JP3449766B2 - Motilin antagonist - Google Patents

Motilin antagonist

Info

Publication number
JP3449766B2
JP3449766B2 JP33872893A JP33872893A JP3449766B2 JP 3449766 B2 JP3449766 B2 JP 3449766B2 JP 33872893 A JP33872893 A JP 33872893A JP 33872893 A JP33872893 A JP 33872893A JP 3449766 B2 JP3449766 B2 JP 3449766B2
Authority
JP
Japan
Prior art keywords
motilin
compound
present
added
contraction
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.)
Expired - Fee Related
Application number
JP33872893A
Other languages
Japanese (ja)
Other versions
JPH07138284A (en
Inventor
榮五郎 村山
昌幸 原村
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.)
Chugai Pharmaceutical Co Ltd
Original Assignee
Chugai Pharmaceutical 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 Chugai Pharmaceutical Co Ltd filed Critical Chugai Pharmaceutical Co Ltd
Priority to JP33872893A priority Critical patent/JP3449766B2/en
Publication of JPH07138284A publication Critical patent/JPH07138284A/en
Application granted granted Critical
Publication of JP3449766B2 publication Critical patent/JP3449766B2/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Landscapes

  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)

Description

【発明の詳細な説明】 【0001】 【産業上の利用分野】本発明はモチリンアンタゴニスト
に関する。 【0002】 【従来の技術】消化管ホルモンの一つであるモチリン
は、ブタ十二指腸より抽出された22個の直鎖のペプチ
ドであり(Brown et al., Can.
J. Physiol. Pharmacol. 4
9、 399−405 1971)、ヒトを含む哺乳類
動物の消化管運動を調節していることはよく知られてい
る。外因性に与えたモチリンは、ヒトおよびイヌに空腹
期伝播性収縮(Interdigestive Mig
rating Contractions,IMC)と
同様な収縮を引き起こし、胃排出を促進することが報告
されている(Itohet al., Scand.
J. Gastroenterol. 11、93−1
10 1976; Peeters et al.,
Gastroenterology 102、 97−
101 1992)。そのため、モチリンアゴニストで
あるエリスロマイシン誘導体が消化管運動機能促進剤と
して開発が進められている(Inatomi et a
l., J. Pharmacol. Exp.The
r. 251、 707−712 1989; Sat
oh et al., J. Pharmacol.
Exp. Ther.254、 940−944 19
90)。 【0003】またモチリンレセプターは、十二指腸に主
に存在することが知られていたが、最近、下部消化管の
大腸にも存在することが認められ(William e
tal., Am. J. Physiol. 26
2、 G50−G55 1992)、上部消化管運動ば
かりでなく、下部消化管運動調節にもモチリンが関与す
る可能性が示されている。さらに、下痢症状を示す過敏
性腸症候群患者やストレス下の過敏性腸症候群患者が高
モチリン血症を示すことが報告されており(Prest
on et al., Gut 26、 1059−1
064 1985; Fukudo et al.,
Tohoku J. Exp. Med. 151、
373−385 1987)、本病態に血中モチリンの
上昇が関与する可能性が示唆されている。 【0004】 【発明が解決しようとする課題】しかしながら、選択的
なモチリンレセプターアンタゴニストはまだ発見されて
おらず、そのことが、モチリンの消化管運動に対する作
用の研究や、本分野における医薬品の開発研究において
大きな妨げになっている。 【0005】 【課題を解決するための手段】本発明者らは、以上のよ
うな観点からモチリンアンタゴニストについて鋭意研究
を重ねた結果、本発明の化合物がモチリンアンタゴニス
トの作用を有することを見いだし、本発明を完成した。
本発明化合物であるモチリンレセプターアンタゴニスト
は、モチリンおよびモチリンアゴニストの開発研究にお
いて、薬理学的なツールとして使えるばかりでなく、過
敏性腸症候群など消化管運動機能に関連した疾患に対す
る医薬品として開発できる可能性もある。 【0006】本発明化合物は新規なペプチドであり、通
常のペプチド合成法、例えば固相合成法や液相合成法な
どで合成できる。また、アミノ酸上の置換基はペプチド
合成の前後の何れにおいても導入することができる。チ
ロシン残基への置換基の導入は、通常の有機化学的方法
例えばプロトン酸やルイス酸などの酸触媒の存在下での
フリーデルクラフツ反応などにより行うことができる。 【0007】 【実施例】以下に実施例により本発明をさらに詳細に説
明する。 【0008】 【参考例1】化合物1の合成 【化2】 PEPSYN KA樹脂(ミリジェン社製、0.2mm
ol/g)3.5gをDMF中にて膨潤させた後、Fm
oc−βAla−OH 1.74g 、ジイソプロピル
カルボジイミド353mg、4−ジメチルアミノピリジ
ン86mgを加え、室温にて一晩放置し、反応させる。
反応終了後、DMF,メタノール、酢酸で順次洗浄した
後、メタノール、DMFにて再度順次洗浄する。得られ
たFmoc−βAla−PEPSYN KA樹脂を原料
として、ペプチド合成装置(LKB Biolynx4
175)を用いた固相合成法によってペプチド合成を行
う。常法にしたがって、Fmoc−Tyr(tBu)−
PFPエステル、Fmoc−Lys(Boc)−DHB
Tエステルを順次反応、脱保護させた後、Fmoc−P
he−PFPエステルのカップリング反応終了後、DM
F,メタノールにて順次洗浄し、乾燥して、保護ペプチ
ド樹脂3.9gを得る。得られた保護ペプチド樹脂3.
9gに、TFA4.7ml、アニソール2.5ml、
1,2−エタンジチオール0.5mlを加え、室温にて
2時間反応させる。濾過の後、濾液を減圧下にて濃縮
し、氷冷した無水エーテル中にあけ固体化する。得られ
た固体を遠心分離により収集し、無水エーテルにより洗
浄した後、乾燥させる。DMF200ml及びピリジン
200mlを加え、Bop試薬1.55gを加えて、2
4時間反応させる。反応終了後減圧にて濃縮し、水を加
えて固体とし、一晩攪拌後、固体を濾集し乾燥させる。
20%ピペリジン−DMF溶液を加え20分間室温で攪
拌した後、DMFを加え減圧にて濃縮する。得られた濃
縮液をセップパックC18(ウォーターズ社製)で処理
した後、減圧下で濃縮し、高速液体クロマトグラフィー
[カラム;YMC ODS、移動相;アセトニトリル
(0.1%TFA)/蒸留水(0.1%TFA) 10
%→30%グラジェント30min]にて、分離精製す
る。得られたピーク画分を凍結乾燥すると、化合物1
56.9mgを得る。 【0009】アミノ酸分析 測定値(理論値) βAla;1.23(1) Ty
r;1.09(1) Phe;1.00(1) Lys;0.63(1) Fab−Mass m/z 510(M+H) 【0010】 【実施例1】化合物2の合成 【化3】 参考例1で得られた化合物1 170mgに、TFA1
0ml,トリメチルシリルトリフラート0.1mlを加
え、氷−食塩にて冷却しながらイソブテンガスを導入す
る。氷冷した無水エーテル中にあけ固体化し、得られた
固体を遠心分離により収集し、無水エーテルにより洗浄
する。DMFを加え溶解し、セップパックC18(ウォ
ーターズ社製)で処理した後、高速液体クロマトグラフ
ィー[カラム;YMC ODS、移動相;アセトニトリ
ル(0.1%TFA)/蒸留水(0.1%TFA) 2
0%→60%グラジェント40min]にて、分離精製
する。得られたピーク画分を凍結乾燥すると、化合物2
85.5mgを得る。 【0011】NMR(90%HO−10%DO) δ;1.01〜1.11(m,2H) 1.29(s,
9H) 1.38〜1.47(m,2H) 1.49〜
1.63(m,2H) 2.07〜2.31(m,2
H) 2.62〜2.88(m,2H) 2.80〜
2.90(m,2H) 3.97〜4.07(brs,1H) 4.27〜4.
33(m,1H) 4.40〜4.47(m,1H)
6.64(d,J=8Hz,1H) 6.80(d,J
=8Hz,1H) 6.95(s,1H) 7.14〜
7.20(m,2H) 7.21〜7.27(m,2
H) 7.35〜7.40(m,1H) 7.60〜
7.65(dd,J=8Hz,8Hz、1H) 8.0
3(brs,3H)8.39(d,J=8Hz,1H)
8.57(d,J=8Hz,1H) Fab−Mass m/z 566(M+H) 【0012】 【試験例1】モチリン受容体結合実験 モチリン受容体結合実験は次の方法で行った[Vant
rappen etal.,Regul. Pepti
des,15,143(1986)]。屠殺したウサギ
より十二指腸を摘出し、粘膜を剥離後、50mM Tr
is溶液中でhomogenizeして蛋白液とした。
蛋白液を125Iモチリン25pMと共にインキュベー
トした後に、蛋白に結合した放射活性を測定した。イン
キュベート液中に、何も添加しなかった際の放射活性と
大過剰のモチリン(10−7M)を添加した際の放射活
性の差を特異的結合とした。薬物の活性は特異的結合を
50%に減らす濃度(IC50,M)で表した。その結
果、本発明化合物は用量依存的に特異的結合を減少さ
せ、IC50は、1.0±0.3x10−8M(N=
4)と計算された。 【0013】 【試験例2】ウサギ摘出十二指腸縦層筋標本の収縮に対
する作用 モチリンによるウサギ摘出十二指腸縦層筋標本の収縮に
対する本発明化合物の作用を調べた。屠殺したウサギよ
り摘出した十二指腸標本(5x15mm)を、28℃に
加温したkrebs溶液を満たした恒温槽(organ
bath 10ml)中に縦走筋方向に懸垂した。混
合ガス(95% 02,5%C02)をKrebs溶液
に連続的に通気し、十二指腸標本の収縮は、isoto
nictransducer(ME−3407,ME
Commercial,Tokyo,Japan)を介
して等張性(負荷 1g)に記録した。収縮の程度はア
セチルコリン10−4Mの濃度による収縮を100%と
して、それに対する割合で示した。結果を図1に示す。 【図1】恒温槽内に滴下されたモチリンは、十二指腸標
本を濃度依存的に収縮させた。本発明化合物の恒温槽内
への前処置は、モチリンの濃度依存性収縮曲線を右に平
行移動させた。この結果のSchildプロットを図2
に示した。 【図2】これより直線の傾きは1.07、pA2値は
7.17と計算された。また、本発明化合物はアセチル
コリンおよびKClの濃度依存性収縮曲線には影響を与
えなかった。この結果より、本発明化合物は、モチリン
の競合的な拮抗剤と考えられた。
Description [0001] The present invention relates to motilin antagonists. [0002] Motilin, one of the gut hormones, is a 22 linear peptide extracted from pig duodenum (Brown et al., Can.
J. Physiol. Pharmacol. 4
9, 399-405 1971), and is well known to regulate gastrointestinal motility in mammals, including humans. Motilin given exogenously gives fasting-induced contractions (Interdigestive Mig) to humans and dogs.
It has been reported that it causes contraction similar to that of ratting contractions (IMC) and promotes gastric emptying (Itoh et al., Scand.
J. Gastroenterol. 11, 93-1
10 1976; Peters et al. ,
Gastroenterology 102, 97-
101 1992). Therefore, erythromycin derivatives, which are motilin agonists, are being developed as gastrointestinal motility promoters (Inatomi et a).
l. , J. et al. Pharmacol. Exp. The
r. 251, 707-712 1989; Sat
oh et al. , J. et al. Pharmacol.
Exp. Ther. 254, 940-944 19
90). The motilin receptor has been known to mainly exist in the duodenum, but has recently been found to also exist in the large intestine of the lower gastrointestinal tract (Williamme).
tal. , Am. J. Physiol. 26
2, G50-G55 1992), indicating that motilin may be involved not only in upper gastrointestinal motility but also in lower gastrointestinal motility regulation. Furthermore, it has been reported that irritable bowel syndrome patients showing diarrhea symptoms and irritable bowel syndrome patients under stress show hypermotilinemia (Prest
on et al. , Gut 26, 1059-1
064 1985; Fukudo et al. ,
Tohoku J .; Exp. Med. 151,
373-385 1987), and it has been suggested that elevated blood motilin may be involved in this disease state. [0004] However, a selective motilin receptor antagonist has not yet been discovered, which has led to studies on the effects of motilin on gastrointestinal motility and research and development of pharmaceuticals in this field. Is a major hindrance. The present inventors have conducted intensive studies on motilin antagonists from the above viewpoints, and as a result, have found that the compound of the present invention has the action of a motilin antagonist. Completed the invention.
The motilin receptor antagonist of the present invention can be used not only as a pharmacological tool in the development research of motilin and motilin agonists, but also as a drug for diseases related to gastrointestinal motility such as irritable bowel syndrome. There is also. The compound of the present invention is a novel peptide and can be synthesized by an ordinary peptide synthesis method, for example, a solid phase synthesis method or a liquid phase synthesis method. In addition, a substituent on an amino acid can be introduced before or after peptide synthesis. Introduction of a substituent to a tyrosine residue can be carried out by a conventional organic chemical method such as a Friedel-Crafts reaction in the presence of an acid catalyst such as a protonic acid or a Lewis acid. The present invention will be described in more detail with reference to the following examples. Reference Example 1 Synthesis of Compound 1 PEPSYN KA resin (Milligen, 0.2 mm
ol / g) After swelling 3.5 g in DMF, Fm
1.74 g of oc-βAla-OH, 353 mg of diisopropylcarbodiimide, and 86 mg of 4-dimethylaminopyridine are added, and left at room temperature overnight to react.
After the completion of the reaction, the resultant is sequentially washed with DMF, methanol and acetic acid, and then sequentially again with methanol and DMF. Using the obtained Fmoc-βAla-PESYN KA resin as a raw material, a peptide synthesizer (LKB Biolynx4) was used.
The peptide is synthesized by the solid phase synthesis method using 175). According to a conventional method, Fmoc-Tyr (tBu)-
PFP ester, Fmoc-Lys (Boc) -DHB
After sequentially reacting and deprotecting the T ester, Fmoc-P
After completion of the coupling reaction of the he-PFP ester, DM
It wash | cleans in order by F and methanol, and it dries, and obtains 3.9 g of protected peptide resins. 2. Obtained protected peptide resin
In 9 g, 4.7 ml of TFA, 2.5 ml of anisole,
0.5 ml of 1,2-ethanedithiol is added and reacted at room temperature for 2 hours. After filtration, the filtrate is concentrated under reduced pressure, poured into ice-cooled anhydrous ether, and solidified. The resulting solid is collected by centrifugation, washed with anhydrous ether and dried. 200 ml of DMF and 200 ml of pyridine were added, 1.55 g of Bop reagent was added, and 2
Incubate for 4 hours. After completion of the reaction, the reaction solution is concentrated under reduced pressure, and solidified by adding water. After stirring overnight, the solid is collected by filtration and dried.
After adding a 20% piperidine-DMF solution and stirring for 20 minutes at room temperature, DMF is added and concentrated under reduced pressure. The obtained concentrate is treated with Seppak C 18 (manufactured by Waters), concentrated under reduced pressure, and subjected to high-performance liquid chromatography [column: YMC ODS, mobile phase: acetonitrile (0.1% TFA) / distilled water] (0.1% TFA) 10
% → 30% gradient 30 min]. When the obtained peak fraction was lyophilized, Compound 1 was obtained.
56.9 mg are obtained. Amino acid analysis measured value (theoretical value) βAla; 1.23 (1) Ty
r; 1.09 (1) Phe; 1.00 (1) Lys; 0.63 (1) Fab-Mass m / z 510 (M + H) + Example 1 Synthesis of Compound 2 ] 170 mg of the compound 1 obtained in Reference Example 1 was added to TFA1
0 ml and 0.1 ml of trimethylsilyl triflate are added, and isobutene gas is introduced while cooling with ice-salt. The solid is poured into ice-cold anhydrous ether and the resulting solid is collected by centrifugation and washed with anhydrous ether. DMF was added and dissolved, treated with Seppak C 18 (manufactured by Waters), and then subjected to high performance liquid chromatography [column: YMC ODS, mobile phase: acetonitrile (0.1% TFA) / distilled water (0.1% TFA) ) 2
[0% → 60% gradient 40 min]. The resulting peak fraction was lyophilized to give Compound 2
85.5 mg are obtained. NMR (90% H 2 O-10% D 2 O) δ; 1.01-1.11 (m, 2H) 1.29 (s,
9H) 1.38-1.47 (m, 2H) 1.49-
1.63 (m, 2H) 2.07 to 2.31 (m, 2H)
H) 2.62 to 2.88 (m, 2H) 2.80 to
2.90 (m, 2H) 3.97-4.07 (brs, 1H) 4.27-4.
33 (m, 1H) 4.40 to 4.47 (m, 1H)
6.64 (d, J = 8 Hz, 1H) 6.80 (d, J
= 8 Hz, 1H) 6.95 (s, 1H) 7.14-
7.20 (m, 2H) 7.21 to 7.27 (m, 2H)
H) 7.35-7.40 (m, 1H) 7.60-
7.65 (dd, J = 8 Hz, 8 Hz, 1H) 8.0
3 (brs, 3H) 8.39 (d, J = 8 Hz, 1H)
8.57 (d, J = 8 Hz, 1 H) Fab-Mass m / z 566 (M + H) + [Test Example 1] Motilin receptor binding experiment The motilin receptor binding experiment was performed by the following method [Vant
rappen et al. , Regul. Pepti
des, 15, 143 (1986)]. The duodenum was removed from the sacrificed rabbit, the mucous membrane was detached, and 50 mM Tr was removed.
It was homogenized in an is solution to obtain a protein solution.
After incubating the protein solution with 25 pM of 125 I motilin, the radioactivity bound to the protein was measured. The difference between the radioactivity when nothing was added and the radioactivity when a large excess of motilin (10 −7 M) was added to the incubation solution was defined as specific binding. Drug activity was expressed as the concentration that reduced specific binding to 50% (IC 50 , M). As a result, the compound of the present invention reduced specific binding in a dose-dependent manner, and IC 50 was 1.0 ± 0.3 × 10 −8 M (N =
4) was calculated. Test Example 2 Effect on contraction of a rabbit isolated duodenal longitudinal muscle sample The effect of the compound of the present invention on contraction of a rabbit isolated duodenal longitudinal muscle sample by motilin was examined. A duodenal specimen (5 × 15 mm) extracted from a sacrificed rabbit was placed in a thermostat (organ) filled with a krebs solution heated to 28 ° C.
bath 10 ml) in the longitudinal muscle direction. A gas mixture (95% 0,5% C02) was continuously bubbled through the Krebs solution and the contraction of the duodenal specimen was
nictransducer (ME-3407, ME
It was recorded isotonic (load 1 g) via Commercial (Tokyo, Japan). The degree of contraction was expressed as a percentage with respect to the contraction due to the concentration of acetylcholine 10 −4 M as 100%. The results are shown in FIG. FIG. 1. Motilin dropped into a thermostat shrinks a duodenal specimen in a concentration-dependent manner. Pretreatment of the compound of the present invention in a thermostatic bath translated the concentration-dependent contraction curve of motilin to the right. The resulting Schild plot is shown in FIG.
It was shown to. FIG. 2 From this, the slope of the straight line was calculated to be 1.07, and the pA2 value was calculated to be 7.17. Further, the compound of the present invention did not affect the concentration-dependent contraction curves of acetylcholine and KCl. From these results, the compound of the present invention was considered to be a competitive antagonist of motilin.

【図面の簡単な説明】 【図1】 本発明化合物のウサギ摘出十二指腸縦層筋標
本の収縮に対する作用を示す。 【図2】 本発明化合物のSchildプロットを示
す。
BRIEF DESCRIPTION OF THE DRAWINGS FIG. 1 shows the effect of the compound of the present invention on contraction of a rabbit isolated duodenal longitudinal muscle sample. FIG. 2 shows a Schild plot of the compound of the present invention.

───────────────────────────────────────────────────── フロントページの続き (56)参考文献 Am J Physiol,1989年, 257,G470−G474 J Med Chem,2002年,45, 670−675 Bioorg Med Chem L ett,1999年,9(24),3441−3446 (58)調査した分野(Int.Cl.7,DB名) C07K 5/023 MEDLINE(STN) CA(STN)────────────────────────────────────────────────── ─── Continuation of the front page (56) References Am J Physiol, 1989, 257, G470-G474 J Med Chem, 2002, 45, 670-675 Biomed Med Chem Lett, 1999, 9 (24), 3441-3446 (58) Field surveyed (Int. Cl. 7 , DB name) C07K 5/023 MEDLINE (STN) CA (STN)

Claims (1)

(57)【特許請求の範囲】 【請求項1】 式(1) 【化1】 で示される化合物またはその塩。(57) [Claims] [Claim 1] Formula (1) Or a salt thereof.
JP33872893A 1993-11-19 1993-11-19 Motilin antagonist Expired - Fee Related JP3449766B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP33872893A JP3449766B2 (en) 1993-11-19 1993-11-19 Motilin antagonist

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
JP33872893A JP3449766B2 (en) 1993-11-19 1993-11-19 Motilin antagonist

Publications (2)

Publication Number Publication Date
JPH07138284A JPH07138284A (en) 1995-05-30
JP3449766B2 true JP3449766B2 (en) 2003-09-22

Family

ID=18320908

Family Applications (1)

Application Number Title Priority Date Filing Date
JP33872893A Expired - Fee Related JP3449766B2 (en) 1993-11-19 1993-11-19 Motilin antagonist

Country Status (1)

Country Link
JP (1) JP3449766B2 (en)

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TW460478B (en) 1997-08-15 2001-10-21 Chugai Pharmaceutical Co Ltd Phenethylamine derivatives
US5972939A (en) * 1997-10-28 1999-10-26 Ortho-Mcneil Pharmaceutical, Inc. Cyclopentene derivatives useful as antagonists of the motilin receptor
TW509699B (en) 1998-09-24 2002-11-11 Chugau Pharmaceutical Co Ltd Ethylamine derivatives
KR100440643B1 (en) 1999-01-28 2004-07-21 쥬가이 세이야쿠 가부시키가이샤 Substituted phenethylamine derivatives
JP4800558B2 (en) * 2000-08-24 2011-10-26 中外製薬株式会社 Cyclic peptide derivative
WO2002059141A1 (en) * 2001-01-25 2002-08-01 Chugai Seiyaku Kabushiki Kaisha Peptide derivatives
US20080312209A1 (en) 2005-07-12 2008-12-18 Glaxo Group Limited Piperazine Heteroaryl Derivatives as Gpr38 Agonists
JO2747B1 (en) 2005-07-26 2014-03-15 جلاكسو جروب ليمتد Compounds
US9133235B2 (en) 2006-09-11 2015-09-15 Ocera Therapeutics, Inc. Macrocyclic antagonists of the motilin receptor for treatment of gastrointestinal dysmotility disorders
CA2750699C (en) 2009-02-27 2015-12-29 Raqualia Pharma Inc. Oxyindole derivatives with motilin receptor agonistic activity

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Am J Physiol,1989年,257,G470−G474
Bioorg Med Chem Lett,1999年,9(24),3441−3446
J Med Chem,2002年,45,670−675

Also Published As

Publication number Publication date
JPH07138284A (en) 1995-05-30

Similar Documents

Publication Publication Date Title
JP3449766B2 (en) Motilin antagonist
FI88400B (en) FRAMEWORK FOR THE FRAME STATION INHIBITION OF PEPTIDER
US7355079B2 (en) Thyronamine derivatives and analogs and methods of use thereof
JP2010511687A (en) Chelating agent
Wagner et al. The (2‐phenyl‐2‐trimethylsilyl) ethyl‐(PTMSEL)‐linker in the synthesis of glycopeptide partial structures of complex cell surface glycoproteins
CA3105786A1 (en) (r)-4-(1-(1-(4-(trifluoromethyl)benzyl)pyrrolidine-2-carboxamide)cyclopropyl)-benzoic acid as ep4 receptor antagonist
JPS5883700A (en) Novel derivative of 3-aminopregn-5-ene, salt of same, manufacture, use as drug and composition
JP2008505180A (en) Process for producing amino acids useful for the production of peptide receptor modulators
TW200820988A (en) Radiofluorination
JP3519367B2 (en) Ethylamine derivative
US6979750B1 (en) Thyronamine derivatives and analogs and methods of use thereof
CN117460720A (en) Preparation method of heterocycloalkyl compound, intermediate and application thereof
US6610502B1 (en) Compound 2-amino-3-[2-(α-mannopyranosyl)indol-3-yl]propionic acid, process for preparing the same, and method for inspecting function of living body with the novel compound
JPH06211886A (en) Erythromycin derivative
Matsumoto et al. Synthesis and biological evaluation of cytogenin derivatives
JP4800558B2 (en) Cyclic peptide derivative
JP4008446B2 (en) Β-Benzyloxyaspartic acid derivatives having disubstituents on the benzene ring
EP2952504B1 (en) Method for producing desmosine, isodesmosine and derivatives thereof
Hong et al. Design, synthesis and pharmacological evaluation of active pyrrole based, nonpeptidic analogues of neurotensin (8-13)
JPH04308538A (en) Halogenation
CN112341464A (en) Halogen-substituted Latricinib compound
CN111358785A (en) Use of compound RADNOVA for treating enteritis
JP3879473B2 (en) Monoclonal antibody against human bcl10
JPH04356438A (en) Method for producing acetophenone compounds and intermediate therefor and method for producing the same
KR100599255B1 (en) Novel Resin Solid-phase Synthesis and Process of Hydroxylamine Derivatives on the Solid-phase

Legal Events

Date Code Title Description
R250 Receipt of annual fees

Free format text: JAPANESE INTERMEDIATE CODE: R250

LAPS Cancellation because of no payment of annual fees