JP6292619B2 - Anti-obesity drugs - Google Patents

Anti-obesity drugs Download PDF

Info

Publication number
JP6292619B2
JP6292619B2 JP2014137892A JP2014137892A JP6292619B2 JP 6292619 B2 JP6292619 B2 JP 6292619B2 JP 2014137892 A JP2014137892 A JP 2014137892A JP 2014137892 A JP2014137892 A JP 2014137892A JP 6292619 B2 JP6292619 B2 JP 6292619B2
Authority
JP
Japan
Prior art keywords
chemical formula
cells
obesity
present
compound represented
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
JP2014137892A
Other languages
Japanese (ja)
Other versions
JP2016013996A (en
Inventor
上村 大輔
大輔 上村
嘉徳 川添
嘉徳 川添
範人 丸
範人 丸
啓太 山本
啓太 山本
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.)
Kanagawa University
Original Assignee
Kanagawa University
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 Kanagawa University filed Critical Kanagawa University
Priority to JP2014137892A priority Critical patent/JP6292619B2/en
Publication of JP2016013996A publication Critical patent/JP2016013996A/en
Application granted granted Critical
Publication of JP6292619B2 publication Critical patent/JP6292619B2/en
Expired - Fee Related legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Landscapes

  • Pyrane Compounds (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Description

本発明は、藍藻から得られた新規天然有機化合物、およびかかる天然有機化合物を有効成分として含有する抗肥満薬に関する。   The present invention relates to a novel natural organic compound obtained from cyanobacteria and an anti-obesity drug containing such a natural organic compound as an active ingredient.

海洋は地球上の表面積の約71%を占め、地球全体の生物の約90%が海洋に棲息するといわれている。海洋生物は、陸上生物と比較すると異なった環境で生存しており、海水という塩濃度の高い系に存在し、温度変化が比較的小さく、高い圧力を受けることがある。こうした特異な環境に生育する生物が生産する化学物質は多彩な構造を持ち、それらの中には、様々な生物活性を持つものが明らかになってきている。
現在、生物活性物質の探索源として有望視されている海洋生物として藍藻が挙げられる。藍藻は光合成を行う原核生物であり、淡水湖沼、汽水域、海洋、土壌など様々な環境下で生育している。生育環境によって特異な化学構造や生理活性を示す物質を生産することが知られており、例えば、発癌プロモーター活性を有するLyngbyatoxin A(非特許文献1)、骨吸収活性を阻害するBiselyngbyaside(非特許文献2)、また強力なプロテインキナーゼの阻害活性を示すBisebromoamide(非特許文献3)などが挙げられ、生物活性物質やそのリード化合物の探索源として藍藻が期待されている。
The ocean occupies about 71% of the surface area of the earth, and it is said that about 90% of all the creatures on the earth live in the ocean. Marine organisms live in a different environment compared to terrestrial organisms, exist in a high salt concentration system called seawater, have a relatively small temperature change, and are subject to high pressure. Chemical substances produced by organisms that grow in such unique environments have a variety of structures, some of which have various biological activities.
Currently, cyanobacteria are listed as promising marine organisms as a search source for bioactive substances. Cyanobacteria are prokaryotes that perform photosynthesis and grow in various environments such as freshwater lakes, brackish waters, oceans, and soil. It is known to produce a substance exhibiting a specific chemical structure or physiological activity depending on the growth environment. For example, Lyngbyoxin A (Non-patent Document 1) having an oncogenic promoter activity, Biselyngbiaside (Non-patent Document) that inhibits bone resorption activity 2) and Bisebromide (Non-patent Document 3), which shows potent protein kinase inhibitory activity, and the like, and cyanobacteria are expected as a search source for bioactive substances and their lead compounds.

ヒトのからだの脂肪組織および種々の臓器における過度の脂肪の蓄積により引き起こされる肥満は、体質的因子、食餌性因子、精神的因子、代謝的因子、運動不足などが要因となり、結果的に摂取カロリーが消費カロリーを上回り、脂肪が蓄積して起こるものである。肥満は糖尿病、高血圧、脂質異常症など多くの生活習慣病の原因となっており、特に近年では高血圧や脂質異常症など複数の生活習慣病を合併していることをメタボリックシンドローム(内臓脂肪症候群)と呼称している。厚生労働省の平成18年度国民健康調査において、40〜74歳のメタボリックシンドロームの該当者数が約960万人、予備群者数が約980万人とされている。そのため、効果的な治療法や予防法が求められている。
また、日本人の死因の第1位はがん、第2位は脳卒中(脳梗塞や脳出血)、第3位は心臓病(心筋梗塞や狭心症)であり、2位と3位はどちらも動脈硬化や高血圧、脂質異常症などが大きな危険因子であるが、これらには食事や肥満が大きく関わっている。さらに、日本人の間で急激に増えている糖尿病、高尿酸血症や痛風、脂肪肝、膵炎なども、肥満との関わりが深い病気といわれている。
肥満の状態では、生体内の脂肪細胞に存在する脂肪滴、すなわちトリグリセリド量が増加し、細胞が肥大している。さらに、最近になって、成人となってからでも脂肪細胞の数が増加することが報告されている。したがって、脂肪細胞への分化を阻害すること、および脂肪細胞内の脂肪滴に作用することの両面から肥満の進行を阻害する試みが期待されている。
Obesity caused by excessive fat accumulation in human body adipose tissue and various organs is caused by constitutional factors, dietary factors, mental factors, metabolic factors, lack of exercise, etc., resulting in calorie intake Is more than calories burned and fat is accumulated. Obesity is the cause of many lifestyle-related diseases such as diabetes, hypertension, and dyslipidemia. In particular, metabolic syndrome (visceral fat syndrome) that multiple lifestyle-related diseases such as hypertension and dyslipidemia are combined. It is called. According to the Ministry of Health, Labor and Welfare's 2006 National Health Survey, the number of people with metabolic syndrome aged 40 to 74 is about 9.6 million, and the number of reserve groups is about 9.8 million. Therefore, effective treatment and prevention methods are required.
The first cause of death among Japanese is cancer, the second is stroke (cerebral infarction and cerebral hemorrhage), the third is heart disease (myocardial infarction and angina), which is second or third? Arteriosclerosis, hypertension, dyslipidemia, etc. are major risk factors, but diet and obesity are greatly involved in these. In addition, diabetes, hyperuricemia, gout, fatty liver, pancreatitis, etc., which are rapidly increasing among Japanese, are said to be deeply related to obesity.
In the state of obesity, the amount of lipid droplets, that is, triglycerides present in fat cells in the body increases, and the cells are enlarged. Furthermore, recently, it has been reported that the number of fat cells increases even after becoming an adult. Therefore, an attempt to inhibit the progression of obesity is expected from both the viewpoint of inhibiting differentiation into fat cells and acting on fat droplets in fat cells.

本発明者らは、前駆脂肪細胞の脂肪細胞への分化阻害または脂肪細胞の脂肪蓄積の阻害を通して抗肥満効果を示す医薬等について研究を続けており、特定のキノコ又は植物から抽出した成分を有効成分とする前駆脂肪細胞分化阻害剤(特許文献1)、環状ヘプタペプチドであるテルナチン:cyclo[−D−Ile1−(N−Me)−L−Ala2−(N−Me)−L−Leu3−L−Leu4−(N−Me)−L−Ala5−(N−Me)−D−Ala6−β―OH−D−Leu7−]を有効成分とする前駆脂肪細胞分化阻害剤(特許文献2)、および植物成分のビサボロールオキシド−A−β−グルコシドを有効成分とする前駆脂肪細胞分化阻害剤(特許文献3)を見出すことに成功した。
また、公知の抗菌作用を有する天然有機化合物であるオーレオシンにはトリグリセリドの生合成阻害活性があり、血液中のトリグリセリド濃度を低下させることが報告されている(特許文献4)。
しかしながら、これまで得られた天然物からの抽出成分や天然有機化合物は、必ずしも抗肥満薬として満足のいくものではなく、とりわけ、脂肪細胞内の脂肪滴に作用することによる抗肥満薬といった観点からは満足のいくものはなく、より優れた効果を発揮する天然有機化合物についての検討が不可欠であった。
The present inventors have continued research on drugs that exhibit an anti-obesity effect through inhibition of preadipocyte differentiation into adipocytes or inhibition of fat cell fat accumulation, and effective use of components extracted from specific mushrooms or plants Preadipocyte differentiation inhibitor as an ingredient (Patent Document 1), cyclic heptapeptide ternatin: cyclo [-D-Ile1- (N-Me) -L-Ala2- (N-Me) -L-Leu3-L A preadipocyte differentiation inhibitor (Patent Document 2) containing -Leu4- (N-Me) -L-Ala5- (N-Me) -D-Ala6-β-OH-D-Leu7-], and The inventors succeeded in finding a preadipocyte differentiation inhibitor (Patent Document 3) containing the plant component bisabolol oxide-A-β-glucoside as an active ingredient.
In addition, aureosin, which is a known natural organic compound having antibacterial action, has triglyceride biosynthesis inhibitory activity and has been reported to reduce the triglyceride concentration in blood (Patent Document 4).
However, the components extracted from natural products and natural organic compounds obtained up to now are not necessarily satisfactory as anti-obesity agents, especially from the viewpoint of anti-obesity agents by acting on lipid droplets in fat cells. However, there were no satisfactory ones, and it was essential to study natural organic compounds that exhibited better effects.

特開2004−075640号公報Japanese Patent Application Laid-Open No. 2004-075640 特開2005−220074号公報Japanese Patent Laid-Open No. 2005-220074 特開2006−213648号公報JP 2006-213648 A 特開平9−194366号公報Japanese Patent Laid-Open No. 9-194366

J.H.Cardellina II,F.J.Marner,R.E.Moore,Science,204,193(1979)J. et al. H. Cardellina II, F.R. J. et al. Marner, R.M. E. Moore, Science, 204, 193 (1979) T.Yonezawa,N.Mase,H.Sasaki,T.Teruya,S.Hasegawa,B.Cha,K.Yagasaki,K.Suenaga,K.Nagai,J.Woo,Cell Biochem.,2012,2,440T.A. Yonezawa, N .; Mase, H .; Sasaki, T .; Teruya, S .; Hasegawa, B.I. Cha, K .; Yagasaki, K .; Suenaga, K .; Nagai, J .; Woo, Cell Biochem. , 2012,2,440 T.Teruya,H.Sasaki,H.Fukazawa,and K.Suenaga, Org.Lett.,2009,11,5062T.A. Teruya, H .; Sasaki, H .; Fukazawa, and K.K. Suenaga, Org. Lett. , 2009, 11, 5062

本発明の課題は、上記に記載した背景の下、前駆脂肪細胞の脂肪細胞への分化阻害活性および脂肪細胞内の脂肪滴の縮小活性を有する新規天然有機化合物、ならびにかかる天然有機化合物を有効成分として含有する医薬を提供することにある。   An object of the present invention is to provide a novel natural organic compound having an activity of inhibiting the differentiation of preadipocytes into adipocytes and an activity of reducing lipid droplets in fat cells, and an active ingredient of such a natural organic compound in the background described above. It is in providing the pharmaceutical containing as.

本発明者らは、沖縄県石垣島米原にて採取した藍藻(Leptolyngbya sp.)から新規天然有機化合物である化学式(1)で示される化合物を単離し、かかる化合物が前駆脂肪細胞の脂肪細胞への分化阻害活性および脂肪細胞内の脂肪滴の縮小活性を有することを見出し、本発明を完成するに至った。

Figure 0006292619
(化学式(1)) The present inventors isolated a compound represented by the chemical formula (1), which is a novel natural organic compound, from a blue-green algae (Leptolingbya sp.) Collected in Ishigaki-jima Yonehara, Okinawa Prefecture, and such a compound becomes a fat cell of a preadipocyte. Have been found to have differentiation-inhibiting activity and reduction activity of lipid droplets in adipocytes, and the present invention has been completed.
Figure 0006292619
(Chemical formula (1))

すなわち、本発明は、化学式(1)で示される化合物に関する。

Figure 0006292619
(化学式(1)) That is, the present invention relates to a compound represented by the chemical formula (1).
Figure 0006292619
(Chemical formula (1))

また、本発明は、上記化学式(1)で示される化合物を有効成分として含有することを特徴とする抗肥満薬に関する。   The present invention also relates to an anti-obesity drug comprising a compound represented by the above chemical formula (1) as an active ingredient.

さらに、本発明は、脂肪細胞内の脂肪滴を縮小させることを特徴とする上記化学式(1)で示される化合物を有効成分として含有する抗肥満薬に関する。   Furthermore, the present invention relates to an anti-obesity drug containing, as an active ingredient, a compound represented by the above chemical formula (1), characterized by reducing fat droplets in fat cells.

本発明によれば、化学式(1)で示される新規化合物は、前駆脂肪細胞の脂肪細胞への分化阻害活性を有すると共に、脂肪細胞内の脂肪滴の縮小活性を有することから、脂肪の蓄積や肥満の予防、改善に寄与するものである。   According to the present invention, the novel compound represented by the chemical formula (1) has the activity of inhibiting the differentiation of preadipocytes into adipocytes, and also has the activity of reducing lipid droplets in adipocytes. It contributes to the prevention and improvement of obesity.

本発明に係る化学式(1)で示される化合物の分画・精製過程を示す図である。It is a figure which shows the fractionation / refinement | purification process of the compound shown by Chemical formula (1) based on this invention. 本発明に係る化学式(1)で示される化合物の分画・精製過程を示す図である。It is a figure which shows the fractionation / refinement | purification process of the compound shown by Chemical formula (1) based on this invention. 本発明に係る化学式(1)で示される化合物の脂肪細胞への分化阻害活性を示すグラフである。It is a graph which shows the differentiation inhibitory activity to the fat cell of the compound shown by Chemical formula (1) which concerns on this invention. 本発明に係る化学式(1)で示される化合物の脂肪細胞内のトリグリセリドレベルへの影響を示すグラフである。It is a graph which shows the influence on the triglyceride level in the fat cell of the compound shown by Chemical formula (1) which concerns on this invention. 本発明に係る化学式(1)で示される化合物の細胞内の乳酸レベルへの影響を示すチャートである。It is a chart which shows the influence on the intracellular lactic acid level of the compound shown by Chemical formula (1) which concerns on this invention.

以下、本発明の実施形態について詳細に説明する。   Hereinafter, embodiments of the present invention will be described in detail.

本発明に係る化学式(1)で示される化合物は藍藻から抽出し、精製することにより得ることができる。より詳細には、本発明に係る化学式(1)で示される化合物は沖縄県石垣島米原にて採取した藍藻(Leptolyngbya sp.)から抽出し、精製することにより得た。   The compound represented by the chemical formula (1) according to the present invention can be obtained by extraction from cyanobacteria and purification. More specifically, the compound represented by the chemical formula (1) according to the present invention was obtained by extracting and purifying from a blue-green algae (Leptolingbya sp.) Collected at Ishigakijima Maibara, Okinawa Prefecture.

藍藻は光合成を行う原核生物であり、淡水湖沼、汽水域、海洋、土壌など様々な環境下で広く生育しているものであるが、本発明で使用する藍藻としては、沖縄県石垣島米原近辺に生息する藍藻(Leptolyngbya sp.)を用いるのが好ましい。本発明に係る化学式(1)で示される化合物を含有する藍藻(Leptolyngbya sp.)またはその部位は、それ自身を乾燥させた乾燥物、その粉砕物、それら自身を圧搾抽出することにより得られる搾汁、水あるいはアルコール、エーテル、アセトンなどの有機溶媒による粗抽出物、および粗抽出物を分配、カラムクロマトなどの各種クロマトグラフィーなどで段階的に精製して得られた抽出物画分など、すべてを利用することができる。これらは単独で用いてもよく、また2種以上混合してもよい。   Cyanobacteria are prokaryotes that carry out photosynthesis and grow widely under various environments such as freshwater lakes, brackish waters, oceans, and soils. The cyanobacteria used in the present invention is near Yonehara, Ishigaki Island, Okinawa Prefecture. It is preferable to use Leptolingbya sp. The blue-green algae (Leptolingbya sp.) Containing the compound represented by the chemical formula (1) according to the present invention or a portion thereof is a dried product obtained by drying itself, a pulverized product thereof, and a squeezed product obtained by compressing and extracting the product All of the crude extract with organic solvents such as juice, water or alcohol, ether, acetone, etc., and the extract fraction obtained by stepwise purification by various chromatographies such as column chromatography etc. Can be used. These may be used alone or in combination of two or more.

例えば、藍藻を採取した後に冷凍保存し、含水メタノールにて粉砕・抽出し、粗抽出物を得、粗抽出物を酢酸エチル層と水層とに分配し、さらに、有機層としての酢酸エチル層をODSシリカゲルカラムクロマトグラフィーにより画分することができる。   For example, the cyanobacteria are collected and stored frozen, crushed and extracted with water-containing methanol to obtain a crude extract, the crude extract is distributed into an ethyl acetate layer and an aqueous layer, and an ethyl acetate layer as an organic layer is further obtained. Can be fractionated by ODS silica gel column chromatography.

本発明に係る化学式(1)で示される化合物は新規な化合物であり、本発明により前駆脂肪細胞の分化阻害活性、および脂肪細胞内の脂肪滴の縮小活性に優れることを見出されたものである。   The compound represented by the chemical formula (1) according to the present invention is a novel compound, and has been found by the present invention to be excellent in the differentiation inhibiting activity of preadipocytes and the activity of reducing lipid droplets in fat cells. is there.

本発明に係る化学式(1)で示される化合物は、優れた前駆脂肪細胞の脂肪細胞への分化阻害活性および脂肪細胞内の脂肪滴の縮小活性を有することから、抗肥満剤として使用可能である。   The compound represented by the chemical formula (1) according to the present invention can be used as an anti-obesity agent because it has excellent activity of inhibiting the differentiation of preadipocytes into adipocytes and the activity of reducing lipid droplets in fat cells. .

本発明に係る化学式(1)で示される化合物は、特に脂肪細胞内の脂肪滴の縮小活性を有することから、脂肪細胞内の脂肪滴を縮小させることによる抗肥満薬として使用可能である。   Since the compound represented by the chemical formula (1) according to the present invention has the activity of reducing lipid droplets in fat cells, it can be used as an anti-obesity drug by reducing the lipid droplets in fat cells.

本発明に係る化学式(1)で示される化合物を有効成分として含む医薬組成物は抗肥満薬として使用され得るものであるが、ここで「肥満」とは一般的には体内に脂肪組織が一定以上多量に蓄積した状態をいう。本明細書においては「肥満」は広義に解釈されるものとし、その概念に肥満症を含む。「肥満症」とは肥満に起因ないし関連する健康障害(合併症)を有するか又は将来的に有することが予測される場合であって、医学的に減量が必要とされる病態をいう。
肥満の判定法としては、通常、国際的に広く使用されているBMI(Body Mass Index)を尺度としたものが用いられている。BMIは、体重(kg)を身長(m)の二乗で除した数値(BMI=体重(kg)/身長(m))である。BMI<18.5は低体重、18.5≦BMI<25は普通体重、25≦BMI<30は肥満(1度)、30≦BMI<35は肥満(2度)、35≦BMI<40は肥満(3度)、40≦BMIは肥満(4度)と判定される。もっとも、標準体重(理想体重)は性別、年齢、職業または生活習慣の差異などによって個人ごとに相違するものであることから、肥満の判定をこの方法で一律に行い、本発明の抗肥満薬を厳格に適用することを意図するものではない。
The pharmaceutical composition containing the compound represented by the chemical formula (1) according to the present invention as an active ingredient can be used as an anti-obesity agent. Here, “obesity” generally means that adipose tissue is constant in the body. A state where a large amount is accumulated. In the present specification, “obesity” is to be interpreted broadly, and the concept includes obesity. “Obesity” refers to a medical condition that is or is predicted to have a health disorder (complication) due to or related to obesity, and that requires medical weight loss.
As a method for determining obesity, a method based on BMI (Body Mass Index), which is widely used internationally, is usually used. BMI is a numerical value obtained by dividing body weight (kg) by the square of height (m) (BMI = weight (kg) / height (m) 2 ). BMI <18.5 is low weight, 18.5 ≦ BMI <25 is normal weight, 25 ≦ BMI <30 is obesity (1 degree), 30 ≦ BMI <35 is obesity (2 degrees), 35 ≦ BMI <40 is Obesity (3 degrees), 40 ≦ BMI is determined to be obese (4 degrees). However, since the standard body weight (ideal body weight) is different for each individual due to differences in gender, age, occupation or lifestyle, obesity is uniformly determined by this method, and the anti-obesity drug of the present invention is used. It is not intended to be strictly applied.

本発明の抗肥満薬の製剤化は常法に従って行うことができる。製剤化する場合には、製剤上許容される他の成分(例えば、担体、賦形剤、崩壊剤、緩衝剤、乳化剤、懸濁剤、無痛化剤、安定剤、保存剤、防腐剤、生理食塩水など)を含有させることができる。賦形剤としては乳糖、デンプン、ソルビトール、D−マンニトール、白糖等を用いることができる。崩壊剤としてはデンプン、カルボキシメチルセルロース、炭酸カルシウム等を用いることができる。緩衝剤としてはリン酸塩、クエン酸塩、酢酸塩等を用いることができる。乳化剤としてはアラビアゴム、アルギン酸ナトリウム、トラガント等を用いることができる。懸濁剤としてはモノステアリン酸グリセリン、モノステアリン酸アルミニウム、メチルセルロース、カルボキシメチルセルロース、ヒドロキシメチルセルロース、ラウリル硫酸ナトリウム等を用いることができる。無痛化剤としてはベンジルアルコール、クロロブタノール、ソルビトール等を用いることができる。安定剤としてはプロピレングリコール、アスコルビン酸等を用いることができる。保存剤としてはフェノール、塩化ベンザルコニウム、ベンジルアルコール、クロロブタノール、メチルパラベン等を用いることができる。防腐剤としては塩化ベンザルコニウム、パラオキシ安息香酸、クロロブタノール等と用いることができる。   Formulation of the anti-obesity drug of the present invention can be performed according to a conventional method. In the case of formulating, other pharmaceutically acceptable ingredients (for example, carriers, excipients, disintegrants, buffers, emulsifiers, suspending agents, soothing agents, stabilizers, preservatives, preservatives, physiological Saline solution and the like). As the excipient, lactose, starch, sorbitol, D-mannitol, sucrose and the like can be used. As the disintegrant, starch, carboxymethylcellulose, calcium carbonate and the like can be used. Phosphate, citrate, acetate, etc. can be used as the buffer. As the emulsifier, gum arabic, sodium alginate, tragacanth and the like can be used. As the suspending agent, glyceryl monostearate, aluminum monostearate, methyl cellulose, carboxymethyl cellulose, hydroxymethyl cellulose, sodium lauryl sulfate and the like can be used. As the soothing agent, benzyl alcohol, chlorobutanol, sorbitol and the like can be used. As the stabilizer, propylene glycol, ascorbic acid or the like can be used. As preservatives, phenol, benzalkonium chloride, benzyl alcohol, chlorobutanol, methylparaben, and the like can be used. As preservatives, benzalkonium chloride, paraoxybenzoic acid, chlorobutanol and the like can be used.

製剤化する場合の剤型も特に限定されず、例えば錠剤、散剤、細粒剤、顆粒剤、カプセル剤、シロップ剤、注射剤、外用剤、および座剤などとして本発明の医薬を提供できる。
本発明の医薬組成物には、期待される治療効果(予防効果も含む)を得るために必要な量(即ち治療上有効量)の有効成分が含有される。本発明の医薬組成物中の有効成分量は一般に剤型によって異なるが、所望の投与量を達成できるように有効成分量を例えば約0.1重量%〜約95重量%の範囲内で設定する。
The dosage form for formulation is not particularly limited, and the medicament of the present invention can be provided as tablets, powders, fine granules, granules, capsules, syrups, injections, external preparations, suppositories, and the like.
The pharmaceutical composition of the present invention contains an active ingredient in an amount necessary for obtaining an expected therapeutic effect (including a preventive effect) (that is, a therapeutically effective amount). The amount of the active ingredient in the pharmaceutical composition of the present invention generally varies depending on the dosage form, but the amount of the active ingredient is set within a range of, for example, about 0.1 wt% to about 95 wt% so as to achieve a desired dose. .

本発明の抗肥満薬はその剤型に応じて経口投与又は非経口投与(静脈内、動脈内、皮下、筋肉、又は腹腔内注射、経皮、経鼻、経粘膜など)によって対象に適用される。ここでの「対象」は特に限定されず、ヒトおよびヒト以外の哺乳動物(ペット動物、家畜、実験動物を含む。)を含む。好ましい態様としては、本発明の抗肥満薬はヒトに対して適用される。   The anti-obesity drug of the present invention is applied to a subject by oral administration or parenteral administration (intravenous, intraarterial, subcutaneous, intramuscular or intraperitoneal injection, transdermal, nasal, transmucosal, etc.) depending on the dosage form. The The “subject” here is not particularly limited, and includes humans and non-human mammals (including pet animals, farm animals, and laboratory animals). In a preferred embodiment, the antiobesity agent of the present invention is applied to humans.

本発明の抗肥満薬(有効成分)の投与量は、期待される治療効果が得られるように設定される。治療上有効な投与量の設定においては一般に症状、患者の年齢、性別、および体重などが考慮される。なお、当業者であればこれらの事項を考慮して適当な投与量を設定することが可能であり、例えば、成人(体重約60kg)を対象として一日当たりの有効成分量が約1mg〜約6g、好ましくは約6mg〜約600mgとなるよう投与量を設定することができる。投与スケジュールとしては例えば一日一回〜数回、二日に一回、或いは三日に一回などを採用できる。投与スケジュールの作成においては、患者の病状や有効成分の効果持続時間などを考慮することができる。   The dose of the anti-obesity drug (active ingredient) of the present invention is set so as to obtain the expected therapeutic effect. In setting a therapeutically effective dose, symptoms, patient age, sex, weight, etc. are generally considered. A person skilled in the art can set an appropriate dose in consideration of these matters. For example, the amount of active ingredient per day for an adult (body weight: about 60 kg) is about 1 mg to about 6 g. The dose can be set to be preferably about 6 mg to about 600 mg. As the administration schedule, for example, once to several times a day, once every two days, or once every three days can be adopted. In preparing the administration schedule, the patient's medical condition and the duration of effect of the active ingredient can be taken into consideration.

以下、実施例を挙げて、本発明の具体的態様を示すものであるが、本発明の技術的範囲は実施例の記載により何ら限定されるものではない。   EXAMPLES Hereinafter, although an Example is given and the specific aspect of this invention is shown, the technical scope of this invention is not limited at all by description of an Example.

本発明に係る化学式(1)で示される化合物の単離・精製・構造決定
沖縄県石垣島米原で採取した藍藻(Leptolyngbya sp.)600g(湿重量)を用いた。採取後は冷凍保存し、小石を取り除いてから含水メタノール中にてブレンダーで粉砕した。抽出液を吸引濾過し、ロ液をエバポレーターを用いて濃縮した。粗抽出物を酢酸エチル(400ml×3)と水(400ml)とで分配した。得られた酢酸エチル層を減圧濃縮し、濃縮物を80%メタノール水溶液(250ml)とヘキサン(250ml×3)で脱脂した。得られた80%メタノール水溶液層を減圧濃縮し、濃縮物をODSカラムクロマトグラフィー[Cosmosil 75C18−OPN(φ 20×85mm)、メタノール/水(20/80→40/60→60/40→80/20→メタノール)]により5画分(Fr.1〜5)に分離した。
(図1参照)
得られた各溶出画分の内、60%メタノール溶出画分(Fr.3)15.2mgを逆相高速液体クロマトグラフィー[Develosil ODS HG−5(φ 10×250mm)、アセトニトリル/水(10/90)→(グラジェント30min)→(100/0)、検出UV215nm、流速 4.6mL/min]により9画分(Fr.3−1〜9)に分離した。得られた画分(Fr.3−5)において脂肪細胞への分化阻害活性が認められたため、この画分の精製を行った。得られたFr.3−5を逆相高速液体クロマトグラフィー[Develosil ODS HG−5(φ 10×250mm)、アセトニトリル/水(30/70)→(グラジェント30min)→(55/45)→(グラジェント5min)→(100/0)、検出UV215nm、流速 4.6mL/min]により9画分(Fr.3−5−1〜3−5−9)に分離した。得られた画分(Fr.3−5−5,7)において阻害活性が認められた。Fr.3−5−7(1.0mg、不定形白色粉末)は本発明に係る化学式(1)で示される化合物であった。
(図2参照)
Isolation / Purification / Structural Determination of Compound Represented by Chemical Formula (1) According to the Present Invention 600 g (wet weight) of blue-green algae (Leptolingya sp.) Collected from Ishigakijima Maibara, Okinawa Prefecture was used. After collection, the sample was stored frozen, pebbles were removed, and the mixture was pulverized in water-containing methanol with a blender. The extract was filtered with suction, and the filtrate was concentrated using an evaporator. The crude extract was partitioned between ethyl acetate (400 ml × 3) and water (400 ml). The obtained ethyl acetate layer was concentrated under reduced pressure, and the concentrate was degreased with 80% aqueous methanol solution (250 ml) and hexane (250 ml × 3). The obtained 80% methanol aqueous solution layer was concentrated under reduced pressure, and the concentrate was subjected to ODS column chromatography [Cosmosil 75C 18 -OPN (φ 20 × 85 mm), methanol / water (20/80 → 40/60 → 60/40 → 80). / 20 → methanol)] was separated into 5 fractions (Fr. 1 to 5).
(See Figure 1)
Among each of the obtained elution fractions, 15.2 mg of 60% methanol elution fraction (Fr.3) was subjected to reverse phase high performance liquid chromatography [Develosil ODS HG-5 (φ 10 × 250 mm), acetonitrile / water (10 / 90) → (gradient 30 min) → (100/0), detection UV 215 nm, flow rate 4.6 mL / min], and separated into 9 fractions (Fr. 3-1 to 9). Since the obtained fraction (Fr. 3-5) showed activity of inhibiting differentiation into adipocytes, this fraction was purified. The obtained Fr. 3-5 was subjected to reverse phase high performance liquid chromatography [Develosil ODS HG-5 (φ 10 × 250 mm), acetonitrile / water (30/70) → (gradient 30 min) → (55/45) → (gradient 5 min) → (100/0), detection UV 215 nm, flow rate 4.6 mL / min], and separated into 9 fractions (Fr. 3-5-1 to 3-5-9). Inhibitory activity was observed in the obtained fractions (Fr. 3-5-5, 7). Fr. 3-5-7 (1.0 mg, amorphous white powder) was a compound represented by the chemical formula (1) according to the present invention.
(See Figure 2)

図1および図2に示した操作により得られた不定形白色粉末である本発明に係る化学式(1)で示される化合物について、高分解能ESI−MSにより得られた分子イオンピークの値[m/z387.2163(M+Na)+,Δ+1.6mmu]より、分子式を求めたところ、C2132の化合物であることが判明した。
さらに、H−NMR、13C−NMRおよびHMBCを測定し、得られたスペクトルを解析することにより、不定形白色粉末であって、C2132の分子式を有する化合物の構造を以下のとおり決定した。

Figure 0006292619
With respect to the compound represented by the chemical formula (1) according to the present invention, which is an amorphous white powder obtained by the operation shown in FIG. 1 and FIG. 2, the molecular ion peak value [m / z387.1633 (M + Na) +, Δ + 1.6 mmu], the molecular formula was determined and found to be a compound of C 21 H 32 O 5 .
Furthermore, by measuring 1 H-NMR, 13 C-NMR and HMBC, and analyzing the obtained spectrum, the structure of a compound having an amorphous white powder and having a molecular formula of C 21 H 32 O 5 is shown below. It was decided as follows.
Figure 0006292619

図1および図2に示した操作により得られた不定形白色粉末である本発明の化学式(1)で示される化合物についてのH−NMR、および13C−NMRのスペクトルデータを以下に示す。
H−NMR(C,500MHz)
d 5.36(d,1H,J=8.5,H−14),5.16(t,1H,J=7.0,H−8),4.38(q,1H,J=6.5,8.5,H−15),3.29(dd,1H,J=5.5,7.5,H−12),3.23(s,3H,2−OMe),3.03(s,3H,12−OMe),2.86(d,2H,J=7.0,H−7),2.13(s,3H,3−Me),2.10(m,1H,H−10a),2.03(m,1H,H−10b),2.01(s,3H,5−Me),1.79(m,1H,H−11a),1.58(m,1H,H−11b),1.48(s,3H,13−Me),1.48(s,3H,9−Me),1.13(d,3H,J=6.5,15−Me)
13C−NMR(C,125MHz)
d 179.9(C−4),161.7(C−2),156.7(C−6),138.2(C−5),135.0(C−13),134.0(C14),119.9(C9),118.0(C−8),100.1(C−3),86.4(C−12),64.0(C−15),52.8(12−OMe),51.8(2−OMe),35.9(C−10),32.3(C−7),32.1(C−11),23.8(15−Me),15.9(9−Me),10.5(13−Me),10.0(5−Me),7.1(3−Me)
1 H-NMR and 13 C-NMR spectrum data of the compound represented by the chemical formula (1) of the present invention, which is an amorphous white powder obtained by the operations shown in FIGS. 1 and 2, are shown below.
1 H-NMR (C 6 D 6 , 500 MHz)
d 5.36 (d, 1H, J = 8.5, H-14), 5.16 (t, 1H, J = 7.0, H-8), 4.38 (q, 1H, J = 6) .5, 8.5, H-15), 3.29 (dd, 1H, J = 5.5, 7.5, H-12), 3.23 (s, 3H, 2-OMe), 3. 03 (s, 3H, 12-OMe), 2.86 (d, 2H, J = 7.0, H-7), 2.13 (s, 3H, 3-Me), 2.10 (m, 1H) , H-10a), 2.03 (m, 1H, H-10b), 2.01 (s, 3H, 5-Me), 1.79 (m, 1H, H-11a), 1.58 (m , 1H, H-11b), 1.48 (s, 3H, 13-Me), 1.48 (s, 3H, 9-Me), 1.13 (d, 3H, J = 6.5, 15- Me)
13 C-NMR (C 6 D 6 , 125 MHz)
d 179.9 (C-4), 161.7 (C-2), 156.7 (C-6), 138.2 (C-5), 135.0 (C-13), 134.0 ( C14), 119.9 (C9), 118.0 (C-8), 100.1 (C-3), 86.4 (C-12), 64.0 (C-15), 52.8 ( 12-OMe), 51.8 (2-OMe), 35.9 (C-10), 32.3 (C-7), 32.1 (C-11), 23.8 (15-Me), 15.9 (9-Me), 10.5 (13-Me), 10.0 (5-Me), 7.1 (3-Me)

実施例1で得られた本発明の化学式(1)で示される化合物の生物活性
実施例1に記載の方法に従って、得られた本発明の化学式(1)で示される化合物の生物活性として、脂肪細胞への分化阻害、細胞生存率による細胞毒性、脂肪細胞からのトリグリセリドの減少および乳酸の増加を調べた。
実施例には、脂肪細胞への効率的な分化が実証されているマウス繊維芽細胞3T3−L1細胞を下記(A)〜(D)のとおり調整して用いた。
(A)3T3−L1細胞の増殖、保存
3T3−L1細胞は100mmディッシュ上で、5%ウシ血清を添加したDMEM増殖用培地で5%炭酸ガス−空気、飽和水蒸気下、37℃で培養した。なお、3T3−L1細胞は、最初に大量培養しておき、常法に従って、分注して凍結保存した。
(B)継代
PBS(−)5mlで細胞を洗浄し、0.25%トリプトシン1mM EDTA含有生理食塩水を1mlを加えて37℃で3分間静置した。増殖用培地5mlで細胞を懸濁して15mlの遠沈管に移して、1500rpm、3分間の遠心によって細胞を沈殿させた。1/4から1/5に希釈して、ディッシュにまいた。
(C)実施例用細胞の準備
上述と同様の方法でディッシュから剥離、遠心沈殿させた細胞は、適当な量の増殖用培地に懸濁し、細胞数を計測した。それをもとに、5×10個/wellとなるように96穴プレートに播種して48時間培養した。
(D)分化誘導培地の調製
上記増殖用培地100mlに1mMデキサメタゾン溶液1ml、3−イソブチル―1−メチルキサンチン11.1mg、5mg/mlインスリン溶液0.1ml、および抗生物質溶液(ペニシリンGカリウム塩317mg、ストレプトマイシン500mgを滅菌生理食塩水25mlに溶解して調整)0.45mlを添加し、溶解することで調整した。
Biological activity of the compound represented by the chemical formula (1) of the present invention obtained in Example 1 According to the method described in Example 1, as the biological activity of the compound represented by the chemical formula (1) of the present invention obtained, fat Inhibition of cell differentiation, cytotoxicity due to cell viability, reduction of triglycerides from adipocytes, and increase of lactic acid were investigated.
In the examples, mouse fibroblast 3T3-L1 cells, which have been demonstrated to be efficiently differentiated into adipocytes, were used as prepared in the following (A) to (D).
(A) Growth and storage of 3T3-L1 cells 3T3-L1 cells were cultured on a 100 mm dish in a DMEM growth medium supplemented with 5% bovine serum at 37 ° C. under 5% carbon dioxide-air and saturated water vapor. In addition, 3T3-L1 cells were first cultured in large quantities, and dispensed and stored frozen according to a conventional method.
(B) Passage Cells were washed with 5 ml of PBS (−), 1 ml of 0.25% trypticin 1 mM EDTA-containing physiological saline was added, and the mixture was allowed to stand at 37 ° C. for 3 minutes. The cells were suspended in 5 ml of growth medium, transferred to a 15 ml centrifuge tube, and precipitated by centrifugation at 1500 rpm for 3 minutes. Dilute from 1/4 to 1/5 and go to dish.
(C) Preparation of cells for examples The cells peeled from the dish and centrifuged by the same method as described above were suspended in an appropriate amount of growth medium, and the number of cells was counted. Based on this, it was seeded in a 96-well plate at 5 × 10 4 cells / well and cultured for 48 hours.
(D) Preparation of differentiation induction medium Into 100 ml of the above growth medium, 1 ml of 1 mM dexamethasone solution, 11.1 mg of 3-isobutyl-1-methylxanthine, 0.1 ml of 5 mg / ml insulin solution, and antibiotic solution (317 mg of penicillin G potassium salt) The solution was adjusted by adding 0.45 ml and dissolving 500 mg of streptomycin in 25 ml of sterile physiological saline.

(1)脂肪細胞への分化誘導率による分化阻害
3T3−L1細胞の脂肪細胞への分化誘導には、培地を増殖用培地から分化誘導培地に置き換えることで行った。この時、特定の濃度に調整した化学式(1)で示される化合物を分化誘導培地に添加し、ブランクとコントロールには分化誘導培地のみを添加した。プレートは5%炭酸ガス−空気、飽和水蒸気下、37℃で培養した。48時間後に、インスリンを加えた新鮮な増殖培地に置き換えた。その後、2日おきに培地を新鮮な増殖培地に置き換えた。
上記の方法で分化誘導開始より7〜10日間の培養後、細胞を顕微鏡で観察し、コントロールの細胞が十分に分化した時点で評価した。
脂肪細胞への分化誘導率は、細胞内に蓄積されたトリグリセリドの量を定量する事によって算出した。細胞をPBS(−)で洗浄して風乾させ、そこにラボアッセイトリグリセライドキットワコーを100μL/wellずつ添加した。室温で30〜60分間静置し、630nmの吸光度を測定してトリグリセリドを定量した。
結果を図3に示す。本発明に係る化学式(1)で示される化合物は濃度420nMで3T3−L1細胞の脂肪細胞への分化を50%阻害した。一方、Fr.3−5−5から分離された化合物(Fr.3−5−5−2,3−5−5−3)は、脂肪細胞への分化阻害活性を示さなかった。
(1) Inhibition of differentiation by rate of induction of differentiation into adipocytes Induction of differentiation of 3T3-L1 cells into adipocytes was performed by replacing the culture medium with a differentiation-inducing medium. At this time, the compound represented by the chemical formula (1) adjusted to a specific concentration was added to the differentiation induction medium, and only the differentiation induction medium was added to the blank and the control. The plate was cultured at 37 ° C. under 5% carbon dioxide gas-air and saturated water vapor. After 48 hours, it was replaced with fresh growth medium supplemented with insulin. Thereafter, the medium was replaced with fresh growth medium every two days.
After culturing for 7 to 10 days from the start of differentiation induction by the above method, the cells were observed with a microscope and evaluated when the control cells were sufficiently differentiated.
The induction rate of differentiation into fat cells was calculated by quantifying the amount of triglyceride accumulated in the cells. The cells were washed with PBS (−) and air-dried, and Lab Assay Triglyceride Kit Wako was added thereto at 100 μL / well. The mixture was allowed to stand at room temperature for 30 to 60 minutes, and the absorbance at 630 nm was measured to quantify triglycerides.
The results are shown in FIG. The compound represented by the chemical formula (1) according to the present invention inhibited the differentiation of 3T3-L1 cells into adipocytes by 50% at a concentration of 420 nM. On the other hand, Fr. The compound isolated from 3-5-5 (Fr. 3-5-5-2, 3-5-5-3) did not show an activity of inhibiting differentiation into adipocytes.

(2)細胞生存率による細胞毒性
細胞毒性としては細胞生存率を指標として用いて評価した。上記(C)の方法で剥離、遠心分離して細胞数計測を行った細胞は、1×10個/wellとなるように96wellプレートに播種した。16時間培養後、特定濃度に調整した化学式(1)で示される化合物を含む増殖培地に置き換え、さらに48時間培養を続けた。Cell Counting Kit−8を5μL/wellずつ添加して1時間培養後、波長450nmの吸光度を測定し、細胞生存率を算出した。
本発明に係る化学式(1)で示される化合物は3T3−L1細胞に対しては50μMの濃度でも顕著な毒性を示さなかった。また、Hela細胞を用いた実験においても、同様に50μMの濃度で顕著な毒性を示さなかった。
(2) Cytotoxicity by cell viability Cytotoxicity was evaluated using cell viability as an index. Cells that were detached and centrifuged by the method of (C) above and the number of cells was counted were seeded on a 96-well plate at 1 × 10 4 cells / well. After culturing for 16 hours, the culture medium was replaced with a growth medium containing a compound represented by the chemical formula (1) adjusted to a specific concentration, and culturing was continued for further 48 hours. Cell Counting Kit-8 was added in an amount of 5 μL / well and cultured for 1 hour, and then the absorbance at a wavelength of 450 nm was measured to calculate the cell viability.
The compound represented by the chemical formula (1) according to the present invention did not show significant toxicity to 3T3-L1 cells even at a concentration of 50 μM. Similarly, in experiments using Hela cells, no significant toxicity was observed at a concentration of 50 μM.

(3)脂肪細胞内トリグリセリド減少
脂肪細胞内の脂肪滴の縮小を確認するために、脂肪細胞内グリセリドの減少を検討した。上記の方法で完全に分化させた3T3−L1脂肪細胞を、増殖培地で特定濃度に希釈した化学式(1)で示される化合物の存在下で培養を行い、培地は3日おきに試料を含む新鮮なものに置き換えた。7〜9日後に、上述のラボアッセイトリグリセライドキットワコーを用いて細胞内トリグリセリド量を定量した。結果を図4に示す。本発明に係る化学式(1)で示される化合物が脂肪細胞内のトリグリセリドレベルを減少させることから、本発明に係る化学式(1)で示される化合物は脂肪細胞内の脂肪滴の縮小に有効であることが判明した。
(3) Adipocyte triglyceride reduction In order to confirm the reduction of lipid droplets in fat cells, the reduction of fat cell glycerides was examined. The 3T3-L1 adipocytes fully differentiated by the above method are cultured in the presence of the compound represented by the chemical formula (1) diluted to a specific concentration in a growth medium, and the medium is fresh with a sample every 3 days. I replaced it with something. Seven to nine days later, the amount of triglycerides in the cells was quantified using the above-described laboratory assay triglyceride kit Wako. The results are shown in FIG. Since the compound represented by the chemical formula (1) according to the present invention reduces the triglyceride level in the fat cells, the compound represented by the chemical formula (1) according to the present invention is effective for reducing the lipid droplets in the fat cells. It has been found.

(4)乳酸の増加
3T3−L1を分化させた脂肪細胞あるいはHela細胞を、60mmディッシュに1×10個播種し、16時間培養し、その後、任意濃度の化学式(1)で示される化合物の存在下で、さらに72時間培養を続けた。その培養液を回収し、15000rpmで15分間遠心し、細胞残滓を除いた。これをHPLC(Develosil ODS HG−5 4.6X250mm)にて分析したところ、化学式(1)で示される化合物の濃度に依存して乳酸ピークが増加することが観察された。結果を図5に示す。チャートは、下から、それぞれ、0、0.05μM、0.1μMの化学式(1)で示される化合物で処理した3T3−L1を分化させた脂肪細胞、および0.1μMで同様に処理したHela細胞での結果を示す。11分過ぎのピークが乳酸を示しており、細胞種にかかわらず、化学式(1)で示される化合物の濃度に依存して乳酸ピークの増加を示している。
(4) Increase in lactic acid Adipocytes or Hela cells differentiated from 3T3-L1 were seeded in 1 × 10 5 cells in a 60 mm dish, cultured for 16 hours, and then the compound represented by the chemical formula (1) at an arbitrary concentration The culture was continued for an additional 72 hours in the presence. The culture solution was collected and centrifuged at 15000 rpm for 15 minutes to remove cell debris. When this was analyzed by HPLC (Develosil ODS HG-5 4.6 × 250 mm), it was observed that the lactic acid peak increased depending on the concentration of the compound represented by the chemical formula (1). The results are shown in FIG. From the bottom, the chart shows fat cells differentiated from 3T3-L1 treated with the compound represented by chemical formula (1) of 0, 0.05 μM and 0.1 μM, respectively, and Hela cells similarly treated with 0.1 μM. Results are shown. The peak after 11 minutes indicates lactic acid, and the lactic acid peak increases depending on the concentration of the compound represented by the chemical formula (1) regardless of the cell type.

本発明の化学式(1)で示される化合物は、脂肪細胞への分化阻害活性を有すると共に、脂肪細胞内のトリグリセリドレベルを低減する活性を有することから、脂肪細胞内の脂肪滴の縮小活性を有するものであり、抗肥満剤としての医薬組成物として有用である。   The compound represented by the chemical formula (1) of the present invention has an activity of inhibiting differentiation into adipocytes, and also has an activity of reducing triglyceride levels in adipocytes, and thus has a reduction activity of lipid droplets in adipocytes. It is useful as a pharmaceutical composition as an anti-obesity agent.

Claims (3)

次の化学式(1)で示される化合物。
Figure 0006292619
(化学式(1))
The compound shown by following Chemical formula (1).
Figure 0006292619
(Chemical formula (1))
請求項1記載の化合物を有効成分として含有することを特徴とする抗肥満薬。   An anti-obesity drug comprising the compound according to claim 1 as an active ingredient. 脂肪細胞内の細胞滴を縮小させることを特徴とする請求項2記載の抗肥満薬。   3. The anti-obesity drug according to claim 2, wherein cell droplets in fat cells are reduced.
JP2014137892A 2014-07-03 2014-07-03 Anti-obesity drugs Expired - Fee Related JP6292619B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2014137892A JP6292619B2 (en) 2014-07-03 2014-07-03 Anti-obesity drugs

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
JP2014137892A JP6292619B2 (en) 2014-07-03 2014-07-03 Anti-obesity drugs

Publications (2)

Publication Number Publication Date
JP2016013996A JP2016013996A (en) 2016-01-28
JP6292619B2 true JP6292619B2 (en) 2018-03-14

Family

ID=55230511

Family Applications (1)

Application Number Title Priority Date Filing Date
JP2014137892A Expired - Fee Related JP6292619B2 (en) 2014-07-03 2014-07-03 Anti-obesity drugs

Country Status (1)

Country Link
JP (1) JP6292619B2 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111996135B (en) * 2020-07-22 2022-04-08 中国科学院青岛生物能源与过程研究所 High-temperature-resistant high-carbohydrate-yield hot spring cyanobacteria and application thereof

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH09143072A (en) * 1995-11-22 1997-06-03 Yamanouchi Pharmaceut Co Ltd Suppressing agent for cytokine generation
AU2002216364A1 (en) * 2001-12-11 2003-06-23 The Kitasato Institute Novel substance fki-1083 and process for producing the same

Also Published As

Publication number Publication date
JP2016013996A (en) 2016-01-28

Similar Documents

Publication Publication Date Title
CN110105200B (en) Substituted aromatic compounds for treating fibrosis and related methods
JP6620096B2 (en) Pyranochromenylphenol derivative and pharmaceutical composition for treating metabolic syndrome or inflammatory disease
US8119839B2 (en) Carboxylic acid and antidepressant composition containing the same as active ingredient
JP5590286B2 (en) Novel Ca2 + signaling inhibitor
CN111356468B (en) Composition for preventing or treating fibrotic diseases comprising Rhus succedanea extract as active ingredient
JP5777011B2 (en) Composition for preventing or treating bone disease comprising colforsin daropate
JP6292619B2 (en) Anti-obesity drugs
CN106456594A (en) Ppar gamma activating agent
CN110139658A (en) Composition
KR101449575B1 (en) Composition comprising tenuifoliside A for preventing or treating inflammatory diseases
JP2019089759A (en) Ecklonia cava extract and pharmaceutical composition for preventing or treating vascular diseases comprising the same as an active ingredient
KR20140142580A (en) Composition for prevention and treatment of inflammatory diseases comprising N-trans-ρ-caffeoyl tyramine compound isolated from Tribulus terrestris
JP2013227256A (en) Vascular endothelial function improvement agent
KR20180077997A (en) Novel flavonoid compound from Stauntonia hexaphyll leaf extract and composition for anti-inflammatory, and improvement of bone tissue generation or cartilage tissue generation
KR20160125093A (en) Method for Seperation of Compound Derived from Ginseng and Composition for anti-inflammatory Using the same
KR101971438B1 (en) A pharmaceutical composition for preventing or treating vascular inflammation diseases comprising an Lespedeza cuneata ultrasound extract as an active ingredient
KR101305555B1 (en) Composition for treatment and prevention of bone diseases comprising extract of magnoliae flos&#39;s active components
KR101655554B1 (en) Pharmaceutical composition for prevention or treatment bone diseases comprising Alisma canaliculatum extract or fractions thereof, or compounds isolated from therefrom
KR101793654B1 (en) Pharmaceutical composition or functional food containing malaxinic acid for improvement of lipid related metabolic diseases
JP2016124850A (en) Anti-obesity agent
WO2018151334A1 (en) Xanthine oxidase inhibitor and method for producing same
KR20190126214A (en) Composition for preventing and treating cardiovascular disease comprising lipid extract of Mytilus galloprovincialis
KR101470613B1 (en) Composition comprising latifolin for preventing or treating inflammatory diseases
KR101961193B1 (en) Anti-viral Composition Comprising the Extract of Lemongrass Containing Aromatic Essential Oil
KR101320975B1 (en) Composition for treatment and prevention of bone diseases comprising extract of magnoliae flos&#39;s active components

Legal Events

Date Code Title Description
A621 Written request for application examination

Free format text: JAPANESE INTERMEDIATE CODE: A621

Effective date: 20170518

TRDD Decision of grant or rejection written
A977 Report on retrieval

Free format text: JAPANESE INTERMEDIATE CODE: A971007

Effective date: 20171221

A01 Written decision to grant a patent or to grant a registration (utility model)

Free format text: JAPANESE INTERMEDIATE CODE: A01

Effective date: 20180109

A61 First payment of annual fees (during grant procedure)

Free format text: JAPANESE INTERMEDIATE CODE: A61

Effective date: 20180207

R150 Certificate of patent or registration of utility model

Ref document number: 6292619

Country of ref document: JP

Free format text: JAPANESE INTERMEDIATE CODE: R150

R250 Receipt of annual fees

Free format text: JAPANESE INTERMEDIATE CODE: R250

LAPS Cancellation because of no payment of annual fees