=8.9, 3.5 Hz), 11.22 (1H, s).


The 9th step: Compound (32) (100 mg) was dissolved in methanol (2 ml), 10% palladium-carbon powder (50 mg) was added, and the mixture was stirred in a hydrogen atmosphere at room temperature for 18 hours. Insoluble materials were filtered off, the filtrate was evaporated under reduced pressure, sodium carbonate and water were added therein and extracted with ethyl acetate. The organic layer was dried over anhydrous sodium sulfate and the solvent was evaporated under reduced pressure to give compound (33) (68 mg).



1H-NMR (CDCl3) δ: 1.59 (3H, s), 1.81 (1H, ddd, J=14.1, 10.9, 3.5 Hz), 2.47 (1H, ddd, J=14.1, 5.9, 3.5 Hz), 2.71 (1H, td, J=10.9, 3.5 Hz), 2.89 (1H, ddd, J=10.9, 5.9, 3.5 Hz), 3.57 (2H, br s), 6.49 (1H, dt, J=8.5, 3.3 Hz), 6.67 (1H, dd, J=6.9, 3.3 Hz), 6.80 (1H, dd, J=11.8, 8.5 Hz).


Reference Example 6



embedded image


embedded image


The 1st step: A solution of compound (38) (5.00 g), (R)-(+)-2-methyl-2-propanesulfinamide (3.33 g) and tetraethyl orthotitanate (17.11 g) in tetrahydrofuran (50 ml) was reacted under beating to reflux for 7 hours, and then, it was poured portionwise into saturated brine and the resulted insoluble materials were filtered off. It was extracted with ethyl acetate, the organic layer was dried over anhydrous magnesium sulfate and concentrated under reduced pressure. The residue was purified with a silicagel column chromatography to give compound (39) (6.37 g).



1H-NMR (CDCl3) δ: 1.34 (9H, s), 2.79 (3H, s), 8.26 (1H, t, J=2.3 Hz), 8.76 (1H, d, J=2.3 Hz), 8.96 (1H, d, J=23 Hz).


The 2nd Step: A solution of 2.66 M n-butyllithium/n-hexane (32.4 ml) was added dropwise to a solution of diisopropylamine (9.36 g) in tetrahydrofuran (39 ml) cooled in an acetone/dry ice bath with stirring and the mixture was stirred under ice cooling for 30 minutes. The reaction solution was stirred again in an acetone/dry ice bath and a solution of tert-butyl acetate (4.88 g) in tetrahydrofuran (8 ml) was added dropwise. After stirring for 40 minutes, a solution of chlorotitanium triisopropoxide (23.00 g) in tetrahydrofuran (88 ml) was added dropwise. After stirring for 10 minutes, a solution of compound (39) (6.37 g) in tetrahydrofuran (65 ml) was added dropwise. After reacting for 30 minutes, the reaction solution was poured portionwise into an aqueous solution of ammonium chloride and the resulted insoluble materials were filtered off. It was extracted with ethyl acetate and the organic layer was dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure to give compound (40) (8.03 g) as a crude product.


The 3rd step: Lithium aluminium hydride (2.85 g) was added portionwise to a solution of the compound (40) (8.03 g) obtained in the 2nd step in tetrahydrofuran (100 ml) cooled in ice with stirring and the mixture was stirred for 2 hours. Acetone, water, and a 1N aqueous solution of sodium hydroxide were added portionwise and the mixture was stirred at room temperature for 30 minutes. The insoluble materials were filtered off and extracted with ethyl acetate. The organic layer was dried over anhydrous magnesium sulfate and concentrated under reduced pressure to give compound (41) (5.83 g) as a crude product


The 4st step: A solution of 10% HCl/methanol (60 ml) was added to a solution of the compound (41) (5.83 g) obtained in the 3rd step in methanol (60 ml) cooled in ice with stirring and stirred at room temperature for 16 hours. The reaction solution was made alkaline by the addition of water and potassium carbonate, extracted with chloroform, the organic layer was dried over anhydrous sodium sulfate and concentrated under reduced pressure to give compound (42) (5.07 g) as a crude product.


The 5th step: Imidazole (2.24 g) and t-butyldimethylsilyl chloride (3.77 g) were added to a solution of the compound (42) (5.07 g) obtained in the 4th step in N,N-dimethylformamide (26 ml) with stirring at room temperature and the mixture was stirred for 1 hour and 40 minutes. After extraction with ethyl acetate, the organic layer was washed with saturated brine and dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give compound (43) (3.82 g).



1H-NMR (CDCl3) δ: −0.04 (3H, s), −0.01 (3H, s), 0.85 (9H, s), 1.51 (3H, s), 1.98 (2H, t J=6.0 Hz), 3.49-3.54 (1H, m), 3.65 (1H, dt, J11.1, 6.0 Hz), 8.02 (1H, t, J=2.2 Hz), 8.53 (1H, d, J=2.2 Hz), 8.63 (1H, d, J=2.2 Hz).


The 6th step: Toluene (25 ml) and water (13 ml) were added to compound (43) (3.82 g) and stirred under ice cooling. After the addition of potassium carbonate (5.14 g), thiophosgene (1.83 g) was added dropwise. After reacting for 2 hours, water was added, extracted with chloroform and the organic layer was dried over anhydrous magnesium sulfate. A part of the solvent was evaporated under reduced pressure to give compound (44) as a crude product.


The 7th step: Thionyl chloride (4.43 g) and N,N-dimethylformamide (0.08 ml) were added to a solution of the compound (7) obtained in the 6th step in toluene (25 ml) with stirring at room temperature. The mixture was stirred at 80° C. for 5 hours, concentrated under reduced pressure to give compound (45) (5.03 g) as a crude product.


The 8th step: 28% Ammonia water (60 ml) was added to a solution of the compound (45) (5.03 g) obtained in the 7th step in tetrahydrofuran (60 ml) stirred under ice cooling and the mixture was stirred at room temperature for 14 hours. The reaction solution was concentrated under reduced pressure to give compound (46) (4.92 g) as a crude product.


The 9th step: A mixture of the compound (46) (4.92 g) obtained in the 8th step, di-t-butyl dicarbonate (9.28 g), triethylamine (3.23 g), 4-dimethylaminopyridine (0.13 g) and tetrahydrofuran (106 ml) was stirred at room temperature for 3 days. The insoluble materials were filtered off, water was added to the filtrate and extracted with ethyl acetate. The organic layer was dried over anhydrous sodium sulfate and concentrated under reduced pressure to give compound (47) (8.31 g) as a crude product.


The 10th step: A mixture of the compound (47) (8.31 g) obtained in the 9th step, di-t-butyl dicarbonate (6.96 g), triethylamine (3.23 g), 4-dimethylaminopyridine (0.13 g) and tetrahydrofuran (50 ml) was stirred at room temperature for an hour. After the addition of water, it was extracted with ethyl acetate and the organic layer was dried over anhydrous magnesium sulfate. The solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give compound (48) (1.23 g).



1H-NMR (CDCl3) δ: 1.53 (18H, s), 1.60 (3H, s), 1.93 (1H, ddd, J=13.8, 9.4, 3.9 Hz), 2.06 (1H, ddd, J=13.8, 3.9, 1.9 Hz), 2.91 (1H, ddd, J=12.9, 3.9, 1.9 Hz), 3.15 (1H, ddd, J=12.9, 9.4, 3.9 Hz), 7.89 (1H, t, J=2.1 Hz), 8.55-8.57 (2H, m).


The 11th step: Compound (48) (190 mg), trisdibenzylideneacelonedipalladium (54 mg), dicyclohexylbiphenylphosphine (41 mg) were dissolved in toluene (5 ml), stirred at room temperaturea, and 1.6 M solution of lithium hexamethyldisilazide in tetrahydrofuran (0.73 ml) was added therein. The reaction solution was warmed up to 85° C. and stirred for 9 hours, then, it was cooled in ice and diethyl ether and a 1 N solution of hydrochloric acid were added. After stirring for 10 minutes, it was neutralized by the addition of a saturated aqueous solution of sodium carbonate and extracted with dichloromethane. The organic layer was dried over anhydrous sodium sulfate, concentrated under reduced pressure and the residue was purified with a silicagel column chromatography to give compound (49) (27 mg).



1H-NMR (CDCl3) δ: 1.51 (9H, s), 1.68 (3H, s), 2.12 (1H, ddd, J=14.8, 11.0, 3.0 Hz), 2.38-2.47 (1H, m), 2.64-2.70 (1H, m), 2.78-2.82 (1H, m), 3.80 (2H, br s), 6.90 (1H, t, J=2.4 Hz), 7.98 (1H, dd, J=10.4, 2.4 Hz).


Reference Example 7



embedded image


embedded image


The 1st step: A solution of compound (50) (38.93 g), (R)-(+)-2-methyl-2-propanesulfinamide (13.20 g) and tetraethyl orthotitanate (67.76 g) in tetrahydrofuran (389 ml) was reacted under heating to reflux for 4 hours. A saturated aqueous solution of ammonium chloride was added portionwise therein and the resulted insoluble materials were filtered off. The filtrate was concentrated and dried over anhydrous magnesium sulfate. The solvent was evaporated under reduced pressure to give compound (51) (30.52 g) as a crude product.



1H-NMR (CDCl3) δ: 1.32 (9H, s), 2.83 (3H, s), 7.55-7.65 (2H, m), 8.06 (1H, d, J=8.5 Hz).


The 2nd step: A solution of tert-butyl acetate (22.99 g) in tetrahydrofuran (148 ml) was added dropwise to a 2.0 M solution of lithium diisoprorylamide/n-heptane/ethylbenzene/tetrahydrofuran (202.5 ml) cooled in an acetone/dry ice bath with stirring. After stirring for 45 minutes, a solution of chlorotitanium triisopropoxide (108.36 g) in tetrahydrofuran (342 ml) was added dropwise and stirred for 40 minutes. A solution of the compound (51) (30.52 g) in tetrahydrofuran (342 ml) was added dropwise and reacted for an hour. The reaction solution was poured portionwise into an aqueous solution of ammonium chloride with stirring under ice cooling and the resulted insoluble materials were filtered off. It was extracted with ethyl acetate and the organic layer was dried over anhydrous magnesium sulfate. The solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give compound (52) (27.40 g).



1H-NMR (CDCl3) δ: 1.30 (9H, s), 1.35 (9H, s), 1.65 (3H, s), 3.01 (1H, d, J=16.5 Hz), 3.38 (1H, d, J=16.5 Hz), 5.60 (1H, s), 7.31 (1H, dd, J=5.9, 2.7 Hz), 7.48-7.50 (2H, m).


The 3rd step: Lithium aluminium hydride (5.67 g) was added porionwise to a solution of the compound (52) (22.40 g) in tetrahydrofuran (336 ml) stirred in an ice salt bath and stirred for 7 hours. After the addition of acetone, water and a 1 N aqueous solution of sodium hydroxide, the insoluble materials were filtered off and extracted with ethyl acetate. The organic layer was dried over anhydrous sodium sulfate and concentrated under reduced pressure to give compound (53) (18.75 g) as a crude product.


The 4th step: A solution of 10% HCl/methanol (94 ml) was added to a solution of the compound (53) (18.75 g) obtained in the 3rd step in methanol (94 ml) stirred under ice cooling and stirred at room temperature for 1.5 hours. The reaction solution was made alkaline by the addition of water and potassium carbonate and extracted with chloroform. The organic layer was dried over anhydrous sodium sulfate and concentrated under reduced pressure to give compound (54) (21.03 g) as a crude product.


The 5th step: Imidazole (5.49 g) and tert-butyldimethylsilyl chloride (10.53 g) were added to a solution of the compound (54) (21.03 g) in N,N-dimethylformamide (210 ml) stirred at room temperature and the mixture was stirred for an hour. After extraction with ethyl acetate, the organic layer was washed with saturated brine and dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give compound (55) (20.12 g).



1H-NMR (CDCl3) δ: −0.04 (3H, s), −0.02 (3H, s), 0.84 (9H, s), 1.47 (3H, s), 1.95-2.15 (2H, m), 3.54-3.63 (2H, m), 7.29 (1H, dd, J=6.1, 2.6 Hz), 7.45-7.48 (2H, m).


The 6th step: Toluene (66 ml) and water (33 ml) were added to compound (55) (10.06 g) and stirred under ice cooling. After the addition of potassium carbonate (11.13 g), thiophosgene (2.86 ml) was added dropwise. After reacting for an hours, water was added, extracted with chloroform and the organic layer was dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give compound (56) (9.43 g).



1H-NMR (CDCl3) δ: −0.03 (6H, s), 0.82 (9H, s), 1.80 (3H, s), 2.21-2.24 (1H, m), 2.44-2.48 (1H, m), 3.57 (1H, ddd, J=12.0, 5.8, 4.8 Hz), 3.71 (1H, ddd, J=12.0, 5.8, 4.8 Hz), 7.37 (1H, dd, J=7.5, 1.2 Hz), 7.48-7.58 (2H, m).


The 7th step: 28% Ammonia water (47 ml) was added to compound (56) 9.43 g) dissolved in tetrahydrofuran (94 ml) stirred at room temperature. After stining for 16 hours, water was added, extracted with ethyl acetate and the organic layer was dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure to give compound (57) (6.35 g) as a crude product.


The 8th step: Acetic acid (1.09 g) and a 1.0 M solution of tetrabutylammonium fluoride/tetrahydrofuran (18.20 mil) were added to a solution of the compound (57) (6.35 g) obtained in the 7th step in tetrahydrofuran (127 ml) stirred under ice cooling. After stirring at room temperature for 3 hours, water and potassium carbonate were added and extracted with ethyl acetate. The organic layer was dried over anhydrous sodium sulfate, concentrated under reduced pressure and the residue was purified with a silicagel column chromatography to give compound (58) (4.47 g).



1H-NMR (CDCl3) δ: 1.85 (3H, s), 2.27-2.31 (2H, br m), 3.73-3.83 (2H, m), 5.86 (2H, br s), 7.43 (1H, d, J=7.8 Hz), 7.52 (11H, d, J=7.8 Hz), 7.61 (1H, t, J=7.8 Hz), 7.81 (1H, br s).


The 9th step: 1-Chloro-N,N,2-trimethyl-1-propenylamine (2.16 g) was added to compound (58) (4.47 g) dissolved in dichloromethane (89 ml) stirred under ice cooling. After stirring at room temperature for 1.5 hours, water was added and extracted with dichloromethane. The organic layer was dried over anhydrous sodium sulfate, concentrated under reduced pressure and the residue was purified with a silicagel column chromatography to give compound (59) (2.91 g).



1H-NMR (CDCl3) δ: 1.53 (3H, s), 1.88 (1H, ddd, J=13.9, 10.1, 3.8 Hz), 2.40 (1H, ddd, J=13.9, 6.6, 3.8 Hz), 2.71 (1H, ddd, J=13.9, 10.1, 3.8 Hz), 2.95 (1H, tt, J=6.6, 3.8 Hz), 4.33 (2H, br s), 7.29 (1H, dd, J=7.5, 1.2 Hz), 7.41-7.50 (1H, m).


The 10th step: A mixture of compound (59) (2.91 g), di-tert-butyl dicarbonate (5.52 g), 4-dimethylaminopyridine (0.12 g) and tetrahydrofuran (29 ml) was stirred at room temperature for 2.5 hours. The reaction solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give compound (60) (1.23 g).



1H-NMR (CDCl3) δ: 1.53 (23H, s), 1.60 (3H, s), 1.93 (1H, ddd, J=13.8, 9.4, 3.9 Hz), 2.06 (1H, ddd, J=13.8, 3.7, 1.8 Hz), 2.91 (1H, ddd, J=12.7, 3.7, 1.9 Hz), 3.15 (1H, ddd, J=12.9, 9.2, 3.7 Hz), 7.89 (1H, t, J=2.1 Hz), 8.55-8.57 (2H, m).


The 11th step: Compound (60) (3.30 g), trisdibenzylideneacetonedipalladium (0.93 g), dicyclohexylbiphenylphosphine (0.73 g) were dissolved in toluene (66 ml), stirred at room temperature, and a 1.6 M solution of lithium hexamethyldisilazide in tetrahydrofuran (12.7 ml) was added therein. The reaction solution was warmed up to 80° C. and stirred for 8 hours, then, it was cooled in ice and diethyl ether and a 1 N solution of hydrochloric acid were added. After stirring for 5 minutes, it was neutralized by the addition of a saturated aqueous solution of sodium carbonate and extracted with ethyl acetate. The organic layer was dried over anhydrous sodium sulfate, concentrated under reduced pressure and the residue was purified with a silicagel column chromatography to give compound (61) (1.55 g).



1H-NMR (CDCl3) δ: 1.61 (3H, s), 1.74-1.80 (1H, m), 1.96-2.11 (1H, m), 2.64-2.82 (2H, m), 4.41 (2H, br s), 6.39 (1H, dd, J=8.1, 0.6 Hz), 6.71 (1H, dd, J=8.1, 0.6 Hz), 7.42 (1H, t, J=8.1 Hz).


Reference Example 8



embedded image


The 1st step: Compound (63) (3.31 g) prepared in the same manner as Reference Example 6 and 7 described above was dissolved in dichloromethane (16.5 ml), bis(2,4-dimethoxybenzyl)amine (4.45 g) was added and the solvent was evaporated under reduced pressure after stirring at room temperature for an hour and standing for 15 hours. The residue was purified with silicagel chromatography to give compound (63) (5.77 g).



1H-NMR (CDCl3): −0.10 (3H, s), −0.07 (3H, s), 0.77 (9H, s), 1.93 (3H, s), 2.08-2.27 (1H, m), 3.06-3.28 (1H, m), 3.38 (1H, ddd, J=10.8, 6.8, 6.8 Hz), 3.55 (1H, ddd, J=10.8, 6.8, 6.8 Hz), 3.78 (6H, s), 3.79 (6H, s), 4.81-5.05 (1H, br), 6.43-6.50 (4H, m), 7.07 (1H, d, J=1.9 Hz), 7.17 (2H, d, J=7.3; H-z), 8.05-8.16 (2H, m).


The 2nd step: Compound (63) (5.77 g) was dissolved in tetrahydrofuran (60 ml) and acetic acid (1.01 g) and 1 M tetrabutylammonium fluoride/tetrahydrofuran-solution (15 ml) were added and the mixture was stirred at room temperature for 150 minutes. Water was added, extracted with ethyl acetate and the organic layer was dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure and the residue was purified with a silicagel chromatography to give compound (64) (3.94 g).



1H-NMR (CDCl3): 1.99 (3H, s), 1.91-2.02 (1H, m), 3.10 (1H, ddd, J=14.6, 6.6, 5.2), 3.36-3.51 (2H, m), 3.78 (6H, s), 3.80 (6H, s), 4.58 (2H, d, J=15.8 Hz), 4.72 (2H, d, J=15.8 Hz), 6.43-6.51 (4H, m), 7.12 (1H, dd, J=5.3, 2.0 Hz), 7.18-7.29 (3H, m), 8.20 (1H, dd, J=5.3, 0.6 Hz), 8.28-8.31 (1H, br).


The 3rd step: Compound (64) (3.94 g) was dissolved in dichloromethane (20 ml) and 1-chloro-N,N′,2-trimethyl-1-propenylamine (1.86 mil) was added under ice cooling with stirring. After stirring at room temperature for 2 hours, water was added, extracted with chloroform and the organic layer was dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give a compound (65) (3.41 g).



1H-NMR (CDCl3): 1.49 (3H, s), 1.99 (1H, ddd, J=13.3, 7.6, 3.2 Hz), 2.31 (1H, ddd, J=13.3, 8.6, 3.7), 2.78 (1H, ddd, J=12.2, 8.6, 3.2 Hz), 3.04 (1H, ddd, J=12.2, 7.6, 3.7 Hz), 3.77 (6H, s), 3.79 (6H, s), 4.60 (2H, d, J=15.8 Hz), 4.76 (2H, d, J=15.8 Hz), 6.45-6.52 (4H, m), 7.08 (1H, dd, J=5.3, 2.1 Hz), 7.17-7.27 (3H, m), 8.40 (1H, dd, J=5.3, 0.5 Hz).


Reference Example 9



embedded image


The 1st step: Compound (66) (4.72 g) derived by a conventional method from an intermediate prepared in the same manner as the compound (27) described above was dissolved in tetrahydrofuran (150 ml) and a diethylether solution of methyl magnesium bromide (3M, 37 ml) was added dropwise with stirring under ice cooling in a nitrogen stream for 12 minutes. After stirring 3 hours, a saturated aqueous solution of ammonium chloride (190 ml) was added dropwise, extracted with ethyl acetate, the organic layer was washed with saturated brine and dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give a compound (67) (2.11 g).



1H-NMR (DMSO-d6): 0.75 (3H, s), 1.09 (3H, s), 1.21 (9H, s), 1.79 (3H, s), 2.06 (1H, m), 2.29 (1H, m), 4.97 (1H, s), 6.57 (1H, s), 7.17 (1H, dd, J=8.7, 12.0 Hz), 7.48-7.53 (1H, m), 7.99-8.03 (1H, m), 11.26 (1H, bs).


The 2nd step: Compound (67) (2.11 g) was dissolved in methanol (7.8 ml) and hydrochloric acid-methanol solution (5-10%)(15.6 ml) was added with stirring at room temperature, and the mixture was stirred for 1.5 hours. Then the reaction solution was poured into ice water and ethyl acetate (100 ml), a saturated aqueous solution of sodium bicarbonate (50 ml) was added and extracted with ethyl acetate. The aqueous layer was further extracted with ethyl acetate (50 ml), organic layers were combined, washed with saturated brine and dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure and the residue was crystallized from n-hexane to give compound (68) (1.42 g).



1H-NMR (DMSO-d): 0.65 (3H, s), 1.10 (3H, s), 1.43 (3H, s), 1.85 (1H, d, J=14.4 Hz), 2.17 (1H, dd, J=1.5, 14.4 Hz), 7.12 (1H, dd, J2.7, 12.0 Hz), 7.60-7.64 (1H, m), 7.90 (1H, dd, J=2.7, 7.5 Hz), 11.35 (1H, bs).


The 3rd step: Toluene (9.6 ml) and water (4.8 ml) were added to compound (68) (1.42 g) and suspended, potassium carbonate (2.13 g) was added with stirring under ice cooling and 2 minutes later thiophosgene (0.51 ml) was added at once and the stirring was continued.


The temperature was back to room temperature 40 minutes later, toluene (40 ml) and water were added and extracted an hour later. The aqueous layer was further extracted with toluene, organic layers were combined, washed with saturated brine and dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure to give a crude product (69) (2.02 g).


The 4th step: Tetrahydrofuran (17 ml) was added to triphenylphosphine (1.735 g) and N-chlorosuccinimide (833 mg), suspended in a nitrogen stream and stirred at room temperature for 10 minutes. A tetrahydrofuran (21 ml) solution of the crude product (69) (2.02 g) was added dropwise using a dropping funnel for 2 minutes. After stirring for 6 hours, the mixture was left stand at room temperature overnight. The solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give a compound (70) (828 mg).



1H-NMR (DMSO-d6): 1.54 (3H, s), 1.86 (3H, s), 2.81 (1H, d, J=13.8 Hz), 2.92 (1H, d, J=3.8 Hz), 4.73 (1H, s), 4.85 (11H, m), 7.28-7.35 (1H, m), 7.77-7.82 (2H, m), 11.39 (H, bs).


The 5th step: Compound (70) (828 mg) was dissolved in tetrahydrofuran (4 ml), conc. ammonia water (28%) (4 ml) was added with stirring under ice cooling and the temperature was back to room temperature after stirring for 5 minutes. After 25 hours, the reaction mixture was poured into ice water and extracted with ethyl acetate (50 ml). The organic layer was washed with saturated brine and dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure and the residue was purified with a silicagel chromatography to give a compound (71) (260 mg).



1H-NMR (DMSO-d6): 1.47 (3H, bs), 1.66 (3H, bs), 2.58 (1H, d, J=12.3 Hz), 4.71 (1H, s), 4.87 (3H, bs), 6.42 (1H, bs), 6.51 (1H, dd, J=2.7, 7.2 Hz), 6.75 (2H, bs), 7.54 (1H, bs).


The 6th step: Compound (71) (245 mg) was dissolved in chilled conc. sulfuric acid (4.9 ml) and stirred under ice cooling for 2 hours. The reaction solution was poured into ice water with stirring and pH was adjusted to 2-3 by the addition of a 5N aqueous solution of sodium hydroxide. Ethyl acetate (100 ml) and an aqueous solution of potassium carbonate were added and extracted under alkaline condition. The alkaline layer was further extracted with ethyl acetate (50 ml). Organic layers were combined, washed with saturated brine (50 ml) and dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give a compound (72) (101 mg) as a crystal.



1H-NMR (DMSO-d6): 0.83 (3H, s), 1.27 (3H, s), 1.44 (3H, s), 1.54 (1H, d, J=14.1 Hz), 2.45 (1H, d, J=14.1 Hz), 4.79 (2H, s), 5.89 (2H, bs), 6.32-6.37 (1H, m), 6.58 (1H, dd, J=2.7, 7.2 Hz), 6.72 (1H, dd, J=8.7, 12.3 Hz).


Reference Example 10



embedded image


embedded image


The 1st step: Ethyl bromodifluoroacetate (0.77 ml) was added to a suspension of zinc dust (392 mg) in tetrahydrofuran (4 ml) with stirring in a nitrogen stream at room temperature, stirred for 15 minutes, ethyl bromodifluoroacetate (0.29 ml) was added, stirred for 30 minutes to prepare a solution of ethyl bromozincdifluoroacetate. This solution was added to a solution of compound (73) in tetrahydrofuran (3 ml) in a nitrogen stream and stirred for 8 hours. 3% Ammonia water was added to the reaction mixture with stirring under ice cooling, extracted with ethyl acetate, the organic layer was washed with saturated brine and dried over anhydrous magnesium sulfate. The solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give a compound (74) (696 mg).



1H-NMR (DMSO-d6) δ: 1.17 (3H, t, J=7.2 Hz), 1.18 (9H, s), 2.00 (3H, brs), 4.24 (2H, q, J=7.2 Hz), 5.56 (1H, brs), 7.56 (dd, J=9.0, 11.7 Hz), 8.36 (1H, m), 8.49 (1H, dd, J=3.0, 6.6 Hz).


The 2nd step: Compound (74) (670 mg) was dissolved in tetrahydrofuran (6.7 ml) and lithium borohydride (71 mg) was added in a nitrogen stream with stirring under ice cooling. After stirring for 30 minutes, acetic acid (198 mg) and ice water were added to the reaction mixture, extracted with ethyl acetate, the organic layer was washed with brine and dried over anhydrous magnesium sulfate. The solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give a compound (75) (401 mg).



1H-NMR (DMSO-d6) δ: 1.20 (9H, s), 2.00 (3H, d, J=3.6 Hz), 3.80 (1H, m), 4.00 (1H, m), 5.99 (1H, s), 6.34 (1H, 1, J=5.7 Hz), 7.53 (1H, dd, J=9.0, 12.0 Hz), 831 (1H, m), 8.50 (1H, dd, J=2.7, 6.6 Hz).


The 3rd step: Compound (75) (394 mg) was dissolved in methanol (3 ml), and 4N—HCl/1,4-dioxane (1.35 ml) was added with stirring under ice cooling. After stirring for 30 minutes, the mixture was stirred at room temperature for 1.5 hours. Ice water was added to the reaction solution and washed with ethyl acetate. The aqueous layer was made alkaline by the addition of a 2M aqueous solution of potassium carbonate and extracted with ethyl acetate. The organic layer was dried over anhydrous magnesium sulfate, the solvent was evaporated under reduced pressure to give compound (76) (293 mg).



1H-NMR (DMSO-d6) δ: 1.62 (3H, d, J=2.7 Hz), 2.62 (2H, brs), 3.65-3.83 (2H, m), 5.31 (1H, brt), 7.44 (1H, dd, J=9.0, 11.4 Hz), 8.23 (1H, m), 8.59 (1H, dd, J=3.0, 6.9 Hz).


The 4th step: Compound (76) (266 mg) was dissolved in acetone (3 ml) and benzoyl isothiocyanate (164 mg) was added in a nitrogen stream with stirring under ice cooling. After stirring for an hour, the mixture was stirred at room temperature for an hour. The reaction solution was concentrated under reduced pressure and the residue was purified with a silicagel column chromatography to give a compound (77) (353 mg).



1H-NMR (DMSO-d6) δ: 2.30 (3H, brs), 3.65-3.96 (2H, m), 5.90 (1H, brt), 7.42-7.68 (4H, m), 7.93-7.96 (2H, m), 8.17-8.33 (2H, m), 11.42 (1H, brs), 12.31 (1H, brs).


The 5th step: Compound (77) (348 mg) was dissolved in dichloromethane (4 ml) and 1-chloro-N,N-2-trimethyl-1-propenylamine (131 mg) was added in a nitrogen stream with stirring under ice cooling. After stirring for 15 hours at room temperature, ice water was added and neutralized by the addition of potassium carbonate. It was extracted with ethyl acetate, washed with brine and dried over anhydrous magnesium sulfate. The solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give a compound (78) (308 mg).



1H-NMR (CDCl3) δ: 1.89 (3H, d, J=3.0 Hz), 3.17 (1H, ddd, J=8.4, 10.2, 13.2 Hz), 3.51 (1H, ddd, J=6.0, 13.2, 19.2 Hz), 7.23 (1H, dd, J9.0, 10.8 Hz), 7.49-7.64 (3H, m), 7.91 (2H, d, J=7.2 Hz), 8.24 (1H, m), 8.43 (1H, dd, J=3.0, 6.6 Hz), 8.57 (1H, br).


The 6th step: Compound (78) (297 mg) was dissolved in ethanol (4 ml), water (1.5 ml) and conc. hydrochloric acid (1.5 ml) were added and the mixture was stirred at 90° C. for 2.5 hours. Water was added to the reaction solution, washed with ethyl acetate and the aqueous layer was made alkaline by the addition of a 2M aqueous solution of potassium carbonate. It was extracted with ethyl acetate, washed with brine and the organic layer was dried over anhydrous magnesium sulfate. The solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give a compound (79) (89 mg).



1H-NMR (CDCl3) δ: 1.85 (3H, d, J=3.6 Hz), 3.15 (1H, ddd, J=8.7, 10.5, 12.9 Hz), 3.50 (1H, ddd, J=5.4, 12.9, 18.3 Hz), 4.51 (2H, brs), 7.19 (1H, dd, J=9.0, 11.1 Hz), 8.20 (1H, ddd, J=3.0, 6.9, 9.0 Hz), 8.54 (1H, dd, J=3.0, 6.9 Hz).


The 7th step: Compound (79) (82 mg) was dissolved in dichloromethane (1 ml), di-tert-butyldicarbonate (176 mg) and 4-dimethylaminopyridine (4 mg) were added and the mixture was stirred at room temperature for 30 minutes. The reaction solution was concentrated under reduced pressure and the residue was purified with a silicagel column chromatography to give a compound (80) (101 mg).



1H-NMR (CDCl3) δ: 1.56 (18H, S), 1.90 (3H, d, J=3.6 Hz), 3.27 (1H, ddd, J=6.6, 9.3, 12.9 Hz), 3.69 (1H, ddd, J=4.2, 12.9, 17.4 Hz), 7.23 (1H, dd, J=9.0, 12.0 Hz), 8.24 (1H, ddd, J=3.0, 9.0, 12.0 Hz), 8.41 (1H, ddd, J=2.4, 3.0, 6.0 Hz).


The 8th step: Compound (80) (4.76 g) was dissolved in methanol (70 ml), 10% Pd—C(containing 50% water) (238 g) was added and the mixture was stirred in a hydrogen atmosphere for 20 hours. The catalyst was filtered off, the solvent was evaporated under reduced pressure to give compound (81) (4.43 g)



1H-NMR (CDCl3) δ: 1.54 (18H, S), 1.85 (3H, d, J=2.4 Hz), 3.24 (1H, m), 3.44 (1H, m), 3.53 (2H, brs), 6.61 (1H, m), 6.82-6.89 (2H, m).


Reference Example 11



embedded image


embedded image


The 1st step: A solution of 2.0M lithium diisopropylamide/n-heptane/ethyl benzene (182 ml) in tetrahydrofuan (150 ml) was cooled in a dry ice-acetone bath, and a solution of methyl isobutyrate (27.17 g) in tetrahydrofuran (90 ml) was added dropwise with stirring. After stirring for 40 minutes, a solution of chlorotitanium triisopropoxide (97.07 g) in tetrahydrofuran (300 ml) was added dropwise. After stirring for 15 minutes, a solution of compound (86) (25.39 g) in tetrahydrofuran (150 ml) was added dropwise. After the reaction for 2.5 hours, the reaction mixture was poured portionwise into an aqueous solution of ammonium chloride stirred under ice cooling and the formed insoluble materials were filtered. It was extracted with ethyl acetate, and the organic layer was dried over anhydrous magnesium sulfate. The solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give a compound (87) (23.98 g).



1H-NMR (CDCl3) δ: 1.12 (3H, s), 1.22 (3H, s), 1.35 (9H, s), 1.99 (3H, d, J=5.8 Hz), 3.75 (3H, s), 5.65 (1H, s), 7.20 (OH, dd, J=11.5, 8.9 Hz), 8.18-8.21 (1H, m), 8.45 (1H, dd, J=6.9, 2.9 Hz).


The 2nd step: Compound (87) (391 mg) was dissolved in tetrahydrofuran (4 ml) and lithium borohydride (44 mg) was added in 3 minutes in a nitrogen stream with stirring at room temperature. After stirring for 2 hours, lithium borohydride (22 mg) was further added and the stirring was continued. After stirring for 2 hours, a saturated aqueous solution of ammonium chloride was slowly added to the reaction solution with stirring under ice cooling, extracted with ethyl acetate 5 minutes later, the organic layer was washed with saturated brine and dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give a compound (88) (175 mg).



1H-NMR (DMSO-d6) δ: 0.65 (3H, d, J=1.8 Hz), 0.93 (3H, s), 1.22 (9H, s), 1.93 (3H, d, J=6.6 Hz), 3.24 (1H, d, J=8.4 Hz), 3.74 (1H, d, J=8.4 Hz), 5.96 (1H, bs), 6.75 (1H, s), 7.47 (1 μl, dd, J=9.0, 12.0 Hz), 8.23 (1H, ddd, J=3.0, 3.0, 9.0 Hz), 8.39 (1H, dd, J=3.0, 6.9 Hz).


The 3rd step: Compound (88) (331 mg) was dissolved in methanol (1.5 ml), and a hydrogen chloride-methanol solution (5-10%) (3 ml) was added with stirring at room temperature. After stirring for 1.5 hours, the reaction solvent was evaporated under reduced pressure. The residue was dissolved in ethyl acetate-methanol (9:1), poured into ice water, and a saturated aqueous solution of sodium bicarbonate (4 ml) was added, extracted, washed with saturated brine and the organic layer was dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure and the powder obtained by the addition of n-hexane to the solid was filtered to give a compound (89) (207 mg).



1H-NMR (DMSO-d6) δ: 0.80 (6H, s), 1.59 (3H, d, J=4.5 Hz), 3.16 (1H, d, J=10.8 Hz), 7.38 (1H, dd, J=9.0, 12.0 Hz), 8.17 (1H, ddd, J=3.0, 3.0, 9.0 Hz), 8.64 (1H, dd, J=3.0, 6.9 Hz)


The 4th step: Compound (89) (150 mg) was dissolved in acetone (3 ml) and benzoyl isothiocyanate (0.079 ml) was added in a nitrogen stream with stirring under ice cooling. After stirring for 2 hours, the reaction solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give a compound (90) (236 mg).


LCMS: 420 m/z [M+H]+


The 5th step: Compound (90) (233 mg) was dissolved in dichloromethane (4 ml) and chloropropenylamine (0.081 ml) was added at once in a nitrogen stream with stirring at room temperature. After stirring for 23 hours, the reaction solution was poured into ice water, extracted with ethyl acetate, washed with saturated brine and the organic layer was dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give a compound (91) (128 mg).



1H-NMR (DMSO-d6) δ: 0.83 (3H, s), 1.12 (3H, s), 1.72 (3H, s), 2.69 (1H, d, J=13.2 Hz), 2.90-3.10 (1H, m), 7.44-7.58 (4H, m), 8.00 (2H, d, J=7.5 Hz), 8.23-8.35 (2H, m), 10.75 (1H, bs).


The 6th step: Compound (91) (20 mg) was suspended in 99.5% ethanol (0.4 ml), 6N hydrochloric acid (0.2 ml) was added and the mixture was stirred in a oil bath beated to 90° C.


After stirring for 17 hours, the reaction solution was poured into water, and extracted with ethyl acetate. The aqueous layer was made alkaline by the addition of a saturated aqueous solution of potassium carbonate (pH=11), extracted with ethyl acetate, washed with saturated brine and the organic layer was dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure to give compound (92) (14 mg).



1H-NMR (DMSO-d6) δ: 0.72 (3H, s), 1.00 (3H, d, J=3.6 Hz), 1.54 (3H, d, J=4.8 Hz), 2.61 (1H, d, J=12.3 Hz), 3.09 (H, d J=12.3 Hz), 5.98 (2H, s), 7.41 (1H, dd, J=9.0, 11.7 Hz), 8.16-8.21 (1H, m), 8.42 (1H, dd, J=3.0, 6.9 Hz).


The 7th step: Compound (92) (12 mg) was dissolved in dichloromethane (0.1 ml) and a di-tert-butyldicarbonate-dichloromethane solution (0.0966M, 1.2 ml) was added with stirring at room temperature. After stirring for 2 hours, the reaction solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give a compound (93) (15 mg).



1H-NMR (DMSO-d6) δ: 0.70 (3H, s), 1.02 (3H, s), 1.43 (9H, s), 1.56 (3H, bs), 2.61 (1H, d, J=2.9 Hz), 3.16 (1H, m), 7.45 (1H, dd, J=9.0, 11.4 Hz), 8.20-8.24 (1H, m), 8.35 (1H, m), 9.87 (1H, bs).


The 8th step: Methanol (4.1 ml) was added to compound (93) (823 mg), suspended, and 10% Pd—C (50% wet) (412 mg) was added. A catalytic reduction was carried out at normal pressure, and methanol (8.2 ml) was added when a solid was precipitated and the reduction was further continued. After 23 hours, the catalyst was filtered through a Celite pad, washed with warm methanol, and the washings were combined. The solvent was evaporated under reduced pressure and the powder precipitated by the addition of diisopropylether to the residue was filtered to give compound (94) (638 mg).



1H-NMR (DMSO-d6) δ: 0.87 (3H, bs), 1.06 (3H, bs), 1.39 (9H, s), 1.57 (3H, bs), 2.66-2.72 (2H, m), 4.97 (2H, bs), 6.45-6.47 (2H, m), 6.78 (1H, m), 9.65 (1H, bs).


Reference Example 12



embedded image


The 1st step: 3-(Trifluoromethyl)-1H-pyrazole (591 mg) was dissolved in dimethylformamide (7 ml), potassium carbonate (601 mg) and compound (104) (500 mg) were added thereto and stirred at room temperature overnight. The reaction was quenched by an addition of water. The insoluble materials were filtered and washed with diisopropylether. The resulted solid was dried under reduced pressure to give compound (105) (644 mg).



1H-NMR (CDCl3) δ: 4.08 (3H, s), 6.81 (1H, d, J=2.5 Hz), 8.65 (1H, s), 9.14 (1H, s), 9.45 (1H, s).


The 2nd step: Compound (105) (640 mg) was added to a mixed solvent of water-methanol (6 ml, 1:1), lithium hydroxide (84 mg) was added and the mixture was stirred at room temperature for 4 hours. The reaction solution was acidified by the addition of 2N hydrochloric acid, the insoluble materials were filtered off and washed with diisopropylether.


The resulted solid was dried under reduced pressure to give compound (106) (343 mg).



1H-NMR (DMSO-d6) δ: 7.20 (1H, d, J=2.5 Hz), 8.93 (1H, s), 9.12 (1H, s), 9.33 (1H, s).


Reference Example 13



embedded image


The 1st step: A mixture of compound (107) (1000 mg), dioxane (2 ml), and 28% ammonia water (2 ml) was stirred at 50° C. for 19 hours. The reaction solution was concentrated under reduced pressure. Water was added to the residue, extracted with ethyl acetate and dried over anhydrous magnesium sulfate. The solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give a compound (108) (476 mg).



1H-NMR (CDCl3) δ: 1.63 (9H, s), 5.04 (2H, br s), 8.03 (1H, s), 8.69 (1H, s).


The 2nd step: 3-Bromo-2-oxopropanoic acid ethyl ester (1582 mg) was added to compound (108) (475 mg) in dimethoxyethane (4 ml) and the mixture was stirred at 75° C. for 2.5 hours. The reaction solution was diluted with diisopropylether, the insoluble materials were filtered, washed with diisopropylether and hexane, and dried under reduced pressure. The residue was stirred in tert-butyl alcohol (7.5 ml) at 95° C. for 2 hours. The solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give compound (109) (709 mg).



1H-NMR (CDCl3) δ: 1.46 (3H, t, J=7.1 Hz), 1.66 (9H, s), 4.50 (2H, q, J=7.1 Hz), 8.35 (1H, s), 8.89 (1H, s), 9.24 (1H, s).


The 3rd step: A mixture of compound (09) (270 mg), dioxane (3 ml) and 28% ammonia water (2.5 ml) was stirred in a pressure bottle at 50° C. for 6 hours. The reaction solution was concentrated under reduced pressure to give a crude product of compound (110) (249 mg).



1H-NMR of the crude product (CDCl3) δ: 1.67 (9H, s), 5.79 (1H, br s), 8.35 (1H, s), 8.90 (1H, s), 9.15 (1H, s).


The 4th step: 2,2,2-Trichloroacetyl chloride (253 mg) was added at 0° C. to a mixture of compound (10) (146 mg), triethylamine (282 mg) and dimethylaminopyridine (6.8 mg) in tetrahydrofuran (9 ml), and the mixture was stirred at room temperature for 2 hours. The reaction solution was diluted with ethyl acetate and the reaction was quenched by the addition of a saturated aqueous solution of sodium bicarbonate. It was extracted with ethyl acetate, dried over anhydrous magnesium sulfate and the solvent was evaporated under reduced pressure to give compound (111) (99 mg) as a crude product.


The 5th step: Compound (111) (95 mg) was dissolved in chloroform (3 ml), trifluoroacetic acid (1330 mg) was added and the mixture was stirred at room temperature for 4 hours. The reaction solution was concentrated under reduced pressure to give a crude product. The residue was suspended with ethyl acetate and diisopropylether and the insoluble materials were filtered and washed with diisopropylether. The residue was dried under reduced pressure to give a composition including compound (112).


Reference Example 14



embedded image


embedded image


The 1st step: A 2.6M n-butyl lithium/hexane solution (9.38 ml) was added dropwise for 10 minutes to diisopropylamine (2.75 g) dissolved in tetrahydrofuran (25 ml) under stirring in a dry ice/acetone bath. After stirring in a ice bath for 10 minutes and in a dry ice/acetone bath for 10 minutes, tert-butyl α-benzyloxyacetate (5.21 g) dissolved in tetrahydrofuran (25 ml) was added dropwise for 30 minutes. After stirring for 40 minutes, chlorotitaniumtriisopropoxide (6.60 g) dissolved in tetrahydrofuran (50 ml) was added dropwise. After stirring for 30 minutes, compound (73) (2.68 g) dissolved in tetrahydrofuran (50 ml) was added dropwise for 10 minutes and stirred for 90 minutes. A suspension of ammonium chloride (7.52 g) in tetrahydrofuran-water (1:1, 40 ml) was stirred at room temperature, and the reaction mixture was added thereto at once and the precipitated insoluble materials were filtered. The filtrate was extracted with ethyl acetate, the organic layer was dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give compound (4.49 g).



1H-NMR (CDCl3) δ: 1.14 (3.6H, s), 1.22 (3.6H, s), 1.27 (5.4H, s), 1.39 (5.4H, s), 1.96 (1.2H, s), 1.99 (1.8H, s), 4.31 (0.4H, s), 4.34 (0.6H, d, J=1.6 Hz), 4.41 (0.4H, d, J=11.6 Hz), 4.45 (0.6H, s), 4.56 (0.4H, s), 4.68 (0.6H, d, J=11.6 Hz), 4.81 (0.4H, d, J=11.6 Hz), 5.01 (0.6H, s), 7.06-7.38 (6H, m), 8.18 (0.6H, d, J=8.8 Hz), 8.24 (0.4H, d, J=9.1 Hz), 8.42-8.47 (1H, m).


The 2nd step: Compound (113) (4.49 g) was dissolved in trifluoroacetic acid (44 ml), stirred at room temperature for an hour and the solvent was evaporated under reduced pressure. The resulted residue was dissolved in 10% hydrochloric acid-methanol (44 ml), stirred at room temperature overnight and the reaction solution was concentrated under reduced pressure. The residue was dissolved in tetrahydrofuran (22 ml) and a solution of 1 M-borane-tetrahydrofuran complex in tetrahydrofuran (44.1 mJ) was added dropwise for 15 minutes under ice cooling, and the mixture was stirred at room temperature for 2.5 hours. Water (50 ml) was added therein with stirring under ice cooling, stirred for 15 minutes and ethyl acetate (50 ml) and potassium carbonate (16 g) were added. It was extracted with ethyl acetate and the organic layer was dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure and the obtained compound (114) (3.27 g) was used in the next step without purification.


The 3rd step: Benzoyl isothiocyanate (1.41 ml) was added to compound (114) (3.27 g) in methylenechloride (16.5 ml), stirred at room temperature for an hour and the solvent was evaporated under reduced pressure. The residue was purified with a silicagel column chromatography to give compound (115) (3.14 g).



1H-NMR (CDCl3) δ: 2.14 (1.35H, s), 2.21 (1.65H, s), 3.73-4.07 (3H, m), 4.43 (0.55H, d, J=11.5 Hz), 4.63 (0.55H, d, J=11.5 Hz), 4.74 (0.45H, d. J=11.5 Hz), 4.78 (0.45H, d, J=11.5 Hz), 7.20-7.38 (4H, m), 7.43-7.51 (2H, m), 7.56-7.63 (1H, m), 7.75-7.86 (2H, m), 8.08-8.17 (1H, m), 8.24-8.34 (1H, m), 8.91-9.01 (1H, m), 11.81 (0.55H, s), 11.90 (0.45H, s).


The 4th step: α-Chlorotetramethylenamine (1.67 ml) was added to compound (115) (3.14 g) in methylenechloride (15.5 ml), stirred at room temperature for 30 minutes and pH was adjusted to over 11 by the addition of water (15 ml) and potassium carbonate. It was extracted with chloroform and the organic layer was dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give compound (116) (2.66 g).



1H-NMR (CDCl3) δ: 1.58 (3 bH, s), 1.81 (3 aH, s), 2.76 (bH, dd, J=13.4, 1.8 Hz), 3.09 (bH, dd, J=13.4, 6.1 Hz), 3.16 (aH, dd, J=13.8, 3.9 Hz), 3.35 (aH, dd, J=13.8, 1.8 Hz), 4.21-4.25 (aH, m), 4.28 (aH, d, J=12.4 Hz), 4.33-4.38 (bH, m), 4.49-4.56 (a+bH, m), 4.73 (bH, d, J=11.9 Hz), 6.83-7.60 (10H, m), 7.91-8.23 (3H, m), 8.25-8.30 (bH, m), 8.74 (aH, m).


The 5th step: Hydrazine monohydrate (0.73 ml) was added to compound (116) (1.44 g) in ethanol (7.2 ml) and stirred at room temperature for 2 hours. Water was added, extracted with ethyl acetate and the organic layer was dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure and the obtained compound (117) (1.14 g) was used in the next step without purification.


The 6th step: A solution of di-tert-butyl dicarbonate (1.65 g) in methylenechloride (5.5 ml) and 4-dimethylaminopyridine (37 mg) were added to compound (117) (1.14 g) in methylenechloride (5.5 ml) and stirred at room temperature for an hour. The solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give compound (118) (1.52 g).



1H-NMR (CDCl3) δ: 1.47 (8.1H, s), 1.51 (9.9H, s), 1.53 (1.35H, s), 1.75 (1.65H, s), 3.01-3.49 (2H, m), 3.81-3.86 (0.55H, m), 4.07-4.09 (0.45H, m), 4.17 (0.45H, d, J=12.1 Hz), 4.25 (0.55H, d, J=1.16 Hz), 4.41 (0.45H, d, J=12.1 Hz), 4.49 (0.55H, d, J=11.6 Hz), 6.73-6.78 (1H, m), 6.94-7.23 (5H, m). 8.11-8.18 (1H, m), 8.22-8.27 (0.55H, m), 8.51-8.55 (0.45H, m).


The 7th step: 20 w/w % Palladium hydroxide supported by carbon (40 mg) was added to a solution of compound (118) (211.3 mg) in ethanol (2 ml), stirred in a hydrogen atmosphere of 1 atom at room temperature for 22 hours and filtered through a Celite pad. The filtrate was concentrated under reduced pressure and the residue was purified with a silicagel column chromatography to give compound (119) (149.1 mg).



1H-NMR (CDCl3) δ: 1.46 (8.1H, s), 1.51 (9.9H, s), 1.54 (1.35H, s), 1.74 (1.65H, s), 2.91 (0.55H, d, J=12.9 Hz), 3.02 (0.55H, dd, J=12.9, 6.3 Hz), 3.15 (0.45H, dd, J=13.3, 3.0 Hz), 3.37-3.73 (2H, br), 3.43 (0.45H, d, J=13.3 Hz), 4.13-4.18 (1H, m), 4.22 (0.45H, d, J=11.9 Hz), 4.34 (0.45H, d, J=11.9 Hz), 4.49 (0.55H, d, J=11.6 Hz), 4.59 (0.55H, d, J=11.6 Hz), 6.45-6.61 (1H, m), 6.71-7.39 (7H, m).


Example 1
Preparation of Compound 46



embedded image


Compound (34) (125 mg) and DMT-MM (162 mg) were suspended in methanol (1.2 ml), stirred at room temperature for 30 minutes and compound (33) (117 mg) was added therein. After stirring for 5 hours, the product was isolated by a silicagel thin-layer chromatography to give the objective compound (46) (13.5 mg).



1H-NMR (DMSO-d6) δ: 1.47 (3H, s), 1.81 (1H, d, J=11.6 Hz), 2.12 (1H, bs), 2.54-2.59 (1H, m), 2.97 (1H, bs), 3.28 (2H, d, 6.4 Hz), 3.52 (2H, d, 6.5 Hz), 3.88 (3H, s), 5.69 (2H, s), 7.09 (1H, dd, J=11.8, 6.8 Hz), 7.50 (1H, s), 7.60 (1H, d, J=7.8 Hz), 7.67 (1H, s), 10.06 (1H, s).


Example 2
Preparation of Compound 86



embedded image


The 1st step: Sodium hydride (302 mg) is added to DMF (3.0 ml) and 3-butene alcohol (3.0 ml) under ice cooling in a nitrogen atmosphere. After stirring at room temperature for 1.0 hour, compound (35) (300 mg) was added and stirred under heating at 65° C. After 7 hours, the reaction solution was neutralized by the addition of 2M hydrochloric acid and concentrated under reduced pressure. Water was added to the resulted residue and filtered to give compound (36) (87 mg, 23.7%).


The 2nd step: Compound (36) (65.8 mg) and compound (37) (50 mg) were dissolved in methanol (2.0 ml), DMT-MM (93.7 mg) was added and the mixture was stirred at room temperature. After 6 hours, the solvent was evaporated under reduced pressure and the residue was purified with a column chromatography using chloroform/methanol to give compound (86) (40 mg, 44.5%).



1H-NMR (DMSO-d6) δ: 1.65 (3H, s), 2.03-2.09 (1H, m), 2.34-2.38 (1H, m), 2.51-2.61 (2H, m), 3.10-3.13 (1H, m), 3.57 (2H, t, J=4.4 Hz), 4.45 (2H, t, J=6.4 Hz), 5.13 (21, dd, J=29.1, 13.9 Hz), 5.83-5.92 (1H, m), 7.08 (1H, d, J=7.8 Hz), 7.40 (1H, t, J=8.0 Hz), 7.84 (1H, s), 7.91 (1H, d, J=8.1 Hz), 8.36 (1H, s), 8.87 (1H, s), 10.56 (1H, s).


Example 3



embedded image


A carboxylic acid, R—COOH corresponding to the objective compound (0.115 mmol) and 4-(4,6-dimethoxy-1,3,5-triazin-2-yl)-4-methylmorpholinium chloride (0.106 mmol) were dissolved in methanol (0.4 ml) and stirred by shaking at room temperature for 1.5 hours. A solution of compound A (0.0884 mmol) in methanol (0.4 ml) was added and the mixture was stirred for 8 hours. The reaction solvent was concentrated, dissolved in ethyl acetate (1 ml) and dimethylsulfoxide (0.5 ml), a 2N aqueous solution of sodium hydroxide (1 ml) was added and stirred by shaking for 2 hours. The organic layer was separated and concentrated to give a crude product of compound B. Trifluoroacetic acid (0.3 ml) was added and stirred by shaking at room temperature for 14 hours, dimethylsulfoxide (0.4 ml) was added and the product was purified with preparative LC/MS to give the objective compound C.


Example 4
Preparation of Compound 668



embedded image


The 1st step: Compound (82) (506 mg) was dissolved in chloroform (30.0 m), an aqueous solution (10.0 ml) of sodium bicarbonate (851 mg) and thiophosgene (0.111 ml) were added and stirred under ice cooling for 40 minutes. The organic layer was separated from the reaction solution and dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure to give compound (83) (457 mg).



1H-NMR (DMSO-d6) δ: 1.44 (9H, s), 1.51 (3H, s), 1.60 (1H, s), 2.17 (1H, s), 2.68 (1H, s), 3.05 (1H, s), 7.30 (1H, t, J=10.1 Hz), 7.42 (1H, s), 7.58 (1H, s).


The 2nd step: Compound (83) (240 mg) was dissolved in methylelechloride (3.60 ml), pyridine-2-ylmethanamine (74.8 mg) and triethylamine (0.192 ml) were added and the mixture was stirred at room temperature for 40 minutes. The reaction solution was washed with distilled water, the separated organic layer was dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure to give compound (84) (210 mg).



1H-NMR (DMSO-d6) δ: 1.46 (9H, s), 1.65 (3H, s), 2.05 (2H, s), 2.57 (1H, s), 2.97 (1H, s), 4.86 (2H, s), 7.29 (3H, m), 7.40 (1H, d, J=7.3 Hz), 7.66 (1H, s), 7.84 (1H, s), 8.27 (1H, s), 8.58 (1H, s), 9.96 (1H, s).


The 3rd step: Compound (84) (95.1 ml) was dissolved in toluene (1.50 ml), dicyclohexylcarbodiimide (40.1 mg) was added and the mixture was stirred under irradiation of microwave at 100° C. for 20 minutes. The reaction solution was concentrated under reduced pressure and the residue was purified with a column chromatography to give compound (85) (38.0 mg).



1H-NMR (DMSO-d6) δ: 1.40 (9H, s), 1.61 (3H, s), 1.94 (2H, s), 2.57 (1H, s), 2.88 (1H, s), 6.55 (1H, d, J=6.3 Hz), 6.59 (1H, d, J=8.6 Hz), 7.07 (1H, d, 8.6 Hz), 7.13 (2H, s), 7.29 (1H, s), 7.41 (1H, s, J=9.3; H), 7.96 (1H, d, J=6.8 Hz), 8.85 (1H, s).


The 4h step: Compound (85) (38.0 mg) was dissolved in chloroform (0.50 ml), trifluoroacetic acid (1.00 ml) was added and stirred at room temperature for 2 hours. The reaction solution was extracted with a mixture of chloroform/methanol and washed with an aqueous solution of potassium carbonate and distilled water. The separated organic layer was dried over anhydrous sodium sulfate and the solvent was evaporated under reduced pressure. Diisopropylether was added to the residue and the precipitated powder was filtered to give compound (668) (9.39 mg).



1H-NMR (DMSO-d6) δ: 1.58 (3H, s), 1.90 (1H, s), 2.44 (1H, s), 2.62 (1H, t, J=9.7 Hz), 3.06 (1H, s), 6.57 (2H, td, J−15.0, 6.3 Hz), 7.05 (1H, dd, J=12.1, 10.6 Hz), 7.15 (1H, s), 7.24 (1H, d, J=5.3 Hz), 7.31 (1H, dd, J=7.7, 3.7 Hz), 7.42 (1H, d, J-8.8 Hz), 7.99 (1H, d, J=6.8 Hz), 8.85 (1H, s).


Example 5
Preparation of Compound 674



embedded image


embedded image


The 1st step: A solution of 20M lithium diisopropylde/n-hepetane/ethylbenzene (172 ml) in tetrahydrofuran (280 m) was cooled in a dry ice/acetone bath and a solution of with stirring. After stirring for an hour, a solution of chlorotitanium triisopropoxide (92) compound (73) (24.56 g) in tetrahydrofuran (120 ml) was added dropwise. After reaction for 2 hours, the reaction solution was added portionwise to an aqueous solution of ammonium chloride with stirring under ice cooling and the precipitated insoluble materials were filtered. It was extracted with ethyl acetate, the organic layer was dried over anhydrous sodium sulfate.


The solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give compound (95) (15.49 g).



1H-NMR (CDCl3) δ: 1.16-1.19 (2H, m), 1.24 (9H, s), 1.28-1.32 (2H, m), 1.36 (9H, s), 1.46 (3H, s), 1.50-1.55 (2H, m), 1.64-1.72 (2H, m), 5.45 (1H, s), 7.11-7.16 (1H, m), 8.11-8.16 (1H, m), 8.67 (1H, dd, J=6.9, 2.9 Hz).


The 2nd step: 2.0 M Hydrochloric acid/ethyl acetate (30 ml) was added to compound (95) (2.48 g) and stirred at 65° C. for 5.5 hours. Diisopropylether was added and the precipitated solid was filtered to give a crude product of compound (96) (1.66 g).


The 3rd step: A solution of compound (96) (1.66 g) in tetrahydrofuran (8.3 ml) was stirred under ice cooling and a solution of 1M borane/tetrahydrofuran (21.8 ml) was added and the mixture was stirred at room temperature for 2 hours and 45 minutes. Ice and sodium bicarbonate were added, extracted with ethyl acetate and the organic layer was dried over anhydrous magnesium sulfate. The solvent was evaporated under reduced pressure to give a crude product of compound (97) (1.36 g).


The 4th step: A solution of compound (97) (1.36 g) in acetone (20 ml) was stirred under ice cooling, a solution of benzoyl isothiocyanate (0.92 g) in acetone (6 ml) was added and the mixture was stirred for 40 minutes. After the addition of water, the reaction solution was extracted with ethyl acetate and the organic layer was dried over anhydrous magnesium sulfate. The solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give compound (98×) (1.68 g).



1H-NMR (CDCl3) δ: 0.67-0.73 (3H, m), 0.84-0.88 (1H, m), 1.73 (1H, t, J=5.6 Hz), 2.29 (3H, d, J=2.0 Hz), 3.44 (1H, dd, J=12.2, 5.1 Hz), 3.82 (1H, dd, J=12.2, 5.1 Hz), 7.14 (1H, dd, J=11.0, 9.0 Hz), 7.52 (2H, t, J=7.6 Hz), 7.63 (1H, t, J=7.6 Hz), 7.87 (2H, d, J=7.6 Hz), 8.17 (1H, ddd, J=9.0, 3.9, 2.9 Hz), 8.27 (1H, dd, J=6.8, 2.9 Hz), 8.82 (1H, s), 11.75 (1H, s).


The 5th step: Compound (98) (1.68 g) was dissolved in dichloromethane (17 ml), stirred under ice cooling and 1-chloro-N,N,2-trimethyl-1-propenylamine (0.60 g) was added. After stirring at room temperature for an hour, water was added, the reaction solution was extracted with ethyl acetate and the organic layer was dried over anhydrous magnesium sulfate. The solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give compound (99) (1.34 g).



1H-NMR (CDCl3) δ: 0.77-0.82 (1H, m), 0.95-1.07 (2H, m), 1.38-1.40 (1H, m), 1.52 (3H, d, J=1.1 Hz), 2.25 (1H, d. J=13.0 Hz), 3.05 (1H, d, J=13.0 Hz), 7.27 (1H, dd, J=10.8, 8.9 Hz), 7.40-7.54 (3H, m), 8.18-8.27 (3H, m), 8.36 (1H, dd, J=6.7, 2.7 Hz).


The 6th step: Hydrazine monohydrate (038 g) was added to a solution of compound (99×) (1.00 g) in ethanol (10 ml) under stirring at room temperature. After stirring for 4 hours, it was stirred under heating at 50° C. for 2 hours. The reaction solution was concentrated under reduced pressure, water was added, and the mixture was extracted with ethyl acetate and the organic layer was dried over anhydrous magnesium sulfate. The solvent was evaporated under reduced pressure to give compound (100) (0.69 g) as a crude product.


The 7th step: A mixture of compound (100) (0.91 g), di-tert-butyldicarbonate (1.55 g), 4-dimethylaminopyridine (0.04 g) and tetrahydrofuran (9.1 ml) was stirred at room temperature for 1 hour and 15 minutes. Water was added, the reaction solution was extracted with ethyl acetate and the organic layer was dried over anhydrous magnesium sulfate. The solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give compound (101) (1.28 g).



1H-NMR (CDCl3) δ: 0.37-0.41 (1H, m), 0.50-0.54 (1H, m), 0.68 (2H, t, J=7.7 Hz), 1.56 (18H, s), 1.78 (3H, d, J=4.0 Hz), 2.35 (1H, d, J=12.7 Hz), 3.57 (1H, dd, J=12.7, 1.8 Hz), 7.12-7.21 (1H, m), 8.15 (1H, ddd, J=8.9, 3.9, 3.0 Hz), 8.39 (1H, dd, J=6.7, 3.0 Hz).


The 8th step: Compound (101) (1.28 g) was dissolved in ethyl acetate (13 ml), 10% Pd—C (0.64 g) was added and the mixture was stirred at room temperature for 13 hours and 30 minutes. The insoluble materials were filtered, the filtrate was concentrated under reduced pressure and the residue was purified with a silicagel column chromatography to give compound (102) (1.07 g).



1H-NMR (CDCl3) δ: 0.51-0.58 (2H, m), 0.81-0.86 (2H, m), 1.54 (18H, s), 1.64 (3H, d, J=3.0 Hz), 2.60 (1H, d, J=12.4 Hz), 3.08 (1H, d, J=12.4 Hz), 3.50 (2H, s), 6.51 (1H, ddd, J=8.6, 3.7, 3.0 Hz), 6.78-6.84 (2H, m), 7.18-7.21 (1H, m).


The 9th step: A solution of 5-methylpyrazine-2-carboxylic acid (59 mg) in N,N-dimethylformamide (1.5 ml) was stirred under ice cooling, and 1-[bis(dimethylamino)methylene]-1H-1,2,3-triazolo(4,5-b)pyridinium-3-oxide hexafluorophosphate (196 mg) and triethylamine (61 mg) were added and the mixture was stirred for 10 minutes. A solution of compound (102) (200 mg) in N,N-dimethylformamide (3 ml) was added and the mixture was stirred at room temperature for 4 hours. Water was added, extracted with ethyl acetate and the organic layer was dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give compound (103) (170 mg).


The 10th step: Compound (103) (170 mg) was dissolved in dichloromethane (0.75 ml), stirred under ice cooling, trifluoroacetic acid (0.75 ml) was added and the mixture was stirred at room temperature for 3 hours. After concentration of the reaction solution under reduced pressure, ice water was added, potassium carbonate was added with stirring under ice cooling and extracted with ethyl acetate. The organic layer was dried over anhydrous sodium sulfate, the solvent was evaporated under reduced pressure and ether/hexane was added to the residue. The precipitated solid was filtered to give compound (674) (104 mg)



1H-NMR (CDCl3) δ: 0.53-0.59 (1H, m), 0.65-0.72 (1H, m), 0.85-0.91 (1H, m), 1.14-1.17 (1H, m), 1.47 (3H, d, J=2.0 Hz), 2.46 (1H, d, J=12.1 Hz), 2.69 (3H, s), 2.89 (1H, dd, J 12.1, 1.3 Hz), 7.06 (1H, dd, J=11.5, 8.8 Hz), 7.45 (1H, dd, J=6.8, 2.8 Hz), 7.94 (1H, ddd, J=8.8, 4.0, 2.8 Hz), 8.44 (1H, d, J=1.3 Hz), 9.36 (1H, d, J=1.3 Hz), 9.60 (1H, s).


Example 6
Preparation of Compound 687



embedded image


The 1st step: Dichloromethane-trifluoroacetic acid (1:1, 1 ml) was added to compound (120) (49 mg) and stirred at room temperature for an hour. The reaction solution was concentrated under reduced pressure, dimethyl sulfoxide-acetic anhydride (1:1, 1 ml) was added to the residue, stirred at 50° C. for 1.5 hours and the solvent was evaporated under reduced pressure. Hydrochloric acid (1M, 0.5 ml) was added to the residue and stirred at 50° C. for 1 hours. A saturated aqueous solution of sodium bicarbonate was added, extracted with chloroform, and the organic layer was dried over anhydrous sodium sulfate.


The solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography. A mixture of chloroform-diethyl ether/ethyl acetate was added and the precipitated slid was filtered to give compound 687 (17 mg)



1H-NMR (CDCl3) δ: 1.78 (3H, s), 3.52 (1H, d, J=15.1 Hz), 3.73 (1H, d, 15.1 Hz), 7.06 (1H, dd, J=10.4, 8.6 Hz), 7.73 (1H, dd, J=6.6, 1.3 Hz), 7.82-7.86 (1H, m), 7.90 (1H, d, J=1.3 Hz), 8.48 (1H, d, J=1.3 Hz), 9.79 (1H, s).


Example 7
Preparation of Compound 680,681 and 682



embedded image


embedded image


The 1st step: In a nitrogen atmosphere, diisopropylamine (20.3 ml) and tetrahydrofuran (83.5 ml) were added and cooled to −60° C. in an acetone/dry ice bath and a solution of 1.63M n-butyl lithium/n-hexane was added dropwise while stirring was continued until the temperature rose to 0° C. After stirring for 30 minutes, the reaction solution was cooled to −60° C. in an acetone/dry ice bath and a solution of tert-butyl acetate (16.8 ml) in tetrahydrofuran (22.2 ml) was added dropwise with stirring. After stirring for 45 minutes, a solution of compound (121) (11.1 g) in tetrahydrofuran (22.2 ml) was added dropwise. After 2.5 hours, a saturated aqueous solution of ammonium chloride (100 ml) was stirred under ice cooling and the reaction solution was poured portionwise therein, extracted with ethyl acetate, and the organic layer was dried over anhydrous magnesium sulfate. The solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give compound (122) (8.83 g).



1H-NMR (CDCl3) δ: 1.32 (17H, s), 1.93 (3H, s), 3.15 (1H, d, J=16.4 Hz), 3.66 (1H, d, J=16.2 Hz), 5.50 (1H, s), 8.12 (1H, s), 8.36 (1H, s).


The 2nd step: Lithium aluminium hydride (0.76 g) and tetrahydrofuran (5 ml) were cooled in an ice-salt bath in a nitrogen atmosphere and a solution of compound (122) (3.03 g) in tetrahydrofuran (10 ml) was added dropwise with stirring. After stirring for 15 minutes, acetone (4 ml) was added, insoluble materials were filtered, extracted with ethyl acetate, and the organic layer was washed with water and dried over anhydrous magnesium sulfate. The solvent was evaporated under reduced pressure to give compound (123) (2.30 g) as a crude product.


MS: 385 m/z [M+H]+


The 3rd step: 10% Hydrochloric acid/methanol solution (30 ml) was added to compound (123) (2.2 g) and stirred at room temperature for 1.5 hours. The solvent was evaporated under reduced pressure, the residue was basified with a 2.0M aqueous solution of potassium carbonate and extracted with ethyl acetate (100 ml). The organic layer was washed with water, dried over anhydrous magnesium sulfate and the solvent was evaporated under reduced pressure. Toluene (30 ml) and water (15 ml) were added to the resulted crude product (2.25 g), cooled in a ice bath and potassium carbonate (1.58 g) and thiophosgene (0.656 ml) were added with stirring. After stirring at room temperature for 30 minutes, the reaction solution was extracted with toluene, the organic layer was washed with water and dried over anhydrous magnesium sulfate. The solvent was evaporated under reduced pressure, toluene (30 ml), thionyl chloride (1.25 ml) and N,N-dimethylformamide (0.044 ml) were added to the resulted residue and the mixture was stirred under heating at 80° C. for 1.5 hours. The solvent was evaporated under reduced pressure, ice water was added, extracted with ethyl acetate, and the organic layer was washed with water and dried over anhydrous magnesium sulfate. The solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give compound (124) (1.26 g).



1H-NMR (CDCl3) δ: 1.56 (3H, s), 2.54-2.64 (1H, m), 3.07-3.17 (1H, m), 3.29-3.38 (1H, m), 3.50-3.57 (11H, m), 8.13 (1H, d, J=2.4 Hz), 8.44 (1H, d, J=2.4 Hz).


The 4th step: Tetrahydrofuran (12.6 ml) and 28% ammonia water (6.3 ml) were added to compound (124) (1.26 g) and stirred at room temperature for 1.5 hours. The reaction solution was extracted with ethyl acetate, the organic layer was washed with saturated brine and dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure to give compound (125) (1.13 g) as a crude product.



1H-NMR (CDCl3) δ: 1.70 (3H, s), 2.15-2.21 (1H, m), 2.52-2.58 (1H, m), 2.70-2.77 (1H, m), 3.05-3.11 (1H, m), 4.44 (2H, br s), 8.12 (1H, s), 8.34 (1H, s).


The 5th step: Tetrahydrofuran (11.3 ml) and di-tert-butyldicarbonate (0.89 ml) were added to compound (125) (1.13 g) and stirred at room temperature for an hour.


Di-tert-butyldicarbonate (1.13 ml) and 4-dimethylaminopyridine (0.086 g) were added and further stirred at room temperature for 2 hours. The solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give compound (126) (1.59 g).



1H-NMR (CDCl3) δ: 1.53 (18H, s), 1.73 (3H, s), 1.90-1.97 (1H, m), 2.63-2.69 (1H, m), 2.93-2.99 (1H, m), 3.21-3.28 (1H, m), 8.24 (1H, d, J=2.3 Hz), 8.36 (1H, d, J=2.3 Hz).


The 6th step: N,N-Dimethylformmamide (40 ml) was added to compound (126) (2.00 g) in a nitrogen stream, cooled in an ice bath with stirring and sodium methoxide (2.074 g) was added therein. After stirring at room temperature for 1.5 hours, the reaction solution was warmed up to 60° C. and stirred for 2 hours. It was cooled in a ice bath, neutralized by the addition of 2N hydrochloric acid and extracted with ethyl acetate. The organic layer was washed with 2M aqueous solution of potassium carbonate and brine and dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give compound (127) (1.69 g).



1H-NMR (CDCl3) δ: 1.52 (9H, s), 1.70 (3H, s), 1.96-2.03 (1H, m), 2.54-2.61 (1H, m), 2.80-2.85 (1H, m), 2.97-3.00 (1H, m), 3.97 (3H, s), 7.62 (1H, d, J=1.5 Hz), 8.15 (1H, d, J=1.5 Hz).


The 7th step: Compound (127) (1.571 g), trisdibenzylideneacetonedipalladium (0.414 g) and butynyl-1-adamantylphosphine (0.324 g) were dissolved in toluene under a nitrogen stream, and a solution of 1.6M lithium hexamethyldisilazide/tetrahydrofuran (5.66 ml) was added at room temperature with stirring. The reaction solution was warmed up to 80° C. and stirred for 3 hours. Then diethyl ether and 1 N hydrochloric acid were added with stirring under ice cooling. After stirring for 5 minutes, it was neutralized by the addition of a saturated aqueous solution of sodium carbonate, extracted with ethyl acetate and dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give compound (128) (1.55 g).



1H-NMR (CDCl3) δ: 1.52 (9H, s), 1.72 (3H, s), 1.86-1.93 (1H, m), 2.02 (2H, s), 2.52-2.59 (1H, m), 2.74-2.79 (1H, m), 3.13-3.18 (1H, m), 3.90 (3H, s), 6.96 (1H, d, J=2.3 Hz), 7.59 (1H, d, J=1.8 Hz).


The 8th step: Compound (128) (0.20 g), 5-methylpyridine-2-carboxylic acid (0.10 g) and O-(7-azabenzotriazole-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (HATU)(0.28 g) were dissolved in N,N-dimethylformamide (2 ml), triethylamine (0.119 ml) was added and the mixture was stirred at room temperature for 1.0 hours. A 2M aqueous solution of potassium carbonate was added, extracted with ethyl acetate, and the organic layer was washed with brine and dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure and the resulted residue was dissolved in chloroform (4.0 ml), trifluoroacetic acid (1.0 ml) was added and the mixture was stirred at room temperature for 3 hours. The solvent was evaporated under reduced pressure, the residue was made basic by the addition of a 2.0M aqueous solution of potassium carbonate, extracted with ethyl acetate and the organic layer was washed with water and dried over anhydrous magnesium sulfate. The solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give compound (680) (0.096 g).



1H-NMR (DMSO-d6) δ: 1.47 (3H, s), 1.77-1.83 (1H, m), 2.34-2.39 (1H, m), 2.48-2.53 (1H, m), 2.63 (3H, s), 2.89-2.96 (1H, m), 3.90 (3H, s), 5.86 (2H, br s), 8.10 (1H, d, J=2.3 Hz), 8.47 (1H, d, J=2.5 Hz), 8.69 (1H, s), 9.14 (1H, s), 10.69 (1H, s).


The 9th step: Compound (680) (0.096 g) and sodium iodide (0.193 g) were dissolved in acetonitrile (5.0 ml), trimethylsilylchloride (0.164 ml) was added and the mixture was stirred at room temperature for 2.5 hours. Sodium iodide (0.193 g) and trimethyl silylchloride (0.164 ml) were added and stirring was continued at room temperature for 12 hours. A 2.0M aqueous solution of potassium carbonate, extracted with ethyl acetate and the organic layer was washed with water and dried over anhydrous magnesium sulfate. The solvent was evaporated under reduced pressure to give compound (681) (0.073 g) as a crude product.



1H-NMR (DMSO-d6) δ: 1.52 (3H, s), 1.80-1.85 (1H, m), 2.62 (3H, s), 2.64-2.69 (2H, m), 2.96-3.01 (1H, m), 7.77 (1H, d, J=2.5 Hz). 7.96 (1H, d, J=2.3 Hz), 8.67 (1H, s), 9.10 (1H, s), 10.58 (1H, s).


The 10th step: Compound (681) (0.031 g) was dissolved in tetrahydrofuran (2.0 ml), di-tert-butyldicarbonate (0.030 ml) was added and the mixture was stirred at room temperature for 1.5 hours. Di-tert-butyldicarbonate (0.030 ml) was further added and the stirring was continued at room temperature for 2.0 hours. The reaction solution was concentrated under reduced pressure, the resulted residue was dissolved in N, N-dimethylformamide (0.5 ml) and potassium carbonate (23.9 mg) was added. A solution of methyl iodide (12.2 mg) in N,N-dimethylformamide (0.5 ml) was added with stirring at room temperature. After stirring at room temperature for 3 hours, methyl iodide (11.05 mg) was added and the mixture was stirred at room temperature for 2 hours. Brine was added, extracted with ethyl acetate and the organic layer was washed with water and dried over anhydrous magnesium sulfate. The solvent was evaporated under reduced pressure, the resulted residue was dissolved in chloroform (2.0 ml), trifluoroacetic acid (0.5 ml) was added and the mixture was stirred at room temperature for 3 hours. The reaction solution was concentrated under reduced pressure, the resulted residue was made alkaline by the addition of a 2.0M aqueous solution of potassium carbonate, extracted with ethyl acetate and the organic layer was washed with water and dried over anhydrous magnesium sulfate. The solvent was evaporated under reduced pressure and the residue was purified with a silicagel column chromatography to give compound (682) (4.2 mg).



1H-NMR (DMSO-d6) δ: 1.46 (3H, s), 1.95-2.01 (1H, m), 2.33-2.39 (1H, m), 2.62 (3H, s), 2.64-2.69 (1H, m), 2.74 (3H, s), 2.92-2.98 (1H, m), 7.90 (1H, d, J=2.5 Hz), 7.94-7.95 (1H, m), 8.67 (1H, s), 9.09 (1H, s), 10.57 (1H, s).


The other compounds are prepared in the same manner. Chemical structures and physical constants are shown below.












TABLE 1







Com-




pound




No.
Structure









1


embedded image









2


embedded image









3


embedded image









4


embedded image









5


embedded image






















TABLE 2







Com-




pound




No.
Structure









 6


embedded image









 7


embedded image









 8


embedded image









 9


embedded image









10


embedded image




















TABLE 3





Com-



pound



No.
Structure







11


embedded image







12


embedded image







13


embedded image







14


embedded image







15


embedded image



















TABLE 4





Compound



No.
Structure







16


embedded image







17


embedded image







18


embedded image







19


embedded image







20


embedded image





















TABLE 5







Com-




pound




No.
Structure









21


embedded image









22


embedded image









23


embedded image









24


embedded image









26


embedded image




















TABLE 6





Com-



pound



No.
Structure







27


embedded image







28


embedded image







29


embedded image







30


embedded image







31


embedded image



















TABLE 7





Compound



No.
Structure







32


embedded image







33


embedded image







34


embedded image







35


embedded image







36


embedded image



















TABLE 8





Compound



No.
Structure







37


embedded image







38


embedded image







39


embedded image







40


embedded image







41


embedded image





















TABLE 9







Com-




pound




No.
Structure









42


embedded image









43


embedded image









44


embedded image









45


embedded image









46


embedded image




















TABLE 10





Compound



No.
Structure







47


embedded image







48


embedded image







49


embedded image







50


embedded image







51


embedded image



















TABLE 11





Com-



pound



No.
Structure







52


embedded image







53


embedded image







54


embedded image







55


embedded image







56


embedded image



















TABLE 12





Com-



pound



No.
Structure







57


embedded image







58


embedded image







59


embedded image







60


embedded image







61


embedded image



















TABLE 13





Com-



pound



No.
Structure







62


embedded image







63


embedded image







64


embedded image







65


embedded image







66


embedded image



















TABLE 14





Compound



No.
Structure







67


embedded image







68


embedded image







69


embedded image







70


embedded image







71


embedded image



















TABLE 15





Compound



No.
Structure







72


embedded image







73


embedded image







74


embedded image







75


embedded image







76


embedded image



















TABLE 16





Compound



No.
Structure







77


embedded image







78


embedded image







79


embedded image







80


embedded image







81


embedded image



















TABLE 17





Compound



No.
Structure







82


embedded image







83


embedded image







84


embedded image







85


embedded image







86


embedded image



















TABLE 18





Compound



No.
Structure







87


embedded image







88


embedded image







89


embedded image







90


embedded image







91


embedded image



















TABLE 19





Compound



No.
Structure







92


embedded image







93


embedded image







94


embedded image







95


embedded image







96


embedded image



















TABLE 20





Compound



No.
Structure







 97


embedded image







 98


embedded image







 99


embedded image







100


embedded image







101


embedded image



















TABLE 21





Compound



No.
Structure







102


embedded image







103


embedded image







104


embedded image







105


embedded image







106


embedded image



















TABLE 22





Compound



No.
Structure







107


embedded image







108


embedded image







109


embedded image







110


embedded image







111


embedded image



















TABLE 23





Compound



No.
Structure







112


embedded image







113


embedded image







114


embedded image







115


embedded image







116


embedded image



















TABLE 24





Compound



No.
Structure







117


embedded image







118


embedded image







119


embedded image







120


embedded image







121


embedded image



















TABLE 25





Compound



No.
Structure







122


embedded image







123


embedded image







124


embedded image







125


embedded image







126


embedded image



















TABLE 26





Compound No.
Structure







127


embedded image







128


embedded image







129


embedded image







130


embedded image







131


embedded image



















TABLE 27





Compound No.
Structure







132


embedded image







133


embedded image







134


embedded image







135


embedded image







136


embedded image



















TABLE 28





Compound



No.
Structure







137


embedded image







138


embedded image







139


embedded image







140


embedded image



















TABLE 29





Compound



No.
Structure







141


embedded image







142


embedded image







143


embedded image







144


embedded image







145


embedded image



















TABLE 30





Compound No.
Structure







146


embedded image







147


embedded image







148


embedded image







149


embedded image







150


embedded image



















TABLE 31





Compound



No.
Structure







151


embedded image







152


embedded image







153


embedded image







154


embedded image







155


embedded image



















TABLE 32





Compound



No.
Structure







156


embedded image







157


embedded image







158


embedded image







159


embedded image







160


embedded image



















TABLE 33





Compound



No.
Structure







161


embedded image







162


embedded image







163


embedded image







164


embedded image







165


embedded image



















TABLE 34





Compound No.
Structure







166


embedded image







167


embedded image







168


embedded image







169


embedded image







170


embedded image







171


embedded image






















TABLE 35





Compound
MS





No.
(M + 1)
MP
NMR (solvent, shift value: ascending order)
uv



















1


1H-NMR (CDCl3) δ: 8.69 (1.0H, br s), 8.57 (1.0H, s),






8.53-8.41 (1.0H, m), 8.36-8.33 (1.0H, m), 7.90-7.81 (1.0H, m),






7.56 (1.0H, br s), 7.41-7.29 (2.0H, m), 7.00-6.88 (2.0H, m),






2.88-2.63 (2.0H, m), 2.50-2.38 (1.0H, m), 2.06-2.00 (1.0H, m), 1.66 (3.0H,



2
344





3


1H-NMR (CDCl3) δ: 7.79 (1.0H, br s), 7.65-7.64 (1.0H, m),






7.48-7.41 (1.0H, m), 7.31 (1.0H, t, J = 8.01 Hz),






7.04-7.01 (1.0H, m), 6.23 (1.0H, br s), 2.93-2.65 (2.0H, m), 2.57 (3.0H, br






s), 2.40 (1.0H, ddd, J = 14.11, 5.34, 3.43 Hz), 2.27 (3.0H, br s),






2.09-1.92 (1.0H, m), 1.67 (3.0H, s).



4


1H-NMR (CDCl3) δ: 7.86-7.83 (1.0H, m), 7.45-7.42 (1.0H, m),






7.35 (1.0H, t, J = 12.96 Hz), 7.21 (1.0H, br s), 7.04-7.01 (1.0H,






m), 4.23 (3.0H, s), 2.90-2.86 (1.0H, m), 2.77-2.61 (1.0H, m),






2.38-2.30 (1.0H, m), 1.99-1.89 (1.0H, m), 1.60 (3.0H, s).



5


1H-NMR (DMSO-d6) δ: 9.81 (1.0H, br s), 7.70-7.65 (2.0H, m),






7.22 (1.0H, t, J = 7.85 Hz), 7.06-7.03 (1.0H, m), 6.53 (1.0H, s),






2.92-2.85 (1.0H, m), 2.61-2.52 (1.0H, m), 2.28 (3.0H, s),






2.02-1.97 (1.0H, m), 1.73-1.67 (1.0H, m), 1.39 (3.0H, s).



6
425





7
415





8
361





9
331





10
347





11
360





12
379





13
367





14
331





15


1H-NMR (DMSO-d6) δ: 10.02 (1.0H, s), 7.61-7.55 (2.0H, m),






7.25 (1.0H, t, J = 7.93 Hz), 7.09 (1.0H, d, J = 7.78 Hz),






6.26 (1.0H, s), 3.86 (3.0H, s), 2.91-2.87 (1.0H, m), 2.59-2.54 (1.0H,






m), 2.00-1.96 (1.0H, m), 1.75-1.62 (1.0H, m), 1.39 (3.0H, s).



16
404





17
422





18
360





19
349





20
349





21
388





22
365





23
392





24
385





26


1H-NMR (MeOD) δ: 1.73 (1H, s), 1.98 (1H, s), 2.29 (1H, s),






2.76 (1H, s), 6.58 (1H, s), 6.79 (1H, s), 6.92 (1H, s), 7.13 (1H, s),






8.01 (1H, s), 8.55 (1H, s)



27


1H-NMR (DMSO-d6) δ: 1.65 (1H, s), 2.07 (1H, t, J = 13.1 Hz),






2.57 (1H, d, J = 11.6 Hz), 3.10 (1H, s), 7.10 (1H, d, J = 7.1 Hz),






7.42 (1H, t, J = 7.5 Hz), 7.72 (2H, s), 7.84 (1H, d, J = 8.3 Hz),






7.92 (1H, t, J = 9.5 Hz), 8.55 (1H, s), 10.74 (1H, s)



28
358





29


1H-NMR (CDCl3) δ: 1.71 (s3H, s), 2.00 (1H, d, J = 8.8 Hz),






2.45 (1H, d, J = 12.4 Hz), 2.78 (1H, t, J = 12.5 Hz), 2.88 (aH, s, J = 13.6 Hz),






3.94 (3H, s), 5.30 (1H, s) 7.05 (1H, d, J = 7.8 Hz),






7.35 (1H, t, J = 8.2 Hz), 7.48 (1H, s), 7.56 (1H, s), 7.65 (1H, d, J = 7.8 Hz),






9.58 (1H, s)




















TABLE 36





Compound
MS





No.
(M + 1)
MP
NMR (solvent, shift value: ascending order)
uv



















30
376





31


1H-NMR (MeOD) δ: 1.75 (3H, s), 2.10-2.13 (1H, m),






2.49-2.62 (1H, m), 2.65-2.71 (2H, m), 2.80 (3H, s), 7.02 (1H, d, J = 8.6 Hz),






7.41 (1H, t, J = 7.8 Hz), 7.53 (1H, s), 8.12 (1H, d, J = 8.1 Hz),






9.34 (2H, s), 9.79 (1H, s)



32
372





33
373





34


1H-NMR (DMSO-d6) δ: 1.63 (3H, s), 1.70 (3H, s, J = 5.3 Hz),






1.99-2.02 (1H, br m), 2.28 (1H, s), 2.57-2.60 (1H, dr m), 3.07 (1H,






d, J = 10.1 Hz), 4.65 (2H, s), 5.70 (1H, d, 14.1 Hz), 5.91 (1H, d,






7.3 Hz), 7.06 (1H, s), 7.36 (1H, s), 7.59 (1H, d, J = 5.1 Hz),






7.82 (1H, s), 7.89 (1H, s, J 5.1 Hz), 8.36 (1H, s), 10.48 (1H, s)



35
395





36
428





37
412





38
396





39
238





40
408





41
428





42
379





43
390





44
386





45
420





46
435





47


1H-NMR (DMSO-d6) δ: 1.41 (3H, s), 1.69-1.75 (1H, m),






2.01-2.05 (1H, m), 2.55-2.58 (1H, m), 2.74-2.76 (2H, m),






2.88-2.91 (1H, m), 7.06 (1H, d, J = 7.8 Hz), 7.26 (1H, t, J = 7.8 Hz),






7.70-7.73 (2H, m), 7.91 (1H, br s), 7.99 (1H, s), 8.63 (1H, s),






9.97 (1H, s).



48


1H-NMR (DMSO-d6) δ: 1.42 (3H, s), 1.71-1.85 (3H, m),






2.04-2.08 (1H, m), 2.56-2.58 (1H, m), 2.82 (2H, t, J = 7.2 Hz),






2.88-2.93 (1H, m), 3.41-3.43 (2H, m), 7.06 (1H, d, J = 7.8 Hz),






7.26 (1H, t, J = 7.6 Hz), 7.70-7.73 (2H, m), 7.99 (2H, s), 8.65 (1H, s),






10.00 (1H, s).



49


1H-NMR (DMSO-d6) δ: 1.57 (3H, s), 1.62 (3H, s),






1.94-1.97 (1H, m), 2.37-2.40 (1H, m), 2.56-2.60 (1H, m), 2.80-2.82 (2H,






m), 3.03-3.06 (1H, m), 7.04 (1H, d, J = 7.6 Hz), 7.34 (1H, t, J = 7.6 Hz),






7.78-7.82 (2H, m), 8.01 (1H, s), 8.06 (1H, br s),






8.64 (1H, s), 10.13 (1H, s).



50


1H-NMR (DMSO-d6) δ: 1.45 (3H, s), 1.76-1.79 (1H, m),






2.09-2.13 (1H, m), 2.40 (3H, br s), 2.57-2.60 (1H, m), 2.83 (2H, t, J = 5.9 Hz),






2.93-2.94 (1H, m), 3.49-3.51 (2H, m), 5.74 (1H, s),






7.06 (1H, d, J = 7.6 Hz), 7.27 (1H, t, J = 7.8 Hz), 7.74 (1H, br s),






7.77 (1H, s), 7.95 (2H, br s), 8.02 (1H, s), 8.66 (1H, s), 10.02 (1H, s).



51


1H-NMR (DMSO-d6) δ: 1.40 (3H, s), 1.70-1.75 (1H, m),






1.85-1.90 (2H, m), 1.93-2.02 (2H, m), 2.14-2.21 (1H, m),






2.53-2.60 (1H, m), 2.86-2.94 (1H, m), 3.82 (1H, q, J = 7.1 Hz), 3.98 (1H, q,






J = 7.2 Hz), 4.35-4.39 (1H, m), 7.06 (1H, d, J = 7.6 Hz),






7.23 (1H, t, J = 7.8 Hz), 7.56-7.60 (2H, m), 9.57 (1H, s).




















TABLE 37





Compound
MS





No.
(M + 1)
MP
NMR (solvent, shift value: ascending order)
uv







52


1H-NMR (DMSO-d6) δ: 1.38 (3H, s), 1.61-1.81 (5H, m),






1.95-2.08 (2H, m), 2.53-2.58 (1H, m), 2.88 (4H, t, J = 6.6 Hz),






3.65-3.68 (1H, m), 5.67-5.85 (2H, m), 7.04 (1H, d, J = 7.8 Hz),






7.22 (1H, t, J = 7.8 Hz), 7.48 (1H, s), 7.59 (1H, d, J = 7.8 Hz),






9.87 (1H, s).



53


1H-NMR (DMSO-d6) δ: 1.39 (3H, s), 1.62-1.80 (5H, m),






1.99-2.07 (2H, m), 2.52-2.58 (1H, m), 2.87-2.91 (3H, m), 3.68 (2H,






dd, J = 8.7, 5.9 Hz), 7.04 (1H, d, J = 7.6 Hz), 7.23 (1H, t, J = 8.0 Hz),






7.47 (1H, s), 7.61 (1H, d, J = 7.8 Hz), 9.90 (1H, s).



54


1H-NMR (DMSO-d6) δ: 1.40 (3H, s), 1.70-1.75 (1H, m),






1.95-2.05 (2H, m), 2.09-2.37 (2H, m), 2.52-2.57 (1H, m),






2.87-2.94 (1H, m), 4.03 (1H, q, J = 7.1 Hz), 4.19 (1H, dd, J = 8.6, 4.3 Hz),






7.06 (1H, d, J = 7.3 Hz), 7.25 (1H, t, J = 8.0 Hz), 7.49 (1H, s),






7.56 (1H, d, J = 7.8 Hz), 7.86 (1H, s), 10.01 (1H, s).



55


1H-NMR (DMSO-d6) δ: 1.40 (3H, s), 1.69-1.75 (1H, m),






1.97-2.03 (2H, m), 2.09-2.36 (4H, m), 2.52-2.57 (1H, m),






2.87-2.93 (1H, m), 4.17-4.20 (1H, m), 7.06 (1H, d, J = 7.8 Hz), 7.25 (1H, t,






J = 8.0 Hz), 7.54 (2H, t, J = 8.0 Hz), 7.87 (1H, s), 10.00 (1H, s).



56


1H-NMR (DMSO-d6) δ: 1.40 (3H, s), 1.71-1.76 (1H, m),






2.00-2.03 (1H, m), 2.19-2.26 (1H, m), 2.46-2.58 (5H, m),






2.88-2.94 (1H, m), 5.03-5.06 (1H, m), 7.09 (1H, d, J = 7.1 Hz), 7.27 (1H, t,






J = 8.1 Hz), 7.50 (1H, s), 7.56 (1H, d, J = 8.1 Hz), 10.22 (1H, s).



57


1H-NMR (DMSO-d6) δ: 1.40 (3H, s), 1.69-1.75 (1H, m),






2.00-2.04 (1H, m), 2.19-2.27 (1H, m), 2.46-2.57 (6H, m),






2.87-2.94 (1H, m), 5.03-5.06 (1H, m), 7.09 (1H, d, J = 8.3 Hz), 7.26 (1H, t,






J = 8.0 Hz), 7.51 (1H, s), 7.56 (1H, d, J = 8.3 Hz), 10.22 (1H, s).



58


1H-NMR (DMSO-d6) δ: 1.48 (3H, s), 1.78-1.84 (1H, m),






2.11-2.18 (1H, m), 2.55-2.61 (1H, m), 2.93-2.99 (1H, m), 7.14 (1H, d,






J = 7.8 Hz), 7.34 (1H, t, J = 8.0 Hz), 7.77-7.82 (2H, m),






8.93 (1H, s), 9.11 (1H, s).



59


1H-NMR (DMSO-d6) δ: 1.43 (3H, s), 1.73-1.78 (1H, m),






2.01-2.08 (1H, m), 2.54-2.59 (1H, m), 2.89-2.96 (1H, m), 7.16 (1H, d,






J = 8.1 Hz), 7.34 (1H, t, J = 8.0 Hz), 7.57 (1H, s), 7.68 (1H, d, J = 7.1 Hz),






8.70 (1H, d, J = 2.5 Hz), 8.77 (1H, d, J = 2.3 Hz),






10.80 (1H, s).



60


1H-NMR (DMSO-d6) δ: 1.42 (3H, s), 1.70-1.76 (1H, m)






2.01-2.06 (1H, m), 2.54-2.60 (1H, m), 2.88-2.95 (1H, m), 3.97 (3H, s),






7.12 (1H, d, J = 7.8 Hz), 7.30 (1H, t, J = 7.8 Hz), 7.62-7.68 (2H,






m), 8.29 (1H, d, J = 2.3 Hz), 8.40 (1H, d, J = 2.5 Hz), 10.53 (1H,






s).



61


1H-NMR (DMSO-d6) δ: 0.91 (3H, t, J = 7.2 Hz), 1.32-1.40 (5H,






m), 1.51-1.58 (2H, m), 1.68-1.73 (1H, m), 1.97-2.05 (0H, m),






2.55-2.60 (1H, m), 2.85-2.92 (1H, m), 5.77 (2H, br s), 7.07 (1H,






d, J = 7.6 Hz), 7.25 (1H, t, J = 8.0 Hz), 7.73 (2H, t, J = 6.7 Hz),






7.83-7.87 (1H, m), 7.96 (1H, s), 8.64 (1H, s), 9.95 (1H, s).



62


1H-NMR (DMSO-d6) δ: 1.43 (3H, s), 1.71-1.77 (1H, m),






2.02-2.09 (1H, m), 2.55-2.61 (1H, m), 2.87-2.95 (1H, m), 3.31 (3H, s),






3.70-3.74 (2H, m), 4.51-4.54 (2H, m), 7.11 (1H, d, J = 7.1 Hz),






7.29 (1H, t, J = 7.7 Hz), 7.75 (1H, d, J = 8.3 Hz), 7.80 (1H, s),






8.43 (1H, s), 8.87 (1H, s), 10.35 (1H, s).




















TABLE 38





Compound
MS





No.
(M + 1)
MP
NMR (solvent, shift value: ascending order)
uv







63


1H-NMR (DMSO-d6) δ: 1.29 (3H, t, J = 7.5 Hz), 1.43 (3H, s),






1.71-1.77 (1H, m), 2.01-2.09 (1H, m), 2.55-2.61 (1H, m),






2.90-2.96 (3H, m), 7.12 (1H, d, J = 7.8 Hz), 7.30 (1H, t, J = 7.8 Hz),






7.76 (1H, d, J = 8.1 Hz), 7.82 (1H, s), 8.70 (1H, s), 9.18 (1H, s),






10.53 (1H, s).



64


1H-NMR (DMSO-d6) δ: 1.41 (3H, s), 1.70-1.75 (1H, m),






1.99-2.06 (1H, m), 2.52-2.57 (1H, m), 2.87-2.94 (1H, m), 7.11 (1H, d,






J = 7.6 Hz), 7.28 (1H, t, J = 7.8 Hz), 7.47-7.59 (5H, m),






7.65 (2H, d, J = 7.6 Hz), 8.30 (0H, s).



65


1H-NMR (DMSO-d6) δ: 1.38 (3H, s), 1.67-1.72 (1H, m),






1.94-2.00 (1H, m), 2.03 (3H, s), 2.50-2.55 (1H, m), 2.85-2.92 (1H, m),






7.06 (1H, d, J = 7.1 Hz), 7.23 (1H, t, J = 7.7 Hz), 7.48 (2H, t, J = 8.3 Hz),






10.55 (1H, s).



66
707.0 (2M + 1)





67
743.1 (2M + 1)





68


1H-NMR (DMSO-d6) δ: 1.45 (3H, s), 1.74-1.80 (1H, m),






2.06-2.13 (1H, m), 2.56-2.61 (1H, m), 2.90-2.97 (1H, m), 7.15 (1H, d,






J = 7.6 Hz), 7.33 (1H, t, J = 7.7 Hz), 7.79 (1H, d, J = 7.8 Hz),






7.87 (1H, s), 8.04 (1H, s), 8.45 (1H, s), 9.29 (2H, d, J = 8.3 Hz),






10.79 (1H, s).



69


1H-NMR (DMSO-d6) δ: 1.43 (3H, s), 1.71-1.76 (1H, m),






2.02-2.09 (1H, m), 2.55-2.63 (4H, m), 2.88-2.94 (1H, m), 7.12 (1H, d,






J = 8.1 Hz), 7.29 (1H, t, J = 7.8 Hz), 7.74 (1H, d, J = 8.1 Hz),






7.81 (1H, s), 8.70 (1H, s), 9.08 (1H, s), 10.45 (1H, s).



70


1H-NMR (DMSO-d6) δ: 1.43 (3H, s), 1.71-1.77 (1H, m),






2.02-2.09 (1H, m), 2.55-2.61 (1H, m), 2.87-2.95 (1H, m), 3.31 (3H, s),






3.70-3.74 (2H, m), 4.51-4.54 (2H, m), 7.11 (1H, d, J = 7.1 Hz),






7.29 (1H, t, J = 7.7 Hz), 7.75 (1H, d, J = 8.3 Hz), 7.80 (1H, s),






8.43 (1H, s), 8.87 (1H, s), 10.35 (1H, s).



71


1H-NMR (DMSO-d6) δ: 0.94 (3H, t, J = 7.3 Hz), 1.43-1.47 (4H,






m), 1.72-1.79 (3H, m), 2.02-2.09 (1H, m), 2.58 (1H, t, J = 9.7 Hz),






2.91 (1H, s), 4.40 (2H, t, J = 6.6 Hz), 7.11 (1H, d, J = 7.8 Hz),






7.29 (1H, t, J = 8.1 Hz), 7.75 (1H, d, J = 7.3 Hz), 7.80 (1H,






s), 8.37 (1H, s), 8.86 (1H, s), 10.34 (1H, s).



72
771.1 (2M + 1)





73


1H-NMR (DMSO-d6) δ: 1.40 (3H, s) 1.68-1.73 (1H, m),






1.98-2.05 (1H, m), 2.11 (3H, s), 2.55-2.60 (1H, m), 2.69 (2H, t, J = 6.8 Hz),






2.86-2.92 (1H, m), 3.55-3.59 (2H, m), 5.77 (2H, br s),






7.07 (1H, d, J = 8.1 Hz), 7.25 (1H, t, J = 8.0 Hz), 7.71-7.75 (2H,






m), 8.00 (2H, s), 8.65 (1H, s), 9.98 (1H, s).



74


1H-NMR (DMSO-d6) δ: 0.99 (3H, t, J = 7.5 Hz), 1.39 (3H, s),






1.56 (2H, td, J = 14.3, 7.2 Hz), 1.67-1.73 (1H, m),






1.95-2.02 (1H, m), 2.38 (2H, t, J = 6.8 Hz), 2.50-2.55 (1H, m),






2.86-2.93 (1H, m), 7.06 (1H, d J = 7.8 Hz), 7.23 (1H, t, J = 8.3 Hz),






7.47-7.51 (2H, m).



75
839.1 (2M + 1)





76
835.1 (2M + 1)





77
782.9 (2M + 1)





78


1H-NMR (DMSO-d6) δ: 1.50 (3H, s), 1.79-1.84 (1H, m),






2.23-2.30 (1H, m), 2.55-2.60 (1H, m), 2.96-3.02 (1H, m), 7.16 (1H,






dd, J = 11.6, 8.8 Hz), 7.75-7.78 (2H, m), 8.89 (1H, s), 9.07 (1H,






s), 10.74 (1H, br s).




















TABLE 39





Compound
MS





No.
(M + 1)
MP
NMR (solvent, shift value: ascending order)
uv



















79


1H-NMR (DMSO-d6) δ: 1.49 (4H, s), 1.77-1.83 (1H, m),






2.16-2.23 (1H, m), 2.56-2.62 (1H, m), 2.95-3.01 (1H, m), 5.87 (2H, br






s), 7.17 (1H, dd, J = 11.7, 8.5 Hz), 7.76-7.82 (2H, m), 9.96 (2H,






d, J = 3.8 Hz), 10.82 (1H, s).



80


1H-NMR (DMSO-d6) δ: 1.33 (3H, t, J = 7.2 Hz), 1.42 (3H, s),






1.71-1.76 (1H, m), 2.03-2.07 (1H, m), 2.55-2.59 (1H, m),






2.89-2.92 (1H, m), 3.25 (2H, q, J = 7.3 Hz), 7.11 (1H, d, J = 7.8 Hz),






7.29 (1H, t, J = 7.8 Hz), 7.74 (1H, d, J = 8.1 Hz), 7.79 (1H, s),






8.66 (1H, s), 9.07 (1H, s), 10.45 (1H, s).



81
831.1 (2M + 1)





82
850.9 (2M + 1)





83
795.0 (2M + 1)





84
758.8 (2M + 1)





85
750.9 (2M + 1)





86
795.1 (2M + 1)





87


1H-NMR (DMSO-d6) δ: 1.01 (6H, d, J = 6.8 Hz), 1.44 (3H, s),






1.73-1.78 (1H, m), 2.05-2.13 (2H, m), 2.56-2.61 (1H, m),






2.89-2.95 (1H, m), 4.19 (2H, d, J = 6.6 Hz), 7.12 (1H, d, J = 8.1 Hz),






7.29 (1H, t, J = 7.8 Hz), 7.76 (1H, d, J = 7.8 Hz), 7.82 (1H, s),






8.41 (1H, d, J = 1.3 Hz), 8.87 (1H, s), 10.36 (1H, s).



88


1H-NMR (DMSO-d6) δ: 1.70 (3H, s), 2.02-2.08 (1H, m),






2.58-2.64 (2H, m), 3.15-3.19 (1H, m), 5.16 (2H, q, J = 8.8 Hz),






7.27 (1H, dd, J = 11.9, 8.8 Hz), 7.85-7.98 (2H, m), 8.62 (1H, s),






8.92 (1H, s), 10.83 (1H, s).



89


1H-NMR (DMSO-d6) δ: 1.65 (3H, s), 2.04-2.11 (1H, m),






2.54-2.62 (3H, m), 2.83-2.95 (2H, m), 3.11-3.14 (1H, m), 4.65 (2H, t,






J = 5.8 Hz), 7.09 (1H, d, J = 7.6 Hz), 7.42 (1H, t, J = 8.0 Hz),






7.87-7.92 (2H, m), 8.48 (1H, s), 8.91 (1H, s), 10.62 (1H, s).



90


1H-NMR (DMSO-d6) d: 1.41 (3H, s), 1.69-1.74 (1H, m),






2.00-2.04 (1H, m), 2.56-2.61 (1H, m), 2.87-2.92 (1H, m), 3.24 (6H, s),






3.37-3.52 (12H, m), 3.59 (2H, t, J = 4.5 Hz), 3.80 (2H, t, J = 4.3 Hz),






4.52 (2H, t, J = 4.4 Hz), 5.81 (2H, br s), 7.12 (1H, d, J = 7.6 Hz),






7.28 (1H, t, J = 8.0 Hz), 7.75 (1H, d, J = 8.3 Hz), 7.81 (1H,






s), 8.42 (1H, s), 8.87 (1H, s), 10.33 (1H, s).



91


1H-NMR (DMSO-d6) δ: 1.41 (3H, s), 1.69-1.75 (1H, m),






1.85 (3H, s), 1.98-2.05 (1H, m), 2.55-2.61 (1H, m), 2.86-2.93 (1H,






m), 5.09 (2H, d, J = 2.0 Hz), 5.79 (2H, br s), 7.12 (1H, d, J = 7.8 Hz),






7.28 (1H, t, J = 7.8 Hz), 7.74 (1H, d, J = 8.3 Hz), 7.80 (1H,






s), 8.45 (1H, s), 8.90 (1H, s), 10.36 (1H, s).



92


1H-NMR (DMSO-d6) δ: 1.41 (4H, s), 1.69-1.74 (1H, m),






1.98-2.05 (1H, m), 2.56-2.61 (1H, m), 2.87-2.93 (1H, m), 4.74 (2H, td,






J = 15.0, 3.1 Hz), 5.79 (2H, br s), 6.34-6.61 (1H, m), 7.12 (1H,






d, J = 7.8 Hz), 7.29 (1H, t, J = 8.0 Hz), 7.74 (1H, d, J = 8.1 Hz),






7.81 (1H, s), 8.54 (1H, s), 8.90 (1H, s), 10.40 (1H, s).



93
837.0 (2M + 1)





94


1H-NMR (DMSO-d6) δ: 1.41 (4H, s), 1.69-1.74 (1H, m),






1.98-2.05 (1H, m), 2.55-2.61 (2H, m), 2.71-2.75 (1H, m),






2.87-2.93 (1H, m), 4.49-4.61 (4H, m), 5.05-5.11 (1H, m), 5.79 (2H, br s),






7.12 (1H, d, J = 7.3 Hz), 7.26 (1H, t, J = 8.0 Hz), 7.74 (1H, d, J = 8.6 Hz),






7.81 (1H, s), 8.47 (1H, s), 8.88 (1H, s), 10.36 (1H, s).



95
801.0 (2M + 1)




















TABLE 40





Compound
MS





No.
(M + 1)
MP
NMR (solvent, shift value: ascending order)
uv



















96


1H-NMR (DMSO-d6) δ: 1.42 (3H, s), 1.70-1.78 (1H, m),






1.97-2.04 (3H, m), 2.55-2.60 (1H, m), 2.87-2.93 (1H, m), 3.26 (3H, s),






3.49 (2H, t, J = 6.2 Hz), 4.20 (2H, t, J = 6.4 Hz), 5.86 (2H, br s),






7.10 (1H, d, J = 7.8 Hz), 7.28 (1H, t, J = 8.1 Hz), 7.61 (1H, dd, J = 8.8,






2.8 Hz), 7.77-7.78 (2H, m), 8.11 (1H, d, J = 8.6 Hz),






8.38 (1H, d, J = 2.8 Hz), 10.32 (1H, s).



97


1H-NMR (DMSO-d6) δ: 1.41 (3H, s), 1.70-1.75 (1H, m),






1.99-2.06 (1H, m), 2.56-2.61 (1H, m), 2.87-2.93 (1H, m),






4.40-4.50 (2H, m), 4.74-4.88 (2H, m), 5.81 (2H, br s), 7.10 (1H, d, J = 7.6 Hz),






7.28 (1H, t, J = 8.2 Hz), 7.66 (1H, dd, J = 8.8, 2.8 Hz),






7.77-7.78 (2H, m), 8.13 (1H, d, J = 8.8 Hz), 8.43 (1H, d, J = 2.5 Hz),






10.33 (1H, s).



98


1H-NMR (DMSO-d6) δ: 1.41 (3H, s), 1.74-1.76 (7H, m),






1.99-2.06 (1H, m), 2.56-2.61 (1H, m), 2.86-2.93 (1H, m), 4.71 (2H, d,






J = 6.3 Hz), 5.44-5.49 (1H, m), 5.81 (2H, br s), 7.10 (1H, d, J = 7.3 Hz),






7.28 (1H, t, J = 8.1 Hz), 7.60 (1H, d, J = 8.8 Hz),






7.77 (2H, s), 8.11 (1H, d, J = 8.6 Hz), 8.36 (1H, s), 10.30 (1H, s).



99
799.0 (2M + 1)





100
827.0 (2M + 1)





101
867.1 (2M + 1)





102
865.1 (2M + 1)





103
382





104
412





105


1H-NMR (DMSO-d6) δ: 1.41 (4H, s), 1.69-1.74 (1H, m),






1.98-2.05 (1H, m), 2.55-2.60 (1H, m), 2.69-2.75 (2H, m),






2.86-2.93 (2H, m), 4.49 (2H, t, J = 6.4 Hz), 5.82 (2H, br s), 7.12 (1H, d, J = 7.3 Hz),






7.28 (1H, t, J = 7.7 Hz), 7.74 (1H, d, J = 7.8 Hz),






7.80 (1H, s), 8.42 (1H, s), 8.88 (1H, s), 10.34 (1H, s).



106


1H-NMR (DMSO-d6) δ: 1.41 (3H, s), 1.69-1.76 (3H, m),






1.98-2.05 (1H, m), 2.56-2.61 (1H, m), 2.64-2.69 (2H, m),






2.87-2.93 (1H, m), 4.45 (2H, t, J = 6.4 Hz), 5.80 (2H, br s), 7.12 (1H, d, J = 7.8 Hz),






7.28 (1H, t, J = 7.8 Hz), 7.74 (1H, d, J = 8.3 Hz),






7.80 (1H, s), 8.42 (1H, s), 8.88 (1H, s), 10.34 (1H, s).



107


1H-NMR (DMSO-d6) δ: 1.47-1.54 (1H, m), 1.86 (3H, s),






2.03-2.09 (1H, m), 2.88-2.94 (1H, m), 3.09-3.15 (1H, m),






4.43-4.47 (1H, m), 5.08-5.11 (2H, m), 5.76 (2H, br s), 7.04-7.06 (1H, m),






7.27-7.31 (1H, m), 7.68-7.70 (1H, m), 7.79 (1H, s), 8.45 (1H, s),






8.89 (1H, s), 10.39 (1H, s).



108
412





109
398





110


1H-NMR (DMSO-d6) δ: 1.45 (3H, s), 1.75-1.81 (1H, m),






2.08-2.14 (1H, m), 2.56-2.61 (1H, m), 2.90-2.97 (1H, m), 7.08 (1H, d,






J = 6.8 Hz), 7.27-7.36 (2H, m), 7.77-7.79 (2H, m), 8.01 (1H, d, J = 8.6 Hz),






8.22 (1H, d, J = 2.8 Hz), 10.27 (1H, s).



111


1H-NMR (DMSO-d6) δ: 1.45 (3H, s), 1.75-1.80 (1H, m),






2.07-2.14 (1H, m), 2.56-2.61 (1H, m), 2.90-2.97 (1H, m), 5.28 (2H, s),






7.09 (1H, d, J = 7.8 Hz), 7.23-7.32 (3H, m), 7.57 (2H, dd, J = 8.3,






5.6 Hz), 7.70 (1H, dd, J = 8.7, 2.7 Hz), 7.78-7.81 (2H, m),






8.13 (1H, d, J = 8.6 Hz), 8.45 (1H, d, J = 2.8 Hz), 10.36 (1H, s).



112
441




















TABLE 41





Compound
MS





No.
(M + 1)
MP
NMR (solvent, shift value: ascending order)
uv



















113


1H-NMR (DMSO-d6) δ: 1.43 (3H, s), 1.71-1.77 (1H, m),






2.05-2.13 (7H, m), 2.56-2.61 (1H, m), 2.88-2.94 (1H, m), 5.75 (2H, s),






7.12 (1H, d, J = 8.1 Hz), 7.30 (1H, t, J = 7.8 Hz), 7.74-7.80 (2H,






m), 8.66 (1H, s), 8.93 (1H, s), 10.44 (1H, s).



114


1H-NMR (DMSO-d6) δ: 1.47 (3H, s), 1.77-1.82 (1H, m),






2.13-2.20 (1H, m), 2.57-2.62 (1H, m), 2.93-3.00 (1H, m),






3.75-3.79 (2H, m), 4.18 (2H, t, J = 4.4 Hz), 5.00 (1H, br s), 5.89 (2H, br s),






7.12 (1H, dd, J = 11.4, 8.8 Hz), 7.58-7.63 (1H, m),






7.72-7.82 (2H, m), 8.09 (1H, d, J = 8.6 Hz), 8.39 (1H, s), 10.34 (1H, s).



115
416





116


1H-NMR (DMSO-d6) δ: 1.47 (4H, s), 1.76-1.81 (1H, m),






2.15-2.22 (1H, m), 2.55-2.60 (1H, m), 2.93-2.99 (1H, m), 4.47 (3H, s),






5.87 (2H, br s), 7.13 (1H, dd, J = 12.0, 8.7 Hz), 7.79 (2H, ddd, J = 18.3,






8.0, 3.1 Hz), 8.89 (1H, s), 9.23 (1H, s), 10.72 (1H, s).



117


1H-NMR (DMSO-d6) δ: 1.48 (3H, s), 1.77-1.82 (1H, m),






2.14-2.21 (1H, m), 2.57-2.62 (1H, m), 2.93-3.00 (1H, m), 3.32 (3H, s),






4.56-4.59 (2H, m), 5.87 (2H, br s), 6.27-6.33 (1H, m), 6.75 (1H,






d, J = 12.4 Hz), 7.13 (1H, dd, J = 11.9, 8.8 Hz), 7.77-7.83 (2H,






m), 8.77 (1H, s), 9.25 (1H, s), 10.64 (1H, s).



118
418





119
361





120
825.1 (2M + 1)





121
416





122


1H-NMR (DMSO-d6) δ: 1.41 (3H, s), 1.68-1.74 (1H, m),






1.98-2.05 (1H, m), 2.21 (6H, s), 2.55-2.60 (1H, m), 2.67 (2H, t, J = 5.6 Hz),






2.86-2.93 (1H, m), 4.48 (2H, t, J = 5.4 Hz), 5.79 (2H, br






s), 7.11 (1H, d, J = 7.8 Hz), 7.28 (1H, t, J = 7.8 Hz), 7.73 (1H, d,






J = 7.6 Hz), 7.79 (1H, s), 8.39 (1H, s), 8.86 (1H, s), 10.33 (1H,






s).



123
395





124
414





125
417





126


1H-NMR (DMSO-d6) δ: 1.45 (3H, s), 1.75-1.81 (1H, m),






2.13-2.20 (1H, m), 2.55-2.60 (1H, m), 2.92-2.99 (1H, m), 3.88 (6H, s),






5.81 (2H, br s), 7.08 (1H, dd, J = 11.6, 8.8 Hz), 7.50-7.55 (1H,






m), 7.66-7.69 (1H, m), 10.07 (1H, s).



127
380





128
372





129
418





130
412





131


1H-NMR (DMSO-d6) δ: 1.48 (3H, s), 1.77-1.82 (1H, m),






2.18-2.25 (1H, m), 2.55-2.60 (1H, m), 2.94-3.01 (1H, m), 4.72 (2H, t,






J = 13.8 Hz), 6.45 (1H, t, J = 53.9 Hz), 7.10-7.15 (1H, m),






7.74-7.79 (2H, m), 8.50 (1H, s), 8.87 (1H, s), 10.47 (1H, s).



132


1H-NMR (DMSO-d6) δ: 1.49 (3H, s), 1.78-1.83 (1H, m),






2.19-2.26 (1H, m), 2.56-2.60 (1H, m), 2.94-3.01 (1H, m),






4.61-4.86 (4H, m), 7.09-7.14 (1H, m), 7.75-7.79 (2H, m), 8.43 (1H, s),






8.84 (1H, s), 10.43 (1H, s).



133
400




















TABLE 42





Compound
MS





No.
(M + 1)
MP
NMR (solvent, shift value: ascending order)
uv







134


1H-NMR (DMSO-d6) δ: 1.37 (3H, t, J = 7.1 Hz), 1.49 (3H, s),






1.78-1.83 (1H, m), 2.19-2.26 (1H, m), 2.56-2.61 (1H, m),






2.95-3.01 (1H, m), 4.44 (2H, q, J = 7.0 Hz), 7.13 (1H, dd, J = 11.6,






9.1 Hz), 7.73-7.78 (2H, m), 8.35 (1H, s), 8.83 (1H, s), 10.41 (1H,






s)



135
411





136
412





137


1H-NMR (CDCl3) δ: 1.63 (3H, s), 1.81-1.91 (1H, m),






2.21-2.32 (1H, m), 2.56-2.67 (1H, m), 2.75-2.83 (1H, m), 3.77 (3H, s),






5.24 (2H, s), 6.47 (1H, dd, J = 3.2, 0.6 Hz), 6.83 (2H, d, J = 8.9 Hz),






7.02 (1H, dd, 8.0, 1.8 Hz), 7.07 (1H, d, 3.2 Hz), 7.12 (2H, d, J = 8.9 Hz),






7.26 (1H, dd, 1.8, 0.6 Hz), 7.57 (1H, d, J = 8.0 Hz).



138
400





139
246





140
356





141
376





142
410





143
378





144
398





145
432





146
529





147
377





148
438





149
390





150



212.2


151



211.0, 266.3,






301.8


152



285.2


153
403





154
403





155
404





156
388





157
389





158
412





159
380





160
381





161


1H-NMR (DMSO-d6) δ: 1.47 (3H, s), 1.77-1.82 (1H, m),






2.15-2.21 (8H, m), 2.56-2.67 (3H, m), 2.93-3.00 (1H, m), 4.21 (2H, t,






J = 5.4 Hz), 5.88 (2H, br s), 7.11 (1H, dd, J = 11.6, 9.3 Hz),






7.59-7.61 (1H, m), 7.73-7.80 (2H, m), 8.09 (1H, d, J = 8.6 Hz),






8.37 (1H, s), 10.33 (1H, s).



162
402





163
408





164
464





165
459





166
404





167
420





168
375





169
432





170
380





171
376


















TABLE 43





Compound




No.
Structure
MS(M + 1)







172


embedded image








173


embedded image


459





174


embedded image


403





175


embedded image


426





176


embedded image


393


















TABLE 44





Compound No
Structure
MS(M + 1)







177


embedded image


359





178


embedded image


402





179


embedded image


447





180


embedded image


435





181


embedded image


396


















TABLE 45





Compound




No
Structure
MS(M + 1)







182


embedded image


376





183


embedded image


385





184


embedded image


375





185


embedded image


378





186


embedded image


412


















TABLE 46





Compound




No.
Structure
MS(M + 1)







187


embedded image


366





188


embedded image


429





189


embedded image


364





190


embedded image


404





191


embedded image


439


















TABLE 47





Compound No.
Structure
MS(M + 1)







192


embedded image


412





193


embedded image


426





194


embedded image


393





195


embedded image


352





196


embedded image


414


















TABLE 48





Compound




No.
Structure
MS(M + 1)







197


embedded image








198


embedded image


414





199


embedded image


364





200


embedded image


397





201


embedded image


428


















TABLE 49





Compound




No.
Structure
MS (M + 1)







202


embedded image








203


embedded image


398





204


embedded image


410





205


embedded image


422





206


embedded image


395


















TABLE 50





Compound




No.
Structure
MS (M + 1)







207


embedded image


414





208


embedded image


410





209


embedded image


402





210


embedded image








211


embedded image




















TABLE 51





Com-




pound

MS


No.
Structure
(M + 1)







212


embedded image


433





213


embedded image


466





214


embedded image


464





215


embedded image


427





216


embedded image


400


















TABLE 52





Compound




No.
Structure
MS (M + 1)







217


embedded image


442





218


embedded image


386





219


embedded image


402





220


embedded image


362





221


embedded image




















TABLE 53





Com-




pound

MS


No.
Structure
(M + 1)







222


embedded image


399





223


embedded image








224


embedded image


352





225


embedded image


402





226


embedded image


395


















TABLE 54





Com-




pound

MS


No.
Structure
(M + 1)







227


embedded image


362





228


embedded image


375





229


embedded image


380





230


embedded image








231


embedded image


400


















TABLE 55





Compound




No.
Structure
MS (M + 1)







232


embedded image








233


embedded image








234


embedded image


422





235


embedded image


395





236


embedded image


364


















TABLE 56





Compound




No.
Structure
MS (M + 1)







237


embedded image


362





238


embedded image


427





239


embedded image


455





240


embedded image


420





241


embedded image


406


















TABLE 57





Compound




No.
Structure
MS (M + 1)







242


embedded image


471





243


embedded image


406





244


embedded image


420





245


embedded image


383





246


embedded image




















TABLE 58





Compound




No.
Structure
MS (M + 1)







247


embedded image


455





248


embedded image


435





249


embedded image


416





250


embedded image


416


















TABLE 59





Compound




No.
Structure
MS (M + 1)







251


embedded image


402





252


embedded image


388





253


embedded image


420





254


embedded image








255


embedded image


524


















TABLE 60





Compound




No.
Structure
MS (M + 1)







256


embedded image


348





257


embedded image








258


embedded image


395





259


embedded image


395





260


embedded image


402


















TABLE 61





Com-




pound

MS


No.
Structure
(M + 1)







261


embedded image


336





262


embedded image








263


embedded image








264


embedded image


334





265


embedded image


384


















TABLE 62





Com-




pound

MS


No.
Structure
(M + 1)







266


embedded image


402





267


embedded image


402





268


embedded image








269


embedded image


396





270


embedded image


427


















TABLE 63





Compound




No.
Structure
MS (M + 1)







271


embedded image


444





272


embedded image


416





273


embedded image


429





274


embedded image








275


embedded image


376


















TABLE 64





Compound




No.
Structure
MS(M + 1)







276


embedded image


425





277


embedded image


425





278


embedded image


429





279


embedded image


430





280


embedded image




















TABLE 65





Compound




No.
Structure
MS(M + 1)







281


embedded image


448





282


embedded image


411





283


embedded image








284


embedded image


438





285


embedded image




















TABLE 66





Com-




pound

MS


No.
Structure
(M + 1)







286


embedded image


437





287


embedded image


437





288


embedded image


348





289


embedded image


429





290


embedded image


448


















TABLE 67





Com-

MS


pound

(M +


No.
Structure
1)







291


embedded image


398





292


embedded image








293


embedded image


419





294


embedded image








295


embedded image


422


















TABLE 68





Compound




No.
Structure
MS(M + 1)







296


embedded image


430





297


embedded image


410





298


embedded image


410





299


embedded image








300


embedded image


401


















TABLE 69





Compound




No.
Structure
MS(M + 1)







301


embedded image








302


embedded image


400





303


embedded image


349





304


embedded image


426





305


embedded image


363


















TABLE 70





Compound




No.
Structure
MS(M + 1)







306


embedded image


415





307


embedded image








308


embedded image


424





309


embedded image


406





310


embedded image


383


















TABLE 71





Compound




No.
Structure
MS(M + 1)







311


embedded image


470





312


embedded image


422





313


embedded image


476





314


embedded image


401





315


embedded image


428


















TABLE 72





Compound




No.
Structure
MS(M + 1)







316


embedded image


413





317


embedded image


442





318


embedded image


442





319


embedded image


411





320


embedded image


434


















TABLE 73





Compound




No.
Structure
MS(M + 1)







321


embedded image








322


embedded image


463





323


embedded image








324


embedded image








325


embedded image


410


















TABLE 74





Com-

MS


pound

(M +


No.
Structure
1)







326


embedded image


390





327


embedded image


410





328


embedded image


410





329


embedded image


410





330


embedded image


384


















TABLE 75





Compound




No.
Structure
MS(M + 1)







331


embedded image


479





332


embedded image


429





333


embedded image


427





334


embedded image


427





335


embedded image


410


















TABLE 76





Compound




No.
Structure
MS(M + 1)







336


embedded image


428





337


embedded image


426





338


embedded image


401





339


embedded image


400





340


embedded image




















TABLE 77





Compound




No.
Structure
MS(M + 1)







341


embedded image


441





342


embedded image


442





343


embedded image


442





344


embedded image


430





345


embedded image


428


















TABLE 78





Compound




No.
Structure
MS(M + 1)







346


embedded image


430





347


embedded image


411





348


embedded image


413





349


embedded image


478





350


embedded image




















TABLE 79





Compound




No.
Structure
MS(M + 1)







351


embedded image


384





352


embedded image


443





353


embedded image


403





354


embedded image








355


embedded image


421


















TABLE 8





Compound




No.
Structure
MS(M + 1)







356


embedded image


422





357


embedded image


421





358


embedded image


369





359


embedded image


430





360


embedded image


424


















TABLE 81





Compound




No.
Structure
MS(M + 1)







361


embedded image


416





362


embedded image


429





363


embedded image








364


embedded image








365


embedded image


398


















TABLE 82





Compound




No.
Structure
MS(M + 1)







366


embedded image


425





367


embedded image


425





368


embedded image








369


embedded image


424


















TABLE 83





Compound




No.
Structure
MS(M + 1)







370


embedded image


413





371


embedded image


430





372


embedded image


408





373


embedded image


426





374


embedded image


437


















TABLE 84





Compound




No.
Structure
MS(M + 1)







375


embedded image


424





376


embedded image








377


embedded image


427





378


embedded image


424


















TABLE 85





Compound




No.
Structure
MS(M + 1)







379


embedded image


424





380


embedded image


493





381


embedded image


458





382


embedded image


395





383


embedded image


407


















TABLE 86





Com-




pound

MS


No.
Structure
(M + 1)







384


embedded image


416





385


embedded image


364





386


embedded image








387


embedded image








388


embedded image




















TABLE 87





Compound




No.
Structure
MS(M + 1)







389


embedded image








390


embedded image








391


embedded image








392


embedded image


413





393


embedded image


446


















TABLE 88





Compound




No.
Structure
MS(M + 1)







394


embedded image


445





395


embedded image


428





396


embedded image


413





397


embedded image


494





398


embedded image


428


















TABLE 89





Compound




No.
Structure
MS(M + 1)







399


embedded image


404





400


embedded image


375





401


embedded image


444





402


embedded image


444





403


embedded image


448


















TABLE 90





Compound




No.
Structure
MS(M + 1)







404


embedded image


440





405


embedded image


365





406


embedded image


414





407


embedded image


443





408


embedded image


385


















TABLE 91





Com-




pound

MS


No.
Structure
(M + 1)







409


embedded image


423





410


embedded image


410





411


embedded image








412


embedded image


393





413


embedded image


348


















TABLE 92





Compound




No.
Structure
MS(M + 1)







414


embedded image


414





415


embedded image


438





416


embedded image


410





417


embedded image








418


embedded image


464


















TABLE 93





Compound




No.
Structure
MS(M + 1)







419


embedded image


461





420


embedded image


462





421


embedded image


412





422


embedded image


466





423


embedded image


437


















TABLE 94





Compound




No.
Structure
MS (M + 1)







424


embedded image


411





425


embedded image


411





426


embedded image


351





427


embedded image


478





428


embedded image


462


















TABLE 95





Compound




No.
Structure
MS (M + 1)







429


embedded image








430


embedded image


443





431


embedded image


470





432


embedded image








433


embedded image


378


















TABLE 96





Compound




No.
Structure
MS (M + 1)







434


embedded image


451





435


embedded image


355





436


embedded image


351





437


embedded image


509





438


embedded image


420


















TABLE 97





Compound




No.
Structure
MS (M + 1)







439


embedded image


429





440


embedded image


406





441


embedded image


494





442


embedded image


458





443


embedded image


483


















TABLE 98





Compound




No.
Structure
MS (M + 1)







444


embedded image


457





445


embedded image


452





446


embedded image


550





447


embedded image


437


















TABLE 99





Compound




No.
Structure
MS (M + 1)







448


embedded image


495





449


embedded image


455





450


embedded image


481





451


embedded image


426


















TABLE 100





Compound




No.
Structure
MS (M + 1)







452


embedded image


454





453


embedded image


480





454


embedded image


404





455


embedded image


441


















TABLE 101





Compound




No.
Structure
MS (M + 1)







456


embedded image


417





457


embedded image


395





458


embedded image


362





459


embedded image


393





460


embedded image























TABLE 102





Compound

MS

NMR (solvent, shift



No.
Structure
(M + 1)
MP
value: ascending order)
uv







461


embedded image


346








462


embedded image


349

1H-NMR (DMSO-d6) d: 10.02 (1.0H, s), 8.59 (1.0H, s), 7.73-7.66 (2.0H, m), 7.09 (1.0H, dd, J = 12.00, 8.97 Hz), 5.83 (2.0H, br s), 2.97-2.95 (1.0H, m), 2.59-2.56 (1.0H, m), 2.17-2.16 (1.0H, m), 1.79-1.76 (1.0H, m), 1.47 (3.0H, s).






463


embedded image


362








464


embedded image


441








465


embedded image


456








466


embedded image




1H-NMR (DMSO-d6) d: 10.70 (1.0H, s), 8.76 (1.0H, s), 8.36 (1.0H, s), 8.03 (1.0H, s), 6.44 (1.0H, s), 5.93 (2.0H, br s), 3.00- 2.97 (1.0 H, m), 2.63-2.61 (1.0H, m), 2.19 (3.0H, s), 2.00-1.98 (1.0H, m), 1.82-1.80 (1.0H, m), 1.60 (9.0H, s), 1.43 (3.0H, s).





















TABLE 103





Compound

MS

NMR (solvent, shift



No.
Structure
(M + 1)
MP
value: ascending order)
uv










467


embedded image


369








468


embedded image


396








469


embedded image


450








470


embedded image


383








471


embedded image


417








472


embedded image


364





















TABLE 104





Compound

MS

NMR (solvent, shift



No.
Structure
(M + 1)
MP
value: ascending order)
uv







473


embedded image


361








474


embedded image


332








475


embedded image


378








476


embedded image


345








477


embedded image


392








478


embedded image


365








479


embedded image


359





















TABLE 105





Compound

MS

NMR (solvent, shift



No
Structure
(M + 1)
MP
value: ascending order)
uv







480


embedded image


360








481


embedded image


366








482


embedded image


345








483


embedded image


394








484


embedded image


385








485


embedded image


347








486


embedded image


347





















TABLE 106





Compound

MS

NMR (solvent, shift



No
Structure
(M + 1)
MP
value: ascending order)
uv







487


embedded image


347








488


embedded image


362








489


embedded image


405








490


embedded image


381








491


embedded image


379








492


embedded image


421








493


embedded image


379





















TABLE 107





Compound

MS

NMR (solvent, shift



No.
Structure
(M + 1)
MP
value: ascending order)
uv







494


embedded image


426








495


embedded image


363








496


embedded image


378








497


embedded image


426








498


embedded image


374








499


embedded image


374








500


embedded image


363





















TABLE 108





Compound

MS

NMR (solvent, shift



No.
Structure
(M + 1)
MP
value: ascending order)
uv







501


embedded image


400








502


embedded image


384








503


embedded image


359








504


embedded image


367








505


embedded image


365








506


embedded image


365








507


embedded image


365





















TABLE 109





Compound

MS

NMR (solvent,



No.
Structure
(M + 1)
MP
shift value: ascending order)
uv







508


embedded image


365








509


embedded image


411








510


embedded image


363








511


embedded image


363








512


embedded image


393








513


embedded image


408








514


embedded image


413





















TABLE 110





Compound

MS

NMR (solvent,



No.
Structure
(M + 1)
MP
shift value: ascending order)
uv







515


embedded image


411








516


embedded image


413








517


embedded image


441








518


embedded image


348








519


embedded image


429








520


embedded image


394








521


embedded image


402





















TABLE 111





Compound

MS

NMR (solvent,



No.
Structure
(M + 1)
MP
shift value: ascending order)
uv







522


embedded image


378








523


embedded image


441








524


embedded image


380








525


embedded image


379








526


embedded image


414








527


embedded image


428





















TABLE 112





Compound

MS

NMR (solvent,



No.
Structure
(M + 1)
MP
shift value: ascending order)
uv







528


embedded image


433








529


embedded image


362








530


embedded image


392








531


embedded image


426








532


embedded image


364








533


embedded image


364





















TABLE 113





Compound

MS

NMR (solvent,



No.
Structure
(M + 1)
MP
shift value: ascending order)
uv







534


embedded image


404








535


embedded image


394








536


embedded image


383








537


embedded image


428








538


embedded image


404








539


embedded image


401





















TABLE 114





Compound

MS

NMR (solvent,



No.
Structure
(M + 1)
MP
shift value: ascending order)
uv







540


embedded image


384








541


embedded image


442








542


embedded image


401








543


embedded image


404








544


embedded image


511








545


embedded image


400





















TABLE 115





Compound

MS

NMR (solvent,



No.
Structure
(M + 1)
MP
shift value: ascending order)
uv







546


embedded image




1H-NMR (DMSO-d6) d: 10.92 (1H, s), 10.45 (1H, s), 8.45 (1H, s), 8.42 (1H, s), 8.30 (1H, d, J = 8.8 Hz), 7.79-7.78 (3H, m), 7.31 (1H, dd, J = 12.3, 9.2 Hz), 3.22 (1H, d, J = 13.4 Hz), 2.72-2.65 (2H, m), 2.11 (1H, t, J = 11.5 Hz), 1.73 (3H, s).






547


embedded image


359








548


embedded image


359








549


embedded image


403








550


embedded image


343








551


embedded image


343





















TABLE 116





Compound

MS

NMR (solvent,



No.
Structure
(M + 1)
MP
shift value: ascending order)
uv







552


embedded image


363








553


embedded image


348








554


embedded image


363








555


embedded image


374








556


embedded image


383








557


embedded image




1H-NMR (DMSO-d6) δ. 10 72 (1H, s), 8 93 (1H, s), 8.90 (1H, s), 8.49 (1H, s), 8.36 (1H, s), 8.24 (1H, s), 4.88-4 64 (4H, m), 3.03-2.97 (1H, m), 2.65-2.58 (1H, m), 2.13-2.07 (1H, m), 1.89-1.81 (1H, m), 1.48 (3H, s).






558


embedded image


402





















TABLE 117





Compound

MS

NMR (solvent,



No.
Structure
(M + 1)
MP
shift value: ascending order)
uv







559


embedded image


418








560


embedded image


387








561


embedded image


411








562


embedded image


431








563


embedded image


342








564


embedded image


372








565


embedded image


390





















TABLE 118





Compound

MS

NMR (solvent,



No.
Structure
(M + 1)
MP
shift value: ascending order)
uv







566


embedded image


428








567


embedded image


429








568


embedded image


419








569


embedded image


442








570


embedded image


456








571


embedded image


443








572


embedded image


396





















TABLE 119





Compound

MS

NMR (solvent,



No.
Structure
(M + 1)
MP
shift value: ascending order)
uv







573


embedded image


447








574


embedded image


430








575


embedded image


458








576


embedded image


412








577


embedded image


426








578


embedded image


426








579


embedded image


440





















TABLE 120





Compound

MS

NMR (solvent,



No.
Structure
(M + 1)
MP
shift value: ascending order)
uv







580


embedded image


480








581


embedded image


363








582


embedded image


393








583


embedded image


437








584


embedded image


366








585


embedded image


360








586


embedded image


380





















TABLE 121





Compound

MS

NMR (solvent,



No.
Structure
(M + 1)
MP
shift value: ascending order)
uv







587


embedded image


363








588


embedded image


323








589


embedded image





250.9, 288.7 





590


embedded image





298.2 





591


embedded image





252.1, 305.3 





592


embedded image





250.9, 288.7 





593


embedded image





216.9, 292.3 





















TABLE 122





Compound

MS

NMR (solvent,



No.
Structure
(M + 1)
MP
shift value: ascending order)
uv







594


embedded image





214.5, 289.9 





595


embedded image





297   





596


embedded image





250.9, 302.9 





597


embedded image





289.9 





598


embedded image





297   





599


embedded image





214.5, 289.9 





600


embedded image


404, 807 (2M + 1)





















TABLE 123





Compound

MS

NMR (solvent,



No.
Structure
(M + 1)
MP
shift value: ascending order)
uv







601


embedded image


448, 895  (2M + 1)








602


embedded image


389








603


embedded image


391








604


embedded image


391








605


embedded image


436








606


embedded image


388





















TABLE 124





Compound

MS

NMR (solvent,



No
Structure
(M + 1)
MP
shift value:ascending order)
uv







607


embedded image











608


embedded image











609


embedded image


377








610


embedded image











611


embedded image











612


embedded image


332





















TABLE 125





Compound

MS

NMR (solvent,



No
Structure
(M + 1)
MP
shift value:ascending order)
uv







613


embedded image


346








614


embedded image











615


embedded image











616


embedded image











617


embedded image











618


embedded image























TABLE 126





Compound

MS

NMR (solvent,



No.
Structure
(M + 1)
MP
shift value:ascending order)
uv







619


embedded image











620


embedded image











621


embedded image











622


embedded image











623


embedded image











624


embedded image























TABLE 127





Compound

MS

NMR (solvent,



No.
Structure
(M + 1)
MP
shift value:ascending order)
uv







625


embedded image











626


embedded image











627


embedded image











628


embedded image











629


embedded image











630


embedded image























TABLE 128





Compound

MS

NMR (solvent,



No
Structure
(M + 1)
MP
shift value:ascending order)
uv







631


embedded image











632


embedded image











633


embedded image











634


embedded image











635


embedded image











636


embedded image























TABLE 129





Compound

MS

NMR (solvent,



No.
Structure
(M + 1)
MP
shift value:ascending order)
uv







637


embedded image


387








638


embedded image











639


embedded image











640


embedded image











641


embedded image











642


embedded image























TABLE 130





Compound

MS

NMR (solvent,



No
Structure
(M + 1)
MP
shift value:ascending order)
uv







643


embedded image











644


embedded image


475








645


embedded image


397








646


embedded image


414



















TABLE 131





Compound

MS



No.
Structure
[M + 1]
NMR (solvent, shift value)







647


embedded image


404






648


embedded image


377






649


embedded image


388






650


embedded image


389






651


embedded image


453






652


embedded image


399






653


embedded image


371



















TABLE 132





Com-

MS



pound

[M +



No
Structure
1]
NMR (solvent, shift value)







654


embedded image


360






655


embedded image


374






656


embedded image


458






657


embedded image


411






658


embedded image


419






659


embedded image


383






660


embedded image



1H-NMR (CDCl3) δ: 1.84 (3H, d-like), 3.16 (1H, ddd, J = 6.9, 12.6, 14.4 Hz), 3.36 (1H, ddd, J = 6.0, 12.6, 18 9 Hz), , 4.61 (2H, br), 7.07 (1H, dd, J = 8.7, 11.7 Hz), 7.49-7.53 (2H, m), 7.67 (1H, dd, J = 3.0, 6.9 Hz), 7.95 (1H, ddd, J = 3.0, 6.9, 8 7 Hz), 8 41 (1H, m), 9.85 (1H, brs).



















TABLE 133





Com-

MS



pound

[M +



No
Structure
1]
NMR (solvent, shift value)







661


embedded image



1H-NMR (CDCl3) δ: 0.89 (3H, s), 1.11 (3H, d, J = 3.0 Hz), 1.67 (3H, d, J = 4.2 Hz), 2.63 (1H, d, J = 12 0 Hz), 3.12 (1H, d, J = 12.0 Hz), 4.29 (2H, br), 7.02 (1H, dd, J = 8.7, 12.3 Hz), 7.49-7.64 (3H, m), 7.96 (1H, ddd, J = 3.0, 6.6, 8.7 Hz), 8.45 (1H, m), 3.81 (1H, brs).





662


embedded image



1H-NMR (CDCl3) δ: 1.85 (3H, d-like), 2.69 (3H, s), 3.17 (1H, ddd, J = 6.9, 12.6, 14.4 Hz), 3.37 (1H, ddd, J = 6.3, 12.9, 18 9 Hz), 4.54 (2H, brs), 7.08 (1H, dd, J = 8.7, 11.7 Hz), 7.69 (1H, dd, J = 2.7, 6.9 Hz), 7.87 (1H, dd, J = 2.4, 8.4 Hz), 7 93 (1H, ddd, J = 2.7, 6.6, 8.7 Hz), 8.24 (1H, dd, J = 0.6, 8.4 Hz), 8.55 (1H, dd, J = 0.6, 2.4 Hz), 9.82 (1H, brs).





663


embedded image



1H-NMR (CDCl3) δ: 1.85 (3H, d-like), 3.18 (1H, ddd, J = 7.2, 12.9, 15.0 Hz), 3.37 (1H, ddd, J = 6.0, 12.6, 18.9 Hz), 4.60 (2H, br), 7.08 (1H, dd, J = 8.7, 11.7 Hz), 7.71 (1H, dd, J = 3.0, 6.5 Hz), 7.90 (1H, ddd, J = 3.0, 6.6, 8.7 Hz), 8.41 (1H, d, J = 0.9 Hz), 9.35 (1H, d, J = 0.9 Hz), 9.63 (1H, brs).





664


embedded image



1H-NMR (CDCl3) δ: 1.83 (3H, d-like), 2.51 (3H, s), 3.16 (1H, ddd, J = 6.9, 12.9, 15.3 Hz), 3.34 (1H, jddd, J = 6.3, 12.9, 19.2 Hz), 4.53 (2H, brs), 7.05 (1H, dd, J = 8.7, 11.4 Hz), 7.62 (1H, dd, J = 2.7, 5 9 Hz), 7.82 (1H, ddd, J = 2.7, 6.9, 8.7 Hz), 8.16 (1H, s), 8.70 (1H, brs).





665


embedded image



1H-NMR (CDCl3) δ: 0.95 (3H, t, J = 7.5 Hz), 1.32- 1.44 (2H, m), 1.59-1.69 (2H, m), 1.85 (3H, d-like), 2.70 (2H, t, J = 7.5 Hz), 3 17 (1H, ddd, J = 6.9, 12.9, 15.3 Hz), 3.34 (1H, ddd, J = 8.3, 12.9, 19.2 Hz), 4.53 (2H, brs), 7.07 (1H, dd, J = 8.7, 11.7 Hz), 7.87- 7.70 (2H, m), 7.94 (1H, ddd, J = 2.7, 6.6, 8 7 Hz), 8.18 (1H, dd, J = 0.8, 7.8 Hz), 8.40 (1H, dd, J = 0.8, 1.8 Hz), 9.97 (1H, brs).





666


embedded image


396






667


embedded image


365



















TABLE 134





Compound

MS



No
Structure
[M + 1]
NMR (solvent, shift value)







668


embedded image


356






669


embedded image


403






670


embedded image



1H-NMR (CDCl3) δ: 1.38 (3H, t, J = 7.6 Hz), 1.63 (3H, s), 1.88-1.97 (1H, m), 2.41-2.50 (1H, m), 2.69- 2.78 (1H, m), 2.84 (2H, q, J = 7.6 Hz), 2.93-3.01 (1H, m), 7.02 (1H, dd, J = 11.8, 8.8 Hz), 7.34 (1H, dd, J = 7.1, 2.8 Hz), 7 89 (1H, ddd, J = 8.8, 4.3, 2 8 Hz), 8.16 (1H, s), 8.69 (1H, s).





671


embedded image


370






672


embedded image


432






673


embedded image


412






674


embedded image



1H-NMR (CDCl3) δ: 0.53-0.59 (1H, m), 0.65-0.72 (1H, m), 0.85-0 91 (1H, m), 1.14-1.17 (1H, m), 1.47 (3H, d, J = 2.0 Hz), 2.46 (1H, d, J = 12.1 Hz), 2.69 (3H, s), 2.88 (1H, dd, J = 12.1, 1.3 Hz), 7.06 (1H, dd, J = 11.5, 8.8 Hz), 7.45 (1H, dd, J = 6.8, 2.8 Hz), 7 94 (1H, ddd, J = 8.8, 4.0, 2.8 Hz), 8.44 (1H, d, J = 1.3 Hz), 9.36 (1H, d, J = 1.3 Hz), 9.60 (1H, s).



















TABLE 135





Compound

MS



No.
Structure
[M + 1]
NMR (solvent, shift value)







675


embedded image


402






676


embedded image


426






677


embedded image


396






678


embedded image


430






679


embedded image


372






680


embedded image




1H-NMR (DMSO-d6) δ: 1.47 (3H, s), 1.77-1.83 (1H, m), 2.34-2.39 (1H, m), 2.48-2.53 (1H, m), 2.63 (3H, s), 2.89-2.96 (1H, m), 3.90 (3H, s), 5.86 (2H, br s), 8.10 (1H, d, J= 2.3 Hz), 8.47 (1H, d, J = 2.5 Hz), 8.69 (1H, s), 9.14 (1H, s), 10.69 (1H, s).






681


embedded image




1H-MMR (DMSO-d6) δ: 1.52 (3H, s), 1.80-1.85 (1H, m), 2.62 (3H, s), 2.64-2.69 (2H, m), 2.96-3.01 (1H, m), 7.77 (1H, d, J = 2.5 Hz), 7.96 (1H, d, J = 2.3 Hz), 8.67 (1H, s), 9.10 (1H, s), 10.58 (1H, s).




















TABLE 136





Compound

MS



No
Structure
[M + 1]
NMR (solvent, shift value)







682


embedded image




1H-NMR (DMSO-d6) δ: 1.46 (3H, s), 1.95-2.01 (1H, m), 2.33-2.39 (1H, m), 2 62 (3H, s), 2 64-2.69 (1H, m), 2.74 (3H, s), 2.92-2.98 (1H, m), 7.90 (1H, d, J = 2.5 Hz), 7.94-7.95 (1H, m), 8.67 (1H, s), 9.09 (1H, s), 10.57 (1H, s).






683


embedded image


482






684


embedded image


482






685


embedded image


400






686


embedded image


424






687


embedded image


427






688


embedded image


402



















TABLE 137





Com-

MS



pound

[M +



No.
Structure
1]
NMR (solvent, shift value)







689


embedded image


390






690


embedded image


413






691


embedded image


374






692


embedded image


428






693


embedded image



1H-NMR (DMSO-d6) δ: 0.77 (3H, s), 1.30 (3H, s), 1 43 (3H, s), 1.71 (1H, d, J = 13.8 Hz), 2.33 (1H, d, J = 13.8 Hz), 3.65 (1H, t, J = 2.4 Hz), 5.13 (2H, d, J = 2.4 Hz), 6.05 (2H, br), 7.19 (1H, d-like), 7.25 (1H, t, J = 7.8 Hz), 7.71 (1H, d-like), 7.87 (1H, s-like), 8.47 (1H, d, J = 1.2 Hz), 8.90 (1H, d, J = 1.2 Hz), 10.37 (1H, brs).





694


embedded image



1H-NMR (DMSO-d6) δ: 0.77 (3H, s), 1.28 (3H, s), 1 42 (3H, s), 1.67 (1H, d, J = 14.1 Hz), 2.23 (1H, d, J = 14.1 Hz), 4.02 (3H, s), 5.82 (2H, brs), 7.19 (1H, d- like), 7.24 (1H, t, J = 7.8 Hz), 7.70 (1H, d-like), 7.86 (1H, s-like), 8.40 (1H, d, J = 1.2 Hz), 8.89 (1H, d, J = 1.2 Hz), 10.31 (1H, brs).





695


embedded image



1H-NMR (DMSO-d6) δ: 0.76 (3H, s), 1.32 (3H, s), 1 48 (3H, s), 1.78 (1H, d, J = 14.1 Hz), 1.86 (3H, t, J = 2.4 Hz), 2.39 (1H, d, J = 14.1 Hz), 5.90 (1H, q, J = 2.4 Hz), 6 49 (2H, br), 7.18 (1H, d-like), 7.27 (1H, t, J = 7.8 Hz), 7.73 (1H, d-like), 7.87 (1H, s-like), 8.44 (1H, d, J = 1.2 Hz), 8.89 (1H, d, J = 1.2 Hz), 10.38 (1H, brs).



















TABLE 138





Compound

MS



No.
Structure
[M + 1]
NMR (solvent, shift value)







696


embedded image



1H-NMR (DMSO-d6) δ 0.77 (3H, s), 1.30 (3H, s), 1.43 (3H, s), 1.71 (1H, d, J = 14.1 Hz), 2.32 (1H, d, J = 14.1 Hz), 5.95 (2H, br), 7.22 (1H, d-like), 7 27 (1H, t, J = 7.8 Hz), 7.75 (1H, d-like), 7.88 (1H, s-like), 8.29 (1H, dd, J = 0.6, 8.1 Hz), 8.58 (1H, dd, J = 2.1, 8.1 Hz), 9.19 (1H, dd, J = 0.6, 2.1 Hz), 10.65 (1H, brs)





697


embedded image


410






698


embedded image









699


embedded image









700


embedded image









701


embedded image


400






702


embedded image


443









Test Example Assay of β-Secretase-Inhibiting Activity

Forty eight point five μL of substrate peptide solution (Biotin-XSEVNLDAEFRHDSGC-Eu: X-ε-amino-n-capronic acid, Eu=Europium cryptate) was added to each well of 96-hole half-area plate (a black plate: Corning Incorporated), and after addition of 0.5 μl of the test sample (dissolved in N,N′-dimethylformaldehyde) and 1 μl of Recombinant human BACE-1 (R&D Systems), the reaction mixture was incubated at 30° C. for 3 hours. The substrate peptide was synthesized by reacting Cryptate TBPCOOH mono SMP (CIS bio international) with Biotin-XSEVNLDAEFRHDSGC (Peptide Institute, Inc.). The final concentrations of the substrate peptide and Recombinant human BACE-1 were adjusted to 18 nM and 7.4 nM respectively, and the reaction was performed in sodium acetate buffer (50 mM sodium acetate, pH 5.0, 0.008% Triton X-10).


After the incubation for reaction, 50 μl of 8.0 μg/ml Streptavidin-XL665 (CIS bio international) dissolved in phosphate buffer (150 mM K2HPO4—KH2PO4, pH 7.0, 0.008% Triton X-100, 0.8 M KF) was added to each well and left stand at 30° C. for an hour. After then, fluorescence intensity was measured (excitation wavelength: 320 nm, measuring wavelength: 620 nm and 665 nm) using Wallac 1420 multilabel counter (Perkin Elmer life sciences). Enzymatic activity was determined from counting ratio of each wavelength (10,000×Count 665/Count 620) and 50% inhibitory concentration against the enzymatic activity was calculated. IC50 values of the test compounds are indicated in Table 139.











TABLE 139






Compound
IC50



No.
(uM)


















3
0.08



11
0.17



12
0.16



26
4.85



34
0.10



38
0.14



41
0.15



62
0.17



65
0.72



66
0.15



70
0.09



71
0.16



72
0.11



76
0.18



80
0.07



86
0.19



87
0.09



92
0.08



93
0.08



94
0.17



101
0.08



105
0.13



106
0.12



109
0.10



111
0.18



114
0.16



126
2.14



136
0.11



141
0.12



149
9.25



150
2.48



151
6.77



155
5.96



163
6.79



164
0.08









The following compounds have shown IC50 values equal to or under 1 μM in the same assay; same assay;

    • compounds 4, 5, 6, 8, 10, 18, 19, 20, 21, 22, 29, 32, 33, 35, 43, 45, 46, 58, 59, 63, 64, 68, 69, 75, 77, 78, 79, 81, 82, 83, 84, 85, 88, 89, 90, 91, 95, 96, 97, 98, 100, 102, 103, 104, 107, 108, 110, 112, 113, 115, 116, 117, 118, 119, 120, 121, 123, 124, 125, 127, 131, 132, 133, 134, 135, 142, 143, 144, 145, 148, 152, 157, 158, 162 and 165.


Also, compounds 462, 463, 465, 467, 469, 470, 471, 472, 479, 482, 483, 486, 489, 490, 492, 501, 503, 507, 508, 509, 510, 511, 512, 516, 518, 519, 523, 527, 528, 531, 532, 533, 536, 538, 539, 540, 542, 545, 546, 547, 548, 549, 552, 553, 554, 555, 556, 557, 558, 560, 561, 562, 564, 565, 567, 568, 569, 570, 571, 572, 573, 574, 575, 578, 581, 582, 583, 584, 586, 587, 590, 595, 596, 600, 601, 602, 603, 604, 605, 606, 609, 612, 613, 637, 644, 646, 461, 468, 478, 491, 502, 505, 508, 517, 530, 537, 542, 544, 559, 563, 566, 576, 577, 597, 598, 599 and 645 showed IC50 values equal to or under 1 μM in the same assay.


The following compounds also showed IC50 values equal to or under 1 μM in the same assay;


compounds 647, 648, 649, 650, 651, 654, 656, 657, 658, 659, 661, 666, 670, 671, 672, 673, 675, 676, 677, 678, 679, 683, 684, 685, 686, 687, 688, 690, 691, 692, 693, 694, 695, 696 and 697, 652, 655, 660, 662, 664, 665, 667, 669, 674 and 689.


Formulation Example 1
Granular Formulation is Prepared with the Following Ingredients

















Ingredient
compound of the formula (I)
 10 mg




lactose
700 mg




corn starch
274 mg




HPC-L
 16 mg





1000 mg 









Compound of the formula (I) and lactose are put through a sieve of No. 60 mesh. Corn starch is put through a sieve of No. 120 mesh and these are mixed with V-shaped mixer.


An aqueous solution of HPC-L (Hydroxypropyl cellulose of Low viscosity) is added to the mixed powder, kneaded, granulated (extusion granulation; pore diameter 0.5-1 mm) and put into a drying process. The resulted dried granule is sieved with vibrating screen (12/60 mesh) to give a granular formulation.


Formulation Example 2
Granular Formulation for Capsule Filling is Prepared with the Following Ingredients

















Ingredient
compound of the formula (I)
15 mg




lactose
90 mg




corn starch
42 mg




HPC-L
 3 mg





150 mg 









Compound of the formula (I) and lactose are put through a sieve of No. 60 mesh. Corn starch is put through a sieve of No. 120 mesh and these are mixed. An aqueous solution of HPC-L is added to the mixed powder, kneaded, granulated and dried. Particle size of the resulted dried granule is regulated and each of 150 mg is filled in No. 5 hard-gelatin capsule.


Formulation Example 3
Tablet is Prepared with the Following Ingredients

















Ingredient
compound of the formula (I)
10 mg




lactose
90 mg




microcrystalline cellulose
30 mg




CMC-Na
15 mg




magnesium stearate
 5 mg





150 mg 









Compound of the formula (I), lactose, microcrystalline cellulose and CMC-Na (sodium salt of carboxymethylcellulose) are put through a sieve of No. 60 mesh and mixed. Magnesium stearate is mixed with the mixed granule above to give a mixed powder for tablet, which is compressed by a tabletting machine to give a tablet of 150 mg.


Formulation Example 4
The Following Ingredients were Warmed, Mixed and Sterilized to Give an Injection

















Ingredient
compound of the formula (I)
 3 mg




non-ionic surfactant
15 mg




purified water for injection
 1 ml









INDUSTRIAL APPLICABILITY

A compound of the present invention can be a useful drug for treating diseases induced by production, secretion and/or deposition of amyloid β protein.

Claims
  • 1. A compound of the formula ab:
  • 2. The Compound of claim 1, wherein ring A is
  • 3. The Compound of claim 1 wherein ring A is
  • 4. A compound selected from the group consisting of:
Priority Claims (2)
Number Date Country Kind
2007-114288 Apr 2007 JP national
2007-290589 Nov 2007 JP national
Parent Case Info

This application is a Continuation of application Ser. No. 13/417,786, filed Mar. 12, 2012, which is a Divisional of application Ser. No. 12/596,796, filed Dec. 3, 2009, which is a National Stage Application of PCT/JP2008/057847, filed Apr. 23, 2008, which applications are incorporated herein by reference.

US Referenced Citations (67)
Number Name Date Kind
2899426 Bloom Aug 1959 A
3115494 Joseph et al. Dec 1963 A
3227713 Behner Jan 1966 A
3235551 Werner Feb 1966 A
3577428 Suh et al. May 1971 A
3636116 Trepanier Jan 1972 A
3719674 Trepanier Mar 1973 A
3775409 Harsanyi et al. Nov 1973 A
4049807 Paulus et al. Sep 1977 A
4311840 Condon Jan 1982 A
4895841 Sugimoto et al. Jan 1990 A
4906626 Amrein et al. Mar 1990 A
5100901 Sugimoto et al. Mar 1992 A
5236942 Miller Aug 1993 A
5328915 Long et al. Jul 1994 A
5880147 Yoshida et al. Mar 1999 A
5952374 Clarkson, Jr. et al. Sep 1999 A
6096753 Spohr et al. Aug 2000 A
6590123 Bekesi et al. Jul 2003 B2
6713276 Cordell et al. Mar 2004 B2
7183070 Cordell et al. Feb 2007 B2
7309706 Rupp et al. Dec 2007 B2
7326792 Shum et al. Feb 2008 B2
7414050 Illig et al. Aug 2008 B2
7763609 Zhu et al. Jul 2010 B2
7902238 Galley et al. Mar 2011 B2
8173642 Kobayashi et al. May 2012 B2
20020019427 Carry et al. Feb 2002 A1
20050165080 Rupp et al. Jul 2005 A1
20060173006 Sun et al. Aug 2006 A1
20060183790 Cole et al. Aug 2006 A1
20060183792 Fobare et al. Aug 2006 A1
20060183943 Hu Aug 2006 A1
20070004730 Zhou et al. Jan 2007 A1
20070004786 Malamas et al. Jan 2007 A1
20070027199 Malamas et al. Feb 2007 A1
20070224656 Cordell et al. Sep 2007 A1
20080200445 Zhu et al. Aug 2008 A1
20090023729 Nakamuta et al. Jan 2009 A1
20090082560 Kobayashi et al. Mar 2009 A1
20100093999 Motoki et al. Apr 2010 A1
20100125087 Holenz et al. May 2010 A1
20100160290 Kobayashi et al. Jun 2010 A1
20100234365 Liu et al. Sep 2010 A1
20100261727 Chi et al. Oct 2010 A1
20110009395 Audia et al. Jan 2011 A1
20110046122 Andreini et al. Feb 2011 A1
20110065695 Beauchamp et al. Mar 2011 A1
20110190279 Hori et al. Aug 2011 A1
20110237576 Yonezawa et al. Sep 2011 A1
20120015961 Tamura et al. Jan 2012 A1
20120016116 Kobayashi et al. Jan 2012 A1
20120022249 Kobayashi et al. Jan 2012 A1
20120202828 Castro Pineiro et al. Aug 2012 A1
20120238548 Gabellieri et al. Sep 2012 A1
20120238557 Masui et al. Sep 2012 A1
20120245154 Anan et al. Sep 2012 A1
20120245155 Yoshida et al. Sep 2012 A1
20120245157 Masui et al. Sep 2012 A1
20120253035 Narquizian et al. Oct 2012 A1
20120258961 Suzuki et al. Oct 2012 A1
20120258962 Hilpert et al. Oct 2012 A1
20120295900 Hilpert et al. Nov 2012 A1
20130158260 Kobayashi et al. Jun 2013 A1
20130210839 Masui et al. Aug 2013 A1
20130217705 Mitsuoka et al. Aug 2013 A1
20130303755 Kobayashi et al. Nov 2013 A1
Foreign Referenced Citations (182)
Number Date Country
2163724 May 1996 CA
2165386 Jun 1996 CA
140144 Feb 1980 DE
0 798 292 Oct 1995 EP
0 713 704 May 1996 EP
0 718 294 Jun 1996 EP
0717040 Jun 1996 EP
1043312 Oct 2000 EP
1 283 039 Feb 2003 EP
1 577 294 Sep 2005 EP
1 942 105 Jul 2008 EP
1942105 Jul 2008 EP
2147914 Jan 2010 EP
2151435 Feb 2010 EP
2 233 474 Sep 2010 EP
2 305 672 Apr 2011 EP
2332943 Jun 2011 EP
2 360 155 Aug 2011 EP
2415756 Feb 2012 EP
1001093 Aug 1965 GB
62-120374 Jun 1987 JP
8-231521 Sep 1996 JP
9-067355 Mar 1997 JP
10-505862 Jun 1998 JP
11-349572 Dec 1999 JP
2005-509651 Apr 2004 JP
2004-149429 May 2004 JP
2005-517634 Jun 2005 JP
2005-526005 Sep 2005 JP
2005-531520 Oct 2005 JP
2006-519758 Aug 2006 JP
2009-051828 Mar 2009 JP
2009-520685 May 2009 JP
WO 9412165 Jun 1994 WO
WO 9509619 Apr 1995 WO
WO 9609286 Mar 1996 WO
WO 9614842 May 1996 WO
WO 9618608 Jun 1996 WO
WO 9707098 Feb 1997 WO
9714686 Apr 1997 WO
9738977 Oct 1997 WO
9918960 Apr 1999 WO
0000200 Jan 2000 WO
WO 0119788 Mar 2001 WO
WO 0178709 Oct 2001 WO
WO 0187293 Nov 2001 WO
02062766 Aug 2002 WO
02088101 Nov 2002 WO
WO 02096897 Dec 2002 WO
03040096 May 2003 WO
WO 03039446 May 2003 WO
WO 03040115 May 2003 WO
WO 03040142 May 2003 WO
WO 03082191 Oct 2003 WO
2004009549 Jan 2004 WO
WO 2004014843 Feb 2004 WO
2004031154 Apr 2004 WO
2004039404 May 2004 WO
WO 2004043916 May 2004 WO
WO 2004096795 Nov 2004 WO
WO 2005014555 Feb 2005 WO
WO 2005032493 Apr 2005 WO
WO 2005058311 Jun 2005 WO
WO 2005097767 Oct 2005 WO
WO 2005111031 Nov 2005 WO
2005121100 Dec 2005 WO
2006009655 Jan 2006 WO
2006023750 Mar 2006 WO
2006029850 Mar 2006 WO
WO 2006041404 Apr 2006 WO
WO 2006041405 Apr 2006 WO
WO 2006065204 Jun 2006 WO
WO 2006065277 Jun 2006 WO
2006076284 Jul 2006 WO
WO 2006099379 Sep 2006 WO
WO 2006138192 Dec 2006 WO
WO 2006138217 Dec 2006 WO
WO 2006138265 Dec 2006 WO
WO 2006138304 Dec 2006 WO
2007002220 Jan 2007 WO
WO 2007005366 Jan 2007 WO
WO 2007005404 Jan 2007 WO
WO 2007016012 Feb 2007 WO
WO 2007038271 Apr 2007 WO
WO 2007049532 May 2007 WO
WO 2007058580 May 2007 WO
WO 2007058582 May 2007 WO
WO 2007058583 May 2007 WO
WO 2007058601 May 2007 WO
WO 2007058602 May 2007 WO
WO 2007073284 Jun 2007 WO
WO 2007078813 Jul 2007 WO
2007092846 Aug 2007 WO
2007092854 Aug 2007 WO
WO 2007114771 Oct 2007 WO
WO 2007120096 Oct 2007 WO
WO 2007146225 Dec 2007 WO
WO 2008011560 Jan 2008 WO
2008022024 Feb 2008 WO
2008073365 Jun 2008 WO
WO 2008073370 Jun 2008 WO
WO 2008103351 Aug 2008 WO
WO 2008133273 Nov 2008 WO
WO 2008133274 Nov 2008 WO
WO 2009010454 Jan 2009 WO
2009064418 May 2009 WO
WO 2009091016 Jul 2009 WO
2009097278 Aug 2009 WO
2009097401 Aug 2009 WO
2009097578 Aug 2009 WO
2009103626 Aug 2009 WO
2009131974 Oct 2009 WO
2009131975 Oct 2009 WO
WO 2009134617 Nov 2009 WO
WO 2009151098 Dec 2009 WO
2010019392 Feb 2010 WO
2010019393 Feb 2010 WO
WO 2010013302 Feb 2010 WO
WO 2010013794 Feb 2010 WO
WO 2010038686 Apr 2010 WO
WO 2010047372 Apr 2010 WO
2010056194 May 2010 WO
2010056195 May 2010 WO
2010056196 May 2010 WO
WO 2010113848 Oct 2010 WO
2010129864 Nov 2010 WO
WO 2010128058 Nov 2010 WO
WO 2011005738 Jan 2011 WO
WO 2011009897 Jan 2011 WO
WO 2011009898 Jan 2011 WO
WO 2011009943 Jan 2011 WO
WO 2011020806 Feb 2011 WO
WO 2011029803 Mar 2011 WO
WO 2011044181 Apr 2011 WO
WO 2011044184 Apr 2011 WO
WO 2011044185 Apr 2011 WO
WO 2011044187 Apr 2011 WO
2011060207 May 2011 WO
WO 2011058763 May 2011 WO
WO 2011069934 Jun 2011 WO
WO 2011070029 Jun 2011 WO
WO 2011070781 Jun 2011 WO
WO 2011071057 Jun 2011 WO
WO 2011071109 Jun 2011 WO
WO 2011071135 Jun 2011 WO
WO 2011077726 Jun 2011 WO
WO 2011080176 Jul 2011 WO
2011138293 Nov 2011 WO
2011154431 Dec 2011 WO
WO 2011154374 Dec 2011 WO
2012006953 Jan 2012 WO
WO 2012000933 Jan 2012 WO
WO 2012019966 Feb 2012 WO
2012038438 Mar 2012 WO
2012057247 May 2012 WO
2012057248 May 2012 WO
2012085038 Jun 2012 WO
2012093148 Jul 2012 WO
2012098064 Jul 2012 WO
2012098213 Jul 2012 WO
2012098461 Jul 2012 WO
WO 2012095469 Jul 2012 WO
WO 2012095521 Jul 2012 WO
2012104263 Aug 2012 WO
2012107371 Aug 2012 WO
2012110440 Aug 2012 WO
2012110441 Aug 2012 WO
2012110459 Aug 2012 WO
2012117027 Sep 2012 WO
2012120023 Sep 2012 WO
2012126791 Sep 2012 WO
WO 2012119883 Sep 2012 WO
2012136603 Oct 2012 WO
2012139993 Oct 2012 WO
2012147762 Nov 2012 WO
2012147763 Nov 2012 WO
2012156284 Nov 2012 WO
2012162330 Nov 2012 WO
2012162334 Nov 2012 WO
2012163790 Dec 2012 WO
2012168164 Dec 2012 WO
2012168175 Dec 2012 WO
Non-Patent Literature Citations (93)
Entry
Cheong et al., “Pharmacophore elucidation for a new series of 2-aryl-pyrazolo-triazolo-pyrimidines as potent human A3 adenosine receptor antagonists,” Bioorganic & Medicinal Chemistry Letters, 2011, vol. 21, pp. 2898-2905.
Xu et al., “Copper-catalyzed cascade synthesis of benzimidazoquinazoline derivatives under mild condition,” Chemical Communications, 2011, vol. 47, pp. 5596-5598.
Kishore et al., “QSAR of adenosine receptor antagonists: exploring physicochemical requirements for binding of pyrazolo[4,3-e]-1,2,4-triazolo[1,5-c]pyrimidine derivatives with human adenosine A3 receptor subtype,” Bioorganic & Medicinal Chemistry Letters, 2011, vol. 21, No. 2, pp. 818-823.
Dolzhenko et al., “8-methyl-2-[4-(trifluoromethyl)phenyl]-8H-pyrazolo[4,3-e][1,2,4]triazolo[1,5-c]-pyrimidin-5-amine methanol disolvate,” Acta Crystallographica, Section E: Structure Reports Online, E66(7), 12 pages total.
Beaton et al., “3,4-dihydro-1-isoquinolinamines: a novel class of nitric oxide synthase inhibitors with a range of isoform selectivity and potency,” Bioorganic & Medicinal Chemistry Letters, 2001, vol. 11, pp. 1023-1026.
Beaton et al., “The synthesis of 1-aminodihydroisoquinolines by an imine addition cyclisation reaction,” 1998, Tetrahedron Letters, vol. 39, pp. 1227-1230.
Tian et al., “Radiosynthesis of 8-Fluoro-3-(4-[18F]Fluorophenyl)-3,4-Dihydro-1-Isoquinolinamine ([18F]FFDI), a potential PET radiotracer for the inducible nitric oxide synthase,” 2008, Current Radiopharmaceuticals, vol. 1, pp. 49-53.
Weinhardt et al. “Synthesis and antidepressant profiles of phenyl-substituted 2-amino- and 2-((alkoxycarbonyl)amino)11,4,5,6-tetrahydropyrimidines,” Journal of Medicinal Chemistry, vol. 28, No. 6, 1985, pp. 694-698.
Meschino et al., “2-Amino-5,6-dihydro-1,3-oxazines. The reduction of carboxylic esters with sodium borohydride,” J. Org. Chem., vol. 28, 1963, pp. 3129-3134.
Poos et al., “2-amino-5-aryl-2-oxazolines. Potent new anorectic agent,” J. Med. Chem., vol. 6, 1963, pp. 266-272.
Sandin et al., “A fast and parallel route to cyclic isothioureas and guanidines with use of microwave-assisted chemistry,” J. Org. Chem., vol. 69, 2004, pp. 1571-1580.
Weinhardt et al., “Synthesis and central nervous system propreties of 2-[(Alkoxycarbonyl)amino]-4(5)-phenyl-2-imidazolines,” Journal of Medicinal Chemistry, vol. 27, No. 5, 1984, pp. 616-627.
Woodgate et al., “A new synthesis of 2-amino-2-thiazolines,” Heterocycles, vol. 7, No. 1, 1977, pp. 109-112.
Co-pending U.S. Appl. No. 13/881,112, entitled Fused Aminodihydropyrimidine Derivative, filed Apr. 23, 2013.
Co-pending U.S. Appl. No. 13/881,250, entitled Naphthyridine Derivative, filed Apr. 24, 2013.
Portnyagin et al.: Russian Journal of Organic Chemistry, Consultants Bureau, US. vol. 10, Jan. 1, 1974, pp. 95-98, XP009174887.
“Side Reactions in Organic Synthesis: A Guide to Successful Synthesis Design” 2005 Wiley-VCH Verlag GmbH & Co. KGaA, Wienheim, Chapter 1.
English translation of Schubert et al., “Neue synthesen von 2-Amino-5,6-dihydro-4H-1,3-thiazinen,” Archiv der Pharmazie, 1968, vol. 301, No. 10, pp. 750-762.
Edwards, et al., “Application of Fragment-Based Lead Generation to the Discovery of Novel, Cyclic Amidine β-Secretase Inhibitors with Nanomolar Potency, Cellular Activity, and High Ligand Efficiency”, Journal of Medicinal Chemistry, vol. 50, No. 24, 2007, pp. 5912-5925.
Kuo, et al., “A Synthesis of Estrone via Novel Intermediates. Mechanism of the Coupling Reaction of a Vinyl Carbinol with a β Diketone”, The Journal of Organic Chemistry, vol. 33, No. 8, Aug. 1968, pp. 3126-3132.
Cohen, et al., “Synthesis of 2-Amino-5,6-dihydro-4,H-1,3-thiazines and Related Compounds by Acid Catalyzed Cyclizationof Allylic Isothiuronium Salts”, Journal of Heterocyclic Chemistry, vol. 14, 1977, pp. 717-723.
Hünig, et al., “Azofarbstoffe Durch Oxydative Kupplung, XVIII. Synthese von-3-substituierten Thiazolon-(2)-hydrazonen und Thiazolon-(2)-benzolsulfonylhydrazonen”, European Journal of Organic Chemistry, vol. 647, No. 1, May 1961, pp. 66-76.
Schaumann, et al., “Cycloadditionsreaktionen von Heterokumulenen, XXIII. Stickstoffhaltige Fünfring-Heterocyclen aus Carbodiimiden oder Keteniminen mit 3-Dimethylamino-2H-azirinen”, Liebigs Ann. Chem., 1981, pp. 290-305.
Cambie, et al., “vic-Iodothiocyanates and Iodoisothiocyanates. Part 2. New Syntheses of Thiazolidin-2-ones and 2-Amino-2-thiazolines”, Journal of the Chemical Society, Perkin Transactions I, No. 3, 1979, pp. 765-770.
Fernández, et al., “Syntheses of β-iodourea derivatives of carbohydrates and glycosylamino-oxazolines”, Carbohydrate Research, vol. 216, 1991, pp. 21-32.
Fernández, et al., “Syntheses and Spectral Properties of β-Iodoureas and 2-Amino-4,4-diphenyl-2-oxazolines”, Journal of Heterocyclic Chemistry, vol. 28, 1991, pp. 777-780.
Liebscher, et al., “2-Arylimino-3-Thiazolines—Formation of Unusual Tautomers of 2-Arylamino-Thiazolines—a Revision”, Tetrahedron Letters, vol. 26, No. 35, 1985, pp. 4179-4180.
Koriyama et al., “Reversal of diastereofacial selectivity in the nucleophilic addition reaction to chiral N-sulfinimine and application to the synthesis of indrizidine 223AB,” Tetrahedron, vol. 58, 2002, pp. 9621-9628.
Fujisawa et al., “Switchover of diastereofacial selectivity in the condensation reaction of optically active N-sulfinimine with ester enolate,” Tetrahedron Letters, vol. 37, No. 22, 1996, pp. 3881-3884.
Vilaivan et al., “Recent Advances in Catalytic Asymmetric Addition to Imines and Related C=N Systems,” Current Organic Chemistry, vol. 9, 2005, pp. 1315-1392.
Hua et al., “N-Alkylidenesulfinamides,” Sulfur Reports, vol. 21, 1999, pp. 211-239.
Savoca et al., “1,5-Diazabicyclo[4.3.0]non-5-ene1,” Encyclopedia of Reagents for Organic Synthesis, 2006, 10 pages total.
Creeke et al., “Synthesis and elaboration of heterocycles via iodocyclisation of unsaturated thioureas,” Tetrahedron Letters, 1989, vol. 30, No. 33, pp. 4435-4438.
Mellor et al., A general route to spirocycles by radical additions to exocyclic unsaturated sulphides and related compounds, Tetrahedron Letters, 1991, vol. 32, No. 48, pp. 7111-7114.
Murai et al., “Iodo-cyclization of N-homoallyl thioamides leading to 2,4-diaryl-5,6-dihydro-4H-1,3-thiazines,” Chemistry Letters, 2004, vol. 33, No. 5, pp. 508-509.
Singh et al., “Synthesis of heterocyclic compounds via enamines. Part 8.† Acid-catalysed transformations in a 4,4,6-trimethyl-1-4-dihydropyrimidine-2(3H)-thione derivatives and related compounds,” J. Chem. Soc., Perkin Trans. 1, 1980, pp. 1013-1018.
Kondrat'eva et al., “Noncyclic dimer of 4-methyl-2-dimethlaminooxazole,” Izvestiya Akademii Nauk SSSR, Seriya Khimicheskaya, No. 7, Jul. 1977, pp. 1680-1682 (with English translation).
International Search Report for PCT/JP2006/321015.
Database Registry [Online], Chemical Abstracts Service, Columbus, Ohio, US; May 29, 2007, “2H-Indol-2-ome,4-(-2-amino-5,6-dihydro-4-methyl-4H-1,3-thiazin-4-yl)-1,3-dihydro-”, XP002646872, Database accession No. 935998-84-8.
Schubert et al., “Neue syntheses von 2-Amino-5,6-dihydro-4H-1,3-thiazinen,” Archiv der Pharmazie, 1968, vol. 301, No. 10, pp. 750-762.
Co-pending U.S. Appl. No. 13/260,103, entitled Isothiourea Derivatives or Isourea Derivatives Having BACE1 Inhibitory Activity, filed Sep. 23, 2011.
Huang et al., “Pharmacophore model construction of β-secretase inhibitors,” 2008, Acta Chimica Sinica, vol. 66, No. 16, pp. 1889-1897 (English language abstract provided).
Clark, et al., “Antitumor Imidazotetrazines. 32.1 Synthesis of Novel Imidazotetrazinones and Related Bicyclic Heterocycles to Probe the Mode of Action of the Antitumor Drug Temozolomide”, J. Med. Chem., vol. 38, 1995, pp. 1493-1504.
Congreve, et al., “Application of Fragment Screening by X-ray Crystallography to the Discovery of Aminopyridines as Inhibitors of β-Secretase”, J. Med. Chem., vol. 50, 2007, pp. 1124-1132.
Huang, et al., “Progress in the Development of Nonpeptidomimetic BACE 1 Inhibitors for Alzheimer's Disease”, Current Medicinal Chemistry, vol. 16, 2009, pp. 1806-1820.
Goodyer et al., “Synthesis of N-benzyl- and N-phenyl-2-amino-4,5-dihydrothiazoles and thioureas and evaluation as modulators of the isoforms of nitric oxide synthase,” Bioorganic & Medicinal Chemistry, 2003, vol. 11, pp. 4189-4206.
Siddiqui et al., “Some extensions of Von Bruan (BrCN) reaction on organic bases,” Proc. Pakistan Acad. Sci., 1988, vol. 25, No. 3, pp. 231-240.
Ozawa et al., Pharmacological actions of 2-Aminoethylisothiuronium (AET) derivatives. I1), Yakugaku Zasshi, 1968, vol. 88, No. 2, pp. 156-162 (English language abstract provided).
Curtis et al., The byozynsethis of Phenols, Part VIII. The synthesis of (2-carboxy-3,5-dihydroxyphynyl)propan-2-one (C-acetyl-o-orsellinic acid). Journal of the Chemical Society, 1964, pp. 5382-5385.
Burton et al., “Addition reactions of quinones. Part I. The reaction of cysteine and thiourea and its derivatives with some quinones,” Journal of the Chemical Society, 1952, pp. 2193-2196.
Matsui, “Yomo bochuzai no kenkyu (the 6th report) Kagaku kozo to yomo shokugai boshi koka tono kankei (III),” Journal of Synthetic Organic Chemistry, Japan, 1950, vol. 8, No. 10, pp. Ho61-Ho65 (and International Search Report issued in PCT/JP2010/055528, which corresponds to co-pending U.S. Appl. No. 13/260,103).
Desai et al., “The condensation of thiocarbamides with monochloroacetic acide and the conversion of arylformamidinethiolacetic acids into pseudothiohydantoin derivatives,” Recuil des Travaux Chimiques des Pays-Bas et de la Belgique, 1935, pp. 118-121.
Cole et al., “Acylguanidines as small-molecule beta-secretase inhibitors,” J. Med. Chem., 2006, pp. 6158-6161.
Co-pending U.S. Appl. No. 13/508,899, entitled Aminothiazine or Aminooxazine Derivative Having Amino Linker, filed May 9, 2012.
Co-pending U.S. Appl. No. 13/513,839, entitled Oxazine Derivatives , filed Jun. 4, 2012.
Co-pending U.S. Appl. No. 13/514,516, entitled Substituted Aminothiazine Derivative, filed Jun. 7, 2012.
Co-pending U.S. Appl. No. 13/518,285, entitled 4-Amino-1,3-Thiazine or Oxazine Derivative, filed Jun. 21, 2012.
Co-pending U.S. Appl. No. 13/514,907, entitled Fused Heterocyclic Compound Having Amino Group, filed Jun. 8, 2012.
Bol'but et al., “Heterocyclizations of Functionalized Heterocumulenes with C,N- and C,O-Dinucleophiles: III.* Cyclization of N-(1-Aryl-1-chloro-2,2,2-trifluoroethyl)-N′-arylcarbodiimides with 3-Substituted 1-Phenylpyrazol-5-ones” Russian Journal of Organic Chemistry, 2003, vol. 39, No. 12, pp. 1789-1791.
Trepanier et al., “Synthesis and screening for antidepressant activity of some aminoindanooxazolines, aminoindanooxazines, and aminoacenaphthoxazolines,” Journal of Medicinal Chemistry, 1970, vol. 13, No. 4, pp. 729-733.
Sayed et al., “α-Enones in heterocyclic synthesis of indazole, thiazine, chromene and quinolone derivatives with their antimicrobial activities” Journal of Chemical Research, 2009, vol. 12, pp. 726-728.
Database Caplus [Online] Chemical Abstracts Service, Columbus, Ohio, US; Xu, Yungen et al: “Preparation of benzimidazolyl and benzothiazolyl isothiourea derivatives as nitric oxide synthase inhibitors”, XP002679913, retrieved from STN Database accession No. 2005:46620 *abstract* Accessed Jul. 13, 2012.
Bol'but et al., “Synthesis of 4-imino-2-trifluoromethyl-3,4-dihydro-2h-Benzo-[1,3]Thiazines” Chemistry of Heterocyclic Compounds, 2001, vol. 37, No. 4, pp. 522-523.
Vovk et al,, “Intramolecular thermal cyclization of N-(1-Aryl-1-aryloxy-2,2,2-trifluoroethyl)-N′-arylcarbodiimides” Russian Journal of Organic Chemistry, 2000, vol. 36, No. 12, pp. 1739-1742.
Vovk et al., “Regioselective cyclization of 1-chloroalkylcarbodiimides with 1,1- and 1,2-bifunctional nucleophilic reagents” Russian Journal of Organic Chemistry, 1997, vol. 33, No. 1, pp. 96-102.
Potts et al., “N-Acyl-β-enamino Ketones: Versatile Heterocyclic Synthons” J. Org. Chem., 1983, 48, pp. 623-625.
Rivkin et al., “Piperazinyl pyrimidine derivatives as potent γ-secretase modulators” Bioorganic & Medicinal Chemistry Letters, 2010, 20, pp. 1269-1271.
Rivkin et al., “Purine derivatives as potent γ-secretase modulators” Bioorganic & Medicinal Chemistry Letters, 2010, 20, pp. 2279-2282.
STN a the Web, RN 79005-45-1, 1964.
Zhu et al. “Two Novel Diasteroselectric Three-Component Reactions of Alkenes or 3, 4,-Dihydro-(2H)-pyran with Urea/Thiourea-Aldehyde Mixtures: [4+2] Cycloadditional vs Biginelli-Type Reaction”. Organic Letters, vol. 8, No. 12, pp. 2599-2602 (2006).
Calabrese et al. “NO Synthase and NO-Dependent Signal Pathways in Brain Aging and Neurodegenerative Disorders: The Role of Oxidant/Antioxidant Balance”. Neurochemical Balance, vol. 25, No. 9/10, pp. 1315-1341 (2000).
Kavya et al. “Nitric oxide synthase regulation and diversity: Implications in Parkinson's Disease”. Nitric Oxide: Biology and Chemistry, vol. 15, No. 4, pp. 280-294 (2006).
Chiou et al. “Review: Effects of Nitric Oxide on Eye Diseases and Their Treatment”. Journal of Ocular Pharmacology and Therapeutics, vol. 17, No. 2, pp. 189-198 (2001).
Ishii et al. “Subacute NO generation induced by Alzheimer's β-amyloid in the living brain: reversal by inhibition of the inducible NO synthase”. The Federation of American Societies for Experimental Biology Journal, vol. 14, pp. 1485-1489 (2000).
Pak et al. “Morphine via nitric oxide modulates β-amyloid metabolism: a novel protective mechanism for Alzheimer's disease”. Medical Science Monitor, vol. 11, No. 10, pp. BR357-BR366 (2005).
Vippagunta et al. “Crystalline Solids”. Advanced Drug Delivery Reviews, vol. 48, 2001, pp. 3-26.
Gavezzotti. “Are Crystal Structures Predictable”. Accounts of Chemical Research, vol. 27, 1994, pp. 309-314.
Kiselyov et al., “Design and chemical synthesis of [1,2,4]triazol[1,5-c]pyrimidin-5-yl amines, a novel class of VEGFR-2 kinase inhibitors,” 2009, Tetrahedron Letters, vol. 50, pp. 3809-3812.
Mulcahy et al., “A stereoselective synthesis of (+)-Gonyautoxin 3,” Journal of the American Chemical Society, 2008, vol. 130, pp. 12630-12631.
“Diphenyl Cyanocarbonimidate,” e-EROS Encyclopedia of Reagents for Organic Synthesis, 2001, 2 pages total.
Shafik et al., “Synthesis of novel 2-[2-(substituted amino)phenethyl]-1H-benzimidazoles; 3,4-dihydro and 1,2,3,4-tetrahydropyrimido[1,6-α]-benzimidazoles as potential antiulcer agents,” 2004, Pharmazie, vol. 59, No. 12, pp. 899-905.
Pohl et al., “Synthesis of partially saturated condensed triazoles by reaction of ω-Aminoalkyl-1,2,4-triazoles with electrophiles,” Journal fuer Praktische Chemie Chemiker-Zeitung, 1992, vol. 334, pp. 630-636.
Buschauer et al., “Isohistamine und Homologe als Bausteine von H2-Antagonisten,” Arzneimittel-Forschung, 1985, vol. 35, pp. 1025-1029 (English language abstract provided).
Buschauer et al., “7,8-Dihydroimidazo[1,2-c]pyrimidin-5(6H)-one, -5(6H)-thione and -5(6H)-ylidencyanamide,” Chemische Berichte, 1984, vol. 117, pp. 2597-2614.
Borchers et al., “H2-Antihystaminika, 19. Mitt.1) Syntheses und H2-antihistaminische Wirkung Nα-substituierter Histamine,” Archiv der Pharmazie (Weinheim, Germany), 1984, vol. 317, pp. 455-459.
Co-pending U.S. Appl. No. 13/952,073, entitled Sulfur-Containing Heterocyclic Derivative Having Beta Secretase Inhibitory Activity, filed Jul. 26, 2013.
Wermuth; Practice of Medicinal Chemistry, Third Ed., 2008, Elsevier, chapter 15.
Emilio Testa et al.: “Auf das Zentralnervensystem wirkende Substanzen, XXXVI. Weitere Untersuchungen über die 2-substituierten Azetidine; Justus Liebigs Annalen Der Chemie”, vol. 673, No. 1., May 4, 1964, pp. 60-70 XP055091964.
Portnyagin et al.: Russian Journal of Organic Chemistry, Consultants Bureau, US, vol. 10, Jan. 1, 1974, pp. 95-98, XP009174887.
Database Caplus [Online]; Chemical Abstracts Service, Columbus, OH, US; 2006, Bathich, Yaser: “Synthesis of Branched Amino Polyoly and Aminohydroxy Acids: Stereoselective Additiom of C-Nucleophiles ti Isoxazolines and Isoxazolinium Salts and Assignment of Configurations”, XP002717806.
Database Registry [Online]; Chemical Abstracts Service, Columbus, OH, US; 3-Pyridinepropanol, .beta., .gamma.-diamino-6-fluoro-.gamma.-(4-fluorophenyl)-, (.beta-R,.gamma.S)-, Apr. 29, 2004, XP002717807.
Church J et al.: “Anticonvulsant actions of phencyclidine receptor ligands: Correlation with N-Methylaspartate Antagonism in vivo”; General Phamacology, Pergamon Press, Oxford, GB, vol. 21, No. 2, Jan. 1, 1990, pp. 165-170, XP023834032.
Bathich, “Synthesis of Branched Amino Polyols and Amino Hydroxy Acids: Stereoselective Addition of C-Nucleophiles to Isoxazolines and Isoxazolinium Salts and Assignment of Configurations”, pp. 148 (2006).
Related Publications (1)
Number Date Country
20140073815 A1 Mar 2014 US
Divisions (1)
Number Date Country
Parent 12596796 US
Child 13417786 US
Continuations (1)
Number Date Country
Parent 13417786 Mar 2012 US
Child 13887745 US