TW201210602A - Phosphorus-containing prodrugs and their therapeutic use - Google Patents

Phosphorus-containing prodrugs and their therapeutic use Download PDF

Info

Publication number
TW201210602A
TW201210602A TW99129443A TW99129443A TW201210602A TW 201210602 A TW201210602 A TW 201210602A TW 99129443 A TW99129443 A TW 99129443A TW 99129443 A TW99129443 A TW 99129443A TW 201210602 A TW201210602 A TW 201210602A
Authority
TW
Taiwan
Prior art keywords
group
compound
nucleoside
formula
limited
Prior art date
Application number
TW99129443A
Other languages
Chinese (zh)
Inventor
Zheng Wang
Run-Cong Liu
Original Assignee
Zheng Wang
Run-Cong Liu
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 Zheng Wang, Run-Cong Liu filed Critical Zheng Wang
Priority to TW99129443A priority Critical patent/TW201210602A/en
Publication of TW201210602A publication Critical patent/TW201210602A/en

Links

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates to phosphorus-containing prodrug of formula I: wherein the symbols are as defined in the specification, and of pharmaceutically acceptable salts and prodrugs thereof, for effectively delivering biologically active phosphonates including nucleoside phosphonates and nucleoside monophosphates in biological system. The present invention also relates to the therapeutic uses of compound of formula I.

Description

201210602 六、發明說明: 本專利申請案主張美國臨時申請案 6 1 /2 75,72 8 (2 00 9 年9月1日申請)之優先權之利益。上述參照之申請案之揭 示以其全文倂於本文參考。 【發明所屬之技術領域】 本發明一般而言係關於含磷前藥及其用於治療及預防 病毒感染及癌症之治療用途,特別是肝炎感染及包括癌症 之其他肝臟疾病。 【先前技術】 藥劑引起之毒性及藥理副作用通常係由藥劑或藥劑代 謝物與組織中正常細胞生長或維持所需之代謝路徑相互作 用或干擾而引起。·在許多情況下,由於在標的組織中劑量 限制性毒性或不調當之藥劑濃度而難以達到所欲之藥理作 用。 核苷類似物已被硏發作爲抗病毒及抗癌劑。前者中, 相較於細胞酵素,病毒編碼核苷激酶及NA (DNA及RNA) 聚合酶之受質特異性較少被開發。核苷酸激酶將核苷磷酸 化成爲其對應之5’-單磷酸酯,其進一步藉由細胞核苷酸激 酶轉化成其之二-及三-磷酸酯。僞核苷三磷酸酯選擇性地 與病毒NA聚合酶作用;被倂入延伸之NA中,且在許多情 況下,終止ΝΑ鏈(當3’位置無〇Η基時),或含有核苷類似 物的此等ΝΑ喪失其正常功能。在後者,快速分化的癌細 胞以比正常細胞更高的頻率將僞核苷倂入核酸。在任何情 況下,核苷藥劑藉由細胞激酶經單磷酸酯及二磷酸酯轉化 成其之對應三磷酸酯而被活化。 -4- 201210602 疱疹病毒(Herpesvirus)爲一典型之DNA病毒’病 苷激酶相當選擇性地磷酸化核苷類似物,在進一步經 激酶轉化成其對應之三磷酸酯之後,其抑制正常功能 毒核酸之製造。人類免疫缺損病毒(HI V)及B型肝炎 (HBV)爲DNA依賴性RNA病毒,病毒RNA之遺傳信 轉錄至 DNA(反轉錄),然後根據 DNA 製造 RNA(messenger RNA),於此等病毒中經核苷藥劑抑制 標幾乎皆爲反轉錄作用。雖然少數潛能核苷藥劑正 發,但尙未獲得對於C型肝炎病毒(HCV)感染非常有 治療。HCV爲RNA依賴性RNA病毒,在所有情況中 苷藥劑被活化成對應之核苷三磷酸酯。 現已得知一些核苷爲弱活性,僅因爲其不能有效 激酶磷酸化或完全不是激酶的受質,如同一些不活 苷,當化學性轉化成其之三磷酸酯,在活體外成爲強 力有效對抗某些病毒。核苷磷酸酯(核苷酸)就其本身 通常並不能作爲藥劑,因爲其在進入細胞前被膜核苷 其他水解酶去磷酸化,或太過極性而不能進入細胞。 改善核苷之生物活性,其之磷酸酯前藥已被徹底硏究 爲其可潛在性地越過速率限制性第一步驟磷酸化作用 來,胺基磷酸酯前藥路徑已被證實是越過磷酸化作用 率限制性第一步驟將非活化核苷生物轉化成活化核苷 酸酯的有效方法[/. Med. 5 0 (22),5463 - 2007]。核苷胺基磷酸酯已被報導爲有效輸送核苷5’ 酸酯進入肝臟(WO 2008/ 1 2 1 634 ; WO 2008/082601 J 2008/082602)。近年來,有許多專利申請案揭示利用 毒核 細胞 的病 病毒 息被 信使 之目 在硏 效之 ,核 地被 化核 而有 而言 酸及 爲了 ,因 。近 之速 單磷 5470, -單磷 ^ WO 胺基 201210602 磷酸酯作爲前藥以輸送核苷單磷酸酯至組織,特別是輸送 至肝臟(us 6455513、WO 2009/052050、WC) 2008/121634、 WO 200 8/0 8 3 3 1 0 1、WO 2 00 8/0 622 06、WO 2007/00293 1、 WO 200 8/0 8 5 5 0 8 ' W Ο 2 0 0 7 / 0 9 5 2 6 9 ' W Ο 2 0 0 6 / 0 1 2 0 7 8 ' WO 2006/100439)。核苷單磷酸酯可進一步磷酸化成爲二磷酸 酯,之後成爲生物活性之三磷酸酯。 然而,因爲潛在性神經毒性及因由前藥釋放之酚所引 起之肝腎損傷,基於麥氏(McGuigan)技術(US 645 5 5 1 3 )之 胺基隣酸酯路徑是有所限制(Carcinogenesis 1993,14: 2477-24 86 ; Mutat. Res. 1 99 1, 249,1 : 2 Ό 1 - 2 0 9)。 核苷之胺基磷酸酯在細胞株分析中通常可顯示出最大 生物活性,因爲其可快速釋放核苷或核苷酸進入細胞。已 報導d4T之胺基磷酸酯前藥在口服投與後並不能在血漿中 被偵測出(Drug Metab. Dispos. 2001,29,1035)。胺基磷酸 酯在胃液中穩定,且可在胃中被吸收,其於血液中快速水 解。另一方面,胺基磷酸酯在腸液中很快分解成 ala-d4T-MP,此代謝物可在腸道中被吸收,並進一步代謝 而產生核苷d4T。另一可能性爲此代謝物因其極性之性質 而並不能在腸道中被有效吸收。因此,此類胺基憐酸酯前 藥之生物可利用性相對較低,很可能是因爲其之醋酶敏感 性酯基。例如,GS-7340爲一種泰諾福韋(ten0f0vir)之異丙 基丙胺醯基單醯胺酯苯基單酯,相較於泰諾福韋靜脈內投 藥,其之生物可利用性在雄性米格魯(beagle)犬爲 17 % (Gilead Sciences, Antimicrob. Agents Chemother. 2005 49, 1898) » 201210602 核苷膦酸酯之二醯胺前藥亦已被硏究,且在某些情況 下,此路徑呈現改良效力及/或藥物動力學輪廓。然而,在 應用磷酸二醯胺至核苷上僅獲得非常有限的硏究(anti-HI V agents, FLT, AZT ; Polish J. Chem. 1 993, 67, 75 5 ; Drug Design and Discovery 1 99 5, 1 3, 4 3 ; Antiviral Chem.201210602 VI. INSTRUCTIONS: This patent application claims the priority of the US provisional application 6 1 /2 75,72 8 (application dated September 1, 2000). The disclosure of the above-referenced application is hereby incorporated by reference in its entirety. TECHNICAL FIELD OF THE INVENTION The present invention relates generally to phosphorus-containing prodrugs and their therapeutic use for the treatment and prevention of viral infections and cancer, particularly hepatitis infections and other liver diseases including cancer. [Prior Art] The toxicity and pharmacological side effects caused by the agent are usually caused by the interaction or interference of the agent or agent metabolite with the metabolic pathway required for the growth or maintenance of normal cells in the tissue. • In many cases, it is difficult to achieve the desired pharmacological effect due to dose-limiting toxicity or unadjusted drug concentration in the subject tissue. Nucleoside analogs have been used as antiviral and anticancer agents. In the former, the pharmacological specificity of the viral-encoded nucleoside kinase and NA (DNA and RNA) polymerase was less developed than that of the cytogene. Nucleotide kinases phosphorylate nucleosides to their corresponding 5'-monophosphates, which are further converted to their di- and tri-phosphates by cellular nucleotide kinases. The pseudonucleoside triphosphate selectively interacts with the viral NA polymerase; is incorporated into the extended NA, and in many cases terminates the ΝΑ chain (when the 3' position has no sulfhydryl group), or contains a nucleoside similar These defects of the substance lose their normal function. In the latter, rapidly differentiated cancer cells pour pseudonucleotides into nucleic acids at a higher frequency than normal cells. In any case, the nucleoside agent is activated by the conversion of the cellular kinase by the monophosphate and the diphosphate to its corresponding triphosphate. -4- 201210602 Herpesvirus is a typical DNA virus 'glycosidase kinase that phosphorylates nucleoside analogs quite selectively, inhibiting normal functional virulence nucleic acids after further kinase conversion to its corresponding triphosphate Manufacturing. Human immunodeficiency virus (HI V) and hepatitis B (HBV) are DNA-dependent RNA viruses, and the viral letter of the RNA is transcribed to DNA (reverse transcription), and then RNA is produced from DNA (messenger RNA). The nucleoside inhibitors are almost all reverse transcription. Although a small number of potential nucleoside agents are occurring, they have not been treated very well for hepatitis C virus (HCV) infection. HCV is an RNA-dependent RNA virus, and in all cases the glycoside agent is activated to the corresponding nucleoside triphosphate. It has been known that some nucleosides are weakly active, simply because they are not effective for kinase phosphorylation or not at all, and like some inactive glycosides, when chemically converted to their triphosphates, they become potent and effective in vitro. Fight against certain viruses. Nucleoside phosphates (nucleotides) are not normally used as agents because they are dephosphorylated by membrane nucleoside other hydrolases before entering the cell, or too polar to enter the cell. To improve the biological activity of nucleosides, the phosphate prodrugs have been thoroughly investigated for their potential to overcome the rate-limiting first step of phosphorylation, and the aminophosphate prodrug route has been shown to be over-phosphorylation. The rate-limiting first step is an efficient method for the biotransformation of a non-activated nucleoside to an activated nucleotide ester [/. Med. 5 0 (22), 5463 - 2007]. Nucleoside phosphoramidates have been reported to efficiently transport nucleoside 5' acid esters into the liver (WO 2008/1 2 1 634; WO 2008/082601 J 2008/082602). In recent years, there have been many patent applications that reveal that the use of virulence cells is a messenger. The nuclear nucleus is nucleated and the acid is used. Near-speed monophosphorus 5470, -monophosphorus ^ WO amine group 201210602 phosphate ester as a prodrug to transport nucleoside monophosphate to tissues, especially to the liver (us 6455513, WO 2009/052050, WC) 2008/121634, WO 200 8/0 8 3 3 1 0 1 , WO 2 00 8/0 622 06, WO 2007/00293 1, WO 200 8/0 8 5 5 0 8 ' W Ο 2 0 0 7 / 0 9 5 2 6 9 ' W Ο 2 0 0 6 / 0 1 2 0 7 8 ' WO 2006/100439). The nucleoside monophosphate can be further phosphorylated to a diphosphate followed by a biologically active triphosphate. However, due to the potential neurotoxicity and liver and kidney damage caused by the phenol released by the prodrug, the amino-based acid ester pathway based on McGuigan technology (US 645 5 5 13) is limited (Carcinogenesis 1993, 14: 2477-24 86 ; Mutat. Res. 1 99 1, 249,1 : 2 Ό 1 - 2 0 9). Aminophosphates of nucleosides generally exhibit maximum biological activity in cell line assays because they rapidly release nucleosides or nucleotides into cells. It has been reported that d4T amino phosphate prodrugs cannot be detected in plasma after oral administration (Drug Metab. Dispos. 2001, 29, 1035). Amino phosphates are stable in gastric juice and are absorbed in the stomach, which rapidly hydrolyzes in the blood. On the other hand, the amino phosphate is rapidly decomposed into ala-d4T-MP in the intestinal fluid, and this metabolite can be absorbed in the intestinal tract and further metabolized to produce nucleoside d4T. Another possibility is that the metabolite is not effectively absorbed in the intestine due to its polar nature. Therefore, the bioavailability of such amino-pureate prodrugs is relatively low, most likely due to their vinegar-sensitive ester groups. For example, GS-7340 is a isopropyl propyl sulfhydryl monodecyl phenyl monoester of tenofovir (ten0f0vir), which is bioavailable in male rice compared to the intravenous administration of tenofovir. The beagle dog is 17% (Gilead Sciences, Antimicrob. Agents Chemother. 2005 49, 1898) » 201210602 The nucleoside phosphonate diamine prodrug has also been studied and, in some cases, this The pathway presents improved efficacy and/or pharmacokinetic profile. However, only very limited studies have been obtained on the application of diammonium phosphate to nucleosides (anti-HI V agents, FLT, AZT; Polish J. Chem. 1 993, 67, 75 5 ; Drug Design and Discovery 1 99 5 , 1 3, 4 3 ; Antiviral Chem.

Chemother. 1992, 3, 107 ; 1 9 9 1,2, 3 5 ; 1995,6,50)。PMEA 二醯胺前藥並不能提供母藥令人滿意的生物可利用性,可 能是因爲其比泰諾福韋胺基膦酸酯還高的極性(J. Med. Chem. 200 8, 5 1, 4 3 3 1; Antimicrob. Agents Chemother. 2 0 0 5, 49, 1898)° 許多疾病可引起肝臟失調,包括肝炎感染(尤其是B型 及C型肝炎病毒)、肝癌,及代謝疾病,例如糖尿病、高血 脂症、動脈粥狀硬化及肥胖》關於此等疾病之治療,以肝 臟爲標的之前藥可有效降低藥劑暴露於其他器官,因此可 降低藥劑有關之副作用。例如,9_(2_膦醯基甲氧基乙基) 腺嘌呤(PME A)呈現非常低量的分佈,推測是因爲其高的負 電荷。在人類中,由於腎小管細胞底側表面上所存在之有 機陰離子輸送器,可幫助促進PMEA之腎清除,98%的投 藥藥劑經腎臟排泄。Ρ Μ E A治療與嚴重腎毒性有關,可能 是因爲PMEA及相關磷酸化形式暴露並累積於腎臟。 核苷酸或核苷之已知的胺基磷酸酯及二醯胺前藥以類 似之機制釋放母藥(如下反應式,J_ Med. Chem· 2008,51, 2 3 2 8)。例如,酯類前藥之水解(1)經由酯酶酵素開始,咸信 羧酸(2)受到分子間環化作用與酚之置換而形成短暫的五 員環中間產物(3 ), 201210602Chemother. 1992, 3, 107 ; 1 9 9 1, 2, 3 5 ; 1995, 6, 50). The PMEA diamine precursor does not provide satisfactory bioavailability of the parent drug, probably because it is more polar than tenofovir phosphinate (J. Med. Chem. 200 8, 5 1 , 4 3 3 1; Antimicrob. Agents Chemother. 2 0 0 5, 49, 1898) ° Many diseases can cause liver disorders, including hepatitis infections (especially hepatitis B and C viruses), liver cancer, and metabolic diseases, such as Diabetes, hyperlipidemia, atherosclerosis, and obesity. For the treatment of these diseases, the liver-based prodrug can effectively reduce the exposure of the agent to other organs, thereby reducing the side effects associated with the drug. For example, 9-(2-phosphonium methoxyethyl) adenine (PME A) exhibits a very low amount of distribution, presumably because of its high negative charge. In humans, due to the presence of an organic anion conveyor on the basal side of the tubular cells, it helps to promote renal clearance of PMEA, with 98% of the drug being excreted by the kidneys. Ρ Μ E A treatment is associated with severe nephrotoxicity, probably because PMEA and related phosphorylated forms are exposed and accumulate in the kidneys. Known aminophosphates and diamine precursors of nucleotides or nucleosides release the parent drug in a similar mechanism (the following reaction formula, J_ Med. Chem. 2008, 51, 2 3 2 8). For example, hydrolysis of an ester prodrug (1) begins with an esterase, and the carboxylic acid (2) undergoes intermolecular cyclization and phenol substitution to form a transient five-membered ring intermediate (3), 201210602

芳基胺基磷酸酯前藥之活化 其被水解成胺基磷酸(Phosphoramidic acid) (4)。單醯胺醋 裂解成活性形式(5)可經由第二酵素(胺基磷酸酶)催化,或 更酸性之次細胞區室中簡單水解產生(Molecular Pharmacol 1 999,56,69 3 ) ° 對於可被酯酶非依賴性機制裂解之膦酸酯前藥努力搜 尋已導致發現二種前藥類型,其已進展至人體臨床試驗, 即雙苯基酯類與 HepDirect 前藥(J. Med. Chem. 1 994,37, 498; J. Am. Chem. Soc. 2004, 1 26, 5 1 54; J. Pharmacol. Exp. Ther. 2005, 312, 554)。雙苄基酯亦已被硏究,但簡單之未 經取代苄基酯裂解太慢,以致於不能使用作爲前藥(Bioorg. Med. Chem. Lett. 2007,17,3412)。Erion 等人揭示環狀磷 酸酯或膦酸酯前藥,其在酯酶存在下是安定的,可增加肝 臟特定藥劑輸送(Erion,M et al US 73037 3 9及其參考資 料)。E r i ο η的前藥在肝臟經由P 4 5 0活化。然而,此路徑之 臨床應用可能被芳族代謝物引發之潛在有害副作用所限 制,且釋放生物活性磷酸酯或膦酸酯之效率相當不確定。 201210602 近來,雙[(對-甲氧基)苄基]膦酸酯前藥被報導其顯示 改善之安定性及增加細胞滲透性(Bioorg. Med. Chem. Lett. 2007,17,34 12)。然而’並沒有進一步的詳細說明。因爲 苄基醇及其氧化代謝物(苯甲酸)相對而言是非毒性化合 物,吾人硏究具有苄基之磷酸酯或膦酸酯前藥,其具有使 連接苄基至磷酸酯或膦酸酯之鍵不安定的取代基。此等前 藥令人驚訝地在細胞中有效釋放苄基部分,可能涉及經細 胞色素P(450)(CYP)酵素調控之去烷基化作用(J. Med. C h e m. 1967, 10,861; J. Chem. Soc. Perkin Trans. 1992,2, 1 1 4 5 )或其他機制或在更酸性之次細胞區室中之簡單水解 作用。 因此,本發明(1)提供新穎核苷、核苷酸、C-核苷、C-核苷酸之胺基磷酸酯及胺基膦酸酯前藥、膦酸酯及含醇類 之藥劑,與(2)藉由使用此等本發明之胺基磷酸酯及胺基 膦酸酯前藥,有效達成輸送母藥至細胞中,尤其是進入肝 臟細胞目標。本文中此等前藥可對抗酯酶。 【發明內容】 本發明係關於一種化合物,其用途爲具有增強之治療 潛能,特別是具有關於例如白血病之癌症、病毒感染(包括 HIV、HBV及HCV)及肝臟失調,包括肝癌及代謝疾病,例 如糖尿病、高血脂症、動脈粥狀硬化及肥胖之治療潛能。 本發明提供各種治療藥劑之核苷胺基磷酸酯及胺基膦 酸酯化合物,包括其之製造方法及在治療各種通常之失調 及特別是肝臟失調上之用途。在一些具體實施例中可使用 201210602 此化口物在肝臟准予高濃度之治療藥劑。在一具體實施 例’該化合物爲一種經取代苄基胺基磷酸酯及胺基膦酸酯。 如本文中所使用’ 「核苷胺基磷酸酯或胺基膦酸酯作 爲治療藥劑」包括衍生成胺基磷酸酯及胺基膦酸酯之核苷 (a非環狀核甘及C -核苷)治療藥劑,該胺基磷酸酯及胺基 鱗酸醋具有节基與下列取代基(但不以此爲限),例如胺 基、CrCw醯氧基、Cl_c2G烷基、芳基、C|-C2G烷氧基、 芳氧基或芳院氧基’皆可選擇性經取代。治療藥劑可例如 爲一種抗病毒藥劑,其包括或已被衍生成包括一反應基(例 如經基)’用以連接胺基磷酸酯,或胺基膦酸酯基團。此治療 藥劑包括核苷類、核苷類似物,包括非環狀核苷類、c_核 苷類及含醇藥劑,但不以此爲限。在一些具體實施例,亦 提供核苷及核苷酸類似物之胺基磷酸酯,例如1,-、2,-、3,-及4’-支鏈化或經二取代之核苷類之胺基磷酸酯。可投與有 效量之此類化合物以治療感染性疾病、肝臟失調,包括癌 症及感深性疾病,例如B型肝炎及C型肝炎感染,包括其 之抗藥株。 在某些具體實施例,儘管不受限於任何理論,但很可 能母藥在肝臟中主要由胺基磷酸酯或胺基膦酸酯化合物之 選擇性代謝獲得。因此,母藥能夠在宿主肝臟中累積。經 由在肝臟中選擇性標定及活化化合物,可降低活性化合物 在胃腸道可能非所欲的分佈。此外,活性化合物在肝臟感 染位置之治療量可被增加。 在某些具體實施例,母核苷(或核苷衍生物)藥劑之單 磷酸酯或膦酸酯在肝臟由胺基磷酸酯或胺基膦酸酯化合物 -10- 201210602 之代謝形成,使單磷酸酯或膦酸酯形成並在宿主肝臟中累 積。因此,在某些具體實施例,本文所揭示之胺基磷酸酯 或胺基膦酸酯提供一種在肝臟之核苷或核昔類似物的安定 化磷酸酯或膦酸酯前藥。在某些具體實施例中’其中化合 物需要被三磷酸化而激活,此可優異地除去對於最初磷酸 化步驟之需求,促進更簡易地形成活性三磷酸酯’其抑制 標的酵素,並可增強核苷或核苷類似物之整體活性。 在某些具體實施例,本文所揭示含醇藥劑之胺基磷酸 酯或胺基膦酸酯前藥有效地輸送母藥進入肝臟’其增加母 藥之生物可利用性及/或藥物動力,並降低母藥暴露於其他 器官。 —些核苷爲生物上非活性,僅僅是因爲其不能被激酶 磷酸化。在某些具體實施例,衍生自此等核苷之本發明前 藥可變得具生物活性,因爲該前藥直接輸送核苷單磷酸酯 越過磷酸化之速率限制第一步驟。 不受限於任何理論,在一具體實施例中,提供核苷之 胺基磷酸酯或胺基膦酸酯,例如5-氟-2’-去氧尿苷,其在 口服後於肝臟選擇性濃縮,並於肝臟細胞代謝,產生5 ’ -單磷酸酯,其抑制胸腺嘧啶合成酶。如此,相較於投與母 核苷分子,可降低潛在性治療劑量。此等前藥可有效降低 被嘧啶核苷磷酸化酶分解5-氟-2’-去氧尿苷成更具毒性之 5 -氟尿嘧啶(5-f luorour acil),以便此等前藥可以經口投與 取代灌注。 因此,在一些具體實施例中,於經口投與本文所述之 胺基磷酸酯及胺基膦酸酯化合物後,該化合物可有利地於 -11 - 201210602 標的肝臟細胞被濃縮,並轉換成其之單磷酸酯或鱗酸酯, 然後進一步被磷酸化以產生其之治療效果。 因爲此等方法能在宿主肝臟累積本文所述之核苷磷酸 酯或膦酸酯化合物,故本文所述之方法可用於例如治療及/ 或預防肝臟疾病或失調,例如肝癌、B型肝炎或C型肝炎。 亦提供一種治療肝臟失調之方法,包括投與有效量之 本文所提供之化合物,其可選擇在醫藥可接受載劑中單獨 或合倂其他治療有效藥劑投與,或者與其他治療有效藥劑 交替投與。 經由在苯環上之取代基所取得之苄基-OP鍵之不安定 化的優點,已發現母藥之胺基磷酸酯或胺基膦酸酯的此種 類苄基酯爲強效的治療藥劑,其直接輸送母藥進入組織。 具有高親脂性之本發明前藥快速穿過細胞膜,以便改善母 藥之藥力動力學及/或生物可利用性。此等前藥可經由肝臟 中富含之P450及/或其他酵素活化。此化合物亦可用於一 些具體實施例中以增加輸送母藥至肝臟。提供本發明各種 治療藥劑之胺基磷酸酯及胺基膦酸酯化合物形式,及其於 '冶療肝炎感染(特別是B型及C型肝炎病毒)及肝臟失調上 之用途,包括癌症、瘧疾及纖維化及代謝性疾病,例如糖 尿病、高血脂症、動脈粥狀硬化及肥胖。 【實施方式】 在一具體實施例,本發明提供一種式I化合物: -12- 201210602 Ο Μ^Χ—ρ—q R1Activation of the arylaminophosphate prodrug is hydrolyzed to Phosphoramidic acid (4). The cleavage of the monoamidamine into the active form (5) can be catalyzed by a second enzyme (aminophosphatase) or by simple hydrolysis in the cell compartment of the acid (Molecular Pharmacol 1 999, 56, 69 3 ) ° Efforts to search for phosphonate prodrugs that are cleaved by an esterase-independent mechanism have led to the discovery of two prodrug types that have progressed to human clinical trials, namely diphenyl esters and HepDirect prodrugs (J. Med. Chem. 1 994, 37, 498; J. Am. Chem. Soc. 2004, 1 26, 5 1 54; J. Pharmacol. Exp. Ther. 2005, 312, 554). Dibenzyl esters have also been studied, but the simple unsubstituted benzyl ester cleavage is too slow to be used as a prodrug (Bioorg. Med. Chem. Lett. 2007, 17, 3412). Erion et al. disclose cyclic phosphonate or phosphonate prodrugs which are stable in the presence of esterases and which increase the delivery of specific agents in the liver (Erion, M et al US 73037 39 and its reference). The prodrug of E r i ο η is activated in the liver via P 4 50 . However, the clinical application of this pathway may be limited by the potentially harmful side effects caused by aromatic metabolites, and the efficiency of releasing biologically active phosphates or phosphonates is rather uncertain. 201210602 Recently, bis[(p-methoxy)benzyl]phosphonate prodrugs have been reported to exhibit improved stability and increased cell permeability (Bioorg. Med. Chem. Lett. 2007, 17, 34 12). However, there is no further detailed explanation. Since benzyl alcohol and its oxidative metabolite (benzoic acid) are relatively non-toxic compounds, we have a phosphate or phosphonate prodrug with a benzyl group which has a linkage to a benzyl group to a phosphate or phosphonate. The bond is unstable. These prodrugs surprisingly release benzyl moieties in cells, possibly involving dealkylation controlled by cytochrome P(450) (CYP) enzymes (J. Med. C he m. 1967, 10,861; J. Chem. Soc. Perkin Trans. 1992, 2, 1 1 4 5 ) or other mechanisms or simple hydrolysis in the more acidic subcellular compartment. Accordingly, the present invention (1) provides a novel nucleoside, a nucleotide, a C-nucleoside, a C-nucleotide aminophosphate and an aminophosphonate prodrug, a phosphonate, and an alcohol-containing agent, And (2) by using the aminophosphate and aminophosphonate prodrugs of the present invention, the delivery of the parent drug to the cells, particularly into the liver cell target, is effectively achieved. These prodrugs herein are capable of combating esterases. SUMMARY OF THE INVENTION The present invention relates to a compound for use in enhancing therapeutic potential, particularly for cancers such as leukemia, viral infections (including HIV, HBV and HCV) and liver disorders, including liver cancer and metabolic diseases, for example Therapeutic potential of diabetes, hyperlipidemia, atherosclerosis and obesity. The present invention provides nucleoside amino phosphates and aminophosphonate compounds of various therapeutic agents, including methods for their manufacture and use in the treatment of various conventional disorders, particularly liver disorders. In some embodiments, 201210602 can be used to administer a high concentration of therapeutic agent in the liver. In a specific embodiment, the compound is a substituted benzylamino phosphate and an aminophosphonate. As used herein, 'a nucleoside amino phosphate or an amino phosphonate as a therapeutic agent' includes nucleosides derived from amino phosphates and aminophosphonates (a non-cyclic nucleoside and C-nucleus) The therapeutic agent, the amino phosphate and the uric acid vinegar having a benzyl group and the following substituents (but not limited thereto), such as an amine group, a CrCw decyloxy group, a Cl_c2G alkyl group, an aryl group, C| -C2G alkoxy, aryloxy or aryloxy can be optionally substituted. The therapeutic agent can, for example, be an antiviral agent that includes or has been derivatized to include a reactive group (e.g., a trans-group) to attach an amino phosphate, or an amine phosphonate group. Such therapeutic agents include, but are not limited to, nucleosides, nucleoside analogs, including acyclic nucleosides, c-nucleosides, and alcohol-containing agents. In some embodiments, aminophosphates of nucleosides and nucleotide analogs, such as 1,-, 2,-, 3,- and 4'-branched or disubstituted nucleosides, are also provided. Amino phosphate. Effective amounts of such compounds can be administered to treat infectious diseases, liver disorders, including cancers and deep-rooted diseases, such as hepatitis B and hepatitis C infections, including resistant strains thereof. In certain embodiments, although not limited to any theory, it is likely that the parent drug is primarily obtained in the liver by selective metabolism of an amino phosphate or an amino phosphonate compound. Therefore, the parent drug can accumulate in the host liver. By selectively calibrating and activating the compound in the liver, the undesired distribution of the active compound in the gastrointestinal tract can be reduced. In addition, the therapeutic amount of active compound at the site of liver infection can be increased. In certain embodiments, the monophosphate or phosphonate of the parent nucleoside (or nucleoside derivative) agent is formed in the liver by the metabolism of an amino phosphate or an amino phosphonate compound-10-201210602, such that Phosphate or phosphonate forms and accumulates in the host liver. Thus, in certain embodiments, the amino phosphate or aminophosphonate disclosed herein provides a stabilized phosphate or phosphonate prodrug of a nucleoside or a nuclear analog of the liver. In certain embodiments, 'where the compound needs to be activated by triphosphorylation, this advantageously removes the need for an initial phosphorylation step, promotes easier formation of the active triphosphate', its inhibitory enzyme, and enhances the nucleus. The overall activity of the glucoside or nucleoside analog. In certain embodiments, the amine phosphate or alanine phosphonate prodrugs of the alcohol-containing agents disclosed herein effectively deliver the parent drug into the liver, which increases the bioavailability and/or drug motility of the parent drug, and Reduce the exposure of the parent drug to other organs. - Some nucleosides are biologically inactive simply because they are not phosphorylated by kinases. In certain embodiments, prodrugs of the invention derived from such nucleosides can become biologically active because the prodrug directly transports the nucleoside monophosphate to limit the first step by the rate of phosphorylation. Without being bound by any theory, in one embodiment, an aminophosphate or an aminophosphonate of a nucleoside, such as 5-fluoro-2'-deoxyuridine, which is selective for hepatic administration after oral administration is provided. Concentrated and metabolized in liver cells to produce 5'-monophosphate, which inhibits thymidine synthetase. Thus, the potential therapeutic dose can be reduced as compared to the administration of the parent nucleoside molecule. These prodrugs are effective in reducing the decomposition of 5-fluoro-2'-deoxyuridine by pyrimidine nucleoside phosphorylase into a more toxic 5-fluorouracil (5-f luorour acil) so that these prodrugs can be administered orally. Cast and replace perfusion. Thus, in some embodiments, after oral administration of the amino phosphate and alanine phosphonate compounds described herein, the compound can advantageously be concentrated in liver cells of the -11 - 201210602 scale and converted into The monophosphate or sulphate is then further phosphorylated to produce its therapeutic effect. Because such methods are capable of accumulating the nucleoside phosphate or phosphonate compounds described herein in the host liver, the methods described herein can be used, for example, to treat and/or prevent liver disease or disorders, such as liver cancer, hepatitis B or C. Hepatitis. Also provided is a method of treating a liver disorder comprising administering an effective amount of a compound provided herein, optionally in a pharmaceutically acceptable carrier, alone or in combination with another therapeutically effective agent, or alternatively with other therapeutically effective agents. versus. The benzyl ester of this type of aminophosphate or aminophosphonate of the parent drug has been found to be a potent therapeutic agent by virtue of the instability of the benzyl-OP bond obtained by the substituent on the phenyl ring. It directly transports the parent drug into the tissue. The prodrugs of the present invention having high lipophilicity rapidly pass through the cell membrane to improve the pharmacokinetics and/or bioavailability of the parent drug. These prodrugs can be activated by P450 and/or other enzymes enriched in the liver. This compound can also be used in some embodiments to increase delivery of the parent drug to the liver. Providing the form of aminophosphate and alanine phosphonate compounds of various therapeutic agents of the invention, and their use in treating hepatitis infections (especially hepatitis B and C viruses) and liver disorders, including cancer, malaria And fibrotic and metabolic diseases such as diabetes, hyperlipidemia, atherosclerosis and obesity. [Embodiment] In one embodiment, the present invention provides a compound of formula I: -12- 201210602 Ο Μ^Χ—ρ—q R1

或其醫藥可接受前藥、 變異構或多形性形式, 其中: X選自氧(〇)、CH2’但不以此爲限 代表核苷或非環狀 倘若X爲氧(0),Μ-ΧΗ爲Μ-ΟΗ, 核苷、C-核苷或含醇化合物; Μ-0-Ρ(0)(0Η)2代表核苷或非環狀核苷或c_核苷之生 物學上活性的單磷酸酯; M-X可爲環狀或非環狀系統; 包括式 Μ-Χ-Ρ(0)(0Η)2爲一生物活性膦酸酯 M-CH2-P(0)(0H)2之核苷膦酸酯,倘若X及p經 • P鍵 連接; R1選自經基、卞氧基、院氧基、芳氧基、芳院氧基 烷基芳氧基、胺基酸殘基、苄基胺基、烷基胺基、芳基 基、烷基芳基胺基、芳基烷基胺基、二烷基胺基、〜 胺基,但不以此爲限:二烷基胺基之二烷基可形成環,& ^ 較 佳爲4至7員環,例如吡咯啶,其全部可選擇經取代; R2選自Η、烷基、芳基、炔基、烯基、環烷基、或具 有一至三個雜原子(例如0、S、Ν)之雜環基或雜芳基,伸 不以此爲限,其全部可選擇經取代; 201210602 R3爲一取代基,至少一個此類之取代基在各種位置連 接至苯環,較佳位於2及/或4-位置,其選自R2C〇〇 (醯氧 基)、醯基-NH、CH3、甲氧基、烷基、烷氧基、烷基胺基、 環烷基、環烷氧基、環烷基胺基、芳基、芳氧基、芳基胺 基、芳基烷基,但不以此爲限,其全部可選擇經取代; Y代表苯環上各種位置上之Η或一至三個取代基,包 括鹵素(I、Cl、Br、F)、CN、疊氮基 '醯氧趣、醯基胺基、 烷氧基、烷基胺基、烯基、烯基胺基、烯氧基、炔基、炔 氧基、炔基胺基、芳基烷氧基、芳基烷基胺基、烷氧基擬 基,但不以此爲限,其全部可選擇經取代; R1及[M-X]可一起形成環磷酸酯,例如核苷3,,5,_環碟 酸酯; M-X中之胺基及/或羥基可選擇性經保護;式I化合物 可爲一種由對掌性磷中心所造成之非鏡像異構物或該非鏡 像異構物之混合物,例如Ia及1 b :Or a pharmaceutically acceptable prodrug, metameric or polymorphic form thereof, wherein: X is selected from the group consisting of oxygen (oxime), CH2' but not limited thereto to represent nucleoside or acyclic if X is oxygen (0), Μ -ΧΗ is Μ-ΟΗ, nucleoside, C-nucleoside or alcohol-containing compound; Μ-0-Ρ(0)(0Η)2 represents biological activity of nucleoside or acyclic nucleoside or c_nucleoside Monophosphate; MX can be a cyclic or acyclic system; including the formula Μ-Χ-Ρ(0)(0Η) 2 is a biologically active phosphonate M-CH2-P(0)(0H)2 Nucleoside phosphonate, if X and p are linked by a P bond; R1 is selected from the group consisting of a thiol group, a decyloxy group, an alkoxy group, an aryloxy group, an aryloxyalkyl aryloxy group, an amino acid residue, Benzylamino group, alkylamino group, aryl group, alkylarylamino group, arylalkylamino group, dialkylamino group, ~amino group, but not limited to: dialkylamino group The dialkyl group may form a ring, and & ^ is preferably a 4 to 7 membered ring, such as pyrrolidine, all of which may be optionally substituted; R2 is selected from the group consisting of anthracene, alkyl, aryl, alkynyl, alkenyl, cycloalkane a heterocyclic group or a heteroaryl group having one to three hetero atoms (for example, 0, S, fluorene) And all of them may be optionally substituted; 201210602 R3 is a substituent, at least one such substituent is attached to the phenyl ring at various positions, preferably at the 2 and/or 4-position, which is selected from R2C〇〇 (醯Oxy), mercapto-NH, CH3, methoxy, alkyl, alkoxy, alkylamino, cycloalkyl, cycloalkoxy, cycloalkylamino, aryl, aryloxy, aryl Amino group, arylalkyl group, but not limited thereto, all of which may be optionally substituted; Y represents oxime or one to three substituents at various positions on the benzene ring, including halogen (I, Cl, Br, F) ), CN, azido-oxime, mercaptoamine, alkoxy, alkylamino, alkenyl, alkenylamino, alkenyloxy, alkynyl, alkynyloxy, alkynylamino, An arylalkoxy group, an arylalkylamino group, an alkoxy-based group, but not limited thereto, all of which may be optionally substituted; R1 and [MX] may together form a cyclic phosphate such as nucleoside 3, , 5, _ ring disc acid ester; the amine group and / or hydroxyl group in MX can be selectively protected; the compound of formula I can be a non-image isomer or the non-image isomerism caused by the palmitic phosphorus center The mixture, for example Ia and 1 b:

〇 R2 Μ—Χ»-ρ—(〇 R2 Μ—Χ»-ρ—(

Π -14- 201210602 其中: Z爲於苯環各種位置上之一取代基,其選自H、F、Cl、 Br、I、疊氮基、CN、CONH2、COOR2、OR2' SR2、COSR2' O-COR2、NH2、NH-醯基、NR2-醯基、但不以此爲限,其 全部可選擇經取代; R4選自R2C〇〇 (醯氧基)、醯基-NH、烷基、烷氧基、 烷基胺基、環烷基、環烷氧基、環烷基胺基、芳基、芳氧 基、芳基胺基、芳基烷基,但不以此爲限,其全部可選擇 經取代; R2如上述定義; R5選自R2CO (醯基)、CH3、烷基、環烷基、芳基、芳 基烷基、烷基芳基,但不以此爲限,其全部可選擇經取代。 舉例而言,式I化合物較佳可爲式Γ化合物: ηΠ -14- 201210602 wherein: Z is a substituent at various positions of the phenyl ring selected from the group consisting of H, F, Cl, Br, I, azide, CN, CONH2, COOR2, OR2' SR2, COSR2' O -COR2, NH2, NH-fluorenyl, NR2-indenyl, but not limited thereto, all of which may be optionally substituted; R4 is selected from the group consisting of R2C〇〇 (decyloxy), fluorenyl-NH, alkyl, alkane Oxyl, alkylamino, cycloalkyl, cycloalkoxy, cycloalkylamino, aryl, aryloxy, arylamino, arylalkyl, but not limited thereto, all of which are R2 is selected as defined above; R5 is selected from the group consisting of R2CO (fluorenyl), CH3, alkyl, cycloalkyl, aryl, arylalkyl, alkylaryl, but not limited thereto, all of which may be The choice is replaced. For example, the compound of formula I is preferably a compound of formula: η

Μ—X— 熟悉技術者應可理解經由例如於該藥劑羥基上經由例 如縮合或脫水之反應可製備式I化合物。此等衍生物可例 如經由自藥劑之羥基除去氫基而製備,例如在脫水反應 中。關於膦酸酯,例如泰諾福韋,二個羥基係被衍生成雙 胺基膦酸酯。 本文所提供爲化合物、組成物及用於治療肝炎感染 (HBV及HCV)及肝臟失調之方法,例如治療癌症,或代謝 性疾病’例如糖尿病、高血脂症、動脈粥狀硬化及肥胖。 •15- 201210602 當提及本文所提供之化合物,除另有指明外,下列術 語具有下列意義。 如本文中所使用之「烷基」一詞包括飽和直鏈、分支 或環狀之通常爲(:,至C2G之選擇地經取代烴類,具體而言 包括甲基、cf3、cci3、cfci2、cf2ci、乙基、ch2cf3、 cf2cf3、丙基、異丙基、環丙基等。烷基可被取代之基團 的非限制性實例選自由鹵素(氟、氯、溴或碘)、羥基、胺 基、烷基胺基、芳基胺基、烷氧基、芳氧基、硝基、氰基 等所組成之群組。 「烯基」包括單價烯烴類不飽和烴基,在某些具體實 施例,具有多達11個碳原子,其可爲直鏈或分支,且具有 至少1個或1至2個位置之烯類不飽和。例示性烯基包括 乙烯基(ethenyl,-CH = CH2)、正丙烯基(_CH2CH = CH2)、異 丙傭基(-C(CH3) = CH2)、乙烯基(vinyl)及經取代之乙稀基 等。 「快基」包括炔類不飽和烴類,在某些具體實施例中, 具有多達11個碳原子,其可爲直鏈或分支,且具有至少i 個或1至2個位置之快類不飽和。炔基之非限制性實例包 括乙炔基(acetylenic)、乙炔基(ethynyl)、块丙基等。 如本文中所使用之「芳基」一詞包括苯基、聯苯基或 萘基’且較佳爲苯基。該術語包括經取代及非經取代基團。 芳基可以任何所述基團取代’包括〜或多個選自由鹵素 (氟、氯、溴或碘)、烷基、羥基、胺基、烷基胺基、芳基 胺基、烷氧基、芳氧基、硝基、氰基、颯基(sulf〇n〇)'硫 酸根(sulfato)、膦醯基(phosphono)、磷酸根(ph〇sphate)或 -16 - 201210602 膦酸根(ph〇Sph〇n〇xy)所組成群組之基團,但不以此爲限, 而且可視需要非經保護或經保護。 「環院基」包括3_7員環之烴類,例如環丙基,其全 部可選擇經取代。 「雜環」包括於環中具有1-3個例如0、S、N之雜原 子的3-7員環碳化合物,其全部可選擇經取代。 「雜芳基」每括含1-3個例如0、S、N之雜原子的芳 族環’例如吡啶基、嘧啶基。 「烷氧基」包括- OR基,其中R爲烷基。特別是烷氧 基’包括正戊氧基、正己氧基、12·二甲基丁氧基等。 院氧基簾基」包括- C(O) -院氧基之殘基,其中院氧 基如本文之定義。 「胺基」包括- NH2殘基。 「羰基」包括-C(0)0H殘基。 「烷基胺基」或「芳基胺基」一詞包括各具有一或二 個烷基或芳基取代基之胺基。 「鹵素」或「鹵」包括氯(C1)、溴(Br)、氟(F)或碘(I)。 「單院基胺基」包括垸基- NHR’-,其中R,選自院基或 芳基。 「硫烷基」包括-SR基,其中R爲烷基或芳基。 如本文中所使用之「經保護」一詞除另有指明外包括 表示一基團被加至氧、氮或磷原子以防止其進一步反應或 用於其他目的。熟悉有機合成技術者已知有很多種氧及氮 保護基。 -17- 201210602 「核音」一詞包括天然或經改質之核苷、非環狀核苷 及C-核苷。 「C -核苷」一詞表示—種核苷,其中醣基鍵連接至經 修飾之核酸鹼基上的碳,而非—般核苷之氮(詳見對於c_ 核苷評述之參考資料:Chemistry of Nucleosides and Nucleotides by Leroy B Townsend 1 994,Science, Chapter 5Μ—X— It will be understood by those skilled in the art that the compound of formula I can be prepared via, for example, a reaction such as condensation or dehydration on the hydroxyl group of the agent. Such derivatives can be prepared, for example, by removing a hydrogen group from the hydroxyl group of the agent, for example, in a dehydration reaction. With regard to phosphonates, such as tenofovir, the two hydroxy groups are derivatized into bis-amino phosphonates. Provided herein are compounds, compositions, and methods for treating hepatitis infection (HBV and HCV) and liver disorders, such as treating cancer, or metabolic diseases such as diabetes, hyperlipidemia, atherosclerosis, and obesity. •15- 201210602 When referring to a compound provided herein, the following terms have the following meanings unless otherwise indicated. The term "alkyl" as used herein, includes saturated straight chain, branched or cyclic, usually (:, to C2G, optionally substituted hydrocarbons, specifically methyl, cf3, cci3, cfci2 Cf2ci, ethyl, ch2cf3, cf2cf3, propyl, isopropyl, cyclopropyl, etc. Non-limiting examples of groups in which an alkyl group may be substituted are selected from halogen (fluoro, chloro, bromo or iodo), hydroxyl, amine a group consisting of an alkyl group, an alkylamino group, an arylamine group, an alkoxy group, an aryloxy group, a nitro group, a cyano group, etc. "Alkenyl" includes a monovalent olefinic unsaturated hydrocarbon group, in certain embodiments Having up to 11 carbon atoms which may be straight or branched and having at least 1 or 1 to 2 positions of ethylenic unsaturation. Exemplary alkenyl groups include ethenyl (-CH = CH2), N-propenyl (_CH2CH = CH2), isopropyl (-C(CH3) = CH2), vinyl (vinyl) and substituted ethylene, etc. "fast base" includes acetylenically unsaturated hydrocarbons, In certain embodiments, there are up to 11 carbon atoms, which may be straight or branched, and have at least i or 1 to 2 positions. Non-limiting examples of alkynyl groups include acetylenic, ethynyl, propyl propyl, etc. As used herein, the term "aryl" includes phenyl, biphenyl or naphthyl. 'and preferably phenyl. The term includes both substituted and unsubstituted groups. The aryl group may be substituted with any of the groups', including ~ or more selected from halo (fluoro, chloro, bromo or iodo), alkyl , hydroxy, amine, alkylamino, arylamine, alkoxy, aryloxy, nitro, cyano, sulfoxime sulfato, phosphino ), a group of phosphate (ph〇sphate) or -16 - 201210602 phosphonate (ph〇Sph〇n〇xy), but not limited thereto, and may be unprotected or protected as needed. "Circular base" includes hydrocarbons of the 3-7-membered ring, such as cyclopropyl, all of which may be optionally substituted. "Heterocycle" includes 3- to 3, for example, 0, S, N heteroatoms in the ring. a 7-membered ring carbon compound, all of which may be optionally substituted. "Heteroaryl" includes an aromatic ring containing 1-3 hetero atoms such as 0, S, N, for example Alkyl and pyrimidinyl. "Alkoxy" includes -OR group, wherein R is alkyl. In particular, alkoxy' includes n-pentyloxy, n-hexyloxy, 12. dimethylbutoxy and the like. The "base group" includes a residue of -C(O)-homolyl, wherein the alkoxy group is as defined herein. "Amine" includes -NH2 residue. "Carbonyl" includes -C(0)0H residue. The term "alkylamino" or "arylamino" includes the radicals each having one or two alkyl or aryl substituents. "Halogen" or "halo" includes chlorine (C1), bromine (Br). , fluorine (F) or iodine (I). "Single-yard amine group" includes fluorenyl-NHR'-, wherein R is selected from a hospital group or an aryl group. "Thioalkyl" includes -SR groups wherein R is alkyl or aryl. The term "protected" as used herein, unless otherwise indicated, indicates that a group is added to an oxygen, nitrogen or phosphorus atom to prevent further reaction or for other purposes. A wide variety of oxygen and nitrogen protecting groups are known to those skilled in the art of organic synthesis. -17- 201210602 The term "nuclear" includes natural or modified nucleosides, acyclic nucleosides and C-nucleosides. The term "C-nucleoside" means a nucleoside in which a glycosyl bond is attached to a carbon on a modified nucleobase, rather than a nitrogen of a nucleoside (see the reference for c_ nucleoside reviews for details): Chemistry of Nucleosides and Nucleotides by Leroy B Townsend 1 994,Science, Chapter 5

The Chemistry of C-nucleosides, Kyoichi A Watanabe pp421)。C-核苷並不限於該評述中所舉出之化合物。 「醫藥上可接受的鹽類」包括本文所提供之化合物的 任何保留其之生物特性的鹽類,且用於醫藥用途上無毒性 或無其他不欲的性質。此鹽類可衍生自技術中已知之各種 有機及無機抗衡離子(counter-ion)。此鹽類包括:(1)與有 機或無機酸所形成之酸加成鹽,例如氫氯酸、氫溴酸、硫 酸、硝酸、磷酸、胺基磺酸、乙酸、三氟乙酸、三氯乙酸、 丙酸、己酸、環戊基丙酸、羥基乙酸、戊二酸、丙酮酸、 乳酸、丙二酸、琥珀酸、山梨酸、抗壞血酸、蘋果酸、馬 來酸、反丁烯二酸、酒石酸、檸檬酸、苯甲酸、3-(4-羥基 苯甲醯基)苯甲酸、苦味酸、肉桂酸、扁桃酸、酞酸、月桂 酸、甲磺酸、乙磺酸、1,2 -乙院-二磺酸、2-羥基乙磺酸、 苯磺酸、4 -氯苯磺酸、2 -萘磺酸、4_甲苯磺酸、樟腦酸、樟 腦磺酸、4-甲基雙環[2.2.2]-辛_2_烯_卜甲酸、葡萄庚酸、 3 -苯基丙酸、三甲基乙酸、第三丁基乙酸、月桂基硫酸、 葡萄糖酸、苯甲酸、麩胺酸、羥基萘酸、水楊酸、硬脂酸、 環己基胺基磺酸、喹啉酸、黏康酸等酸類;或(2)所形成之 鹽類其係當酸性質子存在於母化合物’不論是(a)或(b) ’(a) -18 - 201210602 被金屬離子置換,例如鹼金屬離子、鹼土金屬離子、鋁離 子’或鹼金屬或鹼土金屬氫氧化物,例如氫氧化鈉、氫氧 化鉀、氫氧化釣、氫氧化鎂、氫氧化鋁、氫氧化鋰、氫氧 化鋅及氫氧化鋇 '氨,或(b)與有機鹼配位,例如脂族、脂 環族或芳族有機胺類’例如氨'甲胺、二甲胺、二乙胺、 甲基吡啶、乙醇胺、二乙醇胺、三乙醇胺、乙二胺、離胺 酸、精胺酸、鳥胺酸、膽鹼、n,n’-二节基伸乙基-二胺、 氯普魯卡因(chloroprocaine)、二乙醇胺、普魯卡因、N_〒 基苯乙胺、N -甲基葡萄糖胺哌阱、三(羥基甲基)胺基甲烷、 氫氧化四甲基銨等。 鹽類更包括(僅爲例示)鈉鹽、鉀鹽、鈣鹽、鎂鹽、銨 鹽、四烷基銨鹽等,及當該化合物含鹼性官能性時,非毒 性有機或無機酸之鹽類,例如鹽酸鹽、氫溴酸鹽、硫酸鹽、 磷酸鹽、胺基磺酸鹽、硝酸鹽、乙酸鹽、三氟乙酸鹽、三 氯乙酸鹽、丙酸鹽、己酸鹽、環戊基丙酸鹽、羥基乙酸鹽、 戊二酸鹽、丙酮酸鹽、乳酸鹽、丙二酸鹽、硬脂酸鹽、琥 珀酸鹽、山梨酸鹽、抗壞血酸、蘋果酸鹽、馬來酸鹽、反 丁烯二酸鹽、酒石酸鹽、檸檬酸鹽、苯甲酸鹽、3-(4-羥基 苯甲醯基)苯甲酸鹽、苦味酸鹽、肉桂酸鹽、扁桃酸鹽、酞 酸鹽、月桂酸鹽、甲磺酸鹽、乙磺酸鹽、1,2-乙烷-二磺酸 鹽、2-羥基乙磺酸鹽、苯磺酸鹽' 4-氯苯磺酸鹽、2-萘磺酸 鹽、4-甲苯磺酸鹽、樟腦酸鹽、樟腦磺酸鹽、4-甲基雙環 [2.2.2]-辛-2-烯-1-甲酸鹽、葡萄庚酸鹽、3-苯基丙酸鹽、 三甲基乙酸鹽、第三丁基乙酸鹽、月桂基硫酸鹽、葡萄糖The Chemistry of C-nucleosides, Kyoichi A Watanabe pp421). The C-nucleoside is not limited to the compounds exemplified in the review. "Pharmaceutically acceptable salts" include any of the salts of the compounds provided herein that retain their biological properties and are non-toxic or otherwise undesirable for medical use. This salt can be derived from various organic and inorganic counter-ions known in the art. The salts include: (1) acid addition salts with organic or inorganic acids, such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, aminosulfonic acid, acetic acid, trifluoroacetic acid, trichloroacetic acid , propionic acid, caproic acid, cyclopentylpropionic acid, glycolic acid, glutaric acid, pyruvic acid, lactic acid, malonic acid, succinic acid, sorbic acid, ascorbic acid, malic acid, maleic acid, fumaric acid, Tartaric acid, citric acid, benzoic acid, 3-(4-hydroxybenzhydryl)benzoic acid, picric acid, cinnamic acid, mandelic acid, citric acid, lauric acid, methanesulfonic acid, ethanesulfonic acid, 1,2-B - Disulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, 4-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, 4-toluenesulfonic acid, camphoric acid, camphorsulfonic acid, 4-methylbicyclo[2.2 .2]-octyl-2-ene-p-formic acid, glucoheptanoic acid, 3-phenylpropionic acid, trimethylacetic acid, tert-butylacetic acid, lauryl sulfate, gluconic acid, benzoic acid, glutamic acid, hydroxyl An acid such as naphthoic acid, salicylic acid, stearic acid, cyclohexylaminosulfonic acid, quinolinic acid or muconic acid; or (2) a salt formed by the presence of an acidic proton in the parent compound Either (a) or (b) '(a) -18 - 201210602 is replaced by a metal ion such as an alkali metal ion, an alkaline earth metal ion, an aluminum ion or an alkali metal or alkaline earth metal hydroxide such as sodium hydroxide or hydrogen Potassium oxide, hydrogen hydroxide, magnesium hydroxide, aluminum hydroxide, lithium hydroxide, zinc hydroxide and barium hydroxide 'ammonia, or (b) coordinated with an organic base, such as an aliphatic, alicyclic or aromatic organic Amines such as ammonia 'methylamine, dimethylamine, diethylamine, picoline, ethanolamine, diethanolamine, triethanolamine, ethylenediamine, lysine, arginine, auramine, choline, n, N'-two-segment ethyl-diamine, chloroprocaine, diethanolamine, procaine, N-mercaptophenethylamine, N-methylglucosamine pipe trap, tris(hydroxymethyl) Aminomethane, tetramethylammonium hydroxide, and the like. Salts include, by way of example only, sodium, potassium, calcium, magnesium, ammonium, tetraalkylammonium salts, and the like, and salts of non-toxic organic or inorganic acids when the compound contains basic functionality. Classes such as hydrochloride, hydrobromide, sulfate, phosphate, amine sulfonate, nitrate, acetate, trifluoroacetate, trichloroacetate, propionate, hexanoate, cyclopentane Propionate, glycolate, glutarate, pyruvate, lactate, malonate, stearate, succinate, sorbate, ascorbic acid, malate, maleate, Fumarate, tartrate, citrate, benzoate, 3-(4-hydroxybenzhydryl)benzoate, picrate, cinnamate, mandelate, citrate , laurate, methanesulfonate, ethanesulfonate, 1,2-ethane-disulfonate, 2-hydroxyethanesulfonate, besylate '4-chlorobenzenesulfonate, 2- Naphthalene sulfonate, 4-toluenesulfonate, camphorate, camphorsulfonate, 4-methylbicyclo[2.2.2]-oct-2-ene-1-carboxylate, grape heptanoate, 3 -phenylpropionate, trimethyl b Salt, tert-butyl acetate, lauryl sulphate, glucose

C -19- 201210602 酸鹽、苯甲酸鹽、麩胺酸鹽、羥基萘酸鹽、水楊酸鹽、硬 脂酸鹽、環己基胺基磺酸鹽、喹啉酸鹽、黏康酸鹽等。 如本文中所使用之「前藥」一詞表示當投與至生物系 統時,由於自發化學反應、酵素催化反應及/或代謝性過程 或其之結合而可產生生物上活性的化合物的任何化合物。 標準前藥係使用連接至例如- OH、-NH2之與藥劑連接之官 能性的基團而形成,其在活體內裂解。本發明所述之前藥 爲例示,並不以此爲限,且熟悉技術者能製備其他已知之 各種前藥。 「L -核苷」一詞表示天然及經改質之β._〇_核苷類似物 之鏡像異構物。 「阿拉伯呋喃糖基(arabinofuranosyl)核苷」一詞表示 核苷類似物含有阿拉伯呋喃糖基糖(即天然(一般)核苷之味 喃核糖基(ribofuranosyl)糖之2,-羥基係位於糖環之對側 面。 「二氧環戊院(dioxolane)糖」一詞表示含有氧原子以 代替呋喃核糖基糖之3,碳的醣類。 「氟化糖」一詞表示具有1-3個氟原子連接至糖基之 碳上的醣類。 「核苷」一詞表示連接至糖基之嘌呤或嘧啶鹼基或其 之類似物’包括其之雜環基及碳環狀類似物。 π治療有效量」一詞表示在治療疾病或症狀上具有任 何有利效果之量。 「磷酸酯」一詞表示-〇-Ρ〇32-。 「胺基磷酸酯」一詞表示_Ν-Ρ〇32-。 -20- 201210602 「膦酸酯」一詞表示-CHR.p〇32-。 「母藥」一詞表示核苷及/或非環狀核韦 劑(m-o-po32·)。 「母藥」一詞亦表示含膦 [r-ch2-p(o)(oh)2]。 「生物上活性的藥劑或劑」一詞表示產 化學物質。在本發明中,生物上活性的| (M-OH)、核苷單磷酸酯(Μ·〇_ρ〇32-)、核 (Μ-0-Ρ2063 ·)、核苷三璘酸酯(μ_〇 Ρ3〇94-) [M_CH2P(〇;K〇H)2、m_ch2p〇32·]、非核苷膦 酯(M-CH2P2〇63·)或其二憐酸醋(M-CH2P3094 物。 「院方基」或「院基方基」一詞包括具 芳基。芳烷基或芳基烷基一詞包括具有芳基g 「嘌呤」或「嘧啶」鹼基一詞包括腺嘌出 胺基嘌呤、N6 -醯基-6-胺基嘌呤(其中醯基爲 芳基、院基芳基或芳基院基)、N6 -苄基- 6-® 乙烯基-6-胺基嘌呤、N6-乙炔基-6_胺基嘌呤 呤、7 -脫氮嘌呤、經改質7_脫氮嘌呤、胸腺pg N4-醯基胞嘧啶、5-氟胞嘧啶、5-甲基胞嘧啶. 尿嘧啶、5 -氟尿嘧啶、5 -烷基尿嘧啶、5 -乙 乙炔基尿嘧啶、5-羥基甲基尿嘧啶、5-醯胺届 尿嘧啶、5-碘尿嘧啶、5-Br-乙烯基尿嘧啶、 5 -氮尿嘧啶、三唑并吡啶、咪唑并吡啶、吡 吡唑并嘧啶基,但不以此爲限。嘌呤鹼基包 1=單-磷酸酯藥 酸酯藥劑 生生物效應之 笔劑表示核苷 苷二磷酸酯 、核苷膦酸酯 酸酯、單磷酸 _)、含醇化合 烷基取代基之 代基之烷基。 $、N6-烷基-6-C(0)(烷基、 g基嘌呤、N6-、6-環胺基嘌 I啶、胞嘧啶、 1 6-氮胞嘧啶、 烯基嘧啶、5 · 丨:赌D定、5 -氰基 5 -氮胞嘧啶、 咯并嘧啶基及 括鳥嘌呤、腺 -21 · 201210602 嘌呤、2·氟腺嘌呤、2 -氯腺嘌呤、次黃嘌呤、7 -脫氮鳥嘌呤、 7-脫氮腺嘌呤、2,6-二胺基嘌呤、及6_氯嘌呤、6_烷氧基嘌 呤、6-去氧鳥嘌呤、6-烷基硫嘌呤,但不以此爲限。在鹼 基上之官能性氧及氮基視需要或所欲時可經保護。適當之 保護基爲熟悉技術者所熟知,且包括三甲基矽烷基、二甲 基己基矽烷基、第三丁基二甲基矽烷基、及第三丁基二苯 基矽烷基、三苯甲基、烷基及醯基,例如乙醯基及丙醯基、 甲磺醯基及對甲苯磺醯基。 「醯基」或「0 -連接之酯」一詞包括式C(0)R’之基, 其中R’爲直鏈、分支或環狀烷基或芳基。 「胺基酸」一詞包括天然發現及合成α-、β-、γ-或δ-胺基酸,且包括蛋白質中所發現之胺基酸,即甘胺酸、丙 胺酸、纈胺酸、白胺酸、異白胺酸、甲硫胺酸、苯基丙胺 酸、色胺酸、脯胺酸、絲胺酸、蘇胺酸 '半胱胺酸、酪胺 酸、天冬胺酸、麩醯胺酸、天冬胺酸酯、麩醯胺酸酯、離 胺酸、精胺酸及組胺酸,但不以此爲限。在較佳具靖實施 例中’胺基酸爲L-組態。或者是,胺基酸可爲丙胺醯基、 纈胺酸基、白胺酸基、異白胺酸基、脯胺酸基、苯基丙胺 酸基、色胺酸基、甲硫胺酸基、甘胺酸基、絲胺酸基、蘇 胺酸基、半胱胺酸基、酪胺酸基、天冬胺酸基、麩醯胺酸 基、天冬胺醯基、麩醯胺醯基、離胺酸基、精胺酸基、組 胺酸基、β-丙胺醯基、β-纈胺酸基、β-白胺酸基、β-異白胺 酸基、β·脯胺酸基、β-苯基丙胺酸基、β-色胺酸基、β-甲硫 胺酸基、β-甘胺酸基、β-絲胺酸基、β-蘇胺酸基、β-半胱胺 酸基、Ρ-酪胺酸基、β-天冬胺酸基、β-麩醯胺酸基、β_天冬 -22- 201210602 胺醯基、β·麩醯胺醯基、P-離胺酸基、P精胺酸基或P-組胺 酸基之衍生物。 「溶劑化物」包括本文所提供之化合物或其鹽’其進 一步包括以非共價分子間作用力結合之計量或非計量數量 之溶劑。當該溶劑爲水時,溶劑化物爲水合物。 如本文中所使用之「個體」及「病患」一詞可交互使 用。「個體」一詞表示動物,例如哺乳類’包括非靈長類(例 如牛、豬、馬、貓、犬、大鼠及小鼠)及靈長類(例如猴, 例如食蟹猴(cynomolgus monkey)、黑猩猩(chimpanzee))。 在一具體實施例,該個體爲人類。 如本文中所使用之「治療劑」及「治療劑類」表示可 用於治療或預防失調或其一或多種徵狀的任何藥劑。在某 些具體實施例,「治療劑」一詞包括本文所提供之化合物。 在一具體實施例,治療劑爲一種已知有用或已被使用或近 來被使用於治療或預防失調或其一或多種徵狀的藥劑。 「治療有效量」包括當投與個,體治療疾病時化合物或 組成物之數量,足以引起治療該疾病。「治療有效量」除 化合物外可根據疾病及其嚴重性、及欲治療個體之年齡、 體重等而變化。 任何疾病或失調之「治療」在一具體實施例表示改善 存於個體之疾病或失調。在另一具體實施例,「治療」包 括改善至少一種不能被個體所識別之身體參數。在另一具 體實施例,「治療」包括調節疾病或失調,身體上(例如穩 定可察覺之徵狀)或是生理上(例如穩定身體參數)或爲二 -23- 201210602 者。在另一具體實施例,「治療」包括延遲疾病或失調之 發生。 在另一具體實施例,較佳的式!化合物爲式ηι化合物:C -19- 201210602 acid salt, benzoate, glutamate, hydroxynaphthoate, salicylate, stearate, cyclohexylamine sulfonate, quinolinate, mucate Wait. The term "prodrug" as used herein denotes any compound which, when administered to a biological system, produces a biologically active compound due to spontaneous chemical reactions, enzyme catalyzed reactions, and/or metabolic processes or combinations thereof. . The standard prodrug is formed using a functional group attached to an agent such as -OH, -NH2, which is cleaved in vivo. The prodrugs of the present invention are illustrated by way of example and are not limited thereto, and those skilled in the art can prepare other prodrugs known in the art. The term "L-nucleoside" means a mirror image isomer of a naturally occurring and modified beta._〇-nucleoside analog. The term "arabinofuranosyl nucleoside" means that the nucleoside analog contains arabinofuranosylose (i.e., the natural (general) nucleoside of the ribofuranosyl sugar 2, the hydroxy group is located in the sugar ring. The term "dioxolane sugar" means a sugar containing an oxygen atom instead of a ribofuranosyl sugar. The term "fluorinated sugar" means having 1-3 fluorine atoms. A saccharide attached to a carbon of a glycosyl group. The term "nucleoside" means a hydrazone or a pyrimidine base or an analog thereof which is attached to a glycosyl group, including a heterocyclic group and a carbon cyclic analog thereof. The term "quantity" means an amount which has any beneficial effect in treating a disease or condition. The term "phosphate" means -〇-Ρ〇32-. The term "amino phosphate" means _Ν-Ρ〇32-. -20- 201210602 The term "phosphonate" means -CHR.p〇32-. The term "parental" means nucleoside and/or acyclic nucleus (mo-po32.). The term also means phosphine-containing [r-ch2-p(o)(oh)2]. The term "biologically active agent or agent" means a chemical substance. In the present invention, biologically active | (M-OH), nucleoside monophosphate (Μ·〇_ρ〇32-), nucleus (Μ-0-Ρ2063 ·), nucleoside tricaprate (μ_ 〇Ρ3〇94-) [M_CH2P(〇; K〇H)2, m_ch2p〇32·], non-nucleoside phospholipid (M-CH2P2〇63·) or its two pity vinegar (M-CH2P3094). The term "base" or "hospital base" includes aryl. The term arylalkyl or arylalkyl includes the aryl group " 嘌呤" or "pyrimidine". The term includes adenine oxime, N6-mercapto-6-aminopurine (wherein the fluorenyl group is an aryl group, a aryl group or an aryl group), N6-benzyl-6-® vinyl-6-amino fluorene, N6-ethynyl -6_aminopurine, 7-deazapurine, modified 7-deazapurine, thymus pg N4-decyl cytosine, 5-fluorocytosine, 5-methylcytosine. Uracil, 5- Fluorouracil, 5-alkyluracil, 5-ethynyl uracil, 5-hydroxymethyluracil, 5-guanamine uracil, 5-iodouracil, 5-Br-vinyl uracil, 5- Nitrouracil, triazolopyridine, imidazopyridine, pyrpyrazolopyrimidinyl, but not limited thereto. The base 1 = mono-phosphate ester acid agent agent biological effect pen agent means nucleoside diphosphate, nucleoside phosphonate ester, monophosphate _), alkenyl group containing alcohol alkyl substituent Alkyl., N6-alkyl-6-C(0) (alkyl, g-based oxime, N6-, 6-cyclic amino oxime I pyridine, cytosine, 16-azacytosine, alkenylpyrimidine, 5 · 丨: bet D, 5-cyano 5-azacytosine, pyridopyrimidinyl and guanine, gland-21 · 201210602 嘌呤, 2 · fluoroadenine, 2-chloroadenine, hypoxanthine, 7-deazaguanine, 7-deaza adenine, 2,6-diaminoguanidine, and 6-chloropurine, 6-alkoxyanthracene, 6-deoxyguanine, 6-alkylthiopurine, But not limited to this. The functional oxygen and nitrogen groups on the base can be protected as needed or desired. Suitable protecting groups are well known to those skilled in the art and include trimethyldecyl, dimethylhexyldecyl, tert-butyldimethylalkyl, and tert-butyldiphenylalkyl, triphenyl Base, alkyl and sulfhydryl groups, such as ethenyl and propyl sulfonyl, methylsulfonyl and p-toluenesulfonyl. The term "mercapto" or "0-linked ester" includes the radical of the formula C(0)R' wherein R' is straight-chain, branched or cyclic alkyl or aryl. The term "amino acid" includes the natural discovery and synthesis of alpha-, beta-, gamma- or delta-amino acids, and includes the amino acids found in proteins, namely glycine, alanine, proline, Aleucine, isoleucine, methionine, phenylalanine, tryptophan, valine, serine, threonine, cysteine, tyrosine, aspartate, bran Promethamine, aspartate, glutamate, lysine, arginine and histidine, but not limited thereto. In a preferred embodiment, the amino acid is in the L-configuration. Alternatively, the amino acid may be an amidino group, a prolyl group, an leucine group, an isoleucine group, a valine group, a phenylalanine group, a tryptophan group, a methionine group, Glycosyl group, serine group, threonate group, cysteine group, tyrosine group, aspartic acid group, glutamic acid group, aspartame group, glutamine sulfhydryl group, Amino acid group, arginine group, histidine group, β-alaninyl group, β-proline group, β-leucine group, β-isoleucine group, β·proline group, --phenylalanine, β-tryptophan, β-methionine, β-glycine, β-serin, β-threonate, β-cystein Base, Ρ-tyrosine group, β-aspartate, β-glutamic acid group, β_asparagus-22- 201210602 Amine thiol, β·glutaramine sulfhydryl, P-lysine a derivative of a P, arginine or P-histidine group. "Solvate" includes a compound provided herein or a salt thereof' which further includes a metered or non-metered amount of solvent combined by non-covalent intermolecular forces. When the solvent is water, the solvate is a hydrate. The terms "individual" and "patient" as used herein are used interchangeably. The term "individual" means that an animal, such as a mammal, includes non-primates (such as cattle, pigs, horses, cats, dogs, rats, and mice) and primates (such as monkeys, such as cynomolgus monkeys). , chimpanzee). In a specific embodiment, the individual is a human. As used herein, "therapeutic agent" and "therapeutic agent" mean any agent that can be used to treat or prevent a disorder or one or more of its symptoms. In certain embodiments, the term "therapeutic agent" includes the compounds provided herein. In a specific embodiment, the therapeutic agent is an agent that is known to be useful or has been used or has recently been used to treat or prevent a disorder or one or more of its symptoms. "Therapeutically effective amount" includes the amount of a compound or composition that is sufficient to cause treatment of the disease when administered to a body. The "therapeutically effective amount" may vary depending on the disease and its severity, and the age, weight, etc. of the individual to be treated, in addition to the compound. The "treatment" of any disease or disorder in a particular embodiment indicates an improvement in the disease or disorder that is present in the individual. In another embodiment, "treatment" includes improving at least one physical parameter that is not recognized by the individual. In another embodiment, "treatment" includes modulating a disease or disorder, either physically (e.g., stabilizing a detectable condition) or physiologically (e.g., stabilizing a body parameter) or being 2-23-201210602. In another embodiment, "treatment" includes delaying the onset of a disease or disorder. In another embodiment, the preferred formula! The compound is a compound of the formula ηι:

或其醫藥可接受前藥、鹽類、溶劑化物、立體異構、互變 異構或多形性形式, 其中: R及汉7各獨立選自Η、烷基、芳基、炔基、烯基、環 院基、具有1至3個雜原子(例如〇、s、Ν)之雜環基 或雜方基’但不以此爲限,其全部可選擇經取代; R及R7可形成4至7員環,例如吡咯啶,其全部可選 擇經取代; R2、R3 、X、Y及JV1-XH如上述定義。 土另 具體實施例’較佳之式III化合物爲式IV化合Or a pharmaceutically acceptable prodrug, salt, solvate, stereoisomeric, tautomeric or polymorphic form thereof, wherein: R and Han 7 are each independently selected from the group consisting of hydrazine, alkyl, aryl, alkynyl, alkenyl , a ring-based group, a heterocyclic group or a heterocyclic group having 1 to 3 hetero atoms (for example, hydrazine, s, hydrazine) 'but not limited thereto, all of which may be optionally substituted; R and R 7 may form 4 to A 7-membered ring, such as pyrrolidine, all of which may be optionally substituted; R2, R3, X, Y, and JV1-XH are as defined above. Soil Specific Embodiments The preferred compound of formula III is a compound of formula IV.

Μ、X、γ 兑 β R如上述定義。 適於本發明能Μ, a即藥衍生作用之母藥Μ, X, γ = β R are as defined above. Suitable for the present invention, a drug that is a drug-derived drug

C -24- 201210602 不同種類之母藥可從本發明前藥方法中獲得。較佳爲 前藥保護基連接至母藥上之羥基。在許多情況,該母藥將 具有許多此類之官能基。被選擇用於連接至前藥的較佳基 團爲對於生物活性最重要的基團,且爲化學上適於連接至 母藥。因此,含磷前藥基團將可避免前藥具有生物活性。 非活性前藥應降低全身性副作用,因爲相對於非肝臟組 織,較高的藥劑濃度會位於目標器官(肝臟)。 有許多種等級之含羥基官能基的治療上有用之藥劑 (包括核苷或非核苷),其可用於衍生成本發明之胺基磷酸 酯或胺基膦酸酯前藥。此等化合物包括核苷、C -核苷、核 苷酸、膦酸酯及其他含醇化合物。衍生自此等化合物的本 文所揭示之前藥被視爲在本發明之範圍內。 下列化合物爲此類化合物之實例。 治療上有用且可衍生成本發明前藥之一類例示性核苷 藥劑(D-及L-)爲式V化合物:C -24- 201210602 Different kinds of parent drugs can be obtained from the prodrug method of the present invention. Preferably, the prodrug protecting group is attached to the hydroxyl group on the parent drug. In many cases, the parent drug will have many such functional groups. The preferred group selected for attachment to the prodrug is the group most important for biological activity and is chemically suitable for attachment to the parent drug. Therefore, the phosphorus-containing prodrug group will prevent the prodrug from being biologically active. Inactive prodrugs should reduce systemic side effects because higher drug concentrations are located in the target organ (liver) relative to non-liver tissues. There are many classes of therapeutically useful agents (including nucleosides or non-nucleosides) containing hydroxyl functional groups which can be used to derivatize the amine phosphate or alanine phosphonate prodrugs of the invention. Such compounds include nucleosides, C-nucleosides, nucleotides, phosphonates, and other alcohol containing compounds. The prodrugs disclosed herein are derived from such compounds and are considered to be within the scope of the invention. The following compounds are examples of such compounds. A therapeutically useful and derivable derivative of one of the class of prodrugs of the invention is an exemplary nucleoside agent (D- and L-) which is a compound of formula V:

或其醫藥可接受前藥, 其中: (A)選自 〇、S、CH2、CHF、C = CH2、C = CHF、CF2 ; r8 及 R9 獨立選自 H、OH、CH30、F、CM、Br、I、CN、 N3、甲基、乙基、乙烯基、乙炔基、氯乙烯基、氟甲 •25- 201210602 基、二氟甲基、三氟甲基,但不以此爲限,其全部可 選擇經取代; R8及R9可形成可選擇經F、二氟、CM、Br、I、CN、 N 3取代之乙烯基; R1G選自Η、甲基、乙基、乙烯基、乙炔基、氯乙烯基、 氟甲基、二氟甲基、三氟甲基,但不以此爲限; R11選自H、CN、Ν3、甲基、乙基、乙烯基、乙炔基、 氯乙烯基、氟甲基、二氟甲基、三氟甲基,但不以此 爲限: R12選自Η、OH、F、氰基及疊氮基,但不以此爲限’ Β選自式B-l、Β-2或Β-3之嘧啶及嘌呤,但不以此胃 限:Or a pharmaceutically acceptable prodrug thereof, wherein: (A) is selected from the group consisting of hydrazine, S, CH2, CHF, C = CH2, C=CHF, CF2; r8 and R9 are independently selected from the group consisting of H, OH, CH30, F, CM, Br , I, CN, N3, methyl, ethyl, vinyl, ethynyl, chlorovinyl, fluoromethyl, 25-201210602, difluoromethyl, trifluoromethyl, but not limited thereto, all Optionally substituted; R8 and R9 form a vinyl group optionally substituted by F, difluoro, CM, Br, I, CN, N 3 ; R1G is selected from the group consisting of hydrazine, methyl, ethyl, vinyl, ethynyl, Chlorovinyl, fluoromethyl, difluoromethyl, trifluoromethyl, but not limited thereto; R11 is selected from the group consisting of H, CN, Ν3, methyl, ethyl, vinyl, ethynyl, chlorovinyl, Fluoromethyl, difluoromethyl, trifluoromethyl, but not limited to this: R12 is selected from the group consisting of hydrazine, OH, F, cyano and azide, but is not limited thereto. Β-2 or Β-3 pyrimidine and guanidine, but not the stomach limit:

其中 X2 爲 H、NH2、NHMe、NMe2 或鹵素(I、ΒΓ、 F),但不以此爲限,其全部可選擇經取代; X4 爲 NH2 或 OH ; X5爲鹵素(I、Br、Cl、F)、OH、NH2、甲基、乙惊基 2-溴乙烯基、乙炔基; 食:、環 X6爲H、OH、OMe、OEt、SMe、烷氧基、芳氣暴 烷氧基、烷基硫基、芳基硫基、環烷基硫基、 呋喃基、烷基胺基 '二烷基胺基、芳基胺基、 基胺基、環烷基胺基、環丙基胺基, -26- 201210602 X7爲Η、乙烯基、乙炔基、鹵素(1、Br、cl' F), 所有皆可選擇經取代; 胺基及羥基可選擇性經保護。 其他經改質嘧啶或嘌呤,例如5 -氮嘧啶、6 -氮嘧啶、 3 -脫氮吡啶、3 -氟-3 -脫氮嘧啶、8 -吖-7 .脫氮嘌呤,但 不以此爲限,其亦被視爲在本發明之範圍內。 式V核苷之第一羥基被衍生成本發明之胺基磷酸酯前 藥。舉例而言,氟尿苷(Floxuridine)可如下述衍生成其之 胺基磷酸酯: 〇Wherein X2 is H, NH2, NHMe, NMe2 or halogen (I, ΒΓ, F), but not limited thereto, all of which may be optionally substituted; X4 is NH2 or OH; X5 is halogen (I, Br, Cl, F), OH, NH2, methyl, ethyl bromo 2-bromovinyl, ethynyl; food: ring X6 is H, OH, OMe, OEt, SMe, alkoxy, aromatoxy, alkane Thiothio, arylthio, cycloalkylthio, furyl, alkylamino 'dialkylamino, arylamino, arylamino, cycloalkylamino, cyclopropylamino, -26- 201210602 X7 is an anthracene, a vinyl group, an ethynyl group, a halogen (1, Br, cl' F), all of which may be optionally substituted; the amine group and the hydroxyl group may be selectively protected. Other modified pyrimidines or purines, such as 5-aziridine, 6-aziridine, 3-deazapyridine, 3-fluoro-3-deazapyrimidine, 8-indol-7, deazapurine, but not It is also considered to be within the scope of the invention. The first hydroxyl group of the nucleoside of the formula V is derived from the aminophosphate prodrug of the invention. For example, Fluxuridine can be derivatized as an amino phosphate as follows:

00

〇 FVV \ y^O-P-O—| dΗ。 氟尿苷 胺基磷酸酯前藥 類例示性 治療上有用且適於衍生成本發明前藥之另一 核苷藥劑(D-及L-)爲式VI化合物:〇 FVV \ y^O-P-O-| dΗ. Fluorouridine Aminophosphate prodrugs An exemplary therapeutically useful and suitable derivative of the prodrug of the invention. Another nucleoside agent (D- and L-) is a compound of formula VI:

VI 或其醫藥可接受前藥, 其中: Β、Α及R11如上述定義; R13及R14獨立選自H、N3、F、CN、CH3、烷基或乙 烯基’但不以此爲限,其全部可選擇經取代。 -27- 201210602 適於本發明之前藥衍生的其他種類之母藥爲式VII及 VIII化合物(D-及L-異構物):VI or a pharmaceutically acceptable prodrug thereof, wherein: ruthenium, osmium and R11 are as defined above; R13 and R14 are independently selected from H, N3, F, CN, CH3, alkyl or vinyl 'but not limited thereto, All can be replaced. -27- 201210602 Other types of parent drugs which are suitable for the prodrugs of the present invention are the compounds of the formulae VII and VIII (D- and L-isomers):

HOHO

HOHO

vm 或其醫藥可接受前藥, 其中: B如上述定義。 適於本發明前藥的其他種類之母藥選自爲核苷膦酸酯 (表 1,Biochem Pharmacol. 2007, 73, 911),但不以此爲限: . 表1非環狀核苷膦酸酯之實例Vm or a pharmaceutically acceptable prodrug thereof, wherein: B is as defined above. Other types of parent drugs suitable for the prodrugs of the invention are selected from the group consisting of nucleoside phosphonates (Table 1, Biochem Pharmacol. 2007, 73, 911), but are not limited thereto: Table 1 Acyclic nucleoside phosphines Acid ester example

-28- 201210602 其他通式m-x_p(o)(oh)2之生物上活性的膦酸酯(其 中 M-X 可爲環系統)包括下式例示性化合物(WO 2008/082602):-28-201210602 Other biologically active phosphonates of the formula m-x_p(o)(oh)2 wherein M-X can be a ring system include the following exemplary compounds (WO 2008/082602):

適於本發明之前藥衍生的其他種類之母藥爲一種非環 狀核苷,包括阿昔洛韋(acyclovir)、更昔洛韋(ganciclovir) 及噴昔洛韋(pencyclovir),但不以此爲限。 適於本發明之前藥衍生的其他種類之母藥爲C-核苷, 一種特殊種類的核苷(詳見對於C-核苷評述之參考資料: Chemistry of Nucleosides and Nucleotides by Leroy B Townsend 1 9 9 4, Science, Chapter 5 The Chemistry of C-nucleosides, Kyoichi A Watanabe pp421),C-核苷並不限 於該評述中所舉出之化合物。 當核苷可被磷酸化成活性三磷酸酯形式時,該核苷被 稱爲母藥。舉例而言,阿昔洛韋(ACV)可被認爲是一種以 M-OH (式I中X = 0)形式之母藥。ACV首先被磷酸化成 ACV-P032_,然後活體中進一步磷酸化而形成ACV-三磷酸 酯,其爲生物上活性的形式。 當一些核苷並不是激酶優異的受質且顯示非生物活 性,而其核苷酸或核苷單磷酸酯爲生物活性時,母藥被稱 爲對應之核苷單磷酸酯。 當含醇化合物被衍生成本發明前藥時,母藥被稱爲對 應之含醇藥劑。 -29- 201210602 下列熟知之藥劑(但不以此爲限)於說明書及申請專利 範圍中提及(亦提供縮寫及普通名稱): araA ; 9-P-D-阿拉伯呋喃糖基腺嘌呤(阿糖腺苷; Vidarabine) araC ; β-D-阿拉伯呋喃糖基胞嘧啶 araU ; 9-β-ϋ-阿拉伯呋喃糖基尿嘧啶 d4T ; 2’,3’-二去氫-3’-去氧胸腺嘧啶(司他夫定; Stavudine) ddl; 2’,3’-二去氧肌苷(去經肌苷;Didanosine) ddA ; 2’,3’-二去氧腺苷 ddC; 2’,3’-二去氧胞苷(札西他賓;Zalcitabine) L-ddC ; L-2’,3,-二去氧胞苷 L-FddC ; L-2’,3’-二去氧-5-氟胞苷 L-d4C ; L-3’-去氧-2’,3’-二去氫胞苷 L-Fd4C; L-3’-去氧-2’,3’-二去氫-5-氟胞苷 L-FMAU;L-p-l-(2_氟_2-去氧-阿拉伯味喃糖基)胸腺嘧 啶(克拉夫定;Clevudine) 3TC ; (-)_2’,3’_二去氧-3’-硫胞苷 2,R,5’S(-)-l-[2-(羥基甲基)氧硫雜環戊烷-5-基]胞嘧 啶(拉米夫定;Lamivudine) Ι-β-D-呋喃核糖基-1,2,4-三唑-3-甲醯胺(利巴韋林;Other types of parent drugs that are suitable for the prodrugs of the present invention are an acyclic nucleoside, including acyclovir, ganciclovir, and pencyclovir, but not Limited. Other classes of parent drugs that are suitable for the prodrugs of the present invention are C-nucleosides, a particular class of nucleosides (for details see C-nucleoside reviews: Chemistry of Nucleosides and Nucleotides by Leroy B Townsend 1 9 9 4, Science, Chapter 5 The Chemistry of C-nucleosides, Kyoichi A Watanabe pp421), C-nucleosides are not limited to the compounds cited in the review. When a nucleoside can be phosphorylated to the active triphosphate form, the nucleoside is referred to as a parent drug. For example, acyclovir (ACV) can be considered a parent drug in the form of M-OH (X = 0 in Formula I). ACV is first phosphorylated to ACV-P032_ and then further phosphorylated in vivo to form ACV-triphosphate, which is in a biologically active form. When some nucleosides are not excellent acceptors of kinases and exhibit abiotic activity, and their nucleotides or nucleoside monophosphates are biologically active, the parent drug is referred to as the corresponding nucleoside monophosphate. When an alcohol-containing compound is derived from a prodrug of the invention, the parent drug is referred to as a corresponding alcohol-containing agent. -29- 201210602 The following well-known pharmaceutical agents (but not limited to them) are mentioned in the specification and patent application (also abbreviated and common names): araA; 9-PD-arabinofuranosyl adenine (arabinary gland) Viranabine) araC; β-D-arabinofuranosylcytosine araU; 9-β-ϋ-arabinofuranosyl uracil d4T; 2',3'-dihydro-3'-deoxythymidine ( Stavudine) ddl; 2',3'-dideoxyinosine (Dardanosine) ddA; 2',3'-dideoxyadenosine ddC; 2',3'- Deoxycytidine (zaxitabine; Zalcitabine) L-ddC; L-2',3,-dideoxycytidine L-FddC; L-2',3'-dideoxy-5-fluorocytidine L-d4C; L-3'-deoxy-2',3'-dihydrocytidine L-Fd4C; L-3'-deoxy-2',3'-dihydro-5-fluorocytidine L-FMAU; Lpl-(2-fluoro-2-deoxy-arabinofuranosyl) thymine (Clavudine; Clevudine) 3TC; (-)_2',3'-dideoxy-3'-sulfur Cytidine 2,R,5'S(-)-l-[2-(hydroxymethyl)oxathiolan-5-yl]cytosine (lamivudine; lamivudine) Ι-β-D-furan Sugar-1,2,4-triazole-3-Amides (ribavirin;

Ribavirin) FIAU -去氧_2_氣- β- D-阿拉伯咲喃糖基)-5 -蛾尿音 FIAC;卜(2_去氧-2-氟- β-D-阿拉伯呋喃糖基)-5-碘胞嘧 啶 -30- 201210602 8狀〇;( + )- (1(1,20,3(1)-9-[2,3-雙(經基甲基)環丁基]鳥 嘌呤 FMAU ; 2’-氟_5-甲基-β-L-阿拉伯-呋喃糖基尿嘧啶 BvaraU ; Ι-β-D-阿拉伯呋喃糖基-£_5_(2_溴乙烯基)尿 喷陡(索立夫定:Sorivudine) E-5-(2-溴乙烯基)-2’-去氧尿苷 TFT;三氟胸腺嘧啶(三氟胸苷;Triflu〇r〇thyrnidine) 5-丙炔-1-阿拉伯糖尿嘧啶 CDG ;碳環2’-去氧鳥苷 DAPD ; (-)-p-D-2,6-二胺基嘌呤二氧環戊烷 FDOC;(-)-p-D-5-氟-卜[2-(羥基甲基二氧環戊烷] 胞嘧啶 d4C ; 3’-去氧-2’,3’·二去氫胞苷 DXG ;二氧環戊烷鳥苷 FEAU ; 2’-去氧- 2’-氟-1-0_〇_阿拉伯呋喃糖基-5·乙基 尿嘧啶 .. FLG; 2’,3’-二去氧- 3’-氟鳥音 FTC; (-)-順-5-氟-1-[2-(羥基甲基)-i,3-氧硫雜戊環- 5-基]胞喷陡5 -基-碳環2’-去氧鳥苷(BmS200,475) [1-(4經基-1,2 -丁 一 燃基)胞喷陡](Cytallene) 奧塔諾新 A(Oxetanocin A); 9-(2_ 去氧·2-羥甲基- β-D-赤型-氧雜環丁糖基(oxetanosyl))腺嘌呤 奧塔諾新G; 9-(2-去氧-2-羥甲基- β- D-赤型-氧雜環丁 糖基)鳥嘌呤 ddAPR : 2,6-二胺基嘌呤-2,,3,_二去氧核苷 -31- 201210602 3TC; (-)-2’,3’_二去氧-3’硫胞苷; (2R,5S)l-[2-(羥甲基)-13-氧硫雜戊環-5-基]胞嘧啶 (拉脈優淀;Lamivudine) 環巴 A(Cyclobut A); ( + )-9-[(1贷,2〇1,30)-2,3-雙(羥甲 基)-1-環丁基]腺嘌呤 環巴 G; ( + )-9-[(4,2(1,30)-2,3-雙(羥甲基)-1-環丁基] 鳥嘌呤 5-氟-2’-去氧尿苷(氟尿苷) dFdC ; 2’,2’-二氟去氧胞苷(吉西他濱;Gemcitabine) araC;阿拉伯糖胞嘧啶(阿糖胞苷;Cytarabine) 溴去氧尿苷 IDU; 5-換-2’-去氧尿苷(姚苷;idoxuridine)Ribavirin) FIAU-deoxy-2_gas-β-D-arabinofuranosyl)-5-moth urinary FIAC; Bu (2_deoxy-2-fluoro-β-D-arabinofuranosyl)- 5-iodocytosine-30- 201210602 8-form 〇; ( + )- (1(1,20,3(1)-9-[2,3-bis(transmethyl)cyclobutyl]guanine FMAU 2'-Fluoro-5-methyl-β-L-arabino-furanosyl uracil BvaraU; Ι-β-D-arabinofuranosyl-£_5_(2_bromovinyl) urinary spray steep (Solif Ding: Sorivudine) E-5-(2-bromovinyl)-2'-deoxyuridine TFT; trifluorothymidine (Triflu〇r〇thyrnidine) 5-propyne-1-arabinose Pyrimidine CDG; carbocyclic 2'-deoxyguanosine DAPD; (-)-pD-2,6-diaminophosphoniumdioxolane FDOC; (-)-pD-5-fluoro-b [2-( Hydroxymethyldioxocyclopentane] cytosine d4C; 3'-deoxy-2',3'.dihydrocytidine DXG; dioxolane guanosine FEAU; 2'-deoxy-2'- Fluor-1-0_〇_arabinofuranosyl-5·ethyluracil: FLG; 2',3'-dideoxy-3'-fluorine bird FTC; (-)-cis-5-fluoro 1-[2-(hydroxymethyl)-i,3-oxothiapentane-5-yl]-cello-deep 5-amino-carbocyclic 2'-deoxyguanosine BmS200, 475) [1-(4-based-1,2-butane-based) cell spray steep] (Cytallene) Otto new A (Oxetanocin A); 9-(2_ deoxy-2-hydroxymethyl - β-D-erythro-oxetanosyl) adenine Otano new G; 9-(2-deoxy-2-hydroxymethyl-β-D-erythro-oxocycle Butyryl) guanine ddAPR: 2,6-diaminopurine-2,,3,-dideoxynucleoside-31- 201210602 3TC; (-)-2',3'-dideoxy-3' Thiocytidine; (2R, 5S) 1-[2-(hydroxymethyl)-13-oxathiapentan-5-yl]cytosine (Lamivudine) Cyclobut A; ( + )-9-[(1 loan, 2〇1,30)-2,3-bis(hydroxymethyl)-1-cyclobutyl]adenine ring G; ( + )-9-[(4 , 2(1,30)-2,3-bis(hydroxymethyl)-1-cyclobutyl]guanine 5-fluoro-2'-deoxyuridine (fluorouridine) dFdC; 2',2' -difluorodeoxycytidine (Gemcitabine) araC; arabinose cytosine (cytarabine) bromodeoxyuridine IDU; 5-exchange-2'-deoxyuridine (yasidine; idoxuridine)

CdA; 2 -氯去氧腺苷(克拉屈濱;Cladribine) 2 -氯-2’-氟-araA (克羅拉濱;Clofarabine) F-ara-A;氟阿拉伯糖腺苷(氟達拉濱;Fludarabine) ACV ; 9-(2_羥碁乙氧基甲基)鳥嘌呤(阿昔洛韋) GCV; 9-(1,3-二羥基-2-丙氧基甲基)鳥嘌呤(更昔洛韋) 9-(4-羥基-3-羥基甲基丁 -1-基)鳥嘌呤(噴昔洛韋; Penciclovir)CdA; 2-chlorodeoxyadenosine (Cladribine; Cladribine) 2-chloro-2'-fluoro-araA (Crofarabine) F-ara-A; fluoroarabinoadenosine (fludarabine; Fludarabine) ACV; 9-(2-hydroxyindoleoxymethyl)guanine (acyclovir) GCV; 9-(1,3-dihydroxy-2-propoxymethyl)guanine (more Lovir) 9-(4-hydroxy-3-hydroxymethylbutan-1-yl)guanine (penciclovir; Penciclovir)

(R)-9-(3,4-二羥基丁基)鳥嘌呤(布昔洛韋;Buciclovir) araT ; 9-P-D-阿拉伯呋喃糖基胸腺嘧啶 FMdC ; (E)-2’_去氧-2’-(氟亞甲基)胞苷 AICAR; 5-胺基咪唑-4-甲醯胺-1-呋喃核糖基 CNDAC ; 2’-氰基-2’-去氧胞苷 棕櫚醯基CNDAC -32- 201210602 2’-氟- 2’-去氧- 4’-疊氮基- β- D-阿拉伯呋喃糖基胞嘧啶 (RO-0622) 2’-氟-2’-去氧-4’-疊氮基-9-卩-0-阿拉伯呋喃糖基尿嘧 啶 2’-氟-2’-去氧-4’-疊氮基-9-卩-〇-阿拉伯呋喃糖基腺嘌 哈 2’-氟-2’-去氧-4’-疊氮基-9-卩-0-阿拉伯呋喃糖基鳥嘌 呤 4疊氮基- β- D-阿拉伯呋喃糖基胞嘧啶 4’-疊氮基- 9-P-D-阿拉伯呋喃糖基尿嘧啶 4’ ·疊氮基- 9- P- D-阿拉伯呋喃糖基腺嘌呤 4 ’ -疊氮基-9 - β - D -阿拉伯呋喃糖基鳥嘌呤 4’-疊氮基尿苷 4’-疊氮基胞苷 4’-疊氮基腺苷 4’.;疊氮基肌苷 4’-疊氮基鳥苷 2’-C-甲基尿苷 2’-C-甲基胞苷 2’-C-甲基腺苷 2’-C-甲基肌苷 2’-C-甲基鳥苷 2’-C-(氟甲基)尿苷 2’-C-(氟甲基)胞苷 2’-C-(氟甲基)腺苷 -33- 201210602 2,-C-(氟甲基)肌苷 2’-C-(氟甲基)鳥苷 4,-疊氣基-2’- C-甲基尿苷 4,-疊氮基_2’-C-甲基胞苷 4’-叠氣基- 2’-C-甲基腺甘 4’-疊氮!基- 甲基肌甘 4,-疊氮基-2’-C-甲基鳥苷 2,-氟-2’-去氧_2’-C-甲基尿苷 2,-氟-2’-去氧_2’-C-甲基胞苷 2,-氟-2’-去氧_2’-C-甲-基腺苷 2,-氟-2’-去氧_2’-C-甲基肌苷 2,-氟-2’-去氧_2’-C-甲基鳥苷 2,-氟-2’-去氧-2’-<:-(氟甲基)尿苷 2,-氟-2’-去氧-2’-C-(氟甲基)胞苷 2,_氟-2,-去氧-2’-€-(氟甲基)腺苷 2,-氟-2’-去氧_2’-(3-(氟甲基)肌苷 2,-氟-2,-去氧_2’-(:-(氟甲基)鳥苷 所有上述鳥苷類似物之所有6_烷氧基及6_去氧類似物 上述核苷之所有單磷酸酯。 所有核苷之胺基及/或羥基可選擇經保護。 本發明之前藥技術可使用於抗癌領域。ara-C (阿糖胞 苷)之數項特徵,例如代謝性去胺基作用、對於dCK之低親 和性及ara-CTP之快速清除,限制了其之細胞毒性的活性 (Lancet Oncol. 2002,3,415)。爲了克服此等問題,數種 ara-C之化學修飾已被合成。在鹼基或在糖基團具有親脂性 -34- 201210602 側鏈之ara-C前藥已被設計來針對增加紐 遲其之去胺基作用及清除(Cancer Res. Nucleosides & Nucleotides 1 999, 18 P h arm ac ο 1. 2 0 0 4 , 6 7,5 0 3 )。此種分子可 性機制進入細胞,並可能增加細胞內 (Biochem. Pharmacol. 2004, 67, 503; Br. 95 7) 〇 與ara-C本身相反,此等前藥具有 之活性,此可能是此等藥劑較佳之滲I 性。此等前藥之一·者CP-4055,正進行 其中6-17位具有固體腫瘤之預治療之病 (J. Clin. Oncol. 2004,22, 2049) ° 前核 ig 類似物類似物UA9 11爲一 5’-ara-CMP 服在人類濾泡性淋巴瘤RL-G細胞之經 抗性(Int. J. Cancer 2003, 1 07, 1 49) ° 應用至核苷藥劑的本發明前藥技術 與此等藥劑;數種此類化合物之性質將 較長之滯留。此等特徵可使此類類似物 療區域。 較佳地,適於本文所揭示前藥衍生 之核苷(包括其之前藥)(表2),但不以此 表2 .母核苷之實例 丨胞攝取ara-C及延 1999, 59, 2944 ; ,877; Biochem. 藉由輸送器非依賴 ara-C之半生期 J. Cancer 1995, 71, 對抗固體腫瘤模式 I性及隨後之滯留 第一期臨床評估, 患顯不穩定的疾病 Γ M (pronucleotide) 之前驅物,其能克 dCK調控之ara-C 能以口服調配劑投 產生母藥於體液中 適於治療較廣的治 之化合物包括下式 爲限: -35- 201210602(R)-9-(3,4-dihydroxybutyl)guanine (Buciclovir) araT; 9-PD-arabinofuranosyl thymine FMdC; (E)-2'-deoxy- 2'-(fluoromethylene) cytidine AICAR; 5-aminoimidazole-4-carboxamide-1-furan ribosyl CNDAC; 2'-cyano-2'-deoxycytidine palmitoyl CNDAC - 32- 201210602 2'-Fluoro-2'-deoxy- 4'-azido-β-D-arabinofuranosylcytosine (RO-0622) 2'-fluoro-2'-deoxy-4'- Azido-9-卩-0-arabinofuranosyluracil 2'-fluoro-2'-deoxy-4'-azido-9-卩-〇-arabinofuranosyl adenine 2'- Fluorin-2'-deoxy-4'-azido-9-oxime-0-arabinofuranosylguanine 4 azide-β-D-arabinofuranosylcytosine 4'-azido- 9 -PD-arabinofuranosyluracil 4'-azido- 9-P-D-arabinofuranosyl adenine 4 '-azido-9 - β-D-arabinofuranosylguanine 4'- Azidouridine 4'-azidocytidine 4'-azidoadenosine 4'.; azidoinosine 4'-azidoguanosine 2'-C-methyluridine 2'- C-methylcytidine 2'-C-methyladenosine 2'-C- Inosine 2'-C-methylguanosine 2'-C-(fluoromethyl)uridine 2'-C-(fluoromethyl)cytidine 2'-C-(fluoromethyl)adenosine-33 - 201210602 2,-C-(fluoromethyl)inosine 2'-C-(fluoromethyl)guanosine 4,-azido-2'-C-methyluridine 4,-azido-2 '-C-Methylcytidine 4'-Laminated 2'-C-methylglycine 4'-azido!-Methylglycine 4,-azido-2'-C-methyl Guanosine 2,-fluoro-2'-deoxy-2'-C-methyluridine 2,-fluoro-2'-deoxy-2'-C-methylcytidine 2,-fluoro-2'- Deoxy-2'-C-methyl-based adenosine 2,-fluoro-2'-deoxy-2'-C-methylinosine 2,-fluoro-2'-deoxy-2'-C- Guanosine 2,-fluoro-2'-deoxy-2'-<:-(fluoromethyl)uridine 2,-fluoro-2'-deoxy-2'-C-(fluoromethyl) Glycoside 2, _fluoro-2,-deoxy-2'-€-(fluoromethyl)adenosine 2,-fluoro-2'-deoxy-2'-(3-(fluoromethyl)inosine 2, -Fluoro-2,-deoxy-2'-(:-(fluoromethyl)guanosine All 6-alkoxy groups of all the above guanosine analogs and 6-deoxy analogs All monophosphates of the above nucleosides The amine groups and/or hydroxyl groups of all nucleosides may be optionally protected. Used in the field of anti-cancer. Several characteristics of ara-C (cytarabine), such as metabolic deamination, low affinity for dCK and rapid clearance of ara-CTP, limit its cytotoxic activity. (Lancet Oncol. 2002, 3, 415). In order to overcome these problems, several chemical modifications of ara-C have been synthesized. The ara-C prodrugs that have a lipophilicity at the base or in the sugar group -34-201210602 side chain have been designed to increase the deaminating action and clearance of neostigler (Cancer Res. Nucleosides & Nucleotides 1 999, 18 P h arm ac ο 1. 2 0 0 4 , 6 7,5 0 3 ). This molecularly motivating mechanism enters the cell and may increase intracellular (Biochem. Pharmacol. 2004, 67, 503; Br. 95 7). In contrast to ara-C itself, these prodrugs have activity, which may be The drug is preferably infiltrated. One of these prodrugs, CP-4055, is undergoing pre-treatment of 6-17 patients with solid tumors (J. Clin. Oncol. 2004, 22, 2049) ° Pronuclear ig analog analogue UA9 11 Resistance to human follicular lymphoma RL-G cells for a 5'-ara-CMP (Int. J. Cancer 2003, 1 07, 1 49) ° Prodrug technology of the invention applied to nucleoside agents With these agents; the properties of several such compounds will be retained for a longer period of time. These features allow for the treatment of such analog areas. Preferably, it is suitable for prodrug-derived nucleosides (including prodrugs thereof) disclosed herein (Table 2), but not as shown in Table 2. Examples of parent nucleosides ingestion of ara-C and extensions 1999, 59, 2944 ; , 877; Biochem. By the non-dependent ara-C half-life J. Cancer 1995, 71, against the solid tumor pattern I and subsequent retention of the first phase of clinical evaluation, suffering from unstable disease Γ M (pronucleotide) Pre-drug, its ara-C can regulate the AK-C can be used as an oral formulation to produce a parent drug in body fluids suitable for the treatment of a wide range of compounds including the following formula: -35- 201210602

-36- 201210602-36- 201210602

201210602201210602

經本文所揭示之技術,衍生自含醇藥劑之前藥亦被認 爲在本發明之範圍內。 治療用途 -38- 201210602 本發明亦提供核苷、核苷酸、C -核苷、C核苷酸、核 苷單磷酸酯、膦酸酯、非核苷膦酸酯及含醇化合物的含磷 前藥。本文前藥之治療用途亦供治療病毒感染、癌症及其 他肝臟失調。此等前藥可用於增進母藥之生物可利用性及/ 或藥物動力學。本文所揭示之此等前藥及組成物可單獨投 與或合倂其他治療上有效之藥劑來投與。 本文所揭示之各種治療藥劑的胺基磷酸酯及胺基膦酸 酯化合物可用於對應母藥所用於治療的疾病。 在一些具體實施例,本文所揭示之前藥亦可用於治療 對母藥有抗性之病毒。 此類胺基磷酸酯及胺基膦酸酯化合物可有利地增強輸 送藥劑至肝臟。在一些具體實施例,化合物允許核苷之活 性5’-單磷酸酯輸送至肝臟,其可增強活性三磷酸化化合物 之形成。 在一具體實施例,本文所提供治療肝臟失調之方法’ 其包括投與有效量之本文所提之化合物,或其醫藥上可接 受的鹽類。在某些具體實施例,該方法包含之步驟爲投與 有效於治療肝臟失調之數量的化合物,合倂有效於治療疾 病之第二藥劑至所需個體。該化合物可爲本文所述之任何 化合物,且該第二藥劑可爲技術中或本文所述之任何第二 藥劑。 本發明之前藥技術可應用於轉換大量非活性核苷成爲 治療上有用之核苷酸的前藥。 因此,對於所有含醇藥劑之本文所揭示之胺基隣酸醋 前藥被認爲在本發明之範圍內。 -39- 201210602 本文之前藥的治療用途亦供作治療肝 HBV及HCV)及肝臟失調,包括肝癌及代謝 糖尿病、高血脂症、動脈粥狀硬化及肥胖。 用於方法中之第二或更多種藥劑 在某些具體實施例,本文所提供之化合 益於治療肝臟失調之方法,其包含另投與所 治療失調(例如肝癌)之第二藥劑。該第二藥 悉技術者已知之藥劑,其有效於治療失調,包 核准者。 在某些具體實施例,本文所提供之化合 劑而投與。在另一具體實施例,第二藥劑合 投與。在另一具體實施例,第二藥劑合倂二 劑而投與。 化合物之製備 本文所提供之化合物可以熟悉技術者顯 製備、分離或獲得。例示之製備方法詳述於 節。 核苷5-氟- 2’-去氧尿苷(氟尿苷)之胺基 製法說明於流程圖1。將含氧氯化磷(1 eq., 以含三乙胺(leq.)及3-氟-4-甲氧基苄基醇(1 溶液於-78 °C處理,獲得單酯2,其再以含苄 三乙胺(1 eq.)之THF處理,獲得氯胺基磷酸 基咪唑(NMI)存在下將3與氟尿苷反應,產生 磷酸酯4。 炎感染(包括 性疾病,例如 物及組成物有 需個體有效於 劑可爲任何熟 括目前被FDA 物合併第二藥 倂另一藥劑而 或多種第二藥 見之任何方法 以下實施例章 磷酸酯的例示 P〇Cl3)之 THF ,1 eq.)之 THF 基胺(1 e q .)及 酯3 。在N-甲 標的核苷胺基 -40- 201210602Prodrugs derived from alcohol-containing agents are also considered to be within the scope of the invention by the techniques disclosed herein. Therapeutic use -38- 201210602 The present invention also provides nucleosides, nucleotides, C-nucleosides, C nucleotides, nucleoside monophosphates, phosphonates, non-nucleoside phosphonates and phosphorus-containing compounds before phosphorus-containing compounds medicine. The therapeutic use of the prodrugs herein is also used to treat viral infections, cancer and other liver disorders. Such prodrugs can be used to enhance the bioavailability and/or pharmacokinetics of the parent drug. The prodrugs and compositions disclosed herein can be administered alone or in combination with other therapeutically effective agents. The aminophosphate and aminophosphonate compounds of the various therapeutic agents disclosed herein can be used in response to diseases for which the parent drug is used for treatment. In some embodiments, the prodrugs disclosed herein can also be used to treat a virus that is resistant to the parent drug. Such amino phosphate and amino phosphonate compounds advantageously enhance delivery of the agent to the liver. In some embodiments, the compound allows for the transport of the active 5'-monophosphate of the nucleoside to the liver, which enhances the formation of the active triphosphorylated compound. In a specific embodiment, provided herein is a method of treating a liver disorder comprising administering an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof. In certain embodiments, the method comprises the steps of administering a compound effective to treat the liver disorder, and combining the second agent effective to treat the disease to the desired individual. The compound can be any of the compounds described herein, and the second agent can be any second agent in the art or described herein. The prodrug technique of the present invention can be applied to prodrugs that convert large amounts of inactive nucleosides into therapeutically useful nucleotides. Thus, the amine-based acid vinegar prodrugs disclosed herein for all alcohol-containing agents are considered to be within the scope of the invention. -39- 201210602 The therapeutic use of the previous drugs is also used to treat liver HBV and HCV) and liver disorders, including liver cancer and metabolic diabetes, hyperlipidemia, atherosclerosis and obesity. Second or More Agents for Use in Methods In certain embodiments, the compositions provided herein are directed to a method of treating liver disorders comprising a second agent that is otherwise administered a disorder of treatment (e.g., liver cancer). The second agent is a pharmaceutical agent known to the physician, which is effective for treating disorders, and is approved by the approver. In certain embodiments, the compounds provided herein are administered. In another embodiment, the second agent is administered in combination. In another embodiment, the second agent is administered in combination with two agents. Preparation of Compounds The compounds provided herein can be prepared, isolated or obtained by those skilled in the art. The exemplified preparation methods are detailed in Sections. The amine-based method for the nucleoside 5-fluoro-2'-deoxyuridine (fluorouridine) is illustrated in Scheme 1. Phosphorus oxychloride (1 eq., containing triethylamine (leq.) and 3-fluoro-4-methoxybenzyl alcohol (1 solution at -78 °C to obtain monoester 2, which Treatment with benzyl triethylamine (1 eq.) in THF to obtain 3 in the presence of chloramine phosphatidyl imidazole (NMI) to react with fluorouridine to produce phosphate 4. Inflammation (including sexual diseases, such as The composition may be an individual effective agent, and may be any of the exemplified P(Cl3) THF of any of the following examples of the first embodiment of the FDA, the second compound, or the second drug. 1 eq.) of THF amine (1 eq.) and ester 3. N-alpha nucleoside amine-40- 201210602

NMI ΟNMI Ο

流程圖1胺基磷酸酯例示性製法 流程圖1可應用於其他含醇化合物之前藥的一般製 法,如流程圖2中所示。Flowchart 1 Exemplary Process for Amino Phosphate Flowchart 1 can be applied to the general process of other alcohol-containing compounds, as shown in Scheme 2.

〇 CI-P-CI R-0H --— Et3N 5〇 CI-P-CI R-0H --- Et3N 5

0 R-0-P-CI I Cl E Η Ν r6'r70 R-0-P-CI I Cl E Η Ν r6'r7

EtsNEtsN

0 R—0 -p—Cl I R-OH: R-〇H爲含醇藥劑 及R7如上述定義0 R—0 —p—Cl I R-OH: R—〇H is an alcohol-containing agent and R7 is as defined above

NMI 〇 R-〇-P-〇-Drug R6 R7 8 流程圖2胺基磷酸酯之一般製法 關於胺基磷酸酯之一般製法(流程圖2),將p〇C13以 對應之醇溶液(5,1 eq)及三乙胺(1 eq)處理,獲得二氯磷酸 化物6。在三乙胺(1 eq)存在下,所產生之6進一步以對應 -41- 201210602 之胺溶液(leq)處理,提供氯胺基磷酸酯 7。在N -甲基咪 唑存在下,將7以包括核苷之含醇藥劑處理,產生胺基磷 酸酯前藥8。 根據專利方法(US 2005/0124583),核苷膦酸酯之胺基 膦酸酯的例示性製法可說明於流程圖3。在DCC及三乙胺 存在下,將磷酸酯9以對應醇處理,獲得化合物1〇。以S0C12 氯化10’之後將所產生之氯化物11與對應之胺反應,獲NMI 〇R-〇-P-〇-Drug R6 R7 8 Flowchart 2 General method for the preparation of aminophosphates For the general preparation of aminophosphates (Scheme 2), p〇C13 is the corresponding alcohol solution (5, Treatment with 1 eq) and triethylamine (1 eq) gave the dichlorophosphate 6. The resulting 6 is further treated with an amine solution (leq) corresponding to -41 to 201210602 in the presence of triethylamine (1 eq) to provide chloroamino phosphate 7. In the presence of N-methylimidazole, 7 is treated with an alcohol-containing agent comprising a nucleoside to produce an aminophosphate prodrug 8. An exemplary method for the preparation of an aminophosphonate of a nucleoside phosphonate according to the patented method (US 2005/0124583) is illustrated in Scheme 3. Phosphate 9 is treated with the corresponding alcohol in the presence of DCC and triethylamine to give the compound 1 oxime. After the chlorination of 10° by S0C12, the produced chloride 11 is reacted with the corresponding amine to obtain

流程圖3膦醯胺之製備 生物學評估 1.抗癌活性分析 將合成作爲抗癌藥劑之化合物各於白血病細胞株中測 試以評估其之抗癌效力。使用獲自Promega (CellTiter96 Aqueous One溶液增殖分析)之MTS分析試劑測試化合物, 於5μΜ濃度測試化合物(WO 2006/100439)。 2 .抗C型肝炎活性 -42- 201210602 本文所揭示之抗HCV活性及化合物之細胞毒性可依γ 列專利方法分析(wo 2007/027248)。 3 .抗Η B V分析 本發明化合物可根據技術中已知之任何分析方法分析 抗HBV活性。 4 ·此外,可根據技術中已知之任何分析方法分析化合 物於個體肝臟細胞中之累積。在某些具體實施例,個體肝 臟細胞可用於分析化合物或其衍生物(例如其之核苷、核苷 磷酸酯或核苷三磷酸酯衍生物)之肝臟累積。 實施例 下列實施例說明本文所提供之代表性化合物之合成, 此等實施例並非意圖或被理解爲限制所請標的之範圍。顯 而易見的是,除非本文有特別指出外,所請標的之範圍可 被實施。鑒於本文之教示,標的可有許多改質及變化,因 此皆爲所請標的之範圍內。 由於新形成之磷對掌性中心,根據本文所製備之胺基 磷酸酯之產物爲非鏡像異構物之混合物,並以混合物於生 物學分析中測試。 本發明前藥之非鏡像異構物可經由層析法單離成單一 異構物。 實施例1氟尿苷胺基磷酸酯之製備(DGX-101) -43- 201210602Flowchart 3 Preparation of phosphoniumamine Biological evaluation 1. Anticancer activity assay Compounds synthesized as anticancer agents were tested in leukemia cell lines to evaluate their anticancer efficacy. Compounds were tested using MTS assay reagents obtained from Promega (CellTiter 96 Aqueous One solution proliferation assay) and tested at 5 μΜ concentration (WO 2006/100439). 2. Anti-Hepatitis C Activity -42- 201210602 The anti-HCV activity and cytotoxicity of the compounds disclosed herein can be analyzed according to the gamma patent method (wo 2007/027248). 3. Anti-HBV Analysis The compounds of the invention can be assayed for anti-HBV activity according to any analytical method known in the art. 4 In addition, the accumulation of the compound in individual liver cells can be analyzed according to any analytical method known in the art. In certain embodiments, individual liver cells can be used to analyze liver accumulation of a compound or a derivative thereof (e.g., a nucleoside, nucleoside phosphate, or nucleoside triphosphate derivative thereof). EXAMPLES The following examples illustrate the synthesis of representative compounds provided herein, and such examples are not intended to be construed as limiting the scope of the invention. It is obvious that the scope of the claimed subject matter can be implemented unless otherwise stated herein. In view of the teachings in this article, there are many modifications and variations to the subject matter, and therefore are within the scope of the subject matter. Due to the newly formed phosphorus-to-palm center, the product of the amino phosphate prepared according to the present invention is a mixture of non-image isomers and tested as a mixture in a biochemical analysis. The non-image isomers of the prodrugs of the invention can be isolated into a single isomer by chromatography. Example 1 Preparation of Fluorouridine Amino Phosphate (DGX-101) -43- 201210602

4 (DGX-101)4 (DGX-101)

在含氧氯化磷(3.07 g,20 mmol)之THF(40 mL)溶液 中,於-78 °C添加含醇1(3.12 g’ 20 mmol)及三乙胺(2.02 g, 20 mmol)之T H F (1 0 m L)溶液,並於-7 8 °C攪拌此混合物3 小時。在所產生之混合物中,於-78 °C添加含苄胺(2.14 g, 20 mmol)及三乙胺(2.02 g,20 mmol)之 THF(10 mL)溶液, 並於-7 8 °C攪拌此混合物1小時,然後於室溫攪拌隔夜。於 真空中移除THF,並將殘餘物過濾並以乙醚(50 mL)洗滌, 將濾液及洗液蒸發,獲得粗產物3,將其溶於CH2C12(10 mL),不再進一步純化而用於下一反應。在含核苷(2.42 g, 10 mml)之 CH2C12(40 mL)懸浮液中,添加N-甲基咪唑(5 mL),並將溶液於冰浴中冷卻。在溶液中添加3之溶液,並 將所產生之溶液於冰浴中攪拌3小時。添加水(5 mL),並 以EtOAc(2 X 200 mL)萃取混合物,有機溶液以0.5N HC1 溶液、aqNaHC03、鹽水洗滌,並在Na2S04上乾燥。於真 空中濃縮溶劑,殘餘物經矽凝膠層析純化(含0-8% Me OH 之 CH2C12),獲得化合物 4(DGX_101,3 5 0 mg,64%)°5HNMR -44 - 201210602In a solution of phosphorus oxychloride (3.07 g, 20 mmol) in THF (40 mL), EtOAc (3········· A solution of THF (10 mL) was added and the mixture was stirred at -7 8 ° C for 3 hours. A solution of benzylamine (2.14 g, 20 mmol) and triethylamine (2.02 g, 20 mmol) in THF (10 mL) was added at -78 ° C and stirred at -7 8 ° C. This mixture was allowed to stand for 1 hour and then stirred at room temperature overnight. The THF was removed in vacuo and EtOAc EtOAc (EtOAc m. Next reaction. In a suspension of CH2C12 (40 mL) containing nucleoside (2.42 g, 10 mm), N-methylimidazole (5 mL) was added and the solution was cooled in an ice bath. A solution of 3 was added to the solution, and the resulting solution was stirred in an ice bath for 3 hours. Water (5 mL) was added and the mixture was crystallised eluted with EtOAc EtOAc EtOAc The solvent was concentrated in vacuo, and the residue was purified by EtOAc EtOAc EtOAc EtOAc (EtOAc (EtOAc)

δΗ yMR (CDCL3): 7.70, 7.6 7 (dd, J = 6.4Hz, 1H),6.90 -7.42 (m, 8H), 6.15 (m, 1H), 4.90 - 5.11 (m, 2H), 3.B8 (s, 3H), 3.71 - 4.46 (m, 6H), 1.96 - 2.41 (m, 2H). LC-MS (ESI) : 5 54 [M+l] + . 依據相同程序,但使用對應之核苷或試劑,製備下列 化合物: 實施例2 DGX-102: δΗ NMR (CDCL3): 7.70, 7.67 (dd, J = 6.4Hz, 1H), 7.13 - 7.31 (m, 9H), 6.15 (m, 1H), 4.90 - 5.11 (m, 2H), 3.71 -4.46 (m, 6H), 2.26, 2.2 5 (ss, 3H), 1.96 - 2.41 (m, 2H). LC-MS (ESI) : 5 2 0 [M+ 1 ] + .δΗ yMR (CDCL3): 7.70, 7.6 7 (dd, J = 6.4Hz, 1H), 6.90 -7.42 (m, 8H), 6.15 (m, 1H), 4.90 - 5.11 (m, 2H), 3.B8 ( s, 3H), 3.71 - 4.46 (m, 6H), 1.96 - 2.41 (m, 2H). LC-MS (ESI): 5 54 [M+l] + . According to the same procedure, but using the corresponding nucleoside or Reagents, the following compounds were prepared: Example 2 DGX-102: δ NMR (CDCL3): 7.70, 7.67 (dd, J = 6.4 Hz, 1H), 7.13 - 7.31 (m, 9H), 6.15 (m, 1H), 4.90 - 5.11 (m, 2H), 3.71 -4.46 (m, 6H), 2.26, 2.2 5 (ss, 3H), 1.96 - 2.41 (m, 2H). LC-MS (ESI) : 5 2 0 [M+ 1 ] + .

OO

實施例3 DGX-104: δΗ NMR (CDC13): 8.93 (ss, 1H), 6.9 5 - 7.40 (m, 9H), 6.11 (d, 1H), 5.44 (dd, 1H), 5.00, 4.85 (mm, 2H), 3.48-4.40 (m, 11H), 1.36 (dd, 3H). LC-MS (ESI) 568 [M + H] + . -45- 201210602Example 3 DGX-104: δΗ NMR (CDC13): 8.93 (ss, 1H), 6.9 5 - 7.40 (m, 9H), 6.11 (d, 1H), 5.44 (dd, 1H), 5.00, 4.85 (mm, 2H), 3.48-4.40 (m, 11H), 1.36 (dd, 3H). LC-MS (ESI) 568 [M + H] + . -45- 201210602

DGX-103 DGX-104 實施例4 DGX-105: NMR (CDC13): 8.90 (br. S, 1H), 7.18-7.38 (m, 10H), 6.12 (d, 1H), 5.42 (dd, 1H), 5.05 (mm, 2H), 3.40-4.40 (m, 7H), 2.55 (s, 3H), 1.36 (dd, 3H). LC-MS (ESI) 534 [M + ] + .DGX-103 DGX-104 Example 4 DGX-105: NMR (CDC13): 8.90 (br. S, 1H), 7.18-7.38 (m, 10H), 6.12 (d, 1H), 5.42 (dd, 1H), 5.05 (mm, 2H), 3.40-4.40 (m, 7H), 2.55 (s, 3H), 1.36 (dd, 3H). LC-MS (ESI) 534 [M + ] + .

DGX-105 實施例5 DGX-106: NMR, (CDC13): 8.91 (br. S, 1H), 7.15-7.36 (m, 11H), 6.11 (d, 1H), 5.43 (dd, 1H), 5.08 (mm, 2H), 3.40-4.40 (m, 7H), 1.36 (dd, 3H). LC-MS (ESI) 5 20 [M + ] + . 實施例6DGX-105 Example 5 DGX-106: NMR, (CDC13): 8.91 (br. S, 1H), 7.15-7.36 (m, 11H), 6.11 (d, 1H), 5.43 (dd, 1H), 5.08 ( Mm, 2H), 3.40-4.40 (m, 7H), 1.36 (dd, 3H). LC-MS (ESI) 5 20 [M + ] + . Example 6

DGX-106 -46 - 201210602 DGX-108: δΗ NMR, (CD3〇D): 7.57 (d, J = 8.0Hz, 1H), T-24 (m,9H), 6.10 (m,1H),5.73 (m,1H),5.00 (m,2 H),4.2 6 O’ 2H), 4.01 (m, 4H), 2.32 (s, 3H). LC-MS (ESI) 5 5 9 [M + ] + ·DGX-106 -46 - 201210602 DGX-108: δΗ NMR, (CD3〇D): 7.57 (d, J = 8.0Hz, 1H), T-24 (m,9H), 6.10 (m,1H), 5.73 ( m,1H), 5.00 (m,2 H),4.2 6 O' 2H), 4.01 (m, 4H), 2.32 (s, 3H). LC-MS (ESI) 5 5 9 [M + ] + ·

OO

DGX-107DGX-107

實施例7化合物DGX-110之製備: 在含 2,-C -甲基-2’,3’,5’-0-三苯甲醯基-6-氯鳥苷 (DGX-109,3.13 g,5 mmol)之 MeOH (30 mL)混合物中’ 添力口NaOMe(4.8M於MeOH中,5mL,24mmol)溶液’並 將所產生之溶液於室溫攪拌1 6小時,蒸發溶劑並將殘餘物 經矽凝膠管柱層析純化(含0-15% MeOH之CHzCh)’獲得 呈白色固體之 DGX-110 (1.45 g,93.5%)之。NMR, (DMSO-d6): 8.19 (s,1H), 6.47 (s,2H), 5.80 (s,1H),5.21 (d, J = 6.8Hz, 1H), 5.14 (t,J = 5.2Hz,1H), 5.08 (s,1H), 3.94 (s, 3H), 3.98 (m, 1H), 3.80 (m, 2H), 3.65 (m, 1H), 〇-77 (s, 3H). LC-MS (ESI) 3 12 [M + ] + .Preparation of the compound of Example 7 DGX-110: containing 2,-C-methyl-2',3',5'-0-tritylmercapto-6-chloroguanosine (DGX-109, 3.13 g, 5 mmol) of MeOH (30 mL) EtOAc (EtOAc EtOAc (EtOAc) Purification by gel column chromatography (CHzCh containing 0-15% MeOH) afforded DGX-110 (1.45 g, 93.5%) as a white solid. NMR, (DMSO-d6): 8.19 (s, 1H), 6.47 (s, 2H), 5.80 (s, 1H), 5.21. (d, J = 6.8 Hz, 1H), 5.14 (t, J = 5.2 Hz, 1H), 5.08 (s,1H), 3.94 (s, 3H), 3.98 (m, 1H), 3.80 (m, 2H), 3.65 (m, 1H), 〇-77 (s, 3H). LC-MS (ESI) 3 12 [M + ] + .

實施例8 DGX-111 : 47- 201210602 根據上述用於胺基磷酸酯之程序,由DGX-110製備呈 白色泡沫之 DGX-111°(5HNMR,(CDC13): 7.80, 7.75 (ss, 1H, H-8), 7.20 (m,10H,NH2,Ph-H), 5.98 (ss,1H,H-l’),5.40 (ss, 2H, PhCH2), 5.00 (m, 2H, PhCH2N), 4.80-4.00 ( 1 0H, OCH3, 20H, 3’-,4,-,5’-H), 2.25 (ss,3H, Me), 0.95 (s, 3H, Me). LC-MS (ESI) 5 8 5 [M + ] + .Example 8 DGX-111: 47-201210602 DGX-111° (5H NMR, (CDC13): 7.80, 7.75 (ss, 1H, H) was prepared as a white foam from DGX-110 according to the procedure described above for the amino phosphate. -8), 7.20 (m, 10H, NH2, Ph-H), 5.98 (ss, 1H, H-l'), 5.40 (ss, 2H, PhCH2), 5.00 (m, 2H, PhCH2N), 4.80-4.00 ( 1 0H, OCH3, 20H, 3'-, 4,-, 5'-H), 2.25 (ss, 3H, Me), 0.95 (s, 3H, Me). LC-MS (ESI) 5 8 5 [ M + ] + .

實施例9 根據上述程序,可製備表2中化合物對應之前藥。 實施例10 抗癌分析(WO 2006/ 1 0043 9) 將合成作爲抗癌藥劑之化合物各於白血病細胞株中測 試以評估其之抗癌效力。使用獲自Promega (CellTiter96 Aqueous Ope溶液增殖分析)之MTS分析試劑測試化合物, 於5μΜ濃度測試化合物(WO 2006/100439)。標記(+ )表示該 化合物抑制細胞生長大於50%。 實施例11 HCV複製子(replicon)分析 例示化合物之抗-HCV活性及毒性可於細胞系HCV複 製子分析及細胞毒性分析(WO 2007/027248)二種生物分析 中測試。 I.抗HCV分析 使用含複製 HCV 次基因組複製子(subgenomic replicon)與螢光素酶(luciferase)報導基因(luc-ubi-neo)之 -48- 201210602 人類肝癌細胞株(Huh-7)評估化合物之抗HCV活性。在此 分析中,螢光素酶信號的程度與病毒RNA直接複製相關。 將HCV複製子-報導細胞株(NK/luc-ubi-neo)培養於補充 10%胎牛血清及 0.5 mg/ml建那黴素(Geneticin)(G418)的 DMEM培養液。將細胞維持於次匯合狀態(subCOnfluent state),以確保高程度之HCV複製子RNA合成。 爲了評估化合物之抗病毒活性,以濃度範圍〇 . 1 4至 300 μΜ製備系列稀釋液。將稀釋之化合物轉移至96孔盤, 之後添加複製子細胞(每孔6000個細胞)。在量測化合物之 螢光素酶活性後,將其與化合物培養4 8小時。螢光素酶信 號之減少反映出在被處理之細胞中H C V複製子RN Α之減 少,並用於測定EC5〇値(產生降低50%螢光素酶活性之濃 度)。 11.細胞毒性分析 將穩定地倂入染色體中之攜帶螢光素酶報導基因(經 HIV LTR啓動子驅動)的Huh-7細胞株用於分析經選擇化合 物之細胞毒性效果。此細胞株(LTR-luc)於具有10% FBS之 DMEM培養基中量測。細胞毒性分析之設計相似於HCV複 製子分析。在被處理之細胞中,螢光素酶活性之減少與測 式化合物之細胞毒性效果相關,且被用於計算CC5Q値(抑 制5 0%細胞生長之濃度)。 使用次基因組基因型la複製子之生物活性及經選擇之 化合物的細胞毒性彙整於表3。 -49- 201210602 表3複製子分析中例示化合物之活性 化合物 複製子(μΜ) CC5〇(pM) DGX-103 > 100 > 100 DGX-104 4.1 > 100 DGX-105 1.9 >100 DGX-106 >100 > 100 DGX-110 0.9 > 100 DGX-111 0.3 69 由彙整於表3中經選擇核苷及其前藥之抗HCV活性之 結果指出,本發明之前藥技術可直接輸送核苷單磷酸酯進 入生物系統,例如細胞及/或組織。本發明之前藥技術可轉 換生物上非活性之核苷(例如DGX-103)成爲具有生物效力 之核苷前藥,如DGX-104及DGX-105。本發明之前藥技術 亦可轉換生物上活性的核苷(如DGX-110)成爲更具有生物 效力之核苷前藥,例如DGX-111。 已發現在苄基酯之苯環上的取代作用戲劇性地改變本 發明胺基磷酸酯之生物特性。舉例而言,苯環上各具有4-甲氧基及2 -甲基取代之化合物DGX-104及DGX-105經證 實具有有效之抗HCV活性’而無取代之化合物DGX-106 則未顯示任何顯著之抗HCV活性。 本發明之前藥技術可應用於大多數之核苷藥劑,以增 進其之治療潛能,或改善其之生物特性。 實施例12抗HBV分析 -50- 201210602 可根據熟悉技術者已知之任何分析分析本發明化合物 之抗HBV活性。 實施例13 可根據熟悉技術者已知之任何分析分析化合物於個體 肝臟細胞中之累積。在某些具體實施例,個體之肝臟細胞 可用於分析化合物或其衍生物(例如核苷、核苷磷酸酯或其 核苷三磷酸酯衍生物)於肝臟之累積。 【圖式簡單說明】 無。 【主要元件符號說明】 無。 -51-Example 9 According to the above procedure, the compound corresponding to the compound in Table 2 can be prepared. Example 10 Anticancer assay (WO 2006/1 0043 9) Compounds synthesized as anticancer agents were each tested in leukemia cell lines to evaluate their anticancer efficacy. Compounds were tested using MTS assay reagents obtained from Promega (CellTiter 96 Aqueous Ope Proliferation Assay) and tested at 5 μΜ concentration (WO 2006/100439). The label (+) indicates that the compound inhibits cell growth by more than 50%. Example 11 HCV Replicon Analysis The anti-HCV activity and toxicity of the exemplified compounds can be tested in two biological assays, cell line HCV replication assay and cytotoxicity assay (WO 2007/027248). I. Anti-HCV analysis Evaluation of compounds using a hepatocellular carcinoma cell line (Huh-7) containing a replication HCV subgenomic replicon and a luciferase reporter gene (luc-ubi-neo) It is resistant to HCV activity. In this assay, the extent of luciferase signaling is associated with direct replication of viral RNA. The HCV replicon-reporter cell line (NK/luc-ubi-neo) was cultured in DMEM medium supplemented with 10% fetal calf serum and 0.5 mg/ml Geneticin (G418). The cells are maintained in a subCOnfluent state to ensure a high degree of HCV replicon RNA synthesis. To assess the antiviral activity of the compounds, serial dilutions were prepared at a concentration range of 11.4 to 300 μΜ. The diluted compound was transferred to a 96-well plate, followed by the addition of replicon cells (6000 cells per well). After measuring the luciferase activity of the compound, it was incubated with the compound for 48 hours. The decrease in the luciferase signal reflects a decrease in the H C V replicon RN in the treated cells and is used to determine EC5 〇値 (which produces a concentration that reduces 50% luciferase activity). 11. Cytotoxicity assay A Huh-7 cell line carrying a luciferase reporter gene (driven by the HIV LTR promoter) stably inserted into the chromosome was used to analyze the cytotoxic effect of the selected compound. This cell strain (LTR-luc) was measured in DMEM medium with 10% FBS. The design of the cytotoxicity assay is similar to the HCV replication subanalysis. In the treated cells, the decrease in luciferase activity was correlated with the cytotoxic effect of the test compound and was used to calculate CC5Q(R) (concentration of inhibition of 50% cell growth). The biological activity of the subgenomic genotype la replicon and the cytotoxicity of the selected compounds were summarized in Table 3. -49- 201210602 Table 3 Reactive compound replicon (μΜ) CC5〇(pM) DGX-103 > 100 > 100 DGX-104 4.1 > 100 DGX-105 1.9 >100 DGX- 106 >100 > 100 DGX-110 0.9 > 100 DGX-111 0.3 69 The result of the anti-HCV activity of the selected nucleoside and its prodrugs summarized in Table 3 indicates that the prodrug technology of the present invention can directly transport the nucleus Monophosphate monophosphates enter biological systems, such as cells and/or tissues. The prodrug technique of the present invention converts biologically inactive nucleosides (e.g., DGX-103) into biologically active nucleoside prodrugs, such as DGX-104 and DGX-105. The prodrug technique of the present invention also converts biologically active nucleosides (e.g., DGX-110) into more biologically active nucleoside prodrugs, such as DGX-111. It has been found that the substitution on the phenyl ring of the benzyl ester dramatically alters the biological properties of the amino phosphate of the present invention. For example, compounds DGX-104 and DGX-105 each having a 4-methoxy and 2-methyl substitution on the phenyl ring have been shown to have potent anti-HCV activity' and the unsubstituted compound DGX-106 does not show any Significant anti-HCV activity. The prodrug technology of the present invention can be applied to most nucleoside agents to increase their therapeutic potential or to improve their biological properties. Example 12 Anti-HBV Assay -50-201210602 The anti-HBV activity of the compounds of the invention can be assayed according to any assay known to those skilled in the art. Example 13 The accumulation of a compound in an individual liver cell can be analyzed according to any assay known to those skilled in the art. In certain embodiments, an individual's liver cells can be used to analyze the accumulation of a compound or a derivative thereof (e.g., a nucleoside, a nucleoside phosphate, or a nucleoside triphosphate derivative thereof) in the liver. [Simple description of the diagram] None. [Main component symbol description] None. -51-

Claims (1)

201210602 七、申請專利範圍: 1 . 一種式I化合物,201210602 VII. Patent application scope: 1. A compound of formula I, 或其醫藥可接受前藥、鹽類、溶劑化物、立· 互變異構物或多形性形式, " 其中: X選自氧(0)、CH2,但不以此爲限, 倘若X爲氧(〇),M_XH爲M_oh,代袠核毛 核苷、C -核苷或含醇化合物; Μ·0·Ρ(0)(〇Η)2代表核苷或非環狀核苷或 生物學上活性之單磷酸酯; Μ-Χ可爲環狀或非環狀系統; Μ·Χ_Ρ(0)(〇η)2爲一生物學上活性的膦酸酉 M_CH2-P(〇)(OH)2之核苷膦酸酯,倘若X C-P鍵連接: R1選自羥基、苄氧基、烷氧基 '芳氧基、5 烷基芳氧基、胺基酸殘基'苄基胺基、烷3 基胺基、烷基芳基胺基、芳基烷基胺基、二; 二芳基胺基,但不以此爲限;二烷基胺基之 开< 成環’較佳爲4至7員環,例如啦咯陡, 選擇經取代; 異構物、 、非環狀 C-核苷之 _,包括式 及p經由 r垸氧基、 &胺基、芳 完基胺基、 二烷基可 其全部可 -52- 201210602 R2選自H、甲基、烷基、芳基、炔基、烯基、環烷 基、或具有一至三個雜原子(例如〇、S、Ν)之雜環基 或雜芳基,但不以此爲限,其全部可選擇經取代; R3爲一取代基,至少一個此類之取代基在各種位置 連接至苯環,較佳位於2及/或4-位置,其選自R2C 00 (醯氧基)、醯基-NH、甲基、烷基、烷氧基、烷基胺 基、環烷基、環烷氧基、環烷基胺基、芳基、芳氧基、 芳基胺基、芳基烷基,但不以此爲限,其全部可選擇 經取代: Y代表苯環各種位置上之Η或一至三個取代基,包 括鹵素(I、Cl、Br、F)、CN、疊氮基、醯氧基、醯基 胺基、烷氧基、烷基胺基、烯基、烯基胺基、烯氧基、 炔基、炔氧基、炔基胺基、芳基烷氧基、芳基烷基胺 基、烷氧基羰基,但不以此爲限,其全部可選擇經取 代; R1及[M-X]可形成環磷酸酯,例如核苷3’,5’-環磷酸 式I化合物之所有胺基及/或羥基可被選擇經地保護; 式I化合物可爲一種由對掌性磷中心所造成之非鏡 像異構物或該非鏡像異構物之混合物,例如I a及I b :Or a pharmaceutically acceptable prodrug, a salt, a solvate, a tautomer or a polymorphic form, " wherein: X is selected from the group consisting of oxygen (0), CH2, but not limited thereto, provided that X is Oxygen (〇), M_XH is M_oh, nucleus nucleus, C-nucleoside or alcohol-containing compound; Μ·0·Ρ(0)(〇Η)2 stands for nucleoside or acyclic nucleoside or biology The active monophosphate; Μ-Χ can be a cyclic or acyclic system; Μ·Χ_Ρ(0)(〇η)2 is a biologically active phosphonium phosphonate M_CH2-P(〇)(OH) a nucleoside phosphonate of 2, if the X CP bond is attached: R1 is selected from the group consisting of a hydroxyl group, a benzyloxy group, an alkoxy 'aryloxy group, a 5 alkylaryloxy group, an amino acid residue 'benzylamine group, an alkane 3 arylamino, alkylarylamino, arylalkylamino, bis; diarylamine, but not limited thereto; dialkylamine group opening < ring formation '4 is preferably 4 Up to 7-membered ring, for example, abruptly selected, substituted; isomer, acyclic C-nucleoside _, including formula and p via r-oxy group, & amine group, aryl-based amine group, The dialkyl group may be all -52 - 201210602 R2 is selected from the group consisting of H, methyl, alkyl, a base, an alkynyl group, an alkenyl group, a cycloalkyl group, or a heterocyclic group or a heteroaryl group having one to three hetero atoms (for example, anthracene, S, anthracene), but not limited thereto, all of which may be optionally substituted; R3 is a substituent, at least one such substituent is attached to the phenyl ring at various positions, preferably at the 2 and/or 4-position, selected from the group consisting of R2C 00 (decyloxy), fluorenyl-NH, methyl , alkyl, alkoxy, alkylamino, cycloalkyl, cycloalkoxy, cycloalkylamino, aryl, aryloxy, arylamino, arylalkyl, but not Limits, all of which may be substituted: Y represents oxime or one to three substituents at various positions of the benzene ring, including halogen (I, Cl, Br, F), CN, azide, decyloxy, decylamine Alkyl, alkoxy, alkylamino, alkenyl, alkenylamino, alkenyloxy, alkynyl, alkynyloxy, alkynylamino, arylalkoxy, arylalkylamino, alkoxy a carbonyl group, but not limited thereto, all of which may be optionally substituted; R1 and [MX] may form a cyclic phosphate such as a nucleoside 3', a 5'-cyclic phosphoric acid of all compounds of the formula I and/or a hydroxyl group Can be selected Protected by the earth; the compound of formula I can be a non-mirror isomer or a mixture of such non-image isomers caused by the palmitic phosphorus center, such as I a and I b : 53- 201210602 基,選自 2 .—種式I化合物之較佳例示性經取代_ $ Ila-d,但不以此爲限:53- 201210602 base, selected from the group 2 - a preferred exemplary substitution of the compound of formula I _ $ Ila-d, but not limited to: 其中: Z爲於苯環各種位置上之一取代基,其胃自 Cl、Br、I、疊氮基、CN、CONH2、COOR2、 SR2、COSR2、0-C0R2、NH2、Nh,基 nr2 但不以此爲限,其全部可選擇經取代: R4選自R2COO (醯氧基)、醯基_Nfl、甲基、 烷氧基、烷基胺基、環烷基、環烷氧基、環院_ 芳基' 芳氧基、芳基胺基、芳基烷基,但不以此 其全部可選擇經取代: R2如上述定義; R5選自R2CO (醯基)、ch3、烷基、環烷基、 芳基烷基、烷基芳基,但不以此爲限,其全部 經取代。 3 ·如申請專利範圍第1項之化合物,較佳爲式11〗{七 Η、F、 OR2、 _醯基、 烷基、 i胺基、 ϋ爲限’ 芳基、 可選擇 合物: o R2Wherein: Z is a substituent at various positions of the benzene ring, and the stomach is derived from Cl, Br, I, azide, CN, CONH2, COOR2, SR2, COSR2, 0-C0R2, NH2, Nh, and the group nr2 but not To this end, all of them may be optionally substituted: R4 is selected from the group consisting of R2COO (decyloxy), fluorenyl-Nfl, methyl, alkoxy, alkylamino, cycloalkyl, cycloalkoxy, ring _ aryl 'aryloxy, arylamino, arylalkyl, but not all of them may be optionally substituted: R2 is as defined above; R5 is selected from R2CO (fluorenyl), ch3, alkyl, naphthenic The group, the arylalkyl group, the alkylaryl group, but not limited thereto, are all substituted. 3. A compound of the first aspect of the patent application, preferably Formula 11 {7, F, OR2, _ fluorenyl, alkyl, i-amino, hydrazine, aryl, optional compound: o R2 -54- 201210602 或其醫藥可接受前藥、鹽類、溶劑化物、立體異構物、 互變異構物或多形性形式, 其中: R6及R7各獨立選自Η、甲基、烷基 '.芳基、炔基、 烯基、環烷基、具有1至3個雜原子(例如〇、S、N) 之雜環基或雜芳基,但不以此爲限,其全部可選擇經 取代; R6及R7可形成4至7員環,例如吡略啶,其全部可 選擇經取代: R3、X、Y及M-X如上述定義。 4.一種式III化合物,較佳爲式IV化合物:-54- 201210602 or a pharmaceutically acceptable prodrug, salt, solvate, stereoisomer, tautomer or polymorphic form thereof, wherein: R6 and R7 are each independently selected from the group consisting of hydrazine, methyl, alkyl. An aryl group, an alkynyl group, an alkenyl group, a cycloalkyl group, a heterocyclic group having 1 to 3 hetero atoms (for example, anthracene, S, N) or a heteroaryl group, but not limited thereto, all of which may be selected Substituted; R6 and R7 may form a 4 to 7 membered ring, such as piropyridine, all of which may be optionally substituted: R3, X, Y and MX are as defined above. 4. A compound of formula III, preferably a compound of formula IV: 或其醫藥可接考前藥、鹽類、溶劑化物、立體異構物、 互變異構物或多形性形式, 其中’ M-X、X、Y及R3如上述定義。 5. —種母藥’其適於衍生成如申請專利範圍第1、3或4項 之化合物,其爲下式化合物(D -及L-):Or a pharmaceutical thereof may be a prodrug, a salt, a solvate, a stereoisomer, a tautomer or a polymorphic form, wherein 'M-X, X, Y and R3 are as defined above. 5. A parent drug which is suitable for derivatization into a compound of the formula 1, 3 or 4 which is a compound of the formula (D- and L-): (A)選自 0、S、CH2、CHF、C = CH2、C = CHF、CF2; -55- 201210602 R8 及 R9 獨立選自 H、OH、CH30、F、Cl、Br、I、 CN'N3、甲基 '乙基、乙烯基、乙炔基、氯乙烯基、 氟甲基、二氟甲基、三氟甲基; R8及R9可形成可選擇經F、二氟、Cl、Br、I、CN、 N3取代之乙烯基; R1G選自Η、甲基、乙基、乙烯基、乙炔基、氯乙烯 基、溴乙烯基、碘乙烯基、氟甲基、二氟甲基、三氟 甲基,但不以此爲限; R11選自1^01^、仏、甲基、乙基、乙烯基、乙炔基、 氯乙烯基、溴乙烯基、碘乙烯基、氟甲基、二氟甲基、 三氟甲基,但不以此爲限; R12選自H、OH、F、氰基及疊氮基,但不以此爲限; B選自包括式B-l、B-2或B-3之嘧啶及嘌呤:(A) is selected from the group consisting of H, OH, CH30, F, Cl, Br, I , methyl 'ethyl, vinyl, ethynyl, chlorovinyl, fluoromethyl, difluoromethyl, trifluoromethyl; R8 and R9 can form F, difluoro, Cl, Br, I, CN, N3 substituted vinyl; R1G is selected from the group consisting of hydrazine, methyl, ethyl, vinyl, ethynyl, chlorovinyl, bromovinyl, iodovinyl, fluoromethyl, difluoromethyl, trifluoromethyl , but not limited to this; R11 is selected from the group consisting of 1^01^, hydrazine, methyl, ethyl, vinyl, ethynyl, chloroethylene, bromovinyl, iodovinyl, fluoromethyl, difluoromethyl , trifluoromethyl, but not limited to; R12 is selected from H, OH, F, cyano and azide, but not limited thereto; B is selected from the group consisting of Formula B, B-2 or B-3 Pyrimidine and hydrazine: 其中: X2 選自 H、NH2、NHMe、NMe2 或鹵素(I、Br、C1、 F),但不以此爲限; X4 爲 NH2 或 OH ; X5選自鹵素(I、Br、Cl、F)、OH、NH2、甲基、烷基、 乙烯基、2 -溴乙烯基、乙炔基,但不以此爲限; X6選自H、OH、OMe、OEt、SMe、烷氧基、芳氧基、 環烷氧基、烷基硫基、芳基硫基、環烷基硫基、噻吩 -56- 201210602 基、呋喃基、烷基胺基、二烷基胺基、 基烷基胺基、環烷基胺基、環丙基胺基 限; 芳基胺基、芳 ,但不以此爲 X7爲Η、乙烯基、乙炔基、齒素(I、^ 所有皆可選擇性經取代; F); 胺基及羥基可選擇性經保護; 其他經改質嘧啶或嘌呤,例如 3 -脫氮吡啶、3 -氟-3 -脫氮嘧啶 亦被視爲在本發明之範圍內。 氮_7-脫氮鳴 6. —種母藥’其適於衍生成如申請專利範圍第 之化合物,其爲下式化合物:Wherein: X2 is selected from H, NH2, NHMe, NMe2 or halogen (I, Br, C1, F), but not limited thereto; X4 is NH2 or OH; X5 is selected from halogen (I, Br, Cl, F) , OH, NH2, methyl, alkyl, vinyl, 2-bromovinyl, ethynyl, but not limited thereto; X6 is selected from H, OH, OMe, OEt, SMe, alkoxy, aryloxy , cycloalkoxy, alkylthio, arylthio, cycloalkylthio, thiophene-56-201210602, furyl, alkylamino, dialkylamino, alkylalkyl, ring Alkylamino group, cyclopropylamine group; arylamine group, aryl, but not X7 is oxime, vinyl, ethynyl, dentate (I, ^ can be selectively substituted; F) The amine group and the hydroxyl group may be selectively protected; other modified pyrimidines or purines such as 3-deazapyridine and 3-fluoro-3-deazapyrimidine are also considered to be within the scope of the present invention. Nitrogen-7-deazaring 6. A parent drug which is suitable for derivatization into a compound of the formula: which is a compound of the formula: 14 其中: B、A及R11如上述定義; R13及R14獨-ϋ*選自Η、N3、F、CN、CH3、可選擇經 取代之烷基或乙烯基,但不以此爲限。 3或4項 7 ·—種母藥,其適於衍生成如申請專利範圍第1 之化合物,其爲下式化合物(D-及L-): HO—I Β Η〇>5 其中: Β如上述定義。 -57- 201210602 8.—種母藥,其適於衍生成如申請專利範圍第1、3或4項 之化合物’其爲下式之生物上活性的鱗酸醋: 0 M-X-P"oh OH M-x可爲環狀或非環狀系統’例如m_ch2-p(〇)(〇h)2 nh2 丄 NH2 cl j Η〇ΗΟ^ν i? V^o Ή H〇J R=CH3 :泰諾福早 Cidofovir (Tenofovir) R=H :阿德福韋 西多福韋 (Adefovir) R=CH2OH : HPMPA nh2 η〇\_ </Νχ^Ν HO Ό nh2 0 N \ F GS-9148 PMDTA 9. 一種母藥,其適於衍生成如申請專利範圍第1' 3或4項 之化α物’其爲C -核甘及非環狀核音,包括阿昔洛韋 (acyclovir)、更昔洛韋(ganeyCi〇vjr)、噴昔洛韋 (pencyclovir)及其前藥,但不以此爲限。 10. —種如申g靑專利範圍第1、3或4項之化合物的醫藥可 接受性則藥、磷酸醋、鹽類、溶劑化物、立體異構物、 -58- 201210602 互變異構物或多形性型式,其亦被認爲包含於本發明之 範圍內。 η.—種使用作爲核苷、非環狀核苷、c-核苷、核苷酸、磷 酸酯及其他含醇藥劑之前藥,其係如申請專利範圍第 1、3或4項之化合物。 1 2 . —種用於治療方法之如申請專利範圍第1、3或4項之 化合物,適於預防或治療癌症,例如白血病、病毒感染 (包括HIV、HBV及HCV)及肝臟失調,包括肝癌及代謝 性疾病,例如糖尿病、高血脂症、動脈粥狀硬化及肥胖。 1 3 . —種如申請專利範圍第1、3或.4項之化合物於製造藥 劑上之用途,該藥劑用於預防或治療癌症,例如白血 病、病毒感染(包括HIV、HBV及HCV)及肝臟失調,包 括肝癌及代謝性疾病,例如糖尿病、高血脂症、動脈粥 狀硬化及肥胖。 14. 一種治療病毒感染(包括HIV、HBV及HCV)及肝臟失調 之方法,該肝臟失調包括肝癌及代謝性疾病,例如糖尿 病、高血脂症、動脈粥狀硬化及肥胖,該方法包含投與 人類個體治療有效量之如申請專利範圍第1、3或4項 之化合物。 15, —種治療病毒感染(包括HIV、HBV及HCV)及肝臟失調 之方法,該肝臟失調包括肝癌及代謝性疾病,例如糖尿 病、高血脂症、動脈粥狀硬化及肥胖,該方法包含合倂 投與人類個體治療有效量之如申請專利範圍第1、3或4 項之化合物及其他治療有效之藥劑。 -59- 201210602 1 6. —種醫藥組成物,其包含如申請專利範圍第1、3或4 項之化合物,並合倂醫藥可接受載劑或稀釋劑。 1 7. —種較佳之母藥,其適於衍生成如申請專利範圍第1、3 或4項之化合物(前藥),其爲表2中之各式之核苷(鳥苷 類似物,包括其之鳥嘌呤前藥)或其之單磷酸酯,但不 以此爲限, 表2.母核苷之實例14 wherein: B, A and R11 are as defined above; R13 and R14 are independently selected from the group consisting of hydrazine, N3, F, CN, CH3, optionally substituted alkyl or vinyl, but not limited thereto. 3 or 4 items of a parent drug which is suitable for derivatization into a compound of the first application formula, which is a compound of the formula (D- and L-): HO—I Β Η〇>5 wherein: Β As defined above. -57- 201210602 8. A parent drug suitable for the derivatization of a compound as described in claim 1, 3 or 4, which is a biologically active scaly vinegar of the formula: 0 MX-P"oh OH Mx can be a cyclic or acyclic system 'e.g. m_ch2-p(〇)(〇h)2 nh2 丄NH2 cl j Η〇ΗΟ^ν i? V^o Ή H〇JR=CH3: Tenofo early Cidofovir (Tenofovir) R=H: Adefovir R=CH2OH : HPMPA nh2 η〇\_ </Νχ^Ν HO Ό nh2 0 N \ F GS-9148 PMDTA 9. A parent drug It is suitable for derivatization into the alpha-like substance of the 1st to the 3rd or 4th of the patent application, which is a C-nuclear and acyclic nuclear tone, including acyclovir, ganciclovir (ganeyCi) 〇vjr), pencyclovir and its prodrugs, but not limited to this. 10. A pharmaceutical acceptable substance, a phosphate vinegar, a salt, a solvate, a stereoisomer, a -0 to 201210602 tautomer or a compound of the compound of claim 1, paragraph 3, or 4 Polymorphic forms are also considered to be within the scope of the invention. The compound used as a nucleoside, an acyclic nucleoside, a c-nucleoside, a nucleotide, a phosphate, and other alcohol-containing agents, which is a compound of the first, third or fourth aspect of the patent application. 1 2 . A compound for use in a therapeutic method as claimed in claim 1, 3 or 4, suitable for the prevention or treatment of cancer, such as leukemia, viral infections (including HIV, HBV and HCV) and liver disorders, including liver cancer And metabolic diseases such as diabetes, hyperlipidemia, atherosclerosis and obesity. 1 3 . The use of a compound as claimed in claim 1, 3 or . 4 for the manufacture of a medicament for the prevention or treatment of cancer, such as leukemia, viral infections (including HIV, HBV and HCV) and liver Disorders, including liver cancer and metabolic diseases such as diabetes, hyperlipidemia, atherosclerosis and obesity. 14. A method of treating viral infections (including HIV, HBV, and HCV) and liver disorders, including liver cancer and metabolic diseases such as diabetes, hyperlipidemia, atherosclerosis, and obesity, the method comprising administering to humans An individual therapeutically effective amount of a compound as claimed in claim 1, 3 or 4. 15. A method of treating viral infections (including HIV, HBV, and HCV) and liver disorders, including liver cancer and metabolic diseases such as diabetes, hyperlipidemia, atherosclerosis, and obesity, the method comprising A therapeutically effective amount of a compound administered in accordance with claim 1, 3 or 4 of the patent and other therapeutically effective agents. -59- 201210602 1 6. A pharmaceutical composition comprising a compound as claimed in claim 1, 3 or 4 in combination with a pharmaceutically acceptable carrier or diluent. 1 7. A preferred parent drug, which is suitable for the derivatization of a compound (prodrug) of the formula 1, 3 or 4, which is a nucleoside of the various formulas in Table 2 (guanosine analogues, Including its guanine prodrugs or their monophosphates, but not limited thereto, Table 2. Examples of parent nucleosides -60- 201210602-60- 201210602 -61 - 201210602-61 - 201210602 -62- 201210602 四、指定代表圖: (一) 本案指定代表圖為:無。 (二) 本代表圖之元件符號簡單說明: te。 五、本案若有化學式時,請揭示最能顯示發明特徵的化學式 M-X-P—Ο R1-62- 201210602 IV. Designated representative map: (1) The representative representative of the case is: None. (2) A brief description of the component symbols of this representative figure: te. 5. If there is a chemical formula in this case, please disclose the chemical formula M-X-P-Ο R1 which best shows the characteristics of the invention.
TW99129443A 2010-09-01 2010-09-01 Phosphorus-containing prodrugs and their therapeutic use TW201210602A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
TW99129443A TW201210602A (en) 2010-09-01 2010-09-01 Phosphorus-containing prodrugs and their therapeutic use

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
TW99129443A TW201210602A (en) 2010-09-01 2010-09-01 Phosphorus-containing prodrugs and their therapeutic use

Publications (1)

Publication Number Publication Date
TW201210602A true TW201210602A (en) 2012-03-16

Family

ID=46764056

Family Applications (1)

Application Number Title Priority Date Filing Date
TW99129443A TW201210602A (en) 2010-09-01 2010-09-01 Phosphorus-containing prodrugs and their therapeutic use

Country Status (1)

Country Link
TW (1) TW201210602A (en)

Similar Documents

Publication Publication Date Title
US9095599B2 (en) O-(substituted benzyl) phosphoramidate compounds and therapeutic use
US9156874B2 (en) Double-liver-targeting phosphoramidate and phosphonoamidate prodrugs
AU2012223012B2 (en) Phosphoramidate derivatives of 5 - fluoro - 2 ' - deoxyuridine for use in the treatment of cancer
ES2401070T3 (en) New prodrugs for phosphorus-containing compounds
JP2005503358A5 (en)
EP1284741B1 (en) 3'-or 2'-hydroxymethyl substituted nucleoside derivatives for treatment of viral infections
EP2619215B1 (en) Azido nucleosides and nucleotide analogs
Nauš et al. 6-(Het) aryl-7-deazapurine ribonucleosides as novel potent cytostatic agents
Mahmoud et al. Antiviral nucleoside and nucleotide analogs: a review
US7247621B2 (en) Antiviral phosphonate compounds and methods therefor
US8895531B2 (en) 2′-fluoronucleoside phosphonates as antiviral agents
JP4639032B2 (en) 3'-prodrug of 2'-deoxy-β-L-nucleoside
EP3546473A1 (en) Cyclic [(2',5')p(3',5')p]-dinucleotides for cytokine induction
JP2004522694A (en) Pyrid [2,3-D] pyrimidine and pyrimide [4,5-D] pyrimidine nucleoside
ES2496946T3 (en) Purine nucleotide derivatives
PT1937825T (en) Modified 4'-nucleosides as antiviral agents
WO2008083465A1 (en) Pyrimidine derivatives as anticancer agents
WO2004080466A1 (en) Cytidine analogs and methods of use
CA2476282A1 (en) Dosing regimen for gemcitabine hcv therapy
WO2003000200A2 (en) β-2'-OR 3'-HALONUCLEOSIDES
JP2008508319A (en) Novel nucleoside derivatives
JPWO2018199048A1 (en) 5'-position monophosphoric acid dibenzyl ester derivative of nucleoside anticancer agent or antiviral agent
TW201210602A (en) Phosphorus-containing prodrugs and their therapeutic use
Tokarenko Novel modified nucleosides with antiviral or cytostatic activity
US20040158054A1 (en) Di-ribonucleotides as specific viral RNA-polymerase inhibitors for the treatment or prevention of viral infections