TW200804411A - Certain lithocholic acid analogues that inhibit sialyltransferase - Google Patents

Certain lithocholic acid analogues that inhibit sialyltransferase Download PDF

Info

Publication number
TW200804411A
TW200804411A TW95124663A TW95124663A TW200804411A TW 200804411 A TW200804411 A TW 200804411A TW 95124663 A TW95124663 A TW 95124663A TW 95124663 A TW95124663 A TW 95124663A TW 200804411 A TW200804411 A TW 200804411A
Authority
TW
Taiwan
Prior art keywords
group
acid
compound
cyclopentane
chemical substance
Prior art date
Application number
TW95124663A
Other languages
Chinese (zh)
Other versions
TWI369361B (en
Inventor
Wen-Shan Li
Yi-Ching Wang
Hsueh-Fen Juan
Original Assignee
Academia Sinica
Univ Nat Taiwan
Univ Nat Taiwan Normal
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 Academia Sinica, Univ Nat Taiwan, Univ Nat Taiwan Normal filed Critical Academia Sinica
Priority to TW095124663A priority Critical patent/TWI369361B/en
Publication of TW200804411A publication Critical patent/TW200804411A/en
Application granted granted Critical
Publication of TWI369361B publication Critical patent/TWI369361B/en

Links

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

Certain chemical entities chosen from compounds of Formula I: and pharmaceutically acceptable salts, solvates, chelates, non-covalent complexes, prodrugs, and mixtures thereof are described. Pharmaceutical compositions comprising at least one chemical entity chosen from compounds of Formula I and a pharmaceutically acceptable vehicle are described. Also described are methods for inhibiting α-2,3-sialyltransferase activity in cells, and methods for treating a patient having a disease responsive to inhibition of α-2,3-sialyltransferase activity.

Description

200804411 九、發明說明· 【發明所屬之技術領域】 提供某種具膜穿透性、可抑制α-2,3唾液酸轉移酶活性 的石膽酸類似物及醫藥組合物,其包含至少一種具膜穿透 性、可抑制α-2,3唾液酸轉移陶:活性的石膽酸類似物;亦提 供抑制細胞内α_2,3唾液酸轉移酶活性的方法及一種治療因 α-2,3唾液酸轉移酶活性遭過度活化的疾病之方法。 【先前技術】 細胞蛋白質與脂質的醣化是許多正常細胞功能的整體 部分,除了醣化之外,包括唾液酸化,亦可能與癌細胞的 形成、轉移與侵襲有關。唾液酸化的過程是透過一種可將 唾液酸(一種在正常生理狀態下,帶負電荷的九碳胺糖) 轉移至糖結合物上的多醣成長鏈終端上的唾液酸轉移酶而 達成。高唾液酸化在細胞附著、免疫防禦及發炎扮演一個 重要的角色,而在許多腫瘤模型中,變化後之唾液酸轉移 酶活性亦涉及腫瘤的形成與侵襲(Harvey et al.,1992; Majuri et al., 1995; DallOlio and Chiricolo, 2001; Wang et al.9 2002)。具體來說,一種亞型的轉移酶-a-2,3唾液酸轉移酶 (a-2,3 ST)係因ras-致癌基因的過度表現而導致其向上調 節(Easton et al·,1991)。有效唾液酸轉移酶抑制劑的確認 代表癌症治療發展上的新興方法,包括針對預防腫瘤轉移 的療法。 許多唾液酸轉移酶抑制劑已經被發展出來,包括結構 上虛擬的過渡狀態磷類似物(Transition state anal〇gue )、雙 200804411 受質類似物(bisubstrate analogues)、donor analogue、基於 胞核苷-單鱗酸-N_乙St神經胺糖酸(cytidine monophosphate N-acetylneuraminic acid ; CMP-Neu5Ac)或雙糖戶斤形成的受 體類似物(acceptor analogues ) ( Skropeta et al·, Glycoconjugate J., 2004, 21:205; Chang, Tao and W.-S. Li, Synlett, 2004, 37; Whalen, McEvoy and Halcomb, Bioorg. Med. Chem. Lett., 2003, 13:301; Schwoerer and Schmidt, J. Am. Chem. Soc., 2002, 124:1632; Muller, Schaub and Schmidt, Anyw· C/zem” /ηί· 1998, 37:2893)。雖然這些化合物可 有效抑制唾液酸轉移酶,但由於對細胞膜的通透性不佳, 因此其臨床上之應用受到限制。同樣地,由於唾液酸係自 天然產物中純化而來,故僅能少量取得,限制了其在臨床 使用上的可利用性(Hsu et al·,2005 )。 有鑑於此,本發明之一目的在提供一新穎的石膽酸類 似物及含有可有效抑制α-2,3唾液酸轉移酶的石膽酸與改善 細胞通透性的醫藥上組合物。本發明進一步之目的在提供 對於α-2,3唾液酸轉移酶活性受過度活化所導致之疾病的治 療方法。 【發明内容】 提供至少一種選自化學式I所示化合物之化學物質: 7 200804411200804411 IX. OBJECTS OF THE INVENTION · Technical Fields Provided by the Invention There is provided a lithocholic acid analog and a pharmaceutical composition having membrane permeability and inhibiting α-2,3 sialyltransferase activity, which comprise at least one Membrane penetrability, inhibition of α-2,3 sialic acid transfer pottery: active choline acid analog; also provides a method for inhibiting intracellular α 2,3 sialyltransferase activity and a treatment for α-2,3 saliva A method in which an acid transferase activity is overactivated. [Prior Art] The saccharification of cellular proteins and lipids is an integral part of many normal cellular functions. In addition to saccharification, including sialylation, it may also be involved in the formation, metastasis and invasion of cancer cells. The process of sialylation is achieved by a sialyltransferase that transfers sialic acid, a negatively charged ninth carbonamine sugar under normal physiological conditions, to the sugar chain end of the sugar conjugate. High sialylation plays an important role in cell attachment, immune defense, and inflammation, and in many tumor models, altered sialyltransferase activity is also involved in tumor formation and invasion (Harvey et al., 1992; Majuri et al). 1995, DallOlio and Chiricolo, 2001; Wang et al. 9 2002). Specifically, a subtype of transferase-a-2,3 sialyltransferase (a-2,3ST) is up-regulated by overexpression of ras-oncogene (Easton et al., 1991). . Confirmation of potent sialyltransferase inhibitors represents an emerging approach to the development of cancer treatment, including therapies for the prevention of tumor metastasis. Many sialyltransferase inhibitors have been developed, including structurally transient transition state anal〇gues, double 200804411 bisubstrate analogues, donor analogues, cytosine-based Cyclic monophosphate N-acetylneuraminic acid (CMP-Neu5Ac) or acceptor analogues formed by disaccharide ( Skropeta et al., Glycoconjugate J., 2004, 21:205; Chang, Tao and W.-S. Li, Synlett, 2004, 37; Whalen, McEvoy and Halcomb, Bioorg. Med. Chem. Lett., 2003, 13:301; Schwoerer and Schmidt, J. Am. Chem. Soc., 2002, 124:1632; Muller, Schaub and Schmidt, Anyw·C/zem” /ηί· 1998, 37:2893). Although these compounds are effective in inhibiting sialyltransferase, they are mediated by cell membranes. Poor permeability, so its clinical application is limited. Similarly, since sialic acid is purified from natural products, it can only be obtained in small quantities, which limits its availability in clinical use (Hsu et al ·, 2005). In view of An object of the present invention is to provide a novel choline acid analog and a medicinal composition comprising choline acid which is effective for inhibiting α-2,3 sialyltransferase and improving cell permeability. Further, the present invention The object of the invention is to provide a method for the treatment of a disease caused by excessive activation of α-2,3 sialyltransferase activity. SUMMARY OF THE INVENTION At least one chemical selected from the compounds of formula I is provided: 7 200804411

及其醫藥上可接受之鹽類、溶劑化物、螯合劑、非共價複 合物、前驅藥物及其混合物。其中Ri為選擇性取代之被烷 基;R2係選自羥基及醯氧基;R3係為氫,或R2與R3連同 其所附著的竣,形成一個氧基(0X0 group);當Ri為 (/?)-4-carboxybutan-2-yl 時,貝1J R2 非為經基。進一步提供一 種醫藥組合物,該醫藥組合物係含有至少一種有療效性的 有效量之選自化學式I所示化合物之化學物質及其醫藥上 可接受之載體。 提供一種抑制α-2,3唾液酸轉移酶活性之方法,係包含 以一足以察覺使糖結合物唾液酸化減少之至少一種選自化 學式I所示化合物之化學物質的量與表現α-2,3唾液酸轉移 酶之細胞相接觸。進一步提供一種治療對α-2,3唾液酸轉移 酶過度活化活性減低有反應的病患的方法’該方法包含施 予病患至少一種選自化學式I所示化合物之化學物質之有 療效性有效量。 本發明之其他目的及優點將在以下的部分描述中提 出,熟悉該項技術人士可清楚的暸解,在不脫離下述申請 專利範圍的與精神下進行各種變化與改變,且本發明亦不 8 200804411 受限於說明書中所舉實施例的實施方式。 揭露的所有特徵都可能與其他方法結合,且呪^曰^所 個特徵都可能被其他具有相同、相等^相斤揭露的每一 取代。於說明書及所附申請專利範圍巾,的的特徵所 式,v,、,,an”及W,係包含複數,除非的單一形 定。 1谷有其他明確之規 【實施方式】 本發明之化學物質包括,但不限於化學式j所示化人 物,例如表一所示及其所有醫藥上可接受的形式:化二 物。在此所稱「醫藥上可接受的形式之化合物」包括醫越 上可接受的鹽類、溶劑化物、結晶體(包括多形體及 (clathrates)化合物)、螯合物、非共價化合物前驅藥物 混合物。在某些實施態樣中所描述的化合物係為醫藥上^ 接受之鹽類,因此,名詞「化學物質(chemical entity)」及「化 學物質群(chemicalentities)」亦包括醫藥上可接受的鹽類、 溶劑化物、結晶體(包括多形體及晶籠化合物)、螯合物、 非共價化合物前驅藥物及其混合物。 醯基係指化學基-C(〇)R,在此所指的R為烷基、取代 烧基、取代環烧基、取代雜環烷基、取代芳基或取代雜芳 基。代表例包括,但不限於乙醯基、環己基幾基 (cylcohexylcarbonyl)、 環己基曱基緩又 (cyclohexylmethylcarbonyl)、苯甲醯、笨基羰美 (benzylcarbonyl)及其類似物。 200804411 烧屬烴基(Alkanyl)指一飽和支鏈、直鏈或藉由從母炫 屬烴上的單一碳原子移除一氫原子所衍生而來的環烧基。 典型的烷屬烴基官能基包括,但不限於次甲基(methanyl)、 次乙基(ethanyl)、次丙基(pr〇panyis)(例如:丙燒+基、丙 烧-2-基(即異丙基)、環丙烷小基;次丁基(butanyls)例如: 丁烧-1-基、丁烧-1-基(此c-butyl)、2-曱基-丙烧小基(即異 丁基)、2-曱基-丙烷-2-基〇butyl)、環丁烷-1-基及其類似物。 烯烴基(Alkenyl)指一不飽和支鏈、直鏈或具有至少一由 母烯烴上的單一碳原子移除一氫原子所衍生而來具有碳一 1 炭雙鍵的環烧基。這些官能基的雙鍵可以是順式或反式組 態。典型的烯烴基官能基包括,但不限於乙烯基、丙烯基, 例如:丙-1-烯-1-基、丙小烯-2_基、丙_2_烯小基⑷lyl)、丙_2_ 烯-2-基、環丙-1-烯-;^基、環丙-^烯—卜基;丁烯基例如:丁 小稀-1-基、丁小晞-2-基、2-甲基-丙小烯小基、丁-2·烯-1-基、丁-2-烯-2-基、丁-i,3-二烯小基、丁 4,3_二烯-2-基、環 丁-1-烯-1-基、環丁-1-烯_3_基、環丁二烯―卜基,及其類 似物。在某些實施態樣中,一烯烴基官能基具有2-20個碳 原子;在其他實施態樣中,則具有2-6個碳原子。 烷氧基(Alkoxy)指一化學基—〇R,在此所指的r代表烷 基、、取代烷基、取代環烷基、取代雜環烷基、取代芳基或 ,代雜芳基。代表例包括,但不限於甲氧基、乙氧基、丙 虱基、丁氧基、環己氧基及其類似物。 烷氧羰基(AlkoxyCarb〇nyl)指一化學基_c(0)〜烷氧 土,其中烷氧基即上述所指的烷氧基。 200804411 烧基(Alkyl)指一飽和或不飽和支鏈、直鏈或由母烧屬 烴、,烴、或炔烴上的單一碳原子移除一氫原子所衍生而 來的環狀一價烴基。典型的烷基包括,但不限於甲烷基、 乙烷基(例如:次乙基、乙烯基、乙炔基)、丙烷基(例 如:丙烷-1-基、丙烷-2-基、環丙烷-1—基、丙_1_烯-1-基、 丙-l-烯士基、丙烯小基(allyl)、環丙小稀小基、環丙^ 烯-1-基、丙-丨-炔—:^基、丙炔基、丙-^炔“-基、丁烷 基例如:丁烷-1-基、丁烷_2_基)、2-甲基-丙烷基、孓甲基-丙燒-2-基、環丁统小基、丁小烯+基、丁+稀·基、2· :基-丙-1-烯-1·基、丁-2-烯-1-基、丁-2-烯-2-基、丁-U一二 =卜,、丁一1,3-二烯1基、環丁小·1基、環丁-1·烯-3-土 %丁-1,3-二烯-1-基、丁炔基、丁炔基、丁_3一 炔-1-基,及其類似物。 名詞烷基」特別指任何飽和程度的官能基,例如··具 特碳·碳單鍵的官能基、具有-個或多個碳·碳雙鍵的ΐ 絲具有一個或多個碳-碳三鍵的官能基及具有混合碳_ :早鍵、雙鍵及三鍵的官能基。依照一定的飽和度 屬烴基、烯烴基、及快烴基。在某些實施態樣中, 一 土匕έ 1-20個石反原子;在其他實施態樣中,烧基則包含 -個碳原子,被稱為低碳數烷基。 基(Alkynyl)指一未飽和支鏈、直鏈或由母炔烴上 鍵:了反原子移除一氫原子所衍生、具有至少-個碳-碳: =诚基。㈣㈣烴基包括,但不限於乙絲、= ^-执列如丙小块小基、丁快基,例如:丁小快小基、丁 、、-3-基、丁炔基及其類似物。在某些實施態樣中, 11 200804411 炔基具有2·2〇個碳原子;在其他貧於处 個碳原子。 、也恕樣中,則具有3_6 氨基係指一化學基-NH2。 月女基幾基(Aminocarbonyl)係指〜 此所指之R與R,係個別選自氫、埝(O)NRR官能基,在 環烷基、取代雜環烷基、取代芳基、、取代烷基、取代 地將氮原子附著於R與R,上,形^代雜芳基·,或任意 代雜環。 個或多個雜環或取 芳香基(Aryl)包含:五元及六元锼戸Μ 雙環系統,其中至少一環是碳環及〜香環,例如:苯; 滿及四氫萘;及三環系統,其中至少,例如:萘、茚 例如:芴。舉例來說,芳香環包括與^^是碳環及芳香環, 基環相接的五元及六元芳香破環,緊,元至七元雜環院 一個或多個選自氮、氧、硫的雜原:处,環烷基環係包含 合,在雙環系統中,只有一個是碳環I進行這樣的接 在碳環芳香環或雜環烷基環。二價美,接合點可能 形成且環原子具有自由價,被稱之;衍生物處 係衍生自單價的多環烴,其命名 代〖生本基。二價基 ?的:子原子上移除一個氫原子;添加字自具有j由 符合單價基。例如:具有兩個接合點的萃|en:」則是為 然而,芳香基並不包含或與 ^之為亞萘基。 覆,因此,若一個或多個…^&二二斤界疋之雜芳基重 則在此所μ驗統為麵相接合, 基所坑取芳代基==^燒基(缝州係指-環炫基被芳 厅取代騎絲上的氫原子結合至一個典型末端碳原 12 200804411 子或w3碳原子。典型芳烷基包括但不限於苯甲基、2 # 烷二ι-基、苯乙烯+基、萘基曱基、2_萘基乙垸土 =乙 2-萘基乙烯m、祕苯甲基、2·⑽笨基乙二ς、 其類似物。其中,特定㈣基團係以芳烧屬烴基t 及 ,士/或一芳炔烴基命名之。在某些實施態樣中,;烷:可烴 ifi三十個碳的芳烷基,例如:芳烷基的烷烴二二: 炔烴團y以是-至十個碳,芳烧基的芳香基團可以=或 二十個碳。 疋/、至 务氧幾基(Aryloxycarbonyl) 係指— 基-C(0)-〇-R,其中,在此所指之R係選自 ^予 芳香基。 曰方㈢基或取代 羰基係指一化學基—c(〇)。 在此所使用的名詞「癌症」,係指或描 生理狀況,其細胞群係具有不正常細 植動物的 所揭露以化合物治療的癌症’包括 it锊徵。目前 瘤、印巢癌、乳癌、子宮内膜癌括神=細胞 大腸直腸癌、結腸癌、消化道、肺癌、腎細^ 、、邑素瘤、 淋巴、攝護腺癌、胰臟癌、末期癌症、髮狀細岳曱狀腺、 chronic myelygenous leukemia、末期-g 巨 田胞白血病、 ,腎細胞癌、非何杰金氏淋巴瘤:二=胞^轉移 瘤、末期黑色素瘤、胃癌、非小細胞性肺癌腺細胞 癌、腎細胞癌、多種實體性腫瘤、多發性$ :、、、田胞性肺 攝護腺癌、惡性神經膠質瘤、腎多種實酽性θ⑺瘤、轉移性 難治癒的轉移性疾病、難治癒的多發二=髓痄瘤、淋巴癌、 卡波西氏肉瘤、反覆性退行性神經膠、 13 200804411 更特別的是,目前所揭露以化合物治療的癌症,包括 但不限於心臟:肉瘤(血管肉瘤、纖維肉瘤、横紋肌肉瘤、 脂肪肉瘤)、黏液瘤、橫紋肌瘤、纖維瘤、脂肪瘤、畸形 瘤;肺臟:支氣管肺癌(鱗狀細胞、未分化小細胞、未分 化大細胞、腺癌)、肺泡(細支氣管)癌、支氣管腺瘤、 肉瘤、淋巴瘤、軟骨錯構瘤、間皮瘤;腸胃道:食道(鱗 狀細胞瘤、腺癌、平滑肌肉瘤、淋巴瘤)、胃(癌、淋巴 瘤、平滑肌肉瘤)、胰臟(胰臟管腺癌、胰島素瘤、胰升 糖素瘤、胃泌素瘤、類癌、卡波西氏肉瘤、子宮肌瘤、血 管瘤、脂肪瘤、神經纖維瘤、纖維瘤)、大腸(腺癌、管 狀腺瘤、絨毛狀腺瘤、錯構瘤、子宮肌瘤)、生殖泌尿道: 腎臟(腺瘤、威姆氏腫瘤(腎芽細胞瘤)、淋巴瘤、血癌)、 膀胱及尿道(鱗狀細胞瘤、移行上皮細胞癌、腺癌)、前 列腺(腺癌、肉瘤)、睾丸(精母細胞癌、畸胎瘤、胚胎 細胞癌、畸胎癌、絨毛癌、肉瘤、間質細胞瘤、纖維瘤、 纖維腺瘤、腺癌樣瘤、脂肪瘤)、肝臟:肝癌(肝細胞癌)、 膽管癌、肝母細胞瘤、血管肉瘤、肝細胞腺瘤、血管瘤; 骨骼:骨源性肉瘤(骨肉瘤)、纖維肉瘤、惡性纖維組織 細胞瘤、軟骨肉瘤、尤文氏肉瘤、惡性淋巴瘤(網狀細胞 肉瘤)、多發性骨體瘤、惡性脊索鉅細胞瘤、osteochronfroma (軟骨外生性骨疲)、良性chrodroma、軟骨母細胞瘤、軟 骨黏液樣纖維瘤、骨樣骨瘤及鉅細胞瘤;神經系統:頭骨 (骨瘤、血管瘤、肉芽腫、黃瘤、畸形性骨炎);腦膜··(腦 膜瘤、腦膜肉瘤、神經膠質瘤)、腦(星狀細胞瘤、神經 管母細胞瘤、膠質瘤、室管膜瘤、生殖細胞瘤(松果腺瘤)、 多形性神經膠質母細胞瘤、寡樹突膠質瘤、神經鞘瘤、視 網膜母細胞瘤、遺傳性腫瘤)、脊髓腫瘤、神經線纖維瘤、 14 200804411 腦膜瘤、神經膠質瘤、肉瘤);婦科:子宮(内膜癌)、 子宮頸(子宮頸癌、前期腫瘤子宮頸細胞病變(Pre-tumor cervical dsplasia))、卵巢(卵巢瘤(漿液性囊腺癌、钻液 性囊腺癌、顆粒鞘膜細胞腫瘤(granulose-thecal cell tumors)、支持細胞睾丸間質細胞瘤、胚胎瘤、惡性畸胎瘤)、 外陰(鱗狀細胞癌、上皮内癌、腺癌、firosarcoma、黑色 素瘤)、陰道(亮細胞癌、鱗狀細胞癌葡萄狀肉瘤(胚胎 性橫紋肌肉瘤)、輸卵管瘤);血液:血液(骨髓性白血病 (急性及慢性)、急性淋巴細胞白血病、慢性淋巴細胞白 血病、骨髓增生性疾病、多發性骨髓瘤、骨髓發育不良徵 候群)、霍金森氏症、非霍金森氏症(惡性淋巴瘤);皮 膚:惡性黑色素瘤、基底細胞癌、鱗狀細胞癌、卡波西氏 肉瘤、增生痣、脂肪瘤、血管瘤、皮膚纖維瘤、瘢瘤、牛 皮癬;及腎上腺:神經母細胞瘤。 魏基(Carboxy)係指一化學基-C(0)0H。 「裂解」係指化學鍵的破壞,除非内容中明確指示,否 則並不限於化學性或酵素反應或機制。 名詞「螯合」係指藉由將一化合物之一或多的接觸點 與金屬配位後形成之化學基團。 ^ 環烷基(Cycloalkyl)指一個非芳香族碳環,通常具有 3-7個環碳原子。前述環碳可為飽和或具有—個或多個'碳-石反雙鍵。環烧基的例子包括環丙基、環丁基、環戊美、環 戊烯基、環己基、環己烯基及橋鍵及蘢蔽飽和之環基 and caged saturated ring groups),如:去甲苗(n〇rb〇r腿e)。 15 200804411 膦酸二烧基酯(Dialkylphosphonate)係指一自由 基一 P〇(〇Ra)(〇Rb) ’其中Ra與Rb為相同或不同的烧基。 「疾病」係指任何疾病、異常、症狀、徵候及徵兆。 「鹵素」或「鹵元素」係指氟、氣、溴、碘官能基。 「雜芳基」包括:五元至七元芳香、單環,包含一個 或多個(例如··--四,或在某些實施態樣為一〜三)選 自氮、氧、硫的雜原子,其餘的環狀原子則為碳;以及雙 雜烧環包含一個或多個(例如:一〜四,或在某些實施態 心為一〜二)選自氮、氧、硫的雜原子,i餘的璞狀爲+ 則為碳,其中至少一個雜原子存在於芳香^衷/舉例來說, ,芳基包括一個接合到五至七元烷環的五至七元雜烷芳香 環。為了達到這樣的接合,雙雜芳環系統中僅一個環含有 個或多個雜原子’而接合點可位在雜芳環或烧環上。當 錶芳基上的硫、氧原子總數為超過一,這些雜原子並不與 其他雜原子相接。在某些實施態樣中,雜絲上的硫、& 原子總數不超過二;在某些實施態樣中,芳香雜環上的硫、 氧原子總數不超過一。舉例來說,雜芳基包括一個接合到 :至=環的五至七元雜烧芳香環。為了達到這樣的接 方環系統中僅—個環含有—個或多個雜原子,而 ^,可位在雜芳環統環上。當雜芳基上的硫、氧原子 j為超過-,這祕原子並不與其他雜原子相接。 二,,中’雜芳基上的硫、氧原子總數不超過二;在 中’芳香雜環上的硫、氧原子總數不超過一。 ^方基的例子包括,但不限於(從接合 定基、3·㈣基、咖基、基、3=2 16 200804411 2,4-嘧啶基、3,5-嘧啶基、2,3-吡唑啉基、2,4-咪唑啉基、 異嗓°坐°林基、σ惡唾琳基、σ塞σ坐基、喧唾二基(thiadiazolinyl)、 四氮σ坐基、σ塞吩基、笨售吩基(benzothiophenyl)、吱喃基、 苯並吱°南基、苯咪唾基(benzoimidazolinyl)、二氫°?卜朵基、 pyridiziny卜三唑基、喹啉基、吡唑基及5,6,7,8-四氫異喹啉。 二價基係衍生自單價的雜芳基,其命名字尾「yl」係指自具 有自由彳貝的原子上移除一個氳原子;添加字尾r idene」則 疋為符合單價基。例如:具有兩個接合點的π比η定稱之為吼 咬基(pyridylidene)。然而,芳香基並不包含或與以上所指之 芳香基重覆。在某些實施態樣中,雜芳基可衍生自噻吩、 °比17各、苯並嗔峻、苯並吱喃、叫卜朵、ϋ比咬、喧琳、味唆、 嗓唑、吡嗓、苯並嗟唑、異噁唑及σ塞唑。 「雜烧芳基」及「雜芳烧基」係指一被雜芳基所取代 之環狀烷基,該環烷基上的氫原子結合至一個典型末端碳 原子或孕3碳原子。在某些實施態樣中,雜芳烷基可以是六 至三十元的雜芳烷基,例如:雜芳烷基及芳烷基係指一被 雜芳基所取代之環狀烧基,該環烧基上的氫原子結合至一 個典型末端碳原子或W3碳原子。在某些實施態樣中7雜芳 烧基可以疋六至二十元的雜芳燒基,例如··雜芳烧基的烷 屬fe基、烯基或炔基基團可以是一至十元的雜芳基及雜烷 基基團可以是五至二十元的雜芳基。 「雜環烷基」係指一脂肪單環,通常具有三至七個環 原子,該環原子包含至少兩個碳原子、一至三個個別選自 氧、硫、氮的雜原子及至少一個前述雜原子的组合。合適 的雜環烷基包括,例如(從接合點算起的數字作為1} : 1吡 咯基、2,4-imidaZ〇lidiny卜2,3“比唑烷基,2·呱啶基、3_ 17 200804411 呱咬基、4-呢咬基及2,5_piperzinyl。嗎淋基也是列入考量 的遙擇,包括2-嗎琳基及3_嗎琳基(從接合點算起的數字 作為1)。取代雜環炫基亦包括由—個或多個。如(=〇)或 oxide (-0)取^基取代所形成的環狀系統,例如:&氧化娘 σ疋基、N-氧化嗎啉基、^氧+硫基嗎啉基 (l-〇x〇-l-thi〇morph〇linyl)& U_二氧小硫基嗎琳基。 名祠「轉移」及語法上相同者係指癌症在體内由原發 部位擴散或轉移到其他部位的過程,並且在新的部位也產 生癌症病變的發展。 名闺「非共價化合物」係指由化合物與其他分子相互作 用所形成的化學基團,而在化合物與分子間並不會形成共 價鍵。舉例來說,錯合反應可因凡得瓦爾交互作用(VanDer Waals interactions)、氫鍵及靜電作用力(又稱離子鍵)而 產生。 「選擇性的」或「選擇性地」意指之後所描述事件或情 況可能但非一定會發生,因此敘述包括事件或情況可能發 生或不可能發生的例子。舉例來說,以下所指之「選擇性 地取代烷基」包含「烷基」及「取代烷基」。發明相同領域 中’熟悉此項技術之人都瞭解任何包含一個或多個取代基 的官能基在空間上有障礙、合成上不可行及先天不穩定的 情況下,是不可能產生任何取代反應或取代方式 「患者」或「個體」係指曾接受或將接受治療、觀察或 實驗的動物,例如哺乳動物。本發明之方法對於人類治療 及獸醫應用皆為有效。在某些實施態樣中,「病患」為哺 乳動物,在其他實施態樣中,「患者」則為人類。And pharmaceutically acceptable salts, solvates, chelating agents, non-covalent complexes, prodrugs, and mixtures thereof. Wherein Ri is an optionally substituted alkyl group; R2 is selected from a hydroxyl group and a decyloxy group; R3 is hydrogen, or R2 and R3 together with the oxime to which they are attached form an oxy group (0X0 group); when Ri is ( /?)-4-carboxybutan-2-yl, shell 1J R2 is not a meridian. Further provided is a pharmaceutical composition comprising at least one therapeutically effective amount of a chemical selected from the compounds of Formula I and a pharmaceutically acceptable carrier therefor. Provided is a method for inhibiting the activity of α-2,3 sialyltransferase, comprising an amount and a performance of α-2, wherein at least one chemical selected from the compound of Formula I is sufficient to detect a decrease in sialylation of a sugar conjugate. 3 The cells of the sialyltransferase are in contact. Further provided is a method for treating a patient responsive to a decrease in α-2,3 sialyltransferase overactivation activity. The method comprises administering to the patient at least one chemical selected from the group consisting of a chemical compound of formula I. the amount. Other objects and advantages of the present invention will be set forth in the description which follows. 200804411 is limited to the embodiments of the embodiments set forth in the specification. All of the features disclosed may be combined with other methods, and all of the features may be replaced by other ones that have the same and equal disclosure. The features of the specification and the scope of the appended claims are v,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,, The chemical substance includes, but is not limited to, a person of the formula j, such as shown in Table 1, and all pharmaceutically acceptable forms thereof: a compound referred to herein as "a pharmaceutically acceptable form of a compound" includes medical Acceptable salts, solvates, crystals (including polymorphs and clathrates), chelates, non-covalent compound precursor drug mixtures. The compounds described in certain embodiments are pharmaceutically acceptable salts. Therefore, the terms "chemical entity" and "chemicalentities" also include pharmaceutically acceptable salts. Solvents, crystals (including polymorphs and cage compounds), chelate compounds, non-covalent compound precursor drugs, and mixtures thereof. The fluorenyl group refers to the chemical group -C(〇)R, and R as used herein refers to an alkyl group, a substituted alkyl group, a substituted cycloalkyl group, a substituted heterocycloalkyl group, a substituted aryl group or a substituted heteroaryl group. Representative examples include, but are not limited to, ethyl ketone, cylcohexylcarbonyl, cyclohexylmethylcarbonyl, benzamidine, benzylcarbonyl, and the like. 200804411 Alkanyl refers to a saturated chain, a straight chain or a cycloalkyl group derived by the removal of a hydrogen atom from a single carbon atom on a parent Hyun hydrocarbon. Typical paraffinic group functional groups include, but are not limited to, methanyl, ethanyl, pr〇panyis (eg, propyl ketone + propyl, propyl-2-yl (ie, Isopropyl), cyclopropane small group; butanyls such as: butyl-1-yl, butan-1-yl (this c-butyl), 2-mercapto-propanyl group (ie different) Butyl), 2-mercapto-propan-2-ylindolebutyl), cyclobutane-1-yl and the like. Alkenyl refers to an unsaturated group, a straight chain or a cycloalkyl group having at least one carbon atom removed by a single carbon atom on a parent olefin to have a carbon-carbon double bond. The double bonds of these functional groups may be in cis or trans configuration. Typical olefin-based functional groups include, but are not limited to, ethenyl, propenyl, for example: prop-1-en-1-yl, propyl-2-enyl, prop-2-enyl (4) lyl), c-_2 Alken-2-yl, cycloprop-1-enyl-yl, cyclopropanyl-- yl-yl; butenyl group, for example: butyl succinyl-1-yl, butyl hydrazino-2-yl, 2-methyl- Propylene small group, butane-2·en-1-yl, but-2-en-2-yl, butyl-i,3-diene small group, butyl 4,3-dien-2-yl, ring But-1-en-1-yl, cyclobut-1-ene-3-yl, cyclobutadiene-b-yl, and the like. In certain embodiments, the monoolefin functional group has from 2 to 20 carbon atoms; in other embodiments, it has from 2 to 6 carbon atoms. Alkoxy refers to a chemical group - 〇R, and r as used herein refers to an alkyl group, a substituted alkyl group, a substituted cycloalkyl group, a substituted heterocycloalkyl group, a substituted aryl group or a heteroaryl group. Representative examples include, but are not limited to, methoxy, ethoxy, propyl, butoxy, cyclohexyloxy, and the like. AlkoxyCarb〇nyl refers to a chemical group of _c(0)~alkane, wherein the alkoxy group is the alkoxy group referred to above. 200804411 Alkyl refers to a saturated monovalent hydrocarbon group derived from a saturated or unsaturated branched chain, a straight chain or a single hydrogen atom removed from a parent hydrocarbon, a hydrocarbon or an alkyne. . Typical alkyl groups include, but are not limited to, methyl, ethane (eg, hypoethyl, vinyl, ethynyl), propane (eg, propan-1-yl, propan-2-yl, cyclopropane-1) —yl, prop-1-en-1-yl, propan-1-isoenyl, allyl, propylene small dilute, cyclopropen-1-yl, propyl-hydrazine-alkyne— :^, propynyl, propyl-- alkyne "-yl, butane alkyl (eg butane-1-yl, butan-2-yl), 2-methyl-propanyl, fluorenylmethyl-propan -2-yl, cyclobutyl small group, butadiene + group, butyl + dilute base, 2: base-prop-1-en-1yl, but-2-en-1-yl, butyl 2-en-2-yl, butyl-U bis=b, butyl-1,3-diene 1-yl, cyclobutane-1, cyclobut-1·ene-3-indolyl-1 3-dien-1-yl, butynyl, butynyl, butyl-3-alkyn-1-yl, and the like. The term "alkyl" particularly refers to any degree of saturation of a functional group, such as a functional group of a carbon-carbon single bond, a fluorene having one or more carbon-carbon double bonds having one or more carbon-carbon triple bonds and having a mixed carbon _: early bond, double bond, and triple bond Functional group. It is a hydrocarbon group, an alkene group, and a fast hydrocarbon group according to a certain degree of saturation. In some embodiments, a soil has 1-20 stone anti-atoms; in other embodiments, the alkyl group contains - carbon atoms and is referred to as a lower alkyl group. Alkynyl refers to an unsaturated branch, a straight chain or a parent alkyne bond: an anti-atomic removal of a hydrogen atom derived from at least one carbon-carbon: = honesty. (d) (4) Hydrocarbyl groups include, but are not limited to, Ethyl wire, = ^-exemplified as a small small group of propyl groups, such as: butyl small base, butyl, -3-yl, butynyl and the like. In certain embodiments, 11 200804411 alkynyl has 2 to 2 carbon atoms; other carbon atoms are depleted. Also, in the sample, the 3_6 amino group means a chemical group -NH2. Aminocarbonyl refers to R and R, which are each selected from hydrogen, 埝(O)NRR functional groups, in cycloalkyl, substituted heterocycloalkyl, substituted aryl, substituted The alkyl group is substituted with a nitrogen atom attached to R and R, a heteroaryl group, or an arbitrary hetero ring. One or more heterocyclic or aromatic groups (Aryl) comprising: a five- and six-membered fluorene bicyclic system wherein at least one ring is a carbocyclic ring and a non-fragrance ring, for example: benzene; tetralin; and tricyclic; A system, at least, for example, naphthalene, anthracene, for example: hydrazine. For example, the aromatic ring includes a five-membered and six-membered aromatic ring-breaking ring that is attached to the carbocyclic ring and the aromatic ring, and the one to more than one or more selected from the group consisting of nitrogen and oxygen. Sulfur heterogene: where the cycloalkyl ring system contains a bond, in the double ring system, only one carbocyclic ring I is attached to the carbocyclic aromatic ring or heterocycloalkyl ring. Bivalent beauty, the junction may form and the ring atom has a free valence, which is called; the derivative is derived from a monovalent polycyclic hydrocarbon, which is named after the living base. Divalent group: A hydrogen atom is removed from a subatomic atom; the added word has a value from j to a unit price. For example, the extraction |en:" with two junctions is, however, the aromatic group does not contain or is a naphthylene group. Cover, therefore, if one or more of the ... ^ & 2 jin 疋 疋 疋 杂 杂 芳 在 μ μ μ μ μ μ μ μ μ μ μ μ μ μ μ μ μ μ μ μ μ μ μ μ μ μ μ μ μ μ μ μ μ μ μ 芳The cyclo-ring cyclist is substituted by a aryl group to a hydrogen atom on the lanyard to a typical terminal carbonogen 12 200804411 or w3 carbon atom. Typical aralkyl groups include, but are not limited to, benzyl, 2 # alkanedi-, Styrene + yl, naphthyl fluorenyl, 2 - naphthylethyl bromide = ethyl 2-naphthylethylene m, benzylidene, 2 (10) phenyl quinone, an analog thereof, wherein the specific (four) group It is named after an aromatic hydrocarbon group t and a s- or an aryl-alkyne group. In certain embodiments, an alkane: an aralkyl group of a hydrocarbon of thirty carbons, for example, an alkane of an aralkyl group Two: The alkyne group y is - to ten carbons, and the aromatic group of the aryl group can be = or twenty carbons. 疋 /, yl 、,,,,,,,,,,,,,,,,,,,,,, 〇-R, wherein R is selected from the group consisting of aryl groups. 曰 (三) or substituted carbonyl means a chemical group - c (〇). The term "cancer" as used herein refers to Or physiological state, its cells A cancer with compound treatment disclosed in an abnormally finely planted animal, including it sputum. Current tumor, Indian cancer, breast cancer, endometrial cancer, cell colorectal cancer, colon cancer, digestive tract, lung cancer, Renal kidney, sputum tumor, lymph, prostate cancer, pancreatic cancer, terminal cancer, hairy sputum, chronic myelygenous leukemia, terminal-g mega-cell leukemia, renal cell carcinoma, non-ho Jigkin's lymphoma: two = cell metastasis, terminal melanoma, gastric cancer, non-small cell lung cancer, adenocarcinoma, renal cell carcinoma, multiple solid tumors, multiple m:,,, cytoplasmic lung prostate cancer , malignant glioma, multiple renal sputum θ (7) tumors, metastatic refractory metastatic disease, refractory multiple secondary = medullary tumor, lymphoma, Kaposi's sarcoma, recurrent degenerative nerve glue, 13 200804411 More specifically, cancers currently treated with compounds are disclosed, including but not limited to the heart: sarcoma (vascular aneurysm, fibrosarcoma, rhabdomyosarcoma, liposarcoma), myxoma, rhabdomyoma, fiber , lipoma, malformation; lung: bronchial lung cancer (squamous cells, undifferentiated small cells, undifferentiated large cells, adenocarcinoma), alveolar (bronchiolar) cancer, bronchial adenoma, sarcoma, lymphoma, cartilage hamartoma , mesothelioma; gastrointestinal tract: esophagus (squamous cell tumor, adenocarcinoma, leiomyosarcoma, lymphoma), stomach (cancer, lymphoma, leiomyosarcoma), pancreas (pancreatic ductal adenocarcinoma, insulinoma, pancreatic Glycanoma, gastrinoma, carcinoid, Kaposi's sarcoma, uterine fibroids, hemangioma, lipoma, neurofibromatosis, fibroids, large intestine (adenocarcinoma, tubular adenoma, villous adenoma, Hamartoma, uterine fibroids, genitourinary tract: kidney (Adenoma, Wilms' tumor (renal blastoma), lymphoma, blood cancer), bladder and urethra (squamous cell tumor, transitional epithelial cell carcinoma, gland Cancer), prostate (adenocarcinoma, sarcoma), testis (sperm cell carcinoma, teratoma, embryonic cell carcinoma, teratoma, villus, sarcoma, stromal cell tumor, fibroid, fibroadenomas, adenocarcinoma Tumor, lipoma), liver: Cancer (hepatocellular carcinoma), cholangiocarcinoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, hemangioma; bone: osteogenic sarcoma (osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, Juventus Sarcoma, malignant lymphoma (reticulocyte sarcoma), multiple bone tumor, malignant notochord giant cell tumor, osteochronfroma (chondral exogenous bone fatigue), benign chrodroma, chondroblastoma, chondromyxoid fibroid, bone Osteomas and giant cell tumors; nervous system: skull (osteoma, hemangioma, granuloma, xanthoma, deformity osteitis); meninges · (meningioma, meningioma, glioma), brain (stellate cells) Tumor, neuroblastoma, glioma, ependymoma, germ cell tumor (pine pine adenoma), glioblastoma multiforme, oligodendroglioma, schwannomas, retinoblastoma, Hereditary tumor), spinal cord tumor, neurofibromatosis, 14 200804411 meningioma, glioma, sarcoma); gynecology: uterus (endometrial cancer), cervix (cervical cancer, pre-neoplastic Cervical cytopathic (Pre-tumor cervical dsplasia), ovary (ovarian tumor (serous cystic adenocarcinoma, drilling fluid cystic adenocarcinoma, granulose-thecal cell tumors), supporting cell Leydig cells Tumor, embryonal tumor, malignant teratoma), vulva (squamous cell carcinoma, intraepithelial neoplasia, adenocarcinoma, firosarcoma, melanoma), vaginal (leukocytic carcinoma, squamous cell carcinoma, sarcoma of squamous cell (embryonic rhabdomyosarcoma) , fallopian tube tumors; blood: blood (myeloid leukemia (acute and chronic), acute lymphocytic leukemia, chronic lymphocytic leukemia, myeloproliferative diseases, multiple myeloma, myelodysplastic syndrome), Hawking's disease, Non-Hodgkin's disease (malignant lymphoma); skin: malignant melanoma, basal cell carcinoma, squamous cell carcinoma, Kaposi's sarcoma, hyperplasia, lipoma, hemangioma, cutaneous fibroids, neoplasms, psoriasis; Adrenal gland: neuroblastoma. Carboxy refers to a chemical group -C(0)0H. "Cleavage" refers to the destruction of a chemical bond and is not limited to chemical or enzymatic reactions or mechanisms unless explicitly indicated in the context. The term "chelating" refers to a chemical group formed by the coordination of one or more points of a compound with a metal. ^ Cycloalkyl refers to a non-aromatic carbocyclic ring, usually having from 3 to 7 ring carbon atoms. The aforementioned ring carbon may be saturated or have one or more 'carbon-stone reverse double bonds. Examples of the cycloalkyl group include a cyclopropyl group, a cyclobutyl group, a cyclopentamethylene group, a cyclopentenyl group, a cyclohexyl group, a cyclohexenyl group, and a bridged and caged saturated ring groups, such as: A seedling (n〇rb〇r leg e). 15 200804411 Dialkylphosphonate refers to a free radical -P〇(〇Ra)(〇Rb)' where Ra and Rb are the same or different. "Disease" means any disease, abnormality, symptom, sign and sign. "Halogen" or "halogen" means a fluorine, gas, bromine or iodine functional group. "Heteroaryl" includes: a five- to seven-membered aromatic, monocyclic ring containing one or more (eg, four, or in some embodiments, one to three) selected from the group consisting of nitrogen, oxygen, and sulfur. a hetero atom, the remaining ring atoms are carbon; and the double miscellaneous ring contains one or more (eg, one to four, or in some embodiments, the core is one to two) selected from nitrogen, oxygen, sulfur The atom, i is in the form of a carbon, wherein at least one hetero atom is present in the aromatic group. For example, the aryl group includes a five- to seven-membered heteroalkyl aromatic ring bonded to a five- to seven-membered alkane ring. . To achieve such a bond, only one ring in the double heteroaromatic ring system contains one or more heteroatoms and the junction can be on the heteroaryl ring or the burn ring. When the total number of sulfur and oxygen atoms on the aryl group exceeds one, these heteroatoms are not bonded to other heteroatoms. In certain embodiments, the total number of sulfur, & atoms on the filament is no more than two; in certain embodiments, the total number of sulfur and oxygen atoms on the aromatic heterocycle is no more than one. For example, a heteroaryl group includes a five to seven member miscellaneous aromatic ring bonded to: to a ring. In order to achieve such a ring system, only one ring contains one or more heteroatoms, and ^ can be located on the heteroaryl ring. When the sulfur and oxygen atom j on the heteroaryl group exceeds -, the secret atom does not meet other heteroatoms. Second, the total number of sulfur and oxygen atoms in the medium heteroaryl group is not more than two; the total number of sulfur and oxygen atoms in the medium aromatic heterocycle is not more than one. Examples of square radicals include, but are not limited to, (from a bonding group, a 3 (tetra) group, a ke group, a group, 3 = 2 16 200804411 2, 4-pyrimidinyl, 3,5-pyrimidinyl, 2,3-pyrazole Ortholinyl, 2,4-imidazolinyl, isoindole, sigma, sigma, sigma, thiadiazolinyl, tetrazolium, sigma, Substituting benzothiophenyl, fluorenyl, benzoxanthyl, benzoimidazolinyl, dihydro-bromo, pyridiziny, triazolyl, quinolyl, pyrazolyl and 5 , 6,7,8-tetrahydroisoquinoline. The divalent group is derived from a monovalent heteroaryl group whose suffix "yl" refers to the removal of a deuterium atom from an atom having free mussels; The tail r idene 疋 is in accordance with the monovalent group. For example, a π ratio η having two joints is called a pyridylidene. However, the aromatic group does not contain or overlap with the above-mentioned aromatic group. In some embodiments, the heteroaryl group can be derived from thiophene, a ratio of 17 to each, a benzophenanthrene, a benzopyrene, a bud, a bismuth, a quinone, a miso, a carbazole, a pyridyl Bismuth, benzoxazole, isoxazole And σ- oxazole. "Hybrid aryl" and "heteroalkyl" refer to a cyclic alkyl group substituted by a heteroaryl group, the hydrogen atom of which is bonded to a typical terminal carbon atom or pregnant. 3 carbon atoms. In certain embodiments, the heteroarylalkyl group may be a six to thirty membered heteroarylalkyl group, for example, a heteroarylalkyl group and an aralkyl group refer to a ring substituted with a heteroaryl group. a hydrogen group on the cycloalkyl group bonded to a typical terminal carbon atom or a W3 carbon atom. In some embodiments, the 7 heteroaryl group may be a six to twenty membered heteroaryl group, for example The heteroaryl, alkenyl or alkynyl group of the heteroarylalkyl group may be a one to ten membered heteroaryl group and the heteroalkyl group may be a five to twenty membered heteroaryl group. """ refers to a fatty monocyclic ring, usually having three to seven ring atoms, the ring atom comprising at least two carbon atoms, one to three heteroatoms individually selected from oxygen, sulfur, nitrogen, and combinations of at least one of the foregoing heteroatoms Suitable heterocycloalkyl groups include, for example, (the number from the junction as 1}: 1 pyrrolyl, 2,4-imidaZ〇lidiny, 2,3" ratio Azolidinyl, 2·acridinyl, 3_ 17 200804411 呱 base, 4-bite base and 2,5_piperzinyl. 淋 基 也是 is also included in the consideration of choice, including 2-lineline and 3_linen (The number from the junction is taken as 1.) The substituted heterocyclic thiol also includes a ring system formed by one or more substitutions such as (=〇) or oxide (-0), for example: & Oxidation 疋 疋 、, N-oxidized morpholino, ^ oxy + thiomorpholinyl (l-〇x〇-l-thi〇morph〇linyl) & U_dioxosylthiopyrine . The term "metastasis" and grammatically identical means the process by which cancer spreads or metastasizes from the primary site to other sites, and the development of cancerous lesions also occurs in new sites. The term "non-covalent compound" refers to a chemical group formed by the interaction of a compound with other molecules, and does not form a covalent bond between the compound and the molecule. For example, mismatch reactions can result from VanDer Waals interactions, hydrogen bonding, and electrostatic forces (also known as ionic bonds). "Optional" or "optionally" means that the event or circumstance described later may, but does not necessarily occur, such that the narration includes instances in which an event or circumstance may or may not occur. For example, "selectively substituted alkyl" as used hereinafter includes "alkyl" and "substituted alkyl". In the same field of the invention, 'a person familiar with the art knows that any functional group containing one or more substituents is sterically hindered, synthetically unfeasible, and inherently unstable, and it is impossible to produce any substitution reaction or The alternative means "patient" or "individual" means an animal, such as a mammal, that has received or will be treated, observed or tested. The method of the invention is effective for both human and veterinary applications. In some embodiments, the "patient" is a mammal, and in other embodiments, the "patient" is a human.

1R 200804411 「醫藥上可接受」係指已被或可被聯邦管理機構、州政 府所認可或列入美國藥典或其他一般被認可、用於動物(包 括人類)之藥典。 「醫藥上可接受之鹽類」係指一種醫藥上可接受之鹽 類化合物,而該化合物具有特定的醫藥活性。此種鹽類包 括與:(1)由無機酸(如:鹽酸、氫溴酸、硫酸、琐酸、 磷酸及類似物)或有機酸(如:醋酸、丙酸、己酸、環戊 基丙酸、甘醇酸、丙酮酸、乳酸、丙二酸、破站酸、蘋果 酸、馬來酸、富馬酸、酒石酸、檸檬酸、苯甲酸、3-(4-羥 基苯曱酰基)苯甲酸、桂皮酸、杏仁酸、曱基磺酸、乙基 石黃酸、1,2-乙烧-二石黃酸、2-經乙基石黃酸、苯石黃酸、4-鼠本石黃 酸、2-萘磺酸、4-甲笨磺酸、樟腦磺酸、4-甲基雙環[2.2.2]-辛-2-烯-1-羧酸、葡萄庚糖酸、3-苯基丙酸、三曱基醋酸、 三級丁基醋酸、月桂醯醚硫酸、葡萄糖酸、麩胺酸、羥萘 甲酸、水揚酸、硬脂酸、已二烯二酸及類似物)所形成的 酸性附加鹽;或(2)因化合物中存在或酸性質子被金屬離子 (如:驗金屬離子、驗土金屬離子或紹離子)所取代或與 有機鹼(如:乙醇胺、二乙醇胺、三乙醇胺、N-葡甲胺、 二環己胺及類似物)相作用而形成的鹽類。 「醫藥上可接受之賦形劑、載體或佐劑」係指一種可 與至少一種目前已揭露之化學物質一同施予給一個體而不 破壞其醫藥活性的賦形劑、載體或佐劑,並在施與一足已 產生有效劑量量的化合物時,並不會對該個體造成毒性。 「醫藥上可接受之載體」係指與至少一種目前已揭露之 化學物質一同施打之稀釋劑、佐劑、賦形劑或載體。 19 200804411 「前驅藥物」係指有療效化合物之衍生物,該衍生物在 體内必須進行轉形作用,以便產生有療效的化合物。前驅 藥物在轉化為母體化合物前,可以是藥學上的不活化狀 態。 「保護基」係指當一群原子附著於一分子的反應基時 ,反應力會被遮蔽、減低或抑制。保護基的例子可在Green 專人所著之“Protective Groups in Organic Chemistry,,(Wiley, 2 ed· i991)及 Harrison 等人所著之 “Compendium of Synthetic Organic Methods,,Vols· 1-8 (John Wiley and Sons, 1971-1996)找到。代表性的氨基保護基包括但不限於甲醯 基、乙醯基、三氟乙醯、苄基、苄氧羰基(“CBZ”)、叔丁氧 羰基(“Boc”)、三甲基硅烷基(“TMS,,)、 2-trimethylsilyl-ethanesulfonyl (“SES,,)、三苯甲基及取代三 苯曱基、丙稀氧獄基(allyloxycarbonyl)、9-芴曱氧幾基 (“FMOC”)、nitro-veratryloxycarbonyl (“NVOC”)及其類似 物。代表性的氫氧基包括但不限於(這些氫氧基係已醯化 或院化),例如:T基、三苯曱基醚、烷基醚、四氳吡喃 基醚、三烧石夕醚(trialkylsilylethers)及烯丙峻。 本發明之化學物質之「治療上有效量」意指當施予一 有效量於人類或非人類患者時,可提供療效,例如:改善 症狀、減緩病程及預防疾病。「治療上有效量」可能是足 以減少對α-2,3唾液酸轉移酶過度活化所導致的疾病症狀的 量。在某些實施態樣中 治療上有效量」,係足以減少 癌症症狀的置,在某些貫施樣中’ 「治療上有效量」, 係足以減少一有機體中’可測得之癌細胞數;在苹此實施 悲樣中’冶療上有效里」’係足以測出擴散的腫瘤減緩 20 200804411 或停止生長;在某些實施態樣中,「治療上有效量」,係 足以使腫瘤縮小的量;在某些情況下,罹患癌症的患者可 能未出現已患病的症狀;在某些實施態樣中,一化學物質 的有效量係足以預防腫瘤明顯增加或減少可測得的癌細胞 數或腫瘤標記在患者的血液、血清或組織中出現。 /口療(Treatment)」或「治療(Treating)」意指任何對 於疾病患者的任何處置,包括疾病預防,也就是使至少一 種疾病的臨床症狀不會繼續發展;抑制疾病;減緩或阻礙 至y 種疾病的〖品床症狀的發展;以及解除疾病,也就是 治癒至少一種疾病的臨床症狀。「治療(Treating)」或「治 療(Treatment)」亦指抑制一種身體上(例如:穩定可識別的 症狀)或生理上(穩定生理指標)或兩者兼具的疾病或抑 制至少一種個體未能識別出的生理指標。進一步來說,治 療(Treatment)」或「治療(Treating)係指延缓疾病或一可能 接觸或易於患病的個體’其至少一種臨床症狀的開展,即 使該個體尚未體驗或顯露出疾病的症狀。 「取代」係指一官能基中,一個或多個氫原子分別被相 同或相異的取代基所取代。典型的取代基包括但不限 於,-X,-R33, -0·,=0, -OR33, -SR33, —S,=S,-NR33R34, =NR33,-CX3,-CF3,- CN, -0CN,- SCN,-NO,-N02, =N29 -N35 -S(0)20\ -S(0)20H, -S(0)2R33? ~0S(02)0\ -0S(0)2R33? 一P(〇X〇-)2, -p(o)(or33)(o ),-op(o)(or33)(or34),-c(o)r33, -c(s)1R 200804411 “Pharmaceutically acceptable” means a Pharmacopoeia that has been or is approved by the federal regulatory agency, the state government, or listed in the US Pharmacopoeia or other generally recognized animal (including human). "Pharmaceutically acceptable salt" means a pharmaceutically acceptable salt compound which has specific pharmaceutical activity. Such salts include: (1) from inorganic acids (such as: hydrochloric acid, hydrobromic acid, sulfuric acid, tribasic acid, phosphoric acid and the like) or organic acids (such as: acetic acid, propionic acid, caproic acid, cyclopentyl Acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, catalyzed acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, 3-(4-hydroxybenzoyl)benzoic acid , cinnamic acid, mandelic acid, mercapto sulfonic acid, ethyl tarnishic acid, 1,2-ethane-di-dilybetic acid, 2-ethyl-ethyl-retinic acid, benzoic acid, 4-rhamninoic acid, 2-naphthalenesulfonic acid, 4-methylsulfonic acid, camphorsulfonic acid, 4-methylbicyclo[2.2.2]-oct-2-ene-1-carboxylic acid, glucoheptonic acid, 3-phenylpropionic acid Acid addition of trimethyl hydroxyacetate, tertiary butyl acetate, lauryl ether sulphuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, hexadienedioic acid and the like a salt; or (2) due to the presence of a compound or an acidic proton that is replaced by a metal ion (eg, a metal ion, a soil metal ion, or a sulphur ion) or with an organic base (eg, ethanolamine, diethanolamine, triethanolamine, a salt formed by the action of N-glucamine, dicyclohexylamine, and the like. "Pharmaceutically acceptable excipient, carrier or adjuvant" means an excipient, carrier or adjuvant that can be administered to a body with at least one of the currently disclosed chemical substances without destroying its pharmaceutically active activity. It does not cause toxicity to the individual when a sufficient amount of the compound has been administered. "Pharmaceutically acceptable carrier" means a diluent, adjuvant, excipient or carrier with which at least one of the currently disclosed chemical substances is applied. 19 200804411 "Precursor drug" means a derivative of a therapeutic compound which must be transformed in vivo to produce a therapeutic compound. The prodrug may be pharmaceutically inactive prior to conversion to the parent compound. "Protecting group" means that when a group of atoms are attached to a molecule of a reactive group, the reaction force is blocked, reduced or suppressed. Examples of protecting groups can be found in "Protective Groups in Organic Chemistry," by Wiley, 2, ed. i991, and Harrison et al., "Compendium of Synthetic Organic Methods," Vols 1-8 (John Wiley). And Sons, 1971-1996) found. Representative amino protecting groups include, but are not limited to, indolyl, ethyl fluorenyl, trifluoroacetamidine, benzyl, benzyloxycarbonyl ("CBZ"), tert-butoxycarbonyl ("Boc"), trimethylsilyl ("TMS,,), 2-trimethylsilyl-ethanesulfonyl ("SES,,), trityl and substituted triphenyl fluorenyl, allyloxycarbonyl, 9-anthoxycarbonyl ("FMOC") ), nitro-veratryloxycarbonyl ("NVOC") and its analogs. Representative hydroxyl groups include, but are not limited to, (these hydroxyl groups have been deuterated or denatured), for example: T-based, triphenyl decyl ether, alkyl ether, tetrapyridyl pyridyl ether, three burnt stone eve Trialkylsilylethers and olefins. The "therapeutically effective amount" of the chemical substance of the present invention means that when an effective amount is administered to a human or non-human patient, the therapeutic effect can be provided, for example, improving symptoms, slowing the course of the disease, and preventing the disease. A "therapeutically effective amount" may be sufficient to reduce the symptoms of the disease caused by excessive activation of alpha-2,3 sialyltransferase. In some embodiments, a therapeutically effective amount, which is sufficient to reduce the symptoms of cancer, is 'a therapeutically effective amount' in some embodiments, which is sufficient to reduce the number of measurable cancer cells in an organism. In the implementation of the sadness in this practice, the 'effectiveness in treatment' is sufficient to detect the spread of tumors. 20200804411 or stop growing; in some embodiments, the "therapeutically effective amount" is sufficient to shrink the tumor. In some cases, patients with cancer may not have symptoms of the disease; in some embodiments, an effective amount of a chemical is sufficient to prevent tumors from significantly increasing or decreasing measurable cancer cells. The number or tumor marker appears in the blood, serum or tissue of the patient. /"Treatment" or "Treating" means any treatment of a patient with a disease, including disease prevention, that is, the clinical symptoms of at least one disease will not continue to develop; inhibit the disease; slow or hinder to y The development of the disease symptoms of the disease; and the relief of the disease, that is, the clinical symptoms of at least one disease. "Treating" or "Treatment" also means inhibiting a physical (eg, stable and identifiable symptom) or physiological (stable physiological indicator) or both, or inhibiting at least one individual. Identifyed physiological indicators. Further, "treating" or "treating" refers to delaying the progression of a disease or an individual who is likely to be exposed to or susceptible to disease, at least one clinical symptom, even if the individual has not experienced or revealed symptoms of the disease. "Substitution" means that in a monofunctional group, one or more hydrogen atoms are each replaced by the same or a different substituent. Typical substituents include, but are not limited to, -X, -R33, -0., = 0, -OR33, -SR33, -S, =S, -NR33R34, =NR33, -CX3, -CF3, - CN, - 0CN,- SCN,-NO,-N02, =N29 -N35 -S(0)20\ -S(0)20H, -S(0)2R33? ~0S(02)0\ -0S(0)2R33? A P(〇X〇-)2, -p(o)(or33)(o), -op(o)(or33)(or34), -c(o)r33, -c(s)

R33, -C(0)0R33, -C(0)NR33R34, -C(0)0\ -C(S)OR33, -NR35C(0)NR 33r34, 一nr35c(s)nr33r34, 一nr35c(nr33)nr33r34, —c(nr33)nr33r34, -s(o)2nr33r34, -nr35s(o)2r33, —nr35c(o)r33 及—s(o)r33,其中 每一個X分別是鹵素;每一個R33、R34分別是氩、烷基、 21 200804411 A、ΐ二方基、取代芳基、芳燒基、取代芳烧基、環垸 其㈣Ϊ、雜環絲、取代雜環院基、雜芳基、取 土、35雜芳烷基、取代雜芳烷基、nr35r36,—C(〇)R35 或-S(〇)2r或r33與r34選擇性地與附著直上的原子形 環,严乂代,烷環、雜芳環、取代㈣環1 ^A —舁R分別疋氫、烷基、取代烷基、芳基、取代 =2燒基,代芳烧基、環燒基、取代環&基、_ 代雜魏基』料基、取代雜絲、雜芳烧基或 =^方烧基;或R及R36選擇性地與附著其上的氮原 =形士-個或多個雜環院環、取代雜環院環、雜芳環、取 =雜芳環。在某些實施例’三級胺或含氮芳香基化合物可 被一個或多個氧原子所取代而形成對應的氧化氮。 基、-s(o)2nr33r34 、-nr35s(o)2r33、—nr35c(o)r 方基、c5_1()雜芳基 ,33 -nr33r34 35〜a、d33 在某些實施例,取代芳基與取代雜芳基包括一個或多 個下列的取代基:氟、氯、漠、C13燒基、取代炫基、Ci 3 燒 氧 -CF3、一0CF3、-CN, C5-H)烷基、C5_1G取代 基、c5_1()雜芳基、c5,取代雜芳 基一C(0)0R,-N〇2, -C(0)R,-C(〇)nr33r34, —〇cHF2, Cu 基、-SR33,、-S(0)20H、-S(〇)2R33、_s(〇)r33、_c(S) R33、-c(0)0-、-c(s)〇R33、-NR35C(〇)NR33R34、_nr3 =(s)nr33r34及 _c(nr35)nr33r34、c3 8 環烷基及 C3 8 取代 %燒基、Cs-8雜環烧基及C3·8取代雜環烧基。 在某些實施例’取代芳烧基與取代雜芳烧基包括一個 或多個下列的取代基:氟、氣、溴、Cl_3烷基、取代烷基、 22 200804411R33, -C(0)0R33, -C(0)NR33R34, -C(0)0\ -C(S)OR33, -NR35C(0)NR 33r34, one nr35c(s)nr33r34, one nr35c(nr33) Nr33r34, —c(nr33)nr33r34, -s(o)2nr33r34, -nr35s(o)2r33, —nr35c(o)r33 and —s(o)r33, each of which is a halogen; each R33, R34 Are argon, alkyl, 21 200804411 A, ruthenium diaryl, substituted aryl, aryl, substituted aryl, cyclopentadienyl (tetra), heterocyclic, substituted heterocyclic, heteroaryl, desorbed , 35 heteroaralkyl, substituted heteroaralkyl, nr35r36, -C(〇)R35 or -S(〇)2r or r33 and r34 are selectively attached to the atomic ring, straightening, alkane, Heteroaromatic ring, substituted (tetra) ring 1 ^A - 舁R, respectively, hydrogen, alkyl, substituted alkyl, aryl, substituted = 2 alkyl, arylalkyl, cycloalkyl, substituted ring & a hetero Wei group, a substituted heterofilament, a heteroaryl or a aryl group; or R and R36 are optionally substituted with a nitrogen atom attached thereto or a plurality of heterocyclic rings Heterocyclic ring, heteroaryl ring, and = heteroaryl ring. In certain embodiments, the tertiary amine or nitrogen-containing aromatic compound can be substituted with one or more oxygen atoms to form the corresponding nitrogen oxide. , -s(o)2nr33r34, -nr35s(o)2r33, -nr35c(o)r, cyclyl, c5_1()heteroaryl, 33-nr33r34 35~a, d33 In certain embodiments, substituted aryl Substituted heteroaryl groups include one or more of the following substituents: fluoro, chloro, dimethyl, C13 alkyl, substituted leuco, Ci3 oxy-CF3, =0CF3, -CN, C5-H) alkyl, C5_1G substituted Base, c5_1()heteroaryl, c5, substituted heteroaryl-C(0)0R, -N〇2, -C(0)R, -C(〇)nr33r34, -〇cHF2, Cu group, -SR33 ,, -S(0)20H, -S(〇)2R33, _s(〇)r33, _c(S) R33, -c(0)0-, -c(s)〇R33, -NR35C(〇)NR33R34 _nr3 = (s) nr33r34 and _c(nr35)nr33r34, c3 8 cycloalkyl and C3 8 substituted % alkyl, Cs-8 heterocycloalkyl and C3. 8 substituted heterocycloalkyl. In certain embodiments, the substituted aryl and substituted heteroaryl groups include one or more of the following substituents: fluorine, gas, bromine, Cl_3 alkyl, substituted alkyl, 22 200804411

Ci.3 烷 氧 基、-S(0)2NR33R34, -nr33r34, -CF3, 一OCF3, —CN,一nr35s( 0)2R33,-NR35C(O)R33,C5-10 芳基、取代烷基、c5_10 取代 芳基 、C5-H)雜芳基、C5-10 取代雜芳 基、-C(0)0R33,-N〇2, -C(0)R33,-C(0)NR33R34,一〇CHF2,Ci.3 alkoxy, -S(0)2NR33R34, -nr33r34, -CF3, -OCF3, -CN,-nr35s(0)2R33,-NR35C(O)R33,C5-10 aryl, substituted alkyl, C5_10 substituted aryl, C5-H)heteroaryl, C5-10 substituted heteroaryl, -C(0)0R33, -N〇2, -C(0)R33, -C(0)NR33R34, mono-CHF2 ,

Cl-3 酿 基、一SR33, -S(0)2〇H,-S(0)2R33, -S(0)R33, —C(S)R33, —C(0 )0\ -C(S)OR33, 一nr35c(o)nr33r34,-nr35c(s)nr33r34 及-c(nr35)nr33r34、C3_8環烷基及C3-8取代環烷基。 在某些實施例,取代芳烷基與取代雜芳烷基包括一個 或多個下列的取代基:Cm烷氧基、-NR33R34、c5_1()取代雜 芳基、-SR33、Cm烷氧基、-S(0)2NR33R34、氰基、氟、氯、 -CF3, -OCF3, -NR35S(0)2R33, —nr35c(o)r33、c5-10 芳基、c5_10 取 代芳基、c5_1()雜芳基、c5_1()取代雜芳基、 C(0)0R33,-N02,- C(0)R33,—C(0)NR33R34,—0CHF2,Cm acyl,一S(0)20H,一S(0)2R33, 一S(0)R33, —C(S)R,-C(0)0 , 一C(S)OR33, 一NR35C(0)NR33R34, —NR35C(S)NR33R34, and —C(NR35)NR33R34、 C3-8環烷基及C3-8取代環烷基。 在某些實施例,取代烯基包括一個或多個下列的取代 基· C1-8烧基、取代院基、C5-10芳基、C5-10取代芳基、 C5-H)雜芳基、C5_1()取代雜芳基、c3_8環烷基、c3_8取代環烷 基、雜環就基烧基(cycloheteroalkylalkyl)及取代之雜環烷基烧 基(substituted cycloheteroalkylalkyl)。 名詞「取代氨基」係指官能基_NHRd或一NRdRd,其中每一 R係選自烷基、取代烷基、環烷基、取代環烷基、醯基、取代醯 23 200804411 基芳基、取代芳基、雜芳基、取代雜芳基、雜環炫基、取代雜 環烧基、烧氧基羰基及硫基。代表例包括,但不限於二甲胺基、 甲基乙基胺基、二-(1-甲基乙炔基)胺基、(環己基)(甲基)胺基、(環 己基)(乙基)胺基、(環己基)(丙基)胺基及其類似物。 提供至少一種選自化學式I所示化合物之化學物質:Cl-3, a group of SR33, -S(0)2〇H,-S(0)2R33, -S(0)R33, —C(S)R33, —C(0 )0\ -C(S OR33, an nr35c(o)nr33r34, -nr35c(s)nr33r34 and -c(nr35)nr33r34, a C3_8 cycloalkyl group and a C3-8 substituted cycloalkyl group. In certain embodiments, substituted aralkyl and substituted heteroarylalkyl include one or more of the following substituents: Cm alkoxy, -NR33R34, c5_1() substituted heteroaryl, -SR33, Cm alkoxy, -S(0)2NR33R34, cyano, fluoro, chloro, -CF3, -OCF3, -NR35S(0)2R33, —nr35c(o)r33, c5-10 aryl, c5_10 substituted aryl, c5_1() heteroaryl Substituent, c5_1() substituted heteroaryl, C(0)0R33, -N02, -C(0)R33, -C(0)NR33R34, -0CHF2, Cm acyl, one S(0)20H, one S(0 ) 2R33, a S(0)R33, —C(S)R, -C(0)0, a C(S)OR33, an NR35C(0)NR33R34, —NR35C(S)NR33R34, and —C(NR35 NR33R34, C3-8 cycloalkyl and C3-8 substituted cycloalkyl. In certain embodiments, a substituted alkenyl group includes one or more of the following substituents: C1-8 alkyl, substituted institutional, C5-10 aryl, C5-10 substituted aryl, C5-H) heteroaryl, C5_1() substituted heteroaryl, c3-8 cycloalkyl, c3-8 substituted cycloalkyl, cycloheteroalkylalkyl and substituted cycloheteroalkylalkyl. The term "substituted amino" refers to a functional group -NHRd or an NRdRd wherein each R is selected from the group consisting of alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, fluorenyl, substituted oxime 23 200804411 aryl, substituted Aryl, heteroaryl, substituted heteroaryl, heterocyclo, substituted heterocycloalkyl, alkoxycarbonyl and thio. Representative examples include, but are not limited to, dimethylamino, methylethylamino, bis-(1-methylethynyl)amino, (cyclohexyl)(methyl)amine, (cyclohexyl) (ethyl) Amino, (cyclohexyl) (propyl) amine groups and analogs thereof. Providing at least one chemical selected from the group consisting of the compounds of Formula I:

及其醫藥上可接受之鹽類、溶劑化物、螯合劑、非共價複 合物、前驅藥物及其混合物。其令心為任意烧基;R2可選 自經基及酸氧基;R3係為氫,或r2與r3連同其所附著的 碳’形成一個氧基(OXO group);當Ri為羧基-2-基, 則R2非為經基。 在某些實施例,艮選自-CHR4(CH2)nR5,η為2跟3 ; R4選自氫或任意取代的低碳數烷基;及R5選自羧基、曱醯 胺基及選擇性取代的雜芳基。在某些實施例,n為2 ;在某 些實施例,η為3;在某些實施例,R4選自氩或低碳數烷基; 在某些實施例,r4為低碳數烷基;在某些實施例,R4為甲 基;在某些實施例,R5選自羧基、-(C0)-NHR6及選擇性取 代的三唑,其中r6選自選擇性取代的低碳數烷基。 24 200804411 在某些實施例,R^-CHR4(CH2)nR5,其中,η為2及 3,&選自氫或選擇性取代的低碳數烧基;及&選自叛基、 -(CO)-NHR6及取代三唑,其中三唑環係為一官能基r?所取 代,而R7為選擇性取代的低碳數烷基。在某些實施例,R6 係選自被一個或兩個羧基所選擇性取代的低碳數烷基。在 某些實施例,&為取代三唑,其取代基係選自被一個或兩 個叛基、氣基及雙烧基碟酸(dialkylphosphonate)所選擇性 取代的低碳數烧基’且該低碳數烷基係被至少一個羧基所 取代。 在某些實施例,R!係選自: (及)-5-羥基戊烷-2-基; (R)-5-(叛基甲基胺)-5-氧戊垸_2_基; (R)-5-((S)-l,2c叛基乙基胺)·5-氧戊烷基; (R)-5-四-丁氧基-5-氧戊烧-2-基; (R)-4-羧基丁烷-2-基; (R)-5-(4-(2_羥基乙烷唑小基)戊烷_2_基; (R)-5-(4-(2-胺-2-羧基乙燒)切4,2,3_三唑小基)戊烷_2_基; (R)-5-(4-(2-魏基-2-(二乙氧膦酸)乙基)三唑小基)戊烷-2_ 基; ⑻-5-(4-(2-羧基乙烷)-讯1,2,3-三唑小基)戊院-2-基; (R)_5K3-羧基丙统)-讯1,2,3-三唑小基)戊烧-2-基;及 (R)-5-(二苯氧膦酸)戊烷:基. 25 200804411 在某些實施例,R2係選自羥基、3-羧基丙醯基氧基及 (R)-2-胺-3-羧基丙醯基氧基。 在某些實施例,R3係為氫。 在某些實施例,R2及R3 —起附著於碳上,形成氧基。 進一步提供一醫藥組合物,係包含至少一種化學物質及一醫 藥上可接受之載體,前述具有治療上有效量之化學物質係選自化 學式I。 進一步提供一種抑制α-2,3唾液酸轉移酶活性之方法, 係包含以一足以察覺使糖結合物唾液酸化減少之至少一種 選自化學式I所示化合物之化學物質的量與表現性α-2,3唾 液酸轉移酶之細胞相接觸。在某些實施例,細胞係為癌細 胞;在某些實施例,細胞係經細胞遷移過程;在某些實施 例,前述細胞遷移係伴隨癌細胞遷移;在某些實施例,該 細胞出現於哺乳動物;在某些實施例,該哺乳動物為人類。 進一步提供一種治療患有對抑制α-2,3唾液酸轉移酶過 度活化有反應之疾病的患者之方法,包括施予患者至少一 種具有治療上有效量之化學物質,該化學物質係選自化學 式I。在某些實施例,前述至少一種選自化學式I的化學物 質包含一醫藥組合物。在某些實施例,對於α-2,3唾液酸轉 移酶活性抑制有所反應之疾病為癌症;在某些實施例,細 胞係經細胞遷移過程;在某些實施例,前述細胞遷移係伴 隨癌細胞遷移;在某些實施例,該細胞出現於哺乳動物; 在某些實施例,該哺乳動物為人類。在某些實施例,治療 患有對抑制α-2,3唾液酸轉移酶過度活化有反應之疾病的患 26 200804411 者之方法,包括額外施予患者至少一種能對合併治療有所 影響的治療劑。 在某些實施例,醫藥組合物可包含至少一種目前已揭 露之化學物質及一種對合併治療有影響的治療劑。本發明 所揭露之化學物質亦可與習知的治療劑及抗癌藥物合併使 用。熟習此項技術之人能夠分辨藥物的併用是基於藥物的 某種特性以及涉及何種癌症。在此領域中,已有存在多種 化療方法,此種抗癌藥物包括,但不限於雌激素受體調節 劑、細胞生長抑制劑/細胞毒劑、抗增殖劑、cell Cycle checkpoint inhibitors、血管生成抑製劑、單株抗體靶向治療 別(monoclonal antibody targeted therapeutic agents)、路月安 酸激酶抑制劑、絲胺酸/蘇鞍酸激酶抑制劑、組蛋白去乙^ 化酶抑制劑、熱休克蛋白抑制劑及法尼西基轉移酶抑^ 劑。本發明所揭露之化學物質亦可用於放射線治療。衣 為了在應用於人體或或其他哺乳動物之前確認治療戈 預防的作用,本發明所揭露之化學物質可先進行體外及 内试驗。舉例來說,體外試驗可用以決定是否施予—目& 揭露之特定化合物或合併使用一個可有效抑制α_2,3唾液^ 轉移酶活性或至少治療一種疾病的化合物。在某些實扩 例,在此所指可抑制α-2,3唾液酸轉移酶活性之化學&質^ 體外生化活性分析中的半抑制濃度%。少於25() μΜ,·在 些實施例,在此所指可抑制 物質在體:生化活性分析中的半抑制濃度lC5〇少: ΙΟΟμΜ,在某些實施例,在此所指可抑制α_2,3唾液酸. 酶活性之化學物質在體外生化活性 二夕 (IC5〇)少於25μΜ; 辰度 27 200804411 本發明所揭露之化學物質亦可顯示利用動物模型系統 是有效、安全的。在某些實施例,本發明所揭露之至少一 種化學物質的治療上有效量能在不產生實質毒性的狀況 下,提供治療上的益處。目前所揭露之化學物質的毒性可 利用一藥學上的標準程序,由熟習該項技術之人很快地加 以確認。毒性與療效性之劑量比係為治療指數,本發明所 揭露之化學物質可能在治療疾病或異常上顯示出高治療指 數。本發明所揭露之化合物之劑量可介在一少量或無毒性 循環濃度的範圍。 在作為藥物時,本發明所揭露之化學物質可以醫藥組 合物的形式施用。此類組合物可利用製藥領域中所熟知的 方式製備,並可包含至少一種本發明所揭露之化學物質。 在治療疾病時,可施打本發明所揭露之化學物質之治療上 有效量。可被瞭解的是,醫生可依照相關的條件決定化合 物施打的量,包括治療情況、給藥途徑的選擇、實際施打 的化合物、年齡、體重、個體施打後的反應、個體症狀的 嚴重度及類似情況。 本發明所揭露之醫藥組合物可包含目前所揭露的化學 物質之治療上的有效量及一醫藥上可接受之載體。本發明 所揭露之醫藥組合物可額外包含一種可提高本發明所揭露 之一種或多種化學物質療效的其他化合物。例如,但不受 限於學理,藉由有效增加此類化合物於血漿中的濃度;或 於施打前、輸送於血漿的過程中、在血漿中減少目前所揭 露之化學物質的降解;或藉由提高腸胃道對化合物的吸收 以增加血漿中的濃度,都可提高目前所揭露之化學物質之 療效。本發明所揭露之醫藥組合物亦可額外包含一治療藥 28 200804411 物,其係可正常施打以治療疾病或異常,以提供上述之合 併治療。 目前所揭露的某些實施例係關於包含一種或多種作 為活性成分的化學物質之組合物,可與醫藥上可接受之賦 形劑一起使用。在製作本發明所揭露之某些化合物時,該 活性成分可與賦形劑混合、以賦形劑稀釋、或以膠囊、小 袋(sachet)、紙或其他類似容器為載體封裹住活性成分。 當賦形劑作為稀釋劑時,其可為固態、半固態或液態物質, 其係作為一種活性成分的載具(例如:載體)。因此,舉 例來說,前述組合物呈現的形式可以是藥片、藥丸、粉末、 錠劑、小袋、膠囊、萬能藥(elixirs )、懸浮物、乳劑、溶 液、軟膠囊、硬膠囊及包含例如··約佔至少一種本發明所 揭露之化合物的1-90%重量比的糖漿。 製備化合物時,與其他成分結合之前,必須先將活性 化合物研磨成適當的大小。若活性化合物不可溶,該活性 化合物一般可被研磨至顆粒大小小於200目(mesh);若 該活性化合物為水溶性,其顆粒大小可藉由研磨方式,提 供單一形式的顆粒大小,例如:40目。 適當的賦形劑的例子包括,但不限於乳糖、葡萄糖、 蔗糖、山梨糖醇、甘露糖醇、澱粉、阿拉伯樹膠、磷酸鈣、 藻酸鹽、黃蓍膠、明膠、石圭酸妈、微晶纖維素、聚乙稀σ比 咯烷酮、修飾環糊精、纖維素、水、糖漿及甲基纖維素。 某些組合物可額外包含:潤滑劑,如:滑石、硬脂酸鎂、 礦物油;溼潤劑;乳化劑及懸浮劑;防腐劑,如:甲基及 propylhydroxy-benzoates ;甜味劑;及調味劑。目前已揭露 29 200804411 的化合物可以配製,以便利用習知的程序施打化合於個體 時’能提供快速、持續或緩慢釋放地活性物質。(到076段 結束) ' 某些已揭露之化合物可以單位劑裂的方式調配,每一 個劑量包括,例如:約0.1 mg〜2 g的活性成分。在此所使 用之「單位劑型」係指適合作為人體與其他哺乳動物之單 一劑量的物理上可分離單位;每單位包含一預先決定、經 计异可產生預定效果的活性物質的量,並與一合適的醫藥 上賦形劑、稀釋劑、載體及佐劑搭配使用。在某些實施態 樣,目前所揭露的化合物可以多次劑量加以配製。目前所 揭露的化學物質的量可與其他物質及治療藥物搭配使用, 以產製目前所揭露、可隨疾病、個體及特定給藥模式的不 同而變化的單一劑型組合物。 為了製備如藥片的固態組合物,主要活性成分可與醫 藥上可接受之賦形劑混合,形成一包含目前所揭露之化合 物之均質混合物之固態前配方組合物。談到這些均質前配 方化合物’意指其中的活性成分以均勻分佈於組合物中, 以便该組合物能被很快地分成如:藥片、藥丸及膠囊的相 同功效單一劑型。固態前配方可以再細分如:治療上有效 量約0.1 mg到2 g的單一劑型。 目鈾所揭露含有某些組合物的藥片、藥丸可以利用包覆 或其他方式提供長效的劑型。例如:藥片或藥丸可包含一 内部劑量成分(inner dosage component)及一外部劑量成分 (outer dosage component ),而後者封裹住前者。^兩種成 分由-腸内層(entede layer)所區隔’以便抵抗在胃内所 30 200804411 產生的崩解並允許内部劑量成分完整到達十二指腸或#、緩 内部劑量成分的釋放。多種材料可用作此種腸内層或包覆 層’這類的材料包括聚合物酸或聚合物酸與蟲膠、·錄鼠醇 及醋酸纖維素等材料的混合物。 ^ 目前所揭露之液狀組合物可以口服、注射及其他類似方 式’包括水狀溶液、適當調味的糖漿、水狀或油狀懸浮物 及以食用油(如:棉籽油、麻油、椰子油、花生油)調配 的調味乳劑、萬能藥及類似載體。 在某些實施態樣,目前所揭露之醫藥組合物可利用口 服非口服、鼻吸入、於局部、直腸、鼻部、頻内或陰道 藉由灌注器或其他適當途徑給藥。目前所揭露之醫藥組合 物可包含一個或多個醫藥上可接受之載體,在某些實施態 樣,配方的pH直可利用醫藥上可接受之酸、鹼或缓衝劑^ 行調整,以提高調配後化合物或輸送型態的穩定性。在此 所使用的名詞「非口服」包括皮下、眼内、皮内、靜脈内、 腹腔内、肌肉内、關節内、門脈内、動脈内、滑膜内、胸 骨内、鞠膜内、病灶内、顱内(間質内)、腦室内、腦内注射 或灌注技術。 在某些貫施態樣,目前所揭露之醫藥組合物可利用口 服方式傳遞;在某些實施態樣,目前所揭露、不論是否含 至少一種的額外治療藥物之化合物,該化合物可以被調配 成=有載體或不含載體兩種形式;前述載體通常用以合成 ϋ怨劑型,如樂片及膠囊。在某些實施態樣,膠嚢可能設 什成能夠在腸胃道巾釋放配方裡的活性料,使該處的生 體利用率達到最大,並使藥物在循環全身前所產生的降解 200804411 達到最小。在某些實施態樣,至少一種的額外治療藥物可 包含於配方之中,以促進目前所揭露的化合物的吸收與額 外治療藥物進入全身循環。在某些實施態樣,稀釋劑、調 味劑、低熔點壤、植物油、潤滑劑、懸浮劑、藥片崩解劑 及黏結劑皆可加以運用。適當的口服劑量範圍係按化合物 的藥效而定,一般劑量約為每公斤體重0.1 mg〜20/mg。 在某些實施態樣,目前所揭露之醫藥組合物可選擇以非 口服方式傳遞;在某些實施態樣,在考慮選擇非口服方式 時,治療性組合物可於載體中,以無熱原、包含至少一種 目前所揭露之化學物質的非口服方式可接受之水狀溶液及 不論是否含額外治療藥物的方式存在;在某些實施態樣, 非口服注射式載體可以調製成無菌等張溶液,以利保存; 而前述非口服注射劑可以是含有至少一種目前所揭露之化 學物質、不論是否含額外治療藥物的無菌蒸餾水。在某些 實施態樣,醫藥組合物可包括至少一種目前所揭露的、包 含一種藥劑的化學物質,此類藥劑如:可注射均質微粒、 生物溶蝕性顆粒、聚合物,如:聚乙酸或聚羥基乙酸、小 球或微脂粒,以藉由一種長效注射劑來提供目前所揭露之 化合物的控制或持續釋放。在某些實施態樣,植入型給藥 裝置可以將目前所揭露之化合物傳輸至個體標的器官的血 漿及體内的特定部位。 在某些實施態樣,目前所揭露之醫藥組合物可調製成吸 入型;在某些實施態樣,目前所揭露、不論是否含額外治 療藥物的化合物可調製成吸入式乾燥粉末型;在某些實施 態樣,含至少一種目前所揭露、不論是否含額外治療藥物 的化合物可調製成喷劑輸送的推進劑;在某些實施態樣, 32 200804411 喷劑可以是氣霧狀;在某些實施態樣,溶液、粉末或目前 所揭露之化學物質所形成的乾膜,可以如喷霧式或蒸汽 式,以便傳送到肺部。 在某些實施態樣,醫藥組合物可調製供局部給藥使 用,包含在此所指之至少一種目前所揭露之化學物質的局 部型組合物可與相同領域中所熟知的多種載體物質混合, 例如:水、醇類、蘆薈、尿囊素、甘油、維生素A、E油、 礦物油、丙二醇、PPG-2十四酿丙酸鹽及類似物。其他適 合作為局部型之載體包括,例如,潤膚劑、溶劑、保濕劑、 增稠劑及粉末。 在某些實施態樣,組合物可經由植入已吸收、包覆了目 前所揭露之化合物的膜、海綿、或其他適當材料來進行局 部給藥。在某些實施態樣,可以將植入裝置至於任何合適 的組織或器官中,並經由擴散、長效緩釋的藥丸或持續給 藥的方式傳輸特定的藥物分子。 目前所揭露之醫藥組合物可包含任何醫藥上可接受之 鹽類、酯類、salt of an ester、或其他目前所揭露之化合物 的衍生物及前驅藥物,一旦將其施予給一接受者,就能夠 直接或間接地將目前所揭露之化合物或抑制型代謝物或其 殘餘物提供給該名接受者。衍生物及前驅藥物的例子包 括、但不限於醋酸、甲酸、苯甲酸及存在於目前所揭露之 化合物上類似的官能基衍生物(如:醇類或氨類官能基)。 衍生物及前驅藥物包括那些施打於哺乳動物時即可增加目 前所揭露之化學物質的生體利用率之化合物,例如:相對 於親本種(parent species),可使口服給藥的化合物能更快於 200804411 淋巴腺、及收或提鬲母化合向生物腔室之傳送,如:腦部或 ^ Jtb ^ » 以修飾1、Γ只轭態樣,目前所揭露之醫藥組合物可包含用 遷壓、、維持及保存的配方材料,包括組合物的pH值、滲 減#声度、’爭度、顏色、同向性(is〇t〇nicity )、氣味、 實^ 2 *铋定度、溶解及釋放率、吸收及穿透度。在某些 甘,,合適的配方材料包含,但不限於氨基酸(如: 胺酸、麵胺酸、天門冬胺酸、精胺酸、離胺酸)、抗微 物副、抗氧化劑(如:抗壞血酸、亞硫酸納及亞硫酸氫 辟1、緩衝劑(如:硼酸、碳酸氫鹽、Tris-HCl、檸檬酸、 $酸鹽及其他有機酸)、膨脹劑(如··甘露醇及甘胺酸)、 口合刎(如··乙烯二胺四乙酸(EDta))、錯合劑(如··咖 作因、聚乙烯吼咯烷酮、β_環糊精、羥丙基|環糊精及磺 丁基醚-β-環糊精)、填充劑、單醣、雙醣及其他碳水化合 物(如··葡萄糖、甘露糖及糊精)、蛋白質(如:如清白 ,白、明膠及免疫球蛋白)、著色劑、調味劑及稀釋劑、 =化劑、疏水性聚合物(如:聚乙烯吡咯烷酮)、低分子 里夕肽、鹽形成反離子(salt-forming counterions,如: 納)、防腐劑(如:氯化苯二甲烴銨、苯甲酸、水揚酸、 碲柳汞(thimerosal)、苯乙醇、對羥基苯曱酸甲酯、對羥 基笨甲酸丙酯、氯己啶、山梨酸及過氧化氫)、溶劑(如: 甘泊、丙二醇及聚乙二醇)、糖醇(如··甘露糖醇及山梨 糖醇)、懸浮劑、界面活性劑及濕潤劑(如:普朗尼克類 (Pluronics) 、PEG、山梨醇醣脂、聚山梨醇酯(如:聚 山梨醇酯20、聚山梨醇醋80、triton、氨基丁三醇、卵鱗 赌、膽固醇、tyloxapal)、穩定性增強劑(如:蔗糖及山梨 轉醇)、張力增強劑(如:鹼金屬鹵化物,例如:氣化鈉 34 200804411 及氣化鉀、甘露糖醇、山梨糖醇)、輸送載體、稀釋劑、 賦形劑及醫藥佐劑(Remington’s Pharmaceutical Sciences, 18th Edition, A.R. Gennaro, ed.? Mack Publishing Company (1990)) 〇 在某些實施態樣,最佳的醫藥組合物可由熟悉此項技 術之人依照如:所欲採取的給藥途徑、輸送型態劑量來決 定(例如可參照前述 Remington 的 Pharmaceutical Sciences 書中的内容)。在某些實施態樣,這樣的組合物可能影響 物理狀態、穩定性、體内釋放率及體内對於目前所揭露之 化學物質的廓清率。適當的劑量範圍約在25〜500 mg/ 天,同時,給藥劑量可隨著在個體的血漿中的莫耳量而調 整。組合物中劑量的輸送可經由單次給藥、多次給藥、持 續釋放、控制式持續释放或任何以適當間隔與速率所進行 藥物釋放。劑量範圍及療程由本領域中熟習該項技術之人 決定。在某些實施態樣,給藥頻率需考慮目前所揭露的化 學物質的藥學動力學參數以及醫藥組合物中所使用的任何 額外的治療藥物。在某些實施態樣,醫生可以持續給藥至 產生預定療效為止。組合物可單劑給藥、二劑或多劑給藥, 施予療效性活性化合物的時間可以相同、也可以不同,或 者,也可以利用植入裝置或導管進行連續性輸注,熟習該 項技術之人並可由例行性地進行劑量的進一步精算。舉例 來說,治療上有效量及攝取可由適當的劑量反應資料來決 定。 ' 治療特定疾病時,對於目前所揭露之組合物的需要量 可隨化合物及個體所需治療的疾病而定。一般來說,每日 劑量範圍約在100 ng/kg〜100 mg/kg,例如口服或頰部給藥 35 200804411 劑量為每公斤體重0.01 mg〜40 mg ;非口服劑量為每公斤 體重0.001 mg/kg〜20 mg/kg •,以及鼻腔給藥、吸入給藥、 通氣給藥劑量為0.05 mg〜1,000 mg 。 在某些實施態樣,可以在體外利用包含目前所揭露之 化合物、不論是否含額外治療藥物的醫藥組合物。舉例來 說,將以自個體取出的細胞、組織或器官接觸一含有至少 一種目前所揭露之化學物質、不論是否含額外治療藥物的 醫藥組合物,之後再將前述細胞、組織或器官植回個體。 如果需要,可利用包裝或分藥裝置將目前所揭露之組 合物分裝成含有活性成分之單一劑型,前述包裝或分藥裝 置同時有給藥的用法說明。這樣的包裝過配方包含至少一 種目前所揭露之化學物質及用以治療哺乳動物(尤其是人 類)之用藥指示;同時也提供處方資訊,例如··向患者或 照護者提供或作為包裝後的醫藥製劑上的一種標示。處方 資訊包含,例如··效果、劑型、給藥方式、禁忌症、不良 反應等關於藥物製劑的資訊。 目前所揭露之具體實施例將由以下實施例進一步加 以界定,該實施例將詳述目前所揭露之化學物質製備的過 程及利用目前所揭露之化學物質所為的試驗過程。在不脫 離目前揭露的範圍内,任何熟習此項技術之人,都能對材 料及方法加以修飾。 36 200804411 實施例 實施例一:新穎的可抑制α-2,3唾液酸轉移酶之膜通透性石膽 酸類似物 本例係描述石膽酸類似物的合成,其係利用類固醇基 團(steroidal moiety)改善石膽酸的膜通透性,並顯示出非 競爭型α-2,3唾液酸轉移酶(a-2,3-ST)會出現在胞苷一磷酸 -N —乙醯神經氨酸(CMP-Neu5Ac)類似物的過渡狀態中。大 豆皂甙I (化合物1)是一種具有大豆皂苷中的參醣的牢固 的五環系統(a rigid pentacyclic system),該系統已被確認 為在體内可產生明顯抑制唾液酸轉移酶作用(Ki = 2.1 μΜ) (Hsu et al., Gynecol. Oncol., 2005, 96:415) 〇 為了確認與 類固醇相關的化合物可抑制α-2,3唾液酸轉移酶的活性,將 對不同的類固醇化合物進行篩選,而半抑制濃度(IC50) 21 μΜ的石膽酸(化合物3 )已被確認是一種有效的抑制劑。 反應圖I顯示牢固的五環系統、大豆皂戒I (化合物1 )的 參醣結構及石膽酸(化合物3)的架構。當D環轉換為五元 環時,a鍵與b鍵會斷裂,化合物1會轉變為化合物3,同 時,末端的醇類氧化成羧酸。 ΎΊ 200804411 反應式1And pharmaceutically acceptable salts, solvates, chelating agents, non-covalent complexes, prodrugs, and mixtures thereof. The core is an arbitrary alkyl group; R2 may be selected from a trans group and an acidoxy group; R3 is hydrogen, or r2 and r3 together with the carbon to which it is attached form an oxy group (OXO group); when Ri is a carboxyl group-2 -Base, then R2 is not a warp group. In certain embodiments, the oxime is selected from the group consisting of -CHR4(CH2)nR5, η is 2 and 3; R4 is selected from hydrogen or an optionally substituted lower alkyl group; and R5 is selected from the group consisting of a carboxyl group, a guanamine group, and a selective substitution. Heteroaryl. In certain embodiments, n is 2; in certain embodiments, η is 3; in certain embodiments, R4 is selected from argon or a lower alkyl group; in certain embodiments, r4 is a lower alkyl group. In certain embodiments, R4 is methyl; in certain embodiments, R5 is selected from the group consisting of carboxy, -(C0)-NHR6, and an optionally substituted triazole, wherein r6 is selected from an optionally substituted lower alkyl group. . 24 200804411 In certain embodiments, R^-CHR4(CH2)nR5, wherein η is 2 and 3, & is selected from hydrogen or an optionally substituted lower carbon number; and & is selected from the group consisting of: (CO)-NHR6 and a substituted triazole wherein the triazole ring system is substituted with a monofunctional group r?, and R7 is an optionally substituted lower alkyl group. In certain embodiments, R6 is selected from the group consisting of lower alkyl groups that are optionally substituted with one or two carboxyl groups. In certain embodiments, & is a substituted triazole whose substituent is selected from the group consisting of a low carbon number alkyl group selectively substituted with one or two thiol, gas groups, and dialkylphosphonate The lower alkyl group is substituted with at least one carboxyl group. In certain embodiments, R! is selected from the group consisting of: (and)-5-hydroxypentan-2-yl; (R)-5-(demethylmethylamine)-5-oxopent-2-yl; (R)-5-((S)-l, 2c tracylethylamine)·5-oxopentanyl; (R)-5-tetra-butoxy-5-oxopentan-2-yl; (R)-4-carboxybutan-2-yl; (R)-5-(4-(2-hydroxypyrazolyl)pentan-2-yl; (R)-5-(4-( 2-amine-2-carboxyethyl bromide) cut 4,2,3-triazole small group)pentane_2-yl; (R)-5-(4-(2-weiyl-2-(diethoxy) Phosphonic acid)ethyl)triazole small)pentan-2-yl; (8)-5-(4-(2-carboxyethane)-in-1,2,3-triazole small) pentan-2-yl (R)_5K3-carboxypropyl)-- 1,2,3-triazole small group) amyl-2-yl; and (R)-5-(diphenylphosphinic acid)pentane: yl. 25 200804411 In certain embodiments, R2 is selected from the group consisting of hydroxyl, 3-carboxypropenyloxy, and (R)-2-amine-3-carboxypropenyloxy. In certain embodiments, R3 is hydrogen. In certain embodiments, R2 and R3 are attached to carbon to form an oxy group. Further provided is a pharmaceutical composition comprising at least one chemical and a pharmaceutically acceptable carrier, wherein said therapeutically effective amount of the chemical is selected from the group consisting of Formula I. Further provided is a method for inhibiting the activity of α-2,3 sialyltransferase, comprising an amount and a performance α of at least one chemical selected from the compound of formula I, which is sufficient to detect a reduction in sialylation of the sugar conjugate. The cells of the 2,3 sialyltransferase are in contact. In certain embodiments, the cell line is a cancer cell; in certain embodiments, the cell line undergoes a cell migration process; in certain embodiments, the aforementioned cell line is accompanied by cancer cell migration; in certain embodiments, the cell is present in Mammal; in certain embodiments, the mammal is a human. Further provided is a method of treating a patient suffering from a disease responsive to inhibition of alpha-2,3 sialyltransferase overactivation, comprising administering to the patient at least one therapeutically effective amount of a chemical selected from the group consisting of: I. In certain embodiments, at least one of the aforementioned chemical species selected from Formula I comprises a pharmaceutical composition. In certain embodiments, the disease responsive to inhibition of alpha-2,3 sialyltransferase activity is cancer; in certain embodiments, the cell line undergoes a cell migration process; in certain embodiments, the aforementioned cell migration system is accompanied Cancer cell migration; in certain embodiments, the cell is present in a mammal; in certain embodiments, the mammal is a human. In certain embodiments, the method of treating a patient having a response to inhibition of alpha-2,3 sialyltransferase overactivation comprises administering to the patient at least one treatment that affects the combination therapy Agent. In certain embodiments, the pharmaceutical composition can comprise at least one currently disclosed chemical and a therapeutic agent that has an effect on the combination therapy. The chemical substances disclosed in the present invention can also be used in combination with conventional therapeutic agents and anticancer drugs. Those skilled in the art are able to distinguish between the combination of drugs and the nature of the drug and the type of cancer involved. In this field, there are a variety of chemotherapy methods, including, but not limited to, estrogen receptor modulators, cytostatic/cytotoxic agents, antiproliferative agents, cell cycle checkpoint inhibitors, angiogenesis inhibitors. Monoclonal antibody targeted therapeutic agents, lunar acid kinase inhibitors, serine/sarranic acid kinase inhibitors, histone deacetylase inhibitors, heat shock protein inhibitors And farnesyl transferase inhibitor. The chemical substances disclosed in the present invention can also be used for radiation therapy. In order to confirm the effect of treatment prevention before application to humans or other mammals, the chemical substances disclosed in the present invention may be tested in vitro and in vivo first. For example, in vitro assays can be used to determine whether to administer a specific compound disclosed or to use a compound that is effective in inhibiting alpha 2,3 salivary transferase activity or at least treating a disease. In some of the examples, the percent inhibition concentration in the chemical & mass in vitro biochemical activity assay for inhibition of alpha-2,3 sialyltransferase activity is referred to herein. Less than 25 () μΜ, in some embodiments, the semi-inhibitory concentration of the inhibiting substance in the body: biochemical activity analysis is less than: ΙΟΟμΜ, in some embodiments, the α_2 is inhibited herein. 3 sialic acid. The chemical activity of the enzyme activity in vitro biochemical activity (IC5〇) is less than 25μΜ; 辰度27 200804411 The chemical substance disclosed in the present invention can also show that the animal model system is effective and safe. In certain embodiments, a therapeutically effective amount of at least one of the chemicals disclosed herein provides a therapeutic benefit without substantial toxicity. The toxicity of the currently disclosed chemical substances can be quickly confirmed by a person skilled in the art using a standard pharmaceutical procedure. The dose ratio between toxic and therapeutic is a therapeutic index, and the chemical disclosed herein may exhibit a high therapeutic index in treating a disease or abnormality. The dosage of the compounds disclosed herein may be in the range of a small or non-toxic circulating concentration. In the case of a medicament, the chemical substance disclosed in the present invention can be administered in the form of a pharmaceutical composition. Such compositions may be prepared in a manner well known in the pharmaceutical art and may comprise at least one of the chemicals disclosed herein. In the treatment of a disease, a therapeutically effective amount of the chemical disclosed in the present invention can be administered. It can be understood that the doctor can determine the amount of the compound to be administered according to the relevant conditions, including the treatment situation, the choice of the administration route, the actual compound to be administered, the age, the body weight, the reaction after the individual is beaten, and the serious symptoms of the individual. Degree and similar situations. The pharmaceutical compositions disclosed herein may comprise a therapeutically effective amount of a chemical as disclosed herein and a pharmaceutically acceptable carrier. The pharmaceutical compositions disclosed herein may additionally comprise an additional compound which enhances the efficacy of one or more of the chemicals disclosed herein. For example, but not limited to the theory, by effectively increasing the concentration of such compounds in plasma; or reducing the degradation of currently exposed chemicals prior to application, during plasma delivery, or in plasma; or By increasing the absorption of the compound by the gastrointestinal tract to increase the concentration in the plasma, the efficacy of the currently disclosed chemical substances can be improved. The pharmaceutical composition disclosed herein may additionally comprise a therapeutic agent 28 200804411 which is normally administered to treat a disease or disorder to provide a combination therapy as described above. Certain embodiments disclosed herein are directed to compositions comprising one or more chemical substances as active ingredients for use with pharmaceutically acceptable excipients. In making certain of the compounds disclosed herein, the active ingredient may be mixed with excipients, diluted with excipients, or enclosed in a capsule, sachet, paper, or other similar container. When the excipient serves as a diluent, it may be a solid, semi-solid or liquid material which is a carrier (e.g., carrier) as an active ingredient. Thus, for example, the foregoing compositions may be in the form of tablets, pills, powders, lozenges, sachets, capsules, elixirs, suspensions, emulsions, solutions, soft capsules, hard capsules, and, for example, A syrup having from about 1 to 90% by weight of at least one of the compounds disclosed herein. When preparing a compound, the active compound must be ground to an appropriate size before being combined with other ingredients. If the active compound is insoluble, the active compound can generally be ground to a particle size of less than 200 mesh; if the active compound is water soluble, the particle size can be provided by milling to provide a single form of particle size, for example: 40 Head. Examples of suitable excipients include, but are not limited to, lactose, glucose, sucrose, sorbitol, mannitol, starch, gum arabic, calcium phosphate, alginate, tragacanth, gelatin, sarcophagus, micro Crystalline cellulose, polyethylene σ pyrrolidone, modified cyclodextrin, cellulose, water, syrup and methyl cellulose. Some compositions may additionally comprise: lubricants such as: talc, magnesium stearate, mineral oil; wetting agents; emulsifying and suspending agents; preservatives such as methyl and propylhydroxy-benzoates; sweeteners; Agent. It has now been disclosed that the compounds of 29 200804411 can be formulated to provide rapid, sustained or slow release of the active substance when administered to a subject using conventional procedures. (To the end of paragraph 076) 'Some of the disclosed compounds can be formulated in unit dosage form, each dose including, for example, about 0.1 mg to 2 g of active ingredient. As used herein, "unit dosage form" refers to a physically separable unit that is suitable as a single dosage of the human body and other mammals; each unit contains a predetermined amount of active substance that is predetermined to produce a predetermined effect, and A suitable pharmaceutical excipient, diluent, carrier and adjuvant are used in combination. In certain embodiments, the presently disclosed compounds can be formulated in multiple doses. The amount of chemical currently disclosed can be used in conjunction with other substances and therapeutic agents to produce a single dosage form composition that is disclosed as being variable with the disease, the individual, and the particular mode of administration. To prepare a solid composition such as a tablet, the principal active ingredient may be combined with apharmaceutically acceptable excipient to form a solid pre-formulation composition comprising a homogeneous mixture of the presently disclosed compounds. Reference to these pro-fermentation formulas' means that the active ingredients therein are evenly distributed throughout the composition so that the composition can be quickly divided into single dosage forms of the same efficacy as tablets, pills and capsules. The pre-solids formulation can be subdivided into a single dosage form that is therapeutically effective from about 0.1 mg to 2 g. Tablets and pills containing certain compositions disclosed by uranium can be coated or otherwise provided with a long-lasting dosage form. For example, a tablet or pill may contain an inner dosage component and an outer dosage component, while the latter encloses the former. ^The two components are separated by an entere layer to resist disintegration in the stomach 30 200804411 and allow the internal dose component to reach the duodenum or #, slow release of internal dose components. A variety of materials can be used as such an enteric layer or coating. Materials such as polymeric acids or polymeric acids are blended with materials such as shellac, hammer, and cellulose acetate. ^ The liquid compositions disclosed herein may be administered orally, by injection and in other similar manners, including aqueous solutions, suitably flavored syrups, aqueous or oily suspensions, and edible oils (eg, cottonseed oil, sesame oil, coconut oil, Peanut oil) formulated flavoring emulsion, panacea and similar carriers. In certain embodiments, the presently disclosed pharmaceutical compositions can be administered orally, nasally, topically, rectally, nasally, intravitally, or vaginally by a perfusion device or other suitable route. The presently disclosed pharmaceutical compositions may comprise one or more pharmaceutically acceptable carriers. In certain embodiments, the pH of the formulation may be adjusted using a pharmaceutically acceptable acid, base or buffer. Improve the stability of the compound or delivery profile after formulation. The term "non-oral" as used herein includes subcutaneous, intraocular, intradermal, intravenous, intraperitoneal, intramuscular, intraarticular, intraportal, intraarterial, intrasynovial, intrasternal, intraperorbital, lesions. Internal, intracranial (interstitial), intraventricular, intracerebral injection or perfusion techniques. In certain embodiments, the presently disclosed pharmaceutical compositions can be delivered orally; in certain embodiments, the compounds disclosed herein, whether or not comprising at least one additional therapeutic agent, can be formulated into = There are two forms of carrier or carrier; the aforementioned carriers are usually used to synthesize sputum dosage forms such as tablets and capsules. In some embodiments, the capsule may be designed to release the active ingredient in the gastrointestinal tract release formulation, maximizing the bioavailability of the drug, and minimizing the degradation of the drug before it circulates throughout the system. . In certain embodiments, at least one additional therapeutic agent can be included in the formulation to facilitate absorption of the presently disclosed compounds and additional therapeutic agents into the systemic circulation. In certain embodiments, diluents, flavoring agents, low melting point soils, vegetable oils, lubricants, suspending agents, tablet disintegrating agents, and binders can be utilized. The appropriate oral dosage range will depend on the potency of the compound, which is generally about 0.1 mg to 20 mg per kg of body weight. In certain embodiments, the presently disclosed pharmaceutical compositions can optionally be delivered in a non-oral manner; in certain embodiments, the therapeutic composition can be in a carrier, without pyrogens, when considering a non-oral regimen. a non-oral acceptable aqueous solution comprising at least one of the presently disclosed chemistries and whether or not it contains an additional therapeutic agent; in certain embodiments, the parenteral injectable carrier can be formulated as a sterile isotonic solution The above parenteral injection may be sterile distilled water containing at least one of the currently disclosed chemicals, whether or not containing additional therapeutic agents. In certain embodiments, the pharmaceutical composition can include at least one currently disclosed chemical comprising a pharmaceutical agent such as: injectable homogeneous microparticles, bioerodible particulates, polymers such as polyacetic acid or poly Glycolic acid, pellets or vesicles to provide controlled or sustained release of the presently disclosed compounds by a long-acting injection. In certain embodiments, an implantable drug delivery device can deliver a compound disclosed herein to a plasma of an individual's target organ and to a particular site in the body. In certain embodiments, the presently disclosed pharmaceutical compositions are adjustable to be inhaled; in certain embodiments, the presently disclosed compounds, whether or not containing additional therapeutic agents, can be formulated as inhaled dry powders; In some embodiments, at least one of the presently disclosed compounds, whether or not containing additional therapeutic agents, can be formulated as a propellant for spray delivery; in certain embodiments, 32 200804411 sprays can be aerosolized; In the embodiment, the dry film formed by the solution, powder or the currently disclosed chemical may be sprayed or vaporized for delivery to the lungs. In certain embodiments, the pharmaceutical compositions can be formulated for topical administration, and the topical compositions comprising at least one of the currently disclosed chemistries can be combined with a variety of carrier materials well known in the art. For example: water, alcohols, aloe vera, allantoin, glycerin, vitamin A, E oil, mineral oil, propylene glycol, PPG-2 fourteen propionate and the like. Other suitable carriers for topical type include, for example, emollients, solvents, humectants, thickeners, and powders. In certain embodiments, the composition can be administered topically via implantation of a film, sponge, or other suitable material that has absorbed, coated the compound disclosed herein. In certain embodiments, the implant device can be delivered to any suitable tissue or organ and the particular drug molecule delivered via a diffusing, long-acting sustained-release pill or sustained administration. The presently disclosed pharmaceutical compositions may comprise any pharmaceutically acceptable salts, esters, salts of an ester, or other derivatives and precursors of the presently disclosed compounds, once administered to a recipient, It is possible to provide the currently disclosed compound or inhibitory metabolite or residue thereof to the recipient directly or indirectly. Examples of derivatives and prodrugs include, but are not limited to, acetic acid, formic acid, benzoic acid, and similar functional derivatives (e.g., alcohol or amine functional groups) present in the presently disclosed compounds. Derivatives and prodrugs include those compounds which increase the bioavailability of the currently disclosed chemical when applied to a mammal, for example, a compound that can be administered orally relative to a parent species Faster than 200804411 Lymphatic gland, and transfer or transfer of the progeny to the biological chamber, such as: brain or ^ Jtb ^ » to modify 1, Γ yoke, the currently disclosed pharmaceutical composition can be used Formulation materials for migration, maintenance and preservation, including pH value of the composition, permeability, sound, 'contention, color, isotropic (is〇t〇nicity), odor, real ^ 2 *铋, dissolution and release rate, absorption and penetration. In some cases, suitable formulations include, but are not limited to, amino acids (eg, amino acids, face acid, aspartic acid, arginine, lysine), anti-microbial agents, antioxidants (eg: Ascorbic acid, sodium sulfite and hydrogen sulfite, buffers (eg boric acid, bicarbonate, Tris-HCl, citric acid, acid salts and other organic acids), bulking agents (eg · mannitol and glycine) Acid), mouth 刎 (such as · ethylene diamine tetraacetic acid (EDta)), a mixture of agents (such as · caffeine, polyvinyl pyrrolidone, β_cyclodextrin, hydroxypropyl | cyclodextrin And sulfobutyl ether-β-cyclodextrin), fillers, monosaccharides, disaccharides and other carbohydrates (such as · glucose, mannose and dextrin), proteins (such as: white, white, gelatin and immune Globulin, coloring agents, flavoring agents and diluents, chemical agents, hydrophobic polymers (eg polyvinylpyrrolidone), low molecular weight peptides, salt-forming counterions (eg, nano), Preservatives (eg, dimethylammonium chloride, benzoic acid, salicylic acid, thimerosal, benzene) Ethanol, methyl p-hydroxybenzoate, propyl p-hydroxybenzoate, chlorhexidine, sorbic acid and hydrogen peroxide), solvents (eg, ganpo, propylene glycol and polyethylene glycol), sugar alcohols (eg ··· Mannitol and sorbitol), suspending agents, surfactants and wetting agents (eg, Pluronics, PEG, sorbitol glycolipids, polysorbates) (eg polysorbate 20, poly Sorbitol vinegar 80, triton, tromethamine, egg gambling, cholesterol, tyloxapal), stability enhancers (such as: sucrose and sorbitol), tension enhancers (such as: alkali metal halides, such as: gasification Sodium 34 200804411 and potassium carbonate, mannitol, sorbitol), delivery vehicles, diluents, excipients and pharmaceutical adjuvants (Remington's Pharmaceutical Sciences, 18th Edition, AR Gennaro, ed.? Mack Publishing Company (1990) In certain embodiments, the optimal pharmaceutical composition can be determined by a person skilled in the art in accordance with, for example, the route of administration to be administered, the type of delivery mode (for example, reference to the aforementioned Pharmaceutical Sciences of Remington). In certain embodiments, such compositions may affect physical state, stability, in vivo release rate, and clearance of the currently disclosed chemical species in the body. Suitable dosage ranges are about 25 ~500 mg / day, at the same time, the dose can be adjusted with the amount of mole in the plasma of the individual. The delivery of the dose in the composition can be sustained by a single administration, multiple administrations, sustained release, controlled Release or any drug release at appropriate intervals and rates. The dosage range and course of treatment are determined by those skilled in the art. In certain embodiments, the frequency of administration will take into account the pharmacokinetic parameters of the currently disclosed chemical materials as well as any additional therapeutic agents used in the pharmaceutical compositions. In certain embodiments, the physician can continue to administer until a predetermined therapeutic effect is achieved. The composition may be administered in a single dose, in two or more doses, and the time of administration of the therapeutically active compound may be the same or different, or the implant or catheter may be used for continuous infusion, and the technique is familiar to the technology. The person can be routinely dosed for further actuarial. For example, therapeutically effective amounts and ingestions can be determined by appropriate dose response data. When treating a particular disease, the amount of the presently disclosed composition may depend on the compound and the condition to be treated by the individual. In general, the daily dose range is from about 100 ng/kg to 100 mg/kg, for example, oral or buccal administration 35 200804411. The dose is 0.01 mg to 40 mg per kg of body weight; the non-oral dose is 0.001 mg per kg of body weight. Kg~20 mg/kg •, as well as nasal, inhaled, and ventilated doses of 0.05 mg to 1,000 mg. In certain embodiments, pharmaceutical compositions comprising the presently disclosed compounds, whether or not containing additional therapeutic agents, can be utilized in vitro. For example, a cell, tissue or organ removed from an individual is contacted with a pharmaceutical composition containing at least one of the currently disclosed chemicals, whether or not containing an additional therapeutic agent, and then the aforementioned cells, tissues or organs are returned to the individual. . If desired, the presently disclosed compositions can be packaged into a single dosage form containing the active ingredient by means of a package or dispensing device, and the aforementioned packaging or dispensing device also has instructions for administration. Such packaged formulations comprise at least one of the currently disclosed chemicals and instructions for the treatment of a mammal, especially a human; and also provide prescription information, for example, to a patient or caregiver or as a packaged medicine A label on the preparation. Prescription information includes information about pharmaceutical preparations such as effects, dosage forms, administration methods, contraindications, and adverse reactions. The specific embodiments disclosed herein will be further defined by the following examples which illustrate the process of preparing the presently disclosed chemical materials and the testing procedures utilized by the presently disclosed chemistries. Anyone familiar with the art can modify the materials and methods without departing from the current disclosure. 36 200804411 EXAMPLES Example 1: Novel Membrane Permeability Corrosion Inhibition of α-2,3 Sialyltransferases This example describes the synthesis of lithocholic acid analogs using steroid groups ( The steroidal moiety) improves the membrane permeability of lithocholic acid and shows that non-competitive α-2,3 sialyltransferase (a-2,3-ST) occurs in cytidine-phosphate-N-acetamidine In the transition state of the (TC-Neu5Ac) analog. Soybean saponin I (Compound 1) is a robust pentacyclic system with a ginseng in soy saponin, which has been identified to produce significant inhibition of sialyltransferase activity in vivo (Ki = 2.1 μΜ) (Hsu et al., Gynecol. Oncol., 2005, 96:415) 〇 In order to confirm that steroid-related compounds inhibit the activity of α-2,3 sialyltransferase, different steroid compounds will be screened. A semi-inhibitory concentration (IC50) of 21 μΜ of lithocholic acid (Compound 3) has been identified as a potent inhibitor. Reaction Scheme I shows the structure of a strong pentacyclic system, the sugar structure of soy soap ring I (compound 1) and the structure of lithocholic acid (compound 3). When the D ring is converted to a five-membered ring, the a bond and the b bond are broken, and the compound 1 is converted into the compound 3, and at the same time, the terminal alcohol is oxidized to a carboxylic acid. ΎΊ 200804411 Reaction 1

隨後進行膽酸類似物(化合物7-22)合成反應,以確 遇a-2,3-ST抑制劑之膜通透性。化合物的合成是利用以下 材料、根據以下的一般流程所進行,所有的化學藥品購自 Sigma、Aldrich或Acros Organics,以及所有的氨基酸購 自Advanced Chemtech。CMP-Neu5Ac合成反應是以唾液酸 為起始物(Traving & Schauer,CW/· Mo/· 1>汾 Scz··,1998, 54:1330; Schwarzkopf et al., Proc. Natl. Acad. Sci. USA, 2002, 99:5267);乳糖受器的合成在Harduin-Lepers等人的書中有 所描述(Harduin-Lepers et al·,β沁c/i/mie,2001,83:727 )。老 鼠肝的2,3-ST取自CalBiochem,其濃度為3.7 mU/pL、儲 存於-80°C,於一週内使用。以溶點測定器(Electrothermal MEL-TEMP)記錄溶點;以Perkin Elmer FT-IR紅外光譜儀紀 錄 FT-IR 光譜(Paragon 1000,以 KBr 壓片);以 Bruker AMX400或500 MHz 儀器紀錄1H and 13C核磁共振光 譜;質子化學位移(δ)之記述係以百萬分之一為單位,該單 位係相對比較氘-氣仿中之氣仿之次曱基殘基在7.24 ppm之 單峰訊號,或氘甲醇(CD3OD-d4 )中之1-H-氘曱醇 38 200804411 (CHD2OD)之甲基殘基在3.30 ppm之五重峰訊號定義而 得。碳化學位移之記述係相對比較CDC13及CD3OD-d4中之 13C 訊號(77.0ppm,49.0ppm)之定義而得。以 FAB JMS-700 雙聚焦質譜儀(JEOL,Tokyo,Japan), MALDI Voyager DE-PRO (Applied Biosystem Houston,USA)及 ESI Finnigan LCQ 質譜儀 ESI ( Finnigan LCQ mass spectrometer,Thermo Finnigan,San Jose,CA,United States)在負極模式測得質 譜。以使用 Vydac 214TP510 C4 管柱(1 cm x 25 cm)之逆 向高效液相層析來純化胜肽化合物,以獲取高於95%的純 度。分離的過程係以1%水、1%三氟醋酸及乙腈/0.05% TFA (B)作為沖提液。 類固醇磷酸化的大致流程如下··將三(對—硝基苯紛)石舞 酸酯(1.30 mmol)、類固醇(1.43 mmol)溶解於二氯甲烧(2〇 cm3),再加入 7,1 l-diazabicyclo[5A0]undec-ll«ene (1 82 mmol),於室溫下攪拌十二小時後,以飽和的碳酸氫納溶液 洗提數次,再以硫酸納乾燥此有機萃取物,並於過滤後, 進行濃縮。將最後取得的淡黃色固體溶於二氣甲炫^ ( 1 ^ cm3),並添加甲醇(13.00 mmol ,0·54 cm3)及 7,ll-diazabicyclo[5.4.0]undec-ll-ene (6·5 mmol),於室、、西下 攪拌十五小時後,以飽和的碳酸氫納溶液洗提數z欠,#& 硫酸鈉乾燥此有機萃取物,並於過濾後,進行濃縮;並以 乙酸乙酯與己烷作為洗提液、利用矽膠純化殘餘物,以# 得終產物。 類固醇磷酸鹽甲氧基去保護化過程如下:鱗酸化類@ 醇化合物(0.25 mmol)溶於二氯甲烧(1.50cm3),添加二甲美 溴矽烷(0.13 cm3,1·00 mmol),於室溫下攪拌三十分鐘後二 39 200804411 添加飽和的碳酸氫納溶液終止反應,並以減壓濃縮 evaporation)移除溶劑。再以二氯甲烷與己烷沈澱混合物, 過濾、濃縮後;於真空系統中進行乾燥,以獲得終產物。 琥珀酸酐與類固醇偶合的大致流程如下:將類固醇(〇 69 mmol) >谷於口比口定(1〇 cm3) ’隨後,加入二甲氦基口比口定(〇 69 mmol)及琥珀酸酐(2·〇7 mmol)後,回流十五小時。以真空幫 浦移除吡啶後,將混合物溶於二氯甲烷,以飽和的碳酸氫 鈉及6% HC1氯化氫洗提。萃取物乾燥蒸發後產生粗產物, 如·稠油,係利用乙酸乙酯與己烷、以矽膠管柱層析獲取 產物。 3-輕基-石膽酸與氨基酸的酯化過程如下:在含保護基 的石膽酸(0.35 mmol)、氨基酸(〇·45 mmol)及4-二甲氨基吡 啶(0.10 mmol)的二氣甲烷(8 cm3)溶液中,二環己基^二亞 胺添加於二氯甲烷(2 cm3),於室溫下攪拌三十分鐘後,以 rotary evaporation移除溶劑後,再利用乙酸乙酯盥己烷 矽膠管柱層析獲取產物。 石膽酸與氨基酸胜肽鍵的形成過程如下··在含有石膽 酸(1.3^ mmol)與氨基酸α·39 mmol)的二曱基曱醯胺(DMF) (5 cm3)中加入二異丙基乙胺(3·99 mm〇l)及2-(lf苯并三唑 -1-基)-1,1,3,3-四甲基脲六氟磷酸酯(HBTU) (1牝 mmol),於室溫下攪拌一小時後,以真空移除溶劑。將混合 物溶於二氯甲烷(50 cm3),並以水洗提兩次,萃取物乾燥蒸 發後產生粗產物,如:稠油,係利用乙酸乙酯與己烷、以 石夕膠管柱層析獲取產物。 200804411The synthesis of the cholic acid analog (compound 7-22) is then carried out to confirm the membrane permeability of the a-2,3-ST inhibitor. The synthesis of the compounds was carried out according to the following general procedure using the following materials, all of which were purchased from Sigma, Aldrich or Acros Organics, and all of the amino acids were purchased from Advanced Chemtech. The CMP-Neu5Ac synthesis reaction is based on sialic acid (Traving & Schauer, CW/. Mo/·1> 汾Scz··, 1998, 54:1330; Schwarzkopf et al., Proc. Natl. Acad. Sci USA, 2002, 99: 5267); The synthesis of lactose receptors is described in Harduin-Lepers et al. (Harduin-Lepers et al., β沁c/i/mie, 2001, 83:727). The 2,3-ST of the old rat liver was taken from CalBiochem at a concentration of 3.7 mU/pL and stored at -80 ° C for use within one week. The melting point was recorded with an Electrothermal MEL-TEMP; the FT-IR spectrum was recorded on a Perkin Elmer FT-IR infrared spectrometer (Paragon 1000, compressed in KBr); 1H and 13C nuclear magnetic were recorded on a Bruker AMX400 or 500 MHz instrument. Resonance spectrum; the proton chemical shift (δ) is expressed in parts per million, which is a relatively single peak signal at 7.24 ppm, or 氘 in comparison with the gas-like imitation of the sulfhydryl group. The methyl residue of 1-H-nonanol 38 200804411 (CHD2OD) in methanol (CD3OD-d4) was defined by a five-fold signal of 3.30 ppm. The description of the carbon chemical shift is based on the comparison of the 13C signal (77.0 ppm, 49.0 ppm) in CDC13 and CD3OD-d4. FAB JMS-700 dual focus mass spectrometer (JEOL, Tokyo, Japan), MALDI Voyager DE-PRO (Applied Biosystem Houston, USA) and ESI Finnigan LCQ mass spectrometer ESI (Finnigan LCQ mass spectrometer, Thermo Finnigan, San Jose, CA, United States) measured mass spectra in negative mode. The peptide compound was purified by reverse-phase high performance liquid chromatography using a Vydac 214TP510 C4 column (1 cm x 25 cm) to obtain a purity higher than 95%. The separation was carried out with 1% water, 1% trifluoroacetic acid and acetonitrile/0.05% TFA (B) as the extract. The general procedure for steroid phosphorylation is as follows: · Dissolve tris(p-nitrophenyl) stone oxalate (1.30 mmol) and steroid (1.43 mmol) in methylene chloride (2〇cm3), then add 7,1 L-diazabicyclo[5A0]undec-ll«ene (1 82 mmol), after stirring for 12 hours at room temperature, eluting several times with saturated sodium bicarbonate solution, and drying the organic extract with sodium sulfate, and After filtration, it was concentrated. The finally obtained pale yellow solid was dissolved in dioxane (1 ^ cm 3 ), and methanol (13.00 mmol, 0·54 cm 3 ) and 7, ll-diazabicyclo [5.4.0] undec-ll-ene (6) were added. · 5 mmol), after stirring for 15 hours in the chamber and the west, eluting with a saturated sodium bicarbonate solution, drying the organic extract with #& sodium sulfate, and concentrating after filtration; Ethyl acetate and hexane were used as the eluent, and the residue was purified using yttrium gel to give the final product. The deprotection process of steroid phosphate methoxy is as follows: sulphate class @ alcohol compound (0.25 mmol) is dissolved in methylene chloride (1.50 cm3), and dimethyl bromide (0.13 cm3, 1.000 mmol) is added. After stirring for 30 minutes at room temperature, the reaction was terminated by adding a saturated sodium hydrogencarbonate solution, and the solvent was removed by evaporation under reduced pressure. The mixture was again precipitated with dichloromethane and hexanes, filtered and concentrated, and dried in vacuo to give the final product. The general procedure for the coupling of succinic anhydride with steroids is as follows: steroid (〇69 mmol) > gluten in the mouth (1〇cm3)' followed by the addition of dimethylhydrazine to the mouth (〇69 mmol) and succinic anhydride After (2·〇7 mmol), reflux for 15 hours. After the pyridine was removed by vacuum pumping, the mixture was dissolved in dichloromethane and eluted with saturated sodium hydrogen carbonate and 6% EtOAc. The extract is dried and evaporated to give a crude product, e.g., heavy oil, which is obtained by column chromatography using ethyl acetate and hexane. The esterification process of 3-light-lithocholic acid with amino acids is as follows: in the protective gas-containing lithocholic acid (0.35 mmol), amino acid (〇·45 mmol) and 4-dimethylaminopyridine (0.10 mmol) In a solution of methane (8 cm3), dicyclohexyldiimide was added to dichloromethane (2 cm3), stirred at room temperature for 30 minutes, then the solvent was removed with rotary evaporation, and then ethyl acetate was used. The product was obtained by chromatography on an alkane rubber column. The formation process of lithocholic acid and amino acid peptide bonds is as follows: Add diisopropyl ester to dimercaptodecylamine (DMF) (5 cm3) containing lithocholic acid (1.3^mmol) and amino acid α·39 mmol) Ethylethylamine (3·99 mm〇l) and 2-(lfbenzotriazol-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate (HBTU) (1牝mmol) After stirring at room temperature for one hour, the solvent was removed in vacuo. The mixture was dissolved in dichloromethane (50 cm3) and washed twice with water. The extract was dried and evaporated to give a crude product, e.g., heavy oil, obtained from ethyl acetate and hexanes. product. 200804411

Fmoc group去保護化大致的過程如下··在含有Fmoc 保護類固醇化合物(Fmoc-protected steroidal compound) (0.15 mmol)的二氣 f 烷(5 cm3)加入 7,ll-diazabicyclo[5.4.0]undec-ll-ene (0·15 mmol),於室溫下 攪拌三十分鐘,以減壓濃縮移除反應溶劑後產生粗產物, 如··稠油,之後係利用乙酸乙酯與己烷、以快速管柱層析 純化產物。The general process of deprotection of Fmoc group is as follows: Adding 1,ll-diazabicyclo[5.4.0]undec- in dioxane (5 cm3) containing Fmoc-protected steroidal compound (0.15 mmol) Ll-ene (0·15 mmol), stirred at room temperature for 30 minutes, concentrated under reduced pressure to remove the reaction solvent to give a crude product, such as heavy oil, followed by ethyl acetate and hexane. The product was purified by column chromatography.

Boc及tBu groups去保護基的大致過程如下:含保護 基化合物(0.15 mmol)的TFA溶液(2 cm3)在室溫下以2% 的水(0.04 cm3)進行處理,於室溫下攪拌三十分鐘,以減壓 濃移除反應溶劑,以飽和的碳酸氫鈉中和混合物後,以逆 相高效液相層析獲取純產物。 甲酯基還原的大致過程如下··將溶於四氫呋喃(1·6 cm3) 的類固醇甲酷(0·56 mmol),於〇〇c下,一滴一滴小心加入 含氫化鋁鋰(1.13 mmol)的四氫呋喃(〇·6 cm3)(所需時間超過 十分鐘)。混合物於室溫下攪拌十分鐘後,添加6% HC1終 止反應,將〃b合物過;慮並以rotary evaporation移除溶劑, 利用乙酸乙酯與己烷、以矽膠管柱層析純化最後的固體 物,以獲取終產物。 二苯磷酸酯類固醇合成的一般過程如下:將一級類固 醇留醇(2.02 mmol)、溶於含有二曱基磷酸基疊氮化物的四 氣咬喃(12 cm3) ’再一滴一滴地添加 7,1 l-diazabicycloDAOhndec-i Lene (5 〇5 mm〇1),於室溫下 攪拌=分鐘後,添加6% HC1終止反應;以減壓濃縮移除溶 劑,並以水及二氣曱烷萃取混合物。在萃取物於減壓環境 41 200804411 下乾煉療:發後’得到一粗萃取物,之後再利用乙酸乙酯與 己烧、以快速管柱層析純化出終產物。 類固醇®氮化物(steroidal azide)合成的一般過程如 下·將溶於1,4-二噁烷(15 cm3)的二苯基磷酸酯類固醇 (0.841 mmol)添加疊氮化納(4 2 mmol)、四丁基破化銨(〇 〇84 mmol)及15—冠—5醚(〇1 cm3),於氮氣壓力下利用回流 將混合物加熱一夜,待冷卻後,以減壓濃縮移除溶劑,並 j水及二氣甲烷萃取殘餘物。在萃取物於減壓環境下乾燥 蒸务後,仟到一粉末狀粗萃取物,利用乙酸乙酯與己烧、 以石夕膠管柱層析獲得產物。 一 雙取代1,2,3-三唑類固醇合成的一般過程如下:將 類固醇《氮化物(〇·26 mmol)及炔烴(〇·283 mmol)溶於四氫 呋喃與水的混合溶液中(4cm3, 1:1),再將硫酸銅(催化量)及 ^壞血酸鈉(催化量)加入,於室溫下攪拌三小時後,以減壓 濃縮移除溶劑,並以水及二氯$烷萃取混合物。在萃取物 於減壓裱境下乾燥蒸發後,得到一粉末狀粗萃取物,利用 乙酸乙酯與F醇、以矽膠管柱層析獲得產物。 速配接合組合化學(click chemistry)觀念的概念方法 大致如下··多種利用銅_化疊氮化合物中間產物盘適當 j炔烴接合成的1,4-雙取代U,3三唾,係根據R〇st_ev 寻人書中所述方法(Rostovtsev et al , (2〇〇2) , /狀祝” 41:2596)所產製而成。 42 200804411 化合物7之合成:(3/?,5反105,1311,145)-17-((/?)-5-羥基戊烷 -2-基)-10,13-二甲基-十六烧鼠-1//-¾戊烧[β]非-3-酉同The general procedure for deprotecting the Boc and tBu groups is as follows: a TFA solution (2 cm3) containing a protecting group compound (0.15 mmol) is treated with 2% water (0.04 cm3) at room temperature and stirred at room temperature. The reaction solvent was concentrated under reduced pressure, and the mixture was neutralized with saturated sodium hydrogen carbonate. The general procedure for the reduction of the methyl ester group is as follows: steroids (0·56 mmol) dissolved in tetrahydrofuran (1·6 cm3) are carefully added to the lithium aluminum hydride (1.13 mmol) in a drop of one drop. Tetrahydrofuran (〇·6 cm3) (more than ten minutes required). After the mixture was stirred at room temperature for ten minutes, the reaction was terminated by adding 6% HCl, and the hydrazine compound was passed; the solvent was removed by rotary evaporation, and the residue was purified by column chromatography using ethyl acetate and hexane. Solid matter to obtain the final product. The general procedure for the synthesis of diphenyl phosphate steroids is as follows: a primary steroid alcohol (2.02 mmol), dissolved in a tetrachabitane (12 cm3) containing dimercaptophosphate azide, and then added 7,1 drop by drop. l-diazabicycloDAOhndec-i Lene (5 〇5 mm〇1), after stirring at room temperature = minute, the reaction was quenched by the addition of 6% HCl. The solvent was concentrated under reduced pressure and the mixture was extracted with water and dioxane. The extract was subjected to dry refining in a reduced pressure environment 41 200804411: After the hair was taken, a crude extract was obtained, and then the ethyl acetate was burned with hexane to purify the final product by flash column chromatography. The general procedure for the synthesis of steroidal azide is as follows: Add a sodium azide (4 2 mmol) to a diphenyl phosphate steroid (0.841 mmol) dissolved in 1,4-dioxane (15 cm3). Tetrabutylammonium chloride (〇〇84 mmol) and 15-crown-5 ether (〇1 cm3) were heated under reflux under nitrogen pressure for one night. After cooling, the solvent was removed by concentration under reduced pressure. Water and two gas methane extraction residues. After the extract was dried under reduced pressure, the crude product was taken up in a powdery form, and the product was obtained by column chromatography using ethyl acetate and hexane. The general procedure for the synthesis of a double substituted 1,2,3-triazole steroid is as follows: The steroid "nitride (〇·26 mmol) and alkyne (〇·283 mmol) are dissolved in a mixed solution of tetrahydrofuran and water (4 cm3, 1:1), then add copper sulfate (catalytic amount) and sodium ascorbate (catalytic amount), stir at room temperature for three hours, then concentrate under reduced pressure to remove the solvent, and water and dichloro-alkane Extract the mixture. After the extract was dried and evaporated under reduced pressure, a crude powdery extract was obtained, which was obtained from ethyl acetate and ethyl alcohol. The conceptual method of the concept of "click chemistry" is as follows: a variety of 1,4-disubstituted U, 3, three saliva, which are formed by the use of a copper-azide azide intermediate product disc. St_ev The method described in the tracing book (Rostovtsev et al, (2〇〇2), /like" 41:2596). 42 200804411 Synthesis of compound 7: (3/?, 5 anti-105, 1311,145)-17-((/?)-5-hydroxypentan-2-yl)-10,13-dimethyl-hexadecanthazole-1//-3⁄4 pentyl[beta]non-3 - Same as

化合物7係由石膽酸(化合物3)經反應流程2所示之 兩個步驟來合成。在步驟(a)中,化合物3先以強酸型離子 交換樹脂Amberlite IR120/曱醇在80°C處理12小時,之後 以步驟(b)之氫化鋁鋰(LAH)、四氫呋喃在0°C至室溫下處 理10分鐘,藉以還原其所獲得的甲酸酯類。 反應式2Compound 7 was synthesized from lithocholic acid (Compound 3) by two steps shown in Reaction Scheme 2. In the step (a), the compound 3 is first treated with a strong acid type ion exchange resin Amberlite IR120/nonanol at 80 ° C for 12 hours, followed by the lithium aluminum hydride (LAH) of step (b), tetrahydrofuran at 0 ° C to the chamber. The mixture was treated for 10 minutes under temperature to reduce the formate obtained therefrom. Reaction formula 2

200804411 化合物 8 之合成:(R)-4-((3R,5R,10S,13R,14S)-3-羥基-10,13-二甲基-十六烷氫-lH-環戊烷[a]菲-17-基)戊基二苯基磷酸200804411 Synthesis of compound 8: (R)-4-((3R,5R,10S,13R,14S)-3-hydroxy-10,13-dimethyl-hexadecanehydro-lH-cyclopentane [a] Phenyl-17-yl)pentyldiphenylphosphoric acid

化合物8係根據反應流程3由化合物7來合成。特別 地,在步驟(c)中,化合物7之經基係在疊氮基填酸二苯S旨 存在及室溫下反應10小時轉換為二苯基磷酸官能基。 反應式3Compound 8 was synthesized from Compound 7 according to Reaction Scheme 3. Specifically, in the step (c), the base of the compound 7 is converted into a diphenylphosphoric acid functional group in the presence of an azide-based diphenyl sulfonate at room temperature for 10 hours. Reaction formula 3

ΛΔ 200804411 化合物 9 之合成:2-((R)-4-((3R,5R,10S,13R,14S)-3-·* -10,13-二甲基-十六烷氫-1H-環戊烷[a]菲-17-基)戊醯胺)乙 酸ΛΔ 200804411 Synthesis of compound 9: 2-((R)-4-((3R,5R,10S,13R,14S)-3-·* -10,13-dimethyl-hexadecane-1H-ring Pentane [a]phenanthrene-17-yl)pentanylamine)acetic acid

化合物9 化合物9係由化合物3依據反應式4來合成。執行化 合物3及被保護胺基酸H-Gly-OBut的縮合係在步驟⑷中, 其係在Ν,Ν-二甲基曱醯胺中使用2-(1Η-苯并三唾-1-基)-1,1,3,3-四甲基脲六氟磷酸(《^1!1)/二異丙基乙基胺 (DIPEA)做為偶合劑,在室溫下反應30分鐘得到被保護 的共軛中間體。化合物9係在後續的步驟(f)中透過使用TFA 及DCM在室溫下反應1小時移去tBu官能基後獲得。 45 200804411 反應式4Compound 9 Compound 9 was synthesized from Compound 3 according to Reaction Scheme 4. The condensation of the compound 3 and the protected amino acid H-Gly-OBut is carried out in the step (4), which is based on 2-(1Η-benzotris-l-yl) in hydrazine, hydrazine-dimethyl decylamine. -1,1,3,3-tetramethyluronium hexafluorophosphate ("1!!") / diisopropylethylamine (DIPEA) as a coupling agent, reacted at room temperature for 30 minutes to be protected Conjugated intermediates. Compound 9 was obtained in the subsequent step (f) by reacting TFA and DCM at room temperature for 1 hour to remove the tBu functional group. 45 200804411 Reaction 4

化合物 10 之合成:(S)-2-((R)-4-((3R,5R,10S,13R,14S)-3^^ 基-10,13-二曱基-十六烷氫-1H-環戊烷[a]菲-17-基)戊醯胺) 琥珀酸 〇Synthesis of Compound 10: (S)-2-((R)-4-((3R,5R,10S,13R,14S)-3^^yl-10,13-didecyl-hexadecanehydrol-1H -cyclopentane [a]phenanthroline-17-yl)pentaamine) bismuth succinate

46 200804411 化合物10係由化合物3依據反應流程5來合成。執行 化合物3及被保護胺基酸L-H-Asp(OBut)-OBut的縮合係在 步驟(e)中,其係在N,N-二甲基甲醯胺中使用2-(1Η-苯并三 唑-1-基)-1,1,3,3-四甲基脲六氟磷酸(113丁1;)/二異丙基乙 基胺(DIPEA)做為偶合劑,在室溫下反應30分鐘得到被 保護的共軛中間體。化合物10係在後續的步驟⑴中透過使 用TFA及DCM在室溫下反應1小時移去tBu官能基後獲得。 反應式546 200804411 Compound 10 was synthesized from Compound 3 according to Reaction Scheme 5. The condensation of the compound 3 and the protected amino acid LH-Asp(OBut)-OBut is carried out in step (e) using 2-(1Η-benzotriene) in N,N-dimethylformamide Zin-1-yl)-1,1,3,3-tetramethylurea hexafluorophosphate (113 butyl 1;) / diisopropylethylamine (DIPEA) as a coupling agent, reacting at room temperature 30 The protected conjugated intermediate is obtained in minutes. Compound 10 was obtained in the subsequent step (1) by reacting TFA and DCM at room temperature for 1 hour to remove the tBu functional group. Reaction formula 5

化合物 11 之合成:(R)-4-((5R,10S,13R,14Sl·l0,13·二曱基-3-氧-十六烷氫-lH-環戊烷[a]菲·17_基)戊酸 47 200804411 么 ΟSynthesis of Compound 11: (R)-4-((5R,10S,13R,14Sl·l0,13·didecyl-3-oxo-hexadecanehydro-lH-cyclopentane[a]phenanthrene-17_ Valentate 47 200804411

化合物11係由化合物3依據反應式6來合成。在步驟 ⑴之氧化反應中,化合物3在醋酸中、100°C利用氧化鉻反 應30分鐘生成化合物11。 反應式6Compound 11 was synthesized from Compound 3 according to Reaction Scheme 6. In the oxidation reaction of the step (1), the compound 3 was reacted with acetic acid in acetic acid at 100 ° C for 30 minutes to give a compound 11. Reaction formula 6

200804411 化合物12之合成:(1〇-4-((3尺,5艮103,13艮143)-3-(3-羧基丙 醯基氧基)-10,13-二甲基-十六烷氫-1H-環戊烷[a]菲-17-基) 戊酸 么 〇200804411 Synthesis of compound 12: (1〇-4-((3 ft, 5艮103,13艮143)-3-(3-carboxypropenyloxy)-10,13-dimethyl-hexadecane) Hydrogen-1H-cyclopentane [a]phenanthrene-17-yl) valeric acid

化合物12係由化合物3依據反應流程7來合成。特別 地,在步驟⑴之酯化反應中,化合物3與琥ίό酸酐、DMAP 及吡啶在60°C反應15小時,以製得化合物12。 反應式7Compound 12 was synthesized from Compound 3 according to Reaction Scheme 7. Specifically, in the esterification reaction of the step (1), the compound 3 was reacted with succinic anhydride, DMAP and pyridine at 60 ° C for 15 hours to obtain a compound 12. Reaction 7

200804411 化合物 13 的合成:(R)-4-((3R,5R,10S,13R,14S)-3-((S)-21$ 基-3-羧基丙醯基氧基)-10,13-二曱基-十六烷氳-1H-環戊烷 [a]菲-17-基)戊酸200804411 Synthesis of compound 13: (R)-4-((3R,5R,10S,13R,14S)-3-((S)-21$-yl-3-carboxypropenyloxy)-10,13- Dimercapto-hexadecane-1H-cyclopentane [a]phenanthrene-17-yl)pentanoic acid

化合物13係由化合物3依據反應流程8來合成。為製 備化合物13,在步驟(k)中化合物3的羥基官能基先利用醋 酸酐、吡啶在室溫下反應5小時先轉換為乙醯基,接著在 後續利用第三丁醇、DMAP、DCC、DCM,在室溫下反應 30分鐘進行酯化反應步驟(步驟(I))獲得反應流程8的第 一中間體。步驟(m)的去乙醯基反應係利用甲氧基鈉、甲醇 在室溫下反應2小時’以生成第二中間體’之後猎由二ί辰 己烷基碳二亞胺(DCC)保護之偶合反應,以及後續三步 驟的Fmoc、Boc及tBu官能基的去保護反應: (n)Fmoc-L-ASP(OBut)-OH、DMAP、DCC、DCM 在室溫下 反應30分鐘;(o)DBU、DCM在室溫下反應1小時;及 (h)TFA,2%水在室溫下反應1小時,來製得預期的產物13。 200804411 反應式8Compound 13 was synthesized from Compound 3 according to Reaction Scheme 8. For the preparation of compound 13, the hydroxy function of compound 3 in step (k) is first converted to an acetamyl group by reaction with acetic anhydride or pyridine at room temperature for 5 hours, followed by subsequent use of third butanol, DMAP, DCC, DCM, the esterification reaction step (step (I)) is carried out by reacting at room temperature for 30 minutes to obtain the first intermediate of the reaction scheme 8. The deacetylation reaction of step (m) is carried out by using sodium methoxide and methanol at room temperature for 2 hours to form a second intermediate, which is protected by a butyl hexyl carbodiimide (DCC). Coupling reaction, and deprotection of Fmoc, Boc and tBu functional groups in the next three steps: (n) Fmoc-L-ASP(OBut)-OH, DMAP, DCC, DCM reacted at room temperature for 30 minutes; (o) DBU and DCM were reacted at room temperature for 1 hour; and (h) TFA, 2% water was reacted at room temperature for 1 hour to obtain the desired product 13. 200804411 Reaction 8

ο 化合物14之合成:4-((3尺,5艮103,13尺,145)-17-((1〇-5-第三 丁氧基-5-氧戊烷-2-基)-10,13-二甲基-十六烷氫-1Η-環戊烷 [a]非-3-基氧基)-4-氧丁酸ο Synthesis of compound 14: 4-((3 ft, 5 艮 103, 13 ft, 145)-17-((1〇-5-t-butoxy-5-oxopentan-2-yl)-10 ,13-dimethyl-hexadecanehydro-1Η-cyclopentane [a]non-3-yloxy)-4-oxobutanoic acid

51 200804411 化合物14係由化合物3依據反應流程9來合成。為製 備化合物14,在步驟(k)中化合物3的羥基官能基先利用醋 酸酐、吡啶在室溫下反應5小時先轉換為乙醯基,接著在 後續利用第三丁醇、DMAP、DCC、DCM,在室溫下反應 30分鐘進行酯化反應步驟(步驟⑴)獲得反應流程8的第 一中間體。步驟(m)的去乙醯基反應係利用甲氧基鈉、甲醇 在室溫下反應2小時,以生成第二中間體,之後藉由與琥 珀酸酐、DMAP及吡啶在60°C,15小時下的酯化反應來製 得化合物14。 反應式951 200804411 Compound 14 was synthesized from Compound 3 according to Reaction Scheme 9. To prepare compound 14, the hydroxy function of compound 3 in step (k) is first converted to an oxime group by reaction with acetic anhydride or pyridine at room temperature for 5 hours, followed by subsequent use of third butanol, DMAP, DCC, DCM, reacting at room temperature for 30 minutes to carry out an esterification reaction step (step (1)) to obtain a first intermediate of the reaction scheme 8. The deacetylation reaction of the step (m) is carried out by using sodium methoxide and methanol at room temperature for 2 hours to form a second intermediate, followed by succinic anhydride, DMAP and pyridine at 60 ° C for 15 hours. The esterification reaction is carried out to obtain a compound 14. Reaction formula 9

200804411 化合物 15 之合成:4-((311,511,103,1311,145)-17-((1〇-5-(羧基 甲基胺基)-5-氧戊烷-2-基)-10,13-二曱基-十六烷氫-1H_環戊 烷[a]菲-3-基氧基)-4-氧丁酸200804411 Synthesis of compound 15: 4-((311,511,103,1311,145)-17-((1〇-5-(carboxymethylamino)-5-oxopentan-2-yl)-10 ,13-dimercapto-hexadecanehydro-1H-cyclopentane [a]phenanthr-3-yloxy)-4-oxobutanoic acid

化合物15係依據反應流程10由化合物3來合成。執 行化合物3及被保護胺基酸L-H-Gly-OBut的縮合係在步驟 (e)中,其係在N,N-二曱基曱醯胺中使用2-(1Η-苯并三唑-1-基)-1,1,3,3-四甲基脲六氟磷酸(HBTU) /二異丙基乙基胺 (DIPEA)做為偶合劑,在室溫下反應30分鐘得到被保護 的共扼中間體。中間體係與號珀酸酐、DMAP、吼啶在80°C, 15小時(步驟g)中酯化,接著以TFA、2%水在室溫下反 應1小時(步驟h)獲得化合物15。 200804411 反應式10Compound 15 was synthesized from Compound 3 according to Reaction Scheme 10. The condensation of the compound 3 and the protected amino acid LH-Gly-OBut is carried out in step (e) using 2-(1 - benzotriazole-1) in N,N-didecylguanamine. -yl)-1,1,3,3-tetramethyluronium hexafluorophosphate (HBTU) / diisopropylethylamine (DIPEA) as a coupling agent, reacted at room temperature for 30 minutes to obtain a protected total扼 intermediates. The intermediate system was esterified with phenolic anhydride, DMAP, acridine at 80 ° C for 15 hours (step g), followed by reaction with TFA, 2% water at room temperature for 1 hour (step h) to obtain compound 15. 200804411 Reaction 10

化合物 16 之合成:(S)-2-((R)-4-((3R,5R,10S,13R,14S)-3-(3-羧基丙醯基氧基:M0,13-二甲基-十六烷氫-1H-環戊烷[a]菲 -17-基)戊醯胺)琥珀酸Synthesis of Compound 16: (S)-2-((R)-4-((3R,5R,10S,13R,14S)-3-(3-carboxypropenyloxy: M0,13-dimethyl -hexadecanehydro-1H-cyclopentane [a]phenanthrene-17-yl)pentanylamine succinic acid

54 200804411 化合物16係由化合物3依據反應流程11來合成。執行 化合物3及被保護胺基酸L-H-AspCOButhOBut的縮合係在 步驟(e)中,其係在N,N-二甲基甲醯胺中使用2-(1Η-苯并三 唑-1-基)-l,l,3,3-四曱基脲六氟磷酸(HBTU)/二異丙基乙 基胺(DIPEA)做為偶合劑,在室溫下反應30分鐘得到被 保護的共軛中間體。令間體係與琥珀酸酐、DMAP、吡啶在 80°C,15小時(步驟g)中酯化,接著以TFA、2%水在室 溫下反應1小時(步驟h)獲得化合物16。 反應式1154 200804411 Compound 16 was synthesized from Compound 3 according to Reaction Scheme 11. The condensation of the compound 3 and the protected amino acid LH-AspCOButhOBut is carried out in step (e) using 2-(1Η-benzotriazol-1-yl) in N,N-dimethylformamide -l,l,3,3-tetradecylurea hexafluorophosphate (HBTU) / diisopropylethylamine (DIPEA) as a coupling agent, reacted at room temperature for 30 minutes to obtain a protected conjugate intermediate body. The interstage system was esterified with succinic anhydride, DMAP, pyridine at 80 ° C for 15 hours (step g), followed by reaction with TFA, 2% water at room temperature for 1 hour (step h) to obtain compound 16. Reaction formula 11

200804411 化合物 17 之合成:(R)-4-((3R,5R,10S,13R,14S)-3-((R)-2j$ 基-3-羧基丙醯基氧基)-10,13-二曱基-十六烷氫-1H-環戊烷 [a]菲-17-基)戊酸200804411 Synthesis of compound 17: (R)-4-((3R,5R,10S,13R,14S)-3-((R)-2j$yl-3-carboxypropenyloxy)-10,13- Dimercapto-hexadecanehydrol-1H-cyclopentane [a]phenanthrene-17-yl)pentanoic acid

化合物17係由化合物3依據反應流程12來合成。為 製備化合物17,在步驟(k)中化合物3的羥基官能基先利用 醋酸酐、吡啶在室溫下反應5小時先轉換為乙醯基,接著 在後續利用第三丁醇、DMAP、DCC、DCM,在室溫下反 應30分鐘進行酯化反應步驟(步驟⑴)獲得反應流程8的 第一中間體。步驟(m)的去乙醯基反應係利用甲氧基鈉、甲 醇在室溫下反應2小時,以生成第二中間體,之後藉由二 環己烷基碳二亞胺(DCC)保護之偶合反應,以及後續三 步驟的Fmoc、Boc及tBu官能基的去保護反應: (lOFmoc-L-ASPCOButO-OH、DMAP、DCC、DCM 在室溫下 反應30分鐘;(〇)DBU、DCM在室溫下反應1小時;及 (h)TFA,2%水在室溫下反應1小時,來製得預期的產物16。 200804411 反應式12Compound 17 was synthesized from Compound 3 according to Reaction Scheme 12. For the preparation of compound 17, the hydroxy function of compound 3 in step (k) is first converted to an acetamyl group by reaction with acetic anhydride or pyridine at room temperature for 5 hours, followed by subsequent use of third butanol, DMAP, DCC, DCM, reacting at room temperature for 30 minutes to carry out an esterification reaction step (step (1)) to obtain a first intermediate of the reaction scheme 8. The deacetylation reaction of the step (m) is carried out by using sodium methoxide and methanol at room temperature for 2 hours to form a second intermediate, which is then protected by dicyclohexylcarbodiimide (DCC). Coupling reaction, and deprotection of Fmoc, Boc and tBu functional groups in the next three steps: (lOFmoc-L-ASPCOButO-OH, DMAP, DCC, DCM reaction at room temperature for 30 minutes; (〇) DBU, DCM in the chamber The reaction was carried out for 1 hour at a temperature; and (h) TFA, 2% water was reacted at room temperature for 1 hour to obtain the intended product 16. 200804411

ΗΗ

.OH Ο 200804411 化合物 18 之合成:(3R,5R,10S,13R,14S)-17-((R)-5-(4-(2·· 基乙基)-1Η-(1,2,3-三唑-1-基)戊-2-基)-10,13-二曱基-十六烷 氫-1H-環戊烷[a]菲-3-酮.OH Ο 200804411 Synthesis of Compound 18: (3R, 5R, 10S, 13R, 14S)-17-((R)-5-(4-(2··ylethyl)-1Η-(1,2,3 -triazol-1-yl)pentan-2-yl)-10,13-dimercapto-hexadecanehydro-1H-cyclopentane [a]phenanthrene-3-one

化合物18係由化合物8依據反應式13來合成。轉換 化合物8成為對應之疊氮中間體係可透過步驟(d)達成,其 係於過量疊氮化鈉、一足以催化之劑量之鐵化四乙基敍、 及15-冠-5-醚在110°C下反應15小時,而製備1,4-雙取代 -1,2,3-三唑化合物18係可透過步驟(q),其係利用速配接合 組合化學(click chemistry)將亞銅催化連結之疊氮中間體 與適當的炔類反應來獲得。 200804411 反應式13Compound 18 was synthesized from Compound 8 according to Reaction Scheme 13. Conversion of compound 8 to the corresponding azide intermediate system can be achieved by step (d), which is carried out in excess of sodium azide, a sufficiently catalyzed dose of ferric tetraethyl sulphate, and 15-crown-5-ether at 110 The reaction was carried out at ° C for 15 hours, and the preparation of the 1,4-disubstituted-1,2,3-triazole compound 18 was carried out through the step (q), which catalyzed the coupling of the cuprous copper by the use of click chemistry. The azide intermediate is obtained by reacting with an appropriate alkyne. 200804411 Reaction formula 13

化合物 19 之合成 : 2- 胺基 -3-(1-((11)-4-((3尺,511,103,1311,145)-3-羥基-10,13-二曱基-十 六烷氫-1H-環戊烷[a]菲-17-基)戊基)-1Η-1,2,3-三唑-4-基)丙 酸Synthesis of Compound 19: 2-Amino-3-(1-((11)-4-((3 ft, 511,103,1311,145)-3-hydroxy-10,13-didecyl-16 Alkanohydro-1H-cyclopentane [a]phenanthrene-17-yl)pentyl)-1Η-1,2,3-triazol-4-yl)propionic acid

200804411 化合物19係由化合物8根據反應式14所合成。在步 驟(d)當中,於過量疊氮化鈉(s〇diumazideNaN3)、催化 劑蛾化四丁|安(tetrabutylammoniumiodide ; TBAI)、15 — 冠一5醚(15 — Crown-5)存在下,在110°C將化合物8轉 換為相對應的疊氮化合物中間產物15小時。於步驟(q)當 中’利用銅(I)催化疊氮化合物中間產物與適當的炔烴接合 為— 1,4二取代1,2,3三唑中間產物。步驟(h)中,第三-丁基氧幾基(tert-butoxy carbonyl group,Boc)於 TFA 2%水 中於室溫進行一小時去保護產生外消旋衍生物19。 200804411 反應式14200804411 Compound 19 was synthesized from Compound 8 according to Reaction Scheme 14. In step (d), in the presence of excess sodium azide (s〇diumazide NaN3), catalyst tetrabutylammonium iodide (TBAI), 15-crown-5 ether (15 - Crown-5), at 110 Compound 8 was converted to the corresponding azide intermediate for 15 hours at °C. In step (q), the copper (I) catalyzed azide intermediate is bonded to the appropriate alkyne to a 1,4 disubstituted 1,2,3 triazole intermediate. In step (h), the tert-butoxy carbonyl group (Boc) is deprotected in TFA 2% water at room temperature for one hour to give the racemic derivative 19. 200804411 Reaction 14

Step h 50% HO、、、' 200804411 化合物 20 之合成 :2-(二乙氧基磷醯 基)各(1-((尺)-4-((3艮5反1(^,13艮145)-3-羥基-10,13-二曱基-十六烷 氳-1H-環戊烷[a]菲-17-基)戊基-1H-1,2,3-三唑-4-基)丙酸Step h 50% HO,,, ' 200804411 Synthesis of compound 20: 2-(diethoxyphosphonium) each (1-(())-4-((3艮5反1(^,13艮145) )-3-hydroxy-10,13-dimercapto-hexadecane-1H-cyclopentane [a]phenanthrene-17-yl)pentyl-1H-1,2,3-triazol-4-yl Propionic acid

化合物20係由化合物8根據反應式15所合成。在步 驟(d)當中,於過量疊氮化納(sodium azide ; NaN3)、催 化劑破化四丁銨(tetrabutylammoniumiodide ; TBAI)、15 —冠一5醚(15 — Crown-5)存在下,在110 QC將化合物8 轉換為相對應的疊氮化合物中間產物15小時。於步驟(q) 當中,利用銅(I)催化疊氮化合物中間產物與適當的炔烴接 合成為一 1,4二取代1,2,3三唑中間產物。步驟(r)中,於 NaOH,EtOH/H20 (1:1)以及室溫當中進行皂化5小時形成一 外消旋衍生物20。 200804411 反應式15Compound 20 was synthesized from Compound 8 according to Reaction Scheme 15. In step (d), in the presence of excess sodium azide (NaN3), catalyst tetrabutylammonium iodide (TBAI), 15-crown-5 ether (15 - Crown-5), at 110 QC converted compound 8 to the corresponding azide intermediate for 15 hours. In step (q), the copper (I) catalyzed azide intermediate is combined with the appropriate alkyne to form a 1,4 disubstituted 1,2,3 triazole intermediate. In the step (r), saponification is carried out for 5 hours at NaOH, EtOH/H20 (1:1) and at room temperature to form a racemic derivative 20. 200804411 Reaction 15

HO、、、、HO,,,,

HH

Step q 55%Step q 55%

Step r 30% HO、、, 200804411 化合物 21 之合成:3-(l-((R)-4-((3R,5R,10S,13R,14S)-3-· 基-10,13-二甲基-十六烷氫-1H-環戊烷[a]菲-17-基)戊 基)-1Η-1,2,3-三唑_4_基)丙酸Step r 30% HO,,, 200804411 Synthesis of compound 21: 3-(l-((R)-4-((3R,5R,10S,13R,14S)-3-)yl-10,13-dimethyl -hexadecanehydro-1H-cyclopentane [a]phenanthrene-17-yl)pentyl)-1Η-1,2,3-triazole-4-yl)propionic acid

化合物21係由化合物8根據反應式16所合成。在步 驟(d)當中,於過量疊氮化鈉(sodium azide ; NaN3)、催化 劑蛾化四丁銨(tetrabutylammonium iodide ; TBAI)、15 — 冠一 5醚(15 — Crown-5 )存在下,在110 °C將化合物8轉 換為相對應的疊氮化合物中間產物15小時。於步驟(q)當 中,利用銅(I)催化疊氮化合物中間產物與適當的炔烴接合 成為一 1,4二取代1,2,3三唑化合物21。 200804411 反應式16Compound 21 was synthesized from Compound 8 according to Reaction Scheme 16. In step (d), in the presence of excess sodium azide (NaN3), tetrabutylammonium iodide (TBAI), 15 - crown-5 ether (15 - Crown-5), Compound 8 was converted to the corresponding azide intermediate for 15 hours at 110 °C. In the step (q), the copper (I)-catalyzed azide compound intermediate is bonded to a suitable alkyne to form a 1,4 disubstituted 1,2,3 triazole compound 21. 200804411 Reaction 16

化合物 22 之合成:4-(l-((R)-4-((3R,5R,10S,13R,14S)-3^|;& -10,13-二甲基-十六烷氫-1H-環戊烷[a]菲-17-基)戊 基)_1H-1,2,3-三唑-4-基)丁酸 HO〆Synthesis of Compound 22: 4-(l-((R)-4-((3R,5R,10S,13R,14S)-3^|;& -10,13-Dimethyl-hexadecane Hydrogen- 1H-cyclopentane [a]phenanthroline-17-yl)pentyl)_1H-1,2,3-triazol-4-yl)butyric acid HO〆

化合物22 200804411 化合物22係由化合物8根據反應式17所合成。在步 驟(d)當中,於過量疊氮化鈉(sodium azide ; NaN3)、催化 劑蛾化四丁銨(tetrabutylammonium iodide ; TBAI)、15 — 冠一5醚(15 — Crown-5)存在下,在110 °C將化合物8轉 換為相對應的疊氮化合物中間產物15小時。於步驟(q)當 中,利用銅(I)催化疊氮化合物中間產物與適當的炔烴接合 成為一 1,4二取代1,2,3三唑化合物22。 反應式17Compound 22 200804411 Compound 22 was synthesized from Compound 8 according to Reaction Scheme 17. In step (d), in the presence of excess sodium azide (NaN3), tetrabutylammonium iodide (TBAI), 15 - crown-5 ether (15 - Crown-5), Compound 8 was converted to the corresponding azide intermediate for 15 hours at 110 °C. In step (q), the copper (I) catalyzed azide intermediate is bonded to a suitable alkyne to form a 1,4 disubstituted 1,2,3 triazole compound 22. Reaction formula 17

200804411 每一化合物以a-2,3-ST進行抑制測試,使用 CMP-Neu5Ac作為受質以及一修飾過的雙醣和一 4,5-二甲 氧基2-硝基溴基團作為UV-標識受體。特別是,抑制試驗 係以三重複於含有200 mM MES缓衝液、100 mM氯化鈉、 0.5 mM 乙二胺四乙基二鈉、〇·〇1% Triton X-100、20 μΜ 修 飾過的雙醣、1·4 mU a-2,3-ST以及抑制劑(於DMS0 (dimethyl sulfoxide)二曱基石黃酸)總體積為55μΐ的反應液 中進行。反應係於37°C進行10分鐘後加入CMP-Neu5Ac(最 終濃度3 μΜ))。接著溶液於37°C反應直到產生一可偵測量 之化合物產生。反應以100°C加熱混和液分鐘終止反應。酵 素活性以 RP-HPLC (Supelco Discovery® HS C18,5 μιη,4.6 mm x 25 cm)|i控唾液酸化雙醣產物於348nm處,滯留時間 (retention time)為 20.38 分鐘。 曰一上述16種合成類似物相較於石膽酸(Hth〇ch〇lic此⑷ 顯不一降低的IQo (表一)。最終乙醇7與其衍生物8相較 於石膽酸降低5·17倍,細擅酸基對於促進親和性有其重 ,進一步以胜肽偶合延長末端的叛酸進行確認,化 i抑制特性相較於化合物7-8可被完全回復。 取代氫氧基的化合物虱== 生,我們以具有-酮基以 果發現化合物,抑制a-2,3-ST的活性。實驗結 七倍的潛力。更、隹_ 50為139μΜ,’相較於石膽酸具有低 示高於兩倍的潛力:步石於石膽酸化合物12與13顯 附親和性作用而Α ^不s酸的3_氫氧基的建構可隨著吸 物14-16似乎遠i。丨正。虽化合物12末端羧酸變化時,類似 一)。令人驚微莫耳親和平原現象(Pla_)(表 則相較於化合物^ 合物17 ’將D-AsP取代為L-Asp, 3有純兩倍的改善。此外,石膽酸進- 200804411 步將羧酸取代為1,4二取代1,2,3三唑配位基。化合物20與 21相較於化合物19有一增加8-12倍的效果,推測加入一 正電胺基酸可以破壞其親和力。 原始乙醇18低於化合物21至少10倍,低於化合物22 至少20倍,與先前觀察末端羧酸吸附的值一致。類似物22 是活性最大的化合物具有一 IC5〇為5 μΜ。 表一200804411 Each compound was tested for inhibition by a-2,3-ST using CMP-Neu5Ac as a substrate and a modified disaccharide and a 4,5-dimethoxy 2-nitrobromo group as UV- Identify the receptor. In particular, the inhibition assay was performed in triplicate with a double modified with 200 mM MES buffer, 100 mM sodium chloride, 0.5 mM ethylenediamine tetraethyl disodium, 〇·〇1% Triton X-100, 20 μΜ. The sugar, 1·4 mU a-2,3-ST and an inhibitor (in DMS0 (dimethyl sulfoxide) dimercaptolithic acid) were reacted in a total volume of 55 μM. The reaction was carried out at 37 ° C for 10 minutes and then CMP-Neu5Ac (final concentration 3 μΜ) was added. The solution is then reacted at 37 ° C until a detectable amount of compound is produced. The reaction was terminated by heating the mixture at 100 ° C for a few minutes. The enzyme activity was controlled by RP-HPLC (Supelco Discovery® HS C18, 5 μιη, 4.6 mm x 25 cm)|i sialylation of the disaccharide product at 348 nm with a retention time of 20.38 minutes. 16The above 16 synthetic analogues compared to choline acid (Hth〇ch〇lic this (4) significantly reduced IQo (Table 1). Finally, ethanol 7 and its derivative 8 are lower than lithocholic acid 5.17 In addition, the fine acid group has a heavier effect on promoting affinity, and further confirms that the peptide is extended by the end of the peptide, and the inhibitory property is completely restored compared to the compound 7-8. == Raw, we found the compound with a ketone group to inhibit the activity of a-2,3-ST. The experiment has a potential of seven times. More, 隹 _ 50 is 139 μΜ, which is lower than lithocholic acid. Shows a potential of more than twice: the step stone has a affinitive affinity for the choline acid compounds 12 and 13 and the construction of the 3 _ hydroxy group which does not s acid can appear as far as the sorbent 14-16. Although the carboxylic acid at the end of compound 12 changes, it resembles a). Amazing monaural and plain phenomenon (Pla_) (the table replaces D-AsP with L-Asp compared to compound 17', 3 has a pure improvement of 2. In addition, lithocholic acid enters - 200804411 The step of replacing the carboxylic acid with a 1,4 disubstituted 1,2,3 triazole ligand. Compounds 20 and 21 have an 8-12-fold increase in effect compared to compound 19, presumably adding a positively charged amino acid can destroy The affinity of the original ethanol 18 is at least 10 times lower than that of the compound 21 and at least 20 times lower than the compound 22, which is consistent with the value of the adsorption of the terminal carboxylic acid previously observed. The analog 22 is the most active compound having an IC5 〇 of 5 μΜ. One

200804411200804411

200804411200804411

70 20080441170 200804411

抑制常數(Ki)以最大活性的化合物22來分析。抑制 測試反應溶液之製備如上述,在加入0.3, 0.15,與0.037 mMCMP-Neu5Ac 於現有的抑制劑 5,4·5,3·5,2·5,及 ΟμΜ 之後反應10分鐘。試驗中每一受質與抑制劑進行二重複。 在IC5〇試驗條件下得到一速率以及每一抑制劑的濃度,速 率的倒數與受質濃度的倒數呈現線性關係。Ki絕對值係由 速度為垂直軸而抑制劑濃度在水平轴的雙倒數關係圖 (Lineweaver-Burk plot)的水平交點而來(第一圖)。化合物 200804411 22 對 CMP-Neu5A 競爭抑制。 c 有一 Ki 為 2.2 μΜ, 顯示其作用為非 Ο) 卜細胞毒性及附著性分析。在 穷二、=T ’將CL1-5細胞以每60mm、1χ1〇6個細胞的 山又、,接種於含10%胎牛血清(FBS)的DMEM培養基,隔 2以不含胎牛血清、含DMS0及5 _ 4 10 _抑制 劑的培養基取代。控制組的細胞則以含01% DMS0、無抑 制劑的培養基培養,48小時後,再以錐藍排除法(trypan blue exclusion)決定其其細胞數目及細胞存活率。分別以三種抑 制劑·· N-Pemyonic (化合物 21)、Lith_Triethyl de、(化合物 20)及Lith+Asp (化合物13)處理後,相較於控制組,CL1-5 細胞的生長受到10-20%的抑制(如第二a圖所示)。 除了對細胞存活率的影響,以a-2,3-ST抑制劑處理過 後,可明顯增加CL1-5細胞對於細胞外基質膠原蛋白j (extracellular matrix protein collagen I)的附著力。96 孔的 微孔板上先塗覆膠原蛋白typel (100 pg/ml,40 μΐ/孔),再於 4C下加入5% +血清蛋白(BSA)。每次試驗取5χ104個細 胞,以不同劑量的a_2,3-ST抑制劑或DMS0處理過後,接 種於先前已經過塗覆的微孔中;在5% C02、37°C的條件下, 培養四小時。之後,以不含血清的DMEM培養基洗提兩次 細胞,除去為附著的細胞;並於4°C下,以1%的曱醛進行 200804411 十分鐘的固疋。固定後的細胞以結晶紫染色二十五分鐘後, 以1% SDS溶液進行細胞溶解。附著的細胞數目以59〇nm 吸光度的相關性加以決定。以a_2,3_ST抑制劑處理過後, CL1-5細胞對於膠原蛋白I的附著增加了 L2-15倍(如第 二Β圖所示)。 實施例三CL1-5肺癌細胞遷移的抑制 為評估a-2,3-ST對於抑制細胞遷移的影響,使用間隙 癒合(wound healing assay)分析。在5% C〇2的條件下, 高度轉移性CL1-5肺癌細胞在含1〇%胎牛血清(FBS)的 DMEM 培養基中以 1〇 μ]ν[的 a-2,3-ST 抑制劑-Lith-o-Asp 進行前處理。將部分細胞從培養板刮下,形成一個「間隙」, 在a-2,3-ST抑制劑持續存在的情況下,觀察細胞遷移至「間 隙」處達48小時的距離(如第三圖所示)。 實施例四:a-2,3-ST減/>肺痛細胞上a-2,3唾液酸化抗原在細腧表 面表現的抑制 為了確定a-2,3-ST抑制作用會改變細胞表面的醣化, 以Lith-o-Asp處理肺癌細胞,並以流式細胞儀偵測a_2,3-唾液酸化細胞表面抗原。收集lxlO6個肺癌細胞,以漢克斯 平衡鹽類溶液(HBSS)洗提兩次後,將細胞以a-2,3-ST抑制 劑進行五天的處理。在37°C下,將收集來的細胞以 FITC-conjugatedMAL(l:50)培養 15-20 分鐘;未結合的 MAL 再以HBSS洗提時即被移除’ FACScalibur instrument (Becton Dickinson)共測得3 x 1〇4個結合細胞。取低轉移潛 能的CL1-1及高轉移潛能的CL1-5肺癌細胞株進行測試, 由於 CL1-5 係利用 matrix gel invasion assay,由 CL1-1 衍生 200804411 而來(Chen et al·,Cancer Res·, 2001,61:5223),這些細胞 株都有相同的遺傳背景。以Utlw>Asp處理之CL1^細胞 比以5 μΜ、20 μΜ DMSO處理的控制組細胞,其.2,3_唾 液酸化抗原的表現約減少15%(如第四Α圖所示)。相較 之下,Lith-o-Asp對於高轉移性的肺癌細胞中,心2,3一 唾液酸化細胞表面抗原的抑制,與劑量的關聯則高於15% (如第四B圖所示)。 _f_施例五:在不同細胞蜂的表現及之德a_2,3_ST的抑制 為了評估a-2,3-ST抑制作用對内源性基因表現的影 響,以半定量逆轉錄酶(RT)-PCR測定各種具有不同轉移潛 能之細胞株的/ mRNA表現。細胞株的分析包括: 正常的肺上皮BES-6細胞、低轉移性CL1-1細胞及H1299 肺癌細胞、高轉移性CL1-5、CL1-5 F4及A549肺癌細 胞。將細胞以a-2,3-ST抑制劑培養48小時,收集後以TRIzol (Invitrogen)、盼及氯仿(phenol/chloroform)進行 RNA 萃 取。完成逆轉錄酶(RT)反應後,使用5T3GW/-特異性引子 (ST3Gal /-specific primers ) - 川貝 向 : 5? -GGACCCTGAAAGTGCTCA-35 及反向 : 5’-TCTCCAGCATAGGGTCCA-3,進行互補 DNA 的擴增。 PCR條件如下··在95Qc下變性、61°C下黏合及72°C下聚 合’共35個循環。mRNA表現的相對量則與反應的内在標 準(β·肌動蛋白)值相比較來決定。SrJGW/基因的PCR產物 與肌動蛋白序列混合後,加入〇·5χ TBE、以120V的電 置’利用1.5%溴化乙錠染色凝膠(ethidium bromide stained gel)進行3〇分鐘的電泳。在正常細胞中,並不能測得 7基因的表現,但相對於低轉移潛能細胞,基因於 74 200804411 高轉移潛能細胞中的表現則明顯增加(如第五A圖所示)。 以a-2,3-ST抑制劑Lith-o-Asp處理後,也不會明顯降低 CL1-5肺癌細胞中基因mRNA的表現。 實施例六·· a-2,3-ST ALTERS PROTEIN在CL1-5肺癌細胞中表現 的抑制 為了確定a-2,3-ST抑制作用對CL1-5肺癌細胞中的整 體蛋白質表現的影響,使用一個對於磷酸蛋白具有特異性 的染色系統來進行二維電泳。每次實驗時,先收集1 X 1〇7 個CL1-5肺癌細胞,以1〇 μΜ的Lith-o-Asp或DMSO處理 四十八小時,再以含有8%尿素(Boehringer Mannheim, Germany)、2M 硫脲(Aldrich,WI,USA)及 4% CHAPS (J· Τ· Baker,NJ,USA)的溶解緩衝液(lysis buffer)來溶解此細胞 沈澱物。經超音波(sonication)破碎細胞後,將總重1 mg 的蛋白載入pH 3 -10、長度18公分的固相pH梯度電泳膠片 (immobilized pH gradient (IPG) gel strips, Amersham Pharmacia Biotech,Uppsala,Sweden)上;前述電泳膠片在使 用前需先置於7M尿素、2M硫脲、4% CHAPS、40 mM三 羥甲基氨基曱烷、2% IPG雙性電解質、65 mM DTE及 0.0002%演紛藍中水化(rehydrated)。進行第一次的二維分 離時’係在20。(1;時,以7000 V、總量65kVh的電量,利用 IPGphor system (Amersham Pharmacia Biotech)進行等焦電The inhibition constant (Ki) was analyzed with the most active compound 22. Inhibition The test reaction solution was prepared as described above by adding 0.3, 0.15, and 0.037 mM CMP-Neu5Ac to the existing inhibitors 5, 4·5, 3·5, 2.5, and ΟμΜ for 10 minutes. Each receptor and the inhibitor were subjected to two repetitions in the test. A rate and the concentration of each inhibitor were obtained under the IC5(R) test conditions, and the reciprocal of the rate was linearly related to the reciprocal of the substrate concentration. The Ki absolute value is derived from the horizontal intersection of the velocity as the vertical axis and the inhibitor concentration in the horizontal axis of the Lineweaver-Burk plot (first image). Compound 200804411 22 Competition inhibition of CMP-Neu5A. c has a Ki of 2.2 μΜ, showing its role as non-Ο) cytotoxicity and adhesion analysis. In the poor two, = T ', CL1-5 cells were inoculated in DMEM medium containing 10% fetal bovine serum (FBS) at a rate of 60 mm, 1χ1〇6 cells, separated by 2 to contain fetal bovine serum. Replaced with medium containing DMS0 and 5 _ 4 10 _ inhibitor. The cells in the control group were cultured in a medium containing 01% DMS0 and no inhibitor. After 48 hours, the cell number and cell survival rate were determined by trypan blue exclusion. After treatment with three inhibitors, N-Pemyonic (Compound 21), Lith_Triethyl de, (Compound 20) and Lith+Asp (Compound 13), the growth of CL1-5 cells was 10-20% compared to the control group. Suppression (as shown in Figure 2a). In addition to the effect on cell viability, treatment with a-2,3-ST inhibitor significantly increased the adhesion of CL1-5 cells to extracellular matrix protein collagen I. The 96-well microplate was first coated with collagen typel (100 pg/ml, 40 μΐ/well), and then 5% + serum protein (BSA) was added at 4C. 5χ104 cells per experiment, treated with different doses of a_2,3-ST inhibitor or DMS0, inoculated into previously coated microwells; cultured at 5% C02, 37 °C hour. Thereafter, the cells were eluted twice in serum-free DMEM medium to remove adherent cells; and solidified at 200804411 for 10 minutes at 1 °C with 1% furfural. After the fixed cells were stained with crystal violet for twenty-five minutes, the cells were lysed with a 1% SDS solution. The number of attached cells is determined by the correlation of absorbance at 59 〇 nm. After treatment with the a_2,3_ST inhibitor, the adhesion of CL1-5 cells to collagen I increased L2-15 times (as shown in the second panel). Example 3 Inhibition of migration of CL1-5 lung cancer cells To evaluate the effect of a-2, 3-ST on inhibition of cell migration, a wound healing assay was used. Highly metastatic CL1-5 lung cancer cells in a DMEM medium containing 1% fetal bovine serum (FBS) at 1 5% C〇2 with 1〇μ]ν[a-2,3-ST inhibitor -Lith-o-Asp for pre-processing. Scratch some of the cells from the plate to form a "gap". Observe the migration of the cells to the "gap" for 48 hours in the presence of a-2,3-ST inhibitor (as shown in Figure 3). Show). Example 4: a-2,3-ST minus/> Inhibition of a-2,3 sialylation antigen on the surface of fine sputum on lung pain cells In order to determine the inhibition of a-2, 3-ST, the cell surface is altered. Glycation, lung cancer cells were treated with Lith-o-Asp, and a2,3-sialylated cell surface antigen was detected by flow cytometry. After lxlO6 lung cancer cells were collected and eluted twice with Hanks Balanced Salt Solution (HBSS), the cells were treated with a-2,3-ST inhibitor for five days. The collected cells were cultured at FITC-conjugatedMAL (1:50) for 15-20 minutes at 37 °C; unbound MAL was removed by elution with HBSS' FACScalibur instrument (Becton Dickinson) 3 x 1 〇 4 binding cells. CL1-1 lung cancer cell lines with low metastatic potential and high metastatic potential were tested, and CL1-5 was derived from CL1-1 using the macro gel invasion assay (Chen et al., Cancer Res·). , 2001, 61: 5223), these cell lines all have the same genetic background. The CL1 cells treated with Utlw>Asp showed a 15% reduction in the performance of the .2,3_sialylated antigen compared to the control group treated with 5 μΜ, 20 μΜ DMSO (as shown in the fourth panel). In contrast, Lith-o-Asp inhibits the surface 2,3-sialylated cell surface antigen in high-metastatic lung cancer cells, and the dose-related association is higher than 15% (as shown in Figure 4B). . _f_Example 5: Inhibition of different cell bees and their inhibition of a_2,3_ST In order to assess the effect of a-2,3-ST inhibition on endogenous gene expression, semi-quantitative reverse transcriptase (RT)- The mRNA expression of various cell lines with different metastatic potentials was determined by PCR. Analysis of the cell lines included: normal lung epithelial BES-6 cells, low metastatic CL1-1 cells and H1299 lung cancer cells, high metastatic CL1-5, CL1-5 F4 and A549 lung cancer cells. The cells were cultured for 48 hours with a-2,3-ST inhibitor, collected and extracted by TRIzol (Invitrogen), chloroform (chloroform). After completing the reverse transcriptase (RT) reaction, use 5T3GW/-specific primers (ST3Gal /-specific primers) - Chuanbei: 5? -GGACCCTGAAAGTGCTCA-35 and reverse: 5'-TCTCCAGCATAGGGTCCA-3 for complementary DNA Amplification. The PCR conditions were as follows: · Denaturation at 95Qc, adhesion at 61 ° C, and polymerization at 72 ° C for a total of 35 cycles. The relative amount of mRNA expression is determined by comparison with the intrinsic standard (β·actin) value of the reaction. The PCR product of the SrJGW/gene was mixed with the actin sequence, and then 〇·5χ TBE was added, and electrophoresis was carried out for 3 minutes using a 1.5% ethidium bromide stained gel at 120 V. In normal cells, the expression of the 7 gene was not measured, but compared with the low metastatic potential cell, the gene showed a significant increase in the high metastatic potential cells in 74 200804411 (as shown in Figure 5A). Treatment with the a-2,3-ST inhibitor Lith-o-Asp did not significantly reduce the expression of gene mRNA in CL1-5 lung cancer cells. Example 6 · a-2,3-ST ALTERS PROTEIN inhibition in CL1-5 lung cancer cells To determine the effect of a-2,3-ST inhibition on overall protein performance in CL1-5 lung cancer cells, use A staining system specific for phosphoproteins for two-dimensional electrophoresis. In each experiment, 1 X 1〇7 CL1-5 lung cancer cells were collected and treated with 1 μμ of Lith-o-Asp or DMSO for 48 hours, followed by 8% urea (Boehringer Mannheim, Germany), 2M sulfur. A lysis buffer of urea (Aldrich, WI, USA) and 4% CHAPS (J. Baker, NJ, USA) was used to dissolve the cell pellet. After disrupting the cells by sonication, a total weight of 1 mg of protein was loaded into an immobilized pH gradient (IPG) gel strips, Amersham Pharmacia Biotech, Uppsala, pH 3 -10, 18 cm in length. Sweden); the aforementioned electrophoresis film should be placed in 7M urea, 2M thiourea, 4% CHAPS, 40 mM trishydroxymethylamino decane, 2% IPG ampholyte, 65 mM DTE and 0.0002% before use. Blue hydrated (rehydrated). When the first two-dimensional separation is performed, it is at 20. (1; at a time of 7000 V, a total of 65 kVh, using the IPGphor system (Amersham Pharmacia Biotech) for isoelectric power

泳。完成等焦電泳後,IPG膠片先置於平衡溶液(5〇 mMswimming. After the isocratic electrophoresis is completed, the IPG film is first placed in an equilibrium solution (5 mM

Tris-HCl, pH 8·8, 6 Μ 尿素、2% SDS、30%甘油、2% DTE ) 中十五分鐘進行平衡,將IPG膠片附著於垂直的12·5%線 性梯度聚丙烯醯胺凝膠電泳膠片上,再以〇·5%壤脂固定 IPG膠片。進行第二次二維電泳時,每塊凝膠係以45mA的 75 200804411 電量跑五小時電泳,直到溴酚藍到達凝膠底部。以Pro-Q二 維石粦蛋白膠片染色法(Pro-Q Diamond PhosphoProtein Gel Stain ,Molecular Probes,OR,USA)染色三小時,以 10% 曱醇與7 %醋酸的混合容易中固定三十分鐘,之後浸泡在去 離子水中二十分鐘,以便去除剩餘的染劑。 經Lith-o-Asp處理後的CL1-5肺癌細胞,在二維凝膠 上會出現四個強度增加的點潰,以及四十九個強度減少的 點潰(如第六圖所示)。利用胰蛋白酶分解膠體内(in_gel) 蛋白質並以串聯式質譜(MSMS)確定其強度,與蛋白質資料 庫比對後鑑定出蛋白質身份。其中,數種涉及訊號及損傷 反應路徑(signaling and damage response pathway)的蛋白 已經被鑑別出來,包括:熱休克蛋白75、填酸甘油酸激酶 1、Ran-GTP 水解酶活化蛋白!( Ran_GTPase activating protein 1 )及含蛋白 1 之 WD 重複片段(WD-repeat c〇ntaining protein 1 ) ° 雖然本發明已將較佳實施例揭露如上,然其並非用以 限定本發明’任何熟悉此技藝者,在不脫離本發明之精神 和範圍内,當可作各種之更動與潤飾。因此,其他實施例 亦包含於以下的申請專利範圍中。 76 200804411 【圖式簡單說明】 第一圖係顯示老鼠的α-2,3唾液酸轉移酶對合成性抑制 劑22的抑制分析結果所形成之Lineweaver-Burk雙倒數關 係圖。 苐二圖(A)為三種α-2,3唾液酸轉移酶抑制劑-N-Pentyonic (化合物 21)、Lith-Triethyl-de (化合物 20)及 Lith-o-Asp (化合物13)分別在以5 μΜ、10 μΜ抑制劑處理 過後48小時之CL1-5細胞内的細胞毒性。存活分數則計算 以DMSO處理過之控制組細胞的存活量表示。第二圖(Β)係 顯示相對於附著於經DMS0處理後之控制組細胞之基本水 平(專於1 ) ’經α-2,3唾液酸轉移酶抑制劑處理後之CL1-5 細胞附著於第一型膠原蛋白之情形。 第三圖係顯示以(A)DMS〇、(Β) 1〇 μΜ Uth 〇 Asp治療 C^L1:5細胞48小時後的間隙癒合分析結果。α_2,3唾液酸轉 私酶抑制劑Lith-0-Asp會抑制細胞因從細胞培養孤中刮除 細胞所致損傷部位的細胞遷移作用。 第四圖係顯不細胞表面之α_2,3唾液酸化抗原在低 =性:U_1細胞及(Β)高轉移性CL1·5細胞的表現量。 ^係^不流式細胞儀顯示之結果:P1係在背景強度下之 疚^ 2及p 3_Λ別為在高強度下表現唾液酸之細胞並從可 ί戶Uit計算出各種細胞的百分比。下圖係顯示依實 ^所件之平均螢光強度值所製成之柱狀圖。 肌動t 在不同轉移潛能肺細胞内的 a的内源性mRNA的表現量。(B)則顯示 200804411 在以DMSO或Lith-o-Asp處理過的CL1-5細胞内,ST3Gal I基因的mRNA表現量未明顯改變。 第六圖係顯示(A)以DMSO控制組及(B) Lith-o-Asp處 理過的CL1-5肺細胞之二維電泳圖。IEF為pH 3-10,分子 量為21.0-97.0 kDa。差異表達蛋白則進一步以質譜及串連 質譜加以鑑別。 【主要元件符號說明】 無 78Tris-HCl, pH 8·8, 6 尿素 urea, 2% SDS, 30% glycerol, 2% DTE) equilibrated in fifteen minutes, attaching IPG film to vertical 12·5% linear gradient polypropylene amide condensation On the gel electrophoresis film, IPG film was fixed with 〇·5% soil grease. For the second two-dimensional electrophoresis, each gel was run for five hours at 45 mA of 75 200804411 until the bromophenol blue reached the bottom of the gel. It was stained with Pro-Q Diamond PhosphoProtein Gel Stain (Molecular Probes, OR, USA) for three hours, and easily fixed for 30 minutes with a mixture of 10% sterol and 7% acetic acid. It was then immersed in deionized water for twenty minutes to remove the remaining dye. After treatment with Lith-o-Asp, CL1-5 lung cancer cells showed four points of increased intensity on the two-dimensional gel and forty-nine points with reduced intensity (as shown in Figure 6). The in-gel protein was decomposed by trypsin and its intensity was determined by tandem mass spectrometry (MSMS), and the protein identity was identified by comparison with the protein database. Among them, several proteins involved in the signal and damage response pathway have been identified, including: heat shock protein 75, glycerate kinase 1, Ran-GTP hydrolase activator protein! (Ran_GTPase activating protein 1 ) and WD-repeat c〇ntaining protein 1 ° ° Although the preferred embodiment of the invention has been disclosed above, it is not intended to limit the invention 'any of the skill Various changes and modifications can be made without departing from the spirit and scope of the invention. Accordingly, other embodiments are also included in the scope of the following claims. 76 200804411 [Simplified Schematic] The first figure shows the Lineweaver-Burk double reciprocal relationship diagram of the inhibition analysis of the synthetic inhibitor 22 by α-2,3 sialyltransferase in mice. Figure 2 (A) shows three α-2,3 sialyltransferase inhibitors - N-Pentyonic (Compound 21), Lith-Triethyl-de (Compound 20) and Lith-o-Asp (Compound 13), respectively. Cytotoxicity in CL1-5 cells 48 hours after treatment with 5 μΜ, 10 μΜ inhibitor. Survival scores were calculated as the survival of cells in the control group treated with DMSO. The second panel (Β) shows the attachment of CL1-5 cells after treatment with α-2,3 sialyltransferase inhibitors relative to the basic level of cells attached to the control group treated with DMSO (specification 1) The case of type 1 collagen. The third panel shows the results of gap healing analysis after 48 hours of treatment of C^L1:5 cells with (A)DMS〇, (Β) 1〇 μΜ Uth 〇 Asp. The α 2,3 sialic acid transaminase inhibitor Lith-0-Asp inhibits cell migration of the injured site due to scraping of cells from cell culture orphans. The fourth panel shows the expression of α_2,3 sialylation antigen on the cell surface at low = sex: U_1 cells and (Β) highly metastatic CL1·5 cells. ^The results of the non-flow cytometry display: P1 is the background intensity of 疚^2 and p3_Λ is the percentage of cells expressing sialic acid at high intensity and calculating the various cells from Uit. The figure below shows a histogram made by the average fluorescence intensity value of the piece. The amount of expression of endogenous mRNA of a in the lung cells of different metastatic potentials. (B) shows that 200804411 did not significantly change the mRNA expression of the ST3Gal I gene in CL1-5 cells treated with DMSO or Lith-o-Asp. The sixth panel shows a two-dimensional electropherogram of (A) CL1-5 lung cells treated with DMSO control and (B) Lith-o-Asp. The IEF is pH 3-10 and the molecular weight is 21.0-97.0 kDa. Differentially expressed proteins were further identified by mass spectrometry and tandem mass spectrometry. [Main component symbol description] None 78

Claims (1)

200804411 十、申請專利範圍: 1. 一種至少一種選自化學式I的化學物質:200804411 X. Patent application scope: 1. A chemical substance selected from at least one chemical formula I: 化學式I 及醫藥上可接受之鹽類、溶劑化物、螯合劑、非共價複合 物、前驅藥物及其混合物,其中 R!為選擇性取代之烷基; R2係選自羥基及醯氧基; R3係為氳,或R2與R3連同其所附著的碳,形成一個氧 基;及 當Ri為(/?)-‘羧基丁烷-2-基時,則R2非為羥基。 2·如申請專利範圍第1項所述之至少一種化學物質,其中前 述Ri選自-CHR4(CH2)nR5,其中,η為2和3 ; R4選自氫或選擇性取代的低碳數烷基;及 R5選自羧基、甲醯胺基及選擇性取代的雜芳基。 3. 如申請專利範圍第2項所述之至少一種化學物質,其中前 述η為2。 4. 如申請專利範圍第2項所述之至少一種化學物質,其中前 述η為3。 5. 如申請專利範圍第2項所述之至少一種化學物質,其中前 述R4選自氳及選擇性取代的低碳數烷基。 79 200804411 6·如申請專利範圍第5項所述之至少一種化學物質,其中前 述R4為低碳數烷基。 ' ’、 7·如申請專利範圍第6項所述之至少一種化學物質,並中. 述r4為甲基。 、 8·如申請專利範圍第2項所述之至少一種化學物質,其中前 述R5選自羧基、-(CO)-NHR6及選擇性取代的三唾R6 選自選擇性取代的低碳數烷基。 6 9·如申請專利範圍第8項所述之至少一種化學物質,其中前 述R5選自羧基、-(CO)-NHR6及選擇性取代的三唾,其中 三唑環係為一官能基&amp;所取代,而&amp;為選擇性取代的 低碳數烷基。 10·如申請專利範圍第8項所述之至少一種化學物質,其中前 述R6選自被一個或兩個羧基所取代的低碳數烷基。 11·如申請專利範圍第8項所述之至少一種化學物質,其中前 述R5為致基。 12·如申請專利範圍第8項所述之至少一種化學物質,其中 前述R5為一取代三嗤,其取代基係選自被一個或兩個叛 基、氨基及雙烷基磷酸所選擇性取代的低碳數烷基,且 该低碳數烧基係被至少一個叛基所取代。 13·至如申請專利範圍第1項所述之至少一種化學物質,其中 前述R3為氫。 14.如申請專利範圍第1項所述之至少一種化學物質,其中前 述R2與R3連同其所附著的碳,形成一個氧基。 15·如申請專利範圍第1項所述之至少一種化學物質,其中前 述之至少一種抑制α-2,3唾液酸轉移酶活性之化學物質,在 體外生化活性分析中的半抑制濃度IC5〇少於約360 μΜ。 80 200804411 16. 如申請專利範圍第15項所述之至少一種化學物質,其中前 述之至少一種抑制α-2,3唾液酸轉移酶活性之化學物質,在 體外生化活性分析中的半抑制濃度(IC5〇)少於約25 μΜ。 17. 如申請專利範圍第1項所述之至少一種化學物質,其中前 述化學式I之化合物係選自: (311,511,103,13尺,145)-17-((11)-5-羥基戊烷-2-基)-10,13-二曱基-十六烷氫-1Η-環戊烷[a]菲-3-酮; (R) -4-((3R,5R,10S,13R,14S)-3-羥基-10,13-二曱基-十六 烷氫-1H-環戊烷[a]菲-17-基)戊基二苯基磷酸; 2-((11)-4-((311,511,103,1311,145)-3-羥基-10,13-二甲基-十 六烷氫-1H-環戊烷[a]菲-17-基)戊醯胺)乙酸; (S) -2-((R)-4-((3R,5R, 10S,13R,14S)-3-羥基-10,13-二曱基-十六烷氫-1H-環戊烷[a]菲-17-基)戊醯胺)琥珀酸; (R)-4-((5R,10S,13R,14S)-10,13-二甲基-3-氧-十六烷氫 -1H-環戊烷[a]菲-17-基)戊酸; (R)-4-((3R,5R,10S,13R,14S)-3-(3-羧基丙醯基氧 基)-10,13-二曱基-十六烷氫-1H-環戊烷[a]菲-17-基)戊 酸; (11)-4-((3尺,511,105,13尺,143)-3-((3)-2-胺基-3-羧基丙醯基 氧基)-10,13-二曱基-十六烷氫-1H-環戊烷[a]菲-17-基)戊 酸; 4-((311,511,103,13艮143)-17-((11)-5-第三丁氧基-5-氧戊-2- 81 200804411 基)-10,13-二甲基-十六烷氫-1H-環戊烷[a]菲-3-基氧 基)-4-氧丁酸; 4-((3R,5R,10S,13R,14S)-17-((R)-5-(羧基甲基胺基)-5-氧 戊-2-基)-10,13-二甲基-十六烷氫-1H-環戊烷[a]菲-3-基氧 基)-4-氧丁酸; (S)-2-((R)-4-((3R,5R,10S,13R,14S)-3-(3-羧基丙醯基氧 基)-10,13-二曱基-十六烷氫-1H-環戊烷[a]菲-17-基)戊醯 胺)琥珀酸; (尺)-4-((3尺,5艮103,13艮145)-3-((11)-2-胺基-3-羧基丙醯基 氧基)-10,13-二甲基-十六烷氫-1H-環戊烷[a]菲-17-基)戊 酸; (3R,5R,10S,13R,14S)-17-((R)-5-(4-(2- 羥基乙 基)-1Η-(1,2,3-三唑-1-基)戊烷-2-基)-10,13-二甲基-十六 烷氳-1H-環戊烷[a]菲-3-酮; 2-胺基 _3-(l-((R)-4-((3R,5R,10S,13R,14S)-3j|*-10,13-二曱基-十六烷氫-1H-環戊烷[a]菲-17-基)戊基)-1Η-1,2,3-三峻-4-基)丙酸; 2-( 二 乙氧基 磷 醯 基)-3-(l-((R)-4-((3R,5R,10S,13R,14S)-3^i*-10,13-:T 基-十六烷氫-1H-環戊烷[a]菲-17-基)戊基-1H-1,2,3-三唑 -4-基)丙酸; 82 200804411 3- (1-((幻-4-((3尺,511,103,13&amp;145)-3-羥基-1〇,13-二甲基-十六炫氮-1H-環戊烷[a]菲-17-基)戊基)-iH-l,2,3-三唑-4-基)丙酸;及 4- (1-((尺)-4-((31511,103,131^,148)-3-經基-1〇,13-二甲基-十六烷氫-1H-環戊烷[a]菲-17-基)戊基)·1Η-1,2,3-三唑_心 基)丁酸。 18. -種醫藥組合物,包含-治療上有效量之如申請專利範圍第 1項所述之化學物質及一醫藥上可接受之载體。 19. 一種抑制α-2,3唾液酸轉移酶活性之方法= -足以祭覺可使糖結合物唾㈣化減少之至少一種如申 1專利範圍第i項所賴示之化學 唾液酸轉移酶之細胞相接觸。 』里/、衣現α 20·如申請專利範圍第19項所述 細胞。 Τ、夂方法,其中前述細胞為癌 21·如申請專利範圍第2〇項所述之 進行細胞遷移。 / /、中前述癌細胞係 22·如申請專利範圍第21項所述之方法,复… 係伴隨癌細胞遷移。 /、中則述細胞遷移 23.如申請專利範圍第19項所述之方法,复 在於哺乳動物。 /、中則述細胞係存 24·如申請專利範圍第23項所 係為人類。 乃&amp;其中珂述哺乳動物 25·-種治療患有對抑帝】α_2,3唾 、 疾病的患者之方法,·軲私_活性有反應之 至少-如申請患者治療上有效量之 26.如申請專利範圍* 25項所述之方法化2質。 ^ 其中前述對抑制 83 200804411 α-2,3唾液酸轉移酶活性有反應之疾病係為癌症。 27. 如申請專利範圍第26項所述之方法,其中前述癌係進行 細胞遷移。 28. 如申請專利範圍第25項所述之方法,其中前述患者係為 人類。 29. 如申請專利範圍第25項所述之方法,係進一步包括施予 至少一種額外的、可影響合併療法之的治療藥物。 84And a pharmaceutically acceptable salt, solvate, chelating agent, non-covalent complex, prodrug, and mixtures thereof, wherein R! is an optionally substituted alkyl group; R2 is selected from the group consisting of a hydroxyl group and a decyloxy group; R3 is hydrazine, or R2 and R3 together with the carbon to which they are attached form an oxy group; and when Ri is (/?)-'carboxybutan-2-yl, then R2 is not a hydroxy group. 2. The at least one chemical substance of claim 1, wherein the Ri is selected from the group consisting of -CHR4(CH2)nR5, wherein η is 2 and 3; and R4 is selected from hydrogen or an optionally substituted lower alkylene. And R5 are selected from the group consisting of a carboxyl group, a formamidine group, and an optionally substituted heteroaryl group. 3. The at least one chemical substance according to item 2 of the patent application, wherein the aforementioned η is 2. 4. At least one chemical substance as claimed in claim 2, wherein the aforementioned η is 3. 5. The at least one chemical substance of claim 2, wherein the aforementioned R4 is selected from the group consisting of hydrazine and an optionally substituted lower alkyl group. 79 200804411 6. The at least one chemical substance of claim 5, wherein the aforementioned R4 is a lower alkyl group. ' </ br /> </ RTI> <RTIgt; </ RTI> <RTIgt; </ RTI> <RTIgt; </ RTI> <RTIgt; 8. The at least one chemical substance of claim 2, wherein the R5 is selected from the group consisting of a carboxyl group, -(CO)-NHR6, and an optionally substituted trisalin R6 is selected from the group consisting of an optionally substituted lower alkyl group. . 6. The at least one chemical substance of claim 8, wherein the R5 is selected from the group consisting of a carboxyl group, -(CO)-NHR6, and an optionally substituted trisal, wherein the triazole ring system is a monofunctional group &amp; Substituted, and &amp; is an optionally substituted lower alkyl group. 10. The at least one chemical substance of claim 8, wherein the aforementioned R6 is selected from the group consisting of a lower alkyl group substituted with one or two carboxyl groups. 11. The at least one chemical substance of claim 8, wherein the aforementioned R5 is a radical. 12. The at least one chemical according to claim 8, wherein the R5 is a monosubstituted triterpene, the substituents being selected from the group consisting of one or two rebel, amino and dialkyl phosphates Lower alkyl, and the lower carbon number is replaced by at least one rebel. 13. The at least one chemical substance of claim 1, wherein the aforementioned R3 is hydrogen. 14. The at least one chemical of claim 1, wherein the foregoing R2 and R3 together with the carbon to which they are attached form an oxy group. 15. The at least one chemical substance according to claim 1, wherein the at least one chemical substance which inhibits the activity of α-2,3 sialyltransferase has a semi-inhibitory concentration IC5 in the in vitro biochemical activity analysis. About 360 μΜ. 80 200804411 16. The at least one chemical substance according to claim 15, wherein the at least one chemical substance inhibiting α-2,3 sialyltransferase activity has a semi-inhibitory concentration in an in vitro biochemical activity assay ( IC5〇) is less than about 25 μΜ. 17. The at least one chemical substance according to claim 1, wherein the compound of the above formula I is selected from the group consisting of: (311, 511, 103, 13 ft, 145) -17-((11)-5-hydroxyl Pentane-2-yl)-10,13-dimercapto-hexadecanehydro-1Η-cyclopentane [a]phenanthrene-3-one; (R) -4-((3R,5R,10S,13R , 14S)-3-hydroxy-10,13-dimercapto-hexadecanehydrol-1H-cyclopentane [a]phenanthrene-17-yl)pentyldiphenylphosphoric acid; 2-((11)-4 -((311,511,103,1311,145)-3-hydroxy-10,13-dimethyl-hexadecanehydrol-1H-cyclopentane [a]phenanthrene-17-yl)pentanylamine)acetic acid (S) -2-((R)-4-((3R,5R, 10S,13R,14S)-3-hydroxy-10,13-didecyl-hexadecanehydrol-1H-cyclopentane [ a]phenanthroline-17-yl)pentaamine)succinic acid; (R)-4-((5R,10S,13R,14S)-10,13-dimethyl-3-oxo-hexadecanehydrol-1H -cyclopentane [a]phenanthrene-17-yl)pentanoic acid; (R)-4-((3R,5R,10S,13R,14S)-3-(3-carboxypropenyloxy)-10, 13-dimercapto-hexadecanehydro-1H-cyclopentane [a]phenanthrene-17-yl)pentanoic acid; (11)-4-((3 ft, 511, 105, 13 ft, 143)-3 -((3)-2-amino-3-carboxypropenyloxy)-10,13-dimercapto-hexadecanehydro-1H-cyclopentane [a]phenanthrene -17-yl)pentanoic acid; 4-((311,511,103,13艮143)-17-((11)-5-t-butoxy-5-oxopen-2- 81 200804411)-10, 13-dimethyl-hexadecanehydrol-1H-cyclopentane [a]phenanthr-3-yloxy)-4-oxobutanoic acid; 4-((3R,5R,10S,13R,14S)-17 -((R)-5-(carboxymethylamino)-5-oxopen-2-yl)-10,13-dimethyl-hexadecanehydro-1H-cyclopentane [a]phenanthrene-3 -yloxy)-4-oxobutyric acid; (S)-2-((R)-4-((3R,5R,10S,13R,14S)-3-(3-carboxypropenyloxy) -10,13-dimercapto-hexadecanehydro-1H-cyclopentane [a]phenanthrene-17-yl)pentanylamine succinic acid; (foot)-4-((3 ft, 5 艮 103, 13艮145)-3-((11)-2-Amino-3-carboxypropenyloxy)-10,13-dimethyl-hexadecanehydro-1H-cyclopentane [a]phenanthrene- 17-yl)pentanoic acid; (3R,5R,10S,13R,14S)-17-((R)-5-(4-(2-hydroxyethyl)-1Η-(1,2,3-triazole) -1-yl)pentan-2-yl)-10,13-dimethyl-hexadecane-1H-cyclopentane [a]phenanthrene-3-one; 2-amino group 3-3-(l- ((R)-4-((3R,5R,10S,13R,14S)-3j|*-10,13-dimercapto-hexadecanehydrol-1H-cyclopentane [a]phenanthrene-17-yl Pentyl)-1Η-1,2,3-trisyl-4-yl)propionic acid; 2-(diethoxyphosphonium) -3(l-((R)-4-((3R,5R,10S,13R,14S)-3^i*-10,13-:T-hexadecanehydrol-1H-cyclopentyl) Alkane [a]phenanthrene-17-yl)pentyl-1H-1,2,3-triazol-4-yl)propanoic acid; 82 200804411 3- (1-((4-((3), 511 ,103,13&amp;145)-3-hydroxy-1〇,13-dimethyl-hexadecane-1H-cyclopentane [a]phenanthyl-17-yl)pentyl)-iH-l,2, 3-triazol-4-yl)propionic acid; and 4-(1-(())-4-((31511,103,131^,148)-3-yl-1, 13-dimethyl -hexadecanehydrol-1H-cyclopentane [a]phenanthrene-17-yl)pentyl)·1Η-1,2,3-triazole-cardio)butyric acid. 18. A pharmaceutical composition comprising - a therapeutically effective amount of a chemical as described in claim 1 and a pharmaceutically acceptable carrier. 19. A method for inhibiting the activity of α-2,3 sialyltransferase = - sufficient to satisfactorily reduce at least one of the reduction of salivation of a sugar conjugate, such as the chemical sialyltransferase represented by the item i of claim 1 The cells are in contact. 』, /, clothing α 20 · as described in the scope of claim 19 cells. A method of sputum and sputum, wherein the aforementioned cells are cancers. 21. Cell migration is carried out as described in the second paragraph of the patent application. / /, the aforementioned cancer cell line 22. The method described in claim 21, which is accompanied by cancer cell migration. /, in the case of cell migration 23. The method of claim 19, which is a mammalian. /, in the middle of the cell line 24. As claimed in the 23rd section of the patent application for humans. It is a method of treating a mammal with a 25--type treatment of a patient suffering from a depression of α 2, 3 saliva, and a disease, and at least responsive to the activity of the patient - such as the therapeutically effective amount of the patient. As described in the scope of application for patents*25, the quality of the method is as follows. ^ The aforementioned disease which is responsive to the inhibition of 83 200804411 α-2,3 sialyltransferase activity is cancer. 27. The method of claim 26, wherein the cancer cell is subjected to cell migration. 28. The method of claim 25, wherein the aforementioned patient is a human. 29. The method of claim 25, further comprising administering at least one additional therapeutic agent that affects the combination therapy. 84
TW095124663A 2006-07-06 2006-07-06 Certain lithocholic acid analogues that inhibit sialyltransferase TWI369361B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
TW095124663A TWI369361B (en) 2006-07-06 2006-07-06 Certain lithocholic acid analogues that inhibit sialyltransferase

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
TW095124663A TWI369361B (en) 2006-07-06 2006-07-06 Certain lithocholic acid analogues that inhibit sialyltransferase

Publications (2)

Publication Number Publication Date
TW200804411A true TW200804411A (en) 2008-01-16
TWI369361B TWI369361B (en) 2012-08-01

Family

ID=44765842

Family Applications (1)

Application Number Title Priority Date Filing Date
TW095124663A TWI369361B (en) 2006-07-06 2006-07-06 Certain lithocholic acid analogues that inhibit sialyltransferase

Country Status (1)

Country Link
TW (1) TWI369361B (en)

Also Published As

Publication number Publication date
TWI369361B (en) 2012-08-01

Similar Documents

Publication Publication Date Title
JP7061161B2 (en) Nuclear transport regulators and their use
JP7195402B2 (en) Silicon-based drug complex and method of using the same
JP6777626B2 (en) Polymorphic form of serinexol
JP5261488B2 (en) Proteasome inhibitor
CN105899493B (en) For inhibiting the active 1- of SHP2 (triazine -3- base/pyridazine -3- base)-piperazine (- piperazine) piperidine derivatives and combinations thereof
JP5372751B2 (en) AZA-peptide protease inhibitor
CN106232612B (en) Bone selectivity osteogenic oxygen sterol diphosphonate is similar to object
EA025845B1 (en) Modulators of pharmacokinetic properties of therapeutics
TW201425321A (en) Proteasome inhibitors
CN106589055B (en) Substituted cell acyl dipeptide compound and preparation method and application thereof
TW200412349A (en) Eponemycin and epoxomicin analogs and uses thereof
CN103896921A (en) Compounds and compositions as protein kinase inhibitors
EA020488B1 (en) Derivatives of purine or deazapurine useful for the treatment of viral infections and other diseases
TW201305185A (en) 2&#39;-fluoro substituted carba-nucleoside analogs for antiviral treatment
PT2069303E (en) Antiviral protease inhibitors
CN116348455A (en) Protein tyrosine phosphatase inhibitors and methods of use thereof
TWI345465B (en) Proteasome inhibitors and methods of using the same
US8940719B2 (en) Lithocholic acid analogues that inhibit sialyltransferase
CN101835747A (en) Benzimidazole derivative
CA3170503A1 (en) Stat degraders and uses thereof
CN107531685B (en) Compounds with antitumor activity
TW200804411A (en) Certain lithocholic acid analogues that inhibit sialyltransferase
CN105960399B (en) Enzyme inhibitor epoxy ketone compound
EP4326260A1 (en) Pi3k inhibitors, nanoformulations, and uses thereof
WO2021104532A1 (en) Brefeldin a derivative, preparation method therefor, and use thereof

Legal Events

Date Code Title Description
MM4A Annulment or lapse of patent due to non-payment of fees