JPWO2005030773A1 - New pyrazolopyrimidine derivatives - Google Patents

New pyrazolopyrimidine derivatives Download PDF

Info

Publication number
JPWO2005030773A1
JPWO2005030773A1 JP2005514269A JP2005514269A JPWO2005030773A1 JP WO2005030773 A1 JPWO2005030773 A1 JP WO2005030773A1 JP 2005514269 A JP2005514269 A JP 2005514269A JP 2005514269 A JP2005514269 A JP 2005514269A JP WO2005030773 A1 JPWO2005030773 A1 JP WO2005030773A1
Authority
JP
Japan
Prior art keywords
group
compound
acceptable salt
pharmaceutically acceptable
added
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
JP2005514269A
Other languages
Japanese (ja)
Inventor
克憲 坪井
克憲 坪井
正志 中塚
正志 中塚
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Sumitomo Pharma Co Ltd
Original Assignee
Sumitomo Dainippon Pharma Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Sumitomo Dainippon Pharma Co Ltd filed Critical Sumitomo Dainippon Pharma Co Ltd
Publication of JPWO2005030773A1 publication Critical patent/JPWO2005030773A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system

Abstract

一般式(1)(式中、R1は水酸基またはカルボキシル基等を表し、mは1〜3の整数を表し、R2はアルキル基またはアルコキシ基等を表し、nは0〜4の整数を表し、環Aは5〜8員の飽和もしくは不飽和含窒素複素環を表し、R3は、ハロアルキル基等を表し、R4およびR5は、独立して、水素原子、ハロゲン原子又はアルキル基等を表し、Xはメチンまたは窒素原子等を表す。)で表される化合物、またはその薬学上許容される塩は、抗掻痒作用等を有し、アレルギー疾患、皮膚疾患等の治療剤もしくは予防剤として有用である。General formula (1) (In formula, R1 represents a hydroxyl group or a carboxyl group, m represents an integer of 1-3, R2 represents an alkyl group, an alkoxy group, etc., n represents an integer of 0-4, Ring A represents a 5- to 8-membered saturated or unsaturated nitrogen-containing heterocycle, R3 represents a haloalkyl group or the like, R4 and R5 independently represent a hydrogen atom, a halogen atom, an alkyl group, or the like, and X Represents a methine or nitrogen atom, etc.), or a pharmaceutically acceptable salt thereof, has an antipruritic action and is useful as a therapeutic or preventive agent for allergic diseases, skin diseases, etc. .

Description

本発明は、PAR2阻害剤、すなわちPAR2が関与する、アレルギー疾患、呼吸器疾患、心血管系疾患、神経系疾患、炎症性疾患、神経性炎症性疾患、皮膚疾患等の、治療剤、予防剤、または進行防止剤として有効な、新規なピラゾロピリミジン誘導体に関する。  The present invention relates to a PAR2 inhibitor, that is, a therapeutic agent or a preventive agent for allergic diseases, respiratory diseases, cardiovascular diseases, nervous system diseases, inflammatory diseases, neuroinflammatory diseases, skin diseases and the like that involve PAR2. Or a novel pyrazolopyrimidine derivative that is effective as a progression inhibitor.

PAR2(proteinase−activated receptor2)は、Gタンパク共役7回膜貫通型受容体の一種であり、セリンプロテアーゼの細胞作用を媒介する受容体である。前記セリンプロテアーゼはPAR2分子の細胞外N末側のペプチド鎖を特定部位で切断することにより、5−6アミノ酸残基からなる受容体活性化配列を有する新しいN末端を露出させる。新たに露出したN末端が、鎖状リガンドとしてPAR2自身の活性部位に結合することにより、PAR2の活性化が起こる。PAR2は具体的には、トリプシン、トリプターゼ、または、血液凝固第VIIaもしくはXa因子によって活性化されることが明らかとなっている。また、受容体活性化配列に基づいて合成した5〜6個のアミノ酸から成る合成ペプチドにより、活性化されることが知られている(Dery,O.ら著、Am.J.Physiol.,274,C1429−52(1998)又はMacfarlane,S.R.ら著、Pharmacol Rev.,53,245−82(2001)を参照)。
PAR2は、ホスホリパーゼCの活性化に伴う細胞内カルシウム濃度の上昇とイノシトール3リン酸の産生、p38 MAPキナーゼの活性化またはc−Jun N−terminalキナーゼの活性化等の細胞内シグナルを活性化することが知られている。また、PAR2は、消化液の分泌を促進することや、神経性炎症もしくはアレルギー性疾患などの種々の疾患において増悪因子であることが知られている。
更に、受容体活性化配列に基づく合成ペプチド誘導体がPAR2アゴニストとして報告されているが、該合成ペプチド誘導体を用いた検討等から、PAR2アンタゴニストが腸疾患治療薬、皮膚色素沈着防止薬、アレルギー性疾患または癌転移抑制剤として有用であることが示唆されている。
一方、ピラゾロピリミジン誘導体としては、ケミカルアブストラクトにおいて、レジストリー番号:334500−35−5、304686−65−5、312634−72−3、312918−87−9、312934−98−6、313987−33−6、または313986−65−1等の化合物が公知であった。また、これらの化合物は、ナトリウムチャンネルの一種であるPN3のブロッカー作用を有することが知られている(特国際公開第03/037900号パンフレットを参照)。しかしながら、本発明優先日において、これらの化合物とPAR2阻害活性との関係は全く知られていなかった。
PAR2 (proteinase-activated receptor 2) is a kind of G protein-coupled seven-transmembrane receptor, and is a receptor that mediates the cellular action of serine protease. The serine protease cleaves the peptide chain on the extracellular N-terminal side of the PAR2 molecule at a specific site, thereby exposing a new N-terminus having a receptor activation sequence consisting of 5-6 amino acid residues. Activation of PAR2 occurs when the newly exposed N-terminus binds to PAR2's own active site as a chain ligand. PAR2 has been shown to be specifically activated by trypsin, tryptase, or blood coagulation factor VIIa or factor Xa. It is also known to be activated by a synthetic peptide consisting of 5 to 6 amino acids synthesized based on the receptor activation sequence (Dery, O. et al., Am. J. Physiol., 274). C1429-52 (1998) or Macfarlane, SR et al., Pharmacol Rev., 53, 245-82 (2001)).
PAR2 activates intracellular signals such as the increase of intracellular calcium concentration and inositol triphosphate production, activation of p38 MAP kinase or c-Jun N-terminal kinase associated with activation of phospholipase C. It is known. Moreover, PAR2 is known to promote secretion of digestive juice and to be an exacerbating factor in various diseases such as neurogenic inflammation or allergic diseases.
Furthermore, synthetic peptide derivatives based on receptor activation sequences have been reported as PAR2 agonists. From studies using such synthetic peptide derivatives, PAR2 antagonists have been shown to be therapeutic agents for intestinal diseases, skin pigmentation inhibitors, allergic diseases. It has also been suggested to be useful as a cancer metastasis inhibitor.
On the other hand, as a pyrazolopyrimidine derivative, in the chemical abstract, registry numbers: 334500-35-5, 304686-65-5, 312634-72-3, 312918-87-9, 319344-98-6, 3139987-33 Compounds such as 6, or 313986-65-1 were known. Further, these compounds are known to have a blocker action of PN3, which is a kind of sodium channel (see Japanese Patent Publication No. 03/037900 pamphlet). However, on the priority date of the present invention, the relationship between these compounds and PAR2 inhibitory activity was not known at all.

本発明が解決しようとする課題は、PAR2阻害活性を有する化合物、すなわちPAR2が関与する疾患の治療剤もしくは予防剤として有用な化合物を提供することにある。
本発明者らは、鋭意検討を重ねた結果、ピラゾロピリミジン骨格またはピラゾロピリジン骨格を有する化合物(以下本発明の化合物と称する場合がある。)が、優れたPAR2阻害活性を示すことを見出した。これらの化合物は、PAR2の機能亢進が原因となっている疾患、具体的には炎症性疾患、アレルギー性疾患、呼吸器疾患、心血管系疾患、神経系疾患、炎症性疾患、神経性炎症性疾患、皮膚疾患等の治療もしくは予防剤として有用である。また、疼痛や掻痒を伴う各種疾患の治療剤として有用である。また、本発明の化合物は、プロスタグランジンE2(PGE2)産生阻害活性を示し、慢性関節リウマチ等のPGE2の機能亢進が原因となっている疾患の治療剤もしくは予防剤としても有用である。
本発明は以上の知見に基づき、完成するに至ったものである。
すなわち、本発明は、
[1] 一般式(1)

Figure 2005030773
〔式中、Rは水酸基、カルボキシル基、メルカプト基、アミノ基、置換アミノ基、または水酸基、カルボキシル基、メルカプト基、アミノ基もしくは置換アミノ基で置換されたアルキル基を表し、
mは1〜3の整数を表し、mが2以上の整数を表す場合Rは同一もしくは異なっていてもよく、
はハロゲン原子、アルキル基、アルケニル基、アルキニル基、シクロアルキル基、アルコキシ基、又はアルキルチオ基を表し、
nは0〜4の整数を表し、nが2以上の整数を表す場合Rは同一もしくは異なっていてもよく、
環Aは5〜8員の飽和もしくは不飽和含窒素複素環を表し、
は、水素原子、アルキル基、ハロアルキル基、または置換もしくは無置換のアリール基を表し、
およびRは、独立して、水素原子、ハロゲン原子、アルキル基、アルケニル基、アルキニル基、シクロアルキル基、アルコキシ基、アルキルチオ基またはハロアルキル基を表し、
XはCR(Rは水素原子またはアルキル基を表す)または窒素原子を表す。〕
で表される化合物、またはその薬学上許容される塩;
[2] 式(1)において、Rがハロアルキル基である、[1]に記載の化合物、またはその薬学上許容される塩;
[3] ハロアルキル基が、トリフルオロメチル基である、[2]に記載の化合物、またはその薬学上許容される塩;
[4] [1]〜[3]のいずれかに記載の化合物又はその薬学上許容される塩を有効成分として含有する、PAR2阻害剤;
[5] [1]〜[3]のいずれかに記載の化合物又はその薬学上許容される塩を有効成分として含有する、医薬組成物;
[6] [1]〜[3]のいずれかに記載の化合物又はその薬学上許容される塩を有効成分として含有する、抗掻痒剤;
[7] [1]〜[3]のいずれかに記載の化合物又はその薬学上許容される塩を有効成分として含有する、鎮痛剤;
[8] [1]〜「3]のいずれかに記載の化合物又はその薬学上許容される塩を有効成分として含有する、PGE2産生抑制剤;及び
[9] [1]〜[3]のいずれかに記載の化合物又はその薬学上許容される塩を有効成分として含有する、抗炎症剤;
に関する。
本発明により、PAR2阻害活性を有し、炎症性疾患、アレルギー性疾患、呼吸器疾患、心血管系疾患、神経系疾患、炎症性疾患、神経性炎症性疾患又は疼痛もしくは掻痒を伴う疾患等の、種々の疾患の治療剤として有用なピラゾロピリジン化合物及びピラゾロピリミジン化合物を提供することが可能になった。The problem to be solved by the present invention is to provide a compound having PAR2 inhibitory activity, that is, a compound useful as a therapeutic or prophylactic agent for diseases involving PAR2.
As a result of intensive studies, the present inventors have found that a compound having a pyrazolopyrimidine skeleton or a pyrazolopyridine skeleton (hereinafter sometimes referred to as the compound of the present invention) exhibits excellent PAR2 inhibitory activity. It was. These compounds are diseases caused by hyperfunction of PAR2, specifically inflammatory diseases, allergic diseases, respiratory diseases, cardiovascular diseases, nervous system diseases, inflammatory diseases, neurogenic inflammatory It is useful as an agent for treating or preventing diseases and skin diseases. It is also useful as a therapeutic agent for various diseases associated with pain and pruritus. In addition, the compound of the present invention exhibits prostaglandin E2 (PGE2) production inhibitory activity, and is also useful as a therapeutic or prophylactic agent for diseases caused by hyperfunction of PGE2, such as rheumatoid arthritis.
The present invention has been completed based on the above findings.
That is, the present invention
[1] General formula (1)
Figure 2005030773
[Wherein R 1 represents a hydroxyl group, a carboxyl group, a mercapto group, an amino group, a substituted amino group, or an alkyl group substituted with a hydroxyl group, a carboxyl group, a mercapto group, an amino group, or a substituted amino group;
m represents an integer of 1 to 3, and when m represents an integer of 2 or more, R 1 may be the same or different;
R 2 represents a halogen atom, an alkyl group, an alkenyl group, an alkynyl group, a cycloalkyl group, an alkoxy group, or an alkylthio group,
n represents an integer of 0 to 4, and when n represents an integer of 2 or more, R 2 may be the same or different,
Ring A represents a 5- to 8-membered saturated or unsaturated nitrogen-containing heterocyclic ring,
R 3 represents a hydrogen atom, an alkyl group, a haloalkyl group, or a substituted or unsubstituted aryl group,
R 4 and R 5 independently represent a hydrogen atom, a halogen atom, an alkyl group, an alkenyl group, an alkynyl group, a cycloalkyl group, an alkoxy group, an alkylthio group or a haloalkyl group,
X represents CR 6 (R 6 represents a hydrogen atom or an alkyl group) or a nitrogen atom. ]
Or a pharmaceutically acceptable salt thereof;
[2] The compound according to [1] or a pharmaceutically acceptable salt thereof, wherein in formula (1), R 3 is a haloalkyl group;
[3] The compound according to [2], wherein the haloalkyl group is a trifluoromethyl group, or a pharmaceutically acceptable salt thereof;
[4] A PAR2 inhibitor containing the compound according to any one of [1] to [3] or a pharmaceutically acceptable salt thereof as an active ingredient;
[5] A pharmaceutical composition comprising the compound according to any one of [1] to [3] or a pharmaceutically acceptable salt thereof as an active ingredient;
[6] An anti-pruritic agent containing the compound according to any one of [1] to [3] or a pharmaceutically acceptable salt thereof as an active ingredient;
[7] An analgesic comprising the compound according to any one of [1] to [3] or a pharmaceutically acceptable salt thereof as an active ingredient;
[8] A PGE2 production inhibitor comprising the compound according to any one of [1] to [3] or a pharmaceutically acceptable salt thereof as an active ingredient; and any of [9] [1] to [3] An anti-inflammatory agent comprising the compound according to claim 1 or a pharmaceutically acceptable salt thereof as an active ingredient;
About.
According to the present invention, it has PAR2 inhibitory activity, such as inflammatory disease, allergic disease, respiratory disease, cardiovascular disease, nervous system disease, inflammatory disease, neuroinflammatory disease or disease with pain or pruritus, etc. It has become possible to provide pyrazolopyridine compounds and pyrazolopyrimidine compounds that are useful as therapeutic agents for various diseases.

本明細書において、アルキル基としては炭素数1〜4の直鎖もしくは分枝のアルキル基が挙げられる。具体的にはメチル基、エチル基、プロピル基、1−メチルエチル基、ブチル基、1−メチルプロピル基、2−メチルプロピル基又は1,1−ジメチルエチル基等が挙げられる。
本明細書において、アルケニル基としては炭素数1〜4の直鎖もしくは分枝のアルケニル基が挙げられる。具体的にはビニル基、1−プロペニル基、2−プロペニル基、1−メチルビニル基、1−ブテニル基、2−ブテニル基、3−ブテニル基、1−メチル−1−プロペニル基、1−メチル−2−プロペニル基、2−メチル−1−プロペニル基、又は2−メチル−2−プロペニル基等が挙げられる。
本明細書において、アルキニル基としては炭素数1〜4の直鎖もしくは分枝のアルキニル基が挙げられる。具体的にはエチニル基、1−プロピニル基、2−プロピニル基、1−ブチニル基、2−ブチニル基、3−ブチニル基、又は1−メチル−2−プロピニル基等が挙げられる。
本明細書において、シクロアルキル基としては、炭素数3〜6のシクロアルキル基が挙げられる。具体的には、シクロプロピル基、シクロブチル基、シクロペンチル基、又はシクロヘキシル基等が挙げられる。
本明細書において、アルコキシ基としては炭素数1〜4の直鎖もしくは分枝のアルコキシ基が挙げられる。具体的にはメトキシ基、エトキシ基、プロポキシ基、1−メチルエトキシ基、ブトキシ基、1−メチルプロポキシ基、2−メチルプロポキシ基又は1,1−ジメチルエトキシ基等が挙げられる。
本明細書において、アルキルチオ基としては炭素数1〜4の直鎖もしくは分枝のアルキルチオ基が挙げられる。具体的にはメチルチオ基、エチルチオ基、プロピルチオ基、1−メチルエチルチオ基、ブチルチオ基、1−メチルプロピルチオ基、2−メチルプロピルチオ基又は1,1−ジメチルエチルチオ基等が挙げられる。
本明細書において、水酸基、カルボキシル基、メルカプト基、アミノ基もしくは置換アミノ基で置換されたアルキル基においては、前記アルキル基の任意の1または複数の水素原子が置換されていてもよい。
本明細書において、ハロゲン原子とは、フッ素原子、塩素原子、臭素原子、またはヨウ素原子を表し、好ましくは、フッ素原子または塩素原子を表す。
本明細書において、ハロアルキル基としては、同一または異なる、1〜3個のハロゲン原子を含む炭素数1〜2のハロアルキル基が挙げられ、具体的には、トリフルオロメチル基、ジフルオロメチル基、2,2,2−トリフルオロエチル基、または2,2−ジフルオロエチル基等が挙げられる。好ましくは、トリフルオロメチル基が挙げられる。
本明細書において、置換アミノ基としては、1もしくは2の、アルキル基、あるケニル基、アルキニル基、シクロアルキル基から選択される1もしくは2個の置換基で置換されたアミノ基が挙げられる。また、1−ピロリジニル基、1−ピペリジニル基、ピペラジノ基、モルホリノ基、もしくはチオモルホリノ基等の環状のアミノ基も、置換アミノ基の範疇に含まれる。
一般式(1)中の環Aにおける「6〜8員の飽和もしくは不飽和含窒素複素環」としては、1〜3個の窒素原子、0もしくは1個の酸素原子及び0もしくは1個の硫黄原子から選択される1〜3個のヘテロ原子を含む飽和もしくは不飽和含窒素複素環が挙げられ、0〜2個の二重結合を有していてもよい。具体的には、ピペリジン、ピペラジン、モルホリン、チオモルホリン、S−オキソチオモルホリン、S,S−ジオキソチオモルホリン、パーヒドロアゼピン(アゼパン)、2,3,6,7−テトラヒドロアゼピン、1,3−パーヒドロオキサゼピン(1,3−オキサゼパン)、パーヒドロアゾシン(アゾカン)、1,2,3,6,7,8−ヘキサヒドロアゾシン、1,4−パーヒドロオキサゾシン(1,4−オキサゾカン)等を例示することができる。
が水酸基、カルボキシル基、メルカプト基、アミノ基、もしくは置換アミノ基を表す場合、環Aを構成する任意の炭素原子に結合することができる。また、Rが水酸基、カルボキシル基、メルカプト基、アミノ基もしくは置換アミノ基で置換されたアルキル基を表す場合、環Aを構成する任意の炭素原子または窒素原子に結合することができる。
は、好ましくは水酸基、カルボキシル基、ヒドロキシメチル基、1−ヒドロキシエチル基、2−ヒドロキシエチル基、1−ヒドロキシ−1−メチルエチル基、2−ヒドロキシ−1−メチルエチル基、1−ヒドロキシプロピル基、2−ヒドロキシプロピル基、3−ヒドロキシプロピル基、1,2−ジヒドロキシエチル基、2,3−ジヒドロキシプロピル基、カルボキシメチル基、1−カルボキシエチル基、2−カルボキシエチル基、1−カルボキシ−1−メチルエチル基、2−カルボキシ−1−メチルエチル基、1−カルボキシプロピル基、2−カルボキシプロピル基、3−カルボキシプロピル基、1,2−ジカルボキシエチル基、2,3−ジカルボキシプロピル基等を表す。
は、好ましくは炭素数1〜2のアルキル基、または炭素数1〜2のアルコキシ基を表す。
一般式(1)中のRにおけるアリール基としては、フェニル基またはナフチル基が挙げられ、好ましくはフェニル基が挙げられる。
該アリール基、例えばフェニル基もしくはナフチル基が置換されている場合の置換基としては、ハロゲン原子、炭素数1〜4のアルキル基、炭素数1〜2のハロアルキル基、または炭素数1〜4のアルコキシ基が挙げられる。
は好ましくは炭素数1〜3のアルキル基、フェニル基、置換フェニル基、または炭素数1〜2のハロアルキル基を表す。特に好ましい例として、トリフルオロメチル基が挙げられる。
一般式(1)で表される化合物におけるRおよびRとして、好ましくは、同一もしくは異なって、水素原子、フッ素原子、塩素原子、メチル基、エチル基、メトキシ基、エトキシ基、トリフルオロメチル基、2,2,2−トリフルオロエチル基、2,2−ジフルオロエチル基等が挙げられる。
一般式(1)で表される本発明の化合物は、以下の方法で製造することができる。

Figure 2005030773
[式中、m、n、R、R、R、RおよびRは前記と同義である。]
式(1−2)のヒドラゾンは、公知化合物である式(1−1)の化合物とヒドラジン1水和物を、酸性条件下、0〜100℃で反応させることで製造できる。溶媒としては、メタノール、エタノールなどのアルコール系溶媒、クロロホルム、ジクロロメタンなどのハロゲン系溶媒、ジメチルホルムアミド、ジメチルスルフォキシド、アセトン、アセトニトリル、ジオキサンやテトラヒドロフランなどのエーテル系溶媒、水またはこれらの混合溶媒等が用いられる。メタノール、エタノールなどのアルコール系溶媒がより好ましい。酸としては、酢酸、塩酸、硫酸、トリフルオロ酢酸などを用いることができる。この反応では、クロロホルム中、酸として硫酸を用いる等の方法により、式(1−2)の化合物を単離することなく、式(1−3)の化合物を製造することもできる。
式(1−2)の化合物を、メタノール、エタノールなどのアルコール系溶媒中、塩基を加えて加熱することにより、式(1−3)の化合物を得ることができる。塩基としては、水酸化ナトリウム、水酸化カリウム、炭酸カリウム、炭酸ナトリウム、もしくは炭酸水素ナトリウムなどの無機塩基や、トリエチルアミン、もしくはピリジンなどの有機塩基が用いられる。好ましくは、炭酸カリウム、または炭酸ナトリウムを用いることができる。
式(1−5)の化合物は、式(1−3)の化合物と式(1−4)で表される1,3−ジケトンを、塩基もしくは酸の存在、または非存在下に、室温から100℃で反応させることにより製造できる。溶媒としては、メタノール、エタノールなどのアルコール系溶媒、クロロホルム、ジクロロメタンなどのハロゲン系溶媒、ジメチルホルムアミド、ジメチルスルフォキシド、アセトン、アセトニトリル、ジオキサンやテトラヒドロフランなどのエーテル系溶媒、水、またはこれらの混合溶媒等が用いられる。このうち、メタノール、エタノールなどのアルコール系溶媒がより好ましい。酸としては、酢酸、塩酸、または、塩化亜鉛などのルイス酸などを用いることができる。塩基として、ピロリジン、またはピペリジンなどの有機塩基を用いることができる。
式(1−4)の化合物は、公知化合物を用いるか、あるいは本明細書実施例に記載された方法で製造することができる。また、式(1−5)の化合物は、式(1−2)の化合物から、式(1−3)の化合物を単離することなく製造することもできる。
式(1−6)の化合物は、式(1−5)の化合物を加水分解してカルボン酸とした後、公知の方法に従い環状アミン化合物と縮合させることにより製造できる。縮合方法としては、本明細書記載の水溶性カルボジイミド(EDC:1−(3−ジメチルアミノプロピル)−3−エチルカルボジイミド・塩酸塩等)を用いる方法、混合酸無水物法、または、酸ハライドを用いる方法等が挙げられる。該縮合方法については「コンプリヘンシブ オーガニック トランスフォーメーション(ラロック,R.C.ら著,VCH Publishers,Inc.1989)」(以下、ラロック文献と称する場合がある。)等に記載されている。
原料となる環状アミン化合物は、市販品を用いるか、当業者にとって公知の方法で製造することができる。例えば以下の方法で製造することができる。
Figure 2005030773
[式中、Xは酸素原子、硫黄原子、NHもしくはCHを表し、m、n、R及びRは前記と同義である。尚、上記Nsについては、本明細書において以下同じ意味を表す。]
式(2−2)の化合物は、公知化合物(2−1)から、公知の方法(Synlett,1998,p1301等を参照)に従い、市販のハライドとのアルキル化、または市販、あるいは公知のアルコールとの光延(Mitsunobu)反応を繰り返すことにより製造できる。式(2−3)の化合物は、式(2−2)の化合物を、ジクロロメタンなどのハロゲン系溶媒中、Grubbs触媒(Acc.Chem.Res.,34,p18(2001)等を参照)を加えて室温または加熱することにより得ることができる。
式(2−4)の化合物は、必要に応じて式(2−3)の化合物をヒドロホウ素化や四酸化オスミウムなどを用いた酸化反応や、接触水素添加反応に付すことで二重結合を単結合に変換後、公知の方法(Synlett,1998,p1301等を参照)により脱保護を行うことで製造することができる。必要ならば塩酸塩として単離することもできる。また、脱保護後、そのまま反応系中でアミド化することにより本発明の化合物を得ることができる。二重結合から所望の構造への変換反応としては、本明細書記載の方法以外にも、ラロック文献等に記載されている。
式(2−5)の化合物は、式(2−3)の化合物を公知の方法(Synlett,1998,p1301等を参照)により脱保護を行うことで製造することができる。必要ならば塩酸塩として単離することもできる。また、脱保護後、そのまま反応系中でアミド化することにより式(1)の化合物を得ることができる。
尚、式(2−2)〜式(2−5)で示される(R及び(Rの結合位置は便宜的なものであって、式(2−4)もしくは式(2−5)における環上の炭素原子または窒素原子上であれば特に限定されない。
また、以下に示す方法によっても、環状アミンを製造することができる。
Figure 2005030773
市販のアリルグリシン(3−1)を保護した式(3−2)の化合物から、市販のアルキルハライドを用い上記と同様の方法で式(3−3)の化合物を製造できる。式(3−4)の化合物は、式(3−3)の化合物をテトラヒドロフランなどのエーテル系溶媒、ジクロロメタンなどのハロゲン系溶媒中、水素化アルミニウムリチウム、水素化ジイソブチルアルミニウム、水素化ホウ素リチウムなどを用いて還元することにより得られる。好ましくは、ジクロロメタン中、水素化ジイソブチルアルミニウムを用いる。式(3−3)や式(3−4)の化合物は上記と同様の方法で式(2−4)もしくは式(2−5)に相当する化合物へ導くことができる。
一般式(1)で表される化合物において、Rがカルボキシル基またはカルボキシル基で置換されたアルキル基を表す場合、上記の製造工程の任意の工程で、エステル等の当業者に良く知られた保護基で保護されていてもよい。あるいは、ヒドロキシメチル基から酸化反応によりカルボキシル基に変換してもよい。例えば、式(4−3)で示される本発明の化合物は、以下の方法により製造できる。
Figure 2005030773
[式中、Rは低級アルキル基等のカルボキシル基の保護基を示し、環A、R、R、Rは前記と同義である。]
例えばRが低級アルキル基の場合には、上記の方法により製造できる式(4−1)の化合物を、メタノール、エタノールなどのアルコール系溶媒や、テトラヒドロフラン、ジオキサンなどのエーテル系溶媒中、塩基として水酸化ナトリウム水溶液や水酸化カリウム水溶液を加え、室温または加熱することで、式(4−3)で示した化合物を製造することができる。または、式(4−2)の化合物を酸化することにより式(4−3)の化合物を得ることができる。アルコールのカルボン酸への酸化方法としては、本明細書記載のように、スワン(Swern)酸化(J.Org.Chem.,43,p2480(1978)等を参照)によりアルデヒドに変換後、亜塩素酸ナトリウムを用いてカルボン酸に酸化する方法(Tetrahedron,43,4767(1987))や、直接アルコールをカルボン酸に酸化する方法が挙げられる。酸化方法についてはラロック文献等に記載されている方法を参照することができる。
Rとしては、t−ブチル基等のトリフルオロ酢酸もしくは4N HCl/ジオキサン等の酸で脱保護できる保護基や、ベンジル基等の接触水素化反応で脱保護できる保護基を用いることも可能であり、保護・脱保護反応については「プロテクティブ グループス イン オーガニンック シンセシス(T.W.グリーンら著、John Wiley & Sons,Inc.発行 1991)」等に記載された方法に準じればよい。
次に、ピラゾロピリジン骨格を有する本発明の化合物の製造方法を示す。
Figure 2005030773
[式中、R、R、R、およびRは前記と同義である。]
公知化合物または公知の方法(J.Org.Chem.,59,p2740(1994))によって製造できる式(5−1)で表される化合物を、公知の方法(Tetrahedron Lett.,40,p4133(1972))に従ってN−アミノ化することにより、式(5−2)で表される化合物を製造できる。式(5−2)の化合物と市販のアセチレン、好ましくはジt−ブチルアセチレンを、N,N−ジメチルホルムアミドやジメチルスルフォキシド、メタノールもしくはエタノールなどのアルコール系溶媒、又はテトラヒドロフランもしくはジオキサンなどのエーテル系溶媒中で、好ましくはN,N−ジメチルホルムアミド中で、塩基の存在下、室温または加熱することによって、式(5−3)で示される化合物を得ることができる。塩基としては、炭酸カリウム、炭酸ナトリウム、炭酸セシウム、水酸化ナトリウム、水酸化カリウムなどの無機塩基や、トリエチルアミン、N−メチルモルホリンなどの有機塩基が用いられる。好ましくは炭酸カリウムを用いる。式(5−3)で示される化合物を、酸性条件下室温または加熱することで脱炭酸と加水分解を行うことにより、式(5−4)で表されるカルボン酸を得ることができる。酸として、硫酸、塩酸、トリフルオロ酢酸などが用いられるが、より好ましくはトリフルオロ酢酸を用いて室温で行う。式(5−4)の化合物は、上記と同様の方法を用いて本発明の化合物へと導くことができる。
本発明の化合物を製造する際、任意の工程で必要に応じて水酸基、カルボキシル基もしくはアミノ基等の官能基を保護・脱保護することができる。保護基の種類、保護・脱保護の方法については、当業者に良く知られたものを用いればよく、例えば「プロテクティブ グループス イン オーガニンック シンセシス(T.W.グリーンら著、John Wiley & Sons,Inc.発行 1991)」等を参考にすればよい。また、本発明の化合物を製造する際、任意の工程で必要に応じて官能基変換等を行うことができる。具体的には、クロロ化もしくはブロモ化等のハロゲン化反応、置換反応、Wittig反応等を挙げることができる。これらについても、当業者に良くしられた方法を用いればよく、例えばラロック文献等を参考にすればよい。
一般式(1)で示される本発明の化合物のうち、塩を形成しうる官能基を有している化合物の薬学的に許容される塩としては、例えば、ナトリウム塩、カリウム塩等のアルカリ金属塩、カルシウム塩、マグネシウム塩等のアルカリ土金属塩、亜鉛塩等の無機金属塩、トリエチルアミン、トリエタノールアミンもしくはトリヒドロキシメチルアミノメタン等有機塩、アンモニウム塩、塩酸塩、臭化水素塩、硫酸塩、リン酸塩、硝酸塩等の無機酸塩、および酢酸塩、プロピオン酸塩、コハク酸塩、乳酸塩、リンゴ酸塩、酒石酸塩、クエン酸塩、マレイン酸塩、フマル酸塩、メタンスルホン酸塩、p−トルエンスルホン酸塩、ベンゼンスルホン酸塩もしくはアスコルビン酸塩等の有機酸塩等が挙げられる。これらの塩は一般式(1)で表される化合物と酸又は塩基を混合した後、再結晶などの常法により得ることができる。
また、本発明には、一般式(1)で示される化合物又は薬学上許容される塩の水和物、エタノール溶媒和物等の溶媒和物も含まれる。さらに、本発明には、一般式(1)で示される化合物のあらゆる互変異性体、光学異性体等の存在するあらゆる立体異性体、およびあらゆる態様の結晶形のものも包含している。これらは当業者に良く知られているシリカゲルカラムクロマトグラフィー、HPLC、イオン交換クロマトグラフィー、再結晶等の方法を用いて、適宜精製することができる。
前記光学異性体を純粋に得るためには、当業者に公知の光学分割法を用いればよい。具体的には、本発明の化合物もしくはその中間体が塩基性官能基を有する場合には不活性溶媒中光学活性な酸(例えば、マンデル酸、N−ベンジルオキシアラニン、乳酸などのモノカルボン酸類、酒石酸、o−ジイソプロピリデン酒石酸、リンゴ酸などのジカルボン酸類、カンファースルフォン酸、ブロモカンファースルフォン酸などのスルフォン酸類)と塩を形成させることができる。また、本発明の化合物もしくはその中間体が酸性置換基を有する場合は光学活性なアミン(例えばα−フェネチルアミン、キニン、キニジン、シンコニジン、シンコニン、ストリキニーネ等の有機アミン類)と塩を形成させることもできる。塩を形成させる温度としては、室温から溶媒の沸点の範囲が挙げられる。
一般式(1)で表される化合物、またはその薬学上許容される塩は、PAR2の機能亢進が関与する、アレルギー疾患、呼吸器疾患、心血管系疾患、神経系疾患、炎症性疾患、神経性炎症性疾患、皮膚疾患等の疾患の治療剤、予防剤、または進行防止剤として有用である。
該疾患として具体的には、関節炎(変形性関節炎、変形性関節症、脊椎関節症、痛風関節炎、全身性エリテマトーデス、若年性関節炎および慢性関節リウマチを含む)、熱(リウマチ熱並びにインフルエンザおよび他のウイルス性感染症関連熱)、一般的な感冒、月経困難、月経痙攣、炎症性腸疾患,クローン病、気腫、急性呼吸窮迫症候群、ぜん息、気管支炎、慢性閉塞性肺疾患、アルツハイマー病、器官移植毒性、悪液質、アレルギー反応、アレルギー性接触過敏症、癌(例えば、結腸癌,乳癌、肺癌および前立腺癌を含めた固形腫瘍癌;白血病およびリンパ腫を含めた造血悪性疾患;ホジキン病;再生不良性貧血、皮膚癌および家族性腺腫ポリポーシス)、組織潰瘍、消化性潰瘍、胃炎、限局性腸炎、潰瘍性大腸炎、憩室炎、再発性胃腸病変、胃腸出血、凝固、貧血、滑膜炎、痛風、強直性脊椎炎、再狭窄、歯周病、表皮水泡症、骨粗しょう症、人工関節インプラントのゆるみ、アテローム硬化症(アテローム硬化症性血小板破壊)、大動脈瘤(腹部大動脈瘤および脳大動脈瘤)、結節性動脈周囲炎、うっ血性心不全、心筋梗塞、発作、大脳虚血、頭部外傷、脊髄損傷、神経痛、神経変性疾患(急性および慢性)、自己免疫疾患、ハンティングトン病、パーキンソン病、片頭痛、うつ病、末梢神経障害、痛み(背の下部および首の痛み、頭痛並びに歯痛)、歯肉炎、大脳アミロイド血管障害、ヌートロピック(nootropic)または認識強化、筋萎縮性側策硬化症、多発性硬化症、目の脈管形成、角膜損傷、黄斑変性、結膜炎、異常創傷治癒、筋肉もしくは関節の捻挫または緊張、腱炎、皮膚疾患(例えば、乾癬、湿疹、強皮症および皮膚炎)、重症筋無力症、多発性筋炎、筋炎、滑液包炎、熱傷、糖尿病(タイプIおよびII糖尿病、糖尿病性網膜症)、腫瘍浸潤、腫瘍成長、腫瘍転移、角膜傷跡、強膜炎、免疫不全疾患(例えば、人のエイズ、ネコのFLV、FIV)、敗血症、早産、低プロトロンビン血症、血友病、甲状腺炎、サルコイドーシス、ベーチェット症候群、過敏症、腎臓疾患等が挙げられる。
好ましくは、関節炎(変形性関節症、脊椎関節症、痛風関節炎、全身性エリテマトーデス、若年性関節炎および慢性関節リウマチ等が挙げられる。)、皮膚炎、発熱、ぜん息、骨吸収、心臓血管疾患、月経困難、早産、腎炎、ネフローゼ、アテローム硬化症、低血圧、ショック、又は疼痛を伴う神経組織由来の神経性炎症、癌、およびアルツハイマー病等の疾患が挙げられる。
また、一般式(1)で表される化合物、またはその薬学上許容される塩は、軟骨や滑膜の細胞でのPGE2放出を抑制し、PGE2産生抑制剤としても有用である。
更に、一般式(1)で表される化合物、またはその薬学上許容される塩は、本明細書実施例に示されるとおり、インビボにおいて優れた抗掻痒活性を示し、抗掻痒剤として、掻痒を伴う各種疾患の治療もしくは予防剤としても有用である。掻痒として、具体的には、眼掻痒、鼻掻痒、皮膚掻痒、全身性掻痒が挙げられる。また、掻痒を伴う疾患としては、アトピー性皮膚炎、皮膚掻痒症、蕁麻疹、接触性皮膚炎、乾癬、乾皮症、脂漏性皮膚炎、神経性皮膚炎、自己感作性皮膚炎、毛虫皮膚炎、虫刺症、光線過敏症、痒疹、湿疹、腎透析時掻痒症、老人性皮膚掻痒症、老人性乾皮症、果肉過敏症、オピオイド系鎮痛剤投与時の掻痒症、皮脂欠乏性湿疹、アトピー性角結膜炎、アレルギー性角結膜炎、感染性角結膜炎、及び春季カタル等の疾患に伴う掻痒等が挙げられる。また、本発明の化合物は、内科系疾患(悪性腫瘍、糖尿病、肝疾患、腎不全、痛風、甲状腺疾患、血液疾患)、寄生虫、真菌、もしくはウイルス等の感染症、心因性のストレス、薬剤の過敏症、又は妊娠などが原因となる掻痒の予防又は治療剤としても有効である。 更に、本発明の化合物は、PAR2阻害活性を有するため、PAR2の生理作用を調べるための研究ツールとしても有用である。
以下、本発明の化合物を有効成分として含有する、上記疾患の治療剤、予防剤、または進行防止剤等を医薬品として用いる場合の投与量および投与形態などにつき記述する。
本発明の化合物は、経口または非経口投与、好ましくは経口投与することができ、適切な添加剤、基材、担体とともに医薬組成物として経口または非経口投与に適した種々の剤型で、ヒトおよびヒト以外の動物に使用される。例えば、経口的に投与する場合、通常用いられる投与形態、例えば錠剤、カプセル剤、シロップ剤、懸濁液などで投与することができる。非経口的に投与する場合、溶液、乳剤、懸濁液などの液剤を注射剤又は点眼剤として投与すること、坐剤の型で直腸投与すること、軟膏剤、クリーム剤、ローションなどの経皮剤として投与すること等ができる。このような投与剤型は、通常の担体、賦型剤、結合剤、安定化剤などの助剤と有効成分を配合することにより一般的方法に従って製造することができる。注射剤型で用いる場合は、水、生理食塩水、油、ブドウ糖水溶液などの生理的に許容しうる担体に溶解または懸濁し、補助剤として乳化剤、安定化剤、浸透圧調整用塩、溶解補助剤、または緩衝剤を必要に応じて含有してもよい。
経皮剤として投与する場合、基剤の他必要に応じて、安定化剤、防腐剤、乳化剤、懸濁化剤安定剤、酸化防止剤、香料、充填剤、あるいは他の経皮吸収促進剤などを添加することができる。軟膏剤の基剤としては、例えば脂肪油、ラノリン、ワセリン、パラフィン、プラスチベース、グリコール類、高級脂肪酸、高級アルコール等が挙げられ、ローション剤の基剤としては、例えばエタノール、グリセリン、グリコール等が挙げられる。液剤の基剤としては、例えばエタノール、水、グリコール等が用いられる。
投与量、投与回数は、対象とする疾患、患者の症状、年齢、体重等、および投与形態などによって異なるが、経口投与する場合、有効成分は通常は成人に対し1日あたり約1〜1000mgの範囲、好ましくは、約10〜500mgの範囲を1回または数回に分けて投与することができる。注射剤として投与する場合、有効成分は約0.1〜約500mgの範囲、好ましくは約3〜約100mgの範囲を1回または数回に分けて投与することができる。
一般式(1)で表される化合物の好ましいものとして、以下の表1〜表2の化合物が例示される。
Figure 2005030773
Figure 2005030773
Figure 2005030773
Figure 2005030773
以下、実施例により本発明をさらに詳細に説明するが、本発明の技術的範囲はこれら実施例に限定されるものではない。
実施例1(参考例1)
(工程1)
(2E)−3−シアノ−2−ヒドラゾノプロパン酸エチル
Figure 2005030773
エチルシアノピルベートナトリウムエノラート(ethyl cyanopyruvate sodium−enolate)(7.8g)を酢酸(40ml)とエタノール(40ml)に溶かし、0℃でヒドラジン1水和物を加えた。30分後室温にし、一晩攪拌した。反応液を濃縮後、水、クロロホルムを加え、分液、抽出した。有機層を乾燥、濃縮し、目的物(5.0g)を得た。
H−NMR(400MHz,CDCl)δppm:1.34−1.41(m,3H),3.46(s),3.63(s),4.26−4.38(m,2H),6.52(brs),8.50(br)
(工程2)
エチル 5−フェニル−7−(トリフルオロメチル)ピラゾロ[1,5−a]ピリミジン−2−カルボキシレート
Figure 2005030773
工程1で得られた化合物(5.0g)をエタノール(50ml)に溶かし、炭酸カリウム(1.0g)を加え、加熱還流した。3時間後、4,4,4−トリフルオロ−1−フェニル−1,3−ブタンジオン(10.5g)を加え、加熱還流した。生じた結晶をろ過、乾燥し、目的物(4.0g)を得た。
H−NMR(400MHz,DMSO−d)δppm:1.37(t,3H,J=7.1Hz),4.42(q,2H,J=7.1Hz),7.44(s,1H),7.57−7.65(m,3H),8.31−8.38(m,3H)
(工程3)
5−フェニル−7−(トリフルオロメチル)ピラゾロ[1,5−a]ピリミジン−2−カルボン酸カリウム
Figure 2005030773
工程2で得られた化合物(4.6g)、エタノール(50ml)、1N水酸化カリウム水溶液(15ml)を混ぜ、加熱還流した。30分後、室温に冷却した。生じた結晶をろ過、乾燥し、目的物(4.2g)を得た。
H−NMR(400MHz,DMSO−d)δppm:6.87(s,1H),7.53−7.59(m,3H),8.02(s,1H),8.24−8.31(m,2H)
実施例2(参考例2)
(工程1)
1−(4−クロロフェニル)−4,4,4−トリフルオロブタン−1,3−ジオン
Figure 2005030773
4−クロロアセトフェノン(4.6ml)をテトラヒドロフラン(50ml)に溶かし、0℃に冷却後、水素化ナトリウム(1.55g;60%)を加えた。15分後室温にし、30分攪拌した。反応液を0℃に冷却し、トリフルオロ酢酸エチルを加えた。2時間後、室温にあげ、さらに1.5時間攪拌した。1N塩酸を反応液に加え、酢酸エチルで抽出した。有機層を水洗、乾燥、濃縮し、目的物(10.6g)を得た。
H−NMR(400MHz,DMSO−d)δppm:7.02(s,1H),7.66(d,2H,J=8.7Hz),8.14(d,2H,J=8.7Hz)
(工程2)
エチル 5−(4−クロロフェニル)−7−(トリフルオロメチル)ピラゾロ[1,5−a]ピリミジン−2−カルボキシレート
Figure 2005030773
工程1で得た化合物と参考例1(実施例1)の工程1で得た化合物を用いて、参考例1の工程2と同様の方法で目的物(4.3g)を得た。
H−NMR(400MHz,CDCl)δppm:1.47(t,3H,J=7.1Hz),4.51(q,2H,J=7.1Hz),7.38(s,1H),7.52−7.57(m,2H),7.71(s,1H),8.07−8.12(m,2H)
(工程3)
5−(4−クロロフェニル)−7−(トリフルオロメチル)ピラゾロ[1,5−a]ピリミジン−2−カルボン酸カリウム
Figure 2005030773
工程2で得た化合物から、参考例1の工程3と同様の方法で目的物(3.5g)を得た。
実施例3(参考例3)
(工程1)
2−ニトロ−N−4−ペンテン−1−イルベンゼンスルフォンアミド
Figure 2005030773
2−ニトロ−N−t−ブトキシカルボニルベンゼンスルフォンアミド(5.0g)をN,N−ジメチルホルムアミド(50ml)に溶かし、炭酸カリウム(6.9g)、5−ブロモ−1−ペンテン(2.2ml)を加え、80℃で加熱した。4時間後、反応液を水中に流し込み、酢酸エチルで抽出した。有機層を水洗、乾燥、濃縮し、残渣をトリフルオロ酢酸(75ml)に溶かし、2時間室温で静置した。反応液を濃縮し、トルエンで共沸した。減圧乾燥し、目的物(3.6g)を得た。
H−NMR(400MHz,CDCl)δppm:1.65(m,2H),2.09(m,2H),3.12(q,2H,J=6.9Hz),4.94−5.03(m,2H),5.29(brt,1H,J=5.6Hz),5.72(m,1H),7.72−7.78(m,2H),7.87(m,1H),8.14(m,1H)
(工程2)
N−{2−[(ベンジルオキシ)メチル]−2−プロペン−1−イル}−2−ニトロ−N−4−ペンテン−1−イルベンゼンスルフォンアミド
Figure 2005030773
工程1で得た化合物(1.8g)をトルエン(50ml)に溶かし、2−[(ベンジルオキシ)メチル]−2−プロペン−1−オール(1.4g)、トリフェニルフォスフィン(2.3g)、ジエチルアゾジカルボキシレート(3.8ml)を混ぜ、室温で攪拌した。1時間後、反応液を水中に注ぎ、酢酸エチルで抽出した。有機層を乾燥、濃縮し、残渣をシリカゲルカラム(300g、酢酸エチル:ヘキサン=1:4〜1:3)で精製し、目的物(2.3g)を得た。
H−NMR(400MHz,CDCl)δppm:1.58(m,2H),1.96(m,2H),3.29(m,2H),3.91(s,2H),4.01(s,2H),4.42(s,2H),4.91−5.00(m,2H),5.17(s,1H),5.26(s,1H),5.68(m,1H),7.26−7.38(m,5H),7.56−7.67(m,3H),8.02(m,1H)
(工程3)
6−[(ベンジルオキシ)メチル]−1−[(2−ニトロフェニル)スルフォニル]−2,3,4,7−テトラヒドロ−1H−アゼピン
Figure 2005030773
工程2で得た化合物(2.3g)をジクロロメタン(400ml)に溶かし、第二世代Grubbs触媒(0.1g)を混ぜ、反応液を還流した。1時間後反応液を濃縮し、残渣をシリカゲルカラム(150g、酢酸エチル:ヘキサン=1:2)で精製し、目的物(2.0g)を得た。
H−NMR(400MHz,CDCl)δppm:1.89(m,2H),2.32(m,2H),3.57(t,2H,J=6.1Hz),3.96(s,2H),3.99(s,2H),4.48(s,2H),5.85(t,1H,J=5.7Hz),7.27−7.37(m,5H),7.57−7.68(m,3H),7.98(m,1H)
(工程4)
tert−ブチル 6−[(ベンジルオキシ)メチル]−2,3,4,7−テトラヒドロ−1H−アゼピン−1−カルボキシレート
Figure 2005030773
工程3で得た化合物(2.0g)をN,N−ジメチルホルムアミド(50ml)に溶かし、炭酸カリウム(2.1g)、ベンゼンチオール(0.77ml)を加え、室温で2時間攪拌した。Boc20(2.2g)を加え、1時間攪拌した。反応液を水中に注ぎ、酢酸エチルで抽出した。有機層を水洗、乾燥、濃縮し、残渣をシリカゲルカラム(200g、酢酸エチル:ヘキサン=1:10〜1:5)で精製し、目的物(1.5g)を得た。
H−NMR(400MHz,CDCl)δppm:1.38−1.46(m,4H),1.75−1.84(m,2H),2.18−2.25(m,2H),3.46−3.62(m,2H),3.85−4.03(m,4H),4.29(s,2H),5.77(t,1H,J=5.6Hz),7.26−7.38(m,5H)
(工程5)
アゼパン−3−イルメタノール 塩酸塩
Figure 2005030773
工程4で得た化合物(1.5g)をエタノール(50ml)に溶かし、10%Pd−Cを加え、水素雰囲気下、5時間攪拌した。反応液をセライトろ過し、エタノールで洗浄した。ろ液を濃縮後、乾燥し、残渣に4N塩酸−ジオキサン溶液(10ml)を加えた。室温で2時間攪拌後、反応液を濃縮した。減圧乾燥し、目的物(0.77g)を得た。
H−NMR(400MHz,DMSO−d)δppm:1.24(m,1H),1.47(m,1H),1.63−1.99(m,5H),2.75(m,1H),2.98−3.35(m,5H),4.86(brs,1H),9.04(brs,1H),9.23(brs,1H)
実施例4(参考例4)
(工程1)
エチル2−{[(2−ニトロフェニル)スルフォニル]アミノ}−4−ペンテノエート
Figure 2005030773
エタノール(20ml)を0℃に冷却し、塩化チオニル(4.4ml)を滴下した。30分後、アリルグリシン(2.0g)を加えた。30分攪拌後室温に上げ、そのまま一晩攪拌した。反応液を濃縮し、減圧乾燥した。得られた粗生成物をクロロホルム(40ml)に溶かし、0℃で塩化o−ニトロベンゼンスルホニル(4.2g)とトリエチルアミン(5.2ml)を加え攪拌した。3時間後、水、クロロホルムを加え、分液、抽出した。有機層を乾燥、濃縮し、残渣をシリカゲルカラム(200g、酢酸エチル:ヘキサン=1:2)で精製し、エステル(5.7g)を得た。
H−NMR(400MHz,CDCl)δppm:1.11(t,3H,J=7.1Hz),2.58(m,2H),3.97(q,2H,J=7.1Hz),4.28(dt,1H,J=8.8,5.9Hz),5.12−5.19(m,2H),5.69(m,1H),6.11(brd,1H,J=8.8Hz),7.70−7.76(m,2H),7.93(m,1H),8.08(m,1H)
(工程2)
エチル 2−{3−ブテン−1−イル[(2−ニトロフェニル)スルフォニル]アミノ}−4−ペンテノエート
Figure 2005030773
参考例3(実施例3)の工程2と同様の方法により、目的物(1.9g)を得た。
H−NMR(400MHz,CDCl)δppm:1.12(t,3H,J=7.2Hz),2.38(m,1H),2.46−2.59(m,2H),2.83(m,1H),3.21(m,1H),3.49(m,1H),4.04(m,2H),4.71(dd,1H,J=9.2,6.0Hz),5.03−5.24(m,4H),5.67−5.88(m,2H),7.59(m,1H),7.69(m,2H),8.06(m,1H)
(工程3)
エチル 1−[(2−ニトロフェニル)スルフォニル]−2,3,6,7−テトラヒドロ−1H−アゼピン−2−カルボキシレート
Figure 2005030773
参考例3(実施例3)の工程3と同様の方法により、目的物(1.5g)を得た。
H−NMR(400MHz,CDCl)δppm:1.16(t,3H,J=7.1Hz),2.35−2.45(m,2H),2.65(m,1H),2.87(m,1H),3.48(m,1H),3.83(m,1H),4.03−4.15(m,2H),4.92(dd,1H,J=6.3,3.8Hz),5.70−5.85(m,2H),7.60−7.72(m,3H),8.10(m,1H)
(工程4)
1−ベンジル2−エチル 2,3,6,7−テトラヒドロ−1H−アゼピン−1,2−ジカルボキシレート
Figure 2005030773
(式中、Zはベンジルオキシカルボニル基を表す。)
工程3で得られた環化体(0.55g)をN,N−ジメチルホルムアミド(20ml)に溶かし、室温で炭酸カリウム(0.64g)、ベンゼンチオール(0.24ml)を加え、45分攪拌した。ベンジルクロロホルメート(0.44ml)を滴下し、1時間攪拌した。反応液に水、酢酸エチルを加え、分液、抽出した。有機層を水洗、乾燥、濃縮後、残渣をシリカゲルカラム(100g、酢酸エチル:ヘキサン=1:6)で精製し、目的物(0.42g)を得た。
H−NMR(400MHz,CDCl)δppm:1.12−1.28(m,3H),2.29−2.80(m,4H),3.65−4.22(m,3H),4.64−4.82(m,1H),5.04−5.22(m,2H),5.55−5.68(m,2H),7.27−7.38(m,5H)
(工程5)
エチル アゼパン−2−カルボキシレート
Figure 2005030773
工程4で得られた化合物(0.42g)をエタノール(15ml)に溶かし、10%水酸化パラジウムを加え、水素雰囲気下室温で3時間激しく攪拌した。反応液をセライトろ過し、ろ液を濃縮後減圧乾燥し、目的物(0.2g)を得た。
H−NMR(400MHz,CDCl)δppm:1.27(t,3H,J=7.1Hz),1.54−1.78(m,7H),2.09(m,1H),2.72(dt,1H,J=14.2,5.9Hz),3.05(dt,1H,J=14.2,5.0Hz),3.49(dd,1H,J=9.3,5.0Hz),4.17(q,2H,J=7.1Hz)
(工程6)
2−({[tert−ブチル(ジメチル)シリル]オキシ}メチル)−1−[(2−ニトロフェニル)スルフォニル]−2,3,6,7−テトラヒドロ−1H−アゼピン
Figure 2005030773
工程3で得られた化合物(1.45g)をジクロロメタン(50ml)に溶かし、−78℃に冷却後、ジイソブチルアルミニウムハイドライド(DIBAL)(0.93M in hexane、9.5ml)を滴下した。30分後、0℃に昇温し、2時間後、DIBAL(3ml)を追加した。30分後、メタノールを加え反応を停止後、反応液を酒石酸NaK(30g)の水溶液中に流し込み、室温で2時間攪拌した。有機層を抽出後、乾燥、濃縮し、粗生成物(1.2g)を得た。粗生成物(1.0g)をN,N−ジメチルホルムアミド(20ml)に溶かし、室温でt−ブチルジメチルクロロシラン(0.63g)、イミダゾール(0.29g)を加えた。1時間攪拌後、反応液を水中に流し込み、酢酸エチル加え分液抽出した。有機層を水洗、乾燥、濃縮後、残渣をシリカゲルカラム(80g、酢酸エチル:ヘキサン=1:4)で精製し、目的物(1.25g)を得た。
H−NMR(400MHz,CDCl)δppm:2.28−2.64(m,4H),3.32(m,1H),3.53(d,1H,J=11.8,5.3Hz),3.77(d,1H,J=11.8,8.4Hz),3.86(dt,1H,J=15.0,4.2Hz),4.14(m,1H),5.64(m,1H),5.75(m,1H),7.60−7.73(m,3H),8.12(m,1H)
(工程7)
2−({[tert−ブチル(ジメチル)シリル]オキシ}メチル)アゼパン
Figure 2005030773
工程6で得られた化合物(1.25g)をN,N−ジメチルホルムアミド(20ml)に溶かし、メルカプト酢酸(0.41ml)、水酸化リチウム(0.28g)を加え、室温で攪拌した。1時間後、反応液を飽和炭酸水素ナトリウム水溶液中に流し込み、酢酸エチルで抽出した。有機層を飽和炭酸水素ナトリウム水溶液、飽和食塩水で洗浄後、乾燥、濃縮し、粗生成物(0.71g)を得た。得られた粗生成物(0.36g)をエタノール(10ml)に溶かし、10%Pd−C(0.1g)を加え、水素雰囲気下6時間攪拌した。反応液をセライトろ過し、ろ液を濃縮、減圧乾燥し、目的物(0.32g)を得た。
H−NMR(400MHz,CDCl)δppm:0.05(s,6H),0.89(s,9H),1.23(m,1H),1.49−1.77(m,7H),2.63−2.78(m,2H),3.08(m,1H),3.37(dd,1H,J=9.8,8.5Hz),3.50(dd,1H,J=9.8,4.5Hz)
実施例5(参考例5)
1−[(2−ニトロフェニル)スルフォニル]アゾカン−4,5−ジオール
Figure 2005030773
上記実施例と同様な方法で製造できる1−[(2−ニトロフェニル)スルフォニル]−1,2,3,4,7,8−ヘキサヒドロアゾシン(0.1g)をアセトニトリル(4ml)、水(1ml)に溶かし、N−メチルモルホリン−N−オキシド(0.08g)、四酸化オスミウム(10%、0.09g)を加え、室温で3日間攪拌した。反応液をろ過し、ろ液に水、酢酸エチルを加え分液、抽出した。有機層を水洗、乾燥、濃縮し、目的物(0.11g)を得た。
H−NMR(400MHz,CDCl)δppm:1.42−1.87(m,6H),3.07−3.20(m,2H),3.28−3.48(m,2H),3.83(m,2H),4.51(d,1H,J=4.6Hz),4.80(d,1H,J=5.2Hz),7.81−8.02(m,4H)
実施例6(参考例6)
ベンジル 4,5−ジヒドロキシアゼパン−1−カルボキシレート
Figure 2005030773
テトラヒドロフラン(16ml)に30%過酸化水素水(0.14ml)、ギ酸(0.70ml)を加え0℃に冷却後、上記実施例と同様の方法で製造できるベンジル 2,3,6,7−テトラヒドロ−1H−アゼピン−1−カルボキシレート(0.27g)を加えた。一晩攪拌後、反応液にチオ硫酸ナトリウム水溶液を加え濃縮した。13N水酸化ナトリウム水溶液(3ml)を加え室温で2時間攪拌後、水、酢酸エチルを加え分液、抽出した。有機層を水洗、乾燥、濃縮し、目的物(0.25g)を得た。
H−NMR(400MHz,CDCl)δppm:1.62−1.78(m,2H),1.99−2.13(m,2H),2.80(brs,2H),3.23−3.70(m,6H),5.12(s,2H),7.26−7.40(m,5H)In the present specification, examples of the alkyl group include linear or branched alkyl groups having 1 to 4 carbon atoms. Specific examples include a methyl group, an ethyl group, a propyl group, a 1-methylethyl group, a butyl group, a 1-methylpropyl group, a 2-methylpropyl group, or a 1,1-dimethylethyl group.
In the present specification, examples of the alkenyl group include linear or branched alkenyl groups having 1 to 4 carbon atoms. Specifically, vinyl group, 1-propenyl group, 2-propenyl group, 1-methylvinyl group, 1-butenyl group, 2-butenyl group, 3-butenyl group, 1-methyl-1-propenyl group, 1-methyl -2-propenyl group, 2-methyl-1-propenyl group, 2-methyl-2-propenyl group, and the like.
In the present specification, examples of the alkynyl group include linear or branched alkynyl groups having 1 to 4 carbon atoms. Specific examples include an ethynyl group, a 1-propynyl group, a 2-propynyl group, a 1-butynyl group, a 2-butynyl group, a 3-butynyl group, and a 1-methyl-2-propynyl group.
In the present specification, examples of the cycloalkyl group include cycloalkyl groups having 3 to 6 carbon atoms. Specific examples include a cyclopropyl group, a cyclobutyl group, a cyclopentyl group, and a cyclohexyl group.
In the present specification, examples of the alkoxy group include linear or branched alkoxy groups having 1 to 4 carbon atoms. Specific examples include methoxy group, ethoxy group, propoxy group, 1-methylethoxy group, butoxy group, 1-methylpropoxy group, 2-methylpropoxy group or 1,1-dimethylethoxy group.
In the present specification, examples of the alkylthio group include linear or branched alkylthio groups having 1 to 4 carbon atoms. Specific examples include a methylthio group, an ethylthio group, a propylthio group, a 1-methylethylthio group, a butylthio group, a 1-methylpropylthio group, a 2-methylpropylthio group, or a 1,1-dimethylethylthio group.
In this specification, in the alkyl group substituted with a hydroxyl group, a carboxyl group, a mercapto group, an amino group or a substituted amino group, any one or more hydrogen atoms of the alkyl group may be substituted.
In the present specification, the halogen atom represents a fluorine atom, a chlorine atom, a bromine atom, or an iodine atom, and preferably represents a fluorine atom or a chlorine atom.
In the present specification, examples of the haloalkyl group include the same or different haloalkyl groups having 1 to 2 carbon atoms containing 1 to 3 halogen atoms, specifically, a trifluoromethyl group, a difluoromethyl group, 2 2,2-trifluoroethyl group or 2,2-difluoroethyl group. Preferably, a trifluoromethyl group is used.
In the present specification, examples of the substituted amino group include 1 or 2 amino groups substituted with 1 or 2 substituents selected from an alkyl group, a kenyl group, an alkynyl group, and a cycloalkyl group. In addition, cyclic amino groups such as 1-pyrrolidinyl group, 1-piperidinyl group, piperazino group, morpholino group, or thiomorpholino group are also included in the category of substituted amino groups.
As the “6- to 8-membered saturated or unsaturated nitrogen-containing heterocycle” in the ring A in the general formula (1), 1 to 3 nitrogen atoms, 0 or 1 oxygen atom, and 0 or 1 sulfur The saturated or unsaturated nitrogen-containing heterocyclic ring containing 1-3 heteroatoms selected from an atom is mentioned, You may have 0-2 double bonds. Specifically, piperidine, piperazine, morpholine, thiomorpholine, S-oxothiomorpholine, S, S-dioxothiomorpholine, perhydroazepine (azepane), 2,3,6,7-tetrahydroazepine, 1,3 Perhydrooxazepine (1,3-oxazepane), perhydroazocine (azocan), 1,2,3,6,7,8-hexahydroazocine, 1,4-perhydrooxazocine (1, 4-oxazocan) etc. can be illustrated.
R 1 When R represents a hydroxyl group, a carboxyl group, a mercapto group, an amino group, or a substituted amino group, it can be bonded to any carbon atom constituting the ring A. R 1 When represents an alkyl group substituted with a hydroxyl group, a carboxyl group, a mercapto group, an amino group or a substituted amino group, it can be bonded to any carbon atom or nitrogen atom constituting the ring A.
R 1 Is preferably a hydroxyl group, carboxyl group, hydroxymethyl group, 1-hydroxyethyl group, 2-hydroxyethyl group, 1-hydroxy-1-methylethyl group, 2-hydroxy-1-methylethyl group, 1-hydroxypropyl group 2-hydroxypropyl group, 3-hydroxypropyl group, 1,2-dihydroxyethyl group, 2,3-dihydroxypropyl group, carboxymethyl group, 1-carboxyethyl group, 2-carboxyethyl group, 1-carboxy-1 -Methylethyl group, 2-carboxy-1-methylethyl group, 1-carboxypropyl group, 2-carboxypropyl group, 3-carboxypropyl group, 1,2-dicarboxyethyl group, 2,3-dicarboxypropyl group Etc.
R 2 Represents preferably an alkyl group having 1 to 2 carbon atoms or an alkoxy group having 1 to 2 carbon atoms.
R in general formula (1) 3 As the aryl group, a phenyl group or a naphthyl group can be mentioned, and a phenyl group is preferable.
When the aryl group, for example, a phenyl group or a naphthyl group is substituted, the substituent is a halogen atom, an alkyl group having 1 to 4 carbon atoms, a haloalkyl group having 1 to 2 carbon atoms, or an alkyl group having 1 to 4 carbon atoms. An alkoxy group is mentioned.
R 3 Preferably represents an alkyl group having 1 to 3 carbon atoms, a phenyl group, a substituted phenyl group, or a haloalkyl group having 1 to 2 carbon atoms. A particularly preferred example is a trifluoromethyl group.
R in the compound represented by the general formula (1) 4 And R 5 Are preferably the same or different and are a hydrogen atom, fluorine atom, chlorine atom, methyl group, ethyl group, methoxy group, ethoxy group, trifluoromethyl group, 2,2,2-trifluoroethyl group, 2,2 -A difluoroethyl group etc. are mentioned.
The compound of the present invention represented by the general formula (1) can be produced by the following method.
Figure 2005030773
[Wherein m, n, R 1 , R 2 , R 3 , R 4 And R 5 Is as defined above. ]
The hydrazone of formula (1-2) can be produced by reacting a known compound of formula (1-1) with hydrazine monohydrate at 0 to 100 ° C. under acidic conditions. Solvents include alcohol solvents such as methanol and ethanol, halogen solvents such as chloroform and dichloromethane, ether solvents such as dimethylformamide, dimethyl sulfoxide, acetone, acetonitrile, dioxane and tetrahydrofuran, water, or a mixed solvent thereof. Is used. Alcohol solvents such as methanol and ethanol are more preferred. As the acid, acetic acid, hydrochloric acid, sulfuric acid, trifluoroacetic acid and the like can be used. In this reaction, the compound of formula (1-3) can also be produced without isolating the compound of formula (1-2) by a method such as using sulfuric acid as an acid in chloroform.
A compound of formula (1-3) can be obtained by adding a base and heating the compound of formula (1-2) in an alcohol solvent such as methanol or ethanol. As the base, an inorganic base such as sodium hydroxide, potassium hydroxide, potassium carbonate, sodium carbonate, or sodium hydrogen carbonate, or an organic base such as triethylamine or pyridine is used. Preferably, potassium carbonate or sodium carbonate can be used.
The compound of formula (1-5) is obtained by reacting the compound of formula (1-3) and the 1,3-diketone represented by formula (1-4) from room temperature in the presence or absence of a base or acid. It can manufacture by making it react at 100 degreeC. Solvents include alcohol solvents such as methanol and ethanol, halogen solvents such as chloroform and dichloromethane, ether solvents such as dimethylformamide, dimethyl sulfoxide, acetone, acetonitrile, dioxane and tetrahydrofuran, water, or a mixed solvent thereof. Etc. are used. Of these, alcohol solvents such as methanol and ethanol are more preferred. As the acid, acetic acid, hydrochloric acid, or Lewis acid such as zinc chloride can be used. As the base, an organic base such as pyrrolidine or piperidine can be used.
As the compound of the formula (1-4), a known compound can be used, or it can be produced by a method described in Examples of the present specification. In addition, the compound of formula (1-5) can also be produced from the compound of formula (1-2) without isolating the compound of formula (1-3).
The compound of the formula (1-6) can be produced by hydrolyzing the compound of the formula (1-5) to give a carboxylic acid and then condensing it with a cyclic amine compound according to a known method. As the condensation method, a method using a water-soluble carbodiimide (EDC: 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide / hydrochloride, etc.) described in the present specification, a mixed acid anhydride method, or an acid halide is used. The method to use etc. are mentioned. The condensation method is described in, for example, “Comprehensive Organic Transformation (Larlock, RC, et al., VCH Publishers, Inc. 1989)” (hereinafter sometimes referred to as Laroc literature).
The cyclic amine compound used as a raw material can be a commercially available product or can be produced by a method known to those skilled in the art. For example, it can be produced by the following method.
Figure 2005030773
[Where X 1 Is oxygen atom, sulfur atom, NH or CH 2 M, n, R 1 And R 2 Is as defined above. In addition, about the said Ns, the same meaning is represented hereafter in this specification. ]
The compound of the formula (2-2) is obtained from the known compound (2-1) according to a known method (see Synlett, 1998, p1301, etc.), alkylation with a commercially available halide, or a commercially available or known alcohol. Can be produced by repeating the Mitsunobu reaction. The compound of the formula (2-3) is obtained by converting the compound of the formula (2-2) into a Grubbs catalyst (Acc. Chem. Res., In a halogen-based solvent such as dichloromethane. 34 , P18 (2001), etc.) is added and heated at room temperature or heated.
If necessary, the compound of formula (2-4) can be converted to a double bond by subjecting the compound of formula (2-3) to an oxidation reaction using hydroboration or osmium tetroxide or a catalytic hydrogenation reaction. After conversion to a single bond, it can be produced by performing deprotection by a known method (see Synlett, 1998, p1301, etc.). If necessary, it can be isolated as a hydrochloride salt. Moreover, the compound of this invention can be obtained by amidating in a reaction system as it is after deprotection. The conversion reaction from the double bond to the desired structure is described in the Laroc literature and the like in addition to the method described in this specification.
The compound of the formula (2-5) can be produced by deprotecting the compound of the formula (2-3) by a known method (see Synlett, 1998, p1301, etc.). If necessary, it can be isolated as a hydrochloride salt. Moreover, the compound of Formula (1) can be obtained by amidating in a reaction system as it is after deprotection.
In addition, (R) shown by Formula (2-2)-Formula (2-5) 1 ) m And (R 2 ) n The bonding position of is not particularly limited as long as it is on the carbon atom or nitrogen atom on the ring in formula (2-4) or formula (2-5).
Moreover, a cyclic amine can be manufactured also by the method shown below.
Figure 2005030773
From the compound of the formula (3-2) in which the commercially available allylglycine (3-1) is protected, the compound of the formula (3-3) can be produced by a method similar to the above using a commercially available alkyl halide. In the compound of formula (3-4), the compound of formula (3-3) is mixed with lithium aluminum hydride, diisobutylaluminum hydride, lithium borohydride and the like in an ether solvent such as tetrahydrofuran and a halogen solvent such as dichloromethane. Obtained by reduction. Preferably, diisobutylaluminum hydride is used in dichloromethane. Compounds of formula (3-3) and formula (3-4) can be led to compounds corresponding to formula (2-4) or formula (2-5) in the same manner as described above.
In the compound represented by the general formula (1), R 1 When represents a carboxyl group or an alkyl group substituted with a carboxyl group, it may be protected with a protecting group well known to those skilled in the art, such as an ester, in any step of the above production process. Alternatively, a hydroxymethyl group may be converted to a carboxyl group by an oxidation reaction. For example, the compound of the present invention represented by the formula (4-3) can be produced by the following method.
Figure 2005030773
[Wherein, R represents a protecting group for a carboxyl group such as a lower alkyl group, and ring A, R 3 , R 4 , R 5 Is as defined above. ]
For example, when R is a lower alkyl group, the compound of the formula (4-1) that can be produced by the above method is prepared by using water as a base in an alcohol solvent such as methanol or ethanol, or an ether solvent such as tetrahydrofuran or dioxane. A compound represented by the formula (4-3) can be produced by adding an aqueous sodium oxide solution or an aqueous potassium hydroxide solution and heating at room temperature or heating. Alternatively, the compound of formula (4-3) can be obtained by oxidizing the compound of formula (4-2). As a method of oxidizing alcohol to carboxylic acid, as described herein, Swern oxidation (J. Org. Chem., 43 , P2480 (1978), etc.), and then oxidized to carboxylic acid using sodium chlorite (Tetrahedron, 43 4767 (1987)) and a method of directly oxidizing an alcohol to a carboxylic acid. For the oxidation method, methods described in Laroc literature and the like can be referred to.
As R, it is also possible to use a protecting group that can be deprotected with an acid such as trifluoroacetic acid such as t-butyl group or 4N HCl / dioxane, or a protecting group that can be deprotected by a catalytic hydrogenation reaction such as a benzyl group. The protection / deprotection reaction may be performed in accordance with the method described in “Protective Groups in Organism Synthesis (TW Green et al., John Wiley & Sons, Inc. 1991)”.
Next, the manufacturing method of the compound of this invention which has a pyrazolopyridine skeleton is shown.
Figure 2005030773
[Wherein R 3 , R 4 , R 5 And R are as defined above. ]
Known compounds or known methods (J. Org. Chem., 59 , P2740 (1994)) by N-amination of the compound represented by the formula (5-1) according to a known method (Tetrahedron Lett., 40, p4133 (1972)) The compound represented by 2) can be produced. A compound of formula (5-2) and a commercially available acetylene, preferably di-t-butylacetylene, an alcohol solvent such as N, N-dimethylformamide or dimethyl sulfoxide, methanol or ethanol, or an ether such as tetrahydrofuran or dioxane. The compound represented by the formula (5-3) can be obtained by heating at room temperature or in the presence of a base in a system solvent, preferably in N, N-dimethylformamide. As the base, inorganic bases such as potassium carbonate, sodium carbonate, cesium carbonate, sodium hydroxide and potassium hydroxide, and organic bases such as triethylamine and N-methylmorpholine are used. Preferably potassium carbonate is used. Carboxylic acid represented by the formula (5-4) can be obtained by decarboxylation and hydrolysis of the compound represented by the formula (5-3) by heating at room temperature or under acidic conditions. As the acid, sulfuric acid, hydrochloric acid, trifluoroacetic acid and the like are used. More preferably, trifluoroacetic acid is used at room temperature. The compound of formula (5-4) can be converted to the compound of the present invention using the same method as described above.
When the compound of the present invention is produced, a functional group such as a hydroxyl group, a carboxyl group or an amino group can be protected / deprotected as required in any step. For the type of protecting group and the method of protection / deprotection, those well known to those skilled in the art may be used. For example, “Protective Groups in Organic Synthesis (written by TW Green et al., John Wiley & Sons, Inc.) .. 1991)). Moreover, when manufacturing the compound of this invention, functional group conversion etc. can be performed as needed at arbitrary processes. Specifically, halogenation reaction such as chlorination or bromination, substitution reaction, Wittig reaction and the like can be mentioned. For these, methods well-known to those skilled in the art may be used, and for example, reference may be made to Lalock literature.
Among the compounds of the present invention represented by the general formula (1), examples of the pharmaceutically acceptable salt of a compound having a functional group capable of forming a salt include alkali metals such as sodium salt and potassium salt. Salts, calcium salts, magnesium salts and other alkaline earth metal salts, zinc salts and other inorganic metal salts, triethylamine, triethanolamine, trihydroxymethylaminomethane and other organic salts, ammonium salts, hydrochlorides, hydrobromides, sulfates Inorganic salts such as phosphate, nitrate, and acetate, propionate, succinate, lactate, malate, tartrate, citrate, maleate, fumarate, methanesulfonate , Organic acid salts such as p-toluenesulfonate, benzenesulfonate or ascorbate. These salts can be obtained by a conventional method such as recrystallization after mixing the compound represented by the general formula (1) with an acid or a base.
The present invention also includes solvates such as a hydrate of the compound represented by the general formula (1) or a pharmaceutically acceptable salt, and an ethanol solvate. Furthermore, the present invention includes all tautomers, all stereoisomers such as optical isomers, etc. of the compound represented by the general formula (1), and crystal forms of all embodiments. These can be appropriately purified using methods such as silica gel column chromatography, HPLC, ion exchange chromatography, and recrystallization well known to those skilled in the art.
In order to obtain the optical isomer purely, an optical resolution method known to those skilled in the art may be used. Specifically, when the compound of the present invention or an intermediate thereof has a basic functional group, an optically active acid in an inert solvent (for example, monocarboxylic acids such as mandelic acid, N-benzyloxyalanine, lactic acid, Salts with tartaric acid, o-diisopropylidenetartaric acid, dicarboxylic acids such as malic acid, sulfonic acids such as camphorsulfonic acid, bromocamphorsulfonic acid) and the like. In addition, when the compound of the present invention or an intermediate thereof has an acidic substituent, a salt may be formed with an optically active amine (for example, organic amines such as α-phenethylamine, quinine, quinidine, cinchonidine, cinchonine, strychnine). it can. The temperature at which the salt is formed includes a range from room temperature to the boiling point of the solvent.
The compound represented by the general formula (1) or a pharmaceutically acceptable salt thereof is an allergic disease, respiratory disease, cardiovascular disease, nervous system disease, inflammatory disease, nerve, which is associated with hyperfunction of PAR2. It is useful as a therapeutic agent, preventive agent, or progression preventive agent for diseases such as inflammatory inflammatory diseases and skin diseases.
The diseases specifically include arthritis (including osteoarthritis, osteoarthritis, spondyloarthritis, gout arthritis, systemic lupus erythematosus, juvenile arthritis and rheumatoid arthritis), fever (rheumatic fever and influenza and other Viral infection-related fever), common cold, dysmenorrhea, menstrual cramps, inflammatory bowel disease, Crohn's disease, emphysema, acute respiratory distress syndrome, asthma, bronchitis, chronic obstructive pulmonary disease, Alzheimer's disease, organ Transplant toxicity, cachexia, allergic reaction, allergic contact hypersensitivity, cancer (eg, solid tumor cancers including colon cancer, breast cancer, lung cancer and prostate cancer; hematopoietic malignancies including leukemia and lymphoma; Hodgkin's disease; regeneration Aplastic anemia, skin cancer and familial adenoma polyposis), tissue ulcer, peptic ulcer, gastritis, localized enteritis, ulcerative colitis, diverticulitis, recurrent stomach Lesions, gastrointestinal bleeding, coagulation, anemia, synovitis, gout, ankylosing spondylitis, restenosis, periodontal disease, epidermolysis bullosa, osteoporosis, loosening of artificial joint implants, atherosclerosis (atherosclerotic platelets) Destruction), aortic aneurysm (abdominal aortic aneurysm and cerebral aortic aneurysm), nodular periarteritis, congestive heart failure, myocardial infarction, stroke, cerebral ischemia, head injury, spinal cord injury, neuralgia, neurodegenerative diseases (acute and chronic ), Autoimmune disease, Huntington's disease, Parkinson's disease, migraine, depression, peripheral neuropathy, pain (lower back and neck pain, headache and toothache), gingivitis, cerebral amyloid angiopathy, neutropic ) Or cognitive enhancement, amyotrophic side sclerosis, multiple sclerosis, eye angiogenesis, corneal damage, macular degeneration, conjunctivitis, abnormal wound healing, muscle or joint sprain Or tension, tendinitis, skin diseases (eg, psoriasis, eczema, scleroderma and dermatitis), myasthenia gravis, polymyositis, myositis, bursitis, burns, diabetes (type I and II diabetes, Diabetic retinopathy), tumor invasion, tumor growth, tumor metastasis, corneal scar, scleritis, immunodeficiency disease (eg, human AIDS, feline FLV, FIV), sepsis, premature birth, hypoprothrombinemia, hemophilia Disease, thyroiditis, sarcoidosis, Behcet's syndrome, hypersensitivity, kidney disease and the like.
Preferably, arthritis (including osteoarthritis, spondyloarthritis, gout arthritis, systemic lupus erythematosus, juvenile arthritis and rheumatoid arthritis), dermatitis, fever, asthma, bone resorption, cardiovascular disease, menstruation Diseases such as difficulty, premature birth, nephritis, nephrosis, atherosclerosis, hypotension, shock, or neurogenic inflammation from painful nerve tissue, cancer, and Alzheimer's disease.
In addition, the compound represented by the general formula (1) or a pharmaceutically acceptable salt thereof suppresses the release of PGE2 from cartilage or synovial cells and is also useful as a PGE2 production inhibitor.
Furthermore, the compound represented by the general formula (1), or a pharmaceutically acceptable salt thereof, exhibits excellent anti-pruritic activity in vivo as shown in the Examples of the present specification. It is also useful as an agent for treating or preventing various diseases. Specific examples of pruritus include eye pruritus, nasal pruritus, skin pruritus and generalized pruritus. In addition, diseases involving pruritus include atopic dermatitis, dermatitis pruritus, hives, contact dermatitis, psoriasis, dry skin, seborrheic dermatitis, neurodermatitis, self-sensitizing dermatitis, Caterpillar dermatitis, insect bite, photosensitivity, prurigo, eczema, pruritus on renal dialysis, senile pruritus, senile xerosis, flesh hypersensitivity, pruritus when opioid analgesics are administered, sebum deficiency Eczema, atopic keratoconjunctivitis, allergic keratoconjunctivitis, infectious keratoconjunctivitis, and pruritus associated with diseases such as spring catarrhism. In addition, the compounds of the present invention can be used for medical diseases (malignant tumors, diabetes, liver diseases, renal failure, gout, thyroid diseases, blood diseases), infectious diseases such as parasites, fungi, or viruses, psychogenic stresses, It is also effective as a preventive or therapeutic agent for pruritus caused by drug hypersensitivity or pregnancy. Furthermore, since the compounds of the present invention have PAR2 inhibitory activity, they are useful as research tools for investigating the physiological effects of PAR2.
Hereinafter, dosages and dosage forms in the case of using the therapeutic agent, prophylactic agent, progression inhibitor and the like for the above-mentioned diseases containing the compound of the present invention as an active ingredient will be described.
The compounds of the present invention can be administered orally or parenterally, preferably orally, in various dosage forms suitable for oral or parenteral administration as a pharmaceutical composition with appropriate additives, bases, carriers, human And used for non-human animals. For example, when administered orally, it can be administered in a commonly used dosage form such as tablets, capsules, syrups, suspensions and the like. When administered parenterally, solutions such as solutions, emulsions and suspensions are administered as injections or eye drops, administered rectally in the form of suppositories, and transdermal such as ointments, creams, lotions, etc. It can be administered as an agent. Such a dosage form can be produced according to a general method by blending an active ingredient such as a usual carrier, excipient, binder and stabilizer and an active ingredient. When used in an injection form, dissolve or suspend in a physiologically acceptable carrier such as water, saline, oil, or aqueous glucose solution, and use emulsifiers, stabilizers, osmotic pressure adjusting salts, dissolution aids as adjuvants. An agent or a buffer may be contained as necessary.
When administered as a transdermal agent, in addition to the base, if necessary, stabilizers, preservatives, emulsifiers, suspending agent stabilizers, antioxidants, perfumes, fillers, or other transdermal absorption enhancers. Etc. can be added. Examples of bases for ointments include fatty oils, lanolin, petrolatum, paraffin, plastibase, glycols, higher fatty acids, higher alcohols, etc. Examples of bases for lotions include ethanol, glycerin, glycols and the like. It is done. As the base of the liquid agent, for example, ethanol, water, glycol or the like is used.
The dose and frequency of administration vary depending on the target disease, the patient's symptoms, age, weight, etc., and the dosage form, but when administered orally, the active ingredient is usually about 1-1000 mg per day for an adult. A range, preferably about 10-500 mg can be administered in one or several divided doses. When administered as an injection, the active ingredient can be administered in the range of about 0.1 to about 500 mg, preferably in the range of about 3 to about 100 mg, in one or several divided doses.
Preferred examples of the compound represented by the general formula (1) include the compounds in Tables 1 and 2 below.
Figure 2005030773
Figure 2005030773
Figure 2005030773
Figure 2005030773
EXAMPLES Hereinafter, although an Example demonstrates this invention further in detail, the technical scope of this invention is not limited to these Examples.
Example 1 (Reference Example 1)
(Process 1)
(2E) -3-Cyano-2-hydrazonopropanoic acid ethyl ester
Figure 2005030773
Ethyl cyanopyruvate sodium enolate (7.8 g) was dissolved in acetic acid (40 ml) and ethanol (40 ml), and hydrazine monohydrate was added at 0 ° C. After 30 minutes, the mixture was brought to room temperature and stirred overnight. The reaction mixture was concentrated, water and chloroform were added, and the mixture was separated and extracted. The organic layer was dried and concentrated to obtain the desired product (5.0 g).
1 H-NMR (400 MHz, CDCl 3 ) Δ ppm: 1.34-1.41 (m, 3H), 3.46 (s), 3.63 (s), 4.26-4.38 (m, 2H), 6.52 (brs), 8.50 (br)
(Process 2)
Ethyl 5-phenyl-7- (trifluoromethyl) pyrazolo [1,5-a] pyrimidine-2-carboxylate
Figure 2005030773
The compound (5.0 g) obtained in Step 1 was dissolved in ethanol (50 ml), potassium carbonate (1.0 g) was added, and the mixture was heated to reflux. After 3 hours, 4,4,4-trifluoro-1-phenyl-1,3-butanedione (10.5 g) was added and heated to reflux. The resulting crystals were filtered and dried to obtain the desired product (4.0 g).
1 H-NMR (400 MHz, DMSO-d 6 ) Δppm: 1.37 (t, 3H, J = 7.1 Hz), 4.42 (q, 2H, J = 7.1 Hz), 7.44 (s, 1H), 7.57-7.65 ( m, 3H), 8.31-8.38 (m, 3H)
(Process 3)
5-phenyl-7- (trifluoromethyl) pyrazolo [1,5-a] pyrimidine-2-carboxylate potassium
Figure 2005030773
The compound obtained in Step 2 (4.6 g), ethanol (50 ml), 1N aqueous potassium hydroxide solution (15 ml) were mixed and heated to reflux. After 30 minutes, it was cooled to room temperature. The resulting crystals were filtered and dried to obtain the desired product (4.2 g).
1 H-NMR (400 MHz, DMSO-d 6 ) Δ ppm: 6.87 (s, 1H), 7.53-7.59 (m, 3H), 8.02 (s, 1H), 8.24-8.31 (m, 2H)
Example 2 (Reference Example 2)
(Process 1)
1- (4-Chlorophenyl) -4,4,4-trifluorobutane-1,3-dione
Figure 2005030773
4-Chloroacetophenone (4.6 ml) was dissolved in tetrahydrofuran (50 ml), cooled to 0 ° C., and sodium hydride (1.55 g; 60%) was added. After 15 minutes, the mixture was brought to room temperature and stirred for 30 minutes. The reaction was cooled to 0 ° C. and ethyl trifluoroacetate was added. After 2 hours, the temperature was raised to room temperature and the mixture was further stirred for 1.5 hours. 1N Hydrochloric acid was added to the reaction mixture, and the mixture was extracted with ethyl acetate. The organic layer was washed with water, dried and concentrated to obtain the desired product (10.6 g).
1 H-NMR (400 MHz, DMSO-d 6 ) Δ ppm: 7.02 (s, 1H), 7.66 (d, 2H, J = 8.7 Hz), 8.14 (d, 2H, J = 8.7 Hz)
(Process 2)
Ethyl 5- (4-chlorophenyl) -7- (trifluoromethyl) pyrazolo [1,5-a] pyrimidine-2-carboxylate
Figure 2005030773
Using the compound obtained in Step 1 and the compound obtained in Step 1 of Reference Example 1 (Example 1), the target product (4.3 g) was obtained in the same manner as in Step 2 of Reference Example 1.
1 H-NMR (400 MHz, CDCl 3 ) Δppm: 1.47 (t, 3H, J = 7.1 Hz), 4.51 (q, 2H, J = 7.1 Hz), 7.38 (s, 1H), 7.52-7.57 ( m, 2H), 7.71 (s, 1H), 8.07-8.12 (m, 2H)
(Process 3)
5- (4-Chlorophenyl) -7- (trifluoromethyl) pyrazolo [1,5-a] pyrimidine-2-carboxylate potassium
Figure 2005030773
The target product (3.5 g) was obtained from the compound obtained in Step 2 by the same method as in Step 3 of Reference Example 1.
Example 3 (Reference Example 3)
(Process 1)
2-Nitro-N-4-penten-1-ylbenzenesulfonamide
Figure 2005030773
2-Nitro-Nt-butoxycarbonylbenzenesulfonamide (5.0 g) was dissolved in N, N-dimethylformamide (50 ml), potassium carbonate (6.9 g), 5-bromo-1-pentene (2.2 ml). ) And heated at 80 ° C. After 4 hours, the reaction solution was poured into water and extracted with ethyl acetate. The organic layer was washed with water, dried and concentrated, and the residue was dissolved in trifluoroacetic acid (75 ml) and allowed to stand at room temperature for 2 hours. The reaction was concentrated and azeotroped with toluene. The product was dried under reduced pressure to obtain the desired product (3.6 g).
1 H-NMR (400 MHz, CDCl 3 ) Δ ppm: 1.65 (m, 2H), 2.09 (m, 2H), 3.12 (q, 2H, J = 6.9 Hz), 4.94-5.03 (m, 2H), 5 .29 (brt, 1H, J = 5.6 Hz), 5.72 (m, 1H), 7.72-7.78 (m, 2H), 7.87 (m, 1H), 8.14 (m , 1H)
(Process 2)
N- {2-[(benzyloxy) methyl] -2-propen-1-yl} -2-nitro-N-4-penten-1-ylbenzenesulfonamide
Figure 2005030773
The compound (1.8 g) obtained in Step 1 was dissolved in toluene (50 ml), and 2-[(benzyloxy) methyl] -2-propen-1-ol (1.4 g), triphenylphosphine (2.3 g) was dissolved. ) And diethyl azodicarboxylate (3.8 ml) were mixed and stirred at room temperature. After 1 hour, the reaction solution was poured into water and extracted with ethyl acetate. The organic layer was dried and concentrated, and the residue was purified with a silica gel column (300 g, ethyl acetate: hexane = 1: 4 to 1: 3) to obtain the desired product (2.3 g).
1 H-NMR (400 MHz, CDCl 3 ) Δ ppm: 1.58 (m, 2H), 1.96 (m, 2H), 3.29 (m, 2H), 3.91 (s, 2H), 4.01 (s, 2H), 4. 42 (s, 2H), 4.91-5.00 (m, 2H), 5.17 (s, 1H), 5.26 (s, 1H), 5.68 (m, 1H), 7.26 -7.38 (m, 5H), 7.56-7.67 (m, 3H), 8.02 (m, 1H)
(Process 3)
6-[(Benzyloxy) methyl] -1-[(2-nitrophenyl) sulfonyl] -2,3,4,7-tetrahydro-1H-azepine
Figure 2005030773
The compound obtained in Step 2 (2.3 g) was dissolved in dichloromethane (400 ml), the second generation Grubbs catalyst (0.1 g) was mixed, and the reaction solution was refluxed. After 1 hour, the reaction solution was concentrated, and the residue was purified by a silica gel column (150 g, ethyl acetate: hexane = 1: 2) to obtain the desired product (2.0 g).
1 H-NMR (400 MHz, CDCl 3 ) Δ ppm: 1.89 (m, 2H), 2.32 (m, 2H), 3.57 (t, 2H, J = 6.1 Hz), 3.96 (s, 2H), 3.99 (s) , 2H), 4.48 (s, 2H), 5.85 (t, 1H, J = 5.7 Hz), 7.27-7.37 (m, 5H), 7.57-7.68 (m). , 3H), 7.98 (m, 1H)
(Process 4)
tert-Butyl 6-[(Benzyloxy) methyl] -2,3,4,7-tetrahydro-1H-azepine-1-carboxylate
Figure 2005030773
The compound (2.0 g) obtained in Step 3 was dissolved in N, N-dimethylformamide (50 ml), potassium carbonate (2.1 g) and benzenethiol (0.77 ml) were added, and the mixture was stirred at room temperature for 2 hours. Boc20 (2.2 g) was added and stirred for 1 hour. The reaction mixture was poured into water and extracted with ethyl acetate. The organic layer was washed with water, dried and concentrated, and the residue was purified with a silica gel column (200 g, ethyl acetate: hexane = 1: 10 to 1: 5) to obtain the desired product (1.5 g).
1 H-NMR (400 MHz, CDCl 3 ) Δ ppm: 1.38-1.46 (m, 4H), 1.75-1.84 (m, 2H), 2.18-2.25 (m, 2H), 3.46-3.62 ( m, 2H), 3.85-4.03 (m, 4H), 4.29 (s, 2H), 5.77 (t, 1H, J = 5.6 Hz), 7.26-7.38 ( m, 5H)
(Process 5)
Azepan-3-ylmethanol hydrochloride
Figure 2005030773
The compound obtained in Step 4 (1.5 g) was dissolved in ethanol (50 ml), 10% Pd—C was added, and the mixture was stirred under a hydrogen atmosphere for 5 hours. The reaction solution was filtered through celite and washed with ethanol. The filtrate was concentrated and dried, and 4N hydrochloric acid-dioxane solution (10 ml) was added to the residue. After stirring at room temperature for 2 hours, the reaction solution was concentrated. It dried under reduced pressure and obtained the target object (0.77g).
1 H-NMR (400 MHz, DMSO-d 6 ) Δ ppm: 1.24 (m, 1H), 1.47 (m, 1H), 1.63-1.99 (m, 5H), 2.75 (m, 1H), 2.98-3.35 (M, 5H), 4.86 (brs, 1H), 9.04 (brs, 1H), 9.23 (brs, 1H)
Example 4 (Reference Example 4)
(Process 1)
Ethyl 2-{[(2-nitrophenyl) sulfonyl] amino} -4-pentenoate
Figure 2005030773
Ethanol (20 ml) was cooled to 0 ° C. and thionyl chloride (4.4 ml) was added dropwise. After 30 minutes, allyl glycine (2.0 g) was added. After stirring for 30 minutes, the mixture was raised to room temperature and stirred as it was overnight. The reaction solution was concentrated and dried under reduced pressure. The obtained crude product was dissolved in chloroform (40 ml), o-nitrobenzenesulfonyl chloride (4.2 g) and triethylamine (5.2 ml) were added and stirred at 0 ° C. Three hours later, water and chloroform were added, followed by liquid separation and extraction. The organic layer was dried and concentrated, and the residue was purified by a silica gel column (200 g, ethyl acetate: hexane = 1: 2) to obtain an ester (5.7 g).
1 H-NMR (400 MHz, CDCl 3 ) Δ ppm: 1.11 (t, 3H, J = 7.1 Hz), 2.58 (m, 2H), 3.97 (q, 2H, J = 7.1 Hz), 4.28 (dt, 1H, J = 8.8, 5.9 Hz), 5.12-5.19 (m, 2H), 5.69 (m, 1H), 6.11 (brd, 1H, J = 8.8 Hz), 7. 70-7.76 (m, 2H), 7.93 (m, 1H), 8.08 (m, 1H)
(Process 2)
Ethyl 2- {3-buten-1-yl [(2-nitrophenyl) sulfonyl] amino} -4-pentenoate
Figure 2005030773
The target product (1.9 g) was obtained in the same manner as in Step 2 of Reference Example 3 (Example 3).
1 H-NMR (400 MHz, CDCl 3 ) Δ ppm: 1.12 (t, 3H, J = 7.2 Hz), 2.38 (m, 1H), 2.46-2.59 (m, 2H), 2.83 (m, 1H), 3 .21 (m, 1H), 3.49 (m, 1H), 4.04 (m, 2H), 4.71 (dd, 1H, J = 9.2, 6.0 Hz), 5.03-5 .24 (m, 4H), 5.67-5.88 (m, 2H), 7.59 (m, 1H), 7.69 (m, 2H), 8.06 (m, 1H)
(Process 3)
Ethyl 1-[(2-nitrophenyl) sulfonyl] -2,3,6,7-tetrahydro-1H-azepine-2-carboxylate
Figure 2005030773
The target product (1.5 g) was obtained in the same manner as in Step 3 of Reference Example 3 (Example 3).
1 H-NMR (400 MHz, CDCl 3 ) Δ ppm: 1.16 (t, 3H, J = 7.1 Hz), 2.35-2.45 (m, 2H), 2.65 (m, 1H), 2.87 (m, 1H), 3 .48 (m, 1H), 3.83 (m, 1H), 4.03-4.15 (m, 2H), 4.92 (dd, 1H, J = 6.3, 3.8 Hz), 5 .70-5.85 (m, 2H), 7.60-7.72 (m, 3H), 8.10 (m, 1H)
(Process 4)
1-benzyl 2-ethyl 2,3,6,7-tetrahydro-1H-azepine-1,2-dicarboxylate
Figure 2005030773
(In the formula, Z represents a benzyloxycarbonyl group.)
The cyclized product (0.55 g) obtained in step 3 was dissolved in N, N-dimethylformamide (20 ml), potassium carbonate (0.64 g) and benzenethiol (0.24 ml) were added at room temperature, and the mixture was stirred for 45 minutes. did. Benzyl chloroformate (0.44 ml) was added dropwise and stirred for 1 hour. Water and ethyl acetate were added to the reaction solution, followed by liquid separation and extraction. The organic layer was washed with water, dried and concentrated, and the residue was purified with a silica gel column (100 g, ethyl acetate: hexane = 1: 6) to obtain the desired product (0.42 g).
1 H-NMR (400 MHz, CDCl 3 ) Δ ppm: 1.12-1.28 (m, 3H), 2.29-2.80 (m, 4H), 3.65-4.22 (m, 3H), 4.64-4.82 ( m, 1H), 5.04-5.22 (m, 2H), 5.55-5.68 (m, 2H), 7.27-7.38 (m, 5H)
(Process 5)
Ethyl azepan-2-carboxylate
Figure 2005030773
The compound (0.42 g) obtained in Step 4 was dissolved in ethanol (15 ml), 10% palladium hydroxide was added, and the mixture was vigorously stirred at room temperature for 3 hours in a hydrogen atmosphere. The reaction solution was filtered through Celite, and the filtrate was concentrated and dried under reduced pressure to obtain the desired product (0.2 g).
1 H-NMR (400 MHz, CDCl 3 ) Δ ppm: 1.27 (t, 3H, J = 7.1 Hz), 1.54-1.78 (m, 7H), 2.09 (m, 1H), 2.72 (dt, 1H, J = 14.2, 5.9 Hz), 3.05 (dt, 1H, J = 14.2, 5.0 Hz), 3.49 (dd, 1H, J = 9.3, 5.0 Hz), 4.17 (Q, 2H, J = 7.1Hz)
(Step 6)
2-({[tert-Butyl (dimethyl) silyl] oxy} methyl) -1-[(2-nitrophenyl) sulfonyl] -2,3,6,7-tetrahydro-1H-azepine
Figure 2005030773
The compound (1.45 g) obtained in Step 3 was dissolved in dichloromethane (50 ml), cooled to −78 ° C., and diisobutylaluminum hydride (DIBAL) (0.93 M in hexane, 9.5 ml) was added dropwise. After 30 minutes, the temperature was raised to 0 ° C., and after 2 hours, DIBAL (3 ml) was added. After 30 minutes, methanol was added to stop the reaction, and the reaction solution was poured into an aqueous solution of NaK tartrate (30 g) and stirred at room temperature for 2 hours. The organic layer was extracted, dried and concentrated to obtain a crude product (1.2 g). The crude product (1.0 g) was dissolved in N, N-dimethylformamide (20 ml), and t-butyldimethylchlorosilane (0.63 g) and imidazole (0.29 g) were added at room temperature. After stirring for 1 hour, the reaction solution was poured into water, and ethyl acetate was added for separation and extraction. The organic layer was washed with water, dried and concentrated, and the residue was purified with a silica gel column (80 g, ethyl acetate: hexane = 1: 4) to obtain the desired product (1.25 g).
1 H-NMR (400 MHz, CDCl 3 ) Δ ppm: 2.28-2.64 (m, 4H), 3.32 (m, 1H), 3.53 (d, 1H, J = 11.8, 5.3 Hz), 3.77 (d, 1H, J = 11.8, 8.4 Hz), 3.86 (dt, 1H, J = 15.0, 4.2 Hz), 4.14 (m, 1H), 5.64 (m, 1H), 5.75 (m, 1H), 7.60-7.73 (m, 3H), 8.12 (m, 1H)
(Step 7)
2-({[tert-Butyl (dimethyl) silyl] oxy} methyl) azepane
Figure 2005030773
The compound (1.25 g) obtained in Step 6 was dissolved in N, N-dimethylformamide (20 ml), mercaptoacetic acid (0.41 ml) and lithium hydroxide (0.28 g) were added, and the mixture was stirred at room temperature. After 1 hour, the reaction solution was poured into a saturated aqueous solution of sodium bicarbonate and extracted with ethyl acetate. The organic layer was washed with saturated aqueous sodium hydrogen carbonate solution and saturated brine, dried and concentrated to give a crude product (0.71 g). The obtained crude product (0.36 g) was dissolved in ethanol (10 ml), 10% Pd—C (0.1 g) was added, and the mixture was stirred under a hydrogen atmosphere for 6 hours. The reaction solution was filtered through Celite, and the filtrate was concentrated and dried under reduced pressure to obtain the desired product (0.32 g).
1 H-NMR (400 MHz, CDCl 3 ) Δ ppm: 0.05 (s, 6H), 0.89 (s, 9H), 1.23 (m, 1H), 1.49-1.77 (m, 7H), 2.63-2.78 (M, 2H), 3.08 (m, 1H), 3.37 (dd, 1H, J = 9.8, 8.5 Hz), 3.50 (dd, 1H, J = 9.8, 4. 5Hz)
Example 5 (Reference Example 5)
1-[(2-Nitrophenyl) sulfonyl] azocan-4,5-diol
Figure 2005030773
1-[(2-Nitrophenyl) sulfonyl] -1,2,3,4,7,8-hexahydroazocine (0.1 g), which can be produced by the same method as in the above example, was added to acetonitrile (4 ml), water (1 ml), N-methylmorpholine-N-oxide (0.08 g) and osmium tetroxide (10%, 0.09 g) were added, and the mixture was stirred at room temperature for 3 days. The reaction solution was filtered, and water and ethyl acetate were added to the filtrate to separate and extract. The organic layer was washed with water, dried and concentrated to obtain the desired product (0.11 g).
1 H-NMR (400 MHz, CDCl 3 ) Δ ppm: 1.42-1.87 (m, 6H), 3.07-3.20 (m, 2H), 3.28-3.48 (m, 2H), 3.83 (m, 2H) 4.51 (d, 1H, J = 4.6 Hz), 4.80 (d, 1H, J = 5.2 Hz), 7.81-8.02 (m, 4H)
Example 6 (Reference Example 6)
Benzyl 4,5-dihydroxyazepane-1-carboxylate
Figure 2005030773
After adding 30% hydrogen peroxide (0.14 ml) and formic acid (0.70 ml) to tetrahydrofuran (16 ml) and cooling to 0 ° C., benzyl 2,3,6,7- Tetrahydro-1H-azepine-1-carboxylate (0.27 g) was added. After stirring overnight, an aqueous sodium thiosulfate solution was added to the reaction solution and concentrated. 13N Aqueous sodium hydroxide solution (3 ml) was added, and the mixture was stirred at room temperature for 2 hr. Water and ethyl acetate were added to separate and extract. The organic layer was washed with water, dried and concentrated to obtain the desired product (0.25 g).
1 H-NMR (400 MHz, CDCl 3 ) Δ ppm: 1.62-1.78 (m, 2H), 1.99-2.13 (m, 2H), 2.80 (brs, 2H), 3.23-3.70 (m, 6H) , 5.12 (s, 2H), 7.26-7.40 (m, 5H)

1−{[5−フェニル−7−(トリフルオロメチル)ピラゾロ[1,5−a]ピリミジン−2−イル]カルボニル}アゾカン−4−オール
1−{[5−フェニル−7−(トリフルオロメチル)ピラゾロ[1,5−a]ピリミジン−2−イル]カルボニル}アゾカン−3−オール

Figure 2005030773
上記実施例と同様の方法で製造できる1−[(2−ニトロフェニル)スルフォニル]−1,2,3,4,5,8−ヘキサヒドロアゼシン(0.5g)をテトラヒドロフラン(10ml)に溶かし、0℃でボラン(テトラヒドロフラン溶液、1.15M、2.2ml)を加えた。2時間攪拌後、水を加え反応を停止し、3N水酸化ナトリウム水溶液(6ml)、30%過酸化水素水(3ml)を加え、室温で5時間攪拌した。酢酸エチル、水を加え、分液、抽出した。有機層をチオ硫酸ナトリウム水溶液、水、飽和食塩水で洗浄後、乾燥、濃縮し、粗生成物(0.56g)を得た。N,N−ジメチルホルムアミド(10ml)に溶かし、室温で、炭酸カリウム(0.7g)、ベンゼンチオール(0.26ml)を加え、40分攪拌した。反応液をセライトろ過し、ろ液に参考例1(実施例1)で得られる化合物(0.59g)、WSCI(0.39g)、HOBt(0.46g)を加え、一晩攪拌した。反応液を飽和塩化アンモニウム水溶液に流し込み、酢酸エチルで抽出した。有機層を飽和炭酸水素ナトリウム水溶液、水、飽和食塩水で洗浄後、乾燥、濃縮した。残渣をシリカゲルカラムで精製し、3−OH体(0.38g)、4−OH体(0.07g)を得た。
4−OH体
H−NMR(400MHz,CDCl)δppm:1.44−2.26(m,8H),3.30−4.37(m,5H),7.25(m,1H),7.54−7.60(m,3H),7.68−7.72(m,1H),8.10−8.16(m,2H)
3−OH体
H−NMR(400MHz,CDCl)δppm:1.45−1.97(m,8H),3.22−4.50(m,5H),7.25(m,1H),7.54−7.60(m,3H),7.69−7.74(m,1H),8.10−8.17(m,2H)1-{[5-phenyl-7- (trifluoromethyl) pyrazolo [1,5-a] pyrimidin-2-yl] carbonyl} azocan-4-ol 1-{[5-phenyl-7- (trifluoromethyl) ) Pyrazolo [1,5-a] pyrimidin-2-yl] carbonyl} azocan-3-ol
Figure 2005030773
1-[(2-Nitrophenyl) sulfonyl] -1,2,3,4,5,8-hexahydroazesin (0.5 g), which can be produced by the same method as in the above example, was dissolved in tetrahydrofuran (10 ml). Borane (tetrahydrofuran solution, 1.15M, 2.2 ml) was added at 0 ° C. After stirring for 2 hours, water was added to stop the reaction, 3N aqueous sodium hydroxide solution (6 ml) and 30% aqueous hydrogen peroxide (3 ml) were added, and the mixture was stirred at room temperature for 5 hours. Ethyl acetate and water were added, and the solution was separated and extracted. The organic layer was washed with aqueous sodium thiosulfate solution, water and saturated brine, dried and concentrated to give a crude product (0.56 g). Dissolved in N, N-dimethylformamide (10 ml), potassium carbonate (0.7 g) and benzenethiol (0.26 ml) were added at room temperature and stirred for 40 minutes. The reaction solution was filtered through Celite, and the compound (0.59 g), WSCI (0.39 g) and HOBt (0.46 g) obtained in Reference Example 1 (Example 1) were added to the filtrate and stirred overnight. The reaction solution was poured into a saturated aqueous ammonium chloride solution and extracted with ethyl acetate. The organic layer was washed with saturated aqueous sodium hydrogen carbonate solution, water and saturated brine, dried and concentrated. The residue was purified by a silica gel column to obtain a 3-OH form (0.38 g) and a 4-OH form (0.07 g).
4-OH body
1 H-NMR (400 MHz, CDCl 3 ) δ ppm: 1.44-2.26 (m, 8H), 3.30-4.37 (m, 5H), 7.25 (m, 1H), 7.54 -7.60 (m, 3H), 7.68-7.72 (m, 1H), 8.10-8.16 (m, 2H)
3-OH body
1 H-NMR (400 MHz, CDCl 3 ) δ ppm: 1.45-1.97 (m, 8H), 3.22-4.50 (m, 5H), 7.25 (m, 1H), 7.54 -7.60 (m, 3H), 7.69-7.74 (m, 1H), 8.10-8.17 (m, 2H)

(1−{[5−(4−クロロフェニル)−7−(トリフルオロメチル)ピラゾロ[1,5−a]ピリミジン−2−イル]カルボニル}アゼパン−3−イル)メタノール

Figure 2005030773
参考例3(実施例3)で得た化合物(0.74g)をN,N−ジメチルホルムアミド(15ml)に溶かし、N−ヒドロキシベンゾトリアゾール(0.41g)、参考例2(実施例2)で得たアミン(0.40g)、1−エチル−3−(3’−ジメチルアミノプロピル)カルボジイミド(0.46g)を加え、室温で一晩攪拌した。反応液に、水、酢酸エチルを加え、分液、抽出した。有機層を、飽和塩化アンモニウム水溶液、飽和重曹水、水、飽和食塩水で洗浄、硫酸ナトリウムで乾燥後、濃縮した。残渣をジイソプロピルエーテルで洗浄、ろ過、乾燥し、目的物(0.71g)を得た。
H−NMR(400MHz,CDCl)δppm:1.33−2.20(m,7H),3.30−3.65(m,4H),4.05−4.43(m,2H),7.22−7.25(m,1H),7.54(d,2H,J=8.5Hz),7.62−7.67(m,1H),8.09(d,2H,J=8.5Hz)(1-{[5- (4-Chlorophenyl) -7- (trifluoromethyl) pyrazolo [1,5-a] pyrimidin-2-yl] carbonyl} azepan-3-yl) methanol
Figure 2005030773
The compound (0.74 g) obtained in Reference Example 3 (Example 3) was dissolved in N, N-dimethylformamide (15 ml), and N-hydroxybenzotriazole (0.41 g) and Reference Example 2 (Example 2) were used. The obtained amine (0.40 g) and 1-ethyl-3- (3′-dimethylaminopropyl) carbodiimide (0.46 g) were added, and the mixture was stirred overnight at room temperature. Water and ethyl acetate were added to the reaction solution, and the solution was separated and extracted. The organic layer was washed with saturated aqueous ammonium chloride solution, saturated aqueous sodium hydrogen carbonate, water and saturated brine, dried over sodium sulfate, and concentrated. The residue was washed with diisopropyl ether, filtered and dried to obtain the desired product (0.71 g).
1 H-NMR (400 MHz, CDCl 3 ) δ ppm: 1.33 to 2.20 (m, 7H), 3.30 to 3.65 (m, 4H), 4.05 to 4.43 (m, 2H) 7.22-7.25 (m, 1H), 7.54 (d, 2H, J = 8.5 Hz), 7.62-7.67 (m, 1H), 8.09 (d, 2H, J = 8.5Hz)

(1−{[5−フェニル−7−(トリフルオロメチル)ピラゾロ[1,5−a]ピリミジン−2−イル]カルボニル}アゼパン−2−イル)メタノール

Figure 2005030773
実施例8に従い縮合後、生成物(0.41g)をテトラヒドロフラン(10ml)に溶かし、室温でテトラブチルアンモニウムフルオライド(1M−テトラヒドロフラン溶液、1.0ml)を加えた。1時間後、反応液に飽和塩化アンモニウム水溶液中に流し込み、酢酸エチルで分液、抽出した。有機層を水洗、乾燥、濃縮し、残渣をジイソプロピルエーテルーエタノールから結晶化し、ろ過、乾燥し、目的物(0.21g)を得た。
H−NMR(400MHz,CDCl)δppm:1.20−1.55(m,3H),1.76−2.20(m,5H),2.90−3.25(m,1H),3.60−3.85(m,2H),4.23−4.38(m,1H),4.44−4.84(m,1H),7.28−7.32(m,1H),7.54−7.60(m,2H),7.69−7.73(m,1H),8.10−8.15(m,2H)(1-{[5-Phenyl-7- (trifluoromethyl) pyrazolo [1,5-a] pyrimidin-2-yl] carbonyl} azepan-2-yl) methanol
Figure 2005030773
After condensation according to Example 8, the product (0.41 g) was dissolved in tetrahydrofuran (10 ml), and tetrabutylammonium fluoride (1M-tetrahydrofuran solution, 1.0 ml) was added at room temperature. After 1 hour, the reaction solution was poured into a saturated aqueous solution of ammonium chloride, and separated and extracted with ethyl acetate. The organic layer was washed with water, dried and concentrated, and the residue was crystallized from diisopropyl ether-ethanol, filtered and dried to obtain the desired product (0.21 g).
1 H-NMR (400 MHz, CDCl 3 ) δ ppm: 1.20-1.55 (m, 3H), 1.76-2.20 (m, 5H), 2.90-3.25 (m, 1H) 3.60-3.85 (m, 2H), 4.23-4.38 (m, 1H), 4.44-4.84 (m, 1H), 7.28-7.32 (m, 1H), 7.54-7.60 (m, 2H), 7.69-7.73 (m, 1H), 8.10-8.15 (m, 2H)

エチル 1−{[5−(4−クロロフェニル)−7−(トリフルオロメチル)ピラゾロ[1,5−a]ピリミジン−2−イル]カルボニル}ピペリジン−2−カルボキシレート

Figure 2005030773
参考例2(実施例2)で得た化合物とピペコリン酸エチル塩酸塩から、実施例8と同様の方法により目的物(40g)を得た。
H−NMR(400MHz,CDCl)δppm:1.26−1.35(m,3H),1.36−1.90(m,5H),2.27−2.42(m,1H),3.00−3.40(m,1H),4.20−4.33(m,2H),4.50−4.80(m,1H),5.50−5.58(m,1H),7.22−7.26(m,1H),7.50−7.56(m,2H),7.62−7.66(m,1H),8.06−8.12(m,2H)Ethyl 1-{[5- (4-chlorophenyl) -7- (trifluoromethyl) pyrazolo [1,5-a] pyrimidin-2-yl] carbonyl} piperidine-2-carboxylate
Figure 2005030773
From the compound obtained in Reference Example 2 (Example 2) and ethyl pipecolic acid hydrochloride, the target product (40 g) was obtained in the same manner as in Example 8.
1 H-NMR (400 MHz, CDCl 3 ) δ ppm: 1.26-1.35 (m, 3H), 1.36-1.90 (m, 5H), 2.27-2.42 (m, 1H) , 3.00-3.40 (m, 1H), 4.20-4.33 (m, 2H), 4.50-4.80 (m, 1H), 5.50-5.58 (m, 1H), 7.22-7.26 (m, 1H), 7.50-7.56 (m, 2H), 7.62-7.66 (m, 1H), 8.06-8.12 ( m, 2H)

1−{[5−(4−クロロフェニル)−7−(トリフルオロメチル)ピラゾロ[1,5−a]ピリミジン−2−イル]カルボニル}ピペリジン−2−カルボン酸

Figure 2005030773
実施例10の化合物(40g)をエタノール(500ml)に溶かし、1N水酸化ナトリウム水溶液を100ml加え、70℃で1時間加熱攪拌した。水を加えエタノールを濃縮後、エーテルを加え、分液、抽出した。水層を1N塩酸で酸性にし、酢酸エチルで抽出した。有機層を水洗、乾燥、濃縮し、生じた結晶をジイソプロピルエーテルでリバルブ洗浄し、目的物(32.5g)を得た。
H−NMR(400MHz,CDCl)δppm:H−NMR(400MHz,DMSO−d6)δppm:1.29−1.78(m,5H),2.10−2.28(m,1H),2.85−3.28(m,1H),4.10−4.53(m,1H),5.02−5.23(m,1H),7.17(m,1H),7.63−7.69(m,2H),8.26−8.40(m,3H)1-{[5- (4-Chlorophenyl) -7- (trifluoromethyl) pyrazolo [1,5-a] pyrimidin-2-yl] carbonyl} piperidine-2-carboxylic acid
Figure 2005030773
The compound of Example 10 (40 g) was dissolved in ethanol (500 ml), 100 ml of 1N aqueous sodium hydroxide solution was added, and the mixture was heated with stirring at 70 ° C. for 1 hour. After adding water and concentrating ethanol, ether was added, followed by liquid separation and extraction. The aqueous layer was acidified with 1N hydrochloric acid and extracted with ethyl acetate. The organic layer was washed with water, dried and concentrated, and the resulting crystals were revalve washed with diisopropyl ether to obtain the desired product (32.5 g).
1 H-NMR (400 MHz, CDCl 3 ) δ ppm: 1 H-NMR (400 MHz, DMSO-d6) δ ppm: 1.29-1.78 (m, 5H), 2.10-2.28 (m, 1H) 2.85-3.28 (m, 1H), 4.10-4.53 (m, 1H), 5.02-5.23 (m, 1H), 7.17 (m, 1H), 7 .63-7.69 (m, 2H), 8.26-8.40 (m, 3H)

1−{[5−(4−クロロフェニル)−7−(トリフルオロメチル)ピラゾロ[1,5−a]ピリミジン−2−イル]カルボニル}アゼパン−3−カルボン酸

Figure 2005030773
ジクロロメタン(15ml)、塩化オキザリル(0.26ml)を混ぜ、−78℃に冷却した。ジメチルスルフォキシド(0.28ml)を滴下し、10分攪拌した。実施例8で得られたアルコール(0.67g)のジクロロメタン(10ml)溶液を滴下し30分攪拌後、トリエチルアミン(1.5ml)を加えた。20分後0℃に温度を上げ、さらに1時間攪拌した。反応液を飽和塩化アンモニウム水溶液に流し込み、酢酸エチルで抽出した。有機層を水洗、乾燥、濃縮し、粗生成物(0.70g)を得た。得られた粗生成物をジオキサン(20ml)、t−ブタノール(5ml)に溶かし、2−メチル−2−ブテン(0.5ml)リン酸二水素ナトリウム(0.64g)を加え、室温で亜塩素酸ナトリウム(0.51g)の水溶液(5ml)を滴下した。30分後、反応液に水中に流し込み酢酸エチルで抽出した。有機層を水洗、乾燥、濃縮し、残渣をジイソプロピルエーテルで結晶化した。ろ過、乾燥し、目的物(0.59g)を得た。
H−NMR(400MHz,CDCl)δppm:H−NMR(400MHz,DMSO−d6)δppm:1.40−2.00(m,6H),2.85(m,1H),3.20−3.54(m,2H),3.83−4.28(m,2H),7.23(s,1H),7.64−7.68(m,2H),8.27−8.40(m,3H)1-{[5- (4-Chlorophenyl) -7- (trifluoromethyl) pyrazolo [1,5-a] pyrimidin-2-yl] carbonyl} azepane-3-carboxylic acid
Figure 2005030773
Dichloromethane (15 ml) and oxalyl chloride (0.26 ml) were mixed and cooled to -78 ° C. Dimethyl sulfoxide (0.28 ml) was added dropwise and stirred for 10 minutes. A solution of the alcohol (0.67 g) obtained in Example 8 in dichloromethane (10 ml) was added dropwise and stirred for 30 minutes, and then triethylamine (1.5 ml) was added. After 20 minutes, the temperature was raised to 0 ° C., and the mixture was further stirred for 1 hour. The reaction solution was poured into a saturated aqueous ammonium chloride solution and extracted with ethyl acetate. The organic layer was washed with water, dried and concentrated to obtain a crude product (0.70 g). The obtained crude product was dissolved in dioxane (20 ml) and t-butanol (5 ml), 2-methyl-2-butene (0.5 ml) sodium dihydrogen phosphate (0.64 g) was added, and chlorine was added at room temperature. An aqueous solution (5 ml) of sodium acid (0.51 g) was added dropwise. After 30 minutes, the reaction solution was poured into water and extracted with ethyl acetate. The organic layer was washed with water, dried and concentrated, and the residue was crystallized with diisopropyl ether. Filtration and drying yielded the desired product (0.59 g).
1 H-NMR (400 MHz, CDCl 3 ) δ ppm: 1 H-NMR (400 MHz, DMSO-d 6) δ ppm: 1.40-2.00 (m, 6H), 2.85 (m, 1H), 3.20 -3.54 (m, 2H), 3.83-4.28 (m, 2H), 7.23 (s, 1H), 7.64-7.68 (m, 2H), 8.27-8 .40 (m, 3H)

5,7−ジフェニルピラゾロ[1,5−a]ピリジン−2−カルボン酸

Figure 2005030773
0−メシチレンスルフォニルヒドロキシアミン(2.0g)をジクロロメタン(20ml)に溶かし、0℃に冷却後、公知化合物であるピリジン(1.2g)を加えた。10分後、室温に上げ、さらに2時間攪拌した。反応液を濃縮、減圧乾燥後、N,N−ジメチルホルムアミド(20ml)に溶かし、室温で、炭酸カリウム(1.4g)、ジ−t−ブチルアセチレンカルボキシレート(2.3g)を加え一晩攪拌した。反応液に水、酢酸エチルを加え、分液、抽出し、有機層を水洗、乾燥、濃縮した。残渣を、シリカゲルカラム(200g、酢酸エチル:ヘキサン=1:5)で精製し、目的物とピリジンの混合物(1.0g)を得た。このまま、ジクロロエタン(10ml)に溶かし、トリフルオロ酢酸(5ml)を加え、5時間、65℃で加熱攪拌した。反応液を濃縮後、水、酢酸エチルを加え分液、抽出した。有機層を水洗、乾燥、濃縮し、残渣をシリカゲルカラム(40g、クロロホルム:メタノール=10:1)で精製し、カルボン酸(0.2g)を得た。 Rf=0.3
(クロロホルム:メタノール=10:1)5,7-Diphenylpyrazolo [1,5-a] pyridine-2-carboxylic acid
Figure 2005030773
0-mesitylensulfonylhydroxyamine (2.0 g) was dissolved in dichloromethane (20 ml), cooled to 0 ° C., and pyridine (1.2 g) as a known compound was added. After 10 minutes, the temperature was raised to room temperature and the mixture was further stirred for 2 hours. The reaction mixture was concentrated and dried under reduced pressure, then dissolved in N, N-dimethylformamide (20 ml), and potassium carbonate (1.4 g) and di-t-butylacetylenecarboxylate (2.3 g) were added at room temperature and stirred overnight. did. Water and ethyl acetate were added to the reaction solution, and the mixture was separated and extracted, and the organic layer was washed with water, dried and concentrated. The residue was purified by a silica gel column (200 g, ethyl acetate: hexane = 1: 5) to obtain a mixture (1.0 g) of the target product and pyridine. This was dissolved in dichloroethane (10 ml), trifluoroacetic acid (5 ml) was added, and the mixture was heated and stirred at 65 ° C. for 5 hours. The reaction mixture was concentrated, water and ethyl acetate were added, and the mixture was separated and extracted. The organic layer was washed with water, dried and concentrated, and the residue was purified with a silica gel column (40 g, chloroform: methanol = 10: 1) to obtain carboxylic acid (0.2 g). Rf = 0.3
(Chloroform: methanol = 10: 1)

1−[(5,7−ジフェニルピラゾロ[1,5−a]ピリジン−2−イル)カルボニル]ピペリジン−4−オール

Figure 2005030773
実施例13で得られたカルボン酸(0.04g)を用い、実施例8と同様の方法により目的物(0.04g)を得た。
1.54−1.69(m,2H),1.87−2.05(m,2H),3.39(m,1H),3.57(m,1H),3.99(m,1H),4.26(m,1H),4.38(m,1H),7.03(s,1H),7.20(d,1H,J=2.0Hz),7.39−7.56(m,6H),7.64−7.71(m,2H),7.75(d,1H,J=2.0Hz),7.92−7.98(m,2H)
上記実施例と同様な方法により、以下の表3〜表15に示す化合物を製造した。尚、上記実施例及び以下の表中の構造式において立体配置の表示がある場合、これらは相対的な立体配置を表しており、本明細書実施例で得た化合物はラセミ体である。
Figure 2005030773
Figure 2005030773
Figure 2005030773
Figure 2005030773
Figure 2005030773
Figure 2005030773
Figure 2005030773
Figure 2005030773
Figure 2005030773
Figure 2005030773
Figure 2005030773
Figure 2005030773
Figure 2005030773
1-[(5,7-Diphenylpyrazolo [1,5-a] pyridin-2-yl) carbonyl] piperidin-4-ol
Figure 2005030773
Using the carboxylic acid (0.04 g) obtained in Example 13, the target product (0.04 g) was obtained in the same manner as in Example 8.
1.54-1.69 (m, 2H), 1.87-2.05 (m, 2H), 3.39 (m, 1H), 3.57 (m, 1H), 3.99 (m, 1H), 4.26 (m, 1H), 4.38 (m, 1H), 7.03 (s, 1H), 7.20 (d, 1H, J = 2.0 Hz), 7.39-7 .56 (m, 6H), 7.64-7.71 (m, 2H), 7.75 (d, 1H, J = 2.0 Hz), 7.92-7.98 (m, 2H)
The compounds shown in Tables 3 to 15 below were produced in the same manner as in the above Examples. In addition, when there exists a display of a configuration in the structural formula in the said Example and the following table | surfaces, these represent relative configuration, and the compound obtained by this specification Example is a racemate.
Figure 2005030773
Figure 2005030773
Figure 2005030773
Figure 2005030773
Figure 2005030773
Figure 2005030773
Figure 2005030773
Figure 2005030773
Figure 2005030773
Figure 2005030773
Figure 2005030773
Figure 2005030773
Figure 2005030773

製剤例1:錠剤の製造
化合物37(20mg/錠剤)、乳糖(70mg/錠剤)、トウモロコシデンプン(17mg/錠剤)、低置換度ヒドロキシプロピルセルロース(8mg/錠剤)、ヒドロキシプロピルセルロース(4mg/錠剤)、及びステアリン酸マグネシウム(1mg/錠剤)を混合し、必要に応じて造粒した後、打錠することで、錠剤を製造することができる。
Formulation Example 1: Production of tablet Compound 37 (20 mg / tablet), lactose (70 mg / tablet), corn starch (17 mg / tablet), low-substituted hydroxypropylcellulose (8 mg / tablet), hydroxypropylcellulose (4 mg / tablet) , And magnesium stearate (1 mg / tablet) are mixed, granulated as necessary, and then tableted to produce a tablet.

製剤例2:錠剤の製造
化合物37(20mg/錠剤)、D−マンニトール(60mg/錠剤)、リン酸水素カルシウム(25mg/錠剤)、カルメロースカルシウム(8mg/錠剤)、ヒドロキシプロピルセルロース(4mg/錠剤)、及びタルク(3mg/錠剤)を混合し、必要に応じて造粒した後、打錠することで、錠剤を製造することができる。
Formulation Example 2: Tablet Production Compound 37 (20 mg / tablet), D-mannitol (60 mg / tablet), calcium hydrogen phosphate (25 mg / tablet), carmellose calcium (8 mg / tablet), hydroxypropylcellulose (4 mg / tablet) ) And talc (3 mg / tablet), granulated as necessary, and then tableted to produce a tablet.

製剤例3:注射用液剤
精製水(2mL)に、化合物37(200mg)およびエリスリトール(250mg)を溶解し、非経口投与用液剤を調製する。
Formulation Example 3: Injection Solution Compound 37 (200 mg) and erythritol (250 mg) are dissolved in purified water (2 mL) to prepare a solution for parenteral administration.

製剤例4:クリーム
化合物37(2g)にクロタミトン5g、ニッコール(TS−10)5g、流動パラフィン3g、ミリスチン酸イソプロピル15gを加え、70℃に加温して溶解する。これにカルボキシビニルポリマー1gを水60gに膨潤した溶液を加え、攪拌して乳化する。次に、ジイソプロパノールアミン0.5gを水9.75gに溶かした溶液を加え、均一になるまで攪拌して化合物37を有効成分として含有するクリームを得る。
Formulation Example 4: Cream To compound 37 (2 g), 5 g of crotamiton, 5 g of Nikkor (TS-10), 3 g of liquid paraffin, and 15 g of isopropyl myristate are added and heated to 70 ° C. to dissolve. A solution obtained by swelling 1 g of carboxyvinyl polymer in 60 g of water is added to this and emulsified by stirring. Next, a solution obtained by dissolving 0.5 g of diisopropanolamine in 9.75 g of water is added and stirred until uniform to obtain a cream containing compound 37 as an active ingredient.

製剤例5:油性軟膏
化合物37(10g)を精製水(30g)に溶解させ、ヘキシレングリコール(120g)と混合する。これを溶融させた白色ワセリン(700g)、白色ワックス(80g)とプロピレングリコールステアレート(20g)の混合物に添加し、温度を下げながら均質に攪拌して化合物37を有効成分として含有する軟膏を得る。
Formulation Example 5: Oily ointment Compound 37 (10 g) is dissolved in purified water (30 g) and mixed with hexylene glycol (120 g). This is added to a mixture of molten white petrolatum (700 g), white wax (80 g) and propylene glycol stearate (20 g), and stirred uniformly while lowering the temperature to obtain an ointment containing compound 37 as an active ingredient. .

Fluorometric Imaging Plate Reader(FLIPR)を用いた細胞内カルシウム濃度測定
[材料と方法]
牛胎児血清10%(10%FBS)添加DMEM培養液(インビトロジェン社)を用いて培養したヒト胎児腎臓由来細胞株HEK293細胞を2.5X10cells/mlに調製し、ポリDリジンでコートした96穴黒色プレート(透明底)(ファルコン社)に80μl/well播いて、炭酸ガス培養装置で一晩培養した。Hanks−20mM Hepes緩衝液(pH7.4)で調製したFLIPR Calcium Assay Kit(Molecular Devices社)を細胞に80μl/well添加して、1時間炭酸ガス培養装置にて培養した。Hanks−20mM Hepes緩衝液(pH7.4)で、0.0005%に調製したtrypsin(SIGMA社 T8642)を、96穴V底ポリプロピレンプレート(ヌンク社)に添加して試薬プレートとした。FLIPR(Molecular Devices社)で測定する直前に、Hanks−20mM Hepes緩衝液(pH7.4)で調製した被検化合物を、細胞に40μl/well添加し、プレートシェーカーで攪拌した。被検化合物を添加した細胞プレートと試薬プレートをFLIPRにセットし、細胞内カルシウム濃度の変化をCCDカメラにて検出した。測定は室温で70秒間行い、試薬プレートから細胞プレートへの試薬の添加(50μl/well)は、FLIPR本体内蔵の96well自動分注機で行った。
[結果]被検化合物の細胞内カルシウム上昇抑制率(%)を、表16に示した。

Figure 2005030773
Figure 2005030773
上記の結果から、本発明の化合物がPAR2阻害活性を示すことがわかる。Intracellular calcium concentration measurement using Fluorometric Imaging Plate Reader (FLIPR) [Materials and Methods]
Human fetal kidney-derived cell line HEK293 cells cultured in DMEM culture medium (Invitrogen) supplemented with fetal bovine serum 10% (10% FBS) were prepared to 2.5 × 10 5 cells / ml and coated with poly-D-lysine 96 80 μl / well was plated on a hole black plate (transparent bottom) (Falcon) and cultured overnight in a carbon dioxide culture apparatus. FLIPR Calcium Assay Kit (Molecular Devices) prepared with Hanks-20 mM Hepes buffer (pH 7.4) was added to the cells at 80 μl / well and cultured in a carbon dioxide culture apparatus for 1 hour. Trypsin (SIGMA T8642) adjusted to 0.0005% with Hanks-20 mM Hepes buffer (pH 7.4) was added to a 96-well V-bottom polypropylene plate (Nunk) to make a reagent plate. Immediately before measurement with FLIPR (Molecular Devices), a test compound prepared with Hanks-20 mM Hepes buffer (pH 7.4) was added to the cells at 40 μl / well and stirred with a plate shaker. The cell plate and reagent plate to which the test compound was added were set in the FLIPR, and changes in intracellular calcium concentration were detected with a CCD camera. The measurement was performed at room temperature for 70 seconds, and the addition of the reagent from the reagent plate to the cell plate (50 μl / well) was performed with a 96-well automatic dispenser with a built-in FLIPR main body.
[Results] Table 16 shows the inhibition rate (%) of intracellular calcium increase of the test compound.
Figure 2005030773
Figure 2005030773
From the above results, it can be seen that the compound of the present invention exhibits PAR2 inhibitory activity.

ヒト滑膜様細胞株SW982を用いたPGE2量の測定
SW982細胞を2.5x10細胞/mlになるように各培地で希釈し、96穴プレートに100μl/wellの細胞を播きこんだ。炭酸ガス培養装置内で一晩前培養した後、培地を除去し、5μg/ml化合物入り培地を80μl/well添加した(コントロール群、コントロールペプチド群、アゴニストペプチド単独群、IL−1β群には0.1%DMSO含有培地を添加した)。化合物を入れたウェルには 500μMアゴニストペプチド入り培地を20μl/well添加した(終濃度は4μg/ml化合物;100μMアゴニストペプチト)。コントロール群のウェルには各培地を20μl/well、コントロールペプチド群のウェルには500μMコントロールペプチド入り培地を20μl/well(終濃度100μM)、アゴニストペプチド単独群のウェルには500μMアゴニストペプチド入り培地を20μl/well(終濃度100μM)、IL−1β群のウェルには50ng/ml IL−1β入り培地を20μl/well(終濃度10ng/ml)の割合で添加した。24時間後(N=3)に各上清を回収し−80℃で測定時まで保管した。各上清中のPGE2量の測定は、プロスタグランジンE2 EIA(Cayman chemical 514010)を用いて行った。コントロールペプチドのPGE2量の値を阻害率100%、アゴニストペプチドのPGE2量を阻害率0%として、各化合物のPGE2産生阻害率を計算した。ヒト滑膜様細胞株SW982の各刺激によるPGE2産生量(pg/ml)を表17に示した。

Figure 2005030773
また、被験化合物のPGE2産生阻害率(%)を表18に示した。
Figure 2005030773
表17および表18の結果から、滑膜細胞のPAR2が活性化されると炎症・疼痛増悪因子であるPGE2の放出がおこること、また本発明により見出されたPAR2阻害剤は、これらのPAR2の活性化に伴って起こるPGE2の産生を阻害可能であることがわかる。Measurement of PGE2 amount using human synovial cell line SW982 SW982 cells were diluted with each medium to 2.5 × 10 5 cells / ml, and 100 μl / well of cells were seeded in a 96-well plate. After overnight preculture in a carbon dioxide culture apparatus, the medium was removed, and 80 μl / well of a medium containing 5 μg / ml compound was added (the control group, the control peptide group, the agonist peptide alone group, and the IL-1β group were 0. 1% DMSO-containing medium was added). To wells containing compounds, 20 μl / well of a medium containing 500 μM agonist peptide was added (final concentration: 4 μg / ml compound; 100 μM agonist peptide). 20 μl / well of each medium in wells of the control group, 20 μl / well of medium containing 500 μM control peptide in the wells of the control peptide group (final concentration 100 μM), and 20 μl of medium containing 500 μM agonist peptide in the wells of the agonist peptide alone group / Well (final concentration 100 μM), 50 ng / ml IL-1β-containing medium was added to the wells of the IL-1β group at a ratio of 20 μl / well (final concentration 10 ng / ml). After 24 hours (N = 3), each supernatant was collected and stored at −80 ° C. until measurement. The amount of PGE2 in each supernatant was measured using prostaglandin E2 EIA (Cayman chemical 51010). The inhibition rate of PGE2 production of each compound was calculated with the value of the PGE2 amount of the control peptide as 100% inhibition rate and the PGE2 amount of the agonist peptide as 0% inhibition rate. Table 17 shows the amount of PGE2 produced (pg / ml) by each stimulation of the human synovial cell line SW982.
Figure 2005030773
In addition, Table 18 shows the inhibition rate (%) of PGE2 production of the test compound.
Figure 2005030773
From the results of Table 17 and Table 18, when PAR2 of synovial cells is activated, PGE2, which is an inflammation / pain exacerbation factor, is released, and the PAR2 inhibitors found by the present invention are those PAR2 It can be seen that the production of PGE2 that occurs with the activation of can be inhibited.

PAR2の痒み惹起活性
7週齢の雄ICRマウスの肩背部皮内にアゴニストペプチドまたはコントロールペプチドを50pmol/20μl投与した。投与後10分間、後肢で投与部付近を引っかく行動を計測した(後肢が地面から離れて、再度戻るまでを1回と計測した)。結果を表19に示した。この結果から、アゴニストペプチド(配列番号:1)を投与した群は、コントロールペプチド(配列番号:2)を投与した群と比較して、著しく投与部位付近を引っかく回数が多いことがわかった。
更に、化合物1の化合物(200mg/kg)を経口で投与した後、投与化合物の血中濃度を考慮して30分後にアゴニストペプチドを投与した。上記と同様にアゴニストペプチド投与後10分間、後肢で投与部付近を引っかく行動を計測した。結果を表20に示した。この結果から、化合物1の化合物により、アゴニストペプチドにより惹起された引っかき行動が抑制されることがわかった。

Figure 2005030773
Figure 2005030773
PAR2 itch-inducing activity A 7-week-old male ICR mouse was administered with 50 pmol / 20 μl of agonist peptide or control peptide into the back of the shoulder. For 10 minutes after administration, the behavior was measured by scratching the vicinity of the administration part with the hind limb (measured once until the hind limb left the ground and returned again). The results are shown in Table 19. From this result, it was found that the group administered with the agonist peptide (SEQ ID NO: 1) had a remarkably large number of scratches around the administration site compared to the group administered with the control peptide (SEQ ID NO: 2).
Further, after the compound 1 (200 mg / kg) was orally administered, the agonist peptide was administered 30 minutes later in consideration of the blood concentration of the administered compound. Similarly to the above, for 10 minutes after administration of the agonist peptide, the behavior was measured by scratching the vicinity of the administration part with the hind limb. The results are shown in Table 20. From these results, it was found that the scratching behavior induced by the agonist peptide was suppressed by the compound of Compound 1.
Figure 2005030773
Figure 2005030773

Substance P誘発マウス痒み関連行動抑制作用
アトピー性皮膚炎患者の病変部では神経ペプチドであるSubstance P(SP)含有神経線維の増加(Tobin D et al.J Allergy Clin Immunol,90,613−22(1992))及びSPに対する反応性の増加(Gianetti A et al.Br J Dermatol,121,681−8(1989))が報告されている。一方、SPをマウス頚背部に投与すると痒み関連行動が誘発(Kuraishi Y et al.Eur J Pharmacol,275,229−33(1995))され、ある種の抗アレルギー剤で抑制されること(Inagaki N et al.Eur J Pharmacol,400,73−9(2000))が報告されている。そこで、本発明化合物の掻痒抑制作用を検証するため、SP誘発痒み関連行動に対する作用を指標として評価した。BALB/c系雄性マウスをあらかじめ観察用ケージに1匹ずつ移し、観察環境に慣れさせた後、頚背部皮下にSPを投与(200μg/mouse)し、投与後観察ケージに戻して、以後60分間の痒み関連行動を計測した。各被験薬物は0.5%MC溶液に懸濁し、20mg/kgと50mg/kgの用量でSP投与60分前に経口投与した。掻痒抑制作用の評価は0.5%MC溶液投与群に対する抑制率を指標とし、下記のように算出した。なお、比較例としては掻痒抑制作用を持つとされる抗アレルギー剤Oxatomideを50mg/kg投与した。

Figure 2005030773
その結果、表21に示す通り本発明化合物の掻痒抑制作用強度は抗アレルギー剤Oxatomideと同等又はそれ以上であることが判明した。
Figure 2005030773
Substance P-induced mouse itching-related behavior inhibitory effect Increased nerve fibers containing Substance P (SP), which is a neuropeptide, in lesions of patients with atopic dermatitis (Tobin D et al. J Allergy Clin Immunol, 90 , 613-22 (1992) )) And increased reactivity to SP (Gianetti A et al. Br J Dermatol, 121 , 681-8 (1989)) has been reported. On the other hand, when SP is administered to the back of the neck of a mouse, itching-related behavior is induced (Kuraishi Y et al. Eur J Pharmacol, 275 , 229-33 (1995)) and is inhibited by a certain antiallergic agent (Inagaki N et al. Eur J Pharmacol, 400 , 73-9 (2000)). Therefore, in order to verify the pruritus-inhibiting action of the compound of the present invention, the action on SP-induced itching-related behavior was evaluated as an index. BALB / c male mice were transferred to the observation cage one by one in advance and acclimatized to the observation environment. Then, SP was administered subcutaneously to the back of the neck (200 μg / mouse). The itch-related behavior was measured. Each test drug was suspended in a 0.5% MC solution and orally administered at a dose of 20 mg / kg and 50 mg / kg 60 minutes before SP administration. The pruritus inhibitory action was evaluated as follows using the inhibition rate for the 0.5% MC solution administration group as an index. In addition, as a comparative example, 50 mg / kg of an antiallergic agent Oxatomide, which is supposed to have an itching inhibitory action, was administered.
Figure 2005030773
As a result, as shown in Table 21, it was found that the pruritus inhibitory action strength of the compounds of the present invention was equivalent to or higher than that of the antiallergic agent Oxatomide.
Figure 2005030773

OVA誘発マウス2相性皮膚炎抑制作用
本発明のPAR−2アンタゴニストの炎症抑制作用を検証するため、ヒスタミン関与の強い即時型及び炎症性細胞の浸潤が強い遅発型の両反応を示すOVA誘発マウス2相性皮膚炎モデルにて評価した。ICR系雄性マウスに卵白アルブミン(OVA)1μgをAl(OH)1mgとともに腹腔内投与して感作を誘導した。感作14日目にOVA10μg(20μl/ear)を耳介内に直接投与してアレルギー反応を惹起した。比較として同容量の生理的食塩水を耳介内に投与した。被験薬物は0.5%MC水溶液に懸濁し、惹起の1時間前に経口投与(10mg/kg,50mg/kg)した。惹起1時間後及び24時間後に耳介厚を測定し、惹起前の耳介厚との差を算出した。皮膚炎抑制作用の評価は0.5%MC溶液投与群に対する抑制率を指標とし、即時型および遅発型ともに下記のように算出した。なお比較例として抗ヒスタミン作用を有する抗アレルギー薬であるオキサトミド(Oxatomide)を用い、0.5%MC水溶液に懸濁し、惹起の1時間前に経口投与した。惹起1時間後の耳介厚の増加はヒスタミンなどの化学伝達物質による浮腫であり、これを即時相と規定した。24時間後のそれは好酸球などの炎症性細胞の浸潤によるものであり、これを遅発相と規定した。

Figure 2005030773
その結果、表22に示す通り本発明の化合物は即時相及び遅発相の両方を抑制した。これに対して、抗ヒスタミン作用を有する抗アレルギー薬であるオキサトミドは、即時相は強く抑制したが、遅発相に対しては無効であった。
Figure 2005030773
OVA-induced mouse biphasic dermatitis inhibitory action In order to verify the inflammation inhibitory action of the PAR-2 antagonist of the present invention, an OVA-induced mouse exhibiting both an immediate type with strong histamine involvement and a delayed type with strong inflammatory cell infiltration Evaluation was performed using a biphasic dermatitis model. Sensitization was induced by intraperitoneally administering 1 μg of ovalbumin (OVA) together with 1 mg of Al (OH) 3 to ICR male mice. On the 14th day of sensitization, 10 μg of OVA (20 μl / ear) was directly administered into the auricle to induce an allergic reaction. For comparison, the same volume of physiological saline was administered into the auricle. The test drug was suspended in a 0.5% MC aqueous solution and orally administered (10 mg / kg, 50 mg / kg) 1 hour before induction. The thickness of the auricle was measured after 1 hour and 24 hours after induction, and the difference from the thickness of the auricle before induction was calculated. The evaluation of the dermatitis inhibitory action was calculated as follows for both the immediate type and the delayed type using the suppression rate for the 0.5% MC solution administration group as an index. As a comparative example, oxatomide (Oxatomide), an antiallergic agent having antihistamine action, was suspended in 0.5% MC aqueous solution and orally administered 1 hour before induction. The increase in auricular thickness 1 hour after induction was due to edema due to a chemical transmitter such as histamine, which was defined as an immediate phase. After 24 hours, it was due to infiltration of inflammatory cells such as eosinophils, which was defined as a late phase.
Figure 2005030773
As a result, as shown in Table 22, the compound of the present invention suppressed both the immediate phase and the delayed phase. In contrast, oxatomide, an antiallergic agent having antihistamine action, strongly suppressed the immediate phase but was ineffective against the late phase.
Figure 2005030773

本発明により、PAR2阻害活性を有し、アレルギー性疾患、皮膚疾患、又は疼痛もしくは掻痒を伴う疾患等の、種々の疾患の治療剤又は予防剤として有用な、ピラゾロピリジン化合物及びピラゾロピリミジン化合物を提供することが可能になった。  According to the present invention, pyrazolopyridine compounds and pyrazolopyrimidine compounds having PAR2 inhibitory activity and useful as a therapeutic or prophylactic agent for various diseases such as allergic diseases, skin diseases, or diseases accompanied by pain or pruritus It became possible to provide.

配列番号1:アゴニストペプチド
配列番号2:コントロールペプチド
SEQ ID NO: 1: Agonist peptide SEQ ID NO: 2: Control peptide

【配列表】

Figure 2005030773
[Sequence Listing]
Figure 2005030773

Claims (9)

一般式(1)
Figure 2005030773
〔式中、Rは水酸基、カルボキシル基、メルカプト基、アミノ基、置換アミノ基、または水酸基、カルボキシル基、メルカプト基、アミノ基もしくは置換アミノ基で置換されたアルキル基を表し、
mは1〜3の整数を表し、mが2以上の整数を表す場合Rは同一もしくは異なっていてもよく、
はハロゲン原子、アルキル基、アルケニル基、アルキニル基、シクロアルキル基、アルコキシ基、又はアルキルチオ基を表し、
nは0〜4の整数を表し、nが2以上の整数を表す場合Rは同一もしくは異なっていてもよく、
環Aは5〜8員の飽和もしくは不飽和含窒素複素環を表し、
は、水素原子、アルキル基、ハロアルキル基、または置換もしくは無置換のアリール基を表し、
およびRは、独立して、水素原子、ハロゲン原子、アルキル基、アルケニル基、アルキニル基、シクロアルキル基、アルコキシ基、アルキルチオ基またはハロアルキル基を表し、
XはCR(Rは水素原子またはアルキル基を表す)または窒素原子を表す。〕
で表される化合物、またはその薬学上許容される塩。
General formula (1)
Figure 2005030773
[Wherein R 1 represents a hydroxyl group, a carboxyl group, a mercapto group, an amino group, a substituted amino group, or an alkyl group substituted with a hydroxyl group, a carboxyl group, a mercapto group, an amino group, or a substituted amino group;
m represents an integer of 1 to 3, and when m represents an integer of 2 or more, R 1 may be the same or different;
R 2 represents a halogen atom, an alkyl group, an alkenyl group, an alkynyl group, a cycloalkyl group, an alkoxy group, or an alkylthio group,
n represents an integer of 0 to 4, and when n represents an integer of 2 or more, R 2 may be the same or different,
Ring A represents a 5- to 8-membered saturated or unsaturated nitrogen-containing heterocyclic ring,
R 3 represents a hydrogen atom, an alkyl group, a haloalkyl group, or a substituted or unsubstituted aryl group,
R 4 and R 5 independently represent a hydrogen atom, a halogen atom, an alkyl group, an alkenyl group, an alkynyl group, a cycloalkyl group, an alkoxy group, an alkylthio group or a haloalkyl group,
X represents CR 6 (R 6 represents a hydrogen atom or an alkyl group) or a nitrogen atom. ]
Or a pharmaceutically acceptable salt thereof.
式(1)において、Rがハロアルキル基である、請求項1に記載の化合物、またはその薬学上許容される塩。The compound according to claim 1, or a pharmaceutically acceptable salt thereof, wherein in formula (1), R 3 is a haloalkyl group. ハロアルキル基が、トリフルオロメチル基である、請求項2に記載の化合物、またはその薬学上許容される塩。The compound according to claim 2, or a pharmaceutically acceptable salt thereof, wherein the haloalkyl group is a trifluoromethyl group. 請求項1〜請求項3のいずれかに記載の化合物又はその薬学上許容される塩を有効成分として含有する、PAR2阻害剤。A PAR2 inhibitor comprising the compound according to any one of claims 1 to 3 or a pharmaceutically acceptable salt thereof as an active ingredient. 請求項1〜請求項3のいずれかに記載の化合物又はその薬学上許容される塩を有効成分として含有する、医薬組成物。A pharmaceutical composition comprising the compound according to any one of claims 1 to 3 or a pharmaceutically acceptable salt thereof as an active ingredient. 請求項1〜請求項3のいずれかに記載の化合物又はその薬学上許容される塩を有効成分として含有する、抗掻痒剤。The antipruritic agent which contains the compound in any one of Claims 1-3, or its pharmacologically acceptable salt as an active ingredient. 請求項1〜請求項3のいずれかに記載の化合物又はその薬学上許容される塩を有効成分として含有する、鎮痛剤。An analgesic comprising the compound according to any one of claims 1 to 3 or a pharmaceutically acceptable salt thereof as an active ingredient. 請求項1〜請求項3のいずれかに記載の化合物又はその薬学上許容される塩を有効成分として含有する、PGE2産生抑制剤。The PGE2 production inhibitor which contains the compound in any one of Claims 1-3, or its pharmacologically acceptable salt as an active ingredient. 請求項1〜請求項3のいずれかに記載の化合物又はその薬学上許容される塩を有効成分として含有する、抗炎症剤。An anti-inflammatory agent comprising the compound according to any one of claims 1 to 3 or a pharmaceutically acceptable salt thereof as an active ingredient.
JP2005514269A 2003-09-26 2004-09-22 New pyrazolopyrimidine derivatives Abandoned JPWO2005030773A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
JP2003334571 2003-09-26
JP2003334571 2003-09-26
PCT/JP2004/014322 WO2005030773A1 (en) 2003-09-26 2004-09-22 Novel pyrazolopyrimidine derivatives

Publications (1)

Publication Number Publication Date
JPWO2005030773A1 true JPWO2005030773A1 (en) 2006-12-07

Family

ID=34386033

Family Applications (1)

Application Number Title Priority Date Filing Date
JP2005514269A Abandoned JPWO2005030773A1 (en) 2003-09-26 2004-09-22 New pyrazolopyrimidine derivatives

Country Status (2)

Country Link
JP (1) JPWO2005030773A1 (en)
WO (1) WO2005030773A1 (en)

Families Citing this family (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1782859A3 (en) * 2003-04-11 2009-09-30 High Point Pharmaceuticals, LLC Pharmaceutical use of substituted pyrazolo [1,5- a]pyrimidines
US20070123508A1 (en) * 2005-05-27 2007-05-31 Roger Olsson PAR2-modulating compounds and their use
WO2007076055A2 (en) * 2005-12-22 2007-07-05 Entremed, Inc. Compositions and methods comprising proteinase activated receptor antagonists
BR112016006151A2 (en) * 2013-09-25 2017-08-01 Vertex Pharma imidazopyridazines useful as inhibitors of the par-2 signaling pathway
US9981976B2 (en) * 2014-12-08 2018-05-29 Janssen Sciences Ireland Uc Piperidine substituted pyrazolo[1,5-A]pyrimidine derivatives with inhibitory activity on the replication of the respiratory syncytial virus (RSV)
SG11201806393QA (en) * 2016-01-27 2018-08-30 Univ Zuerich Use of gabaa receptor modulators for treatment of itch
WO2018043461A1 (en) 2016-08-31 2018-03-08 エーザイ・アール・アンド・ディー・マネジメント株式会社 PYRAZOLO[1,5-a]PYRIMIDINE COMPOUND
WO2018057588A1 (en) * 2016-09-21 2018-03-29 Vertex Pharmaceuticals Incorporated Furo[3,2-b]pyridine compounds useful as inhibitors of the par-2 signaling pathway
EP3548035A4 (en) 2016-11-30 2020-07-22 Case Western Reserve University Combinations of 15-pgdh inhibitors with corcosteroids and/or tnf inhibitors and uses thereof
US11718589B2 (en) 2017-02-06 2023-08-08 Case Western Reserve University Compositions and methods of modulating short-chain dehydrogenase
RU2020125450A (en) * 2018-02-26 2022-03-28 Эйсай Ар Энд Ди Менеджмент Ко., Лтд. SALT OF PYRAZOLO[1,5-a] PYRIMIDINE COMPOUND AND ITS CRYSTALS
WO2019226820A1 (en) 2018-05-22 2019-11-28 Neurocycle Therapeutics, Inc. Gabaa positive allosteric modulator compounds for treatment of itch and/ or dermatitis
JP7150308B2 (en) * 2018-07-06 2022-10-11 国立大学法人京都大学 Composition for external use on the skin
US20240066027A1 (en) 2020-12-03 2024-02-29 Domain Therapeutics Novel par-2 inhibitors
WO2023233033A1 (en) 2022-06-03 2023-12-07 Domain Therapeutics Novel par-2 inhibitors

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6833371B2 (en) * 2001-11-01 2004-12-21 Icagen, Inc. Pyrazolopyrimidines
JP2003286171A (en) * 2002-03-28 2003-10-07 Sumitomo Pharmaceut Co Ltd Par inhibitor
JP2004170323A (en) * 2002-11-22 2004-06-17 Sumitomo Pharmaceut Co Ltd Screening method for cutaneous disease therapeutic agent

Also Published As

Publication number Publication date
WO2005030773A1 (en) 2005-04-07

Similar Documents

Publication Publication Date Title
AU2009331179B2 (en) Novel bicyclic heterocyclic compound
TWI503321B (en) Substituted pyrazolo(1,5-a)pyrimidine compounds as trk kinase inhibitors
RU2642777C2 (en) 2-aminopyrasine derivatives as csf-1r kinase inhibitors
JP5607046B2 (en) TRPV4 antagonist
JP3993651B2 (en) Cyclopropachromene carboxylic acid derivative
JP4493503B2 (en) Heterocyclic compounds and antitumor agents containing the same as active ingredients
WO2006023844A2 (en) Compositions and methods comprising proteinase activated receptor antagonists
JPWO2005030773A1 (en) New pyrazolopyrimidine derivatives
KR20130097776A (en) Cyclopropane compounds
JP4363530B2 (en) Protein kinase inhibitor
JP2018508502A (en) Substituted amino 6-membered saturated heteroalicyclic compounds as long-acting DPP-IV inhibitors
JP2011525924A (en) Prolyl hydroxylase inhibitor
US20040242602A1 (en) Novel thiazolidine compounds as calcium sensing receptor modulators
CA2824536A1 (en) Protease activated receptor 2 (par2) antagonists
JP3269574B2 (en) Methananoanthracene compounds, pharmaceutical compositions containing the same for treating neuropsychiatric disorders, and methods and intermediates for producing the compounds
CA3194843A1 (en) A sortilin antagonist for use in the prevention or treatment of hearing loss
CN117120434A (en) Indoline derivatives as DDR1 and DDR2 inhibitors
TW201206910A (en) Substituted N-heterocycloalkyl bipyrrolidinylphenyl amide derivatives, preparation and therapeutic use thereof
TWI790810B (en) Ret kinase inhibitors
US11970485B2 (en) RET kinase inhibitors
US9371311B2 (en) Benzoimidazol-2-yl pyrimidine derivatives
AU2012309556A1 (en) Pyrrolidine-3-ylacetic acid derivative
WO2024048615A1 (en) Method for producing quinoxaline derivative
JPH11158067A (en) Serotonin 2 receptor antagonist
CA2317760A1 (en) Piperazino derivatives as neurokinin antagonists

Legal Events

Date Code Title Description
A621 Written request for application examination

Free format text: JAPANESE INTERMEDIATE CODE: A621

Effective date: 20070911

A762 Written abandonment of application

Free format text: JAPANESE INTERMEDIATE CODE: A762

Effective date: 20100215