JPH0617304B2 - Anti-cancer drug - Google Patents

Anti-cancer drug

Info

Publication number
JPH0617304B2
JPH0617304B2 JP57157103A JP15710382A JPH0617304B2 JP H0617304 B2 JPH0617304 B2 JP H0617304B2 JP 57157103 A JP57157103 A JP 57157103A JP 15710382 A JP15710382 A JP 15710382A JP H0617304 B2 JPH0617304 B2 JP H0617304B2
Authority
JP
Japan
Prior art keywords
cells
differentiation
activity
added
test
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 - Lifetime
Application number
JP57157103A
Other languages
Japanese (ja)
Other versions
JPS5946217A (en
Inventor
清 磯野
健一 旭
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.)
RIKEN Institute of Physical and Chemical Research
Original Assignee
RIKEN Institute of Physical and Chemical Research
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 RIKEN Institute of Physical and Chemical Research filed Critical RIKEN Institute of Physical and Chemical Research
Priority to JP57157103A priority Critical patent/JPH0617304B2/en
Publication of JPS5946217A publication Critical patent/JPS5946217A/en
Publication of JPH0617304B2 publication Critical patent/JPH0617304B2/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Description

【発明の詳細な説明】 本発明は、癌細胞に対して分化誘導活性を示す下記の化
合物: または を有効成分として含む制癌剤に関するものである。
DETAILED DESCRIPTION OF THE INVENTION The present invention provides the following compounds which show differentiation-inducing activity on cancer cells: Or The present invention relates to an anticancer drug containing as an active ingredient.

従来、癌化学療法剤として、アルキル化剤(ナイトロジ
エンマスタード類、エチレンイミン類、スルホン酸エス
テル類)、代謝拮抗物質(葉酸拮抗剤、プリン拮抗剤、
ピリミジン拮抗剤)、植物性核分裂毒(コルセミド、ビ
ンブラスチン等)、抗生物質(ザルコマイシン、カルチ
ノフイリン、マイトマイシン等)、ホルモン類(副腎ス
テロイド、男性ホルモン、女性ホルモン)及びポルフイ
リン錯酸(マーフイリン、copp)等が用いられている。
しかしながら、その殆んどは、細胞毒型の物質であり、
重大な副作用を呈するため、低毒性で優れた制癌活性を
有する制癌剤の開発が強く望まれている。
Conventionally, as cancer chemotherapeutic agents, alkylating agents (nitrodien mustards, ethyleneimines, sulfonates), antimetabolites (folate antagonists, purine antagonists,
Pyrimidine antagonist), phytofission venom (corsemide, vinblastine, etc.), antibiotics (sarcomycin, carcinophylline, mitomycin, etc.), hormones (adrenal steroids, male hormones, female hormones), and porphyrin complex acid (murphyrin, copp) Etc. are used.
However, most of them are cytotoxic substances,
Since it has serious side effects, it is strongly desired to develop an antitumor agent having low toxicity and excellent antitumor activity.

そこで、本発明者らは、上記の趣旨に鑑み、低毒性で制
癌活性を有する物質について探索、鋭意研究の結果、前
記のフラボノイド又はイソフラボノイド化合物が動物の
腫瘍細胞に対して分化誘導活性を有することを新たに見
出し、且つ該物質が著しく低毒性で、優れた制癌活性を
有することの新たな知見を得て、本発明の制癌剤を完成
するに至つた。本発明の制癌剤の有効成分は、人、家
畜、犬、描等の温血動物に対する優れた癌化学療法剤と
なり得るものである。
Therefore, in view of the above-mentioned gist, the present inventors searched for a substance having low toxicity and antitumor activity, and as a result of earnest research, the flavonoid or isoflavonoid compound has a differentiation-inducing activity on animal tumor cells. The present inventors have newly found that they have, and obtained new knowledge that the substance has extremely low toxicity and excellent antitumor activity, and completed the antitumor agent of the present invention. The active ingredient of the antitumor agent of the present invention can be an excellent cancer chemotherapeutic agent for warm-blooded animals such as humans, livestock, dogs and animals.

本発明の有効成分であるフラボノイド又はイソフラボノ
イドは、次の化合物である。
The flavonoids or isoflavonoids which are the active ingredients of the present invention are the following compounds.

(1)フラボン(Flavone) (2)フイセチン(Fisetin):3,3′,4′,7−tetr
ahydroxyflavone (3)ナリンゲニン(Naringenin):4′,5,7−trihy
droxyflavanone (4)フラバノン(Flavanone) これらの化合物は、第1表に示す如く、いずれも公知の
化合物であり、構造式及び物理的性質は次の通りである
(以下、上記化合物番号をもつて示す。)。
(1) Flavone (2) Fisetin: 3,3 ', 4', 7-tetr
ahydroxyflavone (3) Naringenin: 4 ', 5,7-trihy
droxyflavanone (4) Flavanone (Flavanone) As shown in Table 1, all of these compounds are known compounds, and their structural formulas and physical properties are as follows (hereinafter, indicated by the above compound numbers). .).

本発明の制癌剤は、経口及び非経口投与のいずれも使用
可能であり、経口投与する場合は、軟・硬カプセル剤又
は錠剤、顆粒剤、細粒剤、散剤として投与され、非経口
投与する場合は、水溶性懸濁液、油性製剤などの皮下或
いは静脈注射剤、点滴剤及び固体状又は懸濁粘稠液状と
して持続的な粘膜吸収が維持できるように坐薬のような
剤型で投与され得る。
The carcinostatic agent of the present invention can be used either orally or parenterally, and when orally administered, it is administered as soft / hard capsules or tablets, granules, fine granules, powders, and parenterally. Can be administered in subcutaneous or intravenous injections such as aqueous suspensions, oily preparations, infusions and solid or suspension viscous liquids in the form of suppositories so that sustained mucosal absorption can be maintained. .

本発明の有効成分の製剤化は、界面活性剤、賦形剤、滑
沢剤、佐剤、及び必要に応じて腸溶性製剤とするために
医薬的に許容し得る皮膜形成物質、コーテイング助剤等
を用いて適宜行うことができ、その具体例を挙げれば、
次のとおりである。
The active ingredient of the present invention is formulated by a surfactant, an excipient, a lubricant, an adjuvant, and a pharmaceutically acceptable film-forming substance or a coating aid to form an enteric-coated preparation as required. Can be appropriately performed by using, for example,
It is as follows.

本発明の組成物の崩壊、溶出を良好ならしめるために、
界面活性剤、例えばアルコール、エステル類、ポリエチ
レングリコール誘導体、ソルビタンの脂肪酸エステル
類、硫酸化脂肪アルコール類等の1種又は2種以上を添
加することができる。
In order to ensure good disintegration and dissolution of the composition of the present invention,
Surfactants such as alcohols, esters, polyethylene glycol derivatives, fatty acid esters of sorbitan, and sulfated fatty alcohols can be added alone or in combination of two or more.

また、賦形剤として、例えば蔗糖、乳糖、デンプン、結
晶セルロース、マンニツト、軽質無水珪酸、アルミン酸
マグネシウム、メタ珪酸アルミン酸マグネシウム、合成
珪酸アルミニウム、炭酸カルシウム、炭酸水素ナトリウ
ム、リン酸水素カルシウム、カルボキシメチルセルロー
スカルシウム等の1種又は2種以上を組合せて添加する
ことができる。
Further, as an excipient, for example, sucrose, lactose, starch, crystalline cellulose, mannitol, light anhydrous silicic acid, magnesium aluminate, magnesium metasilicate aluminate, synthetic aluminum silicate, calcium carbonate, sodium hydrogen carbonate, calcium hydrogen phosphate, carboxy One kind or two or more kinds such as methylcellulose calcium can be added in combination.

滑沢剤としては、例えばステアリン剤マグネシウム、タ
ルク、硬化油等を1種又は2種以上添加することがで
き、また矯味剤及び矯臭剤として、食塩、サツカリン、
糖、マンニツト、オレンジ油カンゾウエキス、クエン
酸、ブドウ糖、メントール、ユーカリ油、リンゴ酸等の
甘味剤、香料、着色料、保存料等を含有させてもよい。
As the lubricant, for example, magnesium stearate, talc, hardened oil and the like can be added alone or in combination of two or more, and as a flavoring agent and a flavoring agent, salt, satukaline,
Sweeteners such as sugar, mannitol, orange oil licorice extract, citric acid, glucose, menthol, eucalyptus oil, malic acid, etc., flavors, colorants, preservatives and the like may be contained.

懸濁剤、湿潤剤の如き佐剤としては、例えばココナツト
油、オリーブ油、ゴマ油、落花生油、乳酸カルシウム、
ベニバナ油、大豆リン脂質等を含有させることができ
る。
Examples of adjuvants such as suspending agents and wetting agents include coconut oil, olive oil, sesame oil, peanut oil, calcium lactate,
Safflower oil, soybean phospholipid, etc. can be contained.

また皮膜形成物質としては、セルロース、糖類等の炭水
化物誘導体として酢酸フタル酸セルロース(CAP)、
またアクリル酸系共重合体、二塩基酸モノエステル類等
のポリビニル誘導体としてアクリル酸メチル・メタアク
リル酸共重合体、メタアクリル酸メチル・メタアクリル
酸共重合体が挙げられる。
Further, as a film-forming substance, cellulose, cellulose acetate phthalate (CAP) as a carbohydrate derivative such as saccharide,
Examples of polyvinyl derivatives such as acrylic acid-based copolymers and dibasic acid monoesters include methyl acrylate / methacrylic acid copolymers and methyl methacrylate / methacrylic acid copolymers.

また、上記皮膜形成物質をコーテイングするに際し、通
常使用されるコーテイング助剤、例えば可塑剤の他、コ
ーテイング操作時の薬剤相互の付着防止のための各種添
加剤を添加することによつて皮膜形成剤の性質を改良し
たり、コーテイング操作をより容易ならしめることがで
きる。なお、有効成分を皮膜形成物質を用いてマイクロ
カプセル化してから賦形剤等と混合した剤型としても良
い。
In addition, when coating the above film-forming substance, a coating-forming agent which is usually used, for example, a plasticizer and various additives for preventing mutual adhesion of chemicals during the coating operation are added to form a film-forming agent. You can improve the properties of the and make the coating operation easier. The active ingredient may be microencapsulated using a film-forming substance and then mixed with an excipient or the like to give a dosage form.

特に代表的な剤型における配合比は下記の通りである。In particular, the compounding ratio in a typical formulation is as follows.

特に好ましい賦形剤は、乳糖、結晶セルローズ、カルボ
キシメチルセルロースカルシウムである。
Particularly preferred excipients are lactose, crystalline cellulose and carboxymethyl cellulose calcium.

また、投与量は、対象腫瘍を有効に治療するに十分な量
であり、腫瘍の症状、投与経路、剤型などによつて左右
されるが、一般に、経口投与の場合、大人では1日当
り、約0.01〜100mg/kg体重(小人では、0.01〜60
mg/kg体重)の範囲で、その上限は好ましくは約50mg
/kg体重、更に好ましくは約10mg/kg体重程度であ
り、非経口投与の場合、その上限は約10mg/kg体重程
度であり、好ましくは5mg〜kg体重、更に好ましくは2
mg/kg体重が適当である。
In addition, the dose is an amount sufficient to effectively treat the target tumor and depends on the tumor symptom, the administration route, the dosage form, etc. About 0.01 to 100 mg / kg body weight (0.01 to 60 for dwarfs)
mg / kg body weight), the upper limit is preferably about 50 mg
/ Kg body weight, more preferably about 10 mg / kg body weight. In the case of parenteral administration, the upper limit is about 10 mg / kg body weight, preferably 5 mg to kg body weight, more preferably 2 mg / kg body weight.
mg / kg body weight is appropriate.

次に、本発明の化合物の制癌活性を確認した制癌性試験
について述べる。
Next, a carcinostatic test confirming the carcinostatic activity of the compound of the present invention will be described.

〔1〕フレンド白血病細胞(mouse erythroid leukemia
cell,B8細胞)に対する試験GIBCO製HAMの
F−12培地に、15%の牛胎児血清及び60mg/lの
カナマイシンを加えたものに、25×104cell/mlとな
るようにB8細胞を接種し、これに所定量の被験化合物
を加える(最終容量5ml)。
[1] Friend leukemia cell (mouse erythroid leukemia
cell, B8 cells) GIBCO HAM F-12 medium supplemented with 15% fetal bovine serum and 60 mg / l kanamycin was inoculated with B8 cells at 25 × 10 4 cells / ml. Then, a predetermined amount of the test compound is added thereto (final volume 5 ml).

7.5%CO2中、37℃7日間培養した後、オルキン(Orki
n)のベンジジン染色法により染色し、染色された細胞
数、すなわち、赤血球への分化によりヘモグロビンを生
成するようになつた細胞数を測定し、分化誘導率を求め
る。
After culturing in 7.5% CO 2 at 37 ° C for 7 days,
The number of cells stained by the benzidine staining method of n), that is, the number of cells adapted to produce hemoglobin due to differentiation into erythrocytes is measured, and the differentiation induction rate is obtained.

〔2〕マウス骨髄性白血病細胞(mouse myeloid leukem
ia cell,M1)に対する試験 GIBCO製イーグルMEM培地に、10%の馬血清及
び60mg/lのカナマイシンを加えたものに、5.0×10
4cell/mlとなるようはM1細胞を接種し、これに所定
量の被験化合物を加える(最終容量5ml)。
[2] Mouse myeloid leukem
Test for ia cell, M1) GIBCO Eagle MEM medium supplemented with 10% horse serum and 60 mg / l kanamycin 5.0 × 10
M1 cells are inoculated to obtain 4 cells / ml, and a predetermined amount of the test compound is added thereto (final volume 5 ml).

7.5%CO2中、37℃7日間培養した後、貧食細胞、ある
いは顆粒球への分化により誘導されたリゾチーム活性を
調べる。なお、リゾチーム活性の1単位(unit)とは、
ミクロコツカス・リソデイクテイカス(Micrococcuslys
odeikticus)菌体の懸濁液を基質として、リゾチームを
作用させ、pH6.24、温度25℃で測定し、450mμの
波長の吸光度を毎分0.001減少させるようなリゾチーム
の量をいう。
After culturing in 7.5% CO 2 at 37 ° C. for 7 days, lysozyme activity induced by differentiation into phagocytes or granulocytes is examined. In addition, 1 unit of lysozyme activity (unit),
Micrococcus lycus
odeikticus) A suspension of microbial cells is used as a substrate, and lysozyme is allowed to act on it, and the amount of lysozyme is measured at pH 6.24 and a temperature of 25 ° C. to reduce the absorbance at a wavelength of 450 mμ by 0.001 per minute.

〔3〕マウス奇形腫細胞(mouse teratocarcinoma)に
対する試験:テラトーマ細胞をマウスの腹腔から腹腔へ
移植後、1ケ月経過したものを用いた。テラトーマ細胞
は、腹腔中では初期胚に似た胚様体(embroid body)と
いう細胞塊として存在し、それらをトリプシン処理など
を行うことなく用いた。採取した腹水中で自然沈下させ
て得られる胚様体をダルベコー変法培地、あるいはハン
クス液で3度洗浄後、10%牛胎児血清を含む培地に接
種し、所定量の被験化合物を加え、37℃でCO27.5〜8
%を含む水蒸気を飽和して、空気中で1週間培養する。
遠心分離(2000r.p.m.10分)して得た胚様体を0.
86%NaCl溶液で洗浄後、ナフトールAS−MXホスフエ
ートとジアゾ試薬(Fast Violet B Salt)を加えて1時
間室温で放置する。これを遠心分離(2000r.p.m.、
10分)して胚様体を分離し、エタノールを加えて1時
間室温で放置する。
[3] Test for mouse teratocarcinoma: One month after teratoma cells were transplanted from the abdominal cavity of the mouse to the abdominal cavity, the cells were used. Teratoma cells exist in the abdominal cavity as a cell mass called an embroid body similar to the early embryo, and they were used without trypsin treatment. The embryoid body obtained by spontaneously submerging in the collected ascites was washed 3 times with a modified Darbeco's medium or Hanks' solution, and then inoculated into a medium containing 10% fetal bovine serum, and a predetermined amount of the test compound was added to the medium. CO 2 7.5-8 at ℃
% Of water vapor is saturated and incubated in air for 1 week.
The embryoid body obtained by centrifugation (2000 rpm 10 minutes) was
After washing with 86% NaCl solution, naphthol AS-MX phosphate and diazo reagent (Fast Violet B Salt) are added and left at room temperature for 1 hour. This is centrifuged (2000r.pm,
After 10 minutes), the embryoid bodies are separated, ethanol is added, and the mixture is left at room temperature for 1 hour.

(未分化の細胞は、赤く着色する)。(Undifferentiated cells are colored red).

これを、535nmの吸収を測定し、アルカリホスフアタ
ーゼ活性(分化誘導の程度)を求める。
The absorption at 535 nm is measured to determine the alkaline phosphatase activity (degree of differentiation induction).

ヘキサメチレンビスアセトアミド(HMBA)5mMを加
えた場合(アルカリホスフアターゼ活性を全く示さな
い。)を「++」とし、HMBAを加えない場合(アルカリホ
スフアターゼ活性を極めて強く示す。)を「--」とし、
分化誘導の程度を次の段階で示した。
The case where 5 mM of hexamethylene bisacetamide (HMBA) was added (has no alkaline phosphatase activity at all) was designated as "++", and the case where HMBA was not added (has extremely strong alkaline phosphatase activity). --"age,
The degree of differentiation induction was shown in the next step.

++:アルカリホスフアターゼ活性を全く示さない。++: shows no alkaline phosphatase activity at all.

+:アルカリホスフアターゼ活性をほとんど示さない。+: Almost no alkaline phosphatase activity is shown.

+:アルカリホスフアターゼ活性を若干示す。+: Shows some alkaline phosphatase activity.

−:アルカリホスフアターゼ活性を強く示す。-: Strongly shows alkaline phosphatase activity.

−−:アルカリホスフアターゼ活性を極めて強く示す。-: Extremely strong alkaline phosphatase activity.

なお、後述の試験例では、分化誘導作用をもつて、制癌
活性を示した。
In addition, in the test example described later, it has anti-tumor activity by having a differentiation inducing action.

以下に、本発明を製剤例及び試験例によつて具体的に説
明する。
Hereinafter, the present invention will be specifically described with reference to formulation examples and test examples.

製剤例(腸溶性カプセル剤)化合物(1)5g、乳糖2.46
g及びヒドロキシプロピルセルロース0.04gを各々と
り、よく混合した後、常法に従つて粒状に成形し、これ
をよく乾燥して篩別してビン、ヒートシール包装などに
適した顆粒剤を製造する。次に、酢酸フタル酸セルロー
ス0.5g及びヒドロキシプロピルメチルセルロースフタ
レート0.5gを溶解して被覆基材となし、前記顆粒を浮
遊流動させつゝこの基材を被覆して腸溶性の顆粒剤とす
る。この組成物をカプセルに充填して腸溶性カプセル製
剤100個を製造する。
Formulation example (enteric coated capsule) Compound (1) 5 g, lactose 2.46
g and hydroxypropylcellulose (0.04 g) are mixed well, and then the mixture is shaped into granules by a conventional method, dried well and sieved to produce granules suitable for bottles, heat-sealed packaging and the like. Next, 0.5 g of cellulose acetate phthalate and 0.5 g of hydroxypropylmethylcellulose phthalate are dissolved to form a coated base material, and the above granules are allowed to float and flow, and this base material is coated to form an enteric coated granule. This composition is filled into capsules to produce 100 enteric-coated capsule preparations.

試験例 第1表の化合物を用い、前記試験法〔1〕,〔2〕及び
〔3〕より、フレンド白血病細胞の分化誘導率、マウス
骨髄性白血病細胞の分化誘導によるリゾチーム活性及び
マウス奇形腫細胞の分化誘導程度を調べたところ、それ
ぞれ、第2表、第3表及び第4表に示す結果が得られ
た。
Test Example From the test methods [1], [2] and [3] using the compounds of Table 1, the induction rate of differentiation of friend leukemia cells, lysozyme activity by induction of differentiation of mouse myeloid leukemia cells and mouse teratoma cells When the degree of differentiation induction was examined, the results shown in Table 2, Table 3 and Table 4 were obtained.

上記試験例の結果から明らかなように、本発明の各種フ
ラボノイド及びイソフラボノイドは癌細胞に対して、正
常細胞への分化誘導作用を示すことから、毒性の少ない
優れた制癌活性を示すことが立証された。
As is clear from the results of the above Test Examples, the various flavonoids and isoflavonoids of the present invention show an effect of inducing differentiation into normal cells against cancer cells, and thus exhibit excellent antitumor activity with low toxicity. Proven.

フロントページの続き (56)参考文献 特公 昭53−23832(JP,B1) 特公 昭53−23831(JP,B1) Journal of Nationa l Products vol.42,N o.1,P.85〜P.91,(1979)Front Page Continuation (56) References Japanese Patent Publication No. 53-23832 (JP, B1) Japanese Patent Publication No. 53-23831 (JP, B1) Journal of National Products vol. 42, No. 1, P. 85-P. 91, (1979)

Claims (1)

【特許請求の範囲】[Claims] 【請求項1】癌細胞に対して分化誘導活性を示す下記の
化合物: または を有効成分として含む制癌剤。
1. The following compound showing a differentiation-inducing activity against cancer cells: Or An anticancer agent containing as an active ingredient.
JP57157103A 1982-09-09 1982-09-09 Anti-cancer drug Expired - Lifetime JPH0617304B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP57157103A JPH0617304B2 (en) 1982-09-09 1982-09-09 Anti-cancer drug

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
JP57157103A JPH0617304B2 (en) 1982-09-09 1982-09-09 Anti-cancer drug

Publications (2)

Publication Number Publication Date
JPS5946217A JPS5946217A (en) 1984-03-15
JPH0617304B2 true JPH0617304B2 (en) 1994-03-09

Family

ID=15642295

Family Applications (1)

Application Number Title Priority Date Filing Date
JP57157103A Expired - Lifetime JPH0617304B2 (en) 1982-09-09 1982-09-09 Anti-cancer drug

Country Status (1)

Country Link
JP (1) JPH0617304B2 (en)

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2633182B1 (en) * 1988-06-23 1993-07-23 Beljanski Mirko ANTI-CANCER PHARMACEUTICAL COMPOSITION AND METHOD OF USING THE INVENTION
KR100447918B1 (en) * 1996-07-25 2005-09-28 동아제약주식회사 Flavones and flavanone compounds with protective gastrointestinal tract including large intestine
FR2781153B1 (en) * 1998-07-15 2001-08-03 Lafon Labor FLAVONOID-BASED THERAPEUTIC COMPOSITION FOR USE IN THE TREATMENT OF TUMORS WITH CYTOTOXIC AGENTS
FR2781154B1 (en) * 1998-07-15 2001-09-07 Lafon Labor THERAPEUTIC COMPOSITION BASED ON ISOFLAVONOIDS FOR USE IN THE TREATMENT OF TUMORS WITH CYTOTOXIC AGENTS
CA2346333A1 (en) * 1998-10-06 2000-04-13 Usda-Ars-Ott Compositions and methods of inhibiting neoplastic diseases with compounds related to limocitrin and 5-desmethyl sinensetin
WO2002005811A1 (en) * 2000-07-18 2002-01-24 Nichimo Co., Ltd. Stem cell reinforcing material
US20020160983A1 (en) * 2001-03-16 2002-10-31 Alberto Bargiotti Substituted benzopyranones as telomerase inhibitors
KR100436220B1 (en) * 2001-08-30 2004-06-12 주식회사 네패스 Organic polymers for bottom antireflective coating, processes for preparing the same, and compositions containing the same
US20030229136A1 (en) * 2002-04-18 2003-12-11 Nurulain Zaveri Novel flavanoids as chemotherapeutic, chemopreventive, and antiangiogenic agents
EP1686981A4 (en) * 2003-11-19 2011-02-23 Novogen Res Pty Ltd Combinational radiotherapy and chemotherapy compositions and methods
FR2923718B1 (en) * 2007-11-15 2009-12-18 Caudalie COMPOSITIONS OF FLAVONOIDIC POLYPHENOLIC DERIVATIVES AND THEIR APPLICATIONS TO COMBAT PATHOLOGIES AND AGING LIVING ORGANISMS
US9132117B2 (en) 2013-06-17 2015-09-15 Kgk Synergize, Inc Compositions and methods for glycemic control of subjects with impaired fasting glucose
US10966954B2 (en) * 2016-05-16 2021-04-06 Global Biolife Inc. Electrophilically enhanced phenolic compounds for treating inflammatory related diseases and disorders

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS5323832A (en) * 1976-08-19 1978-03-04 Tokyo Shibaura Electric Co Plating method on titanium or tantalum base
JPS5323831A (en) * 1976-08-19 1978-03-04 Seiko Instr & Electronics Indirect measuring method of gap between steel sheet and nozzle in gas spray system plating quantity controller device

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
JournalofNationalProductsvol.42,No.1,P.85〜P.91,(1979)

Also Published As

Publication number Publication date
JPS5946217A (en) 1984-03-15

Similar Documents

Publication Publication Date Title
JPH0367045B2 (en)
JPH0617304B2 (en) Anti-cancer drug
EP2094715B1 (en) Dipterinyl calcium pentahydrate (dcp) and therapeutic methods based thereon
JP3285410B2 (en) Oral pharmaceutical composition containing anthocyanosides
JPH027573B2 (en)
JPH07507288A (en) Desferrioxamine-B salts and their use as orally effective iron chelators
JPH0656669A (en) Pterine derivative preparation having active oxygen-scavenging action
JPS639493B2 (en)
JPH01203331A (en) Carcinostatic agent
US20200289593A1 (en) Spirulina carotenoid complex
JP3002700B2 (en) Anti-AIDS virus agent
JPH0720860B2 (en) Anti-cancer drug
JPH0249287B2 (en)
JPH09323935A (en) Antiviral agent, its production and application therefor
RU2181051C1 (en) Method to obtain injection preparation based upon a substance of p-vitamin activity
EP0082667A1 (en) Pharmaceutical compositions
JPH0367050B2 (en)
JPH01203330A (en) Carcinostatic agent
JPH01203322A (en) Carcinostatics
JP2768805B2 (en) Diyne compounds and anticancer drugs
JPS6352011B2 (en)
JPH0144196B2 (en)
JPS6261568B2 (en)
JPH0551566B2 (en)
JPS63264409A (en) Carcinostatic agent