TW201444864A - Hexadepsipeptide analogues as anticancer compounds - Google Patents

Hexadepsipeptide analogues as anticancer compounds Download PDF

Info

Publication number
TW201444864A
TW201444864A TW102118552A TW102118552A TW201444864A TW 201444864 A TW201444864 A TW 201444864A TW 102118552 A TW102118552 A TW 102118552A TW 102118552 A TW102118552 A TW 102118552A TW 201444864 A TW201444864 A TW 201444864A
Authority
TW
Taiwan
Prior art keywords
compound
cancer
formula
alkyl
pharmaceutically acceptable
Prior art date
Application number
TW102118552A
Other languages
Chinese (zh)
Inventor
Prabhu Dutt Mishra
Sreekumar Sankaranarayanan Eyyammadichiyil
Saji David George
Shailendra Sonawane
Subhash Narayan Chakor
Abhijit Roychowdhury
Rajiv Sharma
Original Assignee
Piramal Entpr Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Piramal Entpr Ltd filed Critical Piramal Entpr Ltd
Priority to TW102118552A priority Critical patent/TW201444864A/en
Publication of TW201444864A publication Critical patent/TW201444864A/en

Links

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

This invention relates to an isolated compound of Formula (1) or derivatives or pharmaceutically acceptable salts thereof. The invention also includes all isomeric and tautomeric forms of the compound of Formula (1) or the derivatives thereof. The present invention further relates to processes for the production of the compound of Formula (1) by fermentation of the fungal strain of Actinomycetes (PM0895172/MTCC5684), pharmaceutical compositions comprising the compound of Formula (1) as the active ingredient; and use of the said compounds or composition containing them in the treatment of cancer.

Description

六酯肽類似物作為抗癌化合物Hexapeptide analog as an anticancer compound

本發明係相關於結構式(1)之化合物(如同本文中所描述的)、藥學上可接受之鹽類或其衍生物。本發明進一步地相關於來自分離的屬於放射菌株(PM0895172/MTCC 5684)的微生物之結構式(1)之化合物生產程序。本發明亦相關於包含結構式(1)之化合物、其藥學上可接受之鹽類或衍生物作為活性成分的藥學組成物;以及其癌症治療的用途。The invention is related to a compound of formula (1) (as described herein), a pharmaceutically acceptable salt or a derivative thereof. The present invention is further related to a compound production procedure of structural formula (1) from an isolated microorganism belonging to a radiation strain (PM0895172/MTCC 5684). The present invention is also related to a pharmaceutical composition comprising the compound of the formula (1), a pharmaceutically acceptable salt or derivative thereof as an active ingredient; and the use thereof for the treatment of cancer.

癌症是一個大群組疾病的總稱,其由不受控制的細胞生長與擴散而導致,其可影響身體的任何部分。現今對癌症有數個治療選擇是可得的,包括化療、外科手術與對局部病灶的放射或所述治療的結合。療法的選擇視癌症的位置以及亦視癌症於診斷時擴散的程度而定。化療為癌症治療的最常見的形式之一,其涉及可催毀癌症細胞之抗癌藥物的使用。僅管癌症的治療方案持續進步,此疾病仍然是世界上主要死亡原因之一,最可能的理由是可得的治療選擇與不良的副作用與有限的效果相關。因此,需要表現治療癌症之臨床好處的新抗癌劑/藥物。
有來自天然資源的抗癌化合物的報導,例如紫杉酚(taxol)、長春新鹼(vincristine)、torreyanic acid 以及喜樹鹼(camptothecin)(Natural Product Communications, 2009, 4 (11), 1513)。
海生環境,覆蓋了百分之七十的地球表面以及百分之九十五的熱帶生物,其表現了三十六門中的三十四門生命,且提供了各種迷人的生物多樣性,超過了陸地環境表現的生物多樣性(Life Sciences, 2005, 78, 442-453)。為生物多樣性與內在知識的探索、提煉與篩選以供具商業價值的遺傳與生化資源的海生天然產物生物勘探,已產生了相當多的藥物候選者(Drug Discovery Today, 2003, 8 (12), 536-544)。
已經有一些藥劑發現的報導,其涉及來自海生生物與微生物之天然產物的篩選。源自海生來源的抗癌劑包括阿糖胞苷(cytarabine)、苔蘚蟲抑素(bryostatin-1)、阿吡利丁(aplidine)、海兔毒肽10(dolastatin)以及ET-743(Current Opinion in Pharmacology, 2001, 1, 364-369)。
本領域報導了一些六酯肽。舉例來說,JP09040559揭露具有抗發炎活性的六酯肽類化合物,其對過敏性與非過敏性發炎是有效的。
考量許多具有治療活性的化合物已由天然來源獲得,探索此研究領域以獲得可有效用於疾病像是癌症的治療的新化合物是審慎的。本專利說明書的發明者把他們的努力提供發現在癌症治療的用途之新穎化合物。亦提供的是生產此化合物的方法,其經由發酵作用以及進一步地,影響結構式(1)之化合物的化學修飾,以獲得其衍生物。
Cancer is a general term for a large group of diseases caused by uncontrolled cell growth and spread, which can affect any part of the body. Several treatment options are available for cancer today, including chemotherapy, surgery, and radiation to localized lesions or a combination of such treatments. The choice of therapy depends on the location of the cancer and also on the extent to which the cancer spreads at the time of diagnosis. Chemotherapy is one of the most common forms of cancer treatment involving the use of anticancer drugs that can destroy cancer cells. Although the treatment of cancer continues to progress, the disease remains one of the leading causes of death in the world, and the most likely reason is that the available treatment options are associated with poor side effects and limited effects. Therefore, there is a need for new anticancer agents/drugs that exhibit the clinical benefit of treating cancer.
There are reports of anticancer compounds from natural sources, such as taxol, vincristine, torreyanic acid, and camptothecin (Natural Product Communications, 2009, 4 (11), 1513).
The marine environment, covering 70% of the Earth's surface and 95% of tropical life, represents thirty-four of the thirty-six lives and offers a variety of fascinating biodiversity. Biodiversity beyond terrestrial environmental performance (Life Sciences, 2005, 78, 442-453). A number of drug candidates have been produced for the exploration, refinement and screening of biodiversity and intrinsic knowledge for the bio-exploration of marine natural products with commercially valuable genetic and biochemical resources (Drug Discovery Today, 2003, 8 (12) ), 536-544).
There have been reports of drug discovery involving screening of natural products from marine organisms and microorganisms. Anticancer agents derived from marine sources include cytarabine, bryostatin-1, aplidine, dolastatin, and ET-743 (Current) Opinion in Pharmacology, 2001, 1, 364-369).
Some hexaester peptides are reported in the art. For example, JP09040559 discloses hexaester peptide compounds having anti-inflammatory activity which are effective for allergic and non-allergic inflammation.
Considering that many therapeutically active compounds have been obtained from natural sources, it is prudent to explore this field of research to obtain new compounds that are effective for the treatment of diseases such as cancer. The inventors of this patent specification have provided their efforts to provide novel compounds found for use in cancer therapy. Also provided is a method of producing this compound which, via fermentation and, further, chemical modification of a compound of formula (1) to obtain a derivative thereof.

本發明係相關於在本文中指定為結構式(1)之化合物(如同本文所描述)的化合物。
本發明係相關於結構式(1)之化合物或其藥學上可接受之鹽類或其衍生物的同分異構物形式、互變異構物形式。
根據本發明,結構式(1)之化合物為結構式(1a)之化合物或結構式(1b)之化合物。
根據本發明,結構式(1)之化合物的衍生物係包含於結構式(1c)之化合物。
本發明亦相關於純化的結構式(1)之化合物,其係由海生放射菌株(PM0895172/MTCC 5684)或其變異株或突變株中之一的發酵作用獲得的培養液分離。於是,本發明之化合物為分離的結構式(1)之化合物。
本發明亦相關於結構式(1)之化合物及/或其同分異構物或其互變異構物從海生放射菌株(PM0895172/MTCC 5684)的生產程序。
本發明進一步相關於屬於放射菌株(PM0895172/MTCC 5684)的微生物之分離程序,其培養生產結構式(1)之化合物,或其同分異構物或互變異構物。
結構式(1)之化合物或其同分異構物或互變異構物或其一藥學上可接受之鹽類或一衍生物對癌症的治療是有用處的。
本發明係相於病患癌症治療的方法,其包含給藥予病患治療有效量的結構式(1)之化合物或其同分異構物或互變異構物或藥學上可接受之鹽類或衍生物。
本發明亦相關於包含結構式(1)之化合物或其同分異構物或互變異構物或藥學上可接受之鹽類或其衍生物作為活性成分,以及至少一藥學上可接受的賦形劑、載體或媒液的藥學組成物。
The invention relates to compounds which are designated herein as compounds of formula (1) (as described herein).
The present invention relates to the isomeric form, tautomeric form of the compound of the formula (1) or a pharmaceutically acceptable salt thereof or a derivative thereof.
According to the invention, the compound of formula (1) is a compound of formula (1a) or a compound of formula (1b).
According to the invention, the derivative of the compound of the formula (1) is contained in the compound of the formula (1c).
The present invention is also related to a purified compound of the formula (1) which is isolated from a culture solution obtained by fermentation of one of marine growth strains (PM0895172/MTCC 5684) or a variant or mutant thereof. Thus, the compound of the present invention is an isolated compound of the formula (1).
The invention also relates to a process for the production of a compound of formula (1) and/or its isomers or tautomers thereof from a marine radiation strain (PM0895172/MTCC 5684).
The invention further relates to a process for the isolation of microorganisms belonging to a radiation strain (PM0895172/MTCC 5684) which produces a compound of formula (1), or an isomer or tautomer thereof.
The compound of formula (1) or an isomer or tautomer thereof or a pharmaceutically acceptable salt or derivative thereof is useful for the treatment of cancer.
The present invention relates to a method of treating cancer in a patient comprising administering to the patient a therapeutically effective amount of a compound of formula (1) or an isomer or tautomer or pharmaceutically acceptable salt thereof. Or a derivative.
The invention also relates to a compound comprising structural formula (1) or an isomer or tautomer thereof or a pharmaceutically acceptable salt or derivative thereof as an active ingredient, and at least one pharmaceutically acceptable A pharmaceutical composition of a form, carrier or vehicle.

no

第1圖說明結構式(1a)之化合物的1H NMR(CDCl3; 500 MHz;儀器:Bruker)光譜。
第2圖說明結構式(1a)之化合物的13C NMR(CDCl3; 75 MHz;儀器:Bruker)光譜。
第3圖說明結構式(1b)之化合物的1H NMR(CDCl3; 500 MHz;儀器:Bruker)光譜。
第4圖說明結構式(1b)之化合物的13C NMR(CDCl3; 75 MHz;儀器:Bruker)光譜。
第5圖說明結構式(1a)之化合物對HeLa癌細胞的細胞凋亡之影響。
Figure 1 illustrates the 1 H NMR (CDCl 3 ; 500 MHz; instrument: Bruker) spectrum of the compound of formula (1a).
Figure 2 illustrates the 13 C NMR (CDCl 3 ; 75 MHz; instrument: Bruker) spectrum of the compound of formula (1a).
Figure 3 illustrates the 1 H NMR (CDCl 3 ; 500 MHz; instrument: Bruker) spectrum of the compound of formula (1b).
Figure 4 illustrates the 13 C NMR (CDCl 3 ; 75 MHz; instrument: Bruker) spectrum of the compound of formula (1b).
Figure 5 illustrates the effect of a compound of formula (1a) on apoptosis of HeLa cancer cells.

本發明係提供如同以下表示的結構式(1)之化合物:



結構式(1)
其中在結構式(1)之化合物中,當鍵代表單鍵且R1為H;將化合物稱作結構式(1a)之化合物;以及
當鍵為雙鍵且R1不存在時,將化合物稱作結構式(1b)之化合物。於是,結構式(1)之化合物為結構式(1a)之化合物(其中該鍵代表單鍵且R1為H)或結構式(1b)之化合物(其中該鍵為雙鍵且R1不存在)或其混合物。
結構式(1)之化合物屬於六酯肽類化合物。
結構式(1a)之化合物以及結構式(1b)之化合物可以任何一或更多的物理-化學以及光譜特性描述其特徵,例如如同本文以下討論的高效液相層析法(HPLC)、高解析質譜(HR MS)、遠紅外線(IR)以及核磁共振(NMR)光譜數據。
根據本發明的一方面,結構式(1)之化合物為結構式(1a)之化合物,其具有分子式 C37H62N8O13以及分子量826.9。
結構式(1a)之化合物係表示如下:



結構式(1a)
結構式(1a)之化合物的化學名稱為N-(6,18-二羥基-22-異丙基-7,19-二甲基-5,8,11,17,20,24-六氧二十四氫二噠嗪并[6,1-f:6',1'-o][1,4,7,10,13,16]五氮雜氧環十九炔-23-基)-2-羥基-2-(2-羥基-5-異丁基-6-甲基四氫-2H-吡喃-2-基)丙醯胺。
根據本發明的一方面,結構式(1)之化合物為結構式(1b)之化合物,其具有分子式C37H60N8O13,以及824.4的分子量。
結構式(1b)之化合物係表示如下:



結構式(1b)
結構式(1b)之化合物的化學名稱為N-(6,18-二羥基-22-異丙基-7,19-二甲基-5,8,11,17,20,24-六氧-3,4,4a,5,6,7,8,9, 10,11,13,14,15, 16,16a, 17,18,19,20,22,23,24-二十二氫二噠嗪并[6,1-f:6',1'-o][1,4,7,10, 13,16]五氮雜氧環十九炔-23-基)-2-羥基-2-(2-羥基-5-異丁基-6-甲基四氫-2H-吡喃-2-基)丙醯胺。
可使用於結構式(1)之化合物的生產之微生物為放射菌株(PM0895172/MTCC 5684),本文以下稱為編號PM0895172,將其由印度馬哈拉施特拉邦,孟買的近海區域收集的深海沉積物中分離。
本發明的一方面提供來自培養編號PM0895172的結構式(1)之化合物的生產程序,其包含步驟:
a) 將培養編號PM0895172或其變異型或突變型中之一者於水下需氧條件下,於包含一或更多來源的碳與一或更多來源的氮以及營養無機鹽及/或微量元素的營養培養基中生長,以獲得包含結構式(1)之化合物的培養液;
b)從培養液中分離結構式(1)之化合物;以及
c)純化結構式(1)之化合物。
根據上述程序生產的結構式(1)之化合物可為結構式(1a)之化合物或結構式(1b)之化合物。
在一具體實施例中,在步驟(a)中的培養液包含結構式(1a)之化合物與結構式(1b)之化合物的混合物。

在一具體實施例中,在程序的步驟(b)中,結構式(1)之化合物為結構式(1a)之化合物。
在一具體實施例中,在程序的步驟(b)中,結構式(1)之化合物為結構式(1b)之化合物。
在一具體實施例中,在程序的步驟(c)中,結構式(1)之化合物為結構式(1a)之化合物。
在一具體實施例中,在程序的步驟(c)中,結構式(1)之化合物為結構式(1b)之化合物。
在一具體實施例中,本發明提供來自培養編號PM0895172的結構式(1a)之化合物的生產程序,其包含步驟:
a)將培養編號PM0895172或其變異型或突變型中之一者於水下需氧條件下,於包含一或更多來源的碳與一或更多來源的氮以及營養無機鹽及/或微量元素的營養培養基中生長,以獲得包含結構式(1a)之化合物與結構式(1b)之化合物的混合物培養液;
b)從培養液中分離結構式(1a)之化合物;以及
c)純化結構式(1a)之化合物。
在另一具體實施例中,本發明提供來自培養編號PM0895172的結構式(1b)之化合物的生產程序,其包含步驟:
a)將培養編號PM0895172或其變異型或突變型中之一者於水下需氧條件下,於包含一或更多來源的碳與一或更多來源的氮以及營養無機鹽及/或微量元素的營養培養基中生長,以獲得包含結構式(1a)之化合物與結構式(1b)之化合物的混合物培養液;
b)從培養液中分離結構式(1b)之化合物;以及
c)純化結構式(1b)之化合物。
如上述程序中之任一者的步驟(c)涉及結構式(1)之化合物的純化,並且經由使用相關領域中通常使用的純化程序進行。
結構式(1)之化合物,特別是根據本發明的程序產生之結構式(1a)之化合物與結構式(1b)之化合物為大體上純的化合物。因此,結構式(1)之化合物,特別是結構式(1a)之化合物或結構式(1b)之化合物為分離的純化合物。
典型地,結構式(1a)之化合物與結構式(1b)之化合物係經由管柱層析技術純化。
如同本文所使用的,該用語「突變株(mutant)」意指生物體或細胞,其中基因體中的一或更多基因已被以一個或多個基因改變,該基因係負責生物體產生根據本發明之化合物的能力。可以本身已知的方式,使用物理手段例如以紫外線或X光照射或化學誘突變劑的方式產生此類突變株。
如同本文所使用的該用語「變異株(variant)」意指可識別不同於那物種的任意標準型之個別生物體。
該用語「整個培養液(whole broth)」可與該用語「營養培養液(nutrient broth)」、「培養液(culture broth)」或「發酵培養液(fermented broth)」交替使用。
如同本文所使用的該用語「哺乳動物」意指人類以及非人類哺乳動物,其包括但不限於牛、馬、豬、狗與貓。該用語「哺乳動物」可與該用語「病患(patient)」或「受試者(subject)」交替使用。
該用語「活性成分(active ingredient)」或「活性化合物(active compound)」可交替使用,且如同本文所使用的,所述用語意指結構式(1)之化合物或結構式(1a)之化合物或結構式(1b)之化合物或其同分異構物或互變異構物或其藥學上可接受之鹽類或衍生物。在本發明的上下文中,該用語「活性成分」或「活性化合物」亦意指結構式(1c)之化合物,其包含如同本文所描述的結構式(1)之化合物的衍生物。
該用語「結構式(1)之化合物」包括結構式(1a)之化合物或結構式(1b)之化合物或其混合物;以及其同分異構物、互變異構物或藥學上可接受之鹽類。
該用語「結構式(1c)之化合物」包括結構式(1)之化合物的衍生物,其包括結構式(1a)之化合物或結構式(1b)之化合物或其同分異構物、互變異構物或藥學上可接受之鹽類的衍生物。
該用語「大體上純的(substantially pure)」如同本文中所使用的,意指結構式(1)之化合物,特別是結構式(1a)之化合物或結構式(1b)之化合物或其同分異構物是夠純的,使得進一步的純化不會可偵測地改變其物理與化學特性以及化合物的酵素與生物活性。可將結構式(1)之化合物按照本領域的技術人員所知的方法大幅純化。
如同本文所使用的,該用語「治療有效量(therapeutically effective amount)」參考使用結構式(1)之化合物特別是結構式(1a)之化合物或結構式(1b)之化合物或其藥學上可接受之鹽類或衍生物(結構式(1c)之化合物(如同本文所描述))的癌症(如同本文列舉)之治療,意指能夠在接受本發明之化合物的病患引起一或更多下列效果的量:(i) 至某種程度的腫瘤生長抑制作用,其包括減緩與完全的生長停止;(ii)腫瘤細胞的數目減少;(iii)腫瘤大小縮小;(iv) 腫瘤細胞滲透到周圍器官的抑制作用(即,減少、減緩或完全停止);(v)轉移的抑制作用(即,減少、減緩或完全停止);(vi) 抗腫療免疫反應的增強作用,其可能但不一定造成腫瘤的復原;及/或 (vii) 受治療之癌症相關的一或更多症狀某種程度的緩解。
該用語「藥學上可接受之鹽類」如同本文中所使用的,意指包括結構式(1)之化合物特別是結構式(1a)之化合物或結構式(1b)之化合物,以及其衍生物(結構式(1c)之化合物)之本發明化合物的那些鹽類,其在哺乳動物中為安全且有效的並且擁有所希望的生物活性。藥學上可接受之酸加成鹽包括但不限於鹽酸鹽、氫溴酸鹽、氫碘酸鹽、硝酸鹽、硫酸鹽、磷酸鹽、乙酸鹽、乳酸鹽、水楊酸鹽、檸檬酸鹽、酒石酸鹽、抗壞血酸鹽、琥珀酸鹽、順丁烯二酸鹽、反丁烯二酸鹽、蟻酸鹽、苯甲酸鹽、麩胺酸鹽、甲磺酸鹽、苯磺酸鹽、對甲苯磺酸鹽。合適的鹼加成鹽包括但不限於鈣、鋰、鎂、鉀、鈉或鋅鹽。
經由其菌落特徵的評估進行培養編號PM0895172的初步鑑定,結構式(1)之化合物產生自該培養編號PM0895172。於修改過的放射菌分離瓊脂培養基進行對分離的培養編號PM0895172菌株之顯微鏡研究。於20°C至30°C的20-22日培養之後作觀察,直至觀察到菌落。培養編號PM0895172已確認為海生放射菌菌株。
培養編號PM0895172已存放於微生物菌種保存中心(Microbial Type Culture Collection,MTCC,在微生物科技學院(Sector 39-A, Chandigarh -160 036,印度)) (世界智慧財產權組織(World Intellectual Property Organization,WIPO) 認可的國際寄存管理局(International Depository Authority,IDA))並且已被提供登錄號MTCC 5684。
應了解除了本文描述的特定微生物,亦可培養PM0895172的突變株,例如那些由化學或物理誘突變劑(包括X光、U.V.光)等產生者以及已由分子生物技術修改基因構成的生物體,以生產結構式(1)之化合物。
可經由HPLC、NMR、IR、MS及/或累積於培養液中的活性化合物之生物活性測定,舉例來說經由測試化合物抗癌活性或經由其結合而確認可生產根據本發明之化合物的合適突變型與變異型的篩選。
使用以供生產結構式(1)之化合物的培養編號PM0895172分離與培養的培養基及/或營養培養基較佳地包含碳、氮以及營養無機鹽來源。碳源舉例來說,澱粉、葡萄糖、蔗糖、糊精、果糖、糖蜜、甘油、乳糖或半乳糖中的一或更多者。較佳的碳源為葡萄糖。氮源為,舉例來說大豆粉、花生粉、酵母萃取物、牛肉萃取物、蛋白腖、麥芽萃取物、玉米浸液、明膠或酪蛋白胺基酸中的一或更多者。較佳的氮源為蛋白腖與酵母萃取物。營養無機鹽為,舉例來說,氯化鈉、氯化鉀、氯化鈣、氯化錳、氯化鎂、氯化鍶、氯化鈷、溴化鉀、氟化鈉、磷酸氫鈉、磷酸氫鉀、磷酸氫二鉀、磷酸氫二鈉、碳酸鈣、碳酸氫鈉、矽酸鈉、硝酸鈉、硝酸銨、硝酸鉀、硫酸鈉、硫酸銨、七鉬酸銨、檸檬酸鐵、硫酸銅、硫酸鎂、硫酸亞鐵、硫酸鋅或硼酸中的一或更多者氯化鈉與氯化鈣為較佳的。
培養編號PM0895172的維持可於24°C至32°C範圍的溫度下進行。典型地,培養編號PM0895172維持於27°C-29°C。可將生長良好的培養株保存於4°C - 8°C冰箱。
培養編號PM0895172的種菌培養可於27°C至33°C範圍的溫度以及約5.5至8.5 pH下於210-260 rpm(每分鐘轉數)進行60-80小時。典型地,將編號PM0895172種株於29°C - 31°C以及pH約6.5- 7.5下,於230-250 rpm培養70-74小時。
結構式(1)之化合物的生產可經由將培養編號PM0895172 在搖晃的燒瓶中之發酵作用,於27°C至33°C 範圍的溫度以及約5.5至8.5 pH 下於 200-250 rpm 培養 80-110 小時而進行。典型地,將培養編號PM0895172 於29°C - 31°C 以及pH 約6.5- 7.5,於210- 230 rpm 培養90-100 小時。
結構式(1)之化合物的生產可經由將培養編號PM0895172 在發酵槽之發酵作用,於27°C至33°C 範圍的溫度以及約5.5至8.5 pH 於 40- 70 rpm 以及200-300 lpm (每分鐘公升數)曝氣下培養20-40小時以進行。典型地,將培養編號PM0895172 於28°C-31°C 以及pH 約6.5- 7.5,於50- 60 rpm 以及240-260 lpm曝氣下培養20-30 小時。
結構式(1)之化合物的生產可經由將培養編號PM0895172 於合適的營養培養液中在本文所描述的條件下培養進行,較佳地在水下含氧的條件下,舉例來說在搖晃的燒瓶中。發酵作用的進展與結構式(1)之化合物的產生可經由高效液相層析法(HPLC)偵測並且經由對不同癌症細胞株測試來測量培養液的生物活性而偵測。
發酵作用為產生各種化學或藥學化合物的生長微生物之過程。正常地將微生物於營養素存在的特定條件下培養。在發酵程過完成之後獲得培養液。將數個培養液進行離心,其造成細胞團塊與培養濾液的生成,可將其進一步地以本文所描述之程序處理。
存在於培養液中的結構式(1)之化合物,可以結構式(1a)之化合物與結構式(1b)之化合物的混合物之形式存在,且可將所述結構式(1a)之化合物或結構式(1b)之化合物使用不同萃取方法與層析技術分離。
可將結構式(1)之化合物特別是結構式(1a)之化合物或結構式(1b)之化合物從培養濾液中以與水不混溶的溶劑例如石油醚、二氯甲烷、氯仿、乙酸乙酯、二乙基醚或丁醇萃取或經由使用聚合樹脂例如Diaion HP-20R(Mitsubishi Chemical Industries Limited, Japan)、Amberlite XADR(Rohm and Haas Industries, USA)或活性碳吸附作用的疏水性交互作用層析(hydrophobic interaction chromatography)而回收。這些技術可重複地、單獨地或結合使用。
可將結構式(1)之化合物特別是結構式(1a)之化合物或結構式(1b)之化合物由細胞團塊以與水混溶的溶劑例如甲醇、丙酮、乙腈、正丙醇或異丙醇或是以與水不混溶的溶劑例如石油醚、二氯甲烷、氯仿、乙酸乙酯或丁醇之萃取作用而回收。
替代地,以選自石油醚、二氯甲烷、氯仿、乙酸乙酯、甲醇、丙酮、乙腈、正丙醇或異丙醇或丁醇的溶劑萃取培養液。
典型地,將結構式(1)之化合物使用乙酸乙酯作為溶劑由整個培養液中萃取。萃取物的濃度提供具的結構式(1a)之化合物與結構式(1b)之化合物的混合物之活性粗製材料。
可將根據本發明之結構式(1)之化合物特別是結構式(1a)之化合物或結構式(1b)之化合物由粗製材料中使用下列技術的任一者之分餾作用而回收:正相層析法(使用氧化鋁或矽膠作為固定相;沖提液例如石油醚、乙酸乙酯、二氯甲烷、丙酮、氯仿、甲醇、異丙醇或其結合物);反相層析法(使用反相矽膠例如二甲基十八烷基甲矽烷矽膠(RP-18)或二甲基辛烷基甲矽烷矽膠(RP-8)作為固定相;以及沖提液例如水、緩衝液(舉例來說,磷酸鹽、乙酸鹽、檸檬酸鹽(pH 2-8)),以及有機溶劑(舉例來說甲醇、乙腈、丙酮、四氫呋喃或這些溶劑的結合);凝膠滲透層析法(使用樹脂Sephadex LH-20R(Pharmacia Chemical Industries, Sweden)、TSKgelRToyopearl HW (TosoHaas, Tosoh Corporation, Japan)於溶劑例如甲醇、氯仿、丙酮、乙酸乙酯或其結合物);或經由逆流層析法(使用雙相沖提液系統,其組成是二或更多溶劑例如水、甲醇、乙醇、異丙醇、正丙醇、四氫呋喃、丙酮、乙腈、二氯甲烷、氯仿、乙酸乙酯、石油醚、苯以及甲苯)。這些技術可重複地、單獨地或結合使用。典型的方法為經由正相矽膠與反相矽膠例如RP-18的層析法。
如同本文所使用的,該用語「同分異構物」為對結構式(1)之化合物的所有同分異構物的通稱,其僅在它們的原子空間方位不同。該用語同分異構物包括鏡像異構物(enantiomer,鏡像異構物)、鏡像異構物的混合物(消旋物、消旋混合物)以及帶有多於一掌性中心,其彼此互相不為鏡像的化合物同分異構物(非鏡像異構物)。本發明之化合物可具有不對稱中心並且以消旋物、消旋混合物、個別非鏡像異構物或鏡像異構物發生,或是可以幾何異構物存在,所述化合物的所有同分異構物形式包含於本發明中。
如同本文所使用的該用語「互變異構物」意指僅在一(或更多)移動原子的位置以及電子分佈與彼此不同的二(或更多)化合物之共同存在,舉例來說,酮-烯醇互變異構物(keto-enol tautomer)。
結構式(1)之化合物、其同分異構物或互變異構物可轉變為其藥學上可接受之鹽類或衍生物全考量於本發明中。
結構式(1)之化合物的藥學上可接受之鹽類經由本領域之技術人員所知的標準程序製備,舉例來說,鹽類例如鈉與鉀鹽可經由把結構式(1)之化合物或其同分異構物或互變異構物或衍生物以合適的鈉或鉀鹼類處理舉例來說氫氧化鈉、氫氧化鉀以及諸如此類而製備。類似地,鹽類例如鹽酸鹽與硫酸鹽,可經由將結構式(1)之化合物或其同分異構物或互變異構物或衍生物以合適的酸類處理,舉例來說以鹽酸、硫酸以及諸如此類而製備。
所有的結構式(1)之化合物的衍生物,特別是結構式(1a)之化合物或結構式(1b)之化合物的衍生物,係包含於本發明的範圍內。根據本發明特別受關注的結構式(1)之化合物的衍生物,特別是結構式(1a)之化合物或結構式(1b)之化合物的衍生物為那些其中衍生胺基基團以及羥基基團中的一或更多者,特別是結構式(1)之化合物的N-羥基基團,特別是結構式(1a)之化合物或結構式(1b)之化合物的N-羥基基團。
因此,在本發明一方面,提供了結構式(1)之化合物的衍生物,其以下列結構式(1c)表示,



結構式(1c)
其中,
R1與R4獨立地代表H或不存在;
R2與R3係獨立地選自H、羥基、-O(C1-C6)烷基以及-OC(O)(C1-C6)烷基;
表示單鍵或雙鍵;
其中,
(C1-C6)烷基為未被取代的或被以一或更多基團取代,該一或更多基團係獨立地選自鹵素、羥基、-O(C1-C6)烷基、硝基、氰基、-COOH、-NH2、-NH(C1-C6)烷基、-N[(C1-C6)烷基]2、-NHC(O)O(C1-C6)烷基、-NHC(O)O(C1-C6)烷基(C6-C10)芳基、-NH-PEG或(C6-C10)芳基;
(C6-C10)芳基為未被取代的或被以一或更多基團取代,該一或更多基團係獨立地選自鹵素、鹵(C1-C6)烷基、羥基、氰基、硝基、 (C1-C6)烷基、-O(C1-C6)烷基、-COOH與-NH2
以及PEG為聚乙二醇,其係選自O,O'-雙[2-(N-琥珀醯亞胺-琥珀醯胺基)乙基]聚乙二醇(α,ω-雙-NHS-PEG)、甲基-PEG-NHS酯(MS(PEG)n,其中n為24),以及分枝的聚乙二醇的三甲基與琥珀醯亞胺酯衍生物(TMS(PEG)n,其中n為12);
並規定若R2與R3兩者為羥基,那麼R1與R4為不存在的並且表示雙鍵;
以及其所有同分異構與互變異構的形式或藥學上可接受之鹽類。
將理解「取代(substitution)」、「被取代的(substituted)」或「以…取代(substituted with)」表示指定基元的一或更多氫原子被以合適的取代基取代並且包括隱含條件為此類取代係根據被取代原子與取代基的容許性價,並且生成穩定的化合物。
該用語「(C1-C6)烷基」或「烷基」,如同本文單獨或為另一基團的部分使用的,意指包含1至6碳之未被取代的或被取代的直或分枝鏈碳氫化合物,例如甲基、乙基、丙基、異丙基、丁基、叔丁基、異丁基、戊基與己基。被取代的烷基意指被一或更多基團取代的(C1-C6)烷基,該基團係選自但不限於鹵素、羥基、-O(C1-C6)烷基、硝基、氰基、-COOH、-NH2、-N[(C1-C6)烷基]2、-NHC(O)O(C1-C6)烷基、-NHC(O)O(C1-C6)烷基(C6-C10)芳基、-NH-PEG或(C6-C10)芳基。
如同本文所使用的,該用語「(C6-C10)芳基」或「芳基(aryl)」意指具有高達十環碳原子的單環或雙環碳氫環系統,其中至少一碳環之環具有p電子系統。(C6-C10)芳基環系統的範例包括但不限於苯基或萘基。除非另外指明,芳基基團可為未被取代的或被以一或更多基團所取代,該一或更多基團係獨立地選自但不限於鹵素、鹵(C1-C6)烷基、羥基、氰基、硝基、、-O(C1-C6)烷基、-COOH與 -NH2
如同本文所使用的,該用語「-O(C1-C6)烷基」或「烷氧基」意指未被取代的或被取代的 (C1-C6)烷基自由基,其具有氧原子連結其上。代表性的烷氧基基團包括但不限於甲氧基、乙氧基、正丙氧基、1-甲基乙氧基、正丁氧基、1-甲基丙氧基、2-甲基丙氧基或1,1-二甲基乙氧基。被取代的烷氧基基團意指-O(C1-C6)烷基基團,其中烷基被以一或更多基團取代,該一或更多基團係選自但不限於鹵素、羥基、-O(C1-C6)烷基、硝基、氰基、-COOH、-NH2、-NH(C1-C6)烷基、-N[(C1-C6)烷基]2、-NHC(O)O(C1-C6)烷基、-NHC(O)O(C1-C6)烷基(C6-C10)芳基、-NH-PEG或(C6-C10)芳基。
如同本文所使用的,該用語「鹵(C1-C6)烷基」或「鹵烷基」意指自由基,其中烷基基團之氫原子的一或更多者被以一或更多鹵素取代。單鹵烷基自由基,舉例來說,可具有一氯、溴、碘或氟原子。二鹵與多鹵烷基自由基可具有二或更多相同或不同的鹵素原子。「鹵(C1-C6)烷基」或「鹵烷基」的範例包括但不限於氯甲基、二氯甲基、三氯甲基、二氯乙基、二氯丙基、氟甲基、二氟甲基、三氟甲基、五氟乙基、七氟丙基、二氟氯甲基、二氯氟甲基、二氟乙基或二氟丙基。
如同本文所使用的,該用語「鹵素」意指氟、氯、溴與碘。
如同本文所使用的,該用語「PEG」意指聚乙二醇聚合物。PEG為無毒性的、無免疫原性、無抗原性並且在水中高度可溶。多種不同分子量的PEG為商業上可得的,其係根據分子內環氧乙烷(即-OCH2CH2-)的重覆單元之數目。舉例來說,PEG可得自商業供應商例如Sigma-Aldrich Co. LLC或NOF公司。PEG的一些範例為但不限於:(a) O,O'-雙[2-(N-琥珀醯亞胺-琥珀醯胺基)乙基]聚乙二醇(α,ω-雙-NHS-PEG),分子量在2000 Da 至10,000 Da的範圍,將其結構性地表示於下:


(b) 甲基-PEG-NHS酯(MS(PEG)n,其中n為24),分子量1214.39 Da,其結構性地表示於下;

以及;
(c)聚乙二醇的分枝的三甲基與琥珀醯亞胺酯類衍生物(TMS(PEG)n,其中n為12、分子量2420.80 Da)。典型地, PEG 具有1000至20,000 Da範圍之分子量,其較佳地用於如同本文所描述之化合物的PEG共軛物的製備。
在本發明的上下文中,該用語「結構式(1c)之化合物」、「結構式(1)之化合物的衍生物」或「結構式(1c)之衍生物係交替地使用,且這些用語的每個意指由結構式(1c)表示的結構式(1)之化合物;以及其同分異構物、互變異構物或藥學上可接受之鹽類的衍生物。
根據一具體實施例,本發明提供結構式(1c)之化合物,
其中,
R1與 R4為H;
R2為-O(C1-C6)烷基或-OC(O)(C1-C6)烷基;
R3為羥基或-O(C1-C6)烷基;
其中,(C1-C6)烷基為未被取代的或被以-NH2、-NHC(O)O(C1-C6)烷基(C6-C10)芳基或-NH-PEG取代;
以及其所有同分異構或互變異構形式或藥學上可接受的鹽類。
根據另一具體實施例,本發明提供結構式(1c)之化合物,
其中,
R1與R4獨立地表示 H或不存在;
R2與R3係獨立地選自H與羥基;
表示單鍵或雙鍵;
並規定若R2與R3兩者為羥基,那麼R1與 R4為不存在的並且表示雙鍵;
以及其所有同分異構或互變異構形式或藥學上可接受的鹽類。
根據本發明包含的代表性結構式(1c)之化合物包括:









以及其所有同分異構或互變異構形式或藥學上可接受的鹽類。
結構式(1)之化合物的衍生物,其以結構式 (1c)表示,可由本領域所知的方法製備。在結構式 (1c)之化合物製備過程中採用的試劑可為商業上可得的或可由本領域所知的程序製備。
R1與 R4為H;且R2與R3係獨立地選自羥基與-O(C1-C6)烷基,其中R2與 R3中的至少一者為-O(C1-C6)烷基的結構式 (1c)之化合物可經由將結構式(1)之化合物反應而製備,其為結構式(1a)之化合物在選自甲醇、乙醇、丙醇或丁醇的醇類中,結合甲苯或苯,伴隨在有機溶劑例如己烷或甲苯中的烷基化試劑例如三甲基甲矽烷重氮甲烷,於-5 °C至5°C的溫度範圍反應1 至5小時。化合物1與2係以此方法製備。
R1與 R4為H;且R2與R3係獨立地選自羥基與-OC(O)(C1-C6)烷基而使得R2與R3的至少一者為-OC(O)(C1-C6)烷基;其中(C1-C6)烷基被以-NHC(O)O(C1-C6)烷基(C6-C10)芳基所取代的結構式 (1c)之化合物可經由將結構式(1)之化合物反應而製備,其為將結構式(1a)之化合物,在溶劑例如四氫呋喃(THF)、二氯甲烷(DCM)或二甲基甲醯胺(DMF),伴隨試劑例如N-芐氧羰基-L-丙胺酸(Cbz丙胺酸)或N-​(9-​茀基甲氧羰基)​-L-丙胺酸(Fmoc丙胺酸),在耦合劑的存在下例如苯并三唑-1-基-氧三吡咯烷鏻六氟磷酸(PyPOP)、(苯并三唑-1-基氧基)三(二甲基胺基) 鏻六氟磷酸酯(BOP)、N, N'-二環己基碳二亞胺(DCC)或丙烷膦酸酐(T3P),以及鹼類例如N,N-二異丙基乙胺(DIPEA)、三乙基胺或4-二甲基胺基吡啶(DMAP),於-5 °C至5°C的溫度範圍反應1 至20小時。化合物3係以此方法製備。因此獲得的化合物可經由使用催化劑例如Pd/C或Pt/C,在溶劑例如甲醇、乙醇、乙酸乙酯或THF中,以使用酸類例如稀釋HCl創造的酸性環境中的催化還原作用而被去保護,以獲得結構式(1c)之化合物,其中 與R3係獨立地選自羥基與-OC(O)(C1-C6)烷基,以使得R2與R3的至少一者為-OC(O)(C1-C6)烷基;其中該(C1-C6)烷基被-NH2所取代。化合物4係以此方法製備。
R1與 R4為H;且R2與R3係獨立地選自羥基與-OC(O)(C1-C6)烷基,以使得R2與R3的至少一者為 -OC(O)(C1-C6)烷基;其中該(C1-C6)烷基被-NH2所取代之結構式 (1c)之化合物可用於聚乙二醇作用,其經由將所述化合物溶解於有機溶劑例如DCM、甲醇或DMF中,在鹼類例如三乙基胺或DIPEA的存在下,並加入聚乙二醇(PEG)聚合物例如O,O'-雙[2-(N-琥珀醯亞胺-琥珀醯胺基)乙基]聚乙二醇(α,ω-雙-NHS-PEG)或甲基-PEG-NHS酯,並且於25 °C至35°C的溫度範圍下攪拌10-20小時,以獲得結構式(1c)之化合物其中R2與 R3係獨立地選自羥基與-OC(O)(C1-C6)烷基,以使得R2與R3的至少一者為-OC(O)(C1-C6)烷基;其中該(C1-C6)烷基被-NH-PEG所取代。化合物8係以此方法製備。
可將結構式(1)之化合物氧化以移除在R1與R4位置的氫,其經由使用在選自二氯甲烷或二氯乙烷的有機溶劑中的試劑例如間氯過氧苯甲酸,於25 °C至35°C的溫度範圍下10-20小時,以獲得結構式(1c)之化合物其中R1與 R4為不存在,並且R2與 R3為羥基。化合物5係以此方法製備。
可將結構式(1)之化合物的N-羥基基團轉變為NH(脫氧作用),其經由將在選自甲醇、乙醇或DMF的有機溶劑中的結構式(1)之化合物加至在溶劑例如THF、DCM或DMF中的溶液試劑例如雙(環戊二烯)鈦(IV)二氯化物以及活化鋅,並且於-5 °C至-50°C的溫度範圍攪拌0.1 hr至1 hr,以獲得結構式(1c)之化合物其中R1與 R4為H或不存在,並且R2與 R3為H。化合物6係以此方法製備。
可將結構式(1)之化合物的N-羥基基團之醯基衍生物經由將結構式(1)之化合物在溶劑例如二氯甲烷、嘧啶或二氯乙烷中,伴隨試劑例如乙醯氯或乙酸酐,在鹼類的存在下例如嘧啶、TEA或DMAP,於-5 °C至30°C的溫度範圍反應1 hr至5 hr而製備,以獲得結構式(1c)之化合物,其中R2與 R3係獨立地選自羥基與-OC(O)(C1-C6)烷基,以使得R2與R3的至少一者為-OC(O)(C1-C6)烷基。化合物7係以此方法製備。
可將為結構式(1)化合物之衍生物的結構式(1c)之化合物隨選地轉變為其藥學上可接受之鹽類。
結構式(1)之化合物或其藥學上可接受之鹽類或結構式(1c)之衍生物具有抗癌活性。此已經由對廣泛的癌細胞分別測試本發明之代表性的化合物,包括結構式(1)、結構式(1a) 、結構式(1b) 與結構式(1c)之化合物而證實。
可將結構式(1)之化合物或其同分異構物或互變異構物或藥學上可接受之鹽類或結構式(1c)之衍生物作為藥物並且以藥學組成物的形式給藥予對其有需求的病患。可將結構式(1)之化合物或其同分異構物或互變異構物或藥學上可接受之鹽類或結構式(1c)之衍生物給藥予診斷具癌症的病患。
因此,本發明亦相關於結構式(1)之化合物特別是結構式(1a)之化合物或結構式(1b)之化合物或其同分異構物或互變異構物或藥學上可接受之鹽類或結構式(1c)之衍生物供癌症治療藥劑之製造的用途。
本發明進一步地相關於包含治療有效量之結構式(1)之化合物或其同分異構物或互變異構物或藥學上可接受之鹽類或結構式(1c)之衍生物,以及藥學上可接受之賦形劑或載體的藥學組成物。該藥學組成物係提供於癌症之治療。作為藥學製劑活性成分的治療有效量之結構式(1)之化合物或其立體異構物或其互變異構物或其藥學上可接受之鹽類或其結構式(1c)之衍生物可為大約0.01 mg至1000 mg的範圍;或可為大約0.1 mg至750 mg的範圍;或可為大約0.5 mg至500 mg的範圍;或可為大約1 mg至250 mg的範圍。
本發明之化合物,其包括結構式(1)之化合物或其同分異構物或互變異構物或藥學上可接受之鹽類或結構式(1c)之衍生物的發現在癌症治療的用途。本發明之化合物係用以降低、抑制或減少腫瘤細胞的增生,並且可提供腫瘤尺寸的縮小。可以本發明之化合物治療的代表性癌症包括但不限於白血病、肺癌、腦癌、非霍奇金氏淋巴瘤、霍奇金氏病、肝癌、腎臟癌、膀胱癌、泌尿道癌、乳癌、頭頸部癌、子宮內膜癌、淋巴瘤、黑色素瘤、子宮頸癌、甲狀腺癌、胃癌、生殖細胞瘤、膽管癌、顱外癌、肉瘤、間皮瘤、骨惡性纖維組織細胞瘤、視網膜母細胞瘤、食道癌、多發性骨髓瘤、胰腺癌、室管膜瘤、神經母細胞瘤、皮膚癌、卵巢癌、復發性卵巢癌、前列腺癌、睪丸癌、大腸癌、淋巴增生性疾病、難治性多發性骨髓瘤、抗多發性骨髓瘤或骨髓增生性疾病。
根據本發明的一具體實施例,癌症係選自急性淋巴性白血病、急性骨髓性白血病、成人急性骨髓性白血病、急性淋巴胚細胞白血病、慢性淋巴性白血病、慢性骨髓性白血病、毛細胞白血病、非小細胞肺癌、小細胞肺癌、腦幹神經膠瘤、神經膠母細胞瘤、包括小腦心臟細胞瘤與大腦心臟細胞瘤的心臟細胞瘤、視覺通路神經膠瘤、丘腦神經膠瘤、小腦幕上原始神經外胚層的、松果體瘤、髓母細胞瘤、原發性中樞神經系統淋巴瘤、套膜細胞淋巴瘤、霍奇金氏病、肝細胞癌、腎細胞癌、胚性癌肉瘤(Wilms' tumor)、膀胱癌、泌尿道癌、Ewing’s 肉瘤腫瘤家族、骨肉瘤、橫紋肌肉瘤、軟組織肉瘤、間皮瘤、乳癌、子宮內膜癌、口腔癌、黑色素瘤、子宮頸癌、甲狀腺癌、胃癌、生殖細胞瘤、膽管癌、顱外癌、骨惡性纖維組織細胞瘤、視網膜母細胞瘤、食道癌、多發性骨髓瘤、胰腺癌、室管膜瘤、神經母細胞瘤、皮膚癌、卵巢癌、復發性卵巢癌、前列腺癌、睪丸癌、大腸癌、淋巴增生性疾病、難治性多發性骨髓瘤、抗多發性骨髓瘤或骨髓增生性疾病等等。
根據本發明的另一具體實施例,癌症係選自急性淋巴性白血病、急性骨髓性白血病、成人急性骨髓性白血病、急性淋巴胚細胞白血病、慢性淋巴性白血病、慢性骨髓性白血病、毛細胞白血病、非小細胞肺癌、小細胞肺癌、腦幹神經膠瘤、神經膠母細胞瘤、包括小腦心臟細胞瘤與大腦心臟細胞瘤的心臟細胞瘤、視覺通路神經膠瘤、松果體瘤、髓母細胞瘤、原發性中樞神經系統淋巴瘤、套膜細胞淋巴瘤、霍奇金氏病、肝細胞癌、腎細胞癌、膀胱癌、泌尿道癌、骨肉瘤、乳癌、子宮內膜癌、口腔癌、黑色素瘤、子宮頸癌、甲狀腺癌、胃癌、骨惡性纖維組織細胞瘤、視網膜母細胞瘤、食道癌、多發性骨髓瘤、胰腺癌、神經母細胞瘤、皮膚癌、卵巢癌、前列腺癌、睪丸癌、大腸癌、淋巴增生性疾病或骨髓增生性疾病等等。
根據本發明的進一步具體實施例,癌症係選自急性淋巴性白血病、急性骨髓性白血病、成人急性骨髓性白血病、急性淋巴胚細胞白血病、慢性淋巴性白血病、慢性骨髓性白血病、毛細胞白血病、非小細胞肺癌、小細胞肺癌、腦幹神經膠瘤、神經膠母細胞瘤、包括小腦心臟細胞瘤與大腦心臟細胞瘤的心臟細胞瘤、髓母細胞瘤、腎細胞癌、膀胱癌、泌尿道癌、乳癌、口腔癌、黑色素瘤、子宮頸癌、甲狀腺癌、胃癌、胰腺癌、前列腺癌或大腸癌等等。
根據一具體實施例,本發明提供病患癌症治療的方法,其係經由給藥予病患治療有效量之結構式(1)之化合物或其結構式(1c)之衍生物或同分異構物或互變異構物或藥學上可接受之鹽類。
根據另一具體實施例,本發明提供病患癌症治療的方法,其係經由給藥予病患治療有效量之結構式(1a)之化合物或結構式(1b)之化合物或其同分異構物或互變異構物或藥學上可接受之鹽類。
根據又一具體實施例,本發明提供病患癌症治療的方法,其係經由給藥予病患治療有效量之結構式(1c)之衍生物或其同分異構物或互變異構物或藥學上可接受之鹽類。
結構式(1)之化合物或其藥學上可接受之鹽類或結構式(1c)之衍生物;或其同分異構物或互變異構物或藥學上可接受之鹽類可以口服地、鼻吸地、局部地、皮下注射地、肌肉注射地、靜脈注射地或以其他給藥模式給藥。
在適用於特定情況的給藥方法視待治療癌症的類型與癌症的階段而定。進一步地,給藥方法可由醫師使用本領域已知的方法優化。
按照慣例,適用於特定情況的劑量範圍視要治療的癌症類型以及個別症狀或疾病的狀態而定。選擇的劑量程度可由熟練的醫師根據相關的情況,包括待治療的症狀(癌症)、依數個因素例如個別病患的年齡、體重與身體健康及反應、藥代動力學、疾病的嚴重程度等醫學領域所知的因素而定所選擇的給藥途徑而立即地確定。本發明之藥學組成物中的活性成分實際劑量可變化,以獲得有效達到對特定病患(需要治療的病患)之合意治療反應的活性成分之量、組合物以及對病患無毒性的給藥模式。平均而言,病患的活性化合物(本發明化合物中的一或更多者)的每日劑量可為大約每公斤0.05 mg至約500 mg或為大約每公斤0.5 mg至約200 mg或為大約每公斤1 mg至約100 mg的範圍,或是落在如同本文指明的較寬劑量範圍的範圍內之任何劑量範圍。在需要治療的較高體重病患的情況中,活性化合物的劑量可為大約每公斤1 mg至約1000 mg或為大約每公斤5 mg至約500 mg的範圍。活性化合物,即本發明之化合物的所需劑量可在很寬的範圍內選擇。選擇給藥的每日劑量以達到在接受癌症治療的病患之合意治療效果。如有需要,亦可給藥較高或較低的每日劑量。
包含結構式(1)之化合物或其同分異構物或互變異構物或藥學上可接受之鹽類或結構式(1c)之衍生物的藥學組成物,其伴隨其他藥學上可接受之賦形劑例如濕潤劑、增溶劑例如表面活性劑、媒液、等滲劑、填料、著色劑、掩蔽香料、潤滑劑、崩解劑、稀釋劑、粘合劑、增塑劑、乳化劑、軟膏基底、潤膚劑、增稠劑、聚合物、脂質、油、助溶劑、錯合劑或緩衝物質而為片劑、包衣片劑、膠囊、顆粒、粉末、乳霜、軟膏、凝膠、糖漿、乳液、懸浮液或適合用於腸外給藥的溶液的形式。

在本發明的一具體實施例中,包含結構式(1)之化合物或結構式(1c)之衍生物;或其同分異構物或互變異構物、藥學上可接受之鹽類的藥學組成物可用於與一或更多抗癌劑例如天冬醯胺酶(asparaginase)、博萊黴素(bleomycin)、卡鉑(carboplatin)、卡莫司汀(carmustine)、瘤克寧(chlorambucil)、順鉑(cisplatin)、天冬醯胺酶(colaspase)、癌德星錠(cyclophosphamide)、阿糖胞苷、達卡巴嗪、放線菌素D、道諾黴素、多柔比星、表柔比星、依託泊苷(etoposide)、氟脲嘧啶、六甲基蜜胺、羥基尿素(hydroxyurea)、異環磷醯胺、甲醯四氫葉酸(leucovorin)、洛莫司汀(lomustine)、雙氯乙基甲胺(mechlorethamine)、6-巰基嘌呤(6-mercaptopurine)、美司那(mesna)、胺甲蝶呤(methotrexate)、絲裂黴素C、米托蒽醌(mitoxantrone)、氫潑尼松(prednisolone)、潑尼松(prednisone)、丙卡巴肼(procarbazine)、鏈脲佐菌素(streptozocin)、他莫昔芬(tamoxifen)、硫代鳥嘌呤(thioguanine)、長春鹼(vinblastine)、長春新鹼(vincristine)、長春地辛(vindesine)、氨魯米特(aminoglutethimide)、5-氮雜胞苷(5-azacytidine)、克拉曲濱(cladribine)、白消安(busulfan)、己烯雌酚(diethylstilbestrol)、2',2'-二氟去氧胞苷、多烯紫杉醇(docetaxel)、紅-9-(2-羥基-3-壬基)腺嘌呤、乙炔雌二醇(ethinyl estradiol)、5-氟去氧尿苷(5-fluorodeoxyuridine)、5-氟去氧尿苷單磷酸鹽、氟達拉濱(fludarabine phosphate)、氟甲睪酮(fluoxymesterone)、氟他胺(flutamide)、羥基孕酮己酸酯(hydroxyprogesterone caproate)、艾達黴素(idarubicin)、干擾素、甲基乙醯氧孕前酮醋酸鹽(medroxyprogesterone acetate)、甲地孕酮醋酸鹽(megestrol acetate)、左旋溶肉瘤素(melphalan)、米托坦(mitotane)、太平洋紫杉醇(paclitaxel)、噴司他丁(pentostatin)、N-亞膦羧基乙醯基-L-天冬胺酸酯(PALA)、光輝黴素(plicamycin)、替尼泊苷(teniposide)、丙酸睪固酮、硫替帕(thiotepa)、三甲基蜜胺、尿苷、長春瑞濱(vinorelbine)、alsterpaullone、丁內酯I(butyrolactone I)、2-(2-羥乙基胺基)-6-(3-氯苯胺基)-9-異丙基嘌呤、靛玉紅-3'-單肟、kenpaullone、奧羅莫新(olomoucine)、異奧羅莫新、N9-異丙基-奧羅莫新、普伐蘭諾A(purvalanol A)、羅斯維汀(roscovitine)、(S)-同分異構物羅斯維汀以及WHI-P180 [4-(3’-羥苯基)胺基-6,7-二甲氧基喹唑啉]結合;用於如同本文所描述的癌症治療。
要了解不實質地影響本發明的各種具體實施例的活性的修改係包含於本文揭露之發明的範圍內。因此,下列範例為意圖說明但不限制本發明的範圍。

範例
在範例中採用下列用語/符號/縮寫/化學式:


範例1
來自海生來源的培養編號PM0895172 之分離
a)分離培養基組成物:
1% 蛋白腖、2% 葡萄糖、0.2% 碳酸鈣、0.001%氯化鈷六水合物、1.5% 瓊脂、pH(於25°C)7.5。
人造海水(artificial seawater,ASW)組成物:2.46% 氯化鈉、0.067% 氯化鉀、0.136%氯化鈣、0.629% 硫酸鎂、0.466%氯化鎂、0.018% 碳酸氫鈉;水。
b)程序
從印度馬哈拉施特拉邦,孟買的近海區域收集深海沉積物,並且在送至印度孟買,戈勒貢(East)Piramal Enterprises Limited(Formerly Piramal Healthcare Limited)的NCE研究部門(Piramal LifeSciences division)的旅途中於-20°C儲存。
對從收集的沉積物樣本中之未培養海生放射菌分離實施分子生物學方法。對於此,基因體DNA從沉積物樣本中分離並且針對聚酮合成酶(PKS)與非核糖體肽合成酶(NRPS)的存在而擴增。再將所關注培養物自具有NRPS途徑的沉積物樣本中分離。
將樣本儲存於-20°C至22°C並且稍後解凍至室溫(25±2°C)以供微生物分離。將土壤樣本(~2 g)於熱壓處理的臼與杵下懸浮於25 ml 無菌人工海水(ASW),並且將其徹底粉碎。將1 ml的此懸浮液轉移至試管中並且將其渦旋震盪30秒。將試管渦旋震盪30秒。於無菌ASW中製備達10-5的連續稀釋。將100 μl 的10-3之稀釋被表面塗布於包含分離培養基的盤中。將盤於室溫(25±2°C)下培養直到觀察到菌落。在20-22日的培養後,將出現在此培養基的菌落於包含分離培養基的培養皿劃線(如同上述提到的)。分離株被純化並提供培養ID編號PM0895172。從而將培養編號PM0895172從生長的微生物之間分離為單一分離株。

範例2
培養編號PM0895172的純化
a)純化培養基組成物:
1% 蛋白腖、2% 葡萄糖、0.2%碳酸鈣、0.001% 氯化鈷六水合物、1.5%瓊脂;pH (於25°C)7.5。
b)程序:將培養編號PM0895172於包含純化培養基的培養皿劃線。將培養皿於27°C下培養十天。將來自培養皿的一個分離菌落轉移至包含ISP瓊脂(HiMedia, India)的新鮮斜面培養基。將斜面培養基於27°C下培養十天。

範例3
生產者菌株的維持—培養編號PM0895172
將此特定菌株維持於包含ISP4培養基(HiMedia, India)的斜面培養基。在經由加熱溶解培養基之後,將生成的溶液分佈於試管並於121°C滅菌30分鐘。將試管冷卻並使其於傾斜的位置固化。將瓊脂傾斜培養基以生長良好的培養編號PM0895172以金屬線環劃線並且於27°C至29°C培養直到觀察到良好生長。將該良好生長的培養株儲存於4°C至8°C的冰箱中。

範例4
在震盪燒瓶中的培養編號PM0895172發酵作用
a)種培養基(seed medium)組成物:
0.2%碳酸鈣、0.5%氯化鈉、0.5%玉米漿、0.75% 蛋白腖、1.5 % 葡萄糖、0.75% 酵母萃取物;軟化水;pH 7。
b)程序:
將50 ml的上述種培養基分佈於容量500 ml的錐形瓶中並且於121°C 滅菌30分鐘。將錐形瓶冷卻至室溫(25±2°C)並將每個錐形瓶以一環量的生長良好之生產株(培養編號PM895172)接種於斜面並且以旋轉震盪器於30°C(±1°C)以230 rpm至250 rpm 震盪72小時,以提供種培養株。
c)生產培養基組成物:
1% 蛋白腖、2% 葡萄糖、0.2 %碳酸鈣、0.001 % 氯化鈷六水合物、1.5 %瓊脂以及pH (於25°C)7.5。
d)程序:
將在容量500 ml錐形瓶中的100 ml的生產培養基於121°C 滅菌30分鐘,將其冷卻至29°C至30°C並且以3%(v/v)在範例4b中提及的種培養株種植。
e)發酵作用參數:
將生產燒瓶在震盪器上於30°C與220 rpm培養96小時。將生產燒瓶收穫(收穫pH:7.0至8.0)並且將整個培養液以相同體積的溶劑混合物(甲醇:乙酸乙酯(1:9))自每個培養基燒瓶中萃取。將這些收穫的燒瓶維持於室溫下4-6小時以供上清液的分離後之萃取。使用上清液以供測試抗癌活性。

範例 5發酵槽批次的震盪燒瓶中之種培養株的製備
a) 種培養基274(1)組成物:

葡萄糖 15 g、玉米漿 5 g、蛋白腖 7.5 g、酵母萃取物7.5 g、碳酸鈣2 g、氯化鈉5 g、軟化水1.0 L、pH 6.5-7.5(滅菌前)。
b) 程序:
將200 ml的上述培養基分佈於1000 ml錐形瓶並且於121°C滅菌30分鐘。將錐形瓶冷卻至室溫,並且將每個錐形瓶以一環量的生長良好之生產株(PM0895172)接種於斜面並且以旋轉震盪器於29°C -30°C以230 -250 rpm 震盪70-74小時,以獲得種培養株。

範例6
在發酵槽中的培養編號PM0895172培養
a) 生產培養基之組成物:
大豆蛋白腖 10 g、葡萄糖 20 g、碳酸鈣2g、氯化鈷0.001 g、軟化水1.0 L,pH 7.0(滅菌前)。
b) 程序:
將500 L的生產培養基於1000 L發酵槽伴隨作為消泡劑的200 ml desmophen 於121°C下原位滅菌20 分鐘,冷卻至29°C-30°C並且以8-10 L在範例5b中獲得的種培養株種植。
c) 發酵作用參數:
溫度 29°C - 30°C、尖端速率(tip speed)0.94m/s、曝氣250 lpm以及收穫時間24 hr。培養液的收穫pH為6.5-7.5。將在發酵液中具活性的結構式(1a)之化合物或結構式(1b)之化合物的生產經由HPLC測定,並且針對抗癌活性測試生物活性。
範例7      
結構式(1a)之化合物的分離與特徵描述
將在範例6的步驟c)中收穫的全培養液(500 L)使用乙酸乙酯(500 L)萃取。使用疊盤式分離器(Alfa Laval(USA),型號LAPX404)分離有機層並濃縮,以獲得粗製之萃取物(150 g)。

將粗製的萃取物經由管柱層析法(矽膠,溶劑:異丙醇/氯仿)處理。將活性化合物以在氯仿中的1-2%異丙醇沖提,將其濃縮以獲得增濃的化合物(30 g)。
將此材料經由管柱層析純化(RP C-18矽膠,溶劑:水/ 乙腈)。將得到的活性化合物以水中之40%乙腈沖提,將其蒸發以獲得2 g 所需的半純的具活性結構式(1)之化合物(結構式(1a)與結構式(1b)之化合物的混合物)。
如同下述經由反相製備型HPLC(reversed phase preparative HPLC)進行進一步的純化:
管柱:Water’s X-Bridge RP-18 (250 mm X 19 mm, 10 μ)
沖提液:乙腈:水(50:50).
流速:25 ml/min
偵測(UV):220 nm
將收集的活性波峰蒸發至乾燥,以獲得1 g活性化合物(結構式(1a)之化合物)。
如同下述經由矽膠製備型HPLC進行活性化合物的最終純化:
管柱:Water’s sunfire silica, (150 mm X 19 mm, 5 μ)
沖提液:氯仿:甲醇(98.5:1.5)
流速:15 ml/min
偵測(UV):240 nm
Rt 時間: 8-9 min。
將來自重複注入之包含活性化合物的沖提物濃縮,以獲得145 mg的純結構式(1a)之化合物。結構式(1a)之化合物的物理與光譜特性係提供於表格1。
分析HPLC條件係如同下述:
管柱:Kromasil RP-18 (150 mm X 4.6 mm, 3.5 μ)
沖提液:梯度(於20分鐘的10% 乙腈至100% 乙腈對水,接著額外的5分鐘以100% 乙腈)
流速:1 ml/min
偵測(UV):220 nm
停留時間:17-18 min (純度 > 99 %)
The present invention provides a compound of the formula (1) as shown below:



Structural formula (1)
Wherein in the compound of formula (1), when the bondRepresents a single button and R1Is H; a compound is referred to as a compound of formula (1a);
When the keyIs a double bond and R1When not present, the compound is referred to as a compound of formula (1b). Thus, the compound of formula (1) is a compound of formula (1a) wherein the bondRepresents a single button and R1a compound of H) or structural formula (1b) (wherein the bondIs a double bond and R1Not present) or a mixture thereof.
The compound of the formula (1) belongs to a hexaester peptide compound.
The compound of formula (1a) and the compound of formula (1b) can be characterized by any one or more of physico-chemical and spectral properties, such as high performance liquid chromatography (HPLC), high resolution as discussed herein below. Mass spectrometry (HR MS), far infrared (IR), and nuclear magnetic resonance (NMR) spectral data.
According to one aspect of the invention, the compound of formula (1) is a compound of formula (1a) having the formula C37H62N8O13And the molecular weight of 826.9.
The compound of the formula (1a) is expressed as follows:



Structural formula (1a)
The chemical name of the compound of formula (1a) is N-(6,18-dihydroxy-22-isopropyl-7,19-dimethyl-5,8,11,17,20,24-hexa-oxa Tetrahydrodioxazino[6,1-f:6',1'-o][1,4,7,10,13,16]pentazaoxacyclohexadecandin-23-yl)-2 - Hydroxy-2-(2-hydroxy-5-isobutyl-6-methyltetrahydro-2H-pyran-2-yl)propanamine.
According to one aspect of the invention, the compound of formula (1) is a compound of formula (1b) having the formula C37H60N8O13, and the molecular weight of 824.4.
The compound of the formula (1b) is represented as follows:



Structural formula (1b)
The chemical name of the compound of formula (1b) is N-(6,18-dihydroxy-22-isopropyl-7,19-dimethyl-5,8,11,17,20,24-hexaoxy- 3,4,4a,5,6,7,8,9,10,11,13,14,15,16,16a, 17,18,19,20,22,23,24-tetrahydrofuran Pyrazino[6,1-f:6',1'-o][1,4,7,10,13,16]pentazaoxacyclohexenyne-23-yl)-2-hydroxy-2- (2-Hydroxy-5-isobutyl-6-methyltetrahydro-2H-pyran-2-yl)propanamine.
The microorganism which can be used for the production of the compound of the formula (1) is a radioactive strain (PM0895172/MTCC 5684), hereinafter referred to as the number PM0895172, which is collected from the deep sea of the offshore area of Mumbai, Maharashtra, India. Separation in sediments.
An aspect of the invention provides a production procedure for a compound of formula (1) from culture number PM0895172, comprising the steps of:
a) cultivating one of the culture numbers PM0895172 or one of its variants or mutants under water aerobic conditions, comprising one or more sources of carbon and one or more sources of nitrogen and nutritive inorganic salts and/or traces Growing in a nutrient medium of the element to obtain a culture solution containing the compound of the formula (1);
b) separating the compound of formula (1) from the culture broth;
c) Purification of the compound of formula (1).
The compound of the formula (1) produced according to the above procedure may be a compound of the formula (1a) or a compound of the formula (1b).
In a specific embodiment, the culture solution in step (a) comprises a mixture of a compound of formula (1a) and a compound of formula (1b).

In a specific embodiment, in step (b) of the procedure, the compound of formula (1) is a compound of formula (1a).
In a specific embodiment, in step (b) of the procedure, the compound of formula (1) is a compound of formula (1b).
In a specific embodiment, in step (c) of the procedure, the compound of formula (1) is a compound of formula (1a).
In a specific embodiment, in step (c) of the procedure, the compound of formula (1) is a compound of formula (1b).
In a specific embodiment, the invention provides a production procedure for a compound of formula (1a) from culture number PM0895172, comprising the steps of:
a) cultivating one of the culture numbers PM0895172 or one of its variants or mutants under water aerobic conditions, comprising one or more sources of carbon and one or more sources of nitrogen and nutritive inorganic salts and/or traces Growing in a nutrient medium of the element to obtain a mixture culture solution comprising the compound of the formula (1a) and the compound of the formula (1b);
b) separating the compound of formula (1a) from the culture broth;
c) Purification of the compound of formula (1a).
In another embodiment, the invention provides a production procedure for a compound of formula (1b) from culture number PM0895172, comprising the steps of:
a) cultivating one of the culture numbers PM0895172 or one of its variants or mutants under water aerobic conditions, comprising one or more sources of carbon and one or more sources of nitrogen and nutritive inorganic salts and/or traces Growing in a nutrient medium of the element to obtain a mixture culture solution comprising the compound of the formula (1a) and the compound of the formula (1b);
b) separating the compound of formula (1b) from the culture broth;
c) Purification of the compound of formula (1b).
Step (c) of any of the above procedures relates to the purification of the compound of the formula (1), and is carried out by using a purification procedure generally used in the related art.
The compound of the formula (1), in particular the compound of the formula (1a) and the compound of the formula (1b) which are produced according to the procedure of the present invention, are substantially pure compounds. Thus, the compound of formula (1), in particular the compound of formula (1a) or the compound of formula (1b), is an isolated pure compound.
Typically, the compound of formula (1a) and the compound of formula (1b) are purified via column chromatography techniques.
As used herein, the term "mutant" means an organism or a cell in which one or more genes in the genome have been altered by one or more genes responsible for the organism's production. The ability of the compounds of the invention. Such mutant strains can be produced in a manner known per se using physical means such as ultraviolet or X-ray irradiation or chemical mutagen.
As used herein, the term "variant" means an individual organism that recognizes any standard type different from that species.
The term "whole broth" can be used interchangeably with the terms "nutrient broth", "culture broth" or "fermented broth".
As used herein, the term "mammal" means both human and non-human mammals including, but not limited to, cows, horses, pigs, dogs, and cats. The term "mammal" can be used interchangeably with the term "patient" or "subject".
The phrase "active ingredient" or "active compound" may be used interchangeably, and as used herein, the term means a compound of formula (1) or a compound of formula (1a). Or a compound of formula (1b) or an isomer or tautomer thereof, or a pharmaceutically acceptable salt or derivative thereof. In the context of the present invention, the term "active ingredient" or "active compound" also means a compound of formula (1c) which comprises a derivative of a compound of formula (1) as described herein.
The phrase "compound of the formula (1)" includes a compound of the formula (1a) or a compound of the formula (1b) or a mixture thereof; and an isomer, tautomer or pharmaceutically acceptable salt thereof. class.
The term "compound of the formula (1c)" includes a derivative of the compound of the formula (1), which comprises a compound of the formula (1a) or a compound of the formula (1b) or an isomer thereof, tautomer A derivative of a construct or a pharmaceutically acceptable salt.
The term "substantially pure" as used herein, means a compound of formula (1), particularly a compound of formula (1a) or a compound of formula (1b) or an isomer thereof. The isomer is pure enough that further purification does not detectably alter its physical and chemical properties as well as the enzyme and biological activity of the compound. The compound of formula (1) can be substantially purified according to methods known to those skilled in the art.
As used herein, the term "therapeutically effective amount" refers to the use of a compound of formula (1), especially a compound of formula (1a) or a compound of formula (1b) or a pharmaceutically acceptable compound thereof. The treatment of a cancer (as exemplified herein) of a salt or derivative (a compound of formula (1c) (as described herein) means that one or more of the following effects can be caused in a patient receiving the compound of the invention Amount: (i) to some degree of tumor growth inhibition, including slowing and complete growth arrest; (ii) reduction in the number of tumor cells; (iii) tumor size reduction; (iv) tumor cells infiltrating into surrounding organs Inhibition (ie, reduction, slowing or complete cessation); (v) inhibition of metastasis (ie, reduction, slowing or complete cessation); (vi) enhancement of anti-tumor immune response, which may but does not necessarily result Reconstruction of the tumor; and/or (vii) some degree of relief of one or more symptoms associated with the cancer being treated.
The term "pharmaceutically acceptable salts" as used herein, is intended to include a compound of formula (1), especially a compound of formula (1a) or a compound of formula (1b), and derivatives thereof. Those salts of the compounds of the invention (compounds of formula (1c)) which are safe and effective in mammals and possess the desired biological activity. Pharmaceutically acceptable acid addition salts include, but are not limited to, hydrochloride, hydrobromide, hydroiodide, nitrate, sulfate, phosphate, acetate, lactate, salicylate, citrate , tartrate, ascorbate, succinate, maleate, fumarate, formic acid salt, benzoate, glutamate, methanesulfonate, besylate, Tosylate. Suitable base addition salts include, but are not limited to, calcium, lithium, magnesium, potassium, sodium or zinc salts.
Preliminary identification of culture number PM0895172 was carried out via evaluation of its colony characteristics, and the compound of formula (1) was generated from the culture number PM0895172. A microscopic study of the isolated culture number PM0895172 strain was carried out on a modified radiobacteria separation agar medium. Observation was carried out after 20-22 days of incubation at 20 ° C to 30 ° C until colonies were observed. The culture number PM0895172 has been confirmed as a marine radiation strain.
The culture number PM0895172 has been deposited in the Microbial Type Culture Collection (MTCC, School of Microbiology and Technology (Sector 39-A, Chandigarh-160 036, India)) (World Intellectual Property Organization (WIPO)) Approved International Depository Authority (IDA) and has been provided with accession number MTCC 5684.
It should be understood that in addition to the specific microorganisms described herein, mutant strains of PM0895172, such as those produced by chemical or physical mutagenic agents (including X-rays, UV light), and organisms that have been modified by molecular biotechnology, may also be cultured, To produce a compound of formula (1).
The biological activity of the active compound can be determined by HPLC, NMR, IR, MS and/or the accumulation of the active compound, for example, via the test compound anticancer activity or via its binding, it is confirmed that a suitable mutation can be produced for the compound according to the invention. Screening of variants and variants.
The medium and/or nutrient medium isolated and cultured using the culture number PM0895172 for the production of the compound of the structural formula (1) preferably contains carbon, nitrogen and nutrient inorganic salt sources. The carbon source is, for example, one or more of starch, glucose, sucrose, dextrin, fructose, molasses, glycerin, lactose or galactose. A preferred carbon source is glucose. The nitrogen source is, for example, one or more of soy flour, peanut meal, yeast extract, beef extract, peptone, malt extract, corn steep liquor, gelatin or casein amino acid. Preferred nitrogen sources are peptone and yeast extracts. The nutritive inorganic salt is, for example, sodium chloride, potassium chloride, calcium chloride, manganese chloride, magnesium chloride, barium chloride, cobalt chloride, potassium bromide, sodium fluoride, sodium hydrogen phosphate, potassium hydrogen phosphate. , dipotassium hydrogen phosphate, disodium hydrogen phosphate, calcium carbonate, sodium hydrogencarbonate, sodium citrate, sodium nitrate, ammonium nitrate, potassium nitrate, sodium sulfate, ammonium sulfate, ammonium heptamolybdate, iron citrate, copper sulfate, sulfuric acid One or more of magnesium, ferrous sulfate, zinc sulfate or boric acid are preferred to sodium chloride and calcium chloride.
The maintenance of the culture number PM0895172 can be carried out at a temperature ranging from 24 ° C to 32 ° C. Typically, culture number PM0895172 is maintained at 27 °C - 29 °C. The well-grown culture plants can be stored in a refrigerator at 4 ° C - 8 ° C.
The inoculum culture of the culture number PM0895172 can be carried out at a temperature ranging from 27 ° C to 33 ° C and at a pH of about 5.5 to 8.5 at 210 to 260 rpm (revolutions per minute) for 60 to 80 hours. Typically, the number PM0895172 is grown at 29 ° C - 31 ° C and at a pH of about 6.5-7.5 and incubated at 230-250 rpm for 70-74 hours.
The production of the compound of the formula (1) can be carried out by fermenting the culture number PM0895172 in a shaken flask at a temperature ranging from 27 ° C to 33 ° C and at a pH of about 5.5 to 8.5 at 200-250 rpm. It took 110 hours. Typically, culture number PM0895172 is cultured at 210-230 rpm for 90-100 hours at 29 °C - 31 °C and pH of about 6.5-7.5.
The production of the compound of the formula (1) can be carried out by fermenting the culture number PM0895172 in the fermentation tank at a temperature ranging from 27 ° C to 33 ° C and from about 5.5 to 8.5 pH at 40-70 rpm and 200-300 lpm ( The liters per minute is incubated for 20-40 hours under aeration. Typically, culture number PM0895172 is incubated at 28 ° C - 31 ° C and pH between about 6.5 and 7.5, and aerated at 50-60 rpm and 240-260 lpm for 20-30 hours.
The production of the compound of formula (1) can be carried out by culturing the culture number PM0895172 in a suitable nutrient medium under the conditions described herein, preferably under water-containing conditions, for example, shaking. In the flask. The progress of the fermentation and the production of the compound of the formula (1) can be detected by high performance liquid chromatography (HPLC) and detected by measuring the biological activity of the culture solution by testing different cancer cell lines.
Fermentation is the process of growing microorganisms that produce various chemical or pharmaceutical compounds. The microorganisms are normally cultured under specific conditions in which nutrients are present. The culture solution is obtained after the completion of the fermentation process. Several cultures are centrifuged, which results in the formation of cell pellets and culture filtrate, which can be further processed in the procedures described herein.
The compound of the formula (1) present in the culture solution may exist in the form of a mixture of the compound of the formula (1a) and the compound of the formula (1b), and the compound or structure of the formula (1a) may be The compound of formula (1b) is separated from the chromatographic technique using different extraction methods.
The compound of the formula (1), in particular the compound of the formula (1a) or the compound of the formula (1b), can be used as a water-immiscible solvent such as petroleum ether, dichloromethane, chloroform or acetic acid from the culture filtrate. Ester, diethyl ether or butanol extraction or via the use of polymeric resins such as Diaion HP-20R(Mitsubishi Chemical Industries Limited, Japan), Amberlite XADR(Rohm and Haas Industries, USA) or hydrophobic interaction chromatography for activated carbon adsorption. These techniques can be used repeatedly, individually or in combination.
The compound of the formula (1), in particular the compound of the formula (1a) or the compound of the formula (1b), may be derived from a cell mass in a water-miscible solvent such as methanol, acetone, acetonitrile, n-propanol or isopropyl. The alcohol is recovered by extraction with a water-immiscible solvent such as petroleum ether, dichloromethane, chloroform, ethyl acetate or butanol.
Alternatively, the culture solution is extracted with a solvent selected from petroleum ether, dichloromethane, chloroform, ethyl acetate, methanol, acetone, acetonitrile, n-propanol or isopropanol or butanol.
Typically, the compound of formula (1) is extracted from the entire culture using ethyl acetate as a solvent. The concentration of the extract provides an active crude material having a mixture of a compound of the formula (1a) and a compound of the formula (1b).
The compound of the formula (1) according to the invention, in particular the compound of the formula (1a) or the compound of the formula (1b), can be recovered from the crude material by fractional distillation of any of the following techniques: normal phase layer Analytical method (using alumina or tannin as stationary phase; extract such as petroleum ether, ethyl acetate, dichloromethane, acetone, chloroform, methanol, isopropanol or a combination thereof); reversed phase chromatography (using reverse A phase gum such as dimethyl octadecyl decane phthalate (RP-18) or dimethyl octyl decyl phthalate (RP-8) as a stationary phase; and an extract such as water, a buffer (for example , phosphate, acetate, citrate (pH 2-8)), and organic solvents (for example, methanol, acetonitrile, acetone, tetrahydrofuran or a combination of these solvents); gel permeation chromatography (using resin Sephadex LH) -20R(Pharmacia Chemical Industries, Sweden), TSKgelRToyopearl HW (TosoHaas, Tosoh Corporation, Japan) in a solvent such as methanol, chloroform, acetone, ethyl acetate or a combination thereof; or via countercurrent chromatography (using a two-phase extract system, the composition of which is two or more Solvents such as water, methanol, ethanol, isopropanol, n-propanol, tetrahydrofuran, acetone, acetonitrile, dichloromethane, chloroform, ethyl acetate, petroleum ether, benzene, and toluene). These techniques can be used repeatedly, individually or in combination. A typical method is chromatography via a normal phase gel and a reverse phase gel such as RP-18.
As used herein, the term "isomer" is a generic term for all isomers of a compound of formula (1) that differ only in their atomic orientation. The term isomers include enantiomers, mirror image isomers (racemates, racemic mixtures) and more than one palm center, which are not mutually Is a mirrored compound isomer (non-an image isomer). The compounds of the invention may have asymmetric centers and may occur as racemates, racemic mixtures, individual non-image or isomers, or may exist as geometric isomers, all isomeric of the compounds The form of matter is included in the present invention.
As used herein, the term "tautomer" means the coexistence of only one (or more) mobile atoms and the distribution of two (or more) compounds different from each other, for example, a ketone. - keto-enol tautomer.
The compound of formula (1), its isomer or tautomer can be converted into a pharmaceutically acceptable salt or derivative thereof in the present invention.
The pharmaceutically acceptable salts of the compounds of formula (1) are prepared by standard procedures known to those skilled in the art, for example, salts such as sodium and potassium salts can be obtained by formulating compounds of formula (1) or The isomers or tautomers or derivatives thereof are prepared by treatment with a suitable sodium or potassium base such as sodium hydroxide, potassium hydroxide and the like. Similarly, salts such as hydrochlorides and sulfates can be treated with a suitable acid by, for example, hydrochloric acid, a compound of formula (1) or an isomer or tautomer or derivative thereof. Prepared by sulfuric acid and the like.
All derivatives of the compound of the formula (1), particularly a compound of the formula (1a) or a derivative of the compound of the formula (1b), are included in the scope of the present invention. Derivatives of the compound of the formula (1), particularly the compound of the formula (1a) or the compound of the formula (1b), which are derived from those in which the amine group and the hydroxyl group are derived One or more of the compounds, particularly the N-hydroxy group of the compound of formula (1), especially the compound of formula (1a) or the N-hydroxy group of the compound of formula (1b).
Accordingly, in one aspect of the invention, there is provided a derivative of a compound of formula (1) which is represented by the following structural formula (1c),



Structural formula (1c)
among them,
R1With R4Representing H independently or not;
R2With R3Is independently selected from H, hydroxyl, -O (C1-C6)alkyl and -OC(O)(C1-C6)alkyl;
Represents a single or double bond;
among them,
(C1-C6An alkyl group is unsubstituted or substituted with one or more groups independently selected from halogen, hydroxy, -O (C)1-C6)alkyl, nitro, cyano, -COOH, -NH2,-NH(C1-C6)alkyl, -N[(C1-C6)alkyl]2,-NHC(O)O(C1-C6)alkyl, -NHC(O)O(C1-C6)alkyl (C6-C10) aryl, -NH-PEG or (C6-C10)Aryl;
(C6-C10An aryl group is unsubstituted or substituted with one or more groups independently selected from halogen, halo (C)1-C6) alkyl, hydroxy, cyano, nitro, (C1-C6)alkyl, -O(C1-C6)alkyl, -COOH and -NH2;
And PEG is polyethylene glycol, which is selected from O, O'-bis[2-(N-succinimide-succinyl)ethyl]polyethylene glycol (α,ω-bis-NHS- PEG), methyl-PEG-NHS ester (MS (PEG)n, wherein n is 24), and a branched trimethyl and amber succinimide derivative of polyethylene glycol (TMS(PEG)n, where n is 12);
And stipulates if R2With R3Both are hydroxyl, then R1With R4For non-existent andRepresents a double bond;
And all isomeric and tautomeric forms or pharmaceutically acceptable salts thereof.
It will be understood that "substitution", "substituted" or "substituted with" means that one or more hydrogen atoms of a given motif are substituted with a suitable substituent and include implied conditions. Substituting for this type is based on the allowable valence of the substituted atom and the substituent, and produces a stable compound.
The term "(C1-C6"Alkyl" or "alkyl", as used herein alone or as part of another group, means an unsubstituted or substituted straight or branched chain hydrocarbon comprising from 1 to 6 carbons, for example Methyl, ethyl, propyl, isopropyl, butyl, tert-butyl, isobutyl, pentyl and hexyl. Substituted alkyl means substituted by one or more groups (C1-C6An alkyl group, the group being selected from, but not limited to, halogen, hydroxyl, -O (C)1-C6)alkyl, nitro, cyano, -COOH, -NH2,, -N[(C1-C6)alkyl]2,-NHC(O)O(C1-C6)alkyl, -NHC(O)O(C1-C6)alkyl (C6-C10) aryl, -NH-PEG or (C6-C10)Aryl.
As used herein, the term "(C6-C10"Aryl" or "aryl" means a monocyclic or bicyclic hydrocarbon ring system having up to ten ring carbon atoms, wherein at least one carbocyclic ring has a p-electron system. (C6-C10Examples of aryl ring systems include, but are not limited to, phenyl or naphthyl. Unless otherwise indicated, an aryl group can be unsubstituted or substituted with one or more groups independently selected from, but not limited to, halogen, halo (C).1-C6) alkyl, hydroxy, cyano, nitro,, -O(C1-C6)alkyl, -COOH and -NH2.
As used herein, the term "-O(C1-C6"Alkyl" or "alkoxy" means unsubstituted or substituted (C1-C6An alkyl radical having an oxygen atom bonded thereto. Representative alkoxy groups include, but are not limited to, methoxy, ethoxy, n-propoxy, 1-methylethoxy, n-butoxy, 1-methylpropoxy, 2-methyl Propyloxy or 1,1-dimethylethoxy. Substituted alkoxy group means -O(C1-C6An alkyl group wherein the alkyl group is substituted with one or more groups selected from, but not limited to, halogen, hydroxy, -O (C)1-C6)alkyl, nitro, cyano, -COOH, -NH2,-NH(C1-C6)alkyl, -N[(C1-C6)alkyl]2,-NHC(O)O(C1-C6)alkyl, -NHC(O)O(C1-C6)alkyl (C6-C10) aryl, -NH-PEG or (C6-C10)Aryl.
As used herein, the term "halogen (C1-C6"Alkyl" or "haloalkyl" means a radical wherein one or more of the hydrogen atoms of the alkyl group are substituted with one or more halogens. The monohaloalkyl radical, for example, may have a chlorine, bromine, iodine or fluorine atom. The dihalo and polyhaloalkyl radicals may have two or more identical or different halogen atoms. "halogen (C1-C6Examples of "alkyl" or "haloalkyl" include, but are not limited to, chloromethyl, dichloromethyl, trichloromethyl, dichloroethyl, dichloropropyl, fluoromethyl, difluoromethyl, tri Fluoromethyl, pentafluoroethyl, heptafluoropropyl, difluorochloromethyl, dichlorofluoromethyl, difluoroethyl or difluoropropyl.
As used herein, the term "halogen" means fluoro, chloro, bromo and iodo.
As used herein, the term "PEG" means a polyethylene glycol polymer. PEG is non-toxic, non-immunogenic, non-antigenic and highly soluble in water. A variety of different molecular weights of PEG are commercially available, based on intramolecular ethylene oxide (ie, -OCH2CH2-) The number of repeating units. For example, PEG can be obtained from commercial suppliers such as Sigma-Aldrich Co. LLC or NOF Corporation. Some examples of PEG are but are not limited to: (a) O, O'-bis[2-(N-succinimide-succinyl)ethyl]polyethylene glycol (α,ω-bis-NHS- PEG), with a molecular weight in the range of 2000 Da to 10,000 Da, which is structurally represented below:
;

(b) Methyl-PEG-NHS ester (MS (PEG)n, where n is 24) and has a molecular weight of 1214.39 Da, which is structurally represented below;
;
as well as;
(c) Branched trimethyl and amber quinone imide derivatives of polyethylene glycol (TMS (PEG)n, where n is 12 and the molecular weight is 2420.80 Da). Typically, PEG has a molecular weight in the range of 1000 to 20,000 Da, which is preferably used in the preparation of PEG conjugates of compounds as described herein.
In the context of the present invention, the term "compound of formula (1c)", "derivative of compound of formula (1)" or "detail of formula (1c) is used interchangeably, and the terms Each means a compound of the formula (1) represented by the formula (1c); and a derivative thereof, an isomer, a tautomer or a pharmaceutically acceptable salt.
According to a specific embodiment, the present invention provides a compound of formula (1c),
among them,
R1And R4Is H;
R2For -O(C1-C6)alkyl or -OC(O)(C1-C6)alkyl;
R3Hydroxyl or -O(C1-C6)alkyl;
Among them, (C1-C6An alkyl group is unsubstituted or is -NH2,-NHC(O)O(C1-C6)alkyl (C6-C10Aryl or -NH-PEG substituted;
And all isomeric or tautomeric forms or pharmaceutically acceptable salts thereof.
According to another specific embodiment, the present invention provides a compound of formula (1c),
among them,
R1With R4Representing H independently or not;
R2With R3Is independently selected from H and a hydroxyl group;
Represents a single or double bond;
And stipulates if R2With R3Both are hydroxyl, then R1And R4For non-existent andRepresents a double bond;
And all isomeric or tautomeric forms or pharmaceutically acceptable salts thereof.
Representative compounds of formula (1c) according to the invention include:









And all isomeric or tautomeric forms or pharmaceutically acceptable salts thereof.
A derivative of the compound of the formula (1), which is represented by the structural formula (1c), can be produced by a method known in the art. The reagents employed in the preparation of the compounds of formula (1c) may be commercially available or may be prepared by procedures known in the art.
R1And R4H; and R2With R3Is independently selected from hydroxyl and -O (C1-C6)alkyl, where R2And R3At least one of them is -O(C1-C6A compound of the formula (1c) of the alkyl group can be prepared by reacting a compound of the formula (1), which is a compound of the formula (1a) in an alcohol selected from the group consisting of methanol, ethanol, propanol or butanol. In combination with toluene or benzene, an alkylating agent such as trimethylformane diazomethane in an organic solvent such as hexane or toluene is reacted at a temperature ranging from -5 ° C to 5 ° C for 1 to 5 hours. Compounds 1 and 2 were prepared in this manner.
R1And R4H; and R2With R3Is independently selected from hydroxyl and -OC(O) (C1-C6) alkyl to make R2With R3At least one of them is -OC(O)(C1-C6Alkyl; where (C1-C6)alkyl group is -NHC(O)O(C1-C6)alkyl (C6-C10The compound of the formula (1c) substituted with an aryl group can be produced by reacting a compound of the formula (1), which is a compound of the formula (1a) in a solvent such as tetrahydrofuran (THF), dichloromethane ( DCM) or dimethylformamide (DMF) with concomitant reagents such as N-benzyloxycarbonyl-L-alanine (Cbz alanine) or N-(9-fluorenylmethoxycarbonyl)-L-propylamine Acid (Fmoc alanine), in the presence of a coupling agent such as benzotriazol-1-yl-oxytripyrrolidinium hexafluorophosphate (PyPOP), (benzotriazol-1-yloxy) three (two Methylamino) hexafluorophosphate (BOP), N, N'-dicyclohexylcarbodiimide (DCC) or propanephosphonic anhydride (T3P), and bases such as N,N-diisopropyl B The amine (DIPEA), triethylamine or 4-dimethylaminopyridine (DMAP) is reacted at a temperature ranging from -5 ° C to 5 ° C for 1 to 20 hours. Compound 3 was prepared in this way. The compound thus obtained can be deprotected via catalytic reduction in an acidic environment created using an acid such as diluted HCl using a catalyst such as Pd/C or Pt/C in a solvent such as methanol, ethanol, ethyl acetate or THF. To obtain a compound of the formula (1c), wherein With R3Is independently selected from hydroxyl and -OC(O) (C1-C6) alkyl to make R2With R3At least one of them is -OC(O)(C1-C6Alkyne; where the (C1-C6)alkyl group -NH2Replaced. Compound 4 was prepared in this way.
R1And R4H; and R2With R3Is independently selected from hydroxyl and -OC(O) (C1-C6) alkyl to make R2With R3At least one of them is -OC(O)(C1-C6Alkyne; where the (C1-C6)alkyl group -NH2The substituted compound of formula (1c) can be used for the action of polyethylene glycol by dissolving the compound in an organic solvent such as DCM, methanol or DMF in the presence of a base such as triethylamine or DIPEA. And adding a polyethylene glycol (PEG) polymer such as O, O'-bis[2-(N-succinimide-succinyl)ethyl]polyethylene glycol (α,ω-bis-NHS- PEG) or methyl-PEG-NHS ester, and stirred at a temperature ranging from 25 ° C to 35 ° C for 10-20 hours to obtain a compound of formula (1c) wherein R2And R3Is independently selected from hydroxyl and -OC(O) (C1-C6) alkyl to make R2With R3At least one of them is -OC(O)(C1-C6Alkyne; where the (C1-C6The alkyl group is substituted by -NH-PEG. Compound 8 was prepared in this manner.
The compound of formula (1) can be oxidized to remove at R1With R4Position of hydrogen via a reagent such as m-chloroperoxybenzoic acid in an organic solvent selected from dichloromethane or dichloroethane at a temperature ranging from 25 ° C to 35 ° C for 10-20 hours to Obtaining a compound of formula (1c) wherein R1And R4For non-existence, and R2And R3It is a hydroxyl group. Compound 5 was prepared in this way.
The N-hydroxy group of the compound of the formula (1) can be converted to NH (deoxygenation) by adding a compound of the formula (1) in an organic solvent selected from methanol, ethanol or DMF to a solvent For example, a solution reagent in THF, DCM or DMF such as bis(cyclopentadienyl)titanium(IV) dichloride and activated zinc, and stirred at a temperature ranging from -5 ° C to -50 ° C for 0.1 hr to 1 hr, To obtain a compound of formula (1c) wherein R1And R4H or non-existent, and R2And R3H. Compound 6 was prepared in this way.
The mercapto derivative of the N-hydroxy group of the compound of formula (1) can be passed via a compound of formula (1) in a solvent such as dichloromethane, pyrimidine or dichloroethane with an accompanying reagent such as ethyl chloroform. Or acetic anhydride, prepared in the presence of a base such as pyrimidine, TEA or DMAP at a temperature ranging from -5 ° C to 30 ° C for from 1 hr to 5 hr to obtain a compound of formula (1c), wherein R2And R3Is independently selected from hydroxyl and -OC(O) (C1-C6) alkyl to make R2With R3At least one of them is -OC(O)(C1-C6)alkyl. Compound 7 was prepared in this way.
The compound of formula (1c) which is a derivative of the compound of formula (1) can be optionally converted into a pharmaceutically acceptable salt thereof.
The compound of the formula (1) or a pharmaceutically acceptable salt thereof or a derivative of the formula (1c) has an anticancer activity. This has been demonstrated by testing a representative compound of the invention for a wide range of cancer cells, including compounds of structural formula (1), structural formula (1a), structural formula (1b) and structural formula (1c).
The compound of the formula (1) or an isomer or tautomer thereof or a pharmaceutically acceptable salt or a derivative of the formula (1c) can be administered as a medicament and administered as a pharmaceutical composition. Patients who have a need for it. The compound of the formula (1) or an isomer or tautomer thereof or a pharmaceutically acceptable salt or a derivative of the formula (1c) can be administered to a patient diagnosed with cancer.
Accordingly, the invention is also related to a compound of formula (1), in particular a compound of formula (1a) or a compound of formula (1b) or an isomer or tautomer or pharmaceutically acceptable salt thereof Use of a derivative of the class or formula (1c) for the manufacture of a therapeutic agent for cancer.
The invention further relates to a therapeutically effective amount of a compound of formula (1) or an isomer or tautomer thereof or a pharmaceutically acceptable salt thereof or a derivative of formula (1c), and a pharmaceutical A pharmaceutical composition of an acceptable excipient or carrier. The pharmaceutical composition is provided for the treatment of cancer. A therapeutically effective amount of a compound of the formula (1) or a stereoisomer thereof or a tautomer thereof, or a pharmaceutically acceptable salt thereof, or a derivative thereof of the structural formula (1c), which is an active ingredient of a pharmaceutical preparation, may be It may range from about 0.01 mg to 1000 mg; or may range from about 0.1 mg to 750 mg; or may range from about 0.5 mg to 500 mg; or may range from about 1 mg to 250 mg.
The use of the compound of the present invention, comprising a compound of the formula (1) or an isomer or tautomer thereof or a pharmaceutically acceptable salt or a derivative of the formula (1c), for the treatment of cancer . The compounds of the invention are used to reduce, inhibit or reduce the proliferation of tumor cells and provide for a reduction in tumor size. Representative cancers that can be treated with the compounds of the invention include, but are not limited to, leukemia, lung cancer, brain cancer, non-Hodgkin's lymphoma, Hodgkin's disease, liver cancer, kidney cancer, bladder cancer, urinary tract cancer, breast cancer, head and neck Cancer, endometrial cancer, lymphoma, melanoma, cervical cancer, thyroid cancer, gastric cancer, germ cell tumor, cholangiocarcinoma, extracranial cancer, sarcoma, mesothelioma, malignant fibrous histiocytoma, retinoblast Tumor, esophageal cancer, multiple myeloma, pancreatic cancer, ependymoma, neuroblastoma, skin cancer, ovarian cancer, recurrent ovarian cancer, prostate cancer, testicular cancer, colorectal cancer, lymphoproliferative disease, refractory Multiple myeloma, anti-multiple myeloma or myeloproliferative disease.
According to a specific embodiment of the present invention, the cancer is selected from the group consisting of acute lymphocytic leukemia, acute myeloid leukemia, adult acute myeloid leukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia, chronic myelogenous leukemia, hairy cell leukemia, non- Small cell lung cancer, small cell lung cancer, brain stem glioma, glioblastoma, cardiac cell tumor including cerebellar cardiac cell tumor and cerebral heart cell tumor, visual pathway glioma, thalamic glioma, cerebellar primordial Neuroectodermal, pineal tumor, medulloblastoma, primary central nervous system lymphoma, mantle cell lymphoma, Hodgkin's disease, hepatocellular carcinoma, renal cell carcinoma, embryonal carcinosarcoma (Wilms 'tumor', bladder cancer, urinary tract cancer, Ewing's sarcoma tumor family, osteosarcoma, rhabdomyosarcoma, soft tissue sarcoma, mesothelioma, breast cancer, endometrial cancer, oral cancer, melanoma, cervical cancer, thyroid cancer, stomach cancer , germ cell tumor, cholangiocarcinoma, extracranial cancer, bone malignant fibrous histiocytoma, retinoblastoma, esophageal cancer, multiple Myeloma, pancreatic cancer, ependymoma, neuroblastoma, skin cancer, ovarian cancer, recurrent ovarian cancer, prostate cancer, testicular cancer, colorectal cancer, lymphoproliferative disease, refractory multiple myeloma, anti- Multiple myeloma or myeloproliferative diseases, etc.
According to another embodiment of the present invention, the cancer is selected from the group consisting of acute lymphocytic leukemia, acute myeloid leukemia, adult acute myeloid leukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia, chronic myelogenous leukemia, hairy cell leukemia, Non-small cell lung cancer, small cell lung cancer, brain stem neuronoma, glioblastoma, cardiac cell tumor including cerebellar cardiac cell tumor and cerebral heart cell tumor, visual pathway glioma, pineal tumor, medullary cell Tumor, primary central nervous system lymphoma, mantle cell lymphoma, Hodgkin's disease, hepatocellular carcinoma, renal cell carcinoma, bladder cancer, urinary tract cancer, osteosarcoma, breast cancer, endometrial cancer, oral cancer , melanoma, cervical cancer, thyroid cancer, gastric cancer, malignant fibrous histiocytoma, retinoblastoma, esophageal cancer, multiple myeloma, pancreatic cancer, neuroblastoma, skin cancer, ovarian cancer, prostate cancer, Testicular cancer, colorectal cancer, lymphoproliferative disease or myeloproliferative disease, etc.
According to a further embodiment of the invention, the cancer is selected from the group consisting of acute lymphocytic leukemia, acute myeloid leukemia, adult acute myeloid leukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia, chronic myelogenous leukemia, hairy cell leukemia, non- Small cell lung cancer, small cell lung cancer, brain stem neuronoma, glioblastoma, cardiac cell tumor including cerebellar cardiac cell tumor and cerebral heart cell tumor, medulloblastoma, renal cell carcinoma, bladder cancer, urinary tract cancer , breast cancer, oral cancer, melanoma, cervical cancer, thyroid cancer, stomach cancer, pancreatic cancer, prostate cancer or colorectal cancer.
According to a specific embodiment, the present invention provides a method of treating cancer in a patient by administering to a patient a therapeutically effective amount of a compound of the formula (1) or a derivative or isomer thereof of the formula (1c) Or tautomer or pharmaceutically acceptable salt.
According to another embodiment, the invention provides a method of treating cancer in a patient by administering to the patient a therapeutically effective amount of a compound of formula (1a) or a compound of formula (1b) or an isomer thereof Or tautomer or pharmaceutically acceptable salt.
According to yet another embodiment, the invention provides a method of treating cancer in a patient by administering to the patient a therapeutically effective amount of a derivative of formula (1c) or an isomer or tautomer thereof or A pharmaceutically acceptable salt.
a compound of formula (1) or a pharmaceutically acceptable salt thereof or a derivative of formula (1c); or an isomer or tautomer or pharmaceutically acceptable salt thereof, can be administered orally, It is administered nasally, topically, subcutaneously, intramuscularly, intravenously or in other modes of administration.
The method of administration suitable for a particular situation depends on the type of cancer to be treated and the stage of the cancer. Further, the method of administration can be optimized by a physician using methods known in the art.
By convention, the range of dosages that are appropriate for a particular situation depends on the type of cancer being treated and the condition of the individual condition or disease. The selected dose level can be determined by the skilled physician according to the relevant circumstances, including the symptoms to be treated (cancer), factors such as the age, weight and physical health and response of the individual patient, pharmacokinetics, severity of the disease, etc. The factors known to the medical field are determined immediately by the chosen route of administration. The actual dosage of the active ingredient in the pharmaceutical compositions of the present invention can be varied to obtain an amount of the active ingredient effective to achieve a desired therapeutic response to a particular patient (patient in need of treatment), a composition, and a non-toxic to the patient. Medicine mode. On average, the daily dose of the active compound of the patient (one or more of the compounds of the invention) may range from about 0.05 mg to about 500 mg per kg or from about 0.5 mg to about 200 mg per kg or about A range of from 1 mg to about 100 mg per kg, or any dosage range falling within the broader range of dosage ranges as indicated herein. In the case of higher body weight patients in need of treatment, the dosage of the active compound may range from about 1 mg to about 1000 mg per kg or from about 5 mg to about 500 mg per kg. The desired dose of the active compound, i.e., the compound of the present invention, can be selected within a wide range. The daily dose administered is selected to achieve the desired therapeutic effect in patients receiving cancer treatment. Higher or lower daily doses may also be administered if desired.
A pharmaceutical composition comprising a compound of formula (1) or an isomer or tautomer thereof or a pharmaceutically acceptable salt or a derivative of formula (1c), which is accompanied by other pharmaceutically acceptable Excipients such as wetting agents, solubilizing agents such as surfactants, vehicle liquids, isotonic agents, fillers, colorants, masking perfumes, lubricants, disintegrating agents, diluents, binders, plasticizers, emulsifiers, Ointment bases, emollients, thickeners, polymers, lipids, oils, solubilizers, complexing agents or buffer substances for tablets, coated tablets, capsules, granules, powders, creams, ointments, gels, A syrup, emulsion, suspension or form of a solution suitable for parenteral administration.

In a specific embodiment of the invention, a compound comprising a compound of formula (1) or a derivative of formula (1c); or an isomer or tautomer thereof, a pharmaceutically acceptable salt The composition can be used with one or more anticancer agents such as asparaginase, bleomycin, carboplatin, carmustine, chlorambucil , cisplatin, colaspase, cyclophosphamide, cytarabine, dacarbazine, actinomycin D, daunorubicin, doxorubicin, soft Equivalent, etoposide, fluorouracil, hexamethyl melamine, hydroxyurea, ifosfamide, leucovorin, lomustine, double Mechlorethamine, 6-mercaptopurine, mesna, methotrexate, mitomycin C, mitoxantrone, hydrogen splash Nisson (prednisolone), prednisone, procarba Procarbazine, streptozocin, tamoxifen, thioguanine, vinblastine, vincristine, vindesine, Aminoglutethimide, 5-azacytidine, cladribine, busulfan, diethylstilbestrol, 2',2'-difluorodeoxygenate Glycosides, docetaxel, red-9-(2-hydroxy-3-indolyl) adenine, ethinyl estradiol, 5-fluorodeoxyuridine, 5- Fluorodeoxyuridine monophosphate, fludarabine phosphate, fluoxymesterone, flutamide, hydroxyprogesterone caproate, idarubicin , interferon, medroxyprogesterone acetate, megestrol acetate, melphalan, mitotan, pacific violet Alcohol (paclitaxel), pentostatin, N-phosphinium carboxyacetamido-L-aspartate (PALA), lucamycin, teniposide, propionic acid Testosterone, thiotepa, trimethyl melamine, uridine, vinorelbine, alsterpaullone, butyrolactone I, 2-(2-hydroxyethylamino)-6- (3-Chloroanilino)-9-isopropyl hydrazine, indirubin-3'-monohydrazine, kenpaullone, olomoucine, iso-oromoxin, N9-Isopropyl-Oromoxin, purvalanol A, roscovitine,S- the isomer Rosvitine and WHI-P180 [4-(3'-hydroxyphenyl)amino-6,7-dimethoxyquinazoline] binding; for cancer as described herein treatment.
It is to be understood that modifications that do not materially affect the activity of the various embodiments of the invention are included within the scope of the invention disclosed herein. Accordingly, the following examples are intended to illustrate but not to limit the scope of the invention.

example
The following terms/symbols/abbreviations/chemical formulas are used in the examples:


Example 1
Separation of culture number PM0895172 from marine sources
a) Separation medium composition:
1% peptone, 2% glucose, 0.2% calcium carbonate, 0.001% cobalt chloride hexahydrate, 1.5% agar, pH (at 25 ° C) 7.5.
Artificial seawater (ASW) composition: 2.46% sodium chloride, 0.067% potassium chloride, 0.136% calcium chloride, 0.629% magnesium sulfate, 0.466% magnesium chloride, 0.018% sodium hydrogencarbonate; water.
b) procedure
Deep sea sediments were collected from the offshore area of Mumbai, Maharashtra, India, and sent to the Piramal LifeSciences division of Piramal Enterprises Limited (Formerly Piramal Healthcare Limited) in Mumbai, India. Store at -20 °C during the trip.
A molecular biology method is performed on the separation of uncultured marine radiobacteria from the collected sediment samples. For this, genomic DNA was isolated from sediment samples and amplified for the presence of polyketide synthase (PKS) and non-ribosomal peptide synthetase (NRPS). The culture of interest is then separated from the sediment sample with the NRPS pathway.
Samples were stored at -20 ° C to 22 ° C and later thawed to room temperature (25 ± 2 ° C) for microbial isolation. Soil samples (~2 g) were suspended in 25 ml of sterile artificial seawater (ASW) under hot pressed mash and underarm and thoroughly comminuted. 1 ml of this suspension was transferred to a test tube and vortexed for 30 seconds. Vortex the tube for 30 seconds. Prepare up to 10 in sterile ASW-5Serial dilution. Put 100 μl of 10-3The dilution is surface coated in a dish containing the separation medium. The plates were incubated at room temperature (25 ± 2 ° C) until colonies were observed. After 20-22 days of culture, the colonies present in this medium were streaked on a Petri dish containing the separation medium (as mentioned above). The isolate was purified and provided with culture ID number PM0895172. Thus, the culture number PM0895172 was separated from the grown microorganisms into a single isolate.

Example 2
Purification of culture number PM0895172
a) Purification medium composition:
1% peptone, 2% glucose, 0.2% calcium carbonate, 0.001% cobalt chloride hexahydrate, 1.5% agar; pH (at 25 ° C) 7.5.
b) Procedure: The culture number PM0895172 was streaked into a culture dish containing the purification medium. The dishes were incubated at 27 ° C for ten days. One isolated colony from the culture dish was transferred to fresh slant medium containing ISP agar (HiMedia, India). The slant medium was incubated at 27 ° C for ten days.

Example 3
Maintenance of producer strain - culture number PM0895172
This particular strain was maintained in a slant medium containing ISP4 medium (HiMedia, India). After dissolving the medium by heating, the resulting solution was distributed in a test tube and sterilized at 121 ° C for 30 minutes. The tube was cooled and allowed to cure in an inclined position. The agar tilt medium was streaked with well-grown culture number PM0895172 and incubated at 27 ° C to 29 ° C until good growth was observed. The well-grown culture strain was stored in a refrigerator at 4 ° C to 8 ° C.

Example 4
Fermentation of culture number PM0895172 in a shaking flask
a) seed medium composition:
0.2% calcium carbonate, 0.5% sodium chloride, 0.5% corn syrup, 0.75% peptone, 1.5% glucose, 0.75% yeast extract; demineralized water; pH 7.
b) Procedure:
50 ml of the above medium was distributed in a 500 ml Erlenmeyer flask and sterilized at 121 ° C for 30 minutes. The conical flask was cooled to room temperature (25 ± 2 ° C) and each conical flask was inoculated on a bevel with a ring of well-growing production (culture number PM895172) and rotated at 30 ° C with a rotating shaker (± 1 ° C) was shaken at 230 rpm to 250 rpm for 72 hours to provide a seed culture.
c) Production medium composition:
1% peptone, 2% glucose, 0.2% calcium carbonate, 0.001% cobalt chloride hexahydrate, 1.5% agar, and pH (at 25 ° C) 7.5.
d) Procedure:
100 ml of production medium in a 500 ml Erlenmeyer flask was sterilized at 121 °C for 30 minutes, cooled to 29 °C to 30 °C and mentioned in Example 4b at 3% (v/v) Plants are grown.
e) Fermentation parameters:
The production flask was incubated on an shaker at 30 ° C for 96 hours at 220 rpm. The production flasks were harvested (harvest pH: 7.0 to 8.0) and the entire culture was extracted from each of the medium flasks with the same volume of solvent mixture (methanol: ethyl acetate (1:9)). These harvested flasks were maintained at room temperature for 4-6 hours for extraction of the supernatant after separation. The supernatant was used for testing anticancer activity.

Example 5 Preparation of a culture strain in a shake flask of a fermentation tank batch
a) Seed medium 274 (1) composition:

Glucose 15 g, corn syrup 5 g, peptone 7.5 g, yeast extract 7.5 g, calcium carbonate 2 g, sodium chloride 5 g, demineralized water 1.0 L, pH 6.5-7.5 (before sterilization).
b) Procedure:
200 ml of the above medium was distributed in a 1000 ml Erlenmeyer flask and sterilized at 121 ° C for 30 minutes. The conical flasks were cooled to room temperature, and each conical flask was inoculated on a bevel with a ring of well-growing production line (PM0895172) and oscillated at 230-250 rpm at 29 ° C -30 ° C with a rotary shaker. 70-74 hours to obtain a seed culture.

Example 6
Culture number No. PM0895172 in the fermentation tank
a) Composition of the production medium:
Soy protein 腖 10 g, glucose 20 g, calcium carbonate 2 g, cobalt chloride 0.001 g, demineralized water 1.0 L, pH 7.0 (before sterilization).
b) Procedure:
500 L of production medium was sterilized in situ in a 1000 L fermentation tank with 200 ml desmophen as an antifoaming agent at 121 ° C for 20 minutes, cooled to 29 ° C to 30 ° C and in 8 to 10 L in Example 5b The obtained seed culture plants are planted.
c) Fermentation parameters:
Temperature 29 ° C - 30 ° C, tip speed 0.94 m / s, aeration 250 lpm and harvest time 24 hr. The harvested pH of the culture broth was 6.5-7.5. Production of a compound of formula (1a) or a compound of formula (1b) which is active in the fermentation broth is determined via HPLC and the biological activity is tested against anticancer activity.
Example 7
Separation and characterization of compounds of structural formula (1a)
The whole broth (500 L) harvested in step c) of Example 6 was extracted with ethyl acetate (500 L). The organic layer was separated using a stacked disc separator (Alfa Laval (USA), model LAPX404) and concentrated to give a crude extract (150 g).

The crude extract was treated by column chromatography (gel, solvent: isopropanol / chloroform). The active compound was extracted with 1-2% isopropanol in chloroform and concentrated to give a concentrated compound (30 g).
This material was purified by column chromatography (RP C-18 gum, solvent: water / acetonitrile). The obtained active compound is extracted with 40% acetonitrile in water and evaporated to obtain 2 g of the desired semi-pure compound of formula (1) (compounds of formula (1a) and formula (1b)) mixture).
Further purification was carried out via reversed phase preparative HPLC as follows:
Column: Water’s X-Bridge RP-18 (250 mm X 19 mm, 10 μ)
Effervescent solution: acetonitrile: water (50:50).
Flow rate: 25 ml/min
Detection (UV): 220 nm
The collected active peaks were evaporated to dryness to obtain 1 g of the active compound (compound of formula (1a)).
Final purification of the active compound via gelatin preparative HPLC as follows:
Column: Water’s sunfire silica, (150 mm X 19 mm, 5 μ)
Effervescent solution: chloroform: methanol (98.5: 1.5)
Flow rate: 15 ml/min
Detection (UV): 240 nm
Rt time: 8-9 min.
The extract containing the active compound from the repeated injection was concentrated to obtain 145 mg of the compound of the pure structural formula (1a). The physical and spectral properties of the compound of formula (1a) are provided in Table 1.
Analytical HPLC conditions are as follows:
Column: Kromasil RP-18 (150 mm X 4.6 mm, 3.5 μ)
Eluent: Gradient (10% acetonitrile to 100% acetonitrile to water at 20 minutes followed by an additional 5 minutes with 100% acetonitrile)
Flow rate: 1 ml/min
Detection (UV): 220 nm
Residence time: 17-18 min (purity > 99 %)

表格 1:結構式(1a)之化合物的物理與光譜特性Table 1: Physical and spectral properties of compounds of structural formula (1a)

範例8
結構式(1b)之化合物的分離與特徵描述
將在範例7中的RP-18製備型HPLC後獲得的包含結構式(1a)之化合物與結構式(1b)之化合物的半純化合物進行RP-18 製備型HPLC,以獲得15 mg 的純結構式(1b)之化合物。將結構式(1b)之化合物的物理與光譜特性於表格2中提供。
分析 HPLC條件係如同下述:
管柱: Kromasil RP-18(150 mm X 4.6 mm,3.5 m)
沖提液: 梯度(於20分鐘的10% 乙腈至100% 乙腈對水,接著額外的5分鐘以100% 乙腈)

流速:1 ml/min
偵測(UV):220 nm
停留時間:15-17 min(純度> 99 %)
Example 8
Isolation and Characterization of the Compound of the Structural Formula (1b) The semi-pure compound comprising the compound of the formula (1a) and the compound of the formula (1b) obtained after the RP-18 preparative HPLC in Example 7 is subjected to RP- 18 Preparative HPLC to obtain 15 mg of the pure structural formula (1b). The physical and spectral properties of the compound of formula (1b) are provided in Table 2.
Analytical HPLC conditions are as follows:
Column: Kromasil RP-18 (150 mm X 4.6 mm, 3.5 m)
Eluate: Gradient (10% acetonitrile to 100% acetonitrile to water at 20 minutes followed by an additional 5 minutes with 100% acetonitrile)

Flow rate: 1 ml/min
Detection (UV): 220 nm
Residence time: 15-17 min (purity > 99%)

表格2:結構式(1b)之化合物的物理與光譜特性Table 2: Physical and spectral properties of compounds of structural formula (1b)

範例 9
2-羥基-N-(18-羥基-22-異丙基-6-甲氧基-7,19-二甲基-5,8,11,17,20,24-六氧二十四氫二噠嗪并[6,1-f:6',1'-o][1,4,7,10,13,16]五氮雜氧環十九炔-23-基)-2-(2-羥基-5-異丁基-6-甲基四氫-2H-吡喃-2-基) 丙醯胺(化合物1)與
2-羥基-2-(2-羥基-5-異丁基-6-甲基四氫-2H-吡喃-2-基)-N-(22-異丙基-6,18-二甲氧基-7,19-二甲基-5,8,11,17,20,24-六氧二十四氫二噠嗪并[6,1-f:6',1'-o][1,4,7,10,13,16]五氮雜氧環十九炔-23-基)丙醯胺(化合物2)之製備
將TMSCHN2(三甲基甲矽烷基重氮甲烷,48 μl,0.097 mmol,在己烷中的2M溶液)逐滴加至在甲醇(1 ml)與甲苯(1 ml)中之結構式(1a)之化合物(在範例7中描述者,20 mg, 0.024 mmol)的溶液,並且將生成的反應混合物於0°C攪拌3小時,然後升溫至室溫。在反應完成之後,移除溶劑以獲得固體,將其以管柱層析法純化(矽膠,二氯甲烷中之1%甲醇),以獲得5 mg的化合物1 與 3 mg 的化合物2。
化合物1:1HNMR (CDCl3,300 MHz):δ 7.98, 7.10, 6.92, 6.54, 6.20, 5.52, 5.50, 5.30, 5.16, 5.14, 5.07, 4.96, 4.54, 4.51, 4.13, 4.05, 3.80,3.18,3.12, 2.79, 2.14,2.04, 1.98, 1.95, 1.77, 1.71, 1.68, 1.60, 1.52, 1.45, 1.15, 1.10, 1.05, 0.98, 0.95, 0.90, 0.88, 0.85;ESI-MS:m/e 839 (M-H)-
化合物2:1HNMR (CDCl3,300 MHz):δ7.85, 7.15, 7.10, 6.20, 5.82, 5.42, 5.38, 5.30, 5.15, 4.98, 4.80, 4.60, 4.15, 3.95, 3.85, 3.70,3.25, 3.15, 2.85, 2.23, 2.15, 1.98, 1.95, 1.77, 1.71, 1.68, 1.65, 1.60, 1.52, 1.44, 1.15, 1.10, 1.05, 0.98, 0.95, 0.90, 0.88, 0.85;ESI-MS:m/e 853 (M-H)-

範例10
18-羥基-23-(2-羥基-2-(2-羥基-5-異丁基-6-甲基四氫-2H-吡喃-2-基)丙醯胺基)-22-異丙基-7,19-二甲基-5,8,11,17,20,24-六氧二十氫二噠嗪并[6,1-f:6',1'-o][1,4,7,10,13,16]五氮雜氧環十九炔-6(7H,13H,22H)-基 3-(((芐氧基)羰基)胺基)丙酸酯(化合物3)之製備。
方法 I:
將芐氧羰基丙胺酸(Cbz alanine,27 mg,0.12 mmol)與PyBOP(75 mg, 0.14 mmol)於0°C加至在THF(5 ml)中之結構式(1a)之化合物(描述於範例7,40 mg,0.048 mmol)的溶液中,接著DIPEA(51 μl, 0.29 mmol)的逐滴加入。將生成的反應混合物於室溫攪拌隔夜。再將反應混合物以水(5 ml)稀釋並且以乙酸乙酯(3 x 20 ml)萃取。以飽和NaHCO3、鹽水清洗合併的有機層,經由無水Na2SO4乾燥並濃縮,以獲得粗製的化合物,將其經由管柱層析法純化(矽膠,在二氯甲烷中的1%甲醇),以獲得純的化合物3。
產量:12 mg;1HNMR(CDCl3,300 MHz):δ 7.90, 7.39, 7.2, 6.45, 6.34, 6.15, 5.65, 5.50, 5.38, 5.30, 5.15, 5.10, 4.90, 4.60, 4.50, 4.10, 3.85, 3.7, 3.65, 3.55, 3.4, 3.2, 3.15, 2.7, 2.6, 2.5, 2.3, 2.19, 1.98, 1.95, 1.77, 1.71, 1.68, 1.65, 1.60, 1.52, 1.44, 1.15, 1.10, 1.05, 0.98, 0.95, 0.90, 0.88, 0.85; ESI-MS: m/e 1030 (M-H)-

方法 II:
將T3P(0.216 ml, 0.363 mmol, 在乙酸乙酯中50 wt. %)於氮氣環境下加至乙腈(20 ml, 0.121 mmol)中的芐氧羰基丙胺酸(Cbz alanine,67.5 mg, 0.302 mmol)冷卻溶液中,並且攪拌10 min。將乙腈(20 ml, 0.121 mmol)中的結構式(1a)之化合物(描述於範例7,100 mg, 0.121 mmol)加至上述溶液,接著三乙胺(0.051 ml, 0.363 mmol)的逐滴加入。將生成的反應混合物持續攪拌4 hr。在反應完成之後,將水(10 ml)加至反應混合物並且以乙酸乙酯(3 x 20 ml)萃取反應混合物。合併的有機層以飽和NaHCO3、鹽水與水清洗,經由無水Na2SO4乾燥並濃縮以獲得粗製的化合物,將其以管柱層析法純化(矽膠,在二氯甲烷中的1-2%甲醇),以獲得化合物3。產量:38 mg。

範例 11
18-羥基-23-(2-羥基-2-(2-羥基-5-異丁基-6-甲基四氫-2H-吡喃-2-基)丙醯胺)-22-異丙基-7,19-二甲基-5,8,11,17,20,24-六氧二十氫二噠嗪并[6,1-f:6',1'-o][1,4,7,10,13,16]五氮雜氧環十九炔-6(7H,13H,22H)-基3-丙胺酸酯之製備(化合物4)。
將1當量數的HCl與催化量的10 % Pd/C加至在甲醇(3 ml)中的如範例10中獲得之化合物3(30 mg, 0.029 mmol)的溶液中,並且將生成的反應混合物於室溫攪拌2小時。在反應完成之後,經由矽藻土過濾反應混合物並且濃縮過濾物,以獲得化合物4。
產量:15 mg;1HNMR (DMSO-d6;300 MHz):δ 7.92,7.30, 7.10, 6.75, 5.85, 5.45, 5.35, 5.15, 5.00, 4.85, 4.16, 3.86, 3.46, 3.10, 2.85, 2.73, 2.56, 2.15, 1.85, 1.80, 1.70, 1.68, 1.65, 1.60, 1.52, 1.44, 1.15, 1.10, 1.05, 0.98, 0.95, 0.88, 0.82, 0.80; ESI-MS: m/e 896 (M-H)-

範例 12
N-(6,18-二羥基-22-異丙基-7,19-二甲基-5,8,11,17,20,24-六氧-3,4,4a,5,6,7,8,9, 10,11,15,16,16a,17,18,19,20,22,23,24-二十氫二噠嗪并[6,1-f:6',1'-o][1,4,7,10,13,16]五氮雜氧環十九炔-23-基)-2-羥基-2-(2-羥基-5-異丁基-6-甲基四氫-2H-吡喃-2-基)丙醯胺(化合物5)之製備。
將間氯過氧苯甲酸(18 mg, 0.104 mmol)於0°C加至在二氯甲烷(6 ml)中的範例7獲得之結構式(1a)之化合物(30 mg, 0.036 mmol)的溶液中,並且將生成的反應混合物於室溫下攪拌隔夜。以水(20 ml)稀釋反應混合物並且以二氯甲烷(3 x 25 ml)萃取。合併的有機層以鹽水、水清洗,並且經由無水Na2SO4乾燥,並且濃縮以獲得粗製的化合物,將其以管柱層析法純化(矽膠,二氯甲烷中的5%甲醇)以獲得化合物5。
產量:20 mg;1HNMR (DMSO-d6,300 MHz):δ 10.34, 9.62, 7.84, 7.52, 7.45, 7.43, 7.07, 6.68, 6.19, 5.77, 5.64, 5.51, 5.41, 5.04, 5.02, 4.94, 4.17, 4.12, 2.20, 1.98, 1.90, 1.60, 1.51, 1.33, 1.23, 1.04, 0.90, 0.87, 0.82;ESI-MS: 846(M+Na)。

範例 13
2-羥基-2-(2-羥基-5-異丁基-6-甲基四氫-2H-吡喃-2-基)-N-(22-異丙基-7,19-二甲基-5,8,11,17,20,24-六氧二十四氫二噠嗪并[6,1-f:6',1'-o][1,4,7,10,13,16]五氮雜氧環十九炔-23-基)丙醯胺(化合物6)之製備。
將配置攪拌棒的一乾淨、以火焰乾燥的圓底燒瓶抽真空並以Ar沖洗。於室溫、Ar環境下,將Cp2TiCl2(193 mg, 0.774 mmol)與活化鋅(411 mg, 6.29 mmol)的脫氣THF溶液(3 ml)攪拌45 min。反應混合物的顏色從暗紅色變為橄欖綠色。將反應混合物冷卻至-30°C並且以在範例7中獲得的結構式(1a)之化合物(40 mg, 0.048 mmol)的甲醇溶液(3 ml)於3 min內逐滴加料。將反應混合物攪拌45 min,維持水浴溫度介於-30 °C至-10 °C。將反應混合物升溫至室溫並於飽和5 % K2CO3(5 ml)與乙酸乙酯(20 ml)間分隔。由移液管移除有機層並且經由whatman玻璃微纖維濾器(GF/F型)過濾以移除不溶的鈦鹽。以乙酸乙酯(4 × 20 ml)萃取水層並且在每次萃取後經由whatman玻璃微纖維濾器(GF/F型)過濾有機層。經由無水Na2SO4乾燥合併的有機層並且濃縮,以獲得固體,將其經由管柱層析法純化(矽膠,己烷中的70%乙酸乙酯),以獲得化合物6。
產量:8 mg;1HNMR (DMSO-d6,300 MHz): δ 8.32, 7.78, 6.87, 6.65, 6.25, 5.75, 5.59, 5.32, 5.28, 5.21, 5.15, 4.95, 4.77, 4.73, 4.45, 4.23, 4.12, 3.94, 3.34, 2.73, 2.27, 2.18, 2.08, 1.99, 1.88, 1.63, 1.35, 1.30, 1.23, 1.04, 0.90, 0.87; ESI-MS: 817(M+Na)。

範例 14
18-羥基-23-(2-羥基-2-(2-羥基-5-異丁基-6-甲基四氫- 2H-吡喃-2-基)丙醯胺)-22-異丙基-7,19-二甲基-5,8,11,17,20,24-六氧二十氫二噠嗪并[6,1-f:6',1'-o][1,4,7,10,13,16]五氮雜氧環十九炔-6(7H,13H,22H)-基乙酸(化合物7)之製備。
將吡啶(0.0255 ml, 0.30 mmol)於0°C加至在二氯甲烷(10 ml)中的範例7獲得的結構式(1a)之化合物(0.050 g, 0.060 mmol)之溶液,並且攪拌15 min,接著加入乙醯氯(0.043 ml, 0.60 mmol)。將生成的反應混合物於室溫下攪拌2 hr。以乙酸乙酯(3 x 30 ml)萃取反應混合物。將合併的有機層以稀釋1% HCl(5 ml)、水(10 ml)清洗,經由無水Na2SO4乾燥並且濃縮。將粗製產物以管柱層析法純化(二氯甲烷中的2%甲醇),以獲得化合物7。
產量:15mg;1HNMR (CDCl3,300 MHz):δ 7.92, 7.28, 6.85, 6.45, 6.15, 5.45, 5.36, 5.20, 5.15, 4.90, 4.85, 4.65, 4.21, 3.80, 3.15, 2.35, 2.25, 2.12, 1.98, 1.95, 1.77, 1.71, 1.68, 1.60, 1.52, 1.40, 1.15, 1.05, 0.98, 0.95, 0.90, 0.88, 0.86; ESI-MS:891(M+Na)。

範例 15
聚乙二醇化(PEGylated)共軛物(化合物8)之製備。
將N-乙基-N-異丙基丙-2-胺(135 ml, 0.779 mmol)於0°C加至在DMF(10 ml, 0.223 mmol)中之範例11獲得的化合物4(200 mg, 0.223 mmol)之溶液,並且加入一部分O,O'-雙[2-(N-琥珀醯亞胺基-琥珀醯基胺基)乙基]聚乙二醇(722 mg, 1.448 mmol),並且將生成的反應混合物於室溫下攪拌隔夜。於高速真空下移除DMF,以獲得固體,將其經由sephadex G-15尺寸排除層析管柱純化。
標準曲線係使用在DMF與水中的一系列游離結構式(1a)之化合物濃度對其在269 nm的吸光值而建構。聚乙二醇化共軛物的濃度係使用下列公式根據結構式(1a)之化合物的含量測定:
PEG共軛物的濃度= 聚乙二醇化共軛物吸光值-(截距/斜率)
發現聚乙二醇化共軛物形式(化合物8)中的結構式(1a)之化合物含量為10%。

範例16
結構式(1a)之化合物的生物評估:
在藥理試驗中採用下列用語/符號/縮寫/化學:


體外試驗
如同在引用文獻BMC Cancer, 2010, 10, 610, 1-11地設計試驗;將其揭露內容以引用的方式併入以供試驗的指導。
單層細胞試驗
步驟1
細胞株的維持
將人類腫瘤細胞株:Panc-1(胰臟癌)、HCT 116(大腸癌)、ACHN(腎細胞癌)、Calu-1(肺癌)、MiaPaca2(胰臟癌)、FADU(頭頸部癌)、PC3(前列腺癌)、G361(黑色素瘤)、MDA-MB435S(黑色素瘤)、HeLa (子宮頸癌)在由AMIMED(BioConcept –瑞士)獲得的帶有Eagle’s基本鹽類之最低必需培養基(MEM – EBS)中生長。將腫瘤細胞株:MDA-MB231(乳癌)、JURKAT(T細胞白血病)、H460(小細胞肺癌)培養於RPMI 1640(AMIMED, BioConcept,瑞士)培養基。將所有腫瘤細胞株以10% 胎牛血清(FBS)(GIBCO)、1%青黴素/鏈黴素(Sigma)以及1% 抗性抗體(Anti-Anti)(GIBCO)補充並且生長於T-175組織培養瓶(Nunc)。將非致腫瘤細胞株MCF-10A培養於帶有所有標準添加物(Lonza, Catalog. No. CC-3150)的乳腺上皮基礎培養基(Mammary Epithelial Basal Medium Mammary Epithelial Basal Medium ,MEBM)。將所有細胞生長於37℃ 的5% CO2培養箱。於80-90%匯合時將細胞繼代培養。將附著的細胞使用胰蛋白酶-EDTA(Sigma)胰蛋白酶消化並維持。所有細胞株係由ATCC(Rockville, MD, USA)購買。

步驟 2
樣本製備
將結構式(1a)之化合物(描述於範例7)溶解於DMSO,以提供所需的20 mM貯存溶液。經由貯存溶液的連續稀釋製備八個不同濃度的結構式(1a)之化合物,最後造成(相較於測試濃度)200倍較高濃度。分別於0.0001 μM、0.001 μM、0.01 μM、0.03 μM、0.1 μM、0.3 μM、1μM與3μM的濃度測試結構式(1a)之化合物。以一式三份重複評估每個濃度。

步驟 3
試驗
結構式(1a)之化合物的IC50之測定方法:
a)於3000 細胞/200 μL孔的密度種植不同的癌症/正常細胞於組織培養級96孔盤,並且使它們在37 ± 0.5 °C下加溼的5 ± 0.2% CO2培養箱中恢復24小時。
b)在24 hr後,將溶解於DMSO的1 μL 200 X(將比所需濃度高200倍以200 X 表示)結構式(1a)之化合物加至以癌症/正常細胞種植的上述組織培養盤。DMSO的最終濃度為孔中0.5%。使用DMSO 作為媒液控制組。
c)在24 hr後,將盤自CO2培養箱中移出,並且於每孔加入5 μL的CCK-8(Dojindo Molecular Technologies, Inc. USA, catalog. no. CK04-20)。
d)再將盤置於37 °C達2 hr。
e)於450 nm記錄吸光值。
f)使用下列公式計算細胞毒性百分比。

經由濃度-效果曲線的目視檢查評估IC50數值。選擇幾何平均值用於測試的十四個細胞株之平均IC50數值的計算。
結果呈現於表格3,如下述提供。
Example 9
2-hydroxy-N-(18-hydroxy-22-isopropyl-6-methoxy-7,19-dimethyl-5,8,11,17,20,24-hexaethoxytetrahydrogen Pyridazine[6,1-f:6',1'-o][1,4,7,10,13,16]pentazaoxacyclohexadecane-23-yl)-2-(2- Hydroxy-5-isobutyl-6-methyltetrahydro-2H-pyran-2-yl)propanamide (Compound 1)
2-hydroxy-2-(2-hydroxy-5-isobutyl-6-methyltetrahydro-2H-pyran-2-yl)-N-(22-isopropyl-6,18-dimethoxy Base-7,19-dimethyl-5,8,11,17,20,24-hexaoxatetrahydrodiazine-[6,1-f:6',1'-o][1, Preparation of 4,7,10,13,16]pentazaoxycyclohexadecano-23-yl)propanamide (Compound 2) TMSCHN 2 (trimethylformamidinyldiazomethane, 48 μl, 0.097 Methanol (2M solution in hexane) was added dropwise to a compound of formula (1a) (described in Example 7, 20 mg, 0.024 mmol) in methanol (1 ml) and toluene (1 ml) The solution was stirred and the resulting reaction mixture was stirred at 0 ° C for 3 h then warmed to room temperature. After completion of the reaction, the solvent was removed to obtain a solid, which was purified by column chromatography (yield, 1% methanol in dichloromethane) to obtain 5 mg of compound 1 and 3 mg of compound 2.
Compound 1: 1 H NMR (CDCl 3, 300 MHz): δ 7.98, 7.10, 6.92, 6.54, 6.20, 5.52, 5.50, 5.30, 5.16, 5.14, 5.07, 4.96, 4.54, 4.51, 4.13, 4.05, 3.80, 3.18, 3.12, 2.79, 2.14, 2.04, 1.98, 1.95, 1.77, 1.71, 1.68, 1.60, 1.52, 1.45, 1.15, 1.10, 1.05, 0.98, 0.95, 0.90, 0.88, 0.85; ESI-MS: m/e 839 (MH ) - .
Compound 2: 1 H NMR (CDCl 3, 300 MHz): δ 7.85, 7.15, 7.10, 6.20, 5.82, 5.42, 5.38, 5.30, 5.15, 4.98, 4.80, 4.60, 4.15, 3.95, 3.85, 3.70, 3.25, 3.15 , 2.85, 2.23, 2.15, 1.98, 1.95, 1.77, 1.71, 1.68, 1.65, 1.60, 1.52, 1.44, 1.15, 1.10, 1.05, 0.98, 0.95, 0.90, 0.88, 0.85; ESI-MS: m/e 853 ( MH) - .

Example 10
18-Hydroxy-23-(2-hydroxy-2-(2-hydroxy-5-isobutyl-6-methyltetrahydro-2H-pyran-2-yl)propanylamino)-22-isopropyl Base-7,19-dimethyl-5,8,11,17,20,24-hexaoxydisuccinazine [6,1-f:6',1'-o][1,4 ,7,10,13,16]pentazaoxycyclohexadecane-6(7H,13H,22H)-yl 3-(((benzyloxy)carbonyl)amino)propionate (compound 3) preparation.
Method I:
Benzyloxycarbonylalanine (Cbz alanine, 27 mg, 0.12 mmol) and PyBOP (75 mg, 0.14 mmol) were added to a compound of formula (1a) in THF (5 ml) at 0 ° C (described in the example) In a solution of 7,40 mg, 0.048 mmol), DIPEA (51 μl, 0.29 mmol) was added dropwise. The resulting reaction mixture was stirred at room temperature overnight. The reaction mixture was diluted with water (5 mL) andEtOAcEtOAc Saturated NaHCO 3, the organic layers were washed with brine, dried by anhydrous Na 2 SO 4 and concentrated to obtain crude compound which was purified by column chromatography (silica gel in 1% methanol in dichloromethane) To obtain pure compound 3.
Yield: 12 mg; 1 H NMR (CDCl 3, 300 MHz): δ 7.90, 7.39, 7.2, 6.45, 6.34, 6.15, 5.65, 5.50, 5.38, 5.30, 5.15, 5.10, 4.90, 4.60, 4.50, 4.10, 3.85, 3.7, 3.65, 3.55, 3.4, 3.2, 3.15, 2.7, 2.6, 2.5, 2.3, 2.19, 1.98, 1.95, 1.77, 1.71, 1.68, 1.65, 1.60, 1.52, 1.44, 1.15, 1.10, 1.05, 0.98, 0.95, 0.90, 0.88, 0.85; ESI-MS: m/e 1030 (MH) - .

Method II:
Benzyloxycarbonylalanine (Cbz alanine, 67.5 mg, 0.302 mmol) in T3P (0.216 ml, 0.363 mmol, 50 wt.% in ethyl acetate) in acetonitrile (20 ml, 0.121 mmol) Cool the solution and stir for 10 min. The compound of formula (1a) (described in Example 7, 100 mg, 0.121 mmol) in acetonitrile (20 ml, 0.121 mmol) was added to the above solution, followed by dropwise addition of triethylamine (0.051 ml, 0.363 mmol) . The resulting reaction mixture was stirred for 4 hrs. After the reaction was completed, water (10 ml) was added to the reaction mixture and the mixture was extracted with ethyl acetate (3 x 20 ml). The organic layers were washed with saturated NaHCO 3, washed with water, brine, dried and concentrated to obtain crude compound via over anhydrous Na 2 SO 4, which was purified by column chromatography (silica gel, in dichloromethane 1-2 % methanol) to obtain compound 3. Yield: 38 mg.

Example 11
18-Hydroxy-23-(2-hydroxy-2-(2-hydroxy-5-isobutyl-6-methyltetrahydro-2H-pyran-2-yl)propanamine)-22-isopropyl -7,19-Dimethyl-5,8,11,17,20,24-hexaoxydisuccinazine [6,1-f:6',1'-o][1,4, Preparation of 7,10,13,16]pentazaoxycyclohexadecane-6(7H,13H,22H)-yl 3-propionate (Compound 4).
1 equivalent of HCl and a catalytic amount of 10% Pd/C were added to a solution of compound 3 (30 mg, 0.029 mmol) obtained in Example 10 in methanol (3 ml) and the resulting reaction mixture Stir at room temperature for 2 hours. After the reaction was completed, the reaction mixture was filtered through celite and concentrated to give Compound 4.
Yield: 15 mg; 1 H NMR (DMSO-d 6; 300 MHz): δ 7.92, 7.30, 7.10, 6.75, 5.85, 5.45, 5.35, 5.15, 5.00, 4.85, 4.16, 3.86, 3.46, 3.10, 2.85, 2.73, 2.56, 2.15, 1.85, 1.80, 1.70, 1.68, 1.65, 1.60, 1.52, 1.44, 1.15, 1.10, 1.05, 0.98, 0.95, 0.88, 0.82, 0.80; ESI-MS: m/e 896 (MH) - .

Example 12
N-(6,18-dihydroxy-22-isopropyl-7,19-dimethyl-5,8,11,17,20,24-hexaoxy-3,4,4a,5,6,7 ,8,9,10,11,15,16,16a,17,18,19,20,22,23,24-tetrahydrodiazine-[6,1-f:6',1'-o ][1,4,7,10,13,16]pentazaoxycyclohexadecano-23-yl)-2-hydroxy-2-(2-hydroxy-5-isobutyl-6-methyltetra Preparation of hydrogen-2H-pyran-2-yl)propanamide (Compound 5).
A solution of the compound of formula (1a) (30 mg, 0.036 mmol) obtained in Example 7 obtained in Example 7 with m-chloroperoxybenzoic acid (18 mg, 0.104 mmol) at 0 °C The resulting reaction mixture was stirred at room temperature overnight. The reaction mixture was diluted with water (20 ml) and extracted with dichloromethane. The organic layers were washed with brine, washed with water, and dried through anhydrous Na 2 SO 4, and concentrated to obtain crude compound, which was purified by column chromatography (silica gel, 5% methanol in dichloromethane) to obtain to Compound 5.
Yield: 20 mg; 1 H NMR (DMSO-d 6, 300 MHz): δ 10.34, 9.62, 7.84, 7.52, 7.45, 7.43, 7.07, 6.68, 6.19, 5.77, 5.64, 5.51, 5.41, 5.04, 5.02, 4.94, 4.17, 4.12, 2.20, 1.98, 1.90, 1.60, 1.51, 1.33, 1.23, 1.04, 0.90, 0.87, 0.82; ESI-MS: 846 (M+Na).

Example 13
2-hydroxy-2-(2-hydroxy-5-isobutyl-6-methyltetrahydro-2H-pyran-2-yl)-N-(22-isopropyl-7,19-dimethyl -5,8,11,17,20,24-hexaoxytetrahydrotetrapyrazine[6,1-f:6',1'-o][1,4,7,10,13,16 Preparation of pentanitroxacyclohexadecano-23-yl)propanamide (Compound 6).
A clean, flame dried round bottom flask equipped with a stir bar was evacuated and rinsed with Ar. Cp 2 TiCl 2 (193 mg, 0.774 mmol) and a solution of activated zinc (411 mg, 6.29 mmol) in degassed THF (3 ml) were stirred for 45 min. The color of the reaction mixture changed from dark red to olive green. The reaction mixture was cooled to -30 ° C and was added dropwise with a solution of the compound of formula (1a) (40 mg, 0.048 mmol) in methanol (3 ml) obtained in Example 7 over 3 min. The reaction mixture was stirred for 45 min maintaining the bath temperature between -30 °C and -10 °C. The reaction mixture was warmed to room temperature and saturated at 5% K 2 CO 3 (5 ml) and partition between ethyl acetate (20 ml). The organic layer was removed by pipette and filtered through a Whatman glass microfiber filter (GF/F type) to remove the insoluble titanium salt. The aqueous layer was extracted with ethyl acetate (4×20 ml) and the organic layer was filtered through watman glass microfiber filter (GF/F type) after each extraction. The combined organic layers were dried over anhydrous Na 2 SO via and concentrated to obtain a solid, which was purified by column chromatography (silica gel, 70% ethyl acetate in hexanes) via, to obtain Compound 6.
Yield: 8 mg; 1 H NMR (DMSO-d 6, 300 MHz): δ 8.32, 7.78, 6.87, 6.65, 6.25, 5.75, 5.59, 5.32, 5.28, 5.21, 5.15, 4.95, 4.77, 4.73, 4.45, 4.23, 4.12, 3.94, 3.34, 2.73, 2.27, 2.18, 2.08, 1.99, 1.88, 1.63, 1.35, 1.30, 1.23, 1.04, 0.90, 0.87; ESI-MS: 817 (M+Na).

Example 14
18-Hydroxy-23-(2-hydroxy-2-(2-hydroxy-5-isobutyl-6-methyltetrahydro-2H-pyran-2-yl)propanamine)-22-isopropyl -7,19-Dimethyl-5,8,11,17,20,24-hexaoxydisuccinazine [6,1-f:6',1'-o][1,4, Preparation of 7,10,13,16]pentazaoxycyclohexadecane-6(7H,13H,22H)-ylacetic acid (Compound 7).
Pyridine (0.0255 ml, 0.30 mmol) was added to a solution of the compound of formula (1a) (0.050 g, 0.060 mmol) obtained in Example 7 in dichloromethane (10 ml) and stirred for 15 min. Then, acetamidine chloride (0.043 ml, 0.60 mmol) was added. The resulting reaction mixture was stirred at room temperature for 2 hr. The reaction mixture was extracted with ethyl acetate (3 x 30 ml). The combined organic layers were diluted with 1% HCl (5 ml), water (10 ml) wash, dried by anhydrous Na 2 SO 4 and concentrated. The crude product was purified by column chromatography (2% methanol in dichloromethane) to afford compound 7.
Yield: 15 mg; 1 H NMR (CDCl 3, 300 MHz): δ 7.92, 7.28, 6.85, 6.45, 6.15, 5.45, 5.36, 5.20, 5.15, 4.90, 4.85, 4.65, 4.21, 3.80, 3.15, 2.35, 2.25, 2.12 , 1.98, 1.95, 1.77, 1.71, 1.68, 1.60, 1.52, 1.40, 1.15, 1.05, 0.98, 0.95, 0.90, 0.88, 0.86; ESI-MS: 891 (M+Na).

Example 15
Preparation of a PEGylated conjugate (Compound 8).
N-Ethyl-N-isopropylpropan-2-amine (135 ml, 0.779 mmol) was added at 0 ° C to compound 4 (200 mg, obtained in Example 11 in DMF (10 ml, 0.223 mmol) a solution of 0.223 mmol), and a portion of O,O'-bis[2-(N-succinimido]-succinylamino)ethyl]polyethylene glycol (722 mg, 1.448 mmol) was added and The resulting reaction mixture was stirred at room temperature overnight. DMF was removed under high speed vacuum to obtain a solid which was purified via a sephadex G-15 size exclusion chromatography column.
The standard curve was constructed using a series of free structural formula (1a) concentrations in DMF and water for its absorbance at 269 nm. The concentration of the PEGylated conjugate is determined according to the content of the compound of the formula (1a) using the following formula:
Concentration of PEG conjugate = PEGylated conjugate absorbance - (intercept / slope)
The content of the compound of the formula (1a) in the PEGylated conjugate form (Compound 8) was found to be 10%.

Example 16
Biological evaluation of compounds of formula (1a):
The following terms/symbols/abbreviations/chemistry are used in pharmacological tests:


In vitro assays are designed as described in the cited documents BMC Cancer, 2010, 10, 610, 1-11; the disclosures of which are incorporated by reference for the purposes of reference.
Monolayer cell test step 1
Maintenance of cell lines will be human tumor cell lines: Panc-1 (pancreatic cancer), HCT 116 (colorectal cancer), ACHN (renal cell carcinoma), Calu-1 (lung cancer), MiaPaca2 (pancreatic cancer), FADU (head and neck) Carcinoma), PC3 (prostate cancer), G361 (melanoma), MDA-MB435S (melanoma), HeLa (cervical cancer) in minimal essential medium with Eagle's basic salt obtained by AMIMED (BioConcept - Switzerland) Growing in (MEM – EBS). Tumor cell lines: MDA-MB231 (breast cancer), JURKAT (T cell leukemia), H460 (small cell lung cancer) were cultured in RPMI 1640 (AMIMED, BioConcept, Switzerland) medium. All tumor cell lines were supplemented with 10% fetal bovine serum (FBS) (GIBCO), 1% penicillin/streptomycin (Sigma), and 1% anti-anti (GIBCO) and grown in T-175 tissue. Culture flask (Nunc). The non-tumor cell line MCF-10A was cultured in Mammary Epithelial Basal Medium Mammary Epithelial Basal Medium (MEBM) with all standard supplements (Lonza, Catalog. No. CC-3150). All cells were grown in a 5% CO 2 incubator at 37 °C. Cells were subcultured at 80-90% confluence. The attached cells were trypsinized and maintained using trypsin-EDTA (Sigma). All cell lines were purchased from ATCC (Rockville, MD, USA).

Step 2
Sample Preparation Compounds of formula (1a) (described in Example 7) were dissolved in DMSO to provide the desired 20 mM stock solution. Eight different concentrations of the compound of formula (1a) were prepared via serial dilution of the stock solution, resulting in a 200-fold higher concentration (compared to the test concentration). The compound of formula (1a) was tested at concentrations of 0.0001 μM, 0.001 μM, 0.01 μM, 0.03 μM, 0.1 μM, 0.3 μM, 1 μM and 3 μM, respectively. Each concentration was evaluated in triplicate.

Step 3
Method for determining the IC 50 of a compound of the formula (1a):
a) Plant different cancer/normal cells in tissue culture grade 96-well plates at a density of 3000 cells/200 μL well and restore them in a 5 ± 0.2% CO 2 incubator humidified at 37 ± 0.5 °C. hour.
b) After 24 hr, add 1 μL of 200 X (200 times higher than the desired concentration to 200 X) of the compound of formula (1a) to the above tissue culture plate grown in cancer/normal cells. . The final concentration of DMSO was 0.5% in the wells. DMSO was used as the vehicle control group.
c) After 24 hr, the plates were removed from the CO 2 incubator and 5 μL of CCK-8 (Dojindo Molecular Technologies, Inc. USA, catalog. no. CK04-20) was added to each well.
d) Place the plate at 37 °C for 2 hr.
e) Record the absorbance at 450 nm.
f) Calculate the percentage of cytotoxicity using the following formula.

Check visually evaluate the effect IC 50 value of the curve - via the concentration. Select the geometric mean value of 50 for testing IC fourteen cell lines of average calculations.
The results are presented in Table 3, as provided below.

表格3:不同癌細胞與正常細胞中的結構式(1a)之化合物的IC50數值Table 3: IC cancer cell structure different from the compound of formula (1a) of the normal cells 50 value

結論
體外研究顯示,結構式(1a)之化合物在不同癌症細胞中表現0.0013 μM至0.025 μM 範圍的IC50;而在正常細胞(MCF-10A)中,其表現0.019 μM的IC50。此數據顯示結構式(1a)之化合物對高度增生的癌細胞株是具有活性的。

範例17
試驗
FITC 膜聯蛋白 V細胞凋亡的偵測:
偵測細胞凋亡的經典技術—膜聯蛋白 V試驗是以流式細胞儀偵測細胞凋亡最常使用的方法。細胞凋亡最早的特徵之一為磷脂醯絲胺酸(phosphatidylserine)從細胞膜內層至外層的轉位作用,從而暴露磷脂醯絲胺酸於外界環境。
膜聯蛋白 V(cat. no. 556420, BD Biosciences, USA)結合至暴露於細胞表面的磷脂醯絲胺酸並且於細胞凋亡較早階段辨識細胞。
a)將HeLa癌細胞(在範例16的步驟1所述)以0.5 × 106細胞/2000 μL孔的密度種植於組織培養級96孔盤,並且使它們在37 ± 0.5 °C下加溼的5 ± 0.2% CO2培養箱中恢復12小時。
b)在12 hr後,將溶解於DMSO的5 μL 200 X(將比所需濃度高200倍以200 X 表示)結構式(1a)之化合物(範例16步驟2)加至以HeLa癌細胞種植的上述組織培養盤。結構式(1a)之化合物的最終濃度為0.0003 μM、0.003 μM與 0.01 μM。孔中的DMSO的最終濃度為0.5%。
c)在48 hr後,盤自CO2培養箱中移出,並且根據製造商指導使用Apoptosis Detection Kit(目錄編號:556547, BD biosciences, USA)而針對FITC膜聯蛋白染色。
d)將經處理的細胞於BD流式細胞儀中分析供膜聯蛋白V偵測。使用FACS分析測定存在於M1與M2的族群。

結果:媒液控制細胞顯示2.01 % 膜聯蛋白V陽性細胞(M2),而以0.0003 μM、0.003 μM與0.01 μM濃度暴露於結構式(1a)之化合物12 hr的細胞分別顯示33.80 %、40.27 %與59.05 %細胞對於膜聯蛋白V染色呈陽性(M2)(如同第5圖顯示)。
結論:此數據表示結構式(1a)之化合物在HeLa癌細胞中經由細胞凋亡誘發細胞死亡。

範例18
試驗
高含量篩選(high content screening)的蛋白質表現研究
於HeLa細胞中以Cellomics高含量影像進行細胞週期蛋白D、核因子-Κb(p65)[NFkB(p65)]、核糖體蛋白S6(pS6)、腫瘤蛋白53(p53)、蛋白質c-jun N-端激酶(pJNK)、切割半胱天冬酶 9、切割半胱天冬酶 3與切割聚(ADP-核糖)聚合酶(PARP)的蛋白質表現(於範例16之步驟1所述)。這些一級抗體係購買自Santacruz Biotechnology, USA。簡單地說將 1 X 104細胞種植在透明底的96孔組織培養級黑盤(Nunc, USA),並且使其附著24 h,然後以新鮮培養基替換,將細胞以0.01 μM、0.003 μM與0.0003 μM濃度的結構式(1)之化合物(於範例16之步驟2所述)處理,並且分別培養1 h 與 3 h。在各自的時間點,將細胞於RT以3.7%甲醛(Sigma St. Louis, MO)固定10分鐘,接著以0.15% Triton X-100(Sigma St. Louis, MO)透化(permeabilization)10分鐘。再將細胞以5%牛血清白蛋白阻斷2小時。於阻斷步驟之後,加入特定一級抗體1 h,且將不同蛋白質的一級抗體以Dylight548(紅色)標記的二級抗體(Thermofisher, USA)定位。在二級抗體培養之後,將核以Hoechst 3342(藍色)(Sigma, USA)染色。經由在Cellomics Array ScanRVTI HCS讀取儀(Thermo- Fisher Scientific Inc., Waltham, MA)掃描盤以測定免疫螢光。使用Cellomics的Compartmental Analysis bio-algorithm分析所有數據點,並且將定量數據相較於未處理細胞以活化%表示。每個重複孔計數1000細胞,並且將結果以平均± S.E表示。
結果總結於表格4與表格5。
表格4:結構式(1a)之化合物於3小時後對HeLa細胞中p53、pJNK、切割半胱天冬酶 9、切割半胱天冬酶 3與切割PARP蛋白質表現的影響


表格5:結構式(1a)之化合物於3小時後對HeLa細胞中細胞週期蛋白D、NFkB(p65)與pS6蛋白質表現的影響


結論
在癌細胞中,結構式(1a)之化合物顯著地向下調節NFkB(p65)、pS6與細胞週期蛋白D的蛋白質表現。其亦向上調節p53、Pjnk、切割半胱天冬酶 9表現,隨後誘發驅動癌細胞朝向細胞凋亡的切割半胱天冬酶3與切割PARP的顯著向上調節。

範例 19
結構式(1b)之化合物與化合物1至8的生物評估
如同描述於上述範例16設計試驗。如同上述描述於範例16進行體外試驗的樣本製備以及結構式(1b)之化合物(描述於範例8)以及在範例9至15獲得之化合物(與化合物1至8相對應)的IC50數值之測定。
經由濃度-效果曲線的目視檢查評估IC50數值。選擇幾何平均值用於測試的六個細胞株(PC3、MDA MB、HCT116、MCF7、MIAPaCa2、MCF10A)之平均IC50數值的計算。
結果呈現於表格6,提供於下。
表格6:不同癌症細胞株與正常細胞中結構式(1b)之化合與化合物2至9的IC50數值


-未測試
結論
體外試驗顯示(i) 結構式(1b)之化合物在不同癌細胞中顯示0.018 μM至0.071 μM範圍的 IC50;而在正常細胞(MCF-10A)中其顯示0.030 μM的 IC50;(ii) 化合物1至8在不同癌細胞中顯示0.009 μM至1.045 μM範圍的IC50;而在正常細胞(MCF-10A)中其顯示< 0.010 μM至0.830 μM範圍的 IC50
這些數據顯示結構式(1b)之化合物以及化合物1至8對高度增生的癌細胞株為具有活性的。
Conclusion In vitro studies have shown that compounds of structural formula (1a) exhibit an IC 50 in the range of 0.0013 μM to 0.025 μM in different cancer cells; whereas in normal cells (MCF-10A), they exhibit an IC 50 of 0.019 μM. This data shows that compounds of structural formula (1a) are active against highly proliferating cancer cell lines.

Example 17
test
Detection of FITC Annexin V cell apoptosis:
The classic technique for detecting apoptosis, the annexin V assay, is the most commonly used method for detecting apoptosis by flow cytometry. One of the earliest features of apoptosis is the translocation of phospholipidylserine from the inner layer to the outer layer of the cell membrane, thereby exposing the phospholipid lysine to the environment.
Annexin V (cat. no. 556420, BD Biosciences, USA) binds to phospholipid lysine exposed to the cell surface and recognizes cells at an earlier stage of apoptosis.
a) HeLa cancer cells (described in step 1 of Example 16) were planted in tissue culture grade 96-well plates at a density of 0.5 × 10 6 cells/2000 μL well and allowed to humidify at 37 ± 0.5 °C. 5 ± 0.2% CO 2 incubator for 12 hours to recover.
b) After 12 hr, add 5 μL of 200 X (200 times higher than the desired concentration to 200 X) of the compound of formula (1a) (Example 16 Step 2) to HeLa cancer cells The above tissue culture plate. The final concentrations of the compounds of formula (1a) are 0.0003 μM, 0.003 μM and 0.01 μM. The final concentration of DMSO in the wells was 0.5%.
c) After 48 hr, the plates were removed from the CO 2 incubator and stained for FITC annexin using the Apoptosis Detection Kit (catalog number: 556547, BD biosciences, USA) according to the manufacturer's instructions.
d) The treated cells were analyzed for Annexin V detection in a BD flow cytometer. The population present in M1 and M2 was determined using FACS analysis.

RESULTS: The vehicle control cells showed 2.01% Annexin V positive cells (M2), while the cells exposed to the compound of formula (1a) at 0.0003 μM, 0.003 μM and 0.01 μM for 12 hr showed 33.80 %, 40.27 %, respectively. It was positive (M2) for Annexin V staining with 59.05% of cells (as shown in Figure 5).
Conclusion: This data indicates that the compound of formula (1a) induces cell death via apoptosis in HeLa cancer cells.

Example 18
High-content screening of protein expression studies in HeLa cells with high levels of Cellomics images of cyclin D, nuclear factor-Κb (p65) [NFkB (p65)], ribosomal protein S6 (pS6), Protein expression of tumor protein 53 (p53), protein c-jun N-terminal kinase (pJNK), cleaving caspase 9, cleaving caspase 3 and cleaved poly(ADP-ribose) polymerase (PARP) (described in step 1 of Example 16). These primary resistance systems were purchased from Santacruz Biotechnology, USA. Briefly, 1 X 10 4 cells were seeded in clear-bottom 96-well tissue culture grade black plates (Nunc, USA) and allowed to attach for 24 h, then replaced with fresh medium at 0.01 μM, 0.003 μM and 0.0003 The compound of formula (1) at a concentration of μM (described in step 2 of Example 16) was treated and cultured for 1 h and 3 h, respectively. At each time point, cells were fixed with 3.7% formaldehyde (Sigma St. Louis, MO) for 10 minutes at RT, followed by permeabilization with 0.15% Triton X-100 (Sigma St. Louis, MO) for 10 minutes. The cells were blocked with 5% bovine serum albumin for 2 hours. Following the blocking step, a specific primary antibody was added for 1 h, and primary antibodies of different proteins were localized with Dylight548 (red) labeled secondary antibody (Thermofisher, USA). After secondary antibody incubation, the nuclei were stained with Hoechst 3342 (blue) (Sigma, USA). Immunofluorescence was determined by scanning the disk on a Cellomics Array Scan R VTI HCS reader (Thermo-Firis Scientific Inc., Waltham, MA). All data points were analyzed using Cellomics' Analysis Analysis bio-algorithm and the quantitative data was expressed as % activation compared to untreated cells. 1000 cells were counted for each replicate well and the results were expressed as mean ± SE.
The results are summarized in Table 4 and Table 5.
Table 4: Effect of compounds of structural formula (1a) on the expression of p53, pJNK, cleaving caspase 9, cleaving caspase 3 and cleaving PARP proteins in HeLa cells after 3 hours


Table 5: Effect of Compounds of Structural Formula (1a) on Cyclin D, NFkB (p65) and pS6 Protein Expression in HeLa Cells After 3 Hours


Conclusion Compounds of structural formula (1a) significantly downregulate the protein expression of NFkB (p65), pS6 and cyclin D in cancer cells. It also upregulates p53, Pjnk, and cleaved caspase-9 expression, followed by a significant upward regulation of cleavage of caspase 3 and cleavage of PARP that drive cancer cells toward apoptosis.

Example 19
The biological evaluation of the compound of the formula (1b) and the compounds 1 to 8 is as described in the above-mentioned Example 16 design test. Determination of the IC 50 values of the compounds prepared in vitro as described in Example 16 and the compounds of formula (1b) (described in Example 8) and the compounds obtained in Examples 9 to 15 (corresponding to compounds 1 to 8). .
Check visually evaluate the effect IC 50 value of the curve - via the concentration. Calculating an average value for the geometric mean of six selected IC cell lines tested (PC3, MDA MB, HCT116, MCF7, MIAPaCa2, MCF10A) of 50.
The results are presented in Table 6, which is provided below.
IC 50 value of the compound with a compound of different cancer cell lines and normal cells in the structural formula (1b) of 2-9: Table 6


- not tested Conclusion vitro test shows that the compound (i) of the formula (1b) of the display range of 0.018 μM to 0.071 μM of IC 50 in various cancer cells; in normal cells (MCF-10A) which shows the IC 50 of 0.030 μM (ii) Compounds 1 to 8 showed an IC 50 in the range of 0.009 μM to 1.045 μM in different cancer cells; whereas in normal cells (MCF-10A) it showed an IC 50 in the range of < 0.010 μM to 0.830 μM.
These data show that the compound of formula (1b) and compounds 1 to 8 are active against highly proliferating cancer cell lines.

no

Claims (22)

一種結構式(1)之化合物;





結構式(1)
其中在該結構式(1)之化合物中,當該鍵代表一單鍵且R1為H;將該所述化合物稱作結構式(1a)之該化合物;以及
當該鍵為一雙鍵且R1不存在時;將該所述化合物稱作結構式(1b)之該化合物;
或其一同分異構物或一互變異構物或一混合物,或一衍生物;或其一藥學上可接受之鹽類。
a compound of formula (1);





Structural formula (1)
Wherein in the compound of formula (1), when the bond Representing a single bond and R 1 is H; the compound is referred to as the compound of formula (1a); and when the bond When a double bond and R 1 is absent; This compound is referred to the structure of the compound of formula (1b) of the;
Or a homoisomer or a tautomer or a mixture thereof, or a derivative; or a pharmaceutically acceptable salt thereof.
如申請專利範圍第1項所述之化合物,其中該所述化合物為該結構式(1a)之該化合物,其特徵為:
a)分子量為826.9,
b)分子式C37H62N8O13
c)IR(KBr)光譜3325, 2960, 2872, 1755, 1641, 1524, 1445, 1331, 1306, 1288 與 1254 cm-1
d)1H NMR光譜(500 MHz, CDCl3):δ 9.20, 8.00, 7.10, 6.90, 6.40, 6.10, 5.40, 5.38, 5.29, 5.10, 4.90, 4.80, 4.70, 4.60, 3.90, 3.70, 3.15, 2.70, 2.30, 2.10, 1.98, 1.95, 1.77, 1.71, 1.68, 1.60, 1.52, 1.40, 1.15, 1.10, 1.05, 0.98, 0.95, 0.90, 0.88 與 0.85(如同第1圖所提供的),
e)13C NMR光譜(75 MHz, CDCl3):δ 176.0, 172.0 (3), 171.0, 170.0, 169.0, 99.9, 78.9, 77.2, 71.7, 53.5, 52.9, 49.3, 49.0, 47.1, 46.7, 46.2, 42.2, 41.0, 39.8, 29.9, 27.4, 24.8, 24.6, 24.2, 24.1, 23.2, 21.7, 21.5 (2), 20.6, 19.9, 19.4, 18.1, 12.9 與 11.7(如同第2圖所提供的)。
The compound of claim 1, wherein the compound is the compound of the formula (1a), which is characterized by:
a) the molecular weight is 826.9,
b) the molecular formula C 37 H 62 N 8 O 13 ,
c) IR (KBr) spectra 3325, 2960, 2872, 1755, 1641, 1524, 1445, 1331, 1306, 1288 and 1254 cm -1 ,
d) 1 H NMR spectrum (500 MHz, CDCl 3 ): δ 9.20, 8.00, 7.10, 6.90, 6.40, 6.10, 5.40, 5.38, 5.29, 5.10, 4.90, 4.80, 4.70, 4.60, 3.90, 3.70, 3.15, 2.70 , 2.30, 2.10, 1.98, 1.95, 1.77, 1.71, 1.68, 1.60, 1.52, 1.40, 1.15, 1.10, 1.05, 0.98, 0.95, 0.90, 0.88 and 0.85 (as provided in Figure 1),
e) 13 C NMR spectrum (75 MHz, CDCl 3 ): δ 176.0, 172.0 (3), 171.0, 170.0, 169.0, 99.9, 78.9, 77.2, 71.7, 53.5, 52.9, 49.3, 49.0, 47.1, 46.7, 46.2, 42.2, 41.0, 39.8, 29.9, 27.4, 24.8, 24.6, 24.2, 24.1, 23.2, 21.7, 21.5 (2), 20.6, 19.9, 19.4, 18.1, 12.9 and 11.7 (as provided in Figure 2).
如申請專利範圍第1項所述之化合物,其中該所述化合物為結構式(1b)之該化合物,其特徵為:
a)分子量824.4,
b)分子式C37H60N8O13
c)IR(KBr):3342, 3030, 2953, 1752, 1650, 1524, 1421, 1293, 1255 與1239 cm-1
d)1H NMR 光譜(500 MHz, CDCl3):δ9.20, 8.00, 7.19, 7.0, 6.18, 6.14, 5.80, 5.41, 5.39, 5.30, 5.10, 4.80 (2), 4.20, 3.70, 3.19, 2.62, 2.27, 2.10, 2.01, 1.98, 1.77, 1.71, 1.62, 1.58, 1.44, 1.40, 1.39, 1.37, 1.20, 1.10, 1.05, 0.96, 0.91, 0.90, 0.86與 0.85(如同第3圖所提供的),
e)13C NMR光譜(75 MHz, CDCl3):δ176.0, 171.8, 171.7, 170.8, 170.6, 170.3, 168.9, 144.6, 98.8, 79.0, 77.2, 71.7, 53.6, 53.5, 50.6, 49.1, 47.1, 46.4, 42.6, 41.0, 39.8, 29.9, 27.4, 24.7, 24.6, 24.2, 24.0, 22.1, 21.5, 21.4, 19.8, 19.5, 19.4, 18.1, 17.2, 12.9 與 11.8 (如同第4圖所提供的)。
The compound of claim 1, wherein the compound is a compound of the formula (1b), which is characterized by:
a) molecular weight 824.4,
b) the molecular formula C 37 H 60 N 8 O 13 ,
c) IR (KBr): 3342, 3030, 2953, 1752, 1650, 1524, 1421, 1293, 1255 and 1239 cm -1 ,
d) 1 H NMR spectrum (500 MHz, CDCl 3 ): δ 9.20, 8.00, 7.19, 7.0, 6.18, 6.14, 5.80, 5.41, 5.39, 5.30, 5.10, 4.80 (2), 4.20, 3.70, 3.19, 2.62 , 2.27, 2.10, 2.01, 1.98, 1.77, 1.71, 1.62, 1.58, 1.44, 1.40, 1.39, 1.37, 1.20, 1.10, 1.05, 0.96, 0.91, 0.90, 0.86 and 0.85 (as provided in Figure 3),
e) 13 C NMR spectrum (75 MHz, CDCl 3 ): δ 176.0, 171.8, 171.7, 170.8, 170.6, 170.3, 168.9, 144.6, 98.8, 79.0, 77.2, 71.7, 53.6, 53.5, 50.6, 49.1, 47.1, 46.4, 42.6, 41.0, 39.8, 29.9, 27.4, 24.7, 24.6, 24.2, 24.0, 22.1, 21.5, 21.4, 19.8, 19.5, 19.4, 18.1, 17.2, 12.9 and 11.8 (as provided in Figure 4).
如申請專利範圍第1項所述之化合物,其中該所述化合物為結構式(1)之該化合物的一衍生物,其以該結構式(1c)表示,



結構式(1c)
其中,
R1與R4獨立地代表H或不存在;
R2與R3係獨立地選自H、羥基、-O(C1-C6)烷基以及 -OC(O)(C1-C6)烷基;
表示一單鍵或雙鍵;
其中,
(C1-C6)烷基為未被取代的或被以一或更多基團取代,該一或更多基團係獨立地選自鹵素、羥基、 -O(C1-C6)烷基、硝基、氰基、-COOH、-NH2、-NH(C1-C6)烷基、-N[(C1-C6)烷基]2、-NHC(O)O(C1-C6)烷基、-NHC(O)O(C1-C6)烷基(C6-C10)芳基、-NH-PEG 或(C6-C10)芳基;
(C6-C10)芳基為未被取代的或被以一或更多基團取代,該基團係獨立地選自鹵素、鹵(C1-C6)烷基、羥基、氰基、硝基、(C1-C6)烷基、、-COOH以及-NH2
以及PEG為聚乙二醇,其係選自O,O'-雙[2-(N-琥珀醯亞胺-琥珀醯胺基)乙基]聚乙二醇(α,ω-雙-NHS-PEG)、甲基-PEG-NHS酯(MS(PEG)n,其中n為24),以及聚乙二醇一分枝的三甲基與琥珀醯亞胺酯衍生物(TMS(PEG)n,其中n為12);
伴隨一規定若R2與R3兩者為羥基,那麼R1與R4為不存在的並且表示一雙鍵;
或其一同分異構物或一互變異構物或一混合物;或其一藥學上可接受之鹽類。
The compound of claim 1, wherein the compound is a derivative of the compound of the formula (1), which is represented by the formula (1c),



Structural formula (1c)
among them,
R 1 and R 4 independently represent H or are absent;
R 2 and R 3 are independently selected H, hydroxy, -O (C 1 -C 6) alkyl and -OC (O) (C 1 -C 6) alkyl;
Represents a single or double bond;
among them,
(C 1 -C 6 )alkyl is unsubstituted or substituted with one or more groups independently selected from halogen, hydroxy, -O(C 1 -C 6 ) Alkyl, nitro, cyano, -COOH, -NH 2 , -NH(C 1 -C 6 )alkyl, -N[(C 1 -C 6 )alkyl] 2 , -NHC(O)O ( C 1 -C 6 )alkyl, -NHC(O)O(C 1 -C 6 )alkyl(C 6 -C 10 )aryl, -NH-PEG or (C 6 -C 10 )aryl;
(C 6 -C 10 ) aryl is unsubstituted or substituted with one or more groups independently selected from halogen, halo(C 1 -C 6 )alkyl, hydroxy, cyano , nitro, (C 1 -C 6 )alkyl, , -COOH and -NH 2 ;
And PEG is polyethylene glycol, which is selected from O, O'-bis[2-(N-succinimide-succinyl)ethyl]polyethylene glycol (α,ω-bis-NHS- PEG), methyl-PEG-NHS ester (MS(PEG) n , where n is 24), and a polyethylene glycol-branched trimethyl and amber quinone imide derivative (TMS(PEG) n , Where n is 12);
With the proviso that if both R 2 and R 3 are hydroxyl groups, then R 1 and R 4 are absent and Represents a double bond;
Or a homoisomer or a tautomer or a mixture thereof; or a pharmaceutically acceptable salt thereof.
如申請專利範圍第4項所述之化合物,其為結構式(1c)之一化合物;
R1與 R4為H;
R2為-O(C1-C6)烷基或-OC(O)(C1-C6)烷基;
R3為羥基或-O(C1-C6)烷基;
其中,(C1-C6)烷基為未被取代的或被以-NH2或-NH-PEG取代;
或其一同分異構物或一互變異構物或一混合物;或其一藥學上可接受之鹽類。
A compound according to claim 4, which is a compound of the formula (1c);
R 1 and R 4 are H;
R 2 is -O(C 1 -C 6 )alkyl or -OC(O)(C 1 -C 6 )alkyl;
R 3 is hydroxy or -O(C 1 -C 6 )alkyl;
Wherein (C 1 -C 6 )alkyl is unsubstituted or is taken as -NH 2 , Or -NH-PEG substituted;
Or a homoisomer or a tautomer or a mixture thereof; or a pharmaceutically acceptable salt thereof.
如申請專利範圍第4項所述之化合物,
其中,
R1與R4獨立地表示 H或不存在;
R2與 R3係獨立地選自H與羥基;
表示一單鍵或雙鍵;
伴隨一規定若R2與R3兩者為羥基,那麼R1與 R4為不存在的並且表示一雙鍵;
或其一同分異構物或一互變異構物或一混合物;或其一藥學上可接受之鹽類。
For example, the compound described in claim 4,
among them,
R 1 and R 4 independently represent H or absent;
R 2 and R 3 are independently selected from H and hydroxy;
Represents a single or double bond;
With the proviso that if both R 2 and R 3 are hydroxyl groups, then R 1 and R 4 are absent and Represents a double bond;
Or a homoisomer or a tautomer or a mixture thereof; or a pharmaceutically acceptable salt thereof.
如申請專利範圍第4項至第6項中的任一項所述之化合物,其中該化合物為:



或其一同分異構物或一互變異構物或一混合物;或其一藥學上可接受之鹽類。
The compound of any one of clauses 4 to 6, wherein the compound is:



Or a homoisomer or a tautomer or a mixture thereof; or a pharmaceutically acceptable salt thereof.
一種藥學組成物,其包含一治療有效量之如申請專利範圍第1項至第7項中的任一項所述的化合物或其一同分異構物或一互變異構物或一藥學上可接受之鹽類;以及一藥學上可接受之賦形劑、稀釋劑及/或載體。A pharmaceutical composition comprising a therapeutically effective amount of a compound according to any one of claims 1 to 7 or a homoisomer or a tautomer thereof or a pharmaceutically acceptable Accepted salts; and a pharmaceutically acceptable excipient, diluent and/or carrier. 如申請專利範圍第1項至第7項中的任一項所述之化合物,或其一同分異構物或一互變異構物或一藥學上可接受之鹽類;用於癌症治療的用途。A compound according to any one of claims 1 to 7, or a homoisomer or a tautomer or a pharmaceutically acceptable salt thereof; for use in cancer therapy . 一種用於如申請專利範圍第9項所述的用途之化合物,其中該癌症係選自白血病、肺癌、腦癌、非霍奇金氏淋巴瘤、霍奇金氏病、肝癌腎臟癌、膀胱癌、泌尿道癌、乳癌、頭頸部癌、子宮內膜癌、淋巴瘤、黑色素瘤、子宮頸癌、甲狀腺癌、胃癌、生殖細胞瘤、膽管癌、顱外癌、肉瘤、間皮瘤、骨惡性纖維組織細胞瘤、視網膜母細胞瘤、食道癌、多發性骨髓瘤、頭頸部癌、胰腺癌、室管膜瘤、神經母細胞瘤、皮膚癌、卵巢癌、復發性卵巢癌、前列腺癌、睪丸癌、大腸癌、淋巴增生性疾病、難治性多發性骨髓瘤、抗多發性骨髓瘤或骨髓增生性疾病。A compound for use according to the invention of claim 9, wherein the cancer is selected from the group consisting of leukemia, lung cancer, brain cancer, non-Hodgkin's lymphoma, Hodgkin's disease, liver cancer, kidney cancer, bladder cancer , urinary tract cancer, breast cancer, head and neck cancer, endometrial cancer, lymphoma, melanoma, cervical cancer, thyroid cancer, gastric cancer, germ cell tumor, cholangiocarcinoma, extracranial cancer, sarcoma, mesothelioma, bone malignancy Fibrosial cell tumor, retinoblastoma, esophageal cancer, multiple myeloma, head and neck cancer, pancreatic cancer, ependymoma, neuroblastoma, skin cancer, ovarian cancer, recurrent ovarian cancer, prostate cancer, testicular Cancer, colorectal cancer, lymphoproliferative disease, refractory multiple myeloma, anti-multiple myeloma or myeloproliferative disease. 一種用於如申請專利範圍第9項或第10項所述的用途之化合物,其中該癌症係選自急性淋巴性白血病、急性骨髓性白血病、成人急性骨髓性白血病、急性淋巴胚細胞白血病、慢性淋巴性白血病、慢性骨髓性白血病、毛細胞白血病、非小細胞肺癌、小細胞肺癌、腦幹神經膠瘤、神經膠母細胞瘤、包括小腦心臟細胞瘤與大腦心臟細胞瘤的心臟細胞瘤、視覺通路神經膠瘤、松果體瘤、髓母細胞瘤、原發性中樞神經系統淋巴瘤、套膜細胞淋巴瘤、霍奇金氏病、肝細胞癌、腎細胞癌、膀胱癌、泌尿道癌、骨肉瘤、乳癌、子宮內膜癌、口腔癌、黑色素瘤、子宮頸癌、甲狀腺癌、胃癌、骨惡性纖維組織細胞瘤、視網膜母細胞瘤、食道癌、多發性骨髓瘤、胰腺癌、神經母細胞瘤、皮膚癌、卵巢癌、前列腺癌、睪丸癌、大腸癌、淋巴增生性疾病或骨髓增生性疾病。A compound for use according to the invention of claim 9 or claim 10, wherein the cancer is selected from the group consisting of acute lymphocytic leukemia, acute myeloid leukemia, acute myeloid leukemia in adults, acute lymphoblastic leukemia, chronic Lymphocytic leukemia, chronic myelogenous leukemia, hairy cell leukemia, non-small cell lung cancer, small cell lung cancer, brain stem glioma, glioblastoma, cardiac cell tumor including cerebellar cardiac cell tumor and cerebral heart cell tumor, vision Accessory neurofibroma, pineal tumor, medulloblastoma, primary central nervous system lymphoma, mantle cell lymphoma, Hodgkin's disease, hepatocellular carcinoma, renal cell carcinoma, bladder cancer, urinary tract cancer , osteosarcoma, breast cancer, endometrial cancer, oral cancer, melanoma, cervical cancer, thyroid cancer, gastric cancer, bone malignant fibrous histiocytoma, retinoblastoma, esophageal cancer, multiple myeloma, pancreatic cancer, nerve Blastoma, skin cancer, ovarian cancer, prostate cancer, testicular cancer, colorectal cancer, lymphoproliferative disease or myeloproliferative disease. 一種用於如申請專利範圍第9項至第11項中的任一項所述之用途之化合物,其中該癌症係選自急性淋巴性白血病、急性骨髓性白血病、成人急性骨髓性白血病、急性淋巴胚細胞白血病、慢性淋巴性白血病、慢性骨髓性白血病、毛細胞白血病、非小細胞肺癌、小細胞肺癌、腦幹神經膠瘤、神經膠母細胞瘤、包括小腦心臟細胞瘤與大腦心臟細胞瘤的心臟細胞瘤、髓母細胞瘤、腎細胞癌、膀胱癌、泌尿道癌、乳癌、口腔癌、黑色素瘤、子宮頸癌、甲狀腺癌、胃癌、胰腺癌、前列腺癌或大腸癌。A compound for use according to any one of claims 9 to 11, wherein the cancer is selected from the group consisting of acute lymphocytic leukemia, acute myeloid leukemia, adult acute myeloid leukemia, acute lymphatic Embryonic leukemia, chronic lymphocytic leukemia, chronic myelogenous leukemia, hairy cell leukemia, non-small cell lung cancer, small cell lung cancer, brain stem neurotroph, glioblastoma, including cerebellar heart cell tumor and cerebral heart cell tumor Cardiomyoma, medulloblastoma, renal cell carcinoma, bladder cancer, urinary tract cancer, breast cancer, oral cancer, melanoma, cervical cancer, thyroid cancer, gastric cancer, pancreatic cancer, prostate cancer or colorectal cancer. 如申請專利範圍第8項所述之藥學組成物,其用於該癌症治療的用途。The pharmaceutical composition according to item 8 of the patent application, for use in the treatment of cancer. 一種如申請專利範圍第1項至第7項中的任一項所述之化合物;或其一同分異構物或一互變異構物或一藥學上可接受的鹽類之用於癌症治療的一藥劑的製造之用途。A compound according to any one of claims 1 to 7; or an isomer or a tautomer or a pharmaceutically acceptable salt thereof for use in cancer therapy The use of a pharmaceutical agent. 一種在一病患中的癌症治療方法;其包含給藥予該所述病患一治療有效量之如申請專利範圍第1項至第7項中的任一項所述之該化合物或其一同分異構物或一互變異構物或一藥學上可接受之鹽類。A method of treating cancer in a patient; comprising administering to the patient a therapeutically effective amount of the compound or any of the same according to any one of claims 1 to 7 Isomer or a tautomer or a pharmaceutically acceptable salt. 一種如申請專利範圍第1項所述的化合物之製備程序,其包含如下步驟;
a) 將該培養編號PM0895172或其變異型或突變型中之一者於水下需氧條件下,於包含碳、氮以及營養無機鹽及/或微量元素來源的營養培養基中生長,以獲得包含結構式(1)之化合物的一培養液;
b) 從該培養液分離該結構式(1)之化合物;以及
c) 純化該結構式(1)之化合物。
A preparation procedure for a compound as described in claim 1, which comprises the following steps;
a) growing one of the culture number PM0895172 or one of its variants or mutants under aerobic conditions under a nutrient medium comprising carbon, nitrogen and nutritive inorganic salts and/or trace elements to obtain a culture solution of a compound of formula (1);
b) separating the compound of formula (1) from the culture solution;
c) Purification of the compound of formula (1).
如申請專利範圍第16項所述之程序,其進一步包含將結構式(1)之一化合物轉變為其藥學上可接受之鹽類。The procedure of claim 16, further comprising converting a compound of formula (1) to a pharmaceutically acceptable salt thereof. 如申請專利範圍第16項所述之程序,其中在步驟(a)的該培養液包含結構式(1a)之該化合物與結構式(1b)之化合物的一混合物。The process of claim 16, wherein the culture solution in the step (a) comprises a mixture of the compound of the formula (1a) and the compound of the formula (1b). 如申請專利範圍第16項所述之程序,其中在步驟(b)該結構式(1)之化合物為該結構式(1a)之化合物。The process of claim 16, wherein the compound of the formula (1) in the step (b) is a compound of the formula (1a). 如申請專利範圍第16項所述之程序,其中在步驟(b)該結構式(1)之化合物為該結構式(1b)之化合物。The process of claim 16, wherein the compound of the formula (1) in the step (b) is a compound of the formula (1b). 如申請專利範圍第16項所述之程序,其中在步驟(c)該結構式(1)之化合物為該結構式(1a)之化合物。The process of claim 16, wherein the compound of the formula (1) in the step (c) is a compound of the formula (1a). 如申請專利範圍第16項所述之程序,其中在步驟(c)該結構式(1)之化合物為該結構式(1b)之化合物。The process of claim 16, wherein the compound of the formula (1) in the step (c) is a compound of the formula (1b).
TW102118552A 2013-05-24 2013-05-24 Hexadepsipeptide analogues as anticancer compounds TW201444864A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
TW102118552A TW201444864A (en) 2013-05-24 2013-05-24 Hexadepsipeptide analogues as anticancer compounds

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
TW102118552A TW201444864A (en) 2013-05-24 2013-05-24 Hexadepsipeptide analogues as anticancer compounds

Publications (1)

Publication Number Publication Date
TW201444864A true TW201444864A (en) 2014-12-01

Family

ID=52706898

Family Applications (1)

Application Number Title Priority Date Filing Date
TW102118552A TW201444864A (en) 2013-05-24 2013-05-24 Hexadepsipeptide analogues as anticancer compounds

Country Status (1)

Country Link
TW (1) TW201444864A (en)

Similar Documents

Publication Publication Date Title
US20160354430A1 (en) Hexadepsipeptide analogues as anticancer compounds
KR101764349B1 (en) A novel flavimycin compound having peptide deformylayse inhibition and antibacterial activity
KR101723649B1 (en) Novel peptide compound, a preparing method thereof, and a use thereof
JP2003523383A (en) Novel compositions and uses of dictyostatin compounds
WO2013144894A1 (en) Nucleoside analogue as an anticancer compound
TW201444875A (en) Hexadepsipeptide analogues as anticancer compounds
TW201444864A (en) Hexadepsipeptide analogues as anticancer compounds
US8598313B2 (en) Compositions for ameliorating cell proliferative disorders and methods of making and using them
US20030083230A1 (en) Reversal of multidrug resistance in human colon carcinoma cells
CA2649480A1 (en) Antibacterial compounds comprising multiple thiazole rings isolated fromkocuria species
NAKANO et al. Novel inhibitors of superoxide anion generation, OPC-15160 and OPC-15161 taxonomy, fermentation, isolation, physico-chemical properties, biological characteristics and structure determination
JP5461082B2 (en) A novel microorganism belonging to the genus Streptomyces, a novel compound produced by the microorganism, and a pharmaceutical comprising the compound as an active ingredient
WO2014106826A2 (en) Anthracycline analogue and uses thereof
JP2011116662A (en) New antibiotic sf2876 substance, method for producing the same, and pharmaceutical composition
US7470714B2 (en) GM-95-containing antitumor effect potentiator, combined antitumor preparation and antitumor agent
US8263579B2 (en) Anti-inflammatory compounds
Cooreman et al. Emerging pharmaceutical therapies of Ascidian-derived natural products and derivatives
US20130130992A1 (en) Dipeptide derivative for the treatment of cancer
US7439225B2 (en) Substances K01-B0171 and process for producing the same
JP5641716B2 (en) Novel compounds produced by microorganisms belonging to the genus Streptomyces, microorganisms producing the compounds, and pharmaceutical preparations containing the compounds as active ingredients
KR100225458B1 (en) Myxococcus spantitumor compound from myxococcus sp. and process for preparation thereof
JP2007238453A (en) Novel cyclic thiopeptide and method for acquiring the same
JP5578649B2 (en) A novel microorganism belonging to the genus Actinomadura, a novel compound produced by the microorganism, and a pharmaceutical comprising the compound as an active ingredient
JP2020075900A (en) Fki-7019 material and method for producing the same
JP2016179953A (en) Novel sagamilactam substance and method for producing the same