TW200836731A - Amide substituted indazoles as poly (ADP-ribose) polymerase (PARP) inhibitors - Google Patents
Amide substituted indazoles as poly (ADP-ribose) polymerase (PARP) inhibitors Download PDFInfo
- Publication number
- TW200836731A TW200836731A TW097100730A TW97100730A TW200836731A TW 200836731 A TW200836731 A TW 200836731A TW 097100730 A TW097100730 A TW 097100730A TW 97100730 A TW97100730 A TW 97100730A TW 200836731 A TW200836731 A TW 200836731A
- Authority
- TW
- Taiwan
- Prior art keywords
- compound
- pharmaceutically acceptable
- doc
- formula
- acid
- Prior art date
Links
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D401/00—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
- C07D401/02—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
- C07D401/10—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing aromatic rings
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P29/00—Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
Landscapes
- Organic Chemistry (AREA)
- Chemical & Material Sciences (AREA)
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- General Health & Medical Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Pharmacology & Pharmacy (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Animal Behavior & Ethology (AREA)
- Medicinal Chemistry (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Pain & Pain Management (AREA)
- Rheumatology (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Plural Heterocyclic Compounds (AREA)
- Nitrogen Condensed Heterocyclic Rings (AREA)
Abstract
Description
200836731 九、發明說明: 【發明所屬之技術領域】 本發明係關於經醯胺取代之吲唑,其係先前稱作聚 (ADP-核糖)合成酶及聚(ADP-核糖基)轉移酶之酵素聚 (ADP-核糖)聚合酶(PARP)的抑制劑。本發明之化合物可用 作在DNA修復途徑中具有特定缺陷之腫瘤的單一療法及用 作諸如抗癌劑等某些DNA損害劑及放射療法之增強劑。而 且,本發明之化合物可用於減少細胞壞死(在中風及心肌 梗塞中)、下調炎症及組織損傷、治療逆轉錄病毒感染並 抵抗化學療法毒性。 【先前技術】 聚(ADP-核糖)聚合酶(PARP)組成18種含有PARP催化區 域之蛋白的超家族(5/ο α (2004) 26:1148)。此等蛋白 包括PARP-1、PARP-2、PARP-3、端錨聚合酶-1、端錨聚 合酶-2、vaultPARP及TiPARP。PARP-1(基礎成員)係由如 下3個主要結構域構成:含有2個辞指之胺基(N)-端DNA-結 合結構域(DBD)、自身修飾結構域及羧基(C)_端催化結構 域。 PARP係細胞核及細胞質酵素,其可使NAD+裂解成煙醯 胺及ADP-核糖以在目標蛋白上形成具支鏈長ADP-核糖聚 合物,該等目標蛋白包括拓撲異構酶、組蛋白及PARP自 身Ties. (1998) 245:1-10) 〇 (ADP-核糖基)聚合參與若干生物過程,包括DNA修復、 基因轉錄、細胞週期進展、細胞死亡、染色質功能及基因 127700.doc 200836731 組穩定。 已經證實藉由DNA鏈斷裂可迅速激發PARP-1及PARP-2 之催化活性(參見尸(2005) 52:25-33)。作為對DNA損害之響應,PARP-1結合單鏈及雙鏈 DNA缺口。在正常生理條件下,PARP具有最小活性,然 • 而,當DNA受到損害時,PARP活性即刻活化高達500倍。 , PARP-1及PARP-2二者均可檢測DNA鏈間斷,因此其可用 作缺口感受器、為停止轉錄提供快速信號並補充損害位點 ^ DNA修復所需酵素。由於癌症治療之許多化學治療方法及 放射治療藉由產生DNA損害起作用,因此PARP抑制劑可 用作癌症治療之化學增敏劑及放射增敏劑。據報道, PARP抑制劑可有效地放射敏化缺氧腫瘤細胞(美國專利第 US 5,032,617號、美國專利第US 5,215,738號及美國專利第 US 5,041,653 號)° PARP之大多數生物效應係與下列相關:可影響目標蛋 。 白之性質和功能的此(ADP-核糖基)聚合過程;在自(ADP- 核糖基)聚合蛋白裂解時產生不同細胞效應之PAR寡聚物; 形成功能複合體之PARP與核蛋白的物理締合;及其NAD+ . 之細胞水平降低(TVaiwre (2005) 4:421-440)。 除了參與DNA修復以外,PARP亦可用作細胞死亡之介 體。PARP在諸如局部缺血及再灌注損傷等病理條件下會 受到過度活化可導致細胞間NAD +之實質性缺失,進而可 造成若干NAD+依賴性代謝途徑受損並導致細胞死亡(參見 (2005) 52:44-59)。作為PARP活 127700.doc 200836731 化之結果,NAD +水平顯著下降。過度PARP活化可造成遭 受重度DNA損害之細胞的嚴重NAD+缺失。聚(ADP-核糖) 之較短半衰期可產生快速轉換率,此乃因聚(ADP-核糖)一 旦形成,其就會受到組成型活性聚(ADP_核糖)糖水解酶 (PARG)迅速降解。PARP及PARG形成一個循環,該循環將 大量NAD+轉化成ADP-核糖,導致NAD+及ATP降低至小於 正常水平之20%。此情形在局部缺血過程中當氧喪失已經 嚴重危害細胞能量輸出時尤其有害。吾人認為在再灌注過 程中之後續自由基生成係組織損害之主要原因。部分ATP 降低(在局部缺血及再灌注過程中通常見於許多器官中)可 能與由聚(ADP-核糖)轉換引起的NAD +缺失有關。因此, 預計,PARP抑制可維護細胞能量水平,進而增強缺血組 織在受損傷後之存活率。因此,作為PARP抑制劑之化合 物可用於治療由PARP介導之細胞死亡引起的病症,包括 諸如中風等神經性病症、創傷及帕金森氏病(Parkinson’s disease) 〇 已經證明PARP抑制劑可用於特異性殺傷BRCA-1及 BRCA-2 缺陷型腫瘤以π (2005) 434:913-916 和 917-921 ; Cancer Biology & Therapy (2005) 4:934-936) ° 已經證實PARP抑制劑可增進抗癌藥之功效 (Pharmacological Research (2005) 52:25-33),該等抗癌藥 包括諸如順翻及卡始等始化合物C/zemoi/zer P/mrmwo/ (1993) 33:157-162 及 Mo/ C仍cw 772er (2003) 2:371-382)。已經證實PARP抑制劑可增加諸如伊立替康 127700.doc 200836731 (Irinotecan)及拓撲替康(Topotecan)等拓撲異構酶I抑制劑 之抗腫瘤活性(Mo/ C⑽cer 77^r (2003) 2:371-382 ;及 C7z> (2000) 6:2860_2867)且此已在活體内模型中證 明(J Cancer /wi (2004) 96:56-67) 〇 已經證實PARP抑制劑可恢復對替莫唑胺 (temozolomide)(TMZ)細胞毒性及抗增生效應之敏感性(參 見 Cw" Med C/zem (2002) 9:1285-1301 及 MW C/zem (2004) 1:144-150)。此已在許多活體外模型(心J Cancer (1995) 72:849-856; Br J Cancer (1996) 74:1030-1036; Mol Pharmacol (1997) 52:249-258; Leukemia (1999) 13:901-909; G/k (2002) 40:44_54 ;及 C7z> C训cer 及以 (2000) 6:2860-2867 和(2004) 10:881-889)及活體内模型 {Blood (2002) 99:2241-2244; Clin Cancer Res (2003) 9:5370-5379 及 J TVai/ Cancer //7W (2004) 96:56-67)中證 明。亦已證實PAPR抑制劑可防止由諸如Me0S02(CH2)-lexitropsin (Me-Lex)等選擇性#3-腺嘌呤甲基化試劑引發的 壞死出現(尸/zarm/(9g/ca/ Tiaearc/z (2005) 52:25-33) 〇 已經證實PARP抑制劑可用作放射增敏劑。據報道, PARP抑制劑可有效地放射敏化(缺氧)腫瘤細胞並有效地防 止腫瘤細胞在放射治療後自DNA之潛在致死(5r. J· Cancer (1984) 49(增刊 VI):34-42;及/W. J· 山·αί. 5/oz·· (1999) 75:91-100)及亞致死(C7h· Ομο/· (2004) 16(1):29-39)損害 恢復,據推測,此係藉由其防止DNA鏈斷裂再結合之能力 並藉由影響若干DNA損害信號傳導途徑來達成。 127700.doc 200836731 亦已證實PARP抑制劑可用於治療急性及慢性心肌病(參 I Pharmacological (2005) 52:34-43) 〇 舉例而 言,已經證明在兔子中單獨注射PARP抑制劑使由心臟或 骨骼肌之再灌注及局部缺血造成的梗塞面積減少。在此等 研究中,於閉塞前1分鐘或於再灌注前1分鐘單獨注射3-胺 基-苯甲醯胺(10 mg/kg)在心臟中達成類似梗塞面積減小 (32-42%)而另一種PARP抑制劑1,5-二羥基異喹啉(1 mg/kg) 將梗塞面積減小相當程度(38-48%)。根據此等結果可合理 地設定PARP抑制劑可拯救先前局部缺血性心臟或骨骼肌 組織再灌注損傷(户见(1997) 94:679-683)。在豬中(五 J.尸Aarmaeo/. (1998) 359:143-150 及 d训.Swrg. (2002) 73:575-581)及在狗中(572〇ci (2004) 21:426-32)亦報 道有類似發現。 已經證明PARP抑制劑可用於治療某些血管疾病、敗血 性休克、局部缺血性損傷及神經毒性 Acta (1989) 1014:1-7; J. Clin, Invest. (1997) 100: 723-73 5)。導致DNA鏈斷裂之氧自由基DNA損害(其接下來可 藉由PARP識別)係此等病態之主要影響因素,如藉由PARP 抑制劑研究所證實(J. Λα. (1994) 39:38-46 及 尸见45 (1996) 93:468 8-4692)。亦已證明PARP在出血性休克 發病中起重要作用(P见(2000) 97:10203-10208)。 已經證明PARP抑制劑可用於治療炎症性疾病(參見 (2005) 52:72-82矛口 83-92) 〇 亦已證明藉由抑制PARP活性可阻斷哺乳動物細胞之有 127700.doc -10- 200836731 效逆轉錄病毒感染。已經證實此等重組逆轉錄病毒介體感 染之抑制可發生於各種不同的細胞類別中(J. hro/ogj;, (1996) 70(6):3 992-4000)。因此,已經研發出用於抗病毒 治療及癌症治療之PARP抑制劑(WO 91/18591)。 活體外及活體内實驗已證明PARP抑制劑可用於治療或 預防諸如I型糖尿病及糖尿病併發症等自身免疫疾病 (Pharmacological Research (20Q5) 52:60-7 〇 據推測PARP抑制可延遲人類纖維母細胞衰老特性出現 (Biochem. Biophys. Res. Comm· (Ϊ994) 20ί(2)'665-672 反 Pharmacological Research (2005) 52:93-99) ° ilt 可倉匕與 PARP在控制端粒作用中所起作用相關Ge/7·,(1999) 23(1):76-80)。 絕大部分PARP抑制劑與酵素之煙醯胺結合結構域定期 相互作用並相當於NAD+之競爭性抑制劑(五χρπί 77ζπ·尸如(2004) 14:153 1-155 1)。諸如苯甲醯胺及衍生 物等煙醯胺之結構類似物屬於欲經研究作為PARP抑制劑 之首要化合物。然而,此等分子具有較弱抑制活性且擁有 與PARP抑制不相關之其他作用。因此,人們需要提供 PARP酵素之強效抑制劑。200836731 IX. INSTRUCTIONS OF THE INVENTION: TECHNICAL FIELD OF THE INVENTION The present invention relates to guanamine substituted carbazoles, which are previously known as poly(ADP-ribose) synthase and poly(ADP-ribosyl)transferase enzymes. Inhibitor of poly(ADP-ribose) polymerase (PARP). The compounds of the present invention are useful as monotherapy for tumors with specific defects in the DNA repair pathway and as inhibitors of certain DNA damaging agents such as anticancer agents and radiation therapy. Moreover, the compounds of the invention are useful for reducing cell necrosis (in stroke and myocardial infarction), down-regulating inflammation and tissue damage, treating retroviral infections, and resisting chemotherapy toxicity. [Prior Art] Poly(ADP-ribose) polymerase (PARP) constitutes a superfamily of 18 proteins containing the catalytic domain of PARP (5/ο α (2004) 26:1148). Such proteins include PARP-1, PARP-2, PARP-3, tankyrase-1, terminal anchor polymerase-2, vaultPARP and TiPARP. PARP-1 (basal member) is composed of three main domains: an amino group (N)-terminal DNA-binding domain (DBD) containing two words, a self-modifying domain, and a carboxyl group (C)_end Catalytic domain. PARP is a nuclear and cytoplasmic enzyme that cleaves NAD+ into nicotinamide and ADP-ribose to form a branched-length ADP-ribose polymer on the target protein, including topoisomerase, histone, and PARP. Self-Ties. (1998) 245:1-10) Anthraquinone (ADP-ribosyl) polymerization is involved in several biological processes including DNA repair, gene transcription, cell cycle progression, cell death, chromatin function, and gene 127700.doc 200836731 . The catalytic activity of PARP-1 and PARP-2 has been rapidly stimulated by DNA strand breaks (see Corpse (2005) 52: 25-33). In response to DNA damage, PARP-1 binds to single-stranded and double-stranded DNA gaps. Under normal physiological conditions, PARP has minimal activity, however, when DNA is damaged, PARP activity is instantly activated up to 500-fold. Both PARP-1 and PARP-2 can detect DNA strand breaks, so they can be used as gap receptors to provide a fast signal for stopping transcription and to supplement the enzymes required for DNA repair. Since many chemical treatments and radiation treatments for cancer treatment play a role in DNA damage, PARP inhibitors can be used as chemical sensitizers and radiosensitizers for cancer treatment. It is reported that PARP inhibitors are effective for radiosensitizing hypoxic tumor cells (U.S. Patent No. 5,032,617, U.S. Patent No. 5,215,738, and U.S. Patent No. 5,041,653). Related: Can affect the target egg. This (ADP-ribosyl) polymerization process of nature and function of white; PAR oligomers that produce different cellular effects upon cleavage of (ADP-ribosyl) polymeric proteins; physical association of PARP and nuclear proteins that form functional complexes Reduced cell levels; (TVaiwre (2005) 4:421-440). In addition to participating in DNA repair, PARP can also be used as a mediator of cell death. Over-activation of PARP under pathological conditions such as ischemia and reperfusion injury can result in substantial loss of NAD+ between cells, which can result in several NAD+-dependent metabolic pathways being impaired and leading to cell death (see (2005) 52). :44-59). As a result of PARP live 127700.doc 200836731, NAD + levels decreased significantly. Excessive PARP activation can cause severe NAD+ deficiency in cells that are severely damaged by DNA. The shorter half-life of poly(ADP-ribose) produces a rapid conversion rate, which is due to the rapid degradation of the constitutively active poly(ADP-ribose) sugar hydrolase (PARG) as a result of the formation of poly(ADP-ribose). PARP and PARG form a cycle that converts a large amount of NAD+ to ADP-ribose, resulting in a reduction in NAD+ and ATP to less than 20% of normal levels. This condition is particularly detrimental during ischemia, when oxygen loss has severely compromised cellular energy output. I believe that the subsequent free radical generation in the reperfusion process is the main cause of tissue damage. Partial ATP reduction (commonly seen in many organs during ischemia and reperfusion) may be associated with NAD+ deficiency caused by poly(ADP-ribose) turnover. Therefore, it is expected that PARP inhibition can maintain cellular energy levels, thereby enhancing the survival rate of ischemic tissue after injury. Thus, compounds that are PARP inhibitors are useful in the treatment of conditions caused by PARP-mediated cell death, including neurological disorders such as stroke, trauma, and Parkinson's disease. Parkerson's disease has been demonstrated to be useful for PARP inhibitors. Killing BRCA-1 and BRCA-2 deficient tumors with π (2005) 434: 913-916 and 917-921; Cancer Biology & Therapy (2005) 4: 934-936) ° PARP inhibitors have been shown to promote anticancer The efficacy of the drug (Pharmacological Research (2005) 52:25-33), such as the start compound C/zemoi/zer P/mrmwo/ (1993) 33:157-162 and Mo /C still cw 772er (2003) 2:371-382). PARP inhibitors have been shown to increase the antitumor activity of topoisomerase I inhibitors such as irinotecan 127700.doc 200836731 (Irinotecan) and topotecan (Mo/C(10)cer 77^r (2003) 2:371 -382; and C7z> (2000) 6:2860_2867) and this has been demonstrated in an in vivo model (J Cancer /wi (2004) 96:56-67). It has been demonstrated that PARP inhibitors can restore to temozolomide (temozolomide) ( TMZ) Sensitivity to cytotoxic and anti-proliferative effects (see Cw " Med C/zem (2002) 9: 1285-1301 and MW C/zem (2004) 1: 144-150). This has been in many in vitro models (Heart J Cancer (1995) 72: 849-856; Br J Cancer (1996) 74: 1030-1036; Mol Pharmacol (1997) 52: 249-258; Leukemia (1999) 13: 901 -909; G/k (2002) 40:44_54; and C7z> C training cer and (2000) 6:2860-2867 and (2004) 10:881-889) and in vivo models {Blood (2002) 99: 2241-2244; Clin Cancer Res (2003) 9:5370-5379 and J TVai/ Cancer //7W (2004) 96:56-67). It has also been demonstrated that PAPR inhibitors prevent the appearance of necrosis caused by selective #3-adenine methylation reagents such as Me0S02(CH2)-lexitropsin (Me-Lex) (corpse/zarm/(9g/ca/ Tiaearc/z (2005) 52:25-33) PAR has been shown to be useful as a radiosensitizer for PARP inhibitors. It has been reported that PARP inhibitors are effective in radiosensitizing (hypoxic) tumor cells and effectively preventing tumor cells from being treated with radiation. Potential death from DNA (5r. J. Cancer (1984) 49 (Supplement VI): 34-42; and /W. J. Shan·αί. 5/oz·· (1999) 75:91-100) and Sublethal death (C7h· Ομο/· (2004) 16(1): 29-39) damage recovery, presumably by virtue of its ability to prevent re-binding of DNA strand breaks and by affecting several DNA-damaging signaling pathways 127700.doc 200836731 PARP inhibitors have also been shown to be useful in the treatment of acute and chronic cardiomyopathy (see I Pharmacological (2005) 52: 34-43). For example, it has been demonstrated that PARP inhibitors alone in rabbits have been shown to be Reinfusion of the heart or skeletal muscle and reduction of infarct size caused by ischemia. In these studies, 1 minute before occlusion or reperfusion A single injection of 3-amino-benzamide (10 mg/kg) in the first minute resulted in a similar reduction in infarct size (32-42%) in the heart and another PARP inhibitor 1,5-dihydroxyisoquinoline (1 mg/kg) reduces the infarct size to a considerable extent (38-48%). Based on these results, it is reasonable to set the PARP inhibitor to rescue the reperfusion injury of the previously ischemic heart or skeletal muscle tissue. 1997) 94: 679-683). In pigs (five J. corpse Aarmaeo/. (1998) 359: 143-150 and d. Swrg. (2002) 73: 575-581) and in dogs (572 〇) Similar findings have been reported in ci (2004) 21:426-32. PARP inhibitors have been shown to be useful in the treatment of certain vascular diseases, septic shock, ischemic injury and neurotoxicity. Acta (1989) 1014:1-7 J. Clin, Invest. (1997) 100: 723-73 5). Oxygen free radical DNA damage leading to DNA strand breaks (which can be subsequently identified by PARP) is a major contributor to these pathologies, as evidenced by the PARP Inhibitors Institute (J. Λα. (1994) 39:38- 46 and corpse 45 (1996) 93: 468 8-4692). PARP has also been shown to play an important role in the pathogenesis of hemorrhagic shock (see P. (2000) 97: 10203-10208). PARP inhibitors have been shown to be useful in the treatment of inflammatory diseases (see (2005) 52: 72-82 Spear 83-92). It has also been shown that by inhibiting PARP activity, mammalian cells can be blocked by 127700.doc -10- 200836731 Retroviral infection. Inhibition of these recombinant retroviral mediator infections has been shown to occur in a variety of different cell classes (J. hro/ogj;, (1996) 70(6): 3 992-4000). Therefore, PARP inhibitors for antiviral therapy and cancer treatment have been developed (WO 91/18591). In vitro and in vivo experiments have demonstrated that PARP inhibitors can be used to treat or prevent autoimmune diseases such as type 1 diabetes and diabetic complications (Pharmacological Research (20Q5) 52:60-7. It is speculated that PARP inhibition can delay human fibroblasts. Aging characteristics appear (Biochem. Biophys. Res. Comm· (Ϊ994) 20ί(2)'665-672 Anti-Pharmacological Research (2005) 52:93-99) ° ilt can be used in the control of telomere Role-dependent Ge/7·, (1999) 23(1): 76-80). The vast majority of PARP inhibitors interact regularly with the nicotinamide binding domain of the enzyme and are equivalent to a competitive inhibitor of NAD+ (5χρπί 77ζπ·尸如 (2004) 14:153 1-155 1). Structural analogs of the nicotinamide such as benzamide and derivatives belong to the primary compound to be studied as a PARP inhibitor. However, these molecules have weak inhibitory activity and have other effects that are not associated with PARP inhibition. Therefore, there is a need to provide potent inhibitors of PARP enzymes.
先前已經闡述結構相關之PARP抑制劑。WO 1999/59973 揭示與5員雜芳香族環稠合之經醯胺取代之苯環; W0200 1/85687揭示經醯胺取代之吲哚;WO 1997/04771、 WO 2000/26192、WO 2000/32579、WO 2000/64878、WO 2000/68206、WO 2001/21615 、WO 2002/068407、WO 127700.doc -11 - 200836731 2 003/1 06430及WO 2004/096793揭示經醯胺取代之苯并咪 唑;WO 2000/29384揭示經醯胺取代之苯并咪唑及吲哚; 且歐洲專利EP 0879820揭示經醯胺取代之苯并噁唑。 現在,人們驚奇地發現本發明經醯胺取代之吲唑對聚 (ADP-核糖)聚合酶(PARP)活性抑制呈現特別高的水平。因 ^ 此,本發明之化合物尤其可用作PARP-1及/或PARP-2之抑 ^ 制劑。該等化合物亦顯示特別佳的細胞活性水平,證明在 BRCA1及BRCA2缺陷之細胞系中具有良好抗增生作用。 "' 【發明内容】 本發明提供式I之化合物:Structurally related PARP inhibitors have been previously described. WO 1999/59973 discloses a guanamine-substituted benzene ring fused to a 5-membered heteroaromatic ring; W0200 1/85687 discloses a guanidine-substituted oxime; WO 1997/04771, WO 2000/26192, WO 2000/32579 WO 2000/64878, WO 2000/68206, WO 2001/21615, WO 2002/068407, WO 127700.doc -11 - 200836731 2 003/1 06430 and WO 2004/096793 disclose benzimidazole substituted by decylamine; WO 2000/29384 discloses benzimidazole and hydrazine substituted with decylamine; and European Patent EP 0879820 discloses benzoxazole substituted with decylamine. It has now surprisingly been found that the indoleamine substituted oxazoles of the present invention exhibit particularly high levels of inhibition of poly(ADP-ribose) polymerase (PARP) activity. As a result, the compounds of the present invention are especially useful as inhibitors of PARP-1 and/or PARP-2. These compounds also showed particularly good levels of cellular activity, demonstrating good antiproliferative effects in BRCA1 and BRCA2-deficient cell lines. "' [Invention] The present invention provides a compound of formula I:
其中: R1係氫或氟;且 R2係氫或氟; 或其醫藥上可接受之鹽、立體異構體或互變異構體。 在一實施例中,R1係氫。 在另一實施例中,R1係氟。 在一實施例中,R2係氫。 在另一實施例中,R2係氟。 在一實施例中,R1係氫且R2係氫或氟。 在另一實施例中,R1係氟且R2係氫或氟。 127700.doc -12- 200836731 在另一實施例中,R1係氫且R2係氫。 在另一實施例中,R1係氫且R2係氟。 在另一實施例中,R1係氟且R2係氟。 在另一實施例中,R1係氫或氟且R2係氫。 在另一實施例中,R1係氫或氟且R2係氟。 本發明亦提供式II之化合物: CONH.Wherein: R1 is hydrogen or fluorine; and R2 is hydrogen or fluorine; or a pharmaceutically acceptable salt, stereoisomer or tautomer thereof. In one embodiment, R1 is hydrogen. In another embodiment, R1 is fluoro. In one embodiment, R2 is hydrogen. In another embodiment, R2 is fluoro. In one embodiment, R1 is hydrogen and R2 is hydrogen or fluorine. In another embodiment, R1 is fluoro and R2 is hydrogen or fluoro. 127700.doc -12- 200836731 In another embodiment, R1 is hydrogen and R2 is hydrogen. In another embodiment, R1 is hydrogen and R2 is fluorine. In another embodiment, R1 is fluoro and R2 is fluoro. In another embodiment, R1 is hydrogen or fluorine and R2 is hydrogen. In another embodiment, R1 is hydrogen or fluorine and R2 is fluorine. The invention also provides a compound of formula II: CONH.
(Π) 其中R1及R2係如上文所定義; 或其醫藥上可接受之鹽、立體異構體或互變異構體。 本發明亦提供式III之化合物:(Π) wherein R1 and R2 are as defined above; or a pharmaceutically acceptable salt, stereoisomer or tautomer thereof. The invention also provides a compound of formula III:
其中R1及R2係如上文所定義; 或其醫藥上可接受之鹽或互變異構體。 本發明亦提供式IV之化合物:Wherein R1 and R2 are as defined above; or a pharmaceutically acceptable salt or tautomer thereof. The invention also provides a compound of formula IV:
(IV) 127700.doc -13- 200836731 其中R1及R2係如上文所定義; 或其醫藥上可接受之鹽或互變異構體。 【實施方式】 式II、III及IV之較佳身份係如前文對式〗所定義(加以必 要的變通)。 本發明在其範圍内亦涵蓋上式I化合物之N_氧化物。一 般而言,此等N-氧化物可在任一可用氮原子上形成。該等 N-氧化物可藉由習知方法(例如,使式j化合物與過硫酸氫 鉀製劑(oxone)於濕礬土存在下反應)形成。 本發明在其範圍内涵蓋上式Ϊ化合物之前藥。一般而 吕,此等刖藥可為在活體内易於轉化成所需式以匕合物的 式I化合物之功能衍生物。用於選擇及製備適宜前藥衍生 物之習知程序闡述於(例如)”Design 〇f pr〇drugsn,乩(IV) 127700.doc -13- 200836731 wherein R1 and R2 are as defined above; or a pharmaceutically acceptable salt or tautomer thereof. [Embodiment] The preferred identities of the formulae II, III and IV are as defined above for the formula (with necessary modifications). The invention also encompasses within its scope N-oxides of the compounds of formula I above. In general, such N-oxides can be formed on any of the available nitrogen atoms. The N-oxides can be formed by conventional methods (e.g., by reacting a compound of formula j with an oxone in the presence of moist soil). The present invention encompasses, in its scope, prodrugs of the above formula. In general, such peony may be a functional derivative of a compound of formula I which is readily converted in vivo to the desired formula. A conventional procedure for selecting and preparing suitable prodrug derivatives is described, for example, in "Design 〇f pr〇drugsn,"
Bundgaard編寫,Elsevier,1985 中。 前藥可為生物活性物質("母體藥物”或”母體分子,,)之藥 理惰性衍生物,其需要在體内轉化以釋放活性藥物且其較 母體藥物分子具有改良遞送性質。活體内轉化可為(例如) 某些代謝過程(例如,羧酸酯、磷酸酯或硫酸酯之化學或 酶促水解、或敏感性官能團之還原或氧化)之結果。 本發明在其範圍内涵蓋式〗化合物之溶劑合物及其鹽, 例如,水合物。 本發明之化合物可具有不對稱中心、對掌性軸及對掌性 平面(如闡述於下列中者:EL· Eliel與s H. wiien,Written by Bundgaard, Elsevier, 1985. The prodrug may be a pharmacologically inert derivative of a biologically active substance ("parent drug" or "parent molecule,", which is required to be converted in vivo to release the active drug and which has improved delivery properties over the parent drug molecule. In vivo transformation can be the result, for example, of certain metabolic processes (e.g., chemical or enzymatic hydrolysis of a carboxylic acid ester, phosphate or sulfate, or reduction or oxidation of a sensitive functional group). The present invention encompasses within its scope solvates of the compounds and salts thereof, for example, hydrates. The compounds of the present invention may have an asymmetric center, a palm axis, and a palm plane (as described in the following: EL·Eliel and s H. wiien,
Stereochemistry of Carbon Compounds ^ John Wiley & Sons ^? 127700.doc -14- 200836731 紐約,撕’第⑴9-⑽頁),且作為外消旋異構體、外 消旋混合物、及作為個別非對映異構體連同其所有可能異 構體及混合物(包括光學異構體)存在,所有該等立體異構 體皆涵蓋於本發明内。此外,本文所揭示化合物可作為互 變異構體存纟且兩種互變異構形式均⑨涵蓋於本發明之範 圍内,即使僅闡述一種互變異構結構。 該等化合物可以不同的同分異構體形式存在,所有該等 同分異構體形式均涵蓋於本發明中。 該等化合物可以許多不同的多晶型形式存在。 本文所用C!·6烧基表示含有1個、2個、3個、4個、5個或 6個峡原子之具支鏈、直鏈及環狀飽和脂肪族烴基團。舉 例而言,nCi_6烷基”具體包括甲基、乙基、正-丙基、異-丙 基、正-丁基、弟二-丁基、異-丁基、戊基、己基、環丙 基、環丁基、環戊基及環己基等。較佳烷基基團係甲基及 乙基。 屬於本發明範圍之特定化合物係: 氯化3-{4-[7-(胺基羰基)-2//-吲唑-2-基]苯基}六氫吼啶 鏽; 2-{4_[(3R)-六氫吡啶-3-基]苯基吲唑-7-甲醯胺; 2-{4-[(38)_六氫吡啶-3-基]苯基}-2孖-吲唑-7_甲醯胺; 三氟乙酸3-{4-[7-(胺基羰基)-5-氟-27/-吲唑-2-基]苯基} 六氫°比σ定鑌; 5-氟-2-(3-氟-4-六氫吡啶-3-基苯基)-2开-吲唑-7-甲醯胺 三氟乙酸鹽; 127700.doc -15- 200836731 三氟乙酸3-{4-[7-(胺基羰基)-2H-吲唑-2-基]苯基}六氫吧 啶鏽; 5-氟-2-(4-六氫吡啶-3-基苯基)-2Η·吲唑_7_甲醯胺; 氯化(3S)-3-{4-[7-(胺基羰基)-2Η·吲唑-2-基]苯基}六氫 吡咬鑌; 氣化(3R)-3-{4-[7-(胺基羰基)-2Η-吲唑-2-基]苯基}六氫 吡啶鏽; (R) -5_氟-2-(4-六氫吡啶-3-基苯基)-2Η-吲唑-7-甲醯胺; (S) -5-氟-2-(4-六氫吡啶-3-基苯基)-2Η-吲唑-7-甲醯胺; (R) -5-氟-2-{3·氟-4_六氫吼啶-3-基苯基}-2Η-吲唑-7-甲 醯胺; (S) -5-氟-2-{3-氟·4-六氫吡啶-3-基苯基}-2Η-吲唑-7-甲醯 胺; 及其醫藥上可接受之鹽、游離鹼或互變異構體。亦提供此 等化合物之立體異構體。 本發明之特定化合物係: 氯化3-{4-[7-(胺基羰基)-2//-吲唑-2-基]苯基}六氫π比啶 錯; 或其醫藥上可接受之游離鹼或互變異構體。亦提供此化合 物之立體異構體。 本發明之特定化合物係: 2-d[(3R)-六氫咄啶·3_基]苯基卜吲唑·7_甲醯胺; 或其醫藥上可接受之鹽、游離鹼或互變異構體。亦提供此 化合物之立體異構體。 127700.doc -16- 200836731 本發明之特定化合物係: 2-{4-[(3S)-六氫吡啶-3-基]苯基吲唑-7-甲醯胺; 及其醫藥上可接受之鹽、游離鹼或互變異構體。亦提供此 化合物之立體異構體。 本發明之特定化合物係: 三敦乙酸3-{4-[7-(胺基羰基)-5-氟-2//-吲唑-2-基]苯基} 六氫吡啶銪; 或其醫藥上可接受之游離鹼或互變異構體。亦提供此化合 ( 物之立體異構體。 本發明之特定化合物係·· 5_氟-2-(3-氟-4·六氫吡啶-3-基苯基)-2//-吲唑_7_甲醯胺 三氣乙酸鹽; 或其醫藥上可接受之游離鹼或互變異構體。亦提供此化合 物之立體異構體。 本發明之特定化合物係: c; 4·甲基 苯磺酸(3S)-3-{4-[7-(胺基羰基)-2H-吲唑-2-基]苯 基}六氫°比σ定鑌; 或其醫藥上可接受之游離鹼或互變異構體。亦提供此化合 • 物之立體異構體。 。 本發明包括式I化合物之游離鹼以及其醫藥上可接受之 |及立體異構體。本發明之化合物可在胺之汉原子處及/戋 ==分受到質子化以形成鹽。術語”游離鹼,,係指該 己物呈非鹽形式。所涵蓋醫藥上可接受之鹽不僅包 括本文所闡述具體化合物之示例性鹽,而且亦包括所有游 127700.doc -17- 200836731 離形式式i化合物之典型醫藥上 巧文又< 盟。所闡述且體 鹽化合物之游離形式可佳w #语 "" ㈣式了使用6亥項技術已知的技術分離。舉 例而言,該游離形式可藉由用適宜稀驗性水溶液(例如Stereochemistry of Carbon Compounds ^ John Wiley & Sons ^? 127700.doc -14- 200836731 New York, tearing 'Pages (1) 9-(10)), and as racemic isomers, racemic mixtures, and as individual diastereoisomers Isomers are present along with all possible isomers and mixtures thereof, including optical isomers, and all such stereoisomers are encompassed by the present invention. Furthermore, the compounds disclosed herein can be considered as tautomers and both tautomeric forms are encompassed within the scope of the invention, even if only one tautomeric structure is illustrated. The compounds may exist in different isomeric forms, and all such isomeric forms are encompassed by the present invention. These compounds can exist in many different polymorphic forms. As used herein, C!·6 alkyl represents a branched, straight-chain, and cyclic saturated aliphatic hydrocarbon group containing one, two, three, four, five, or six gorgon atoms. For example, nCi_6 alkyl" specifically includes methyl, ethyl, n-propyl, iso-propyl, n-butyl, di-butyl, iso-butyl, pentyl, hexyl, cyclopropyl. , cyclobutyl, cyclopentyl and cyclohexyl, etc. Preferred alkyl groups are methyl and ethyl. Specific compounds belonging to the scope of the invention are: 3-{4-[7-(aminocarbonyl) chloride -2//-carbazol-2-yl]phenyl}hexahydroacridine rust; 2-{4_[(3R)-hexahydropyridin-3-yl]phenylcarbazole-7-formamide; -{4-[(38)_hexahydropyridin-3-yl]phenyl}-2孖-carbazole-7-formamide; 3-{4-[7-(aminocarbonyl)-trifluoroacetate- 5-fluoro-27/-oxazol-2-yl]phenyl} hexahydrogen ratio σ 镔; 5-fluoro-2-(3-fluoro-4-hexahydropyridin-3-ylphenyl)-2 Open-carbazole-7-formamide trifluoroacetate; 127700.doc -15- 200836731 3-{4-[7-(aminocarbonyl)-2H-indazol-2-yl]phenyl trifluoroacetate } hexahydrobaridine rust; 5-fluoro-2-(4-hexahydropyridin-3-ylphenyl)-2Η·carbazole-7-carbamamine; chlorinated (3S)-3-{4-[ 7-(Aminocarbonyl)-2Η·oxazol-2-yl]phenyl}hexahydropyridinium; gasification (3R)-3-{4-[7-(aminocarbonyl)-2Η-carbazole -2-yl]phenyl}hexahydropyridine rust; (R) -5-fluoro-2-(4-hexahydropyridin-3-ylphenyl)-2-indole-indazole-7-carboxamide; (S)-5-fluoro-2-(4-hexahydro) Pyridin-3-ylphenyl)-2indole-carbazole-7-formamide; (R)-5-fluoro-2-{3·fluoro-4_hexahydroacridin-3-ylphenyl}-2Η - carbazole-7-formamide; (S) -5-fluoro-2-{3-fluoro-4-hydropyridin-3-ylphenyl}-2 oxime-carbazole-7-carboxamide; The pharmaceutically acceptable salts, free bases or tautomers thereof. Stereoisomers of such compounds are also provided. The specific compounds of the invention are: 3-{4-[7-(aminocarbonyl)- 2//-carbazol-2-yl]phenyl}hexahydropyridinium pyridine; or a pharmaceutically acceptable free base or tautomer thereof. Also provided are stereoisomers of the compound. The compound is: 2-d[(3R)-hexahydroacridin-3-yl]phenylpyrazole·7-formamide; or a pharmaceutically acceptable salt, free base or tautomer thereof. The stereoisomer of this compound is provided. 127700.doc -16- 200836731 The specific compound of the invention is: 2-{4-[(3S)-hexahydropyridin-3-yl]phenylcarbazole-7-formamidine An amine; and a pharmaceutically acceptable salt, free base or tautomer thereof. The stereoisomer of this compound is provided. The specific compound of the present invention is: 3-D4-[7-(aminocarbonyl)-5-fluoro-2//-oxazol-2-yl]phenyl } Hexahydropyridinium; or a pharmaceutically acceptable free base or tautomer thereof. This compound is also provided as a stereoisomer. The specific compound of the present invention is 5_fluoro-2-(3-fluoro-4·hexahydropyridin-3-ylphenyl)-2//-carbazole _7_methantamine trigas acetate; or a pharmaceutically acceptable free base or tautomer thereof. Also provided are stereoisomers of this compound. The specific compounds of the invention are: c; 4·methylbenzene Sulfonic acid (3S)-3-{4-[7-(aminocarbonyl)-2H-indazol-2-yl]phenyl}hexahydrogen ratio sigma; or its pharmaceutically acceptable free base or Tautomers. Stereoisomers of such compounds are also provided. The present invention includes the free bases of the compounds of Formula I, as well as pharmaceutically acceptable | and stereoisomers thereof. The atom and /戋== are protonated to form a salt. The term "free base" means that the substance is in a non-salt form. The pharmaceutically acceptable salts encompassed include not only exemplary salts of the specific compounds described herein. And also includes all the travels of 127700.doc -17- 200836731 from the typical medicinal formula of the compound of formula i and < 盟. W # language type can be good " ". (Iv) of formula 6 using techniques known in the art items Hai For isolated cases, it may be by the free form with a suitable dilute aqueous transcendental (e.g.
Na〇H、碳酸鉀、氨及碳酸⑭之稀水溶液)處理該鹽再 生。該等游離形式盥J:久白鵾4 # 飞,、/、各自鹽形式在某些物理性質(例 如,在極性溶劑中的溶解性)上可有區別,但對於本發明 之目的而言該等酸式及驗式鹽亦為其各游離形式之醫藥等 效物。 ' 本發明化合物之醫藥上可接受之鹽可自包含鹼性或酸性 部分之本發明化合物藉由習知化學方法來合成。通常,續 等驗性化合物之鹽係藉由離子交換層析或藉由在適宜溶劑 或各種溶劑組合中使該游離驗與化學計量量或與過量形成 期望鹽之無機H有機酸反應來製備H該等酸性化 合物之鹽可藉由與適宜無機或有機鹼反應來形成。 因此’本發明化合物之醫藥上可接受之鹽包括本發明化 合物之習知無#性鹽,如藉由使驗性本發明化合物與無機 酸、有機酸或聚合酸反應所形成者。舉例而言,習知無毒 性鹽包括彼等衍生自無機酸(例如,氫氯酸、氫漠酸、氫 蛾酸、硫酸、亞硫酸、胺基續酸、麟酸、亞填酸、硝酸及 諸如此類)者以及自有機酸(例如,乙酸、丙酸、琥始酸、 乙醇酸、硬脂酸、乳酸、絲酸、酒石酸、棒樣酸、抗壞 金酸、巴莫酸、馬來酸、經基馬來酸、苯基乙酸、麵胺 -文苯甲駄、水揚酸、對胺基苯磺酸、2-乙醯氧基-苯甲 酸、富馬酸 '曱笨績酸、甲烧續酸、乙烧二續酸、草酸、 127700.doc -18· 200836731 羥乙磺酸、棕閭酸、葡萄糖酸、抗壞血酸、苯基乙酸、天 冬胺酸、肉桂酸、丙酮酸、乙烷磺酸、乙烷二磺酸、戊 酸、三氟乙酸及諸如此類)製備的鹽。適宜聚合酸鹽之實 例包括彼等衍生自諸如鞣酸、羧甲基纖維素等聚合物酸 者。較佳地,本發明之醫藥上可接受之鹽包含1當量式⑴ 化合物及1、2或3當量無機酸或有機酸。更具體而言,本 發明之醫藥上可接受之鹽係三氟乙酸鹽、氣化物或甲苯石备 酸鹽。更具體而言,本發明之醫藥上可接受之鹽係三氟乙 酸鹽或氣化物鹽。在一實施例中,該鹽係三氟乙酸鹽。在 另一實施例中,該鹽係氯化物。在另一實施例中,該鹽係 曱苯磺酸鹽。 當本發明化合物為酸性時,適宜”醫藥上可接受之鹽,,係 指自醫藥上可接受之無毒性鹼(包括無機鹼及有機鹼)製備 的鹽。衍生自無機驗之鹽包括鋁鹽、銨鹽、趟鹽、銅鹽、 鐵鹽、亞鐵鹽、鐘鹽、鎂鹽、錳鹽、二價錳鹽、鉀鹽、鈉 鹽、鋅鹽及諸如此類。尤佳者係銨鹽、鈣鹽、鎂鹽、鉀鹽 以及鈉鹽。衍生自醫藥上可接受之有機無毒性鹼的鹽包括 一級胺、二級胺及三級胺、經取代胺(包括天然經取代 )、環胺以及驗性離子交換樹脂的鹽,諸如精胺酸、離 胺酸、甜菜鹼、咖啡因、膽鹼、N,NL二苄基乙二胺、乙 胺、一乙胺、2-二乙基胺基乙醇、2-二甲基胺基乙醇、乙 醇胺、二乙醇胺、乙二胺、N_乙基嗎啉、N_乙基六氫吡 % 、還原葡糖胺、葡萄糖胺、組胺酸、哈胺 (hydrabamine)、異丙胺、離胺酸、甲基還原葡糖胺、嗎 127700.doc -19- 200836731 琳、旅嗓、六氫ϋ比tr定、聚胺樹脂、普魯卡因、嗓呤、可可 鹼、三乙胺、三甲胺、三丙胺、胺丁三醇、二環己基胺、 丁胺、苄胺、苯基苄基胺、胺丁三醇等的鹽。 上述醫藥上可接受之鹽及其他典型醫藥上可接受之鹽的 製備更全面地闡述於Berg等人(1977),J.尸/mrm. T/zarmacezzi/ca/ Χα"〆,66:1 -1 9 中 〇 亦應注意:由於在生理條件下本發明化合物之去質子化 酸性部分(例如,羧基基團)可為陰離子,且該電子電荷隨 後可在内部被經質子化或經烷基化鹼性部分(例如,四級 氮原子)之陽離子電荷平衡掉,故該化合物可能為内鹽或 兩性離子。 在藉由療法治療人類或動物體之方法中可使用本發明之 化合物。 本發明提供用於治療或預防可藉由抑制聚(ADP-核糖)聚 合酶(PARP)加以改善之病況的化合物(參見,例如,The salt is regenerated by treating Na〇H, potassium carbonate, ammonia and a dilute aqueous solution of carbonic acid 14. The free forms 盥J: 久白鵾4 #飞,, /, respective salt forms may differ in certain physical properties (e.g., solubility in polar solvents), but for the purposes of the present invention The acid and test salts are also pharmaceutical equivalents in their free form. The pharmaceutically acceptable salt of the compound of the present invention can be synthesized from a compound of the present invention containing a basic or acidic moiety by a conventional chemical method. In general, the salt of the continuation of the test compound is prepared by ion exchange chromatography or by reacting the free test with a stoichiometric amount or with an inorganic H organic acid which forms a desired salt in a suitable solvent or various solvent combinations. Salts of such acidic compounds can be formed by reaction with a suitable inorganic or organic base. Thus, a pharmaceutically acceptable salt of the compound of the present invention includes a conventional non-salt salt of the compound of the present invention, such as by reacting an inspective compound of the present invention with an inorganic acid, an organic acid or a polymeric acid. For example, conventional non-toxic salts include those derived from inorganic acids (eg, hydrochloric acid, hydrogen acid, hydromolybdic acid, sulfuric acid, sulfurous acid, amino acid, linonic acid, sub-acid, nitric acid, and And such as from organic acids (eg, acetic acid, propionic acid, succinic acid, glycolic acid, stearic acid, lactic acid, silk acid, tartaric acid, bar acid, ascorbic acid, balmoic acid, maleic acid, By kiramic acid, phenylacetic acid, face amine-formyl hydrazine, salicylic acid, p-aminobenzenesulfonic acid, 2-ethyloxy-benzoic acid, fumaric acid, 曱 曱 酸 acid, 甲烧Acid, Ethylene, Sodium Acid, Oxalic Acid, 127700.doc -18· 200836731 Isethionethane, palmitic acid, gluconic acid, ascorbic acid, phenylacetic acid, aspartic acid, cinnamic acid, pyruvic acid, ethanesulfonate Salts prepared from acids, ethane disulfonic acids, valeric acid, trifluoroacetic acid, and the like. Examples of suitable polymeric acid salts include those derived from polymeric acids such as citric acid, carboxymethyl cellulose. Preferably, the pharmaceutically acceptable salt of the present invention comprises 1 equivalent of the compound of the formula (1) and 1, 2 or 3 equivalents of a mineral or organic acid. More specifically, the pharmaceutically acceptable salt of the present invention is a trifluoroacetate, a vapor or a toluene acid. More specifically, the pharmaceutically acceptable salt of the present invention is a trifluoroacetate or a vapor salt. In one embodiment, the salt is a trifluoroacetate salt. In another embodiment, the salt is a chloride. In another embodiment, the salt is an anthene sulfonate. When the compound of the invention is acidic, suitable "pharmaceutically acceptable salts" refers to salts prepared from pharmaceutically acceptable non-toxic bases, including inorganic bases and organic bases. Salts derived from inorganic tests include aluminum salts. , ammonium salts, strontium salts, copper salts, iron salts, ferrous salts, bell salts, magnesium salts, manganese salts, divalent manganese salts, potassium salts, sodium salts, zinc salts, and the like. Especially preferred are ammonium salts, calcium Salts, magnesium salts, potassium salts, and sodium salts. Salts derived from pharmaceutically acceptable organic non-toxic bases include primary amines, secondary amines and tertiary amines, substituted amines (including natural substituted), cyclic amines, and Salts of ion exchange resins, such as arginine, lysine, betaine, caffeine, choline, N, NL dibenzylethylenediamine, ethylamine, monoethylamine, 2-diethylaminoethanol , 2-dimethylaminoethanol, ethanolamine, diethanolamine, ethylenediamine, N-ethylmorpholine, N-ethylhexahydropyridyl, reduced glucosamine, glucosamine, histidine, hexamine Hydrabamine), isopropylamine, lysine, methyl-reducing glucosamine, 127700.doc -19- 200836731 Lynn, tourism, hexahydroquinone Tr, polyamine resin, procaine, guanidine, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine, dicyclohexylamine, butylamine, benzylamine, phenylbenzylamine, Salts of tromethamine, etc. The preparation of the above pharmaceutically acceptable salts and other typical pharmaceutically acceptable salts is more fully described in Berg et al. (1977), J. corp./mrm. T/zarmacezzi/ca/ Χα"〆, 66:1 -1 9 It should also be noted that since the deprotonated acidic moiety (for example, a carboxyl group) of the compound of the present invention under physiological conditions can be an anion, and the electronic charge can then be internally The protonated or alkylated basic moiety (eg, a quaternary nitrogen atom) has a cationic charge that is balanced, so the compound may be an internal salt or a zwitterion. It can be used in a method of treating a human or animal body by therapy. A compound of the present invention. The present invention provides a compound for treating or preventing a condition which can be improved by inhibiting poly(ADP-ribose) polymerase (PARP) (see, for example,
Dbcovery (2005) 4:421- 440)。 因此,本發明提供用於製造藥物之式I化合物,該藥物 可用於治療或預防可藉由抑制聚(ADP-核糖)聚合酶(PARP) 加以改善之病況。 本發明亦提供一種用於治療或預防可藉由抑制聚(ADP-核糖)聚合酶(PARP)加以改善之病況的方法,該方法包括 對有需要的患者投與有效量的式I化合物或包含式I化合物 之組合物。 本發明之PARP抑制劑可用於治療在WO 2005/082368中 127700.doc -20- 200836731 所指明的疾病。 本發明之化合物可用於治療炎症性疾病,包括由器官移 植排斥產生的病況,例如;㈣之慢性炎症性疾病,包括 關節炎、類風濕性關節炎、骨關節炎及與骨吸收增強相關 之骨疾病;炎症性腸病,例%,回腸炎、潰瘍性結腸炎、 巴田特氏症候群(Barrett’s syndr〇me)及克隆氏病(ο—、 心⑽);炎症性肺病,例^,哮喘、成人呼吸窘迫症候 群及慢性阻塞性氣道疾病;眼睛之炎症性疾病,包括角膜 營養不良、顆粒性結膜炎、盤尾絲齒病、葡萄膜炎、交感 性眼炎及眼内1 ;齒齦之慢性炎症性疾病,包括齒齦炎及 牙周炎;肺結核,·麻風病;腎臟之炎症性疾病,包括尿毒 癥性併發症、腎小球腎炎及腎病;皮膚之炎症性疾病,包 括硬化性皮炎、乾癖及濕疹;中樞神經系統之炎症性疾 病,包括神經系統之慢性脫髓鞘病、多發性硬化、AIDS 相關之神經變性及阿兹海默氏病(Alzheimer,s disease)、傳 染性腦膜炎、腦脊髓炎、帕金森氏病(Parkins〇n,s⑴卿㈨、 亨庭頓氏病(Huntington,S disease)、肌萎縮性側索硬化及 病毒性或自身免疫腦炎;糖尿病併發症,包括但不限於免 疫複a體血管炎、全身性紅斑狼瘡(sle);心臟之炎症性 疾病,例如,心肌病、局部缺血性心臟病、高膽固醇血症 及動脈粥樣硬化;以及各種可能具有明顯炎症性組成之其 他疾病’包括先兆子癇、慢性肝臟衰竭、腦和脊髓創傷及 夕器B功能障礙症候群(MODS)(多器官衰竭(m〇f))。該炎 症性疾病亦可為個體之全身性炎症,例如,革蘭氏陽性或 127700.doc 200836731 革蘭氏陰性休克、出血性或過敏性休克、或由癌症化學仏 療對促炎細胞因子反應引發的休克,例如,與促炎細2 子相關之休克。此休克可能(例如)由作為癌症治療: 化學治療劑引發。 “> 因此’本發明提供—種用於製造用於治療或預防炎症性 疾病之藥物的式I化合物。 本發明亦提供一種用於治療或預防炎症性疾病之方法, 胃方法包括對有需要的患者投與有效量的式ί化合物或包 含式I化合物之組合物。 本發明之化合物亦可用於治療或預防由自然發生事件及 在手術操作過程中產生的再灌注損傷,例如,腸再灌注損 傷;心肌再灌注損傷;由心肺分流術、主動脈瘤修復術、 頸動脈動脈内膜切除術或出血性休克產生的再灌注損傷; 及由諸如心臟、肺、肝臟、腎臟、胰腺、腸及角膜等器官 之移植產生的再氧化損傷。 〇 因此’本發明提供—種用於製造用於治療或預防再灌注 損傷之藥物的式I化合物。 ▲本發明亦提供一種用&治療或預防再灌注損傷之方法, 4方法包括對有需要的患者投與有效量的式Ζ化合物或包 含式I化合物之組合物。 /本發明之化合物亦可用於治療或預防缺血性病症,包括 彼等由器官移植產生者,例如,穩定狹心症、不穩定狭心 血 局°卩缺血、肝臟局部缺血、腸系膜動脈局部缺 局。卩缺血、嚴重肢體局部缺血、慢性嚴重肢體局部 127700.doc -22- 200836731 缺血、大腦局部缺血、a ^ 心、性心臟局部缺血、局部 病、局部缺血性肝症 、性月 / 、局部缺血性視網膜病症、敗血性伙 克、及中樞神經系絲+ p 丨生休 ......之局部缺血性疾病,例如,中風$ 士 腦局部缺血。 甲風或大 广广i ^明提供~種用於製造用於治療或預防缺血性 病症之樂物的式I化合物。 本發明亦提供-種用於治療或預防缺血性病症之方法,Dbcovery (2005) 4:421-440). Accordingly, the present invention provides a compound of formula I for use in the manufacture of a medicament for the treatment or prevention of a condition which can be ameliorated by inhibition of poly(ADP-ribose) polymerase (PARP). The invention also provides a method for treating or preventing a condition ameliorated by inhibition of poly(ADP-ribose) polymerase (PARP), the method comprising administering to a patient in need thereof an effective amount of a compound of formula I or comprising A composition of a compound of formula I. The PARP inhibitors of the invention are useful in the treatment of the diseases indicated in 127700.doc -20-200836731 in WO 2005/082368. The compounds of the present invention are useful for the treatment of inflammatory diseases, including those caused by rejection of organ transplants, for example; (d) chronic inflammatory diseases including arthritis, rheumatoid arthritis, osteoarthritis and bones associated with enhanced bone resorption Disease; inflammatory bowel disease, %, ileitis, ulcerative colitis, Barrett's syndr〇me and Crohn's disease (ο-, heart (10)); inflammatory lung disease, case ^, asthma, Adult respiratory distress syndrome and chronic obstructive airway disease; inflammatory diseases of the eye, including corneal dystrophy, granular conjunctivitis, onchocerciasis, uveitis, sympathetic ophthalmia and intraocular lens; chronic inflammation of the gums Diseases, including gingivitis and periodontitis; tuberculosis, leprosy; inflammatory diseases of the kidneys, including uremic complications, glomerulonephritis and kidney disease; inflammatory diseases of the skin, including sclerosing dermatitis, cognac and Eczema; inflammatory diseases of the central nervous system, including chronic demyelinating diseases of the nervous system, multiple sclerosis, AIDS-related neurodegeneration, and Alzheimer's disease Disease (Alzheimer, s disease), infectious meningitis, encephalomyelitis, Parkinson's disease (Parkins〇n, s(1) Qing (9), Huntington's disease (Huntington, S disease), amyotrophic lateral sclerosis and virus Sexual or autoimmune encephalitis; diabetic complications, including but not limited to immune vasculitis, systemic lupus erythematosus (sle); inflammatory diseases of the heart, for example, cardiomyopathy, ischemic heart disease, high cholesterol Hypertension and atherosclerosis; and various other diseases that may have significant inflammatory components' including pre-eclampsia, chronic liver failure, brain and spinal cord trauma, and cerebral dysfunction syndrome (MODS) (multiple organ failure (m〇f) The inflammatory disease may also be a systemic inflammation of the individual, for example, Gram-positive or 127700.doc 200836731 Gram-negative shock, hemorrhagic or anaphylactic shock, or chemo-inflammatory therapy by cancer chemotherapy Shock induced by a factoric reaction, for example, shock associated with proinflammatory fines. This shock may be caused, for example, by treatment as a cancer: a chemotherapeutic agent. "> A compound of formula I for use in the manufacture of a medicament for the treatment or prevention of an inflammatory disease. The invention also provides a method for the treatment or prevention of an inflammatory disease, the method comprising administering an effective amount to a patient in need thereof A compound of the formula or a composition comprising a compound of formula I. The compounds of the invention may also be used to treat or prevent reperfusion injury caused by naturally occurring events and during surgical procedures, for example, intestinal reperfusion injury; myocardial reperfusion injury; Reperfusion injury from cardiopulmonary bypass, aortic aneurysm repair, carotid endarterectomy or hemorrhagic shock; and transplantation from organs such as heart, lung, liver, kidney, pancreas, intestine, and cornea Reoxidation damage. 〇 Therefore, the present invention provides a compound of formula I for use in the manufacture of a medicament for the treatment or prevention of reperfusion injury. ▲ The invention also provides a method of treating or preventing reperfusion injury with & 4 methods comprising administering to a patient in need thereof an effective amount of a compound of formula or a composition comprising a compound of formula I. / The compounds of the invention may also be used for the treatment or prevention of ischemic conditions, including those produced by organ transplants, for example, stable angina, unstable narrow blood vessels, ischemia, liver ischemia, local mesenteric artery Missing.卩 Ischemia, severe limb ischemia, chronic severe limbs 127700.doc -22- 200836731 ischemia, cerebral ischemia, a ^ heart, ischemic heart disease, local disease, ischemic liver disease, sex Months /, ischemic retinal disorders, septic gram, and central nervous system filaments + p sputum sputum ..., ischemic disease, for example, stroke $ cerebral ischemia. A wind or a broad formula provides a compound of formula I for use in the manufacture of a medicament for the treatment or prevention of an ischemic condition. The invention also provides a method for treating or preventing an ischemic condition,
ϋ 該方法包括對有需要的患者投與有效量的式!化合物或包 含式I化合物之組合物。 本發明提供一種用於製造用於治療或預防中風之藥物的 式I化合物。 本發明亦提供一種用於治療或預防中風之方法,該方法 包括對有需要的患者投與有效量的式〗化合物或包含式以匕 合物之組合物。 本發明之化合物亦可用於治療或預防慢性或急性腎衰 竭0 因此’本發明提供一種用於製造用於治療或預防腎衰竭 之藥物的式I化合物。 本發明亦提供一種用於治療或預防腎衰竭之方法,該方 法包括對有需要的患者投與有效量的式I化合物或包含式I 化合物之組合物。 本發明之化合物亦可用於治療或預防除心血管疾病外之 血管疾病,例如,外周動脈閉塞、閉塞性血栓血管炎、雷 諾氏疾病(Reynaud’s disease)及現象、手足發纟甘、紅斑性肢 127700.doc -23- 200836731 痛病、靜脈血栓症、靜脈曲張、動靜脈瘻、淋巴水腫及脂 肪水腫。 因此,本發明提供一種用於製造用於治療或預防除心血 管疾病外之血管疾病之藥物的式合物。 本發明亦提供一種用於治療或預防除心血管疾病外之血 官疾病之方法,該方法包括對有需要的患者投與有效量的 式I化合物或包含式I化合物之組合物。 本發明之化合物亦可用於治療或預防心血管疾病,例 如,忮性心臟衰竭、動脈粥樣硬化、充血性心臟衰竭、循 環性休克、心肌病、心臟移植、心肌梗塞、及心律不整, 例如,心房纖維性顫動、室上性心動過速、心房撲動、及 陣發性房性心動過速。 口此,本發明k供一種用於製造用於治療或預防心血管 疾病之藥物的式I化合物。 本發明亦提供一種用於治療或預防心血管疾病之方法, 該方法包括對有需要的患者投與有效量的式〗化合物或包 含式I化合物之組合物。 本發明之化合物亦可用於治療或預防糖尿病,包括工型 糖尿病(胰島素依賴性糖尿病)、„型糖尿病(非胰島素依賴 性糖尿病)、妊娠糖尿病、自身免疫型糖尿病、胰島素 病、由胰臟疾病引起的糖尿病、與其他内分泌疾病相關之 糖尿病(例如,庫欣氏症候群(Cushing,s Syndr〇me)、肢端 肥大症、嗜鉻細胞瘤、胰高血糖素瘤、原發性駿固嗣增多 症或生長抑素瘤)、A型胰島素抵抗症候群、b型胰島素抵 127700.doc -24- 200836731 抗症候群、脂肪萎縮型糖尿病、 尿病。本發明之化合物亦可用…:;胞…發的糖 症例如,糖尿病性白内障、青# " (例如,微量白蛋白尿及二:=:見網膜病、腎病 經病… 退仃性糖尿病性腎病)、多發性神 矿丙足錢疽、動脈粥樣硬化性管狀 :疾:、非㈣血糖症性他昏迷、單發性神:: 二、自律神經錢、足部潰瘍、_節問題、及皮膚或黏膜ϋ The method comprises administering to a patient in need thereof an effective amount of a compound of the formula or a composition comprising a compound of formula I. The present invention provides a compound of formula I for use in the manufacture of a medicament for the treatment or prevention of stroke. The invention also provides a method for treating or preventing stroke, the method comprising administering to a patient in need thereof an effective amount of a compound of the formula or a composition comprising the compound. The compounds of the present invention are also useful for treating or preventing chronic or acute renal failure. Thus, the present invention provides a compound of formula I for use in the manufacture of a medicament for the treatment or prevention of renal failure. The invention also provides a method for treating or preventing renal failure, the method comprising administering to a patient in need thereof an effective amount of a compound of formula I or a composition comprising a compound of formula I. The compounds of the present invention are also useful for treating or preventing vascular diseases other than cardiovascular diseases, for example, peripheral arterial occlusion, occlusive thromboangiitis, Reynaud's disease and phenomenon, hand and foot rash, erythema limb 127700 .doc -23- 200836731 Pain, venous thrombosis, varicose veins, arteriovenous fistula, lymphedema and fat edema. Accordingly, the present invention provides a formula for the manufacture of a medicament for treating or preventing a vascular disease other than a cardiovascular disease. The invention also provides a method for treating or preventing a blood disorder other than a cardiovascular disease, the method comprising administering to a patient in need thereof an effective amount of a compound of formula I or a composition comprising a compound of formula I. The compounds of the present invention are also useful for treating or preventing cardiovascular diseases such as spastic heart failure, atherosclerosis, congestive heart failure, circulatory shock, cardiomyopathy, heart transplantation, myocardial infarction, and arrhythmia, for example, Atrial fibrillation, supraventricular tachycardia, atrial flutter, and paroxysmal atrial tachycardia. Thus, the present invention k provides a compound of formula I for use in the manufacture of a medicament for the treatment or prevention of cardiovascular diseases. The invention also provides a method for treating or preventing a cardiovascular disease, the method comprising administering to a patient in need thereof an effective amount of a compound of formula or a composition comprising a compound of formula I. The compounds of the present invention are also useful for treating or preventing diabetes, including work type diabetes (insulin dependent diabetes), „type diabetes (non-insulin dependent diabetes), gestational diabetes, autoimmune diabetes, insulin disease, caused by pancreatic diseases Diabetes, diabetes associated with other endocrine diseases (eg, Cushing, s Syndr〇me, acromegaly, pheochromocytoma, glucagonoma, primary hyperplasia) Or somatostatin), type A insulin resistance syndrome, type b insulin to 127700.doc -24- 200836731 anti-symptomatic group, adipose atrophic diabetes, urinary disease. The compound of the present invention can also be used for ...:; For example, diabetic cataract, blue # " (for example, microalbuminuria and two: =: see omental disease, kidney disease, degenerative diabetic nephropathy), multiple minerals, ampoules, atherosclerosis Sclerosing tubular: disease: non-four blood sugar coma, single-shot god:: Second, autonomic nerve money, foot ulcers, _ section problems, and skin or mucous membrane
: = 感染、脛部斑點、念珠菌感染型或類脂性 展死型糖尿病性肥胖症)、高血脂症、高血壓、胰 =抵抗症候群、冠狀動脈疾病、視網膜病變、糖尿病性 ^病變、多發性神經病變、單發性神經病變、自律神經 病變、足部潰癌、關節問題、真菌感染、、細菌感染、及: 肌病症。 ,此,本發明提供一種用於製造用於治療或預防糖尿病 之藥物的式I化合物。 本發明亦提供一種用於治療或預防糖尿病之方法,該方 法包括對有需要的患者投與有效量的式i化合物或包含式1 化合物之組合物。 本發明之化合物亦可用於治療或預防癌症,包括實體 瘤’例如,纖維肉瘤、黏液肉瘤、脂肪肉瘤、軟骨肉瘤、 原丨生肉瘤、脊索瘤、血管肉瘤、内皮肉瘤、淋巴管肉 瘤、淋巴管内皮肉瘤、滑膜瘤、間皮瘤、尤因瘤(Ewing,s tumor)、平滑肌肉瘤、橫紋肌肉瘤、結腸癌、結腸直腸 癌、腎癌、胰腺癌、骨癌、乳癌、卵巢癌、前列腺癌、食 127700.doc -25- 200836731 管癌、胃癌、口癌、鼻癌、喉癌、鱗狀細胞癌、基底細胞 癌、腺癌、汗腺癌、皮脂腺癌、乳頭狀癌、乳頭狀囊腺 瘤、囊腺癌、髓質癌、支氣管原癌、腎細胞癌、肝細胞 瘤、膽官癌、絨膜癌、精原細胞瘤、胚胎性癌、維爾姆斯 瘤(Wilms’tumor)、子宮頸癌、子宮癌、睾丸癌、小細胞肺 癌、膀胱癌、肺癌、上皮癌、皮膚癌、黑色素瘤、神經母 細胞瘤及成視網膜細胞瘤;血液傳播性癌症,例如,急性 成淋巴細胞性白血病(”ALL”)、急性成淋巴細胞性B_細胞 白血病、急性成淋巴細胞T-細胞白血病、急性成髓細胞性 白血病(’ AML’’)、急性前髓細胞性白血病("apl”)、急性單 核母細胞性白血病、急性成紅細胞性白血病、急性巨核母 細胞性白血病、急性骨髓單核細胞性白血病、急性非淋巴 細胞性白血病、未分化急性白血病、慢性髓細胞白血病 ("CML”)、慢性淋巴細胞性白血病(”CLL”)、毛細胞白血病 及多發性骨髓瘤;急性和慢性白血病,例如,成淋巴細胞 性、骨髓性、淋巴細胞性、髓細胞性白血病;淋巴瘤,例 如’何傑金氏病(Hodgkin’s disease)、非何傑金氏(N〇n_ Hodgkin’s)淋巴瘤、多發性骨髓瘤' 瓦爾登斯特倫氏巨球 蛋白血症(Waldenstrom’s macroglobulinemia)、重鏈病及真 性紅細胞增多症;CNS及腦癌,例如,神經膠質瘤、纖維 狀細胞性星形細胞瘤 '星形細胞瘤、多形性成膠質細胞 瘤、多形性膠質母細胞瘤、髓母細胞瘤、顱咽管瘤、室管 膜瘤、松果體瘤、血管母細胞瘤、聽神經瘤、少突神經膠 質瘤、腦膜瘤、前庭神經勒瘤、腺瘤、轉移性腦瘤、腦膜 127700.doc -26- 200836731 瘤、脊髓瘤及髓母細胞瘤。 因此,本發明提供一種用於製造用於治療或預防癌症之 藥物的式I化合物。 本發明亦提供一種用於治療或預防癌症之方法,該方法 包括對有需要的患者投與有效量的式I化合物或包含式I化 ^ 合物之組合物。: = infection, ankle spot, candida infection or lipid-induced obesity obesity), hyperlipidemia, hypertension, pancreas = resistance syndrome, coronary artery disease, retinopathy, diabetic lesions, multiple Neuropathy, single neuropathy, autonomic neuropathy, foot ulceration, joint problems, fungal infections, bacterial infections, and: muscle disorders. Thus, the present invention provides a compound of formula I for use in the manufacture of a medicament for the treatment or prevention of diabetes. The invention also provides a method for treating or preventing diabetes comprising administering to a patient in need thereof an effective amount of a compound of formula i or a composition comprising a compound of formula 1. The compounds of the invention may also be used in the treatment or prevention of cancer, including solid tumors 'eg, fibrosarcoma, mucinous sarcoma, liposarcoma, chondrosarcoma, protozoa sarcoma, chordoma, angiosarcoma, endothelial sarcoma, lymphangiosarcoma, lymphatic vessels Sarcoma, synovial tumor, mesothelioma, Ewing, s tumor, leiomyosarcoma, rhabdomyosarcoma, colon cancer, colorectal cancer, kidney cancer, pancreatic cancer, bone cancer, breast cancer, ovarian cancer, prostate cancer 127700.doc -25- 200836731 Tuberculosis, gastric cancer, oral cancer, nasal cancer, laryngeal cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland cancer, sebaceous gland cancer, papillary carcinoma, papillary cystadenoma , cystadenocarcinoma, medullary carcinoma, bronchial carcinoma, renal cell carcinoma, hepatoma, biliary cancer, choriocarcinoma, seminoma, embryonal carcinoma, Wilms'tumor, cervix Cancer, uterine cancer, testicular cancer, small cell lung cancer, bladder cancer, lung cancer, epithelial cancer, skin cancer, melanoma, neuroblastoma and retinoblastoma; blood-borne cancer, for example, acute drenching Cellular leukemia ("ALL"), acute lymphoblastic B-cell leukemia, acute lymphoblastic T-cell leukemia, acute myeloblastic leukemia ('AML''), acute promyelocytic leukemia (" Apl"), acute mononuclear leukemia, acute erythroblastic leukemia, acute megakaryoblastic leukemia, acute myelomonocytic leukemia, acute non-lymphocytic leukemia, undifferentiated acute leukemia, chronic myeloid leukemia "CML"), chronic lymphocytic leukemia ("CLL"), hairy cell leukemia and multiple myeloma; acute and chronic leukemia, for example, lymphoblastic, myeloid, lymphocytic, myeloid leukemia; Lymphoma, such as 'Hodgkin's disease, non-Hodgkin's lymphoma, multiple myeloma' Waldenstrom's macroglobulinemia, Heavy chain disease and polycythemia vera; CNS and brain cancer, for example, glioma, fibrillar cell Astrocytoma, astrocytoma, glioblastoma multiforme, glioblastoma multiforme, medulloblastoma, craniopharyngioma, ependymoma, pineal tumor, hemangioblastoma, Acoustic neuroma, oligodendroglioma, meningioma, vestibular neuroma, adenoma, metastatic brain tumor, meninges 127700.doc -26- 200836731 Tumor, myeloma and medulloblastoma. Accordingly, the present invention provides a compound of formula I for use in the manufacture of a medicament for the treatment or prevention of cancer. The invention also provides a method for treating or preventing cancer, the method comprising administering to a patient in need thereof an effective amount of a compound of formula I or a composition comprising a compound of formula I.
• 本發明之化合物亦可用於治療同源重組(HR)依賴性DNA DSB修復活性有缺陷之癌症(參見WO 2006/021801)。 ^ HR依賴性DNA DSB修復途徑可藉由同源機制以改造連 續DNA螺旋體來修復DNA雙鏈斷裂(DSB)(#ai. Genet· (2001) 27(3):247-254)。HR 依賴性 DNA DSB 修復途徑之組 成包括但不限於 ATM (NM_000051)、RAD51 (NM- 002875) 、RAD51 LI (NM-002877) > RAD51 C (NM- 002876) 、RAD51L3 (NM-002878)、DMC1 (NM-007068)、 XRCC2 (NM700543 1)、XRCC3 (NM-005432)、RAD52 (NM-002879)、RAD54L (NM-003579)、RAD54B (NM-012415)、BRCA-1 (NM-007295)、BRCA-2(NM-000059)、 RAD50 (NM-005732)、MREI ΙΑ (NM-005590)、NBS1 (NM-002485)、ADPRT (PARP-1)、ADPRTL2、(PARP02) CTPS、RPA、RPA1、RPA2、RPA3、XPD、ERCC1、 XPF、MMS19、RAD51、RAD51p、RAD51C、RAD51D、 DMC1、XRCCR、XRCC3、BRCA1、BRCA2、RAD52、 RAD54、RAD50、MRE11、NB51、WRN、BLMKU70、 RU80、ATM、ATRCHK1、CHK2、FANCA、FANCB、 127700.doc -27- 200836731 FANCC、FANCD1 、FANCD2、FANCE、FANCF、 FANCG、FANCC、FANCD1 、FANCD2、FANCE、 FANCF、FANCG、RADI 及 RAD9。HR 依賴性 DNA DSB 修 復途徑所涉及其他蛋白包括諸如EMSY等調節因子(Ce// (2003) 115:523-535) ° HR依賴性DNA DSB修復有缺陷之癌症可包括一種或多 種癌細胞或由一種或多種癌細胞構成,相對於正常細胞, 該等癌細胞修復DNA DSB之能力降低或消失,即,HR依 賴性DNA DSB修復途徑之活性在該一種或多種癌細胞中降 低或消失。 該HR依賴性DNA DSB修復途徑之一種或多種組成的活 性在具有HR依賴性DNA DSB修復有缺陷之癌症之個體的 一種或多種癌細胞中可能消失。HR依賴性DNA DSB修復 途徑之組成在此項技術中受到良好表徵(參見,例如, (2001) 291:1284 -1289)且包括上文所列示組成。 本發明提供一種用於製造藥物的式I化合物,該藥物用 於治療或預防HR依賴性DNA DSB修復活性有缺陷之癌 症。 本發明亦提供一種用於治療或預防HR依賴性DNA DSB 修復活性有缺陷之癌症之方法,該方法包括對有需要的患 者投與有效量的式I化合物或包含式I化合物之組合物。 在一實施例中,該等癌細胞一個或多個選自下列之表型 的HR依賴性DNA DSB修復活性有缺陷:ATM (NM-000051) 、 RAD51 (NM-002875) 、 RAD51 LI (NM- 127700.doc -28- 200836731 002877) 、RAD51 C (NM-002876)、RAD51L3 (NM- 002878) 、DMC1 (NM-007068)、XRCC2 (NM7005431) > XRCC3 (NM-005432)、RAD52 (NM-002879)、RAD54L (NM-003579)、RAD54B (NM-012415)、BRCA-1 (NM-007295)、BRCA-2 (NM-000059)、RAD50 (NM-005732)、 MREI ΙΑ (NM-005590)、NBS1 (NM-002485) )、ADPRT (PARP-1)、ADPRTL2、(PARP02) CTPS、RPA、RPA1、 RPA2、RPA3、XPD、ERCC1、XPF、MMS19、RAD51、 RAD51p、RAD51C、RAD51D、DMC1 、XRCCR、 XRCC3、BRCA1、BRCA2、RAD52、RAD54、RAD50、 MRE11、NB51、WRN、BLMKU70、RU80、ATM、 ATRCHK1 、CHK2、FANCA、FANCB 、FANCC、 FANCD1 、 FANCD2 、 FANCE 、 FANCF 、 FANCG 、 FANCC、FANCD1 、FANCD2、FANCE、FANCF、 FANCG、RADI 及 RAD9。 在另一實施例中,該等癌細胞具有BRCA1及/或BRCA2 缺陷之表型。此表型之癌細胞在BRCA1及/或BRCA2中可 能有缺陷,即,BRCA1及/或BRCA2在該等癌細胞中之表 現及/或活性可減少或消失,例如,藉助編碼核酸之突變 或多肽現象或藉助編碼調節因子之基因(例如,編碼 BRCA2調節因子之EMSY基因)之擴增、突變或多肽現象 (Ce// (2003) 115:523-535)。 已知BRCA-1及BRCA-2係腫瘤抑制基因,其野生型等位 基因通常在雜合載體之腫瘤中丟失(2002) 127700.doc -29- 200836731 21(58):8981-93; Trends Mol Med., (2002) 8(12):571-6)。 BRCA-1及/或BRCA-2突變體與乳癌之關係已經良好表徵 (五xp C///7 (2002) 21 5吵/;/):9-12)。亦已知 編碼BRCA-2結合因子之EMSY基因的擴增與乳癌及卵巢癌 相關。BRCA-1及/或BRCA-2突變之載體亦處於卵巢癌、前 列腺癌及胰腺癌之高風險下。BRC A-1及BRC Α·2變化之檢 測為此項技術所熟知且闡述於(例如)歐洲專利ΕΡ 699 754、歐洲專利 ΕΡ 705 903、GeM,. Γαί (1992) 1:75-83 ; Cancer Treat Res (2002) 107:29-59 ; Neoplasm (2003) 50(4):246-50 ; CeAa (2003) 68(1): 1 1 -16)中。 BRCA-2結合因子EMSY擴增之測定闡述於Ce// 115:523-535中。已經證明PARP抑制劑可用於特異性地殺傷BRCA-1 及 BRCA-2 缺陷之腫瘤(2005) 434:913-916 及 917-920)。 因此,本發明提供一種用於製造藥物之式I化合物,該 藥物用於治療或預防BRC A-1或BRC A-2缺陷之腫瘤。 本發明亦提供一種用於治療或預防BRCA-1或BRCA-2缺 陷之腫瘤之方法,該方法包括對有需要的患者投與有效量 的式I化合物或包含式I化合物之組合物。 在一實施例中,本發明之PARP抑制劑可用於預防性治 療以消除BRCA2缺陷之細胞(參見,(2005) 65:10145)。 本發明之化合物可用於治療或預防神經退化性疾病,包 括多聚穀胺醯胺擴充相關性神經變性、亨庭頓氏病 127700.doc -30- 200836731 (Huntington s disease)、肯尼迪氏病(Kennedy’s disease)、 脊髓小腦共濟失調、齒狀核紅核蒼白球丘腦下部核萎縮 (DRPLA)、蛋白聚集相關性神經變性、馬查多_約瑟夫病 (Machado-Joseph’s disease)、阿茲海默氏病(Alzheimer,s disease)、帕金森氏病、肌萎縮性側索硬化、海綿狀腦 病、朊病毒相關性疾病及多發性硬化(MS)。 因此,本發明提供一種用於製造用於治療或預防神經退 化性疾病之藥物的式〗化合物。• The compounds of the invention may also be used to treat cancers that are defective in homologous recombination (HR)-dependent DNA DSB repair activity (see WO 2006/021801). ^ HR-dependent DNA DSB repair pathways can repair DNA double-strand breaks (DSBs) by homologous mechanisms to engineer continuous DNA spirochetes (#ai. Genet. (2001) 27(3): 247-254). The components of the HR-dependent DNA DSB repair pathway include, but are not limited to, ATM (NM_000051), RAD51 (NM-002875), RAD51 LI (NM-002877) > RAD51 C (NM-002876), RAD51L3 (NM-002878), DMC1 (NM-007068), XRCC2 (NM700543 1), XRCC3 (NM-005432), RAD52 (NM-002879), RAD54L (NM-003579), RAD54B (NM-012415), BRCA-1 (NM-007295), BRCA -2 (NM-000059), RAD50 (NM-005732), MREI ΙΑ (NM-005590), NBS1 (NM-002485), ADPRT (PARP-1), ADPRTL2, (PARP02) CTPS, RPA, RPA1, RPA2 RPA3, XPD, ERCC1, XPF, MMS19, RAD51, RAD51p, RAD51C, RAD51D, DMC1, XRCCR, XRCC3, BRCA1, BRCA2, RAD52, RAD54, RAD50, MRE11, NB51, WRN, BLMKU70, RU80, ATM, ATRCHK1, CHK2 FANCA, FANCB, 127700.doc -27- 200836731 FANCC, FANCD1, FANCD2, FANCE, FANCF, FANCG, FANCC, FANCD1, FANCD2, FANCE, FANCF, FANCG, RADI and RAD9. Other proteins involved in the HR-dependent DNA DSB repair pathway include regulatory factors such as EMSY (Ce// (2003) 115:523-535) ° HR-dependent DNA DSB repair defective cancer may include one or more cancer cells or One or more cancer cells constitute a reduced or absent ability of the cancer cells to repair DNA DSB relative to normal cells, i.e., the activity of the HR dependent DNA DSB repair pathway is reduced or eliminated in the one or more cancer cells. The activity of one or more of the HR-dependent DNA DSB repair pathways may be lost in one or more cancer cells of an individual having a HR-dependent DNA DSB repair defective cancer. The composition of the HR dependent DNA DSB repair pathway is well characterized in the art (see, for example, (2001) 291:1284-1289) and includes the compositions listed above. The present invention provides a compound of formula I for use in the manufacture of a medicament for the treatment or prevention of a cancer which is defective in HR-dependent DNA DSB repair activity. The invention also provides a method for treating or preventing a cancer which is defective in HR-dependent DNA DSB repair activity, the method comprising administering to a patient in need thereof an effective amount of a compound of formula I or a composition comprising a compound of formula I. In one embodiment, one or more of the cancer cells have one or more HR-dependent DNA DSB repair activities selected from the group consisting of: ATM (NM-000051), RAD51 (NM-002875), RAD51 LI (NM- 127700.doc -28- 200836731 002877) , RAD51 C (NM-002876), RAD51L3 (NM- 002878), DMC1 (NM-007068), XRCC2 (NM7005431) > XRCC3 (NM-005432), RAD52 (NM-002879 ), RAD54L (NM-003579), RAD54B (NM-012415), BRCA-1 (NM-007295), BRCA-2 (NM-000059), RAD50 (NM-005732), MREI ΙΑ (NM-005590), NBS1 (NM-002485) ), ADPRT (PARP-1), ADPRTL2, (PARP02) CTPS, RPA, RPA1, RPA2, RPA3, XPD, ERCC1, XPF, MMS19, RAD51, RAD51p, RAD51C, RAD51D, DMC1, XRCCR, XRCC3 , BRCA1, BRCA2, RAD52, RAD54, RAD50, MRE11, NB51, WRN, BLMKU70, RU80, ATM, ATRCHK1, CHK2, FANCA, FANCB, FANCC, FANCD1, FANCD2, FANCE, FANCF, FANCG, FANCC, FANCD1, FANCD2, FANCE , FANCF, FANCG, RADI and RAD9. In another embodiment, the cancer cells have a phenotype of BRCA1 and/or BRCA2 deficiency. Cancer cells of this phenotype may be defective in BRCA1 and/or BRCA2, ie, the expression and/or activity of BRCA1 and/or BRCA2 in such cancer cells may be reduced or eliminated, for example, by means of a mutation or polypeptide encoding a nucleic acid Amplification or amplification, mutation or polypeptide phenomenon by means of a gene encoding a regulatory factor (eg, an EMSY gene encoding a BRCA2 regulatory factor) (Ce// (2003) 115: 523-535). The BRCA-1 and BRCA-2 tumor suppressor genes are known, and their wild-type alleles are usually lost in tumors of hybrid vectors (2002) 127700.doc -29- 200836731 21(58):8981-93; Trends Mol Med., (2002) 8(12):571-6). The relationship between BRCA-1 and/or BRCA-2 mutants and breast cancer has been well characterized (five xp C///7 (2002) 21 5 noisy/;/): 9-12). Amplification of the EMSY gene encoding the BRCA-2 binding factor is also known to be associated with breast and ovarian cancer. The vector of BRCA-1 and/or BRCA-2 mutations is also at high risk for ovarian cancer, prostate cancer and pancreatic cancer. The detection of BRC A-1 and BRC Α·2 changes is well known in the art and is described, for example, in European Patent 699 699 754, European Patent 705 705 903, GeM,. Γαί (1992) 1:75-83; Treat Res (2002) 107:29-59; Neoplasm (2003) 50(4): 246-50; CeAa (2003) 68(1): 1 1 -16). The assay for BRCA-2 binding factor EMSY amplification is set forth in Ce// 115:523-535. PARP inhibitors have been shown to be useful for specifically killing BRCA-1 and BRCA-2 deficient tumors (2005) 434: 913-916 and 917-920). Accordingly, the present invention provides a compound of formula I for use in the manufacture of a medicament for the treatment or prevention of a tumor deficient in BRC A-1 or BRC A-2. The invention also provides a method for treating or preventing a tumor of a BRCA-1 or BRCA-2 deficiency, the method comprising administering to a patient in need thereof an effective amount of a compound of formula I or a composition comprising a compound of formula I. In one embodiment, the PARP inhibitors of the invention are useful for prophylactic treatment to eliminate BRCA2-deficient cells (see, (2005) 65: 10145). The compounds of the present invention are useful for the treatment or prevention of neurodegenerative diseases, including polyglutamine-expanding-related neurodegeneration, Huntington's disease 127700.doc -30-200836731 (Huntington s disease), Kennedy's disease (Kennedy's disease) Disease), spinal cord cerebellar ataxia, dentate nucleus red nucleus globus pallidus hypothalamic nuclear atrophy (DRPLA), protein aggregation-related neurodegeneration, Machado-Joseph's disease, Alzheimer's disease (Alzheimer, s disease), Parkinson's disease, amyotrophic lateral sclerosis, spongiform encephalopathy, prion-related diseases, and multiple sclerosis (MS). Accordingly, the present invention provides a compound of the formula for use in the manufacture of a medicament for the treatment or prevention of a neurodegenerative disease.
本發明亦提供一種用於治療或預防神經退化性疾病之方 法,該方法包括對有需要的患者投與有效量的式〗化合物 或包含式I化合物之組合物。 本發明之化合物亦可用於治療或預防逆轉錄病毒感染 (美國專利US 5652260)、視網膜損害(CMrr及认 (20〇4)’ 29·403)、皮膚老化及uv引發之皮膚損害(美國專利 US 5589483 及 (2〇〇2) 。 本發明之化合物可用於治療或預防過早衰老及推遲年齡 相關性細胞功能障礙出現(/>w崎㈣—細㈣ (2005) 52:93-99) 〇 很蘇標準醫藥實務,本發 ^ 干判兴晉樂上 :!之_、賦形劑、稀釋劑、佐劑、填充劑、緩衝劑、 無知投藥途彳_受, 身/外周方式還是在期望作用點投與,包括但 127700.doc •31· 200836731 於、.二口(例如’藉由攝取);局部(包括(例如)經皮、經 鼻内/經眼、經口腔及經舌下);肺部(例如,藉由使用(例 、)氣岭膠通過(例如)嘴或鼻吸入或吹入療法);直腸;陰 ^非’、、二腸(例如,藉由注射,包括皮下、皮内、肌内、 靜脈内動脈内、心臟内、鞘内、脊柱内、莢膜内、囊 下眼窩内、腹膜腔内、氣管内、表皮下、關節内、蛛網 膜下及胸骨内注射);及藉由植入儲積物(例如,經皮下或 肌内)。The invention also provides a method for treating or preventing a neurodegenerative disease, the method comprising administering to a patient in need thereof an effective amount of a compound of the formula or a composition comprising a compound of formula I. The compounds of the present invention are also useful for treating or preventing retroviral infection (US Pat. No. 5,652,260), retinal damage (CMrr and recognition (20〇4) '29·403), skin aging and uv-induced skin damage (US Patent US) 5589483 and (2〇〇2). The compounds of the present invention are useful for treating or preventing premature aging and delaying the onset of age-related cell dysfunction (/>waki (4)-fine (4) (2005) 52:93-99) Very standard medical practice, this hair ^ dry judgments on the music:! _, excipients, thinners, adjuvants, fillers, buffers, ignorant drug delivery _ _, body / peripheral way is still in anticipation The point of action is administered, including but 127700.doc • 31· 200836731 in , two (eg 'by ingestion'); local (including, for example, percutaneous, intranasal/transocular, transoral and sublingual) The lungs (for example, by using (eg,) urinary gels through, for example, mouth or nose inhalation or insufflation therapy; rectum; vaginal, non-, and second intestines (eg, by injection, including subcutaneous, Intradermal, intramuscular, intravascular, intracardiac, intrathecal, intraspinal The capsule, subcapsular intraorbital, intraperitoneal, intratracheal, subepidermal, intraarticular, subarachnoid, and intrasternal injection); and by depot implantation (e.g., subcutaneously or intramuscularly).
文武者可為真核生物、動物、脊椎動物、哺乳動物、發 齒類動物(例如’天竺鼠、倉鼠、大鼠、小鼠)、鼠科動物 (^列如,小氣)、犬科動物(例如,狗)、貓科動物(例如, 書田)馬科動物(例如,馬)、靈長類、猿類(例如,猴子或 猿)、猴科動物(例如,絨猴、狒狒)、猿類(例如,大猩 猩、黑猩猩、猩猩、長臂猿)或人類。 本發明亦提供包含-種或多種包含本發明化合物及醫藥 上可接受之載劑的醫藥組合物。包含活性成份之醫藥組合 物可呈適於經口使用之形式’例如,錠劑、片劑、菱形 錠、水性或油性懸浮液、可分散粉劑或顆粒、乳劑、硬或 軟膠囊、或糖漿或酏劑。擬口服使用之組合物可依昭業内 已知用於製造醫藥組合物之任一方法加以製備且該'等:且人 物可包含-種或多種選自由甜味劑、矯味劑1色劑及: 腐劑組成之群的試劑以提供醫藥上美觀且可口之势為丨。錠 劑包含該活性成份與適於製造錠劑且在醫藥上可接1之益 毒賦形劑的混合物。舉例而言’此等賦形劑可為:二生稀 127700.doc -32 - 200836731 釋劑,例如,碳酸鈣、碳酸鈉、乳糖、鱗酸鈣或構酸鈉; 造粒劑及崩解劑,例如,微晶纖維素、交聯羧甲基纖維素 鈉、玉米澱粉或藻酸;黏結劑,例如,澱粉、明膠、聚乙 烯σ比洛唆酮或阿拉伯膠;及潤滑劑,例如,硬脂酸鎂、硬 脂酸或滑石粉。該等錠劑可無包膜,或其可藉由已知技術 包膜以掩蔽令人不愉快之味道或延遲在胃腸道中之崩解及 吸收’並藉此提供長期持續作用。舉例而言,可使用水溶 性味道掩蔽材料(例如,羥丙基-甲基纖維素或羥丙基纖維 素)或時間延遲材料(例如,乙基纖維素、乙酸丁酸纖維 素)。 用於口服使用之調配物亦可作為硬明膠膠囊存在,其中 該活性成份與惰性固體稀釋劑(例如,碳酸鈣、磷酸鈣或 高嶺土)混合;或其可為軟明膠膠囊,其中該活性成份與 水溶性載劑(例如,聚乙二醇)或油性媒介(例如,花生油、 液體石壤或橄欖油)混合。 水性懸料包含該活性材料與適於製備水性t浮液之賦 形劑混合物。此等賦形劑係懸浮劑’例如,羧甲基纖維素 納、甲基纖維素、經丙基甲基-纖維素、藻酸納、聚乙稀 :咯啶_、黃蓍膠、黃原膠及阿拉伯樹膠;分散劑或潤濕 ::為天然磷脂(例如’卵磷脂)、或環氧烷與脂肪酸之縮 (]如&氧乙歸硬脂酸s旨)、或環氧乙烧與長鏈脂 ==合產物(例如,十七伸乙氧基録_)、或環氧 如:TL自脂肪酸與己糖醇之部分酿的縮合產物(例 烯山梨糖醇單油酸酯)、或環氧乙烷與衍生自 127700.doc -33- 200836731 脂肪酸與己糖醇軒之部分醋的縮合產物(例如,聚氧乙稀 山梨醇肝單油酸醋)。該等水性懸浮液亦可包含一種或多 種防腐劑,例如,對羥基苯f酸乙酯或對羥基苯甲酸正丙 酿;:種或多種著色劑、-種或多種矯味劑及一種或多種 諸如庶糖、糖精或阿斯巴甜(aspartame)等甜味劑。 可藉由將該活性成份懸浮於植物油(例如,花生油、撤 欖油、芝麻油或椰子油)中或懸浮於諸如液體石壌等礦物 油中來調配油性懸浮液。該等油性懸浮液可包含增稠劑, •j如蜂it硬石壞或十六燒醇。可加入甜味劑(例如, 上文所述彼等)及續味劑以提供適口口服製劑。此等组入 物可藉由添加抗氧化劑(例如,丁基化經基苯甲趟或= 育紛)來保存。 適於藉由添加水來製備水性懸浮液之可分散粉末及顆粒 可包含活性成份與分散劑或潤濕劑、懸浮劑及一種或多種 防腐劑之混合物。適宜分散劑或潤濕劑及懸浮劑係藉由彼 等已於上文中提及者來示例。亦可存在其他賦形劑,例 1味誠著色劑。此等組合物可藉由添加抗 巩化诏(例如,抗壞血酸)加以保存。 本發明之醫藥組合物亦可呈水包油乳劑形式。 植物峨如,撖欖油或花生油)或礦物油(例如 = 卿等之混合物。適宜乳化劑可為天然碟脂(例如:大 :印=及衍生自脂肪酸與己糖醇肝之醋或部分輯(例 木醇酐單油酸酸酯)及該等部分酯與環氧乙 合產物⑼如,聚氧乙烯山梨醇軒單油酸酷)。該等錢亦 127700.doc •34- 200836731 可包含甜味劑、橋味劑、防腐劑及抗氧化劑。 糖漿及馳劑可使用甜味劑(例如,甘油、丙二醇、山梨 :醇或薦糖)㈣。該等調配物亦可含有緩和劑、防腐 d、矯味劑和著色劑及抗氧化劑。 該等醫藥組合物可為盔菌注射 ^ 勹",、囷,王射水洛液形式。可採用的可 接文媒劑及溶劑有水、妖彡夂· a R S格氏(Rlnger,s)溶液及等滲氯化鈉 溶液。 該無菌注射製劑亦可為其中該活性成份溶於油相中之益 菌注射水包油微乳液。舉例而言,可首切該活性成份: 於大丑油及_磷脂之混合物中。然後將該油性溶液引入水 與甘油混合物中並處理,形成微乳液。 该等可注射溶液或微乳液可藉由局部單次注射引入患者 之^流中°或者’可有利地以維持本發明化合物之怪定循 環濃度之方式投與㈣液或微乳液。為維持此^濃度, 可使用持續靜脈内遞送裝置。此裝置之實例係二“ CADD-PLUS™ 5400型靜脈内幫浦。 該等醫藥組合物可為詩肌岐皮下投與之無g注射水 性或油性懸浮液形式。該懸浮液可根據已知技術使用彼等 已於上文提及之適宜分散劑或潤濕劑及懸浮劑加以調配。 無菌可注射㈣亦可為存於無毒非經腸可接受之稀釋劑或 溶劑中之無菌可注射溶液或懸浮液,例如,溶於丨,3-丁二 醇中之溶液。此外,習慣上採用無以揮發油作為溶劑或 懸浮介質。任何溫和不揮發油(包括合成甘油單酯或甘油 二酯)均可用於此目的。此外’在可注射製劑中可使用諸 127700.doc -35- 200836731 如油酸等脂肪酸。 對於該藥物之直腸投盥 ^ ”而s,式1化合物亦可以栓劑形 式技與。可藉由將該筚 — ^雄 μ物一適且無刺激賦形劑混合來製備 二組合物’該賦形劑在常溫下為固體但在直腸溫度下為 液體且因而在直腸中融化以釋放該藥物。此等材料包括可 可油、/油明膠、氯化植物油、各分子量之聚乙二醇與ί 乙一醇之脂肪酸酯之混合物。 對於局部使用而言,可採用含式I化合物之乳霜、軟 貧、凝膠、溶液或懸浮液等。(對於本申請案而言,局部 應用應包括漱口藥水及漱口藥。) 本發明之化合物可以鼻内形式藉由局部使用適宜鼻内媒 训及遞运4置投與’或藉由經皮途徑使用彼等熟習該項技 術者熟知的經皮皮膚貼片形式投與。當然,對於以經皮遞 送系統形式投藥而言,整個劑量方案投與劑量應為連續的 而非間歇的。本發明之化合物亦可作為检劑採用諸如下列 2基質遞送··可可油、甘油明膠、氫化植物油、各種分子 Ϊ之聚乙二醇與聚乙二醇之脂肪酸酯的混合物。 當對受試者投與本發明之化合物時,該經選擇劑量水平 應端視各種因素而定,該等因素包括但不限於具體化合物 之活性、個體症狀之嚴重性、投與途徑、投與時間、該化 合物之排泄速率、治療持續時間、該組合中所用其他藥 物、化合物及/或材料、及患者之年齡、性別、重量、狀 況、總體健康狀況及先前病史。化合物之數量及投與途徑 最終應由醫師決定,但通常該劑量在作用點應達到達成期 127700.doc •36· 200836731 望效果而不會造成實質性傷害或有害副作用之局部濃度。 活體内投與可在整個治療期間以單劑量連續或間歇方式 (例如’以適宜間隔分劑量)實施。確定最有效投與方法及 劑篁之方法已為彼等熟習該項技術者所習知且應隨用於治 療之调配物、治療目的、所治療靶細胞及所治療受試者而 變化。可根據治療醫師所選擇劑量水平及模式實施單次或 多次投與。Civil and military personnel can be eukaryotes, animals, vertebrates, mammals, hairy animals (eg 'scorpine hamsters, hamsters, rats, mice), murines (such as, stingy), canines (eg , dog), feline (eg, Shutian) equine (eg, horse), primate, scorpion (eg, monkey or lynx), simian (eg, marmoset, lynx), mites (for example, gorillas, chimpanzees, orangutans, gibbons) or humans. The invention also provides a pharmaceutical composition comprising one or more of a compound comprising a compound of the invention and a pharmaceutically acceptable carrier. The pharmaceutical composition comprising the active ingredient may be in a form suitable for oral use such as a tablet, tablet, lozenge, aqueous or oily suspension, dispersible powder or granule, emulsion, hard or soft capsule, or syrup or Tincture. The composition to be used orally can be prepared according to any method known in the art for the manufacture of a pharmaceutical composition and the same: and the person may comprise one or more selected from the group consisting of a sweetener, a flavoring agent, and : Reagents consisting of a mixture of agents to provide a medically pleasing and palatable trend. Tablets comprise a mixture of the active ingredient with a pharmaceutically acceptable excipient which is suitable for the manufacture of a tablet. For example, such excipients may be: diquat 127700.doc -32 - 200836731 release agents, for example, calcium carbonate, sodium carbonate, lactose, calcium sulphate or sodium sulphate; granulating agents and disintegrants , for example, microcrystalline cellulose, croscarmellose sodium, corn starch or alginic acid; a binder such as starch, gelatin, polyethylene σ-pirone or gum arabic; and a lubricant, for example, a hard Magnesium citrate, stearic acid or talc. The lozenges may be uncoated or they may be enveloped by known techniques to mask unpleasant taste or delay disintegration and absorption in the gastrointestinal tract and thereby provide a long lasting effect. For example, a water-soluble taste masking material (e.g., hydroxypropyl-methylcellulose or hydroxypropylcellulose) or a time delay material (e.g., ethylcellulose, cellulose acetate butyrate) can be used. Formulations for oral use may also be presented as a hard gelatin capsule, wherein the active ingredient is mixed with an inert solid diluent (for example, calcium carbonate, calcium phosphate or kaolin); or it may be a soft gelatin capsule, wherein the active ingredient A water-soluble carrier (for example, polyethylene glycol) or an oily vehicle (for example, peanut oil, liquid stone or olive oil) is mixed. The aqueous suspension comprises the active material in admixture with an excipient suitable for preparing an aqueous t-liquid. Such excipients are suspending agents', for example, sodium carboxymethylcellulose, methylcellulose, propylmethyl-cellulose, sodium alginate, polyethylene: pyridinium, xanthan gum, xanthan Gum and gum arabic; dispersant or wetting:: natural phospholipids (such as 'lecithin), or alkylene oxide and fatty acid (such as & oxyethylene stearic acid s), or epoxy a condensation product (for example, ethene monooleate) with a long-chain fat== product (for example, hepta-exetyloxy) or an epoxy such as TL from a fatty acid and a hexitol Or a condensation product of ethylene oxide with a portion of the vinegar derived from 127700.doc -33-200836731 fatty acid and hexitol (for example, polyoxyethylene sorbitan monooleate). The aqueous suspensions may also contain one or more preservatives, for example, p-hydroxyphenyl f-ethyl or p-hydroxybenzoic acid; one or more coloring agents, one or more flavoring agents, and one or more such as A sweetener such as sucrose, saccharin or aspartame. The oily suspension can be formulated by suspending the active ingredient in a vegetable oil (for example, peanut oil, eucalyptus oil, sesame oil or coconut oil) or suspended in a mineral oil such as liquid sarcophagus. These oily suspensions may contain a thickening agent, such as bee, hard stone or hexadecanol. Sweetening agents (e.g., as described above) and flavoring agents can be added to provide a palatable oral preparation. These groups of substances can be preserved by the addition of an antioxidant (for example, butylated benzomethazine or = broth). Dispersible powders and granules suitable for the preparation of aqueous suspensions by the addition of water may contain the active ingredient in admixture with dispersing or wetting agents, suspending agents and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Other excipients may also be present, such as a coloring agent. Such compositions can be preserved by the addition of anti-sclerosing sputum (e.g., ascorbic acid). The pharmaceutical compositions of the invention may also be in the form of an oil-in-water emulsion. For example, a mixture of plants such as eucalyptus oil or peanut oil or mineral oil (for example, qingqing, etc.) The suitable emulsifier can be a natural dish fat (for example: large: India = and vinegar or partial derivative derived from fatty acid and hexitol liver) (for example, wood alcohol anhydride monooleate) and the partial ester and epoxy ethylene product (9), such as polyoxyethylene sorbitol Xuan oleic acid cool. The money is also 127700.doc • 34- 200836731 can include Sweeteners, bridges, preservatives and antioxidants. Sweeteners (for example, glycerin, propylene glycol, sorbitol: alcohol or recommended sugar) can be used for syrups and granules. (4) These formulations may also contain a demulcent and antiseptic. d, flavoring agents and coloring agents and antioxidants. These pharmaceutical compositions can be injected into the form of Helmets, 囷, 囷, 王射水洛液. The available media and solvents are water, enchanting夂· a RS Grignard (Rlnger, s) solution and isotonic sodium chloride solution. The sterile injectable preparation may also be an injection of an oil-in-water microemulsion in which the active ingredient is dissolved in an oil phase. For example, The active ingredient can be first cut: in a mixture of large oil and _phospholipid. Then The oily solution is introduced into a mixture of water and glycerin and treated to form a microemulsion. The injectable solutions or microemulsions can be introduced into the patient by topical single injection or can be advantageously used to maintain the compounds of the invention. The IV fluid or microemulsion is administered in the form of a circulating concentration. To maintain this concentration, a continuous intravenous delivery device can be used. An example of this device is the "CADD-PLUSTM 5400 intravenous pump." The poultry muscles are administered subcutaneously in the form of an aqueous or oily suspension. The suspensions may be formulated according to the known techniques using such suitable dispersing or wetting agents and suspending agents as mentioned above. Injectable (d) may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example, a solution dissolved in hydrazine or 3-butane diol. Use volatile oil as solvent or suspension medium. Any mild fixed oil (including synthetic monoglycerides or diglycerides) can be used for this purpose. In addition, '127700.doc can be used in injectable preparations - 35- 200836731 For example, fatty acids such as oleic acid. For rectal administration of the drug ^" and s, the compound of formula 1 can also be used as a suppository. It can be used as a suitable and non-irritating excipient. Mixing to prepare a two composition' The excipient is solid at ambient temperature but liquid at rectal temperature and thus thaws in the rectum to release the drug. These materials include cocoa butter, oil gelatin, chlorinated vegetable oil, each a mixture of a molecular weight polyethylene glycol and a fatty acid ester of ethoxyethyl alcohol. For topical use, a cream, soft lean, gel, solution or suspension containing the compound of formula I may be employed. For topical application, mouthwashes and mouthwashes should be included.) The compounds of the present invention can be administered intranasally in a form suitable for intranasal training and delivery by topical application and or by transdermal routes. Transdermal skin patch forms well known to those skilled in the art are administered. Of course, for administration in the form of a transdermal delivery system, the dose administered throughout the dosage regimen should be continuous rather than intermittent. The compound of the present invention can also be used as a test mixture using a mixture of a glycol such as the following 2 matrix delivery cocoa butter, glycerin gelatin, hydrogenated vegetable oil, various molecular oximes, and a fatty acid ester of polyethylene glycol. When a subject is administered a compound of the invention, the selected dosage level will depend on various factors including, but not limited to, the activity of the particular compound, the severity of the individual's symptoms, the route of administration, and the administration. Time, rate of excretion of the compound, duration of treatment, other drugs, compounds and/or materials used in the combination, and age, sex, weight, condition, general health and prior medical history of the patient. The amount of the compound and the route of administration should ultimately be determined by the physician, but usually the dose should reach the target concentration at the point of action. 127700.doc •36·200836731 The local concentration that does not cause substantial harm or harmful side effects. In vivo administration can be carried out in a single dose, either continuously or intermittently (e.g., at a suitable interval) throughout the treatment period. Methods for determining the most effective administration methods and agents are well known to those skilled in the art and will vary with the formulation being used for the treatment, the purpose of the treatment, the target cells being treated, and the subject being treated. Single or multiple doses can be administered depending on the dosage level and mode selected by the treating physician.
Ο 般而口 "亥/舌性化合物之適宜劑量係介於約1 〇〇微克 至、、、勺250毫克/公斤又試者體重/天之間。當該活性化合物為 鹽、知、别藥或類似物時,根據母體化合物計算所投與之 S且因此實際所用重量應成正比增加。 本&月之化σ物亦可與抗癌劑或化學治療劑組合使用。 本I月之化a物亦可用作癌症治療之化學增敏劑及放射 增敏劑。其可用於二 兑 、σ療先則已經受或目前正經受癌症治療 之哺乳動物。此等先前治療包括預先化學治療、放射治 療、手術或免疫治療(例如,接種癌症疫苗)。 因此,本發明提供一籀 種用於同時、分開或相繼投藥之式 I化合物及抗癌劑的組合。适宜 The general dose of "Hai/tongue compound is between about 1 〇〇 microgram to , , , spoon 250 mg / kg and the tester weight / day. When the active compound is a salt, a known, a drug or the like, the S is administered in accordance with the parent compound and thus the actual weight used should be proportionally increased. This & month sigma can also be used in combination with an anticancer or chemotherapeutic agent. This I month can also be used as a chemical sensitizer and radiosensitizer for cancer treatment. It can be used in mammals that have been or are currently undergoing cancer treatment. Such prior treatments include pre-chemotherapy, radiation therapy, surgery or immunotherapy (e.g., vaccination against cancer). Accordingly, the present invention provides a combination of a compound of formula I and an anticancer agent for simultaneous, separate or sequential administration.
本發明提供一種用於ρη # X 、问時、分開或相繼投藥之式I化合 物、放射治療及另一化學 予冶療劑之組合。 本發明亦提供一種用认杂丨 用於製造藥物之式I化合物,該藥物 用作癌症治療之佐劑或驻 错由與電離輻射或化學治療劑組合 以加強腫瘤細胞治療。 本發明亦提供式1化合物在製造藥物中之用途,該藥物 127700.doc • 37 - 200836731 用作癌症治療之佐劑或藉由與電離輻射或其他化學治療劑 組合以加強腫瘤細胞治療。該等化合物亦可與電離輕射及 其他化學治療劑組合使用。 本發明亦提供一種化學治療或放射治療之方法,該方法 包括對有11的患者投與有效4的式^匕合物或包含式I化 合物以及電離輻射或化學治療劑之組合物。該等化合物亦 可與電離輻射及其他化學治療劑組合投與。 在組合治療中’本發明之化合物可在投與另—抗癌劑之 前(例如,5分鐘、15分鐘、3〇分鐘、45分鐘、}小時、 時、4小時、6小時、12小時、24小時、48小時、小時、 ^小時、1周、2周、3周、4周、5周、6周、8周、或12周 則)、同時、或之後(例如,5分鐘、15分鐘、3〇分鐘、45分 鐘、1小時、2小時、4小時、6小時、12小時、24小時、μ ^時、72小時、96小時、1周、2周、3周、4周、5周、6 =、8周、或12周後)投與有需要的受試者。在各實施例 二本:明之化合物與另一抗癌劑可間隔!分鐘、間隔⑺ 刀主鐘、間隔30分鐘、間隔不足1小時、間隔i小時至2小 時、間隔2小時至3小時、間隔3小時至叫、 至5小時、間隔5小時至6小時、間隔6小時至蚪日;、門隔日7 [間隔8小時至9小時、間隔9小時: 間小時至u小時、間隔1H、時至12 24小時、或間隔不超過48小時投與。、θ隔不超過 本發明之化合物及另一抗癌劑可以加合方 起作用。本發明化合物盘$ ^ ^或協同方式 月化口物與另一抗癌劑之協同組合可有利於 127700.doc •38· 200836731 使用較低劑量的此等藥冑中的一種或兩種及/或較小頻率 扠予本發明化合物及其他抗癌劑中的一種或兩種及/或以 較小頻率投與該等藥劑,此可在不降低該等藥劑在癌症治 療中之功效的情況下減少與投與給受試者之藥劑相關之任 何毒性。另外,協同效應可改良此等藥劑在癌症治療中之 功效及/或減少與單獨使用任一藥劑相關之任何不良或不 期望副作用。 與本發明化合物組合使用之抗癌劑或化學治療劑之實例 可發現於 Cancer Principles and Practice 〇f 〇nc〇l〇gy ^ V.T. Devita及 S· Heilman (編者),第 6版(2〇〇1 年 2 月 15 曰),Lippincott Williams & Wilkins Publishers中。普通技 術人員應能夠根據該等藥物之具體特性及所涉及癌症辨別 出可使用哪種藥劑組合。該等抗癌劑包括但不限於以下: HDAC抑制劑、雌激素受體調節劑、雄激素受體調節劑、 類視色素受體調節劑、細胞毒性/細胞生長抑制劑、抗增 生樂劑、異戊一稀基蛋白轉移酶抑制劑、HMG_CoA還原 酶抑制劑、HIV蛋白酶抑制劑、逆轉錄酶抑制劑及其他血 管生成抑制劑、細胞增生及存活信號傳導之抑制劑、細胞 凋亡誘導藥劑及干擾細胞週期檢測點之藥劑。本發明化合 物在與輕射療法共同實施時尤其有用。 ”HDAC抑制劑”之實例包括辛二醯基苯胺異羥將酸 (SAHA)、LAQ824、LBH589、PXD1G1、MS275、FK228、 丙戊酸、丁酸及CI-994。 π雌激素受體調節劑”係指干擾或抑制雌激素與受體結合 127700.doc •39- 200836731 (與作用機制無關)之化合物。雌激素受體調節劑之實例包 括但不限於他莫昔芬(tamoxifen)、雷洛昔芬(raloxifene)、 艾多昔芬(idoxifene)、LY35338 1、LY1 17081、托瑞米芬 (toremifene)、氟維司群(fuivestrant)、4-[7-(2,2_二甲基 _ι_ 氧代基丙氧基-4-甲基-2-[4-[2-(1-六氫吼啶基)乙氧基]苯 基]-2仏1·苯并吡喃_3_基苯基-2,2-二甲基丙酸酯、4,4,·二 經基二苯基酮_2,4_二硝基苯基-腙及SH646。 π雄激素受體調節劑”係指干擾或抑制雄激素與受體結合 (與作用機制無關)之化合物。雄激素受體調節劑之實例包 括非那雄胺(finasteride)及其他5α_還原酶抑制劑、尼魯米 特(mlutamide)、氟他胺(flutamide)、比卡魯胺(bicalutainide)、 利阿唾(liar〇z〇le)及乙酸阿比特龍(.加⑽加acetate)。 類視色素受體調節劑”係指干擾或抑制類視色素與受體 結合(與作用機制無關)之化合物。此等類視色素受體調節 劑之實例包括貝沙羅汀(bexar〇tene)、維A酸(tretin〇in)、 oc - —氣甲基鳥胺酸、 13-順式-視黃酸、9-順式_視黃酸、 ILX23-7553、反式|(4,m苯基)視黃醯胺、及^4邊基 苯基視黃醯胺。The present invention provides a combination of a compound of formula I, radiation therapy and another chemical chemotherapeutic agent for ρη # X , time, separate or sequential administration. The invention also provides a compound of formula I for use in the manufacture of a medicament for use as a medicament for the treatment of cancer or for the combination of ionizing radiation or a chemotherapeutic agent to enhance tumor cell therapy. The invention also provides the use of a compound of formula 1 for the manufacture of a medicament for use as an adjuvant for cancer therapy or for enhancing tumor cell therapy by combination with ionizing radiation or other chemotherapeutic agents. These compounds can also be used in combination with ionizing light radiation and other chemotherapeutic agents. The invention also provides a method of chemotherapy or radiation therapy comprising administering to a patient having 11 an effective compound of formula 4 or a composition comprising a compound of formula I and an ionizing radiation or chemotherapeutic agent. These compounds can also be administered in combination with ionizing radiation and other chemotherapeutic agents. In combination therapy, the compound of the invention may be administered prior to administration of another anti-cancer agent (eg, 5 minutes, 15 minutes, 3 minutes, 45 minutes, } hours, hours, 4 hours, 6 hours, 12 hours, 24 hours) Hours, 48 hours, hours, ^ hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks), simultaneously, or after (for example, 5 minutes, 15 minutes, 3 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, μ ^ hour, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, Subjects in need were administered 6 =, 8 weeks, or 12 weeks later. In each of the second embodiment: the compound of Ming can be separated from another anticancer agent! Minutes, interval (7) knife main clock, interval 30 minutes, interval less than 1 hour, interval i hour to 2 hours, interval 2 hours to 3 hours, interval 3 hours to call, to 5 hours, interval 5 hours to 6 hours, interval 6 hours to next day; door every other day 7 [interval 8 hours to 9 hours, interval 9 hours: interval to u hour, interval 1H, hour to 12 24 hours, or no more than 48 hours. The compound of the present invention and the other anticancer agent may act in combination. The synergistic combination of the compound of the invention in the form of a compound or a combination of another anticancer agent may be advantageous for 127700.doc •38·200836731 using a lower dose of one or both of these drugs and/or Or a lesser frequency fork to one or both of the compounds of the invention and other anticancer agents and/or to administer the agents at a lesser frequency, without reducing the efficacy of such agents in the treatment of cancer Any toxicity associated with administration of the agent to the subject is reduced. In addition, synergistic effects may improve the efficacy of such agents in the treatment of cancer and/or reduce any undesirable or undesirable side effects associated with the use of either agent alone. Examples of anticancer or chemotherapeutic agents for use in combination with the compounds of the present invention can be found in the Cancer Principles and Practice 〇f 〇nc〇l〇gy ^ VT Devita and S. Heilman (editor), 6th edition (2〇〇1) February 15 曰), Lippincott Williams & Wilkins Publishers. The ordinarily skilled artisan should be able to discern which combination of agents can be used based on the specific characteristics of the drugs and the cancer involved. Such anticancer agents include, but are not limited to, HDAC inhibitors, estrogen receptor modulators, androgen receptor modulators, retinoid receptor modulators, cytotoxic/cytostatic agents, anti-proliferative agents, Isoprenyl-protein transferase inhibitors, HMG_CoA reductase inhibitors, HIV protease inhibitors, reverse transcriptase inhibitors and other angiogenesis inhibitors, inhibitors of cell proliferation and survival signaling, apoptosis-inducing agents and An agent that interferes with cell cycle checkpoints. The compounds of the invention are especially useful when implemented in conjunction with light radiation therapy. Examples of "HDAC inhibitors" include octanediamine aniline isocyanate (SAHA), LAQ824, LBH589, PXD1G1, MS275, FK228, valproic acid, butyric acid, and CI-994. "π-estrogen receptor modulator" refers to a compound that interferes with or inhibits the binding of estrogen to a receptor 127700.doc • 39- 200836731 (independent of the mechanism of action). Examples of estrogen receptor modulators include, but are not limited to, tamoxifen Tamoxifen, raloxifene, idoxifene, LY35338 1, LY1 17081, toremifene, fuivestrant, 4-[7-(2 , 2_Dimethyl_ι_ oxopropoxy-4-methyl-2-[4-[2-(1-hexahydroacridinyl)ethoxy]phenyl]-2仏1·benzene And pyran_3_ylphenyl-2,2-dimethylpropionate, 4,4,di-diphenyldiphenyl ketone-2,4-dinitrophenyl-anthracene and SH646. A hormone receptor modulator refers to a compound that interferes with or inhibits the binding of androgen to a receptor (independent of the mechanism of action). Examples of androgen receptor modulators include finasteride and other 5α-reductase inhibitors. , mlutamide, flutamide, bicalutainide, liar〇z〇le, and abiraterone acetate (. plus (10) plus acetate). Receptor modulator" means dry Or a compound that inhibits retinoid binding to a receptor (independent of the mechanism of action). Examples of such retinoid receptor modulators include bexarrotene, tretin〇in, oc- - gas methyl ornithine, 13-cis-retinoic acid, 9-cis retinoic acid, ILX23-7553, trans | (4, m phenyl) retinol, and ^4 side groups Phenyl retinoin.
之抑制劑、抗代謝藥; 其包括烧基化劑、腫瘤壞死因子、 合物、微管抑制劑/微管穩定劑、 制劑、有絲分裂進程中所涉及激酶 生物反應調節劑;激素/抗激素類 127700.doc -40- 200836731 治療劑、造血生長因子、單株抗體靶向治療劑、拓撲異構 酶抑制劑、蛋白酶體抑制劑及泛素連接酶抑制劑。 細胞毒性劑之實例包括但不限於:環磷醯胺 (cyclophosphamide)、苯丁 酸氮芥(chlorambucil)、卡莫司 汀(carmustine)(BCNU)、洛莫司汀(lomustine)(CCNU)、白 消安(busulfan)、曲奥舒凡(treosulfan)、塞特那(sertenef)、 惡病質素(cachectin)、異環填酸胺(ifosfamide)、他索那明 (tasonermin)、氯尼達明(lonidamine)、卡始、六甲口密胺 (altretamine)、潑尼莫司汀(prednimustine)、二漠衛矛醇 (dibromodulcitol)、雷莫司汀(ranimustine)、 福莫司汀 (fotemustine)、奈達翻(n e dap 1 at in)、 奥利翻脂質體 (aroplatin)、奥沙利錄(oxaliplatin)、替莫嗤胺、甲烧石黃酸 甲酉旨、丙卡巴肼(procarbazine)、達卡巴喚(dacarbazine)、 庚始(heptaplatin)、雌莫司汀(estramustine)、對苯曱石黃酸英丙 舒凡(improsulfan tosilate)、曲填胺(trofosfamide)、尼莫司 汀(nimustine)、二溴螺氯銨(dibrospidium chloride)、嗓 σ密 替派(pumitepa)、樂顧(lobaplatin)、沙翻(satraplatin)、甲基絲裂 黴素(profiromycin)、順始、伊羅夫文(irofulven)、右異環 構酿胺(dexifosfamide)、順式·胺二氣(2-甲基-°比淀)翻、苄基 鳥σ票呤、葡填醯胺(glufosfamide)、GPX100、四氣化(反, 反,反)-雙-μ-(己烷-1,6-二胺)-μ-[二胺-鉑(II)]雙[二胺(氣)鉑 (II)]、二氮丙咬基精胺(diaziridinylspermine)、三氧化二 砷、1-(11-十二烷基胺基-10-羥基十一烷基)·3,7-二曱基黃 嗓呤、佐柔比星(zorubicin)、伊達比星(idarubicin)、柔紅黴素 127700.doc -41 - 200836731 (daunorubicin)、比生群(bisantrene)、米托蒽酉昆(mitoxantrone)、 σ比柔比星(pirarubicin)、σ比萘非特(pinafide)、伐蘆比星 (valrubicin)、氨柔比星(amrubicin)、多柔比星(doxorubicin)、 表柔比星(epirubicin)、°比柔比星(pirarubicin)、抗贅瘤 物、3 ’ -去胺基-31 -嗎琳基-13 -脫乳-10 -控基洋紅彳致素、脂質 體蒽環黴素(annamycin)、加蘭柔比星(galarubicin)、依利 奈法德(elinafide)、MEN10755、及4-去曱氧基-3-去胺基-3-氮丙啶基_心曱基磺醯基-柔紅黴素(參見WO 00/50032)。 其他實例包括Raf激酶抑制劑(例如,Bay43-9006)及mTOR 抑制劑(例如,Wyethfs CCI-779 及 Ariad AP23573)。其他實 例係PI3K抑制劑(例如,LY294002)。 在一實施例中,本發明之化合物可與烷基化劑組合使 用。 烷基化劑之實例包括但不限於氮芥類:環磷醯胺、異環 磷醯胺、曲磷胺及苯丁酸氮芥;亞硝基脲類:卡莫司汀 (BCNU)及洛莫司汀(CCNU);烷基磺酸酯類:白消安及曲 奥舒凡(treosulfan);三氮烯類:達卡巴嗓(dacarbazine)、 丙卡巴肼(procarbazine)及替莫峻胺;含翻錯合物:順翻、 卡鉑、奥利鉑脂質體及奥沙利鉑(oxaliplatin)。 在一實施例中,該烷基化劑係達卡巴嗪。可以介於約 150 mg/m2 (受試者體表面積)至約250 mg/m2之劑量對受試 者投與達卡巴嗪。在另一實施例中,可連續5天以介於約 1 50 mg/m2至約25 0 mg/m2間之劑量每天一次對受試者經靜 脈内投與達卡巴嗪。 127700.doc -42- 200836731 在一實施例中,該烷基化劑係丙卡巴肼。可以介於約50 mg/m2 (受試者體表面積)至約1〇〇 mg/m2間之劑量對受試者 投與丙卡巴肼。在另一實施例中,可連續5天以介於約50 mg/m2至約1〇〇 mg/m2間之劑量每天一次對受試者經靜脈内 投與丙卡巴肼。 在一實施例中,該烷基化劑係替莫唑胺。可以介於約 150 mg/m2 (受試者體表面積)至約2〇〇 mg/m2間之劑量對受 試者投與替莫唑胺。在另一實施例中,可連續5天以介於 約1 50 mg/m2至約200 mg/m2間之劑量每天一次對動物經口 投與替莫唑胺。 抗有絲分裂劑之實例包括··同分異構秋水仙鹼 (allocolchicine)、軟海綿素B(halichondrin B)、秋水仙鹼、 秋水仙鹼衍生物、多拉司他汀(d〇lstatin)10、美登素 (maytansine)、根黴素(rhizoxin)、硫代秋水仙驗 (thiocolchicine)及半胱胺酸三苯曱基酯。 低氧可活化化合物之實例係替拉紮明⑴rapazarnine)。 蛋白酶體抑制劑之實例包括但不限於乳胞素 (lactacystin)、波替單抗(b〇rtezomib)、伊波索米辛 (epoxomicin)及肽搭類’例如,MG 132、MG 115及 PSI 〇 微管抑制劑/微管穩定劑之實例包括紫杉醇(paclitaxel)、 硫酸長春地辛(vindesine sulfate)、長春新鹼(vincristine)、 長春花鹼(vinblastine)、長春瑞濱(vin〇relbine)、3,,4,·二脫 氫-4’-脫氧-8’-去甲長春鹼、多西紫杉醇(d〇cetax〇1)、根黴 素(rhizoxin)、多拉司他汀(d〇lastatin)、羥乙磺酸米伏布林 127700.doc -43· 200836731 (mivobulin isethionate)、奥裏斯他汀(auristatin)、西馬多 丁(cemadotin)、RPR109881、BMS184476、長春默寧 (vinflunine)、克利特非辛(cryptophycin)、2,3,4,5,6-五氟· #-(3-氟-4-曱氧基苯基)苯磺醯胺、脫水長春花驗 (anhydrovinblastine)、#,#_ 二甲基-L-纈胺醯基-L-纈胺酸 基-N-甲基-L-線胺醯基-L-膽胺酿基-L-膽胺酸-第三"·丁酿 胺、TDX258、埃坡黴素(ep〇thilone)(參見,例如,美國專 利第 6,284,781號及第 6,288,237號)及BMS188797。 拓撲異構酶抑制劑之某些實例係拓撲替康、 hycaptamine、伊立替康、魯比替康(rubitecan)、依克沙替康 (exatecan)、吉馬替康(gimetecan)、雙氟莫替康 (diflomotecan)、曱矽烧基-喜樹驗(silyl-camptothecin)、9-胺基喜樹驗、喜樹驗、克立那托(crisnatol)、絲裂黴素 (mitomycin C)、6-乙氧基丙醯基-3’/-Ο·外-亞苄基-教酒菌 素(chartreusin)、9-甲氧基二甲基-5-硝基吡唑并 [3,4,5-kl]吖啶-2-(6//)丙胺、1·胺基-9-乙基-5-氟-2,3-二氫-9-羥基-4_甲基-1//,12尺_苯并[de]吡喃並[3’,4|:1),7卜吲嗪並 [1,2b]喧琳 _ 1 〇, 13(9丹,1 5/ί)二酮、勒托替康(lurtotecan)、7-[2-(7V·異丙基胺基)乙基]-(20S)喜樹鹼、BNP1350、BNPI1100、 ΒΝ80915、ΒΝ80942、填酸依託泊普(etoposide phosphate)、 替尼泊苷(teniposide)、索布佐生(sobuzoxane)、2’·二甲基胺 基-2f-脫氧-依託泊苷、GL331、7V_[2_(二甲基胺基)乙基]-9-經基-5,6-.—甲基-6//· °比淀弁[4,唆-1 -甲酿胺、 asulacrine、(5a、5aB、8aa,9b)-9-[2-[iV-[2-(二甲基胺基)乙 127700.doc -44- 200836731 基]尽甲基胺基]乙基卜5·[‘羥基_3,5_二甲氧基笨基]- 環戊二烯-6,、2,3-(亞甲基二氧基)巧_甲基冬羥基j甲 氧基苯并[C]_菲咬鑌、6,9_雙[(2_胺基乙基)胺基]苯并⑷異 喧琳-5,l〇-一 _、5-(3_胺基丙基胺基)_7,1〇_二經基窥 基乙基胺基甲基)_6/^比唑并[^丨士]吖啶_6酮、象屮 [2(一乙基胺基)乙基胺基卜7-甲氧基-9-氧代基-9H-噻噸 基甲基]甲醯胺、沁(2-(二甲基胺基)乙基)吖啶_4_甲醯胺、 6-[[2-(二甲基胺基)乙基]胺基]_3_羥基_77/-茚并[2,Kc]喹 啉-7-酮、及地美司鈉(dimesna);非喜樹鹼型拓撲異構酶」 抑制劑,例如,吲哚并咔唑;及雙重拓撲異構酶_1及11抑 制劑,例如,苯并吩嗪、XR2〇 1 15761MLN 576及苯并吡 σ定并吲哚。 在一實施例中,該拓撲異構酶抑制劑係伊立替康。可以 介於約50 mg/m2 (受試者體表面積)至約15〇間之劑 量對文试者投與伊立替康。在另一實施例中,可連續5天 (第1天至第5天)以介於約5〇 mg/m2至約15〇 mg/m2間之劑量 每天一次對受試者經靜脈内投與伊立替康,隨後連續5天 (第28天至第32天)以介於約5〇 mg/m2至約15〇 mg/m2間之劑 量每天一次再次經靜脈内投藥,隨後連續5天(第55天至第 59天)以介於約50 mg/m2至約150 mg/m2間之劑量每天一次 再次經靜脈内投藥。 有絲分裂驅動蛋白(且具體而言,係人類有絲分裂驅動 蛋白KSP)抑制劑之實例闡述於PCT公開案WO 01/30768、 127700.doc -45- 200836731Inhibitors, antimetabolites; include an alkylating agent, a tumor necrosis factor, a microtubule inhibitor/microtubule stabilizer, a preparation, a kinase biological response modifier involved in the process of mitosis; a hormone/antihormone 127700.doc -40- 200836731 Therapeutic agents, hematopoietic growth factors, monoclonal antibody targeted therapeutics, topoisomerase inhibitors, proteasome inhibitors and ubiquitin ligase inhibitors. Examples of cytotoxic agents include, but are not limited to, cyclophosphamide, chlorambucil, carmustine (BCNU), lomustine (CCNU), white. Busulfan, treosulfan, sertenef, cachectin, ifosfamide, tasonermin, lonidamine , card initial, altretamine, prednimustine, dibromodulcitol, ranimustine, fotemustine, neda Ne dap 1 at in), aroplatin, oxaliplatin, temoguanamine, methicillin, procarbazine, dacarbazine ), heptaplatin, estramustine, improsulfan tosilate, trofosfamide, nimustine, dibromospiro Dibrospidium chloride, pumitepa, music (lobaplatin), satraplatin, profiromycin, cis, irofulven, dexifosfamide, cis-amine dioxin (2- Methyl-° ratio precipitation, benzyl bird σ 呤 呤, glufosfamide, GPX100, four gasification (reverse, anti, trans)-bis-μ-(hexane-1,6-two Amine)-μ-[diamine-platinum(II)] bis [diamine (gas) platinum (II)], diaziridinylspermine, arsenic trioxide, 1-(11-dodecylamine) Alkyl-10-hydroxyundecyl)·3,7-dimercaptopurine, zorubicin, idarubicin, daunorubicin 127700.doc -41 - 200836731 (daunorubicin ), bisantrene, mitoxantrone, pirarubicin, σpinafide, valrubicin, amrubicin , doxorubicin, epirubicin, pirarubicin, anti-tumor, 3 '-deamino-31 - morphine-13 - de-emulsion-10 -Controlled erythropoietin, lipid Anthocyanin, galarubicin, elinafide, MEN10755, and 4-demethoxy-3-desamino-3-aziridine Mercaptosulfonyl-daunorubicin (see WO 00/50032). Other examples include Raf kinase inhibitors (e.g., Bay 43-9006) and mTOR inhibitors (e.g., Wyethfs CCI-779 and Ariad AP23573). Other examples are PI3K inhibitors (e.g., LY294002). In one embodiment, the compounds of the invention can be used in combination with an alkylating agent. Examples of alkylating agents include, but are not limited to, nitrogen mustards: cyclophosphamide, ifosfamide, tromethamine, and chlorambucil; nitrosoureas: carmustine (BCNU) and Lo Mostin (CCNU); alkyl sulfonates: busulfan and treosulfan; triazenes: dacarbazine, procarbazine and temoride; Inclusion complexes: cisplatin, carboplatin, orlibolite liposomes and oxaliplatin. In one embodiment, the alkylating agent is dacarbazine. The subject may be administered dacarbazine at a dose of between about 150 mg/m2 (subject surface area) to about 250 mg/m2. In another embodiment, the subject can be intravenously administered dacarbazine once daily for 5 consecutive days at a dose between about 1 50 mg/m2 to about 25 mg/m2. 127700.doc -42- 200836731 In one embodiment, the alkylating agent is procarbazine. The subject may be administered procarbazine at a dose of between about 50 mg/m2 (subject surface area) to about 1 mg/m2. In another embodiment, the subject can be administered intravenously with procarbazine once daily for 5 consecutive days at a dose between about 50 mg/m2 to about 1 mg/m2. In one embodiment, the alkylating agent is temozolomide. The subject may be administered temozolomide at a dose of between about 150 mg/m2 (subject surface area) to about 2 mg/m2. In another embodiment, the animal can be orally administered temozolomide once daily for 5 consecutive days at a dose of between about 1 50 mg/m2 to about 200 mg/m2. Examples of anti-mitotic agents include: allocolchicine, halichondrin B, colchicine, colchicine derivatives, dolastatin (d〇lstatin) 10, beauty Maytansine, rhizoxin, thiocolchicine, and triphenyl decyl cysteate. An example of a hypoxic activatable compound is tirapazamine (1) rapazarnine). Examples of proteasome inhibitors include, but are not limited to, lactacystin, börtezomib, epoxomicin, and peptides, eg, MG 132, MG 115, and PSI. Examples of tube inhibitor/microtubule stabilizers include paclitaxel, vindesine sulfate, vincristine, vinblastine, vin〇relbine, 3, , 4, · didehydro-4'-deoxy-8'-norvinine, docetaxel (d〇cetax〇1), rhizoxin (rhizoxin), dolastatin (d〇lastatin), hydroxy Metbuterin sulfonate 127700.doc -43· 200836731 (mivobulin isethionate), auristatin, cemadotin, RPR109881, BMS184476, vinflunine, clitfecine Cryptophycin), 2,3,4,5,6-pentafluoro·#-(3-fluoro-4-decyloxyphenyl)benzenesulfonamide, anhydrovinblastine, #,#_ --L-Amidoxime-L-proline-N-methyl-L-line amine thiol-L-cholamine aryl-L-cholinic acid-third " butylamine, T DX258, ep〇thilone (see, for example, U.S. Patent Nos. 6,284,781 and 6,288,237) and BMS188797. Some examples of topoisomerase inhibitors are topotecan, hycaptamine, irinotecan, rubiconcan, exatecan, gimetecan, diflupremox (diflomotecan), silyl-camptothecin, 9-amino-based eucalyptus, euphorbia, cristatol, mitomycin C, 6-ethoxy Propionyl-3'/-Ο·Exo-benzylidene-chartreusin, 9-methoxydimethyl-5-nitropyrazolo[3,4,5-kl] Acridine-2-(6//)propylamine, 1·amino-9-ethyl-5-fluoro-2,3-dihydro-9-hydroxy-4_methyl-1//, 12 ft. benzene And [de]pyrano[3',4|:1),7-disoxazin[1,2b]喧琳_ 1 〇, 13(9丹,1 5/ί)dione, letoticon (lurtotecan), 7-[2-(7V·isopropylamino)ethyl]-(20S) camptothecin, BNP1350, BNPI1100, ΒΝ80915, ΒΝ80942, etoposide phosphate, teniposide Teniposide, sobuzuxane, 2'-dimethylamino-2f-deoxy-etoposide, GL331, 7V_[2_(dimethylamino)ethyl]-9-carbyl- 5,6-.-methyl- 6//· ° ratio of yttrium [4, 唆-1 - amide, asulacrine, (5a, 5aB, 8aa, 9b)-9-[2-[iV-[2-(dimethylamino)) 127700.doc -44- 200836731 】] methylamino] ethyl b 5 [[hydroxy_3,5-dimethoxyphenyl]-cyclopentadiene-6,2,3- (Asia Methyldioxy) _methyl-winter hydroxy j-methoxybenzo[C] phenanthrene, 6,9-bis[(2-aminoethyl)amino]benzo(4)isoindene- 5,l〇-一_,5-(3-aminopropylamino)_7,1〇_dipyridylethylaminomethyl)_6/^biszolo[^丨士]吖pyridine _6 ketone, like oxime [2 (monoethylamino) ethylaminophenyl 7-methoxy-9-oxo-9H-thioxylmethyl]carbamamine, hydrazine (2-(two) Methylamino)ethyl)acridine_4_formamide, 6-[[2-(dimethylamino)ethyl]amino]_3_hydroxy_77/-茚[2,Kc] Quinoline-7-one, and dimesna; non-camptothecin-type topoisomerase inhibitors, for example, indolocarbazole; and dual topoisomerases 1 and 11 inhibitors, For example, benzophenazine, XR2〇1 15761MLN 576, and benzopyrazine are deuterated. In one embodiment, the topoisomerase inhibitor is irinotecan. The irinotecan can be administered to the subject at a dose of between about 50 mg/m2 (subject surface area) to about 15 Torr. In another embodiment, the subject can be administered intravenously once daily for 5 consecutive days (Day 1 to Day 5) at a dose between about 5 〇 mg/m2 to about 15 〇 mg/m2. Irinotecan, followed by intravenous administration for once every 5 days (Day 28 to Day 32) at a dose between about 5 〇 mg/m 2 and about 15 〇 mg/m 2 , followed by 5 consecutive days From 55 days to 59 days), intravenous administration once again at a dose of between about 50 mg/m2 and about 150 mg/m2. Examples of inhibitors of mitotic kinesins (and in particular, human mitotic kinesin KSP) are described in PCT Publication WO 01/30768, 127700.doc -45-200836731
WO 01/98278 > WO 02/056880 、WO 03/050,064 、 WO 03/050,122 、WO 03/049,527、 WO 03/049,679 、 WO 03/049,678 、WO 03/039460 > WO 03/079973 > WO 03/099211 、WO 2004/039774 、WO 03/105855 、 WO 03/106417 、WO 2004/087050 ' WO 2004/058700 、 WO 2004/058148和WO 2004/037171及美國專利申請案US 2004/13283 0和US 2004/132719中。在一實施例中,有絲分 裂驅動蛋白之抑制劑包括但不限於KSP之抑制劑、MKLP1 之抑制劑、CENP-E之抑制劑、MCAK之抑制劑、Kifl4之 抑制劑、Mphosphl之抑制劑及Rab6_KIFL之抑制劑° ’’有絲分裂進程中所涉及激酶之抑制劑’’包括但不限於 aurora激酶之抑制劑、Polo樣激酶(PLK)之抑制劑(具體而 言,係PLK-1之抑制劑)、bub-1之抑制劑及bub-Rl之抑制 劑。 π抗增生劑”包括反義RNA及DNA寡核苷酸類,例如, G3139、ODN698、RVASKRAS、GEM231、及 ΙΝΧ3001 ; 及抗代謝物,例如,依諾他濱(enocitabine)、卡莫氟 (carmofur)、替加敦(tegafur)、喷司他丁(pentostatin)、去 氧 II 尿普(doxifluridine)、三甲曲沙(trimetrexate)、氟達拉 濱(fludarabine)、卡培他濱(capecitabine)、加洛他濱 (galocitabine)、阿糖胞皆十八烧基構酸鈉(cytarabine ocfosfate)、(fosteabine sodium hydrate)、雷替曲塞 (raltitrexed)、(paltitrexid)、乙喊替氟(emitefur)、嗟 σ坐吱 林(tiazofurin)、地西他濱(decitabine)、諾拉曲塞 127700.doc •46- 200836731 (nolatrexed)、培美曲塞(pemetrexed)、奈拉濱 (nelzarabine)、2’-脫氧-2,_亞甲基胞苷、2,_氟亞甲基_2、脫 氧胞普、#-[5-(2,3-一 IL ·苯并吱喃基)績醯基]_τν^(3,4-二氯 笨基)脲、屬-[4-脫氧-4-[#2-[2作),4斤)-十四碳二烯醯基]甘 胺醯基胺基]_L-丙三氧基-B-L-甘露-七吡喃糖基]腺嗓呤、 aplidine、海鞘素(ecteinascidin)、曲沙他濱(troxacitabine)、 4-[2-胺基-4-氧代基-4,6,7,8-四氫-3//-嘧啶并[5,4-13][1,4]噻 嗪-6-基_(幻_乙基]-2,5-噻吩基-L-麩胺酸、胺基蝶呤、5-氟 尿’ σ定、阿拉諾新(alanosine)、11-乙醯基-8-(胺甲氧基 甲基)-4-甲醯基-6-甲氧基-14-氧雜基-1,11-二氮雜四環并 (7·4·1·0·0)-十四碳_2,4,6-三烯-9-基乙酸酯、苦馬豆素 (swainsonine)、洛美曲索(lometrexol)、右雷佐生 (dexrazoxane)、甲硫胺酸酶、氰基_2,_脫氧棕櫚醯 基呋喃阿糖基胞嘧啶及弘胺基吡啶_2_甲醛縮胺基 硫脈。 單株抗體乾向治療劑之實例包括彼等具有與癌細胞特異 性或乾細胞特異性單株抗體連接之細胞毒性因子或放射性 同位素的治療劑。實例包括百克沙(Bexxar)。 ’’HMG-CoA還原酶抑制劑”係指3_羥基_3_甲基戊二醯基_ CoA還原酶之抑制劑。可使用的hmG-CoA還原酶抑制劑之 實例包括但不限於洛伐他汀(lovastatin)(MEVAC〇R⑧;參 見美國專利第4,231,938號、第4,294,926號及第4,319,039 號)、辛伐他汀(simvastatin)(ZOCOR⑧;參見美國專利第 4,444,784號、第4,820,850號及第4,916,239號)' 普伐他汀 127700.doc -47- 200836731 (pravastatin)(PRAVACHOL⑧;參見美國專利第 4,346,227 號、第 4,537,859 號、第 4,410,629號、第 5,030,447 號及第 5,180,589號)、氟伐他汀(fluvastatin)(LESCOL®;參見美國 專利第 5,354,772號、第 4,911,165號、第 4,929,437號、第 5,189,164 號、第 5,1 18,853 號、第 5,290,946 號及第 5,356,896號)及阿托伐他汀(atorvastatin)(LIPITOR® ;參見 美國專利第5,273,995號、第4,681,893號、第5,489,691號 及第5,342,952號)。此等及本發明方法可使用的其他HMG-CoA還原酶抑制劑之結構式闡述於M· Yalpani, ”Cholesterol Lowering Drugs’’,Chemistry & Industry,第 8 5-89頁(1996年2月5日)之第87頁及美國專利第4,782,084號 及第4,885,314號中。本文所用術語HMG-CoA還原酶抑制 劑包括具有HMG-CoA還原酶抑制活性之化合物的所有醫 藥上可接受之内酯及開環酸形式(即,其中内酯環打開以 形成游離酸)以及鹽和酯形式,且因此此等鹽、酯、開環 酸及内酯形式之使用屬於本發明之範圍。 ”異戊二烯基-蛋白轉移酶抑制劑”係指一種可抑制任一種 或任一組合的異戊二浠基-蛋白轉移酶酵素之化合物,該 等異戊二稀基-蛋白轉移酶酵素包括法呢基-蛋白轉移酶 (FPTase)、I型尨牛兒基尨牛兒基蛋白轉移酶(GGPTase-I)、及II型尨牛兒基尨牛兒基-蛋白轉移酶(GGPTase-II,亦 稱為 Rab GGPTase)。 異戊二烯基-蛋白轉移酶抑制劑之實例可發現於下列公 開案及專利中:WO 96/30343、WO 97/18813、WO 97/21701、 127700.doc -48- 200836731 WO 97/23478、WO 97/38665、WO 98/28980、WO 98/291 19、WO 95/32987、美國專利第 5,420,245號、美國 專利第5,523,430號、美國專利第5,532,359號、美國專利第 5,510,510號、美國專利第5,589,485號、美國專利第 5,602,098號、歐洲專利公開案0 618 221、歐洲專利公開 • 案 0 675 1 12、歐洲專利公開案 0 604 181、歐洲專利公WO 01/98278 > WO 02/056880, WO 03/050,064, WO 03/050,122, WO 03/049,527, WO 03/049,679, WO 03/049,678, WO 03/039460 > WO 03/079973 > WO 03 </ RTI> </ RTI> </ RTI> </ RTI> </ RTI> </ RTI> </ RTI> </ RTI> </ RTI> </ RTI> </ RTI> </ RTI> </ RTI> </ RTI> </ RTI> </ RTI> </ RTI> </ RTI> </ RTI> </ RTI> </ RTI> WO 2004/058700, WO 2004/058148 and WO 2004/037171 /132719. In one embodiment, the inhibitor of mitotic kinesin includes, but is not limited to, an inhibitor of KSP, an inhibitor of MKLP1, an inhibitor of CENP-E, an inhibitor of MCAK, an inhibitor of Kifl4, an inhibitor of Mphosphl, and a Rab6_KIFL Inhibitors ''Inhibitors of kinases involved in mitotic processes'' include, but are not limited to, inhibitors of aurora kinase, inhibitors of Polo-like kinase (PLK) (specifically, inhibitors of PLK-1), bub -1 inhibitor and inhibitor of bub-Rl. π anti-proliferative agents include antisense RNA and DNA oligonucleotides, for example, G3139, ODN698, RVASKRAS, GEM231, and ΙΝΧ3001; and antimetabolites, for example, enocitabine, carmofur (carmofur) , tegafur, pentostatin, dexifluridine, trimetrexate, fludarabine, capecitabine, gallo Calocitabine, cytarabine ocfosfate, fosteabine sodium hydrate, raltitrexed, (paltitrexid), emitefur, 嗟σ Sitting on tiazofurin, decitabine, nola cexi 127700.doc •46- 200836731 (nolatrexed), pemetrexed, nelzarabine, 2'-deoxy- 2,_methylenecytidine, 2,_fluoromethylene-2, deoxycytopent, #-[5-(2,3-ILO·benzopyranyl) 醯 ]]]_τν^(3 ,4-dichlorophenyl)urea, genus-[4-deoxy-4-[#2-[2], 4 kg)-tetradecadienyl]glycine]aminoamine]_L-propyl Trioxy- BL-mannose-heptanopyranosyl] adenine, aplidine, ecteinascidin, troxacitabine, 4-[2-amino-4-oxo-4,6,7, 8-tetrahydro-3//-pyrimido[5,4-13][1,4]thiazin-6-yl-(phanyl-ethyl)-2,5-thienyl-L-glutamic acid, Aminopterin, 5-fluorourine' sigma, alanosine, 11-acetamido-8-(aminomethoxymethyl)-4-carboxyl-6-methoxy-14 -oxal-1,11-diazatetracyclo(7·4·1·0·0)-tetradecyl-2,4,6-trien-9-yl acetate, hummus Swainsonine, lometrexol, dexrazoxane, methionine, cyano-2, _deoxypalmitosylfuranosylcytosine and arginylpyridine_2_ Formaldehyde Aminothiol. Examples of monoclonal antibody dry therapeutic agents include those having a cytotoxic or radioisotope linked to a cancer cell-specific or stem cell-specific monoclonal antibody. Examples include Bexxar. ''HMG-CoA reductase inhibitor') means an inhibitor of 3-hydroxy-3-methylpentadienyl-CoA reductase. Examples of hmG-CoA reductase inhibitors that may be used include, but are not limited to, lovar Lavastatin (MEVAC® R8; see U.S. Patent Nos. 4,231,938, 4,294,926 and 4,319,039), simvastatin (ZOCOR8; see U.S. Patent Nos. 4,444,784, 4,820,850 and 4,916,239 'Pravastatin 127700.doc -47-200836731 (pravastatin) (PRAVACHOL8; see U.S. Patent Nos. 4,346,227, 4,537,859, 4,410,629, 5,030,447 and 5,180,589), fluvastatin (LESCOL®; see U.S. Patent Nos. 5,354,772, 4,911,165, 4,929,437, 5,189,164, 5,1 18,853, 5,290,946 and 5,356,896) and atorvastatin ( Atorvastatin) (LIPITOR®; see U.S. Patent Nos. 5,273,995, 4,681,893, 5,489,691 and 5,342,952). Other HMG-CoA reductase inhibitors which can be used in the methods of the present invention. Structure set forth in formula M · Yalpani, "Cholesterol Lowering Drugs '', Chemistry & Industry, 8 pages 5-89 (5 February 1996), page 87 of U.S. Patent No. 4,782,084 and No. 4,885,314 second. The term HMG-CoA reductase inhibitor as used herein includes all pharmaceutically acceptable lactone and ring opening acid forms of a compound having HMG-CoA reductase inhibitory activity (ie, wherein the lactone ring is opened to form a free acid) and a salt And ester forms, and thus the use of such salts, esters, ring opening acids and lactone forms are within the scope of the invention. "Prenyl-protein transferase inhibitor" means a compound which inhibits any one or any combination of isoprenyl-protein transferase enzymes, such isoprenyl-protein transferase enzymes Including farnesyl-protein transferase (FPTase), type I geranyl-based geranyl-protein transferase (GGPTase-I), and type II geranyl-based geranyl-protein transferase (GGPTase-II) Also known as Rab GGPTase). Examples of isoprenyl-protein transferase inhibitors can be found in the following publications and patents: WO 96/30343, WO 97/18813, WO 97/21701, 127700.doc -48-200836731 WO 97/23478, WO 97/38665, WO 98/28980, WO 98/291 19, WO 95/32987, U.S. Patent No. 5,420,245, U.S. Patent No. 5,523,430, U.S. Patent No. 5,532,359, U.S. Patent No. 5,510,510, U.S. Patent No. 5,589,485 U.S. Patent No. 5,602,098, European Patent Publication No. 0 618 221, European Patent Publication No. 0 675 1 12, European Patent Publication No. 0 604 181, European Patent
. 開案 〇 696 593、WO 94/19357、WO 95/08542、WO 95/11917、WO 95/12612、WO 95/12572、WO 95/105 14、 ( 美國專利第 5,661,152號、WO 95/10515、WO 95/10516、 WO 95/24612、WO 95/34535、WO 95/25086、WO 96/05529、WO 96/06138、WO 96/06193、WO 96/16443、 WO 96/21701、WO 96/21456、WO 96/22278、WO 96/2461 1、WO 96/24612、WO 96/05168、WO 96/05169、 WO 96/00736、美國專利第 5,571,792號、WO 96/17861、 WO 96/33 159、WO 96/34850、WO 96/34851、WO 96/30017、WO 96/30018、WO 96/30362、WO 96/30363、 ί WO 96/31111 、WO 96/31477、WO 96/31478、WO 96/3 1501、WO 97/00252、WO 97/03047 > WO 97/03050、 WO 97/04785、WO 97/02920、WO 97/17070、WO 97/23478、WO 97/26246 > WO 97/30053、WO 97/44350、 WO 98/02436、及美國專利第5,532,359號。對於異戊二烯 基-蛋白轉移酶抑制劑對血管生成作用之實例,可參見 五J· o/Cancer (1999),35(9):1394-1401。 π血管生成抑制劑”係指一種可抑制新穎血管形成(與作用 127700.doc -49- 200836731 機制無關)之化合物。血管生成抑制劑之實例包括但不限 於諸如酪胺酸激酶受體Flt-l (VEGFR1)及Flk-1/KDR (VEGFR2)之抑制劑等酪胺酸激酶抑制劑、表皮源、纖維 母細胞源或血小板源生長因子之抑制劑、MMP (基質金屬 蛋白酶)抑制劑、整合素阻滯劑、干擾素-α、介白素-12、 多硫酸戊聚糖酯(pentosan polysulfate)、環氧合酶抑制 劑,包括非類固醇抗炎劑(NSAID)(如阿司匹林(aspirin)及 布洛芬(ibuprofen))以及選擇性環氧合酶-2抑制劑(如塞利 西蔔(celecoxib)及羅非考昔(rofecoxit^KPA^S (1992) 89:7384; JNCI (1982) 69:475; Arch. Opthalmol. (1990) 108:573; Anat. Rec· (1994) 238:68; FEES Letters (1995) 372:83; Clin, Orthop.(1995) 313:76; J. MoL Endocrinol. (1996) 16:107; Jpn. J. Pharmacol. (1997) 75:105; Cancer Res.(1997) 57:1625 (1997); Cell (1998) 93:705; Inti J. Mol· Med. (1998) 2:715; J· Biol· Chem. (1999) 274:9116))、類固醇抗炎劑(例如,皮質類固醇、鹽皮質激 素、地塞米松(dexamethasone)、潑尼松(prednisone)、潑尼 松龍(prednisolone)、甲潑尼龍、倍他米松(betamethasone))、 曱醯胺三°坐、康布瑞塔卡汀A-4(combretastatin A-4)、角鯊 胺、6-0-氣乙醯基-羰基)-煙麯黴醇(fumagillol)、沙立度胺 (thalidomide)、血管抑制素、肌妈蛋白-1、血管緊張素II拮 抗劑(參見/· C/M. MeA (1985) 105:14卜 145)、及 VEGF抗體(參見 ⑽(1999) 17:963-968;开 696 593, WO 94/19357, WO 95/08542, WO 95/11917, WO 95/12612, WO 95/12572, WO 95/105 14 (U.S. Patent No. 5,661,152, WO 95/) 10515, WO 95/10516, WO 95/24612, WO 95/34535, WO 95/25086, WO 96/05529, WO 96/06138, WO 96/06193, WO 96/16443, WO 96/21701, WO 96/ 21,456, WO 96/22278, WO 96/2461 1, WO 96/24612, WO 96/05168, WO 96/05169, WO 96/00736, US Patent No. 5,571,792, WO 96/17861, WO 96/33 159, WO 96/34850, WO 96/34851, WO 96/30017, WO 96/30018, WO 96/30362, WO 96/30363, WO 96/31111, WO 96/31477, WO 96/31478, WO 96 /3 1501, WO 97/00252, WO 97/03047 > WO 97/03050, WO 97/04785, WO 97/02920, WO 97/17070, WO 97/23478, WO 97/26246 > WO 97/30053 WO 97/44350, WO 98/02436, and U.S. Patent No. 5,532,359. For examples of the effects of prenyl-protein transferase inhibitors on angiogenesis, see JJ o/Cancer (1999), 35 (9): 1394-1401. π angiogenesis inhibitor means a form of inhibiting novel blood vessel formation 127700.doc -49- 200836731 Mechanism-independent compounds. Examples of angiogenesis inhibitors include, but are not limited to, tyrosine kinase receptor Flt-1 (VEGFR1) and Flk-1/KDR (VEGFR2) inhibitors Amino acid kinase inhibitor, epidermal source, fibroblast source or inhibitor of platelet-derived growth factor, MMP (matrix metalloproteinase) inhibitor, integrin blocker, interferon-α, interleukin-12, polysulfate Pentosan polysulfate, cyclooxygenase inhibitors, including non-steroidal anti-inflammatory agents (NSAIDs) (such as aspirin and ibuprofen) and selective cyclooxygenase-2 inhibitors (eg celecoxib and rofecoxit^KPA^S (1992) 89:7384; JNCI (1982) 69:475; Arch. Opthalmol. (1990) 108:573; Anat. Rec· (1994) 238:68; FEES Letters (1995) 372:83; Clin, Orthop. (1995) 313:76; J. MoL Endocrinol. (1996) 16:107; Jpn. J. Pharmacol. (1997) 75: 105; Cancer Res. (1997) 57:1625 (1997); Cell (1998) 93:705; Inti J. Mol. Med. (1998) 2:715; J. Biol. Chem. (1999) 274:9116) ),Steroid Inflammatory agents (eg, corticosteroids, mineralocorticoids, dexamethasone, prednisone, prednisolone, methylprednisolone, betamethasone, guanamine) °Sit, combretatatin A-4 (combretastatin A-4), squalamine, 6-0-ephthyl-carbonyl) fumagillol (thalidomide), thalidomide, Angiostatin, myoma protein-1, angiotensin II antagonist (see /· C/M. MeA (1985) 105:14 145), and VEGF antibody (see (10) (1999) 17: 963-968;
Kim等人(1993) 362:841-844; WO 00/44777 ;及 WO 127700.doc -50- 200836731 00/61186)° 可調節或抑制血管生成且亦可與本發明化合物組合使用 之其他治療劑包括可調節或抑制凝固及纖維蛋白溶解系統 之藥劑(參見 C7k. Ζα· Med· (2000) 38:679-692)。可 調節或抑制凝固及纖維蛋白溶解途徑之此等藥劑的實例包 ’ 括但不限於肝素(參見 TTzrowZ?· (1998) 80:10- - 23)、低分子量肝素及羧肽酶U抑制劑(亦稱作活性凝血酶 可活化纖維蛋白溶解抑制因子[TAFIa]之抑制劑)(參見 (: 及以·(2001) 101:329-354)。TAFIa抑制劑已闡 述於卩(:丁公開案冒0 03/013,526及美國專利第60/349,925號 (2002年1月18曰提出申請)中。 π干擾細胞週期檢查點之藥劑”係指可抑制轉導細胞週期 檢查點信號之蛋白激酶,藉此致使癌細胞對DNA損害劑敏 感的化合物。此等藥劑包括ATR、ATM、Chkl及Chk2激 酶之抑制劑及cdk和cdc激酶抑制劑且其具體實例為7-經基 星形孢菌素、星形孢菌素、夫拉平度(flavoPirid〇l)、 〇 CYC202 (Cyclacel)及 BMS_387032。 π細胞增生及存活信號傳導途徑之抑制劑’f係指可抑制細 . 胞表面受體及此等表面受體下游信號轉導通路之醫藥劑。 此等藥劑包括EGFR之抑制劑(例如,吉非替尼(gefitinib)及 埃羅替尼(erlotinib))、ERB-2之抑制劑(例如’曲司佐單抗 (trastuzumab))、IGFR之抑制劑(例如,彼等揭示於WO 03/05995 1中者)、細胞因子受體之抑制劑、MET之抑制 劑、PI3K之抑制劑(例如,LY294002)、絲胺酸/蘇胺酸激 127700.doc •51 - 200836731 酶(包括但不限於Akt之抑制劑,例如闡述於WO 03/086404、WO 03/086403、WO 03/086394、WO 03/086279、 WO 02/083675、WO 02/083139、WO 02/083140及WO 02/083138 中者)、Raf激酶之抑制劑(例如,BAY-43-9006 )、MEK之 抑制劑(例如,CI-1040及PD-098059)及mTOR之抑制劑(例 如,Wyeth CCI-779及Ariad AP23 5 73)。此等藥劑包括小分 子抑制劑化合物及抗體拮抗劑。 ’’細胞凋亡誘導劑”包括TNF受體家族成員(包括TRAIL受 體)之激活劑。 在一實施例中,本發明之化合物可聯合一種或多種(特 定言之,一種、兩種或三種)選自替莫唑胺、順鉑、卡 鉑、奥沙利鉑、伊立替康及拓撲替康之藥劑用於治療癌 症。 本發明之化合物亦可聯合任一種或多種下列治療劑用於 治療癌症:阿巴瑞克(abarelix)(Plenaxis depot®);阿地白 介素(aldesleukinMProkine^);阿地白介素(Aldesleukin) (Proleukin );阿佘珠單抗(Alemtuzumabb)(Campath );阿曲 諸英(alitretinoinKPanretin^);別口票呤醇(allopurinol) (Zyloprim®);六甲 口密胺(altretamineXHexalen®);胺碟汀 (amifostine)(Ethyol®);阿那曲唾(anastrozoleXArimidex®);三 氧化二坤(Trisenox®);天冬醯胺酶(Elspar®);阿紮胞苷 (azacitidine)(Vidaza@);貝伐單抗(bevacuzimabXAvastin®); 貝沙羅汀(bexarotene)膠囊(Targretin®);貝沙羅汀凝膠 (Targretin®);博來黴素(bleomycin)(Blenoxane®);波替單 127700.doc •52- 200836731 抗(bortezomib)(Velcade®);靜脈内使用的白消安 (Busulfex®) ; 口服白消安(Myleran®);卡普睾酮 (calusteroneXMethosarb®);卡培他濱(capecitabine)(Xeloda@);卡 鉑(Paraplatin®);卡莫司汀(BCNU®,BiCNU@);卡莫司汀 (Gliadel®);卡莫司汀與聚苯丙生20植入物(Gliadel Wafer®);塞利西蔔(Celebrex®);西土 西單抗(cetuximab) (Erbitux@);苯丁 酸氮芥(chlorambucilXLeukeran®);順翻 (Platinol®);克拉屈濱(cladribine)(Leustatin®,2-CdA@); 氯苯吩嗓(clofarabine)(Clolar@);環填酿胺(Cytoxan®, Neosar®);環填醢胺(Cytoxan Injection⑧);環構醢胺 (Cytoxan Tablet®);阿糖胞苷(Cytosar-U®);阿糖胞苷脂質體 (DepoCyt®);達卡巴嗪(DTIC-Dome®);更生黴素 (dactinomycin),放線菌素 D(actinomycin DXCosmegen®);促紅 血球生成素a(Darbepoetin alfa)(Aranesp⑧);柔紅黴素脂質 體(DanuoXome®);柔紅黴素,道諾黴素(daunomycin) (Daunorubicin®);柔紅黴素,道諾黴素(Cerubidine®);地尼 白介素 2(Denileukin diftitox)(Ontak®);右雷佐生 (dexrazoxane)(Zinecard@);多西他賽(docetaxel)(Taxotere⑧); 多柔比星(Adriamycin PFS®);多柔比星(Adriamycin®, Rubex®);多柔比星(Adriamycin PFS Injection®);多柔比 星脂質體(Doxil®);丙酸甲雄烧酮(Dromostanolone®);丙 酸甲雄烧酮(Masterone Injection®);愛立特氏(Elliott’s)B 溶液(Elliott’s B Solution®);表柔比星(epirubicin)(Ellence®);阿 法依伯汀(Epoetin alfa)(epogen⑧);埃羅替尼(erlotinib)(Tarceva⑧); 127700.doc -53- 200836731 雌莫司汀(estramustineKEmcyt®);鱗酸依託泊普(etoposide phosphate)(Etopophos@);依託泊普(etoposide) ,VP-^(Vepesid®);依西美坦(exemestaneKAromasin®);非格司 亭(FilgrastimXNeupogen®);氟尿苷(動脈内 KFUDR0);氟達拉 濱(fludarabine)(Fludara⑧);氟尿,咬,5-FU(Adrucil@); 說維司群 (fulvestrantXFaslodex®); 吉非替尼 (gefltinib)(Iressa⑧);吉西他濱(gemcitabine)(Gemzar⑧);吉姆 單抗奥佐米星(gemtuzumab ozogamicin)(Mylotarg⑧);乙酸 戈舍瑞林(goserelin acetate)(Zoladex Implant®);乙酸戈舍 瑞林(Zoladex®);乙酸組胺瑞林(histrelin acetate) (Histrelin implant®);經基脲(Hydrea@);替坦異貝莫單抗 (Ibritumomab TiuxetanMZevalin®);伊達比星(idarubicin) (Idamycin,;異環磷醯胺(IFEX®);甲磺酸伊馬替尼 (imatinib mesylateKGleevec®);干擾素a 2a(Roferon A@); 干擾素a-2b(Intron A®);伊立替康(Camptosar®);來那度 胺(lenalidomide)(Revlimid®);來曲唾(letrozoleXFemara®); 甲龜四氫葉酸(leucovorin)(Wellcovorin®,leucovorin®);乙酸亮 丙瑞林(Eligard®);左旋四 口米嗤(levamisole) (Ergamisol®); 洛莫司汀(lomustine),CCNU(CeeBU®);雙氯乙基曱胺 (meclorethamine),氮芬(Mustargen®);乙酸甲地孕酮 (Megace®);美法侖(melphalan),L-PAN^Alkeran®);疏基嗓 呤,ό-ΜΡΡιιιίι^ΙιοΙ®);美司納(mesna)(Mesnex®);美司鈉 (Mesnex tabs®);胺曱蝶吟(Methotrexate®);甲氧沙林 (methoxsalen)(Uvadex®);絲裂黴素(^(Mutamycin®);米托 127700.doc -54- 200836731Kim et al. (1993) 362:841-844; WO 00/44777; and WO 127700.doc-50-200836731 00/61186) other therapeutic agents that modulate or inhibit angiogenesis and which may also be used in combination with the compounds of the invention Includes agents that modulate or inhibit coagulation and fibrinolytic systems (see C7k. Ζα· Med. (2000) 38:679-692). Examples of such agents that modulate or inhibit coagulation and fibrinolytic pathways include, but are not limited to, heparin (see TTzrowZ® (1998) 80:10-23), low molecular weight heparin and carboxypeptidase U inhibitors ( Also known as active thrombin can activate the inhibitor of fibrinolysis inhibitor [TAFIa] (see (: and (2001) 101: 329-354). TAFIa inhibitors have been described in 卩 (: Ding public case 0 03/013, 526 and U.S. Patent No. 60/349, 925, filed Jan. 18, 2002, the disclosure of which is incorporated herein by reference. A compound that causes cancer cells to be sensitive to DNA damaging agents. These agents include inhibitors of ATR, ATM, Chkl, and Chk2 kinases, and cdk and cdc kinase inhibitors, and specific examples thereof are 7-synthesis staurosporin, star Sporomycin, flavopirid〇l, 〇CYC202 (Cyclacel) and BMS_387032. Inhibitors of π cell proliferation and survival signaling pathways, f, can inhibit fine cell surface receptors and these surface receptors. A pharmaceutical agent for downstream signal transduction pathways. Such agents include inhibitors of EGFR (eg, gefitinib and erlotinib), inhibitors of ERB-2 (eg, 'trastuzumab'), inhibition of IGFR Agents (for example, those disclosed in WO 03/05995 1), inhibitors of cytokine receptors, inhibitors of MET, inhibitors of PI3K (eg, LY294002), serine/threonine 127700. Doc • 51 - 200836731 Enzymes (including but not limited to inhibitors of Akt, as described, for example, in WO 03/086404, WO 03/086403, WO 03/086394, WO 03/086279, WO 02/083675, WO 02/083139, WO Inhibitors of Raf kinase (eg, BAY-43-9006), inhibitors of MEK (eg, CI-1040 and PD-098059), and inhibitors of mTOR (eg, Wyeth CCI-779 and Ariad AP23 5 73). These agents include small molecule inhibitor compounds and antibody antagonists. ''Apoptosis inducers' include activators of TNF receptor family members, including TRAIL receptors. In one embodiment, the compounds of the invention may be combined with one or more (specifically, one, two or Species) is selected from temozolomide, cisplatin, carboplatin, oxaliplatin, irinotecan and topotecan Kang medicament for the treatment of cancer. The compounds of the invention may also be used in combination with any one or more of the following therapeutic agents for the treatment of cancer: abarelix (Plenaxis depot®); aldesleukin MProkine^; Aldesleukin (Proleukin); Alemtuzumabb (Campath); altretinoin (KPanretin^); allopurinol (Zyloprim®); altretamine XHexalen®; amifostine (amifostine) Ethyol®); anastrozole XArimidex®; Trisenox®; aspartate (Elspar®); azacitidine (Vidaza@); bevacizimab XAvastin® ); bexarotene capsule (Targretin®); borarotin® (Bargoxin®); bleomycin (Blenoxane®); poltidan 127700.doc •52- 200836731 anti-bortezomib (Velcade®); Busulfex® for intravenous use; Myleran® for oral administration; caluterone XMethosarb®; capecitabine (Xeloda@); Carboplatin (Paraplatin) ®); Carmustine (BCNU®, BiCNU@); Carmustine (Gliadel®); Carmustine and Polyphenylene 20 implants (Gliadel Wafer®); Celebex®; cetuximab (Erbitux@); chlorambucil ( chlorambucilXLeukeran®); Platinol®; cladribine (Leustatin®, 2-CdA@); clofarabine (Clolar@); Cytoxan®, Neosar® Cytoxan Injection8; Cytoxan Tablet®; Cytosar-U®; DepoCyt®; Dcarb-Dome® ); dactinomycin, actinomycin DXCosmegen®; Darbepoetin alfa (Aranesp8); daunorubicin liposome (DanuoXome®); daunorubicin, daunro Daunomycin (Daunorubicin®); daunorubicin, Cerubidine®, Denileukin diftitox (Ontak®); dexrazoxane (Zinecard@); Dorsey Docetaxel (Taxotere8); Adriamycin PFS®; Adriamycin® (Rulex®); Doxorubicin (Adriamycin PFS Inj) Scope®); Doxil®; Dromostanolone®; Masterone Injection®; Elliott's B solution (Elliott's B) Solution®); epirubicin (Ellence®); Epoetin alfa (epogen8); erlotinib (Tarceva8); 127700.doc -53- 200836731 espresso Ester (estramustine KEmcyt®); etoposide phosphate (Etopophos@); etoposide, VP-^ (Vepesid®); exemestane (exemestane KAromasin®); filgrastim (FilgrastimXNeupogen) ®); fluorouridine (intra-arterial KFUDR0); fludarabine (Fludara8); fluorourine, bite, 5-FU (Adrucil@); vesistan (fulvestrantXFaslodex®); gefitinib ( Gefltinib) (Iressa8); gemcitabine (Gemzar8); gemtuzumab ozogamicin (Mylotarg8); goserelin acetate (Zoladex Implant®); goserelin acetate ( Zoladex®); histrelin acetate (Histreli) n implant®); ureua (Hydrea@); tiltanebezumab (Ibritumomab Tiuxetan MZevalin®); idarubicin (Idamycin,; ifosfamide (IFEX®); mesylate Imatinib mesylate KGleevec®; interferon a 2a (Roferon A@); interferon a-2b (Intron A®); irinotecan (Camptosar®); lenalidomide (Revlimid®); Letrozole XFemara®; leucovorin (Wellcovorin®, leucovorin®); leuprolide acetate (Eligard®); levamisole (Ergamisol®); lomustine (lomustine), CCNU (CeeBU®); meclorethamine, Mustargen®; megace® acetate; melphalan, L-PAN^Alkeran® ); 嗓呤基嗓呤,ό-ΜΡΡιιιίι^ΙιοΙ®); mesna (Mesnex®); Mesnex tabs®; Methotrexate®; methoxalin (Uvadex®); mitomycin (^(Mutamycin®); mitto 127700.doc -54- 200836731
坦(mitotane)(Lysodren@);米托蒽 g 昆(mitoxantrone) (Novantrone0);苯丙酸諾龍(nandrolone phenpropionate) (Durabolin-50®);耐拉濱(nelarabine)(Arranon⑧);諾非單 抗(Nofetumomab)(Verluma⑨);奥普瑞白介素(Oprelvekin) (Neumega®);奥沙利舶(Eloxatin@);紫杉醇(Paxene⑧);紫 杉醇(Taxol®);紫杉醇蛋白質結合顆粒(Abraxane®);帕利 夫明(paliferminXKepivance®);帕瑪二磷酸(pamidronate) (Aredia®);培加酶(pegademase) (Adagen(Pegademase Bovine)®);培加帕酶(pegaspargase) (Oncaspar⑧);培非司 亭(Pegnigrastim)(Neulasta®);培美曲塞二納(pemetrexed disodiumXAlimta®);喷司他丁(Nipent®) ; 口辰泊溴烧 (pipobroman)(Vercyte@);普利黴素(plicamycin),光輝黴 素(mithramycin)(Mithracin®) ; 口卜吩姆納(porfimer sodium) (Photofrin®);丙卡巴肼(Matulane®);奎納克林 (quinacrine)(Atabrine@);拉布立酶(RasburicaseKElitek®);利 妥昔單抗(Rituximab) (Rituxan0) 沙格司亭 (sargramostim)(Leukinec§));沙格司亭(Prokine®);索拉非 尼(sorafenib)(Nexavar®);鏈脲黴素(streptozocin) (Zanosar0);馬來酸舒尼替尼(sunitinib maleate) (Sutent®);滑石粉(Sclerosol®);他莫昔芬(Nolvadex®);替 莫峻胺(Temodar®);替尼泊苦(teniposide) ,VM-26(Vumonc§));睾内酯(Teslac®);硫鳥嘌呤,6-TG(Thioguanine@) ; σ塞替派(thiotepa) (Thioplex@);拓撲替 康(Hycamtin®);托瑞米芬(toremifene) (Fareston®);托西 127700.doc -55- 200836731 莫單抗(Tositumomab) (Bexxar®);托西莫單抗/1-13 1托西 莫單抗(Bexxar⑧);曲司佐單抗(Trastuzumab)(Herceptin®); 維 A 酸’ ATRA(Vesanoid®);尿嘴咬氮芬(Uracil Mustard Capsules®);伐蘆比星(valrubicin)(Valstar@);長春花鹼 (Velban®);長春新鹼(Oncovin®);長春瑞濱(Navelbine ⑧); 福瑞斯達(vorinostat)(Zolinza@)、 唾來膦酸鹽 (zoledronate)(Zometa@);尼勒替尼(nii〇tinib)(Tasigna⑧)及 達沙替尼(dasatinibRSprycel®)。Mitotane (Lysodren@); mitoxantrone (Novantrone0); nandrolone phenpropionate (Durabolin-50®); nelarabine (Arranon8); Nofetumomab (Verluma9); Oprelvekin (Neumega®); Oxali@ (Eloxatin@); Paclitaxel (Paxene8); Paclitaxel (Taxol®); Paclitaxel Protein Binding Particles (Abraxane®); paliferminXKepivance®; pamidronate (Aredia®); pegademase (Adagen (Pegademase Bovine)®); pegaspargase (Oncaspar8); pefibrate ( Pegnigrastim) (Neulasta®); pemetrexed disodium XAlimta®; pentastatin (Nipent®); pipobroman (Vercyte@); plicamycin, radiance Mithracin (Mithracin®); porfimer sodium (Photofrin®); procalaquinone (Matulane®); quinacrine (Atabrine@); Rabrizyme (RasburicaseKElitek) ®); Rituximab (Rituxan0) Shags Sargramostim (Leukinec§)); sagstatin (Prokine®); sorafenib (Nexavar®); streptozocin (Zanosar0); sunitinib maleate ( Sunitinib maleate) (Sutent®); stalrosol®; tamoxifen®; Temodar®; teniposide, VM-26 (Vumonc§); Testosterone (Teslac®); thioguanine, 6-TG (Thioguanine@); σ thiotepa (Thioplex@); topotecan (Hycamtin®); toremifene (Fareston®) ); Tosi 127700.doc -55- 200836731 Tositumomab (Bexxar®); tosimozumab/1-13 1 tositumomab (Bexxar8); trastuzumab (Trastuzumab) Herceptin®); Atra (Vesanoid®); Uracil Mustard Capsules®; valrubicin (Valstar@); vinblastine (Velban®); vincristine ( Oncovin®); Navelbine 8; vorinostat (Zolinza@), zoledronate (Zometa@); nii〇tinib (Tasigna8) and Dasatini bRSprycel®).
U 本發明亦涵蓋與作為選擇性COX-2抑制劑之NS AID的組 合。就本說明書而言,將作為COX-2選擇性抑制劑之 NSAID定義為彼等與對cox—!抑制特異性相比對c〇X-2具 有至少100倍抑制特異性的物質,該抑制特異性藉由細胞 或微粒體試驗所評定C0X-22與c〇x_丨之IC〜的比例量 測。此等化合物包括但不限於彼等揭示於美國專利 5,474,995、4 國專利 5,861,419、美國專利 Mqi,843、美 國專利6,020,343、美國專利5,4〇9,944、美國專利 5,436,265、λ 國專利 5,536,752、美國專利 5,55(),142、美 國專利5,6〇4,26〇、美國專利5,69M84、美國專利 5,71〇,14〇、购 94/15932、美國專利 5,344,991、美國專利 5,134,142、λ國專利5,38q,738、美國專利$,州,彻 '美 國專利5,466,823、美國專利5,633,272及美國專利 5,932,598中者,所有該等案件均以引用方式併人本文中。 尤其可用於本發明治療方法之鹽_2抑制劑係%氣_心 甲基磺醯基)苯基_2·(2_甲其ς ^ &盆、 (甲基-5-吡啶基)吡啶或其醫藥上可 127700.doc -56· 200836731 接受之鹽。 作為COX-2特異性抑制劑闡述且因此可用於本發明之化 合物包括但不限於··帕瑞考昔(parec〇xib)、celebrex0及 BEXTRA或其醫藥上可接受之鹽。 血管生成抑制劑之其他實例包括但不限於血管内皮抑 素、ukrain、豹虫圭酶(ranpirnase)、IM862、5-甲氧基 _4-[2_ •曱基-3-(3-曱基-2-丁烯基)環氧乙烷基氧雜螺[2,5]辛_6_ 基(氣乙酸基)胺基甲酸酯、乙醯基地那林 (acetyldinaline)、5-胺基 _1_[[3,5_ 二氯-4-(4-氣苯甲醯基)苯 基]甲基]-1汉-1,2,3-三唑-4-甲醯胺、CM101、角鯊胺、康 布瑞塔卡汀(combretastatin)、RPI4610、NX3 1 838、硫酸化 甘露戊糖磷酸鹽、7,7-(羰基-雙[亞胺基-7V-甲基-4,2-吡咯基 羰基亞胺基[7V-甲基-4,2-吡咯]-羰基亞胺基]-雙_(1,3-萘二 磺酸酯)、及3-[(2,4-二甲基吡咯-5-基)亞甲基]-2-二氫吲哚 酮(SU5416) 〇 本文所用”整合素阻滯劑”係指可選擇性地拮抗、抑制或 對抗生理配體與ay β3整合素結合之化合物,可選擇性地括 抗、抑制或對抗生理配體與ανβ5整合素結合之化合物,可 , 拮抗、抑制或對抗生理配體與ανβ3整合素及ανβ5整合素 .一者結合之化合物,及可拮抗、抑制或對抗在毛細jk管内 皮細胞上表現之特殊整合素之活性的化合物。該術語亦指 ανβ6、ανβ8、αΐβι、〇〇2βΐ、α5βΐ、α6βΐ 及 α6β4 整合素之 拮抗劑。該術語亦指 ανβ3、ανβ5, ανβ6、ανβ8、αιβι、 α2βΐ、β5〇Π、α6βΐ及α6β4整合素之任一組合的拮抗劑。 127700.doc -57- 200836731 酪胺酸激酶抑制劑之某些具體實例包括TV-(三氟甲基笨 基)-5-甲基異嚼唾甲醯胺、3_[(2,4_二甲基吡咯-5_基)亞 甲基)二氫。引哚|酮、η·(稀丙基胺基)_n_去甲氧基袼爾 德黴素、4-(3-氣-4-氟苯基胺基)_7_甲氧基_6-[3 —(‘嗎啉基) 丙氧基]喹唑啉、Y-(3-乙炔基苯基)_6,7·雙(2-甲氧基乙氧 基)-4_喹唑啉胺、ΒΙΒΧ1382、^^仏六氫^^羥 基甲基)-10-羥基-9·甲基_9,12-環氧-1/^二吲哚并π,2,3_ fg:3’,2’,l’_kl]。比咯并苯并二氮雜環辛小酮、 SH268、染料木黃酮(genistein)、STI571、CEp2563、4 氯苯基胺基)-5,6-二甲基-7f吡咯并[2,3-d]嘧啶甲烷磺酸 酯、4-(3-溴-4-羥基苯基)胺基-6,7_二甲氧基喹唑啉、4_(4,_ 經基本基)胺基-6,7-二甲氧基喧嗤琳、SU6668、 STI571A、氣苯基-4·(4-吡啶基甲基)呔嗪胺、及 EMD121974 〇 在一實施例中,本發明之化合物可用於治療或預防由諸 如 MeOS02(CH2)-lexitropsin (Me-Lex)等選擇性 #3-腺嗓 a 甲基化試劑引發之壞死出現。 本發明方法亦涵蓋與除抗癌化合物外之化合物的組合。 舉例而言,本發明所主張化合物與PPAR-γ (即,ΡΡΑκ_γ) 激動劑及PPAR-δ (即,PPAR-δ)激動劑之組合可用於治療 某些惡性腫瘤。PPAR-γ及PPAR-δ係核過氧化物酶體增殖 子激活之受體γ及δ。ΡΡ AR-γ在内皮細胞上之表現及其在血 管生成中之參與已經報道於文獻中(參見乂 Car Pharmacol. (1998) 31:909-913; J. Biol. Chem (1999) 127700.doc • 58 - 200836731 274:9116-9121; Invest, Ophthalmol Vis · Sci, (2000) 41:2309-2317)。最近,已經證實PPAR-γ激動劑在活體内 可抑制VEGF之血管生成反應;曲格列酮(troglitazone)及羅 格列酮(rosiglitazone)馬來酸鹽二者在小鼠中均可抑制視網 膜新血管形成的發展。(drc/2.0p/^/mmo/.(2001) 119:709-7 1 7)。PPAR-γ激動劑及PPAR- γ/α激動劑之實例包括但不 限於噻唑啶二酮類(例如,DRF2725、CS-011、曲格列 酮、羅格列酮、及σ比格列酮(pioglitazone))、非諾貝特 (fenofibrate)、吉非貝齊(gemfibrozil)、氣貝 丁酉旨 (clofibrate)、GW2570、SB219994、AR-H039242、JTT-501、MCC-555、GW2331、GW409544、NN2344、KRP297、 NP0110、DRF4158、NN622、GI262570、PNU182716、 DRF552926、2-[(5,7-二丙基三氟甲基-1,2-苯并異噁唑-6-基)氧基]-2-甲基丙酸(揭示於USSN 09/782,856中)、及 2(7?)·7·(3-(2-氯-4-(4-氟苯氧基)苯氧基)丙氧基)-2-乙基苯 并二氫吡喃-2_甲酸(揭示於USSN 60/23 5,708及 60/244,697 中)。 本發明之另一實施例係本發明所揭示化合物與用於癌症 治療之抗病毒劑(例如,核苷類似物,包括更昔洛韋 (ganciclovir))之組合的用途。參見 WO 98/04290。 本發明之另一實施例係本發明所揭示化合物與用於癌症 治療之基因治療之組合的用途。關於治療癌症之基因策 略,參見Hall 等人(dm Ge ⑽ ί (1997) 61:785-789)及U The present invention also encompasses combinations with NS AIDs as selective COX-2 inhibitors. For the purposes of this specification, an NSAID that is a COX-2 selective inhibitor is defined as a substance that has at least 100-fold inhibition specificity for c〇X-2 compared to cox-! The ratio of C0X-22 to c〇x_丨 IC~ was measured by cell or microsomal assay. Such compounds include, but are not limited to, those disclosed in U.S. Patent No. 5,474,995, U.S. Patent No. 5,861,419, U.S. Patent No. 5, 063, U.S. Patent No. 6,020,343, U.S. Patent No. 5,4,9,944, U.S. Patent No. 5,436,265, U.S. Patent No. 5,536,752, U.S. Patent 5,55(), 142, U.S. Patent 5,6,4,26, U.S. Patent 5,69 M84, U.S. Patent 5,71,14,14,94,932, U.S. Patent 5,344,991, U.S. Patent 5,134 </ RTI> <RTIgt; </ RTI> <RTIgt; </ RTI> <RTIgt; </ RTI> <RTIgt; </ RTI> <RTIgt; </ RTI> <RTIgt; </ RTI> <RTIgt; </ RTI> <RTIgt; </ RTI> </ RTI> </ RTI> </ RTI> </ RTI> </ RTI> <RTIgt; In particular, the salt 2 inhibitor which can be used in the treatment method of the present invention is % gas-methylsulfonyl)phenyl-2·(2-methylpyrazine ^ & pot, (methyl-5-pyridyl)pyridine Or a pharmaceutically acceptable salt thereof 127700.doc -56·200836731. Compounds which are described as COX-2 specific inhibitors and are therefore useful in the present invention include, but are not limited to, parecoxib (celecbrex), celebrex0 And BEXTRA or a pharmaceutically acceptable salt thereof. Other examples of angiogenesis inhibitors include, but are not limited to, endostatin, ukrain, ranpirnase, IM862, 5-methoxy_4-[2_ • Mercapto-3-(3-mercapto-2-butenyl)oxiranyloxaspiro[2,5]octyl-6-yl (gasacetic acid) urethane, acetamidine (acetyldinaline), 5-amino-1—[[3,5-dichloro-4-(4-carbobenzyl)phenyl]methyl]-1 han-1,2,3-triazole-4- Formamide, CM101, squalamine, combretastatin, RPI4610, NX3 1 838, sulfated mannose pentose phosphate, 7,7-(carbonyl-bis[imino-7V-A Base-4,2-pyrrolylcarbonylimino[7V-methyl-4,2-pyrrole]-carbonylimino] -bis-(1,3-naphthalene disulfonate), and 3-[(2,4-dimethylpyrrol-5-yl)methylene]-2-indanone (SU5416) As used herein, an "integrin blocker" refers to a compound that selectively antagonizes, inhibits or counteracts the binding of a physiological ligand to ay beta 3 integrin, and optionally inhibits, inhibits or counteracts the binding of a physiological ligand to an ανβ5 integrin. a compound, a compound that antagonizes, inhibits or inhibits the binding of a physiological ligand to ανβ3 integrin and ανβ5 integrin, and a compound that antagonizes, inhibits or counteracts the activity of a particular integrin expressed on capillary jk endothelial cells The term also refers to antagonists of ανβ6, ανβ8, αΐβι, 〇〇2βΐ, α5βΐ, α6βΐ and α6β4 integrin. The term also refers to ανβ3, ανβ5, ανβ6, ανβ8, αιβι, α2βΐ, β5〇Π, α6βΐ and α6β4 integration. Antagonists of any combination of the compounds. 127700.doc -57- 200836731 Some specific examples of tyrosine kinase inhibitors include TV-(trifluoromethylphenyl)-5-methyl chewing salicylamine, 3_[(2,4_Dimethylpyrrole-5-yl)methylene)哚 哚 | ketone, η · (dilylpropyl) _n_ demethoxy aldostermycin, 4- (3- gas-4-fluorophenylamino)_7_methoxy _6- [3-('morpholinyl)propoxy]quinazoline, Y-(3-ethynylphenyl)-6,7-bis(2-methoxyethoxy)-4-quinazolinamine, ΒΙΒΧ1382,^^仏 hexahydro^^hydroxymethyl)-10-hydroxy-9·methyl_9,12-epoxy-1/^diindole π,2,3_fg:3',2', L'_kl]. Prodoxobenzodiazepine ketone, SH268, genistein, STI571, CEp2563, 4 chlorophenylamino)-5,6-dimethyl-7fpyrrolo[2,3- d] pyrimidine methane sulfonate, 4-(3-bromo-4-hydroxyphenyl)amino-6,7-dimethoxyquinazoline, 4_(4,_ via basic)amino-6, 7-Dimethoxy phthalocyanine, SU6668, STI571A, phenylphenyl-4(4-pyridylmethyl)pyridazinamine, and EMD121974 〇 In one embodiment, the compounds of the invention are useful for the treatment or prevention Necrosis induced by a selective #3-adenine methylation reagent such as MeOS02(CH2)-lexitropsin (Me-Lex) occurs. Combinations of compounds other than anti-cancer compounds are also contemplated by the methods of the invention. For example, a combination of a compound of the invention and a PPAR-y (i.e., ΡΡΑκ_γ) agonist and a PPAR-δ (i.e., PPAR-delta) agonist can be used to treat certain malignancies. PPAR-γ and PPAR-δ are nuclear peroxisome proliferator-activated receptors γ and δ. The expression of AR-γ on endothelial cells and its involvement in angiogenesis has been reported in the literature (see 乂Car Pharmacol. (1998) 31:909-913; J. Biol. Chem (1999) 127700.doc • 58 - 200836731 274:9116-9121; Invest, Ophthalmol Vis · Sci, (2000) 41:2309-2317). Recently, it has been confirmed that PPAR-γ agonists can inhibit the angiogenic response of VEGF in vivo; both troglitazone and rosiglitazone maleate can inhibit retinal neoplasia in mice. The development of angiogenesis. (drc/2.0p/^/mmo/. (2001) 119:709-7 1 7). Examples of PPAR-gamma agonists and PPAR-gamma/alpha agonists include, but are not limited to, thiazolidinediones (eg, DRF2725, CS-011, troglitazone, rosiglitazone, and sigma-glitazone ( Pioglitazone)), fenofibrate, gemfibrozil, clofibrate, GW2570, SB219994, AR-H039242, JTT-501, MCC-555, GW2331, GW409544, NN2344, KRP297, NP0110, DRF4158, NN622, GI262570, PNU182716, DRF552926, 2-[(5,7-dipropyltrifluoromethyl-1,2-benzisoxazole-6-yl)oxy]-2- Methylpropionic acid (disclosed in USSN 09/782,856), and 2(7?)·7·(3-(2-chloro-4-(4-fluorophenoxy)phenoxy)propoxy)- 2-ethylchroman-2 -carboxylic acid (disclosed in USSN 60/23 5,708 and 60/244,697). Another embodiment of the invention is the use of a compound of the invention in combination with an antiviral agent for cancer therapy (e.g., a nucleoside analog, including ganciclovir). See WO 98/04290. Another embodiment of the invention is the use of a compound of the invention in combination with gene therapy for cancer therapy. For a genetic strategy for treating cancer, see Hall et al. (dm Ge (10) ί (1997) 61:785-789) and
Kufe 等人(C⑽cer ΜΜζϋβ,第 5 版,第 876-889 頁,BC 127700.doc -59- 200836731Kufe et al. (C(10)cer ΜΜζϋβ, 5th edition, pp. 876-889, BC 127700.doc -59- 200836731
Decker,Hamilton 2000)。基因治療可用於遞送任一腫瘤抑 制基因。此等基因之實例包括但不限於可藉由重組病毒介 導之基因轉移遞送的p53(例如,參見美國專利第6,069,134 號)、uPA/uPAR拮抗劑(’’Adenovirus-Mediated Delivery of a uPA/uPAR Antagonist Suppresses Angiogenesis-Dependent Tumor Growth and Dissemination in Mice,11 Gene Therapy, August (1998) 5(8):1105-13)及干擾素 γ 加 / (2000) 164:217- 222) ° 本發明之化合物亦可與固有多重藥物抗性(MDR)之抑制 劑組合投與,特定言之,MDR與轉運蛋白之高水平表現有 關。此等MDR抑制劑包括p-糖蛋白(P-gp)抑制劑,例如, LY335979、XR9576、OC144-093、R101922、VX853、維 拉帕米(verapamil)及PSC833 (伐司朴達(valspodar))。 本發明之化合物可結合鎮吐劑單獨或與放射治療一起使 用以治療σ惡心或u區吐,包括可由本發明之化合物使用引起 的急性、延遲、後期及期待性嘔吐。為了預防或治療嘔 吐,本發明之化合物可結合其他鎮吐劑使用,該等鎮吐劑 尤其為神經激肽-1受體拮抗劑、5HT3受體拮抗劑(例如, 昂丹司瓊(ondansetron)、格拉司缓(granisetron)、托烧司填 (tropisetron)、及紮托司瓊(zatisetron))、GABAB 受體激動 劑(例如,巴氯芬(baclofen))、皮質類固醇(例如, Decadron (地塞米松)、Kenalog、Aristocort、Nasalide、 Preferid、Benecorten)或其他(例如,揭示於美國專利第 2,789,118 號、第 2,990,401 號、第 3,048,581 號、第 127700.doc -60- 200836731 3’126’375 號、第 3,929,768 號、第 3,996,359 號、第 3,92 8,326 號及第 3 749 71? 口告 * 土、 ^ ^ 5 ,712就中者)、抗多巴胺劑,例如, 吩噻嗪類(例如,丙氯拉嗪(prochlorperazine)、氟奮乃靜 (flUphenaZlne)、硫利噠嗪(thioridazine)及美索達嗪 (mesoridazine))、甲氧氯普胺(met〇ci〇pramide)或屈大麻紛 (dronabinol)。在一實施例中,投與選自神經激肽_丨受體拮 抗劑、5HT3受體拮抗劑及皮質類固醇之鎮吐劑作為佐劑 以治療或預防可由本發明化合物投藥引起的嘔吐。 結合本發明化合物使用的神經激肽_ i受體拮抗劑全面地 闡述於(例如)美國專利第5,162,339號、第5,232,929號、第 5,242,930 號、第 5,373,003 號、第 5,387,595 號、第 5,459,270 號、第 5,494,926 號、第 5,496,833 號、第 5,637,699號、第5,719,147;歐洲專利公開案第EP 0 360 390號、第 0 394 989號、第 0 428 434號、第 0 429 366號、 第 0 430 771號、第 0 436 334號、第 0 443 132號、第 0 482 539號、第 0 498 069號、第 0 499 313號、第 0 512 901 號、 第 0 512 902號、第 0 514 273號、第 〇 514 274號、第 0 514 275號、第 0 514 276號、第 〇 515 681號、第 0 517 589號、 第 0 520 555號、第 〇 522 808號、第 〇 528 495號、第 〇 532 45ό號、第 0 533 280號、第 〇 536 817號、第 0 545 478號、 第 0 558 156號、第 〇 577 394號、第 〇 585 913號、第 〇 590 152號、第 0 599 538號、第 〇 610 793號、第 0 634 402號、 第 0 086 629號、第 〇 693 489號、第 0 694 535號、第 〇 699 655號、第 0 099 074號、第〇 7〇7 〇〇6號、第 〇 708 1〇1 號、 -61 - 127700.doc 200836731 第 0 709 375號、第 0 709 376號、第 0 714 891號、第 0 723 959號、第0 733 632號及第0 776 893號;PCT國際專利公 開案第 WO 90/05525 號、第 90/05729號、第 91/09844號、 第 91/18899 號、第 92/01688 號、第 92/06079 號、第 92/12151 號、第 92/15585 號、第 92/17449 號、第 92/20661 號、第 92/20676號、第 92/21677號、第 92/22569號、第 93/00330 號、第 93/00331 號、第 93/01 159 號、第 93/01 165 號、第 93/01 169號、第 93/01 170號、第 93/06099號、第 93/091 16 號、第 93/10073 號、第 93/14084 號、第 93/14113 號、第 93/18023 號、第 93/19064號、第 93/21 155 號、第 93/21181 號、第 93/23380 號、第 93/24465 號、第 94/00440 號、第 94/01402號、第 94/02461 號、第 94/02595 號、第 94/03429 號、第 94/03445 號、第 94/04494 號、第 94/〇4496 號、第 94/05625 號、第 94/07843 號、第 94/08997號、第 94/10165 號、第 94/10167 號、第 94/10168 號、第 94/10170 號、第 94/1 1368 號、第 94/13639號、第 94/13663 號、第 94/14767 號、第 94/15903 號、第 94/19320 號、第 94/19323 號、第 94/20500號、第 94/26735 號、第 94/26740號、第 94/29309 號、第 95/02595 號、第 95/04040 號、第 95/04042 號、第 95/06645 號、第 95/07886 號、第 95/07908 號、第 95/08549 號、第 95/1 1880 號、第 95/14017 號、第 95/153 1 1 號、第 95/16679號、第 95/17382號、第 95/18124號、第 95/18129 號、第 95/19344 號、第 95/20575 號、第 95/21819 號、第 95/22525 號、第 95/23798 號、第 95/26338 號、第 95/28418 127700.doc -62- 200836731 號、第 95/30674 號、第 95/30687 號、第 95/33744 號、第 96/05181 號、第 96/05193 號、第 96/05203 號、第 96/06094 號、第 96/07649 號、第 96/10562 號、第 96/16939 號、第 96/18643 號、第 96/20197號、第 96/21661 號、第 96/29304 號、第 96/29317 號、第 96/29326 號、第 96/29328 號、第 96/31214號、第 96/32385 號、第 96/37489號、第 97/01553 號、第 97/01554 號、第 97/03066 號、第 97/08144 號、第 97/14671 號、第 97/17362號、第 97/18206號、第 97/19084 號、第97/19942號及第97/21702號;及英國專利公開案第2 266 529號、第 2 268 931號、第 2 269 170號、第 2 269 590 號、第 2 271 774號、第 2 292 144號、第 2 293 168號、第 2 293 169號、及第2 302 689號中。此等化合物之製備全面 地闡述於上述專利案及公開案中,該等案件均以引用方式 併入本文中。 在一實施例中,結合本發明化合物使用的神經激肽-1受 體拮抗劑係選自:闡述於美國專利第5,719,147號中之2_ (卜(7?)·(3,5-雙(三氟甲基)苯基)乙氧基)-3-(S)-(4-氟苯 基)-4-(3-(5-氧代基-1//,4开-1,2,心三唑基)曱基)嗎啉或其醫 藥上可接受之鹽。 本發明之化合物亦可與用於貧血治療之藥劑一起投與。 此貧血治療劑係(例如)連續紅血球生成受體激活劑(例如, 阿法依伯汀)。 本發明之化合物亦可與用於嗜中性白血球減少症治療之 藥劑一起投與。此嗜中性白血球減少症治療劑係(例如)可 127700.doc •63 - 200836731 調節嗜中性粒細胞生成及功能之造血生長因子,例如,人 類粒細胞集落刺激因子(G-CSF)。G-CSF之實例包括非格 司亭。 本發明之化合物亦可與諸如左旋咪。坐(levamisole)、異丙 肌普(isoprinosine)及日達仙(Zadaxin)等免疫增強藥一起投 與。 本發明之化合物亦可與雙膦酸鹽類(應理解為包括雙膦 酸鹽類、二膦酸鹽類、雙膦酸類及二膦酸類)組合以用於 治療或預防癌症,包括骨癌。雙膦酸鹽類之實例包括但不 限於:依替膦酸鹽(Didronel)、 帕米膦酸鹽 (pamidronate)(Aredia)、阿倫膦酸鹽(alendronate)(Fosamax)、 利塞膦酸鹽(risedronate)(Actonel)、ϋ坐來膦酸鹽 (zoledronate)(Zometa)、伊班膦酸鹽(ibandronate)(Boniva)、英 卡膦酸(incadronate)或斯孟膦酸鹽(cimadronate)、氯膦酸 鹽(clodronate)、EB-1053、米諾膦酸鹽(minodronate)、奈 力膦酸鹽(neridronate)、吼膦酸鹽(piridronate)及替魯膦酸 鹽(tiludronate),包括任一及所有其醫藥上可接受之鹽、 衍生物、水合物及混合物。 因此,本發明之範圍涵蓋本發明所主張化合物與電離輻 射及/或與選自下列之第二化合物之組合的用途:HDAC抑 制劑、雌激素受體調節劑、雄激素受體調節劑、類視色素 受體調節劑、細胞毒性/細胞生長抑制劑、抗增生劑、異 戊二烯基-蛋白轉移酶抑制劑、HMG-CoA還原酶抑制劑、 血管生成抑制劑、PPAR-γ激動劑、PPAR-δ激動劑、抗病 127700.doc -64- 200836731 毋劑、固有多重藥物抗性之抑制劑、鎮吐劑、用於貧血治 療之藥劑、用於嗜中性白血球減少症治療之藥劑、免疫增 強藥、細胞增生及存活信號傳導之抑制劑、干擾細胞週期 松查點之藥劑、細胞凋亡誘導劑及雙膦酸鹽。 與本發明化合物有關之術語,,投與”及其變體(例如,,,投 與’’化合物)意指將該化合物或該化合物之前藥引入需要治 療動物之系統中。當本發明化合物或其前藥與一種或多種 其他活性劑(例如,細胞毒性劑等)組合提供時,”投與”及 ,變體各自應理解為包括同時及相繼引人該化合物或其前 藥與其他藥劑。 本文所用術5吾組合物,,意欲涵蓋包含規定量的規定成份 X及任何可自規定量的規定成份之組合直接或間接 產生之產品。 本文所用術語”治療有效量"意指活性化合物或醫藥試劑 在組織、系統、動物或人類中產生生物或醫學反應之量, ’、係由研究者、獸醫師、醫師或其他臨床醫師來尋求。 術::/σ療係、指受病理病況感染之哺乳動物的治療且係 旨可藉由殺傷癌細胞來減輕該病況之作用而且亦指達成該 ^兄進展抑制之作m括進展速率降低、進展速率停 、病況改善及病況治癒。亦包括作為預防措施之 (即,預防)。 用術w藥上可接受之"係指在合理的藥學判斷 :性二於與受試者(例如,人類)組織接觸使用而無過高 、眭過敏性反應或其他問題或併發症並與合理 127700.doc -65- 200836731 的效益/風哈丄& L + 剞。夂哉 帛相應之化合物、材#、組合物及/或劑 1㈣形劑等就與該調配物之其他成份相容之意 義上而吕亦必須為,,可接受的,,。 :甫助係、指化合物結合已知治療方法之使用。該等 括不同癌症類型治療所用藥物之細胞毒性方式及/ 或電離輻射。牿 ^ 、5之,已知该荨活性化合物可強化許多 癌症化學治疼夕从^_ ^ 作用,該專活性化合物包括拓撲異構酶類 別之毋藥(例^^,士Decker, Hamilton 2000). Gene therapy can be used to deliver any tumor suppressor gene. Examples of such genes include, but are not limited to, p53 which can be delivered by recombinant virus-mediated gene transfer (for example, see U.S. Patent No. 6,069,134), uPA/uPAR antagonists (''Adenovirus-Mediated Delivery of a uPA) /uPAR Antagonist Suppresses Angiogenesis-Dependent Tumor Growth and Dissemination in Mice, 11 Gene Therapy, August (1998) 5(8): 1105-13) and Interferon Gamma Plus / (2000) 164:217- 222) ° The present invention Compounds can also be administered in combination with inhibitors of intrinsic multidrug resistance (MDR), in particular, MDR is associated with high levels of transporter expression. Such MDR inhibitors include p-glycoprotein (P-gp) inhibitors, for example, LY335979, XR9576, OC144-093, R101922, VX853, verapamil, and PSC833 (valspodar) . The compounds of the present invention may be combined with an antiemetic agent, alone or in combination with radiation therapy, to treat sigma nausea or ovulation, including acute, delayed, late, and expectant vomiting that may result from the use of a compound of the invention. For the prevention or treatment of vomiting, the compounds of the invention may be used in combination with other antiemetic agents, especially neurokinin-1 receptor antagonists, 5HT3 receptor antagonists (eg, ondansetron, gera) Granisetron, tropisetron, and zatisetron, GABAB receptor agonists (eg, baclofen), corticosteroids (eg, Decadron (dexamethasone) ), Kenalog, Aristocort, Nasalide, Preferid, Benecorten) or others (for example, disclosed in U.S. Patent Nos. 2,789,118, 2,990,401, 3,048,581, 127700.doc-60-200836731 3'126'375, Nos. 3, 929, 768, 3, 996, 359, 3, 92 8, 326 and 3 749 71? (in the case of ^, 5, 712), anti-dopamines, for example, phenothiazines (eg, propyl chloride) Prochlorperazine, flUphenaZlne, thioridazine and mesoridazine, met〇ci〇pramide or dronabinol . In one embodiment, an anti-emetic agent selected from the group consisting of a neurokinin-丨 receptor antagonist, a 5HT3 receptor antagonist, and a corticosteroid is administered as an adjuvant to treat or prevent vomiting caused by administration of the compound of the present invention. The neurokinin-i receptor antagonists used in connection with the compounds of the present invention are described in, for example, U.S. Patent Nos. 5,162,339, 5,232,929, 5,242,930, 5,373,003, 5,387,595, 5,459,270, No. 5, 494, 926, No. 5, 496, 833, No. 5, 637, 699, No. 5, 719, 147; European Patent Publication No. EP 0 360 390, No. 0 394 989, No. 0 428 434, No. 0 429 366, No. 0 430 771 No. 0 436 334, 0 443 132, 0 482 539, 0 498 069, 0 499 313, 0 512 901, 0 512 902, 0 514 273, 〇 514 274, 0 514 275, 0 514 276, 〇 515 681, 0 517 589, 0 520 555, 522 808, 528 495, 〇 532 45 、, 0 533 280, 〇 536 817, 0 545 478, 0 558 156, 〇 577 394, 〇 585 913, 〇 590 152, 0 599 538 No. 610 793, No. 0 634 402, No. 0 086 629, No. 693 489, 0 694 535, 〇 699 655, 0 099 074, 〇7〇7 〇〇 6, 〇 708 1〇1, -61 - 127700.doc 200836731 No. 0 709 375, No. 0 709 376, 0 714 891, 0 723 959, 0 733 632 and 0 776 893; PCT International Patent Publication Nos. WO 90/05525, 90/05729, 91/09844 No. 91/18899, 92/01688, 92/06079, 92/12151, 92/15585, 92/17449, 92/20661, 92/20676, Nos. 92/21677, 92/22569, 93/00330, 93/00331, 93/01 159, 93/01 165, 93/01 169, 93/01 170 No. 93/06099, 93/091 16 , 93/10073, 93/14084, 93/14113, 93/18023, 93/19064, 93/21 155 No. 93/21181, 93/23380, 93/24465, 94/00440, 94/01402, 94/02461, 94/02595, 94/03429, Nos. 94/03445, 94/04494, 94/〇4496, 94 /05625, 94/07843, 94/08997, 94/10165, 94/10167, 94/10168, 94/10170, 94/1 1368, 94/ No. 13639, Nos. 94/13663, 94/14767, 94/15903, 94/19320, 94/19323, 94/20500, 94/26735, 94/26740 , Nos. 94/29309, 95/02595, 95/04040, 95/04042, 95/06645, 95/07886, 95/07908, 95/08549, 95/1 1880, 95/14017, 95/153 1 1 , 95/16679, 95/17382, 95/18124, 95/18129, 95/19344, Nos. 95/20575, 95/21819, 95/22525, 95/23798, 95/26338, 95/28418 127700.doc-62-200836731, 95/30674, 95/30687, 95/33744, 96/05181, 96/05193, 96/05203, 96/06094, 96/07649, 96/10562, 96/ No. 16939, No. 96/18643, No. 96/20197, No. 96/21661 , Nos. 96/29304, 96/29317, 96/29326, 96/29328, 96/31214, 96/32385, 96/37489, 97/01553, 97/01554, 97/03066, 97/08144, 97/14671, 97/17362, 97/18206, 97/19084, 97/19942 and 97/ No. 21702; and British Patent Publication Nos. 2 266 529, 2 268 931, 2 269 170, 2 269 590, 2 271 774, 2 292 144, 2 293 168, Nos. 2,293,169 and 2,302,689. The preparation of such compounds is fully described in the above-identified patents and publications, which are incorporated herein by reference. In one embodiment, the neurokinin-1 receptor antagonist used in conjunction with a compound of the invention is selected from the group consisting of: 2_(Bu(7?)·(3,5-double) as described in U.S. Patent No. 5,719,147 (trifluoromethyl)phenyl)ethoxy)-3-(S)-(4-fluorophenyl)-4-(3-(5-oxoyl-1//,4 open-1,2 , cardiac triazolyl) indenyl) morpholine or a pharmaceutically acceptable salt thereof. The compounds of the invention may also be administered with an agent for the treatment of anemia. This anemia therapeutic agent is, for example, a continuous erythropoietin receptor activator (e.g., Alfa Iberetine). The compounds of the invention may also be administered with an agent for the treatment of neutropenia. This neutropenic therapeutic agent (for example) can be 127700.doc • 63 - 200836731 A hematopoietic growth factor that regulates neutrophil production and function, for example, human granulocyte colony stimulating factor (G-CSF). Examples of G-CSF include filgrastim. The compounds of the invention may also be associated with, for example, levofloxacin. Immune-enhancing drugs such as levamisole, isoprinosine, and Zadaxin are administered together. The compounds of the invention may also be combined with bisphosphonates (which are understood to include bisphosphonates, bisphosphonates, bisphosphonates, and bisphosphonates) for the treatment or prevention of cancer, including bone cancer. Examples of bisphosphonates include, but are not limited to, etidronate (didronel), pamidronate (Aredia), alendronate (Fosamax), risedronate (riseronate) (Actonel), zoledronate (Zometa), ibandronate (Boniva), incadronate or cimadronate, chlorine Clodronate, EB-1053, minodronate, neridronate, piridronate, and tiludronate, including any All pharmaceutically acceptable salts, derivatives, hydrates and mixtures thereof. Accordingly, the scope of the present invention encompasses the use of the claimed compounds in combination with ionizing radiation and/or in combination with a second compound selected from the group consisting of HDAC inhibitors, estrogen receptor modulators, androgen receptor modulators, and the like. Retinoid receptor modulators, cytotoxic/cytostatic agents, antiproliferative agents, prenyl-protein transferase inhibitors, HMG-CoA reductase inhibitors, angiogenesis inhibitors, PPAR-gamma agonists, PPAR-δ agonist, disease resistance 127700.doc -64- 200836731 Tincture, inhibitor of intrinsic multidrug resistance, antiemetic, agent for anemia treatment, agent for treatment of neutropenia, immunity An agent that enhances drug, cell proliferation, and survival signaling, an agent that interferes with cell cycle checkpoints, an apoptosis inducing agent, and a bisphosphonate. A term associated with a compound of the invention, "administering" and variants thereof (eg, administration of a ''compound) means that the compound or prodrug of the compound is introduced into a system in need of treatment of the animal. Where a prodrug is provided in combination with one or more other active agents (e.g., cytotoxic agents, etc.), "administered" and variants are each understood to include both the simultaneous and sequential introduction of the compound or its prodrugs and other agents. As used herein, it is intended to cover a product comprising a specified amount of the specified ingredient X and any combination of specified ingredients in a specified amount, directly or indirectly. The term "therapeutically effective amount" as used herein means the active compound or A pharmaceutical agent that produces a biological or medical response in a tissue, system, animal, or human, ', is sought by a researcher, veterinarian, physician, or other clinician. Surgery::/σ therapy system, refers to the treatment of mammals infected by pathological conditions and aims to alleviate the condition by killing cancer cells and also means that the progress of inhibition of the progress of the brothers is reduced, The rate of progression is stopped, the condition is improved, and the condition is cured. Also included as a precautionary measure (ie, prevention). "Apply acceptable" means a reasonable pharmaceutical judgment: Sex 2 is used in contact with a subject (eg, human) without excessive, allergic reactions or other problems or complications. Reasonable 127700.doc -65- 200836731 Benefits / Wind Harmony & L + 剞.帛 帛 The corresponding compound, material #, composition and/or agent 1 (four) agent and the like are compatible with the other components of the formulation, and must also be, acceptable,,. : A helper, refers to the use of a compound in combination with known treatments. These include the cytotoxic regimen and/or ionizing radiation of the drugs used in the treatment of different cancer types.牿 ^, 5, it is known that the active compound of sputum can strengthen many cancer chemotherapeutic treatments from ^_ ^, the specific active compounds include topoisomerases (eg ^^, Shi
• 如拓撲替康、伊立替康、魯比替康 ㈣1㈣叫)、大部分已知烧基化劑(例如,DTIC、替莫唑 胺)及在治療癌症中所用基於翻之藥物(例如,卡始、順 翻)〇 申明專利_之範m括—種治療癌症之方法,其包 括將治療有效量之式1化合物與輕射治#組合及/或與選自 以下之化合物組合投與:HDAC抑制劑、雌激素受體調節 刈、雄激素文體調節劑、類視色素受體調節劑、細胞毒性 /細胞生長抑制劑、抗增生劑、異戊二烯基_蛋白轉移酶抑 制y hmg-c〇a還原酶抑制劑、血管生成抑制劑、 γ激動劑、PPAR-δ激動劑、抗病毒劑、固有多耐藥性抑制 劑、鎮吐劑、用於治療貧血之藥劑、用於治療嗜中性白血 球減少症之藥劑、免疫增強藥、細胞增生及存活信號途秤 之抑制劑、干擾細胞週期檢測點之藥劑、凋亡誘導藥劑及 雙膦酸鹽。 β 本發明之该等及其他態樣可自本文所包含教示顯而易 見0 127700.doc -66- 200836731 閣述化學品及在下列實例中所用縮寫係:• Topotecan, irinotecan, Rubiconkine (4) 1 (4), most known alkylating agents (eg, DTIC, temozolomide), and drug-based drugs used in the treatment of cancer (eg, Kashun, Shun A method for treating cancer comprising administering a therapeutically effective amount of a compound of Formula 1 in combination with Light Shot # and/or in combination with a compound selected from the group consisting of: HDAC inhibitors, Estrogen receptor regulation of sputum, androgen style regulators, retinoid receptor modulators, cytotoxicity/cytostatic agents, antiproliferative agents, isoprenyl-protein transferase inhibition y hmg-c〇a reduction Enzyme inhibitors, angiogenesis inhibitors, gamma agonists, PPAR-delta agonists, antiviral agents, intrinsic multidrug resistance inhibitors, antiemetic agents, agents for the treatment of anemia, for the treatment of neutropenia The agent, the immunopotentiator, the inhibitor of cell proliferation and survival signal scale, the agent that interferes with the cell cycle detection point, the apoptosis-inducing agent, and the bisphosphonate. These and other aspects of the invention can be readily ascertained from the teachings herein. 0 127700.doc -66-200836731 The chemicals used and the abbreviations used in the following examples:
AcCl (乙醯氣);(Bz〇)2 (過氧化苯甲醯);Cbz_cl (苄基 氯甲酸酯);DCM (二氯曱烷);DIPEA (二-異-丙基乙基 胺);DMF (二甲基甲醯胺);DMSO (二甲基亞砜);eq·(當 量);ES (電噴射);EtOAc (乙酸乙酯);EtOH (乙醇); mol· Sieves (分子篩);HATU [六氟磷酸0-(7-氮雜苯并三 峻-1-基卜况愚#’,#:四甲基錁];MeCN (乙腈);MeOH (甲 醇);MS (質譜);MW (微波);NBS (沁溴琥珀酸亞胺); NMMO (W_甲基嗎啉-ΛΓ·氧化物);NMR (核磁共振);Pcol (柱壓);iPrOH (異丙醇);RT (室溫);sat· aq·(飽和水溶 液);Si02 (矽膠);及THF (四氫呋喃)。aC2〇 (乙酸);卜 BuOH (第三-丁醇);mPEA (二-異-丙基乙基胺);K〇Ac (乙酸鉀);MW 微波;1ST ISOLUTE⑧ SPE 柱 SCX (International Sorbent Technology ISOLUTE® Solid Phase Extraction柱陽離子交換樹脂);Sfc (超臨界流體層析); TBTU四氟硼酸〇-(1孖,苯并三唑-1-基四甲基錁 及Tcol (柱溫)。 式I化合物可藉由使式IA化合物與氨反應來製備:AcCl (acetamethylene); (Bz〇) 2 (benzaldehyde peroxide); Cbz_cl (benzyl chloroformate); DCM (dichlorodecane); DIPEA (di-iso-propylethylamine) ;DMF (dimethylformamide); DMSO (dimethyl sulfoxide); eq·(equivalent); ES (electrospray); EtOAc (ethyl acetate); EtOH (ethanol); mol·Sieves (molecular sieve) ;HATU [hexafluorophosphoric acid 0-(7-azabenzotrien-1-yl), #: tetramethylhydrazine]; MeCN (acetonitrile); MeOH (methanol); MS (mass); Microwave); NBS (沁 bromide succinimide); NMMO (W_methylmorpholine-ΛΓ·oxide); NMR (nuclear magnetic resonance); Pcol (column pressure); iPrOH (isopropanol); RT (room Temperature); sat· aq·(saturated aqueous solution); SiO 2 (silicone); and THF (tetrahydrofuran). aC2〇 (acetic acid); BuBuOH (third-butanol); mPEA (di-iso-propylethylamine) K;Ac (potassium acetate); MW microwave; 1ST ISOLUTE8 SPE column SCX (International Sorbent Technology ISOLUTE® Solid Phase Extraction column cation exchange resin); Sfc (supercritical fluid chromatography); TBTU bismuth tetrafluoroborate-(1 Helium, benzene And triazol-1-yltetramethylguanidine and Tcol (column temperature). The compound of formula I can be prepared by reacting a compound of formula IA with ammonia:
其中R1及R2係如上文所定義且Rx係C"烷基,例如,甲 基。通常在密封反應容器中使用NH3之水性溶液於諸如 127700.doc -67- 200836731 THF等溶劑中於70°C下實施該反應(慎重)。或者,可添加 諸如NaOH或KOH等鹼以將該酯水解成對應羧酸(RX係 氫),繼而添加NH3,於諸如HATU或TBTU及DIPEA等偶合 劑存在下,在諸如DMF等溶劑中,該反應在約室溫下實 施。或者,可使用(例如)boC2〇活化該羧酸以形成混合酸 酐’且隨後與碳酸氫銨反應,通常在諸如吼咬等溶劑中。 或者,可使用氨在諸如MeOH等溶劑中於約12(TC下在(例 如)MW中將該酯轉化成式^化合物。 在以上合成過程中應藉由(例如)Boc保護式〗八化合物之 六氫啦啶環上的氮原子。 式1A化合物可藉由使式1B化合物與疊氮化物反應來製 備:Wherein R1 and R2 are as defined above and Rx is C" alkyl, for example, methyl. The reaction is usually carried out in a sealed reaction vessel using an aqueous solution of NH3 in a solvent such as 127700.doc-67-200836731 THF at 70 ° C (prudent). Alternatively, a base such as NaOH or KOH may be added to hydrolyze the ester to the corresponding carboxylic acid (RX-based hydrogen), followed by the addition of NH3, in the presence of a coupling agent such as HATU or TBTU and DIPEA, in a solvent such as DMF. The reaction is carried out at about room temperature. Alternatively, the carboxylic acid can be activated, for example, with boC2 oxime to form a mixed acid anhydride' and subsequently reacted with ammonium bicarbonate, typically in a solvent such as a bite. Alternatively, the ester can be converted to a compound of the formula in, for example, MW at about 12 (TC) in a solvent such as MeOH. The above compound should be protected by, for example, Boc. A nitrogen atom on a hexahydropyridinium ring. A compound of formula 1A can be prepared by reacting a compound of formula 1B with an azide:
”中R、R及RX係如上文所定義。可使用諸如NaN3等"R, R and RX are as defined above. Can be used such as NaN3, etc.
且氮化物通常在諸如DMF等溶劑中,於約90。〇至140°C 下亦可使用諸如2,6-二甲基吡啶等添加劑。該反應可於 氮氣氛下實施。 式IB化合物可藉由式…化合物與式ι〇化合物之縮合反應 來製備: 127700.doc -68 - 200836731And the nitride is usually in a solvent such as DMF at about 90. Additives such as 2,6-lutidine can also be used at temperatures up to 140 °C. This reaction can be carried out under a nitrogen atmosphere. The compound of the formula IB can be prepared by a condensation reaction of a compound of the formula with a compound of the formula ι: 127700.doc -68 - 200836731
(ic) R2(ic) R2
例 其中R1、R2及Rx係如上文所定義且Ll係離去基團 如,硝基或鹵素,如,氟。方法包括於諸如MgS〇4或分子 筛等脫水劑存在下縮合或在諸如乙醇等醇溶劑中於回流下 加熱。該反應可於氮氣氛下實施。Wherein R1, R2 and Rx are as defined above and the L1 is a leaving group such as a nitro group or a halogen such as fluorine. The method includes condensation in the presence of a dehydrating agent such as MgS 4 or a molecular sieve or heating under reflux in an alcohol solvent such as ethanol. This reaction can be carried out under a nitrogen atmosphere.
式1C化合物可藉由使用諸如NMMO等氧化劑氧化式ιέ化 合物來製備: C〇2RxThe compound of formula 1C can be prepared by oxidizing a oxime compound using an oxidizing agent such as NMMO: C〇2Rx
R (IE) 其中R1、Rx及L1係如上文所定義及l2係離去基團,例 如,鹵素,如溴,通常在諸如MeCN等溶劑中於約室溫 下。該反應可於氮氣氛下實施。 可藉由於諸如過氧化本甲醯等自由基起始劑存在下使用 諸如NBS等溴化劑氧化式IF化合物來製備其中L2係溴之式 IE化合物: co2rx (IF) 127700.doc -69- 200836731 其中R1、Rx及L1係如上文所定義,通常在諸如cci4等溶 劑中於回流下。該反應可於氮氣氛下實施。 其中L1係氟之式if化合物可藉由式犯化合物之重氮化反 應繼而分解中間體重氮鹽來製備:R (IE) wherein R1, Rx and L1 are as defined above and a l2 leaving group, for example, a halogen such as bromine is usually present in a solvent such as MeCN at about room temperature. This reaction can be carried out under a nitrogen atmosphere. The IE compound of the formula L2 bromine can be prepared by oxidizing an IF compound using a brominating agent such as NBS in the presence of a radical initiator such as peroxybenzate: co2rx (IF) 127700.doc -69- 200836731 Wherein R1, Rx and L1 are as defined above, usually under reflux in a solvent such as cci4. This reaction can be carried out under a nitrogen atmosphere. The L1-based fluorine-type if compound can be prepared by decomposing the intermediate diazonium salt by the diazotization reaction of the compound:
(IG) ,、中R及R係如上文所定義。例如,可使用四氟硼酸亞 硝鑌在諸如DCM等溶劑中於約〇t:T實施該重氮化反應。 隨後可分離對應四氟㈣重氮鹽且接下來在高溫(例如, 藉由加熱至160。〇下在諸如二氯苯等溶劑中將其分解成對 應氟苯衍生物(謹慎)。 其中L1係硝基之式_合物可藉由式賺合物之頌化反 應,繼而進行酯化反應來製備··(IG), , R and R are as defined above. For example, the diazotization reaction can be carried out in a solvent such as DCM at about 〇t:T using nitrosonium tetrafluoroborate. The corresponding tetrafluoro(tetra) diazonium salt can then be separated and subsequently decomposed to the corresponding fluorobenzene derivative (caution) in a solvent such as dichlorobenzene by heating to 160. The L1 system The formula of the nitro group can be prepared by the oximation reaction of the formula, followed by the esterification reaction.
其中R1係如上文所定義。 硝酸鹽及諸如硫酸等酸存在 應步驟可在標準條件下實施 了在約室溫下於諸如頌酸鉀等 下實施該硝化反應。該酯化反 ’例如,藉由與之烷基 127700.doc -70- 200836731 鹵(其中x係鹵素,例如,蛾)於諸如碳酸铯等驗存在下且 在諸如DMF等溶劑中於約室溫下反應。式Rx-OH醇亦可與 諸如自AcCl/MeOH就地產生的HC1等酸觸媒於回流下一起 使用。隨後可藉由使用氫及諸如碳載鈀等觸媒氫化(通常 在諸如MeOH等醇溶劑中)硝基化合物生成對應苯胺來獲得 期望式IF化合物。 或者,式I化合物可藉由還原式IJ化合物來製備: CONH.Wherein R1 is as defined above. The nitrate and the presence of an acid such as sulfuric acid may be carried out under standard conditions under a standard condition such as potassium citrate or the like. The esterification is reversed by, for example, by reacting an alkyl group 127700.doc-70-200836731 with a halogen (wherein x-based halogen, for example, moth) in the presence of, for example, cesium carbonate and in a solvent such as DMF at about room temperature The next reaction. The Rx-OH alcohol of the formula may also be used together with an acid catalyst such as HCl which is produced in situ from AcCl/MeOH under reflux. The desired IF compound can then be obtained by hydrogenation of a nitro compound using hydrogen and a catalyst such as palladium on carbon (usually in an alcohol solvent such as MeOH) to form the corresponding aniline. Alternatively, a compound of formula I can be prepared by reduction of a compound of formula IJ: CONH.
(IJ)(IJ)
其中R1及R2係如上文所定義。可使用諸如CBz-Cl等醯氯 及諸如NaBH4等還原劑以富勒反應(Fowler reaction)方式實 施該還原。碳載鈀上之氫化反應完成該反應並去除CBz-保 護基團。 式IJ化合物可藉由交聯式IK化合物與式IL之3-吡啶基硼 酸來製備:Wherein R1 and R2 are as defined above. The reduction can be carried out in a Fowler reaction using ruthenium chloride such as CBz-Cl and a reducing agent such as NaBH4. Hydrogenation on carbon-supported palladium completes the reaction and removes the CBz-protecting group. The compound of formula IJ can be prepared by crosslinking a IK compound with 3-pyridyl boronic acid of formula IL:
(IK) (IL) 其中R1、R2及L2均為如上文所定義。該反應通常在 127700.doc -71 - 200836731(IK) (IL) wherein R1, R2 and L2 are as defined above. The reaction is usually at 127700.doc -71 - 200836731
Suzuki偶合條件(例如,使用諸如Pd2(dba)3及三(第三-丁基) 膦等觸媒以及諸如碳酸鈉等鹼和諸如DMF及水等溶劑在約 90°C下)下實施。 式IK化合物可藉由式IM化合物與式IN化合物之縮合反 應來製備:The Suzuki coupling conditions are carried out, for example, using a catalyst such as Pd2(dba)3 and tris(t-butyl)phosphine, and a base such as sodium carbonate and a solvent such as DMF and water at about 90 °C. Formula IK compounds can be prepared by condensation of a compound of formula IM with a compound of formula IN:
(IM)(IM)
(IN) 其中R1、R2及L2係如上文所定義且L3係離去基團,例 如,鹵素,如氟;通常在諸如DMF等溶劑中於約180°C下 在MW中。亦可添加諸如K2C03等鹼。 式IM化合物可藉由下列來製備:使其中R1及Rx係如上文 所定義之式IO化合物(IN) wherein R1, R2 and L2 are as defined above and the L3 is a leaving group, for example, a halogen such as fluorine; usually in a solvent such as DMF at about 180 ° C in MW. A base such as K2C03 may also be added. Compounds of formula IM can be prepared by formulating compounds of formula IO wherein R1 and Rx are as defined above
(10) 與諸如KOH或NaOH等鹼在約室溫下反應以將該酯水解 成對應羧酸(Rx係氫),繼而添加NH3,於諸如HATU、 DIPEA及TBTU等偶合劑存在下,在諸如DMF等溶劑中, 該反應在約室溫下實施。 127700.doc •72- 200836731 r^r諸如咖⑶等溶劑中於約抓下使用諸如乙 劑乙酿化苯胺基團來自式ig化合物製備式ι〇化合 ::來:=二用存:酸^ 弓丨坐之%化’通常於諸如甲苯等共溶劑及水 存在下於約〇 °C下。 當未闇明中間體及起始材料之合成時,此等化合物有市 售或可错由標準方法或藉由擴展以上合成、本文反應圖及 實例來自市售化合物製備。 •可藉由已知方法或藉由闌述於以上合成、本文反應圖及 只例中之方法將式j化合物轉化成其他式^匕合物。 在本文所述任-合成順序過程中,可能有必要及/或期 ^保護任—所涉及之分子上之敏感或反應基團。此可藉助 白用保濩基團來達成,例如,彼等闡述於h价…叹 〇〇啊以 办__第3版,Gr_,τ w 及 Wuts,(10) reacting with a base such as KOH or NaOH at about room temperature to hydrolyze the ester to the corresponding carboxylic acid (Rx-based hydrogen), followed by the addition of NH3 in the presence of a coupling agent such as HATU, DIPEA and TBTU, such as in In a solvent such as DMF, the reaction is carried out at about room temperature. 127700.doc •72- 200836731 r^r In a solvent such as coffee (3), use a compound such as B to brew an aniline group from a compound of formula ig to prepare a formula: 来: =: two uses: acid ^ The % of the bow squat is usually at about 〇 ° C in the presence of a co-solvent such as toluene and water. When the synthesis of the intermediate and starting materials is not known, such compounds are commercially available or can be prepared by standard methods or by extending the above synthesis, the reaction schemes herein and the examples from commercially available compounds. • Compounds of formula j can be converted to other formulas by known methods or by the methods described above in the synthesis, in the reaction schemes and in the examples. In the course of any-synthesis sequence described herein, it may be necessary and/or useful to protect any of the sensitive or reactive groups on the molecule involved. This can be achieved with the help of the white-protected group, for example, they are stated at the price of h... sigh, __ 3rd edition, Gr_, τ w and Wuts,
Wiley Interscience,1999 及 Kocienski,P. jWiley Interscience, 1999 and Kocienski, P. j
Pro⑽仏⑽…,Thieme,1994中者。可在適宜後續階段 使用業内已知方法去除該等保護基團。舉例而言,當存在 Boc(弟二·丁氧基羰基)或苄基羧基保護基團時,可藉由在 約室溫下添加諸如TFA、DCM及/或MeCN等溶劑來去除該 保護基團。亦可使用標準方法(例如,使用諸如pd/c等觸 媒、在諸如甲醇等溶劑中於氫氣氛下)氫化該化合物。亦 可於HC1及1,4-二氧雜環己烷存在下添加EtOAc以去除B〇c 或苄基羰基保護基團,在約室溫下。 依照下列反應圖製備本發明之化合物。各式之所有變量 127700.doc -73- 200836731 係如上文所定義。 當本發明之化合物具有對掌性中心時,可藉由標準分離 方法(例如,使用SFC、對掌性HPLC或用對掌性酸溶解)自 外消旋混合物分離各對映異構體。可在製備式I化合物之 過程的任一步驟中實施該分離。因此,可在最後步驟實施 分離,或者可分離中間體且隨後在後續反應中利用特定對 映異構體以產生期望產物。 反應圖1 一種合成彼等本發明化合物之衍生物的程序示於反應圖 1中’其中使用類似於闡述於WO 2005/066136中者之合成 途徑製備經取代2//-吲唑。在2-硝基-3-甲基-苯甲酸衍生物 生成對應酯之初始轉化後,使用如N-溴琥珀酸亞胺及過氧 化笨甲醯等試劑對甲基基團進行基團溴化,產生關鍵苄基 溴衍生物。可使用(例如甲基嗎啉_沁氧化物及分子篩完 成此苄基溴化物至對應苯甲醛的氧化。在該醛與胺縮合 後,可藉由使用疊氮化鈉在高溫下處理該關鍵中間體以引 入最終氮原子及所得氮之逐出以提供吲唑環來完成環閉 合。亦可向此反應中添加諸如二甲基吡啶等鹼。最後,將 該S旨轉化為-級醯胺,產生期望料物。此可藉由在氨溶 液中加熱該酯或藉由轉化成對應羧酸且隨後實施醯胺偶合 127700.doc -74- 200836731Pro(10)仏(10)..., Thieme, 1994. These protecting groups can be removed at a suitable subsequent stage using methods known in the art. For example, when a Boc (dibutyloxycarbonyl) or benzylcarboxy protecting group is present, the protecting group can be removed by adding a solvent such as TFA, DCM and/or MeCN at about room temperature. . The compound can also be hydrogenated using standard methods (e.g., using a catalyst such as pd/c in a solvent such as methanol under a hydrogen atmosphere). EtOAc can also be added in the presence of HCl and 1,4-dioxane to remove the B〇c or benzylcarbonyl protecting group at about room temperature. The compounds of the invention were prepared according to the following reaction schemes. All variables of the formula 127700.doc -73- 200836731 are as defined above. When the compound of the invention has a palmitic center, the individual enantiomers can be separated from the racemic mixture by standard separation methods (e.g., using SFC, palmitic HPLC or by dissolving the palmitic acid). This separation can be carried out in any step of the process for preparing the compound of formula I. Thus, the separation can be carried out in the final step, or the intermediate can be isolated and subsequently utilized in a subsequent reaction to produce the desired product. Reaction Scheme 1 A procedure for the synthesis of derivatives of the compounds of the present invention is shown in Reaction Scheme 1 wherein a substituted 2//-carbazole is prepared using a synthetic route similar to that described in WO 2005/066136. After the initial conversion of the 2-nitro-3-methyl-benzoic acid derivative to the corresponding ester, group bromination of the methyl group using reagents such as N-bromosuccinimide and benzamidine peroxide , producing a key benzyl bromide derivative. Oxidation of the benzyl bromide to the corresponding benzaldehyde can be accomplished using, for example, methylmorpholine-onium oxide and molecular sieves. After condensation of the aldehyde with the amine, the critical intermediate can be treated by using sodium azide at elevated temperatures. The body is introduced by introducing a final nitrogen atom and the resulting nitrogen to provide a carbazole ring to complete the ring closure. A base such as lutidine may also be added to the reaction. Finally, the S is converted to a -grade guanamine. Producing the desired material. This can be accomplished by heating the ester in an ammonia solution or by converting to the corresponding carboxylic acid and subsequently performing a guanamine coupling 127700.doc -74 - 200836731
酉旨化 例如 AcCI,RxOH 回流 co2rxFor example, AcCI, RxOH reflux co2rx
Bromination 例如 NBS,(BzO)2, CCI4) Δ C02Rxj^Yn〇2 Ri^^-CH2Br 氧化 例如,NMMO,分子篩Bromination such as NBS, (BzO)2, CCI4) Δ C02Rxj^Yn〇2 Ri^^-CH2Br oxidation For example, NMMO, molecular sieve
Rx = C^alkylRx = C^alkyl
i) _3, DMF,90oC H)酰胺形成 NH3,THF 或 MeOH, 70°C密封試管,或i) _3, DMF, 90oC H) amide formation NH3, THF or MeOH, 70 ° C sealed test tube, or
NaOH 或 KOH,NH3,HATUNaOH or KOH, NH3, HATU
或 TBTU,DIPEA,DMF,RT 反應圖1 反應圖2 反應圊1之變化形式於下文中在反應圖2中展示且可在該 等吲唑核心上引入取代基。當所需硝基苯甲酸衍生物無市 售時,其可藉由(例如)使用存於濃硫酸中之硝酸鉀硝化對 應苯甲酸衍生物來製備。如上文所述合成操作可形成對應 苯胺,此苯胺亦可藉由首先乙醯化該苯胺且在濃HC1酸中 於〇°C下用亞硝酸鈉環化來經環化生成吲唑。或者,可使 用四氟硼酸亞硝鑌重氮化該苯胺且使對應四氟硼酸重氮鹽 在高溫下分解以藉由希曼反應(Schiemann reaction)(小心) 生成對應二就苯衍生物。按照如在反應圖1中所述合成順 序,可將苄基甲基基團氧化成對應醛並藉由與(雜)醯替苯 127700.doc -75- 200836731 胺偶合並用疊氮化鈉實施環化產生期望α引唑衍 R1 CO# 例如 KN〇jconc h2S〇4 9°2ΗOr TBTU, DIPEA, DMF, RT Reaction Figure 1 Reaction Figure 2 Variations of Reaction 圊 1 are shown below in Reaction Scheme 2 and substituents can be introduced on the oxazole core. When the desired nitrobenzoic acid derivative is not commercially available, it can be prepared, for example, by nitrating a corresponding benzoic acid derivative with potassium nitrate present in concentrated sulfuric acid. The synthesis operation as described above forms the corresponding aniline which can also be cyclized to form the carbazole by first acetylating the aniline and cyclizing it with sodium nitrite at 〇 ° C in concentrated HCl. Alternatively, the aniline may be diazotized using nitrosonium tetrafluoroborate and the corresponding diazonium tetrafluoroborate may be decomposed at a high temperature to produce a corresponding diphenyl derivative by Schiemann reaction (care). According to the synthesis sequence as described in the reaction scheme 1, the benzylmethyl group can be oxidized to the corresponding aldehyde and the ring is formed by sodium azide by combining with the (hetero) quinone 127700.doc-75-200836731 amine couple. To produce the desired α-zolyl derivative R1 CO# such as KN〇jconc h2S〇4 9°2Η
CH3 R1CH3 R1
CH, 生物 酯化 N〇2 例如.AcCI,MeOH,ΔCH, bioesterification N〇2 eg .AcCI, MeOH, Δ
還原 H2> Pd/CRestore H2> Pd/C
CHO C〇2RxCHO C〇2Rx
i) NOBF4, DCM C〇2Rx ϋ)氣苯 ,△i) NOBF4, DCM C〇2Rx ϋ) gas benzene, △
R1’ 、CH3 環化R1', CH3 cyclization
例如 i) AcCI,55〇C ii) NaN02l cone HCI, 0°CFor example i) AcCI, 55〇C ii) NaN02l cone HCI, 0°C
參見反應圖3See reaction diagram 3
反應圖2 反應圖3Reaction Figure 2 Reaction Figure 3
另一程序涉及在後期階段官能團化該吲唑,如在反應 圖3中所示。在此,首先將吲唑酯轉化成對應甲醯胺且隨 後用適當氟(雜)芳香族溴化物實施親核性芳香族取代。此 可製備能夠在Suzuki偶合條件(例如,使用三(第三_ 丁基) 膦及Pd2(dba)3作為觸媒於諸如碳酸鈉等鹼存在下)下交聯 偶合之溴化物衍生物。隨後藉由富勒反應使用諸如CBz_cl 等醯氯及諸如NaBH4等還原劑來完成至期望六氫吡啶部分 的轉化。最終氫化反應可產生對應六氫吡啶衍生物。 127700.doc -76- 200836731Another procedure involves functionalizing the oxazole at a later stage, as shown in Figure 3 of the reaction. Here, the carbazole ester is first converted to the corresponding formamide and then subjected to nucleophilic aromatic substitution with an appropriate fluorine (hetero) aromatic bromide. This can prepare a bromide derivative capable of cross-linking coupling under Suzuki coupling conditions (e.g., using tris(t-butyl)phosphine and Pd2(dba)3 as a catalyst in the presence of a base such as sodium carbonate. Subsequent conversion to the desired hexahydropyridine moiety is accomplished by a Fuller reaction using ruthenium chloride such as CBz_cl and a reducing agent such as NaBH4. The final hydrogenation reaction produces the corresponding hexahydropyridine derivative. 127700.doc -76- 200836731
CO〇RXCO〇RX
酰胺形成 例如,NH3,△ N 或 K〇g,隨後 ΤΒΊ^υ NH - C0NH〇Amide formation, for example, NH3, ΔN or K〇g, followed by ΤΒΊ^υ NH - C0NH〇
S>jAr反應 ^ , MW, 180°CS>jAr reaction ^ , MW, 180 ° C
Suzuki偶合Suzuki coupling
反應圖3 PARP-1 SPA 試驗 在此試驗中測試本文所述例示性化合物且發現該等化合 物具有小於5 μΜ(具體而言,小於50 nM)之IC5〇值。 工作試劑 試驗用緩衝液:100 mM Tris pH 8, 4 mM MgCl2, 4 mM 精胺,200 mM KC1,0.04% Nonidet P-40。Reaction Scheme 3 PARP-1 SPA Assay The exemplary compounds described herein were tested in this assay and found to have an IC5 enthalpy of less than 5 μΜ, specifically less than 50 nM. Working Reagents Test buffer: 100 mM Tris pH 8, 4 mM MgCl2, 4 mM spermine, 200 mM KC1, 0.04% Nonidet P-40.
U 酵素混合物:試驗用缓衝液(12.5 ul),100 mM DTT (0.5 ul),PARP-1 (5 nM,Trevigen 4668-5 00-01),H20 (補足至 35 ul) o 煙醯胺-腺嘌呤二核苷酸(NAD)/DNA混合物·· [3H-NAD] (250 uCi/ml,0.4 ul,Perkin-Elmer NET-443H),NAD (1.5 mM, 0.05 ul,SIGMA N-1511),生物素化NAD (250 uM,0.03 ul, Trevigen 4670-500-01),激活小牛胸腺(1 mg/ml,0.05ul, Amersham Biosciences 27-4575),H20 (補足至 10 ul)。 127700.doc -77- 200836731 顯影混合物:溶於500 mM EDTA中之Streptavidin SPA 珠體(5 mg/ml,Amersham Biosciences RPNQ 0007)。 實驗設計 在96-孔微量培養板中以50 uL/孔最終體積實施該反應。 添加5 ul 5%DMSO/化合物溶液,添加酵素混合物(35 ul) ’ 藉由添加NAD/DNA混合物(10 uL)開始該反應並在RT下培 育2小時。藉由添加顯影混合物(25 ul)終止該反應並在在 RT下培育15 min。使用Packard TOP COUNT儀器進行量 測。 BRCA_1沈默HeLa細胞之增生試驗· 縮寫: IMDM (Iscovei改良之杜貝克氏培養基);MM (Roswell Park Memorial Institute培養基);MOI (感染 ^ 樣 性);GFP (綠色螢光蛋白);PBS (磷酸缓衝鹽溶液);FC:S (胎牛血清);及DMEM (杜貝克氏改良鷹氏培養基)°U Enzyme mixture: test buffer (12.5 ul), 100 mM DTT (0.5 ul), PARP-1 (5 nM, Trevigen 4668-5 00-01), H20 (to 35 ul) o nicotinic amine-gland嘌呤 dinucleotide (NAD) / DNA mixture · [3H-NAD] (250 uCi / ml, 0.4 ul, Perkin-Elmer NET-443H), NAD (1.5 mM, 0.05 ul, SIGMA N-1511), biological The NAD (250 uM, 0.03 ul, Trevigen 4670-500-01) was activated to activate the calf thymus (1 mg/ml, 0.05 ul, Amersham Biosciences 27-4575) and H20 (to 10 ul). 127700.doc -77- 200836731 Development mixture: Streptavidin SPA beads (5 mg/ml, Amersham Biosciences RPNQ 0007) dissolved in 500 mM EDTA. Experimental Design The reaction was carried out in a 96-well microplate at a final volume of 50 uL/well. 5 ul of 5% DMSO/compound solution was added, and the enzyme mixture (35 ul) was added. The reaction was started by adding a NAD/DNA mixture (10 uL) and incubated at RT for 2 hours. The reaction was stopped by the addition of a developing mixture (25 ul) and incubated for 15 min at RT. Measurements were performed using a Packard TOP COUNT instrument. BRCA_1 Silencing HeLa Cell Proliferation Assay Abbreviation: IMDM (Iscovei Modified Dubecca Medium); MM (Roswell Park Memorial Institute); MOI (infection); GFP (green fluorescent protein); PBS (phosphoric acid) Flushing salt solution; FC: S (fetal calf serum); and DMEM (Dubec's modified eagle medium) °
亦可在抗增生試驗中於匹配BRCAlwt及BRCAlJshl^W ^ tIt can also match BRCAlwt and BRCAlJshl^W ^ t in anti-proliferation test.
HeLa細胞中測試本發明化合物。該試驗展示PARP抑带〗# 能夠顯示選擇性地抑制BRCA缺陷之細胞的生長。該等化 合物在BRCA1缺陷之細胞中顯示低於5 μΜ之CC5〇’s及車父田 含BRCA之細胞大10的選擇性。 該試驗係基於活細胞可將氧化還原染劑(^ (resazurin))轉化成榮光終產物(試鹵靈(resofurin))之月匕力 所產生試_靈之量與細胞數量成正比。 細胞系此等係在MOI為1〇0下& 127700.doc -78 - 200836731 含有shRNA(針對BRCA-1)及GFP表現盒之慢病毒轉導的 HeLa細胞。BRCA-1沈默係大於80%,如藉由Taqman分析 所評定且該等細胞穩定地表現GFP。 丑Γ/ΓΜ-GT^P-此等係在MOI為100下經不表現任一 shRNA之對照載體轉導的HeLa細胞。 測試方案 * 在96孔黑色板中於90 μΐ培養基中以300個細胞/孔進行 播種* : 「於37°C下培育4小時,5 % C02 - 以10 ul/孔添加10X化合物(5 % DMSO存於H20中) -於37°C下培育168小時,5 % C02 - 添加10 μΐ預先以1:1在PBSlx中稀釋的Cell titer Blue溶 液(Promega,G8081) -於37°C下將該混合物培育45’,5 % C02 -在RT下於黑暗中培育15,The compounds of the invention were tested in HeLa cells. This test demonstrates that PARP inhibition can show the growth of cells that selectively inhibit BRCA deficiency. These compounds showed less than 5 μΜ of CC5〇's in BRCA1-deficient cells and a selectivity of 20 cells of BRCA-containing cells in the plant. The test is based on the fact that living cells can convert the redox dye (resazurin) into the final product of glory (resofurin). The amount of the test is proportional to the number of cells. Cell lines are HeLa cells transduced with lentiviral transduction of shRNA (for BRCA-1) and GFP expression cassette at MOI of 1〇0 & 127700.doc -78 - 200836731. The BRCA-1 silencing system is greater than 80% as assessed by Taqman analysis and the cells stably express GFP. Ugly/ΓΜ-GT^P- These are HeLa cells transduced with a control vector that does not exhibit any shRNA at an MOI of 100. Test protocol* Seeding at 300 cells/well in 90 μM medium in 96-well black plates*: “4 hours incubation at 37°C, 5% C02 - 10X compound (5 % DMSO) at 10 ul/well Stored in H20) - Incubate at 37 ° C for 168 hours, 5% C02 - Add 10 μM of Cell titer Blue solution (Promega, G8081) pre-diluted in PBSlx at 1:1 - Mix the mixture at 37 °C Incubate 45', 5% C02 - 15 in the dark at RT,
-用勞光計對板進行讀數,ex: 550 nm; em: 590 nm ‘ * 培養基:DMEM (GIBCO,41966-029),10% FCS (GIBCO,10106-169),0.1 mg/ml 青黴素-鏈黴素(GIBCO, 15140-114),2 mM L-麩胺醯胺(GIBCO, 3042190) 天然缺少BRC A之細胞系之增生試驗· 本發明之化合物亦顯不可以小於5微莫耳之C C 5 〇1抑制天 然 BRCA-1 (MDA-MB-436)及 BRCA-2 (CAPAN-1)缺陷之細 胞系之增生。 增生試驗 127700.doc -79- 200836731 在96-孔板中於100 ul適當培養基/孔中以700個細胞/孔播 種細胞。* 次曰,以200 μΐ/孔之最終體積添加系列稀釋化合物。每 一稀釋物按一式三份進行測試。 在 6天後,使用 CellTiter-Blue Cell Viability Assay 按照 生產商說明(Promega)評定細胞存活率。使用Fusi〇I1 Alpha 微量培養板讀數器(Packard Bioscience)對平板進行讀數。 對於低增生細胞系(即’ CAPAN-1),在添加該等化合物 之後14天(且在第7天時改變培養基1次,抽出17〇 μ1培養基 並用170 μΐ含有該等混合物之新鮮培養基代替)測試增生。 *培養基: MDA-MB-436: RPMI (GIBCO), 10 % FBS (5% C02) CAPAN-1: IMDM (GIBCO),20 % FBS (5% C02) 在腫瘤學活體内模型中所測試化合物顯示明顯活性水平。- Reading the plate with a light meter, ex: 550 nm; em: 590 nm ' * Medium: DMEM (GIBCO, 41966-029), 10% FCS (GIBCO, 10106-169), 0.1 mg/ml penicillin-chain Protease (GIBCO, 15140-114), 2 mM L-glutamine (GIBCO, 3042190) Proliferation assay of cell lines naturally lacking BRC A · The compounds of the invention may also be less than 5 micromoles of CC 5 〇1 inhibits proliferation of cell lines deficient in native BRCA-1 (MDA-MB-436) and BRCA-2 (CAPAN-1). Proliferation assay 127700.doc -79- 200836731 Cells were seeded at 700 cells/well in 100 ul of the appropriate medium/well in 96-well plates. * Secondary enthalpy, adding serial dilutions in a final volume of 200 μΐ/well. Each dilution was tested in triplicate. After 6 days, cell viability was assessed using CellTiter-Blue Cell Viability Assay according to the manufacturer's instructions (Promega). Plates were read using a Fusi(R) I1 Alpha microplate reader (Packard Bioscience). For the low proliferative cell line (ie 'CAPAN-1), 14 days after the addition of the compounds (and on the 7th day, the medium was changed once, 17 〇μ1 medium was withdrawn and 170 μ 新鲜 fresh medium containing the mixture was used instead) Test proliferation. *Medium: MDA-MB-436: RPMI (GIBCO), 10% FBS (5% C02) CAPAN-1: IMDM (GIBCO), 20% FBS (5% C02) Compounds tested in oncology in vivo models Significant activity levels.
製備實例 實例A 2 -苯基-2丑-0弓丨嗤-7-甲醯胺(a 6) 步驟1 : 3-甲基-2-硝基苯甲酸甲基酯(A1) 於〇°C下向3-甲基-2-硝基-苯甲酸(ΐ·〇 eq·)存於MeOH (0.4 Μ)之懸浮液中逐滴添加AcCl (3.0 eq.)。將該反應混合物在 回流下攪拌20小時。在真空中減少溶劑並將殘留物溶於 EtOAc中且用飽和NaHC03水溶液、鹽水洗務若干次並乾燥 (NazSO4)。蒸發溶劑,產生白色固體狀(Ai),其未經進一 步純化即用於下一步驟。1H NMR (400 MHz,CDC13, 300K) 127700.doc -80 - 200836731 δ 7.86 (1H,d,J=7.5 Hz),7.53-7.42 (2H,m),3·89 (3H,s) 2.36 (3H,s)。MS (ES) C9H9N04規定值:195,實驗值: 218 (M+Na)、 步称2 · 3-(溪甲基)-2-石肖基苯甲酸甲醋(A 2) 在回流、N2氣氛下,將(A1) (1.0 eq.)、(BzO)2 (〇 〇6 eq ) 及NBS (1.18 eq·)存於CC14 (0.2 Μ)中之混合物加熱12小 時。使該混合物冷卻至RT、用DCM稀釋、在減壓下濃縮 同時乾燥裝填至Si〇2上。藉由Si〇2急驟管柱層析使用1 〇·9〇 EtOAc/石油醚純化該殘留物以產生白色固體狀期望(Α2)。 'Η NMR (400 MHz5 CDC13, 300K) δ 7.93 (1H? d? J = 7.7 Hz), 7.72 (1H,d,J=7.7 Hz),7·57 (1H,t,J=7.7 Hz),4·43 (2H,s), 3·88 (3H,s)。MS (ES) C9H8BrN04規定值:273:275,實驗 值:242:244 (M-MeO)+,227:229 (Μ-Ν02) +。 步驟3 : 3-曱醯基-2-硝基苯曱酸甲酯(A3) 於RT下,向(A2)及4A分子篩(15 g)存於MeCN (0.2M)之 混合物中添加NMMO (2.0 eq·)並在N2氣氛下將該反應混合 物攪拌1.5小時。隨後,用EtOAc稀釋該混合物,過濾並用 出0、IN HC1、鹽水洗滌濾液並乾燥(Na2S〇4)。蒸發溶 劑,產生白色固體狀(A3),其未經進一步純化即用於下一 步驟。b NMR (400 MHz,CDC13, 300K) δ 9.96 (1H,s), 8·26 (1Η,d,J=7.9 Ηζ),8.18 (1Η,d,J=7.9 Hz), 7·77 (1Η,t, J = 7.9 Hz),3·93 (3H,s). MS (ES) C9H7N05規定值:209, 實驗值:208 (M-H)-。 步驟4 : 2-硝基-3-[(苯基亞胺基)甲基】苯曱酸甲酯(A4) 127700.doc -81 - 200836731 在回流下於N2氣氛中將(Α3) (1·〇 eq·)及苯胺(1〇5叫·)存 於EtOH (〇·2 Μ)中之混合物攪拌2小時直至τιχ顯示該反應 完成(己院/EtOAc=75:25)。蒸發溶劑,產生白色固體狀 (A4) ’其未經進一步純化即用於下一步驟。iH NMR (4⑼ MHz5 CDC13,300K) δ 8.51 (1Η5 d, J=7.3 Ηζ)5 8.41 (1Η5 s)5 8·11 (1Η,d,J=7.8 Ηζ),7.67 (1Η,t,J=7.8 Ηζ),7.43 (2Η,t, J 7.8 Hz),7.31 (1H,t,J=7.3 Hz),7.16 (2H,d,J=7.8 Hz), 3.94 (3H,s)。 步驟5 : 2-苯基-2H_吲唑-7_甲酸甲酯(A5) 於 90C 下’在& 氣氛中將(α4) (1·〇 eq)及 NaN3 〇 〇5 eq.) 存於乾燥DMF (0.3 M)中之混合物攪拌過夜。在真空中減 少該粗製物並藉由矽膠急驟管柱層析使用自1〇:9〇至4〇:6〇 之EtOAc/石油醚梯度純化殘留物以產生期望褐色油狀 (A5)。H NMR (400 MHz,CDC13, 300K) δ 8.50 (1H,s), 8」2 (1Η,d,J=7.0 Ηζ),7·96-7·90 (3Η,m),7.49 (2Η,t, J = 7.6 Hz),7·38 (1H,t,J=7.4 Hz),7·15 (1H,t,J=7.4 Hz), 4·〇3 (3H,s)· MS (ES) C15H12N202規定值:252,實驗值: 253 (M+H)+ 〇 步驟6 · 2_苯基-2H_吲唑_7_甲醯胺(A6) 在70 C下,於密封試管中,將酯(A5)在thf及32% NH3 水溶液之混合物中加熱過夜。在真空中減少溶劑並藉由矽 勝急驟管柱層析使用自3〇:7〇至5〇:5〇之扮〇^/石油醚梯度 純化殘留物以產生期望白色固體狀(A6)。lH NMR (4〇〇 MHz,DMSO, 300K) δ 9.33 (1H,s),8·56 (1H,bs),8·16 (2H, 127700.doc -82- 200836731 d5 J=7.9 Hz)5 8.08-8 00 、” •U0 (2H,m),7.88 (1H,bs),7.63 (2H,t, J = 7.7 Hz), 7.50 (1H, t, 7.4 Hz), 7.27 (1H, t, J=7.9 Hz) 〇 MS ㈣ Cl4HllN3〇規定值:237,實驗值:238 (m+h)+。 製備實例 實例1 氣化3-{4-[7·(胺基M基)_2Hn2_基】苯基}六氮吨咬鎮 (B4) 步称1 · 3-[4-({-[3-(甲氧基幾基)-2-確基苯基】亞甲基}胺基) 苯基】六氫吡啶-1-甲酸第三·丁基酯(B1) (B1)係按妝對製備實例a步驟4所報告一般程序使用 及3-(4-胺基笨基)六氫吡啶4•甲酸第三-丁基酯來製備(直 至TLC顯示該反應完成(石油醚:Et〇Ac=4:1))且其未經進 一步純化即用於下一步驟。 步驟2 : 2-{4_[1·(寒三基羰基)六氩0比啶-3_基】苯基卜 2丑吲唑-7-甲酸甲酯(Β2) (Β2))係按照對製備實例a步驟5所報告一般程序來製備 且藉由矽膠急驟管柱層析使用2〇-4〇% EtOAc/石油醚梯度 純化粗製物以產生期望黃色固體狀(B2)。iH NMR (400 MHz,CDC13, 300K) δ 8.51 (1H,s),8.13 (1H,d,J = 7.1 Hz), 7·95 (1H,d,J=8.3 Hz),7.91 (2H,d,J=8.4 Hz),7·39 (2H,d, J = 8.4 Hz),7.18 (1H,t,J=7.2 Hz),4.30-4.10 (2H,m),4.00 (3H,s),2·85,2·70 (3H,m),2.11-2.03 (1H,m),1.83-1.75 (1H,m),1.73-1.53 (2H,m 重疊於 H20 信號),1.48 (9H,s). MS (ES) C25H29N304規定值:435,實驗值:436 (M+H)+。 127700.doc -83 - 200836731 步驟3 · 3-{4-[7_(胺基羰基)_2仏吲唑•基】苯基丨六氫吡啶_ 1-甲酸第三-丁基酯(B3)PREPARATION EXAMPLE EXAMPLE A 2 -Phenyl-2 ugly-0 丨嗤-7-carbamimid (a 6) Step 1: 3-methyl-2-nitrobenzoic acid methyl ester (A1) at 〇 ° C To a suspension of 3-methyl-2-nitro-benzoic acid (ΐ·〇eq·) in MeOH (0.4 Μ) was added dropwise EtCl (3.0 eq.). The reaction mixture was stirred under reflux for 20 hours. The solvent was reduced in vacuo and the residue was taken from EtOAc EtOAc EtOAc EtOAc The solvent was evaporated to give a white solid (Ai), which was used in the next step without further purification. 1H NMR (400 MHz, CDC13, 300K) 127700.doc -80 - 200836731 δ 7.86 (1H,d,J=7.5 Hz), 7.53-7.42 (2H,m),3·89 (3H,s) 2.36 (3H , s). MS (ES) C9H9N04 specified: 195, calcd.: 218 (M+Na), step 2 · 3-(Ximethyl)-2-succinylbenzoic acid methyl acetonate (A 2 ) under reflux, N 2 atmosphere, The mixture of (A1) (1.0 eq.), (BzO)2 (〇〇6 eq) and NBS (1.18 eq·) in CC14 (0.2 Μ) was heated for 12 hours. The mixture was cooled to RT, diluted with DCM, concentrated under reduced pressure and dried and then applied to Si. The residue was purified by flash chromatography eluting with EtOAc EtOAc (EtOAc) 'Η NMR (400 MHz5 CDC13, 300K) δ 7.93 (1H? d? J = 7.7 Hz), 7.72 (1H, d, J = 7.7 Hz), 7·57 (1H, t, J = 7.7 Hz), 4 · 43 (2H, s), 3.88 (3H, s). MS (ES) C9H8BrN04: 273: 275, calc.: 242: 244 (M-MeO)+, 227: 229 (Μ-Ν02) +. Step 3: Methyl 3-mercapto-2-nitrobenzoate (A3) Add NMMO (2.0) to a mixture of (A2) and 4A molecular sieves (15 g) in MeCN (0.2M) at RT. Eq.) and the reaction mixture was stirred under N2 atmosphere for 1.5 hours. Subsequently, the mixture was diluted with EtOAc, filtered and washed with EtOAc EtOAc EtOAc The solvent was evaporated to give a white solid (A3). b NMR (400 MHz, CDC13, 300K) δ 9.96 (1H, s), 8·26 (1Η, d, J=7.9 Ηζ), 8.18 (1Η, d, J=7.9 Hz), 7·77 (1Η, t, J = 7.9 Hz), 3·93 (3H, s). MS (ES) C9H7N05 specified: 209, found: 208 (MH)-. Step 4: Methyl 2-nitro-3-[(phenylimino)methyl]benzoate (A4) 127700.doc -81 - 200836731 (Α3) in a N2 atmosphere under reflux (1· The mixture of 〇eq·) and aniline (1〇5 ··) in EtOH (〇·2 Μ) was stirred for 2 hours until τιχ showed the reaction was completed (H.sub.1/EtOAc = 75:25). The solvent was evaporated to give a white solid (A4), which was used in the next step without further purification. iH NMR (4(9) MHz5 CDC13,300K) δ 8.51 (1Η5 d, J=7.3 Ηζ)5 8.41 (1Η5 s)5 8·11 (1Η,d,J=7.8 Ηζ), 7.67 (1Η,t,J=7.8 Ηζ), 7.43 (2Η, t, J 7.8 Hz), 7.31 (1H, t, J = 7.3 Hz), 7.16 (2H, d, J = 7.8 Hz), 3.94 (3H, s). Step 5: 2-Phenyl-2-H-indazole-7-formic acid methyl ester (A5) is stored in (in the & atmosphere, (α4) (1·〇eq) and NaN3 〇〇5 eq.) at 90C The mixture in dry DMF (0.3 M) was stirred overnight. The crude material was reduced in vacuo and the residue was purified eluting with EtOAc EtOAc (EtOAc) H NMR (400 MHz, CDC13, 300K) δ 8.50 (1H, s), 8"2 (1Η, d, J=7.0 Ηζ), 7·96-7·90 (3Η, m), 7.49 (2Η, t , J = 7.6 Hz), 7·38 (1H, t, J = 7.4 Hz), 7·15 (1H, t, J = 7.4 Hz), 4·〇3 (3H, s)· MS (ES) C15H12N202 Specified value: 252, Experimental value: 253 (M+H) + 〇Step 6 · 2_Phenyl-2H_carbazole_7_Mercaptoamine (A6) At 70 C, in an sealed tube, the ester ( A5) Heating in a mixture of thf and 32% NH3 in water overnight. The solvent was reduced in vacuo and the residue was purified by flash chromatography eluting with EtOAc (EtOAc) eluting lH NMR (4〇〇MHz, DMSO, 300K) δ 9.33 (1H, s), 8.56 (1H, bs), 8.16 (2H, 127700.doc -82- 200836731 d5 J=7.9 Hz)5 8.08 -8 00 ,” • U0 (2H, m), 7.88 (1H, bs), 7.63 (2H, t, J = 7.7 Hz), 7.50 (1H, t, 7.4 Hz), 7.27 (1H, t, J= 7.9 Hz) 〇MS (4) Cl4HllN3 〇 Specified value: 237, Experimental value: 238 (m+h)+. Example of preparation Example 1 Gasification 3-{4-[7·(Amino M group)_2Hn2_yl]phenyl }六氮吨 bite town (B4) Step 1 · 3-[4-({-[3-(Methoxy)-2- sureylphenyl]methylene}amino) phenyl] Hydropyridine-1,3-carboxylic acid tert-butyl ester (B1) (B1) is prepared according to the general procedure reported in Step 4 of Preparation Example a and 3-(4-Amino-phenyl)hexahydropyridine 4•carboxylic acid Tri-butyl ester was prepared (until TLC showed the reaction was completed (petroleum ether: Et EtOAc: 4: 1)) and was used in the next step without further purification. Step 2: 2-{4_[1· (cold triylcarbonyl)hexa-argon 0-pyridin-3-yl]phenyl bromide 2 uglyoxazole-7-formic acid methyl ester (Β2) (Β2)) was prepared according to the general procedure reported in Preparation Example 5, Step 5. And use 2 〇 by gel column chromatography The crude material was purified by EtOAc/EtOAc (EtOAc) (EtOAc) (EtOAc) (HHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHHH ), 7·95 (1H, d, J=8.3 Hz), 7.91 (2H, d, J=8.4 Hz), 7·39 (2H, d, J = 8.4 Hz), 7.18 (1H, t, J= 7.2 Hz), 4.30-4.10 (2H, m), 4.00 (3H, s), 2·85, 2·70 (3H, m), 2.11-2.03 (1H, m), 1.83-1.75 (1H, m) , 1.73-1.53 (2H, m is superimposed on H20 signal), 1.48 (9H, s). MS (ES) C25H29N304 specified value: 435, experimental value: 436 (M+H)+. 127700.doc -83 - 200836731 3 · 3-{4-[7_(Aminocarbonyl)_2carbazole•yl]phenylphosphonium hexahydropyridine_1-carboxylic acid tert-butyl ester (B3)
於密封試管中,在60°C下將(B2)在存於MeOH (0·1 M)之 7Ν ΝΗ3中加熱2天。在真空中減少溶劑並藉由用Et2〇研磨 來純化該粗製產物以獲得期望黃色固體狀(B3)。lH NMR (400 MHz,CDC13, 300K) δ 9.04 (1H,br. s),8·51 (1H,s), 8.31 (1Η,d,J = 6.8 Ηζ),7.91 (1Η,d,J=8.3 Ηζ),7.84 (2Η,d, J一8·2 Hz),7.42 (2H,d,J=8.2 Hz),7.31-7.22 (1H,m與 CDC13信號重疊),5.95(1^1,|^ 3),4 4〇_4 〇5(211,111),2 9〇_ 2·7〇 (3H,m),2.15-2.00 (1H,m),1.85-1.75 (1H,m)5 1.75-1·5〇 (2H,m 與 H20 信號重疊),ι ·48 (9H,s)_ MS (ES) C24H28N4〇3規定值:420,實驗值:421 (m+H)+。 步称4 :氣化3_{4-[7_(胺基幾基)-2丑唑-2-基】苯基}六氫 比啶鑌(B4) 向(B3) (1.0 eq)存於EtOAc之經攪拌溶液(0.2M)中加入4N HC1/1,4- 一氧雜環己烧溶液(1 〇·〇 eq)並將該反應混合物在 RT下撥拌3 h。在低壓下蒸發溶劑並藉由用Et20研磨來純 化粗製產物以產生期望黃色固體狀(B4)。4 NMR (400 MHz,DMSO-d6, 300K) δ 9·32 (1H,s),9.12 (1H,br· s),8.87 (1H,br· s),8.55 (1H,br. s),8.13 (2H,d,J =8.6 Hz),8.06 (1H,J=7.0 Hz),8.02 (1H,d,J=8.4 Hz), 7.89 (1H,br. s), 7.55(2H,d,J = 8.6Hz),7.27(lH,dd,J=8.4,7.0Hz),3.43-3.27(2H,m),3.17-3.03(2H,m),3.00-2.85 (lH,m),2.00· 1.70 (4H,m)· MS (ES) C19H21C1N40規定值:320,實驗 127700.doc -84- 200836731 值:321 (M+H)+。 實例2 2-{4_[(3R)-六氫吡啶·3_基]苯基}-2H-吲唑-7-甲醯胺(C1) 及2-{4-[(3S)-六氫吡啶-3-基】苯基}-2H-吲唑-7-甲醯胺(C2) 藉由對掌性 SFC (管柱:Chiralpak AS-H,1 X 25 mm,流 速:10 ml/min,Tcol : 35°C,Pc〇i : 100巴,改良劑:55% (iprOH+4°/〇 Et2NH))使用C02作為超臨界溶洗劑分離實例 1,B4,獲得兩種純淨對映異構體。 獲得白色粉末狀第一經溶洗對映異構體(C1),滯留時間 (SFC) : 4.80 min。4 NMR (400 MHz,DMSO-d6,300K) δ 9.28 (s,1Η),8.57 (br· s,1Η),8.06 (d,2Η,J=7.2 Ηζ),8·04 (d,2H,J=8.4 Hz),7.88 (br. s,1H),7.49 (d,2H,J = 8.4 Hz), 7.27 (dd,1H,J=8.4, 7.2 Hz),3.08-2.94 (m,2H),2.77-2.67 (m,1H),2.64-2.52 (m,1H),1.98義 1.90 (m,1H),1.75] 47 (m,4H). MS (ES) C19H2〇N40規定值:320,實驗值:321 (M+H)+。將游離鹼轉化成氣化(3及)-3-{4-[7-(胺基幾基)_ 2H-吲唑-2-基]苯基}六氫σ比啶鑌並量測旋光度: [ol]20d=+133.3 ( c 〇·15,MeOH)。 獲得白色粉末狀第二經溶洗對映異構體(C2),滞留時間 (SFC) : 6.51 min。4 NMR (400 MHz,DMSO-d6, 3〇〇Κ) δ 9·28 (s,1Η),8·57 (br· s,1Η),8·06 (d,2Η,J=7.2 Ηζ),8 〇4 (d,2H,J=8.4 Hz),7·88 (br· s,1H),7.49 (d,2H,J = 8 4 Hz) 7.27 (dd,1H,J=8.4, 7.2 Hz),3.08-2.94 (m,2H),2·77·2 67 (m,1H),2.64-2.52 (m,1H),1.98-1.90 (m,1H),〇 127700.doc -85 - 200836731 (m,4H)。MS (ES) C19H2〇N40規定值:320,實驗值:321 (M + H)+。將游離驗轉化成氯化(35)-3-{4_ [7-(胺基羰基)一 2H-吲唑-2-基]苯基}六氫吡啶鑌並量測旋光度:= -137.9 ( c 0.145,MeOH)。 實例3 三氟乙酸3_{4-[7-(胺基羰基)-5-氟-2H-吲唑-2-基】苯基} 六氫11比咬銪(D4) 步驟1 : 5-氟-1H-吲唑_7_甲酸甲酯(D1) 向實例4,E3 (1_0 eq)存於1,2-二氣乙烷(〇.1 μ)之溶液中 加入AcCl (5 eq)並在55C下加熱2 h。此後在減壓下去除 溶劑。 將該白色固體溶於曱苯/水(5/1,0.1 M)中。使該溶液冷 卻至0°C並添加HC1 (10 eq·,3 7%)。隨後,緩慢地添加若干 份NaN02 (10 eq)並將該混合物在〇°C下擾拌3 h。有機相用 水(3x)洗滌,經MgSCU乾燥並在加壓下去除溶劑。 隨後於90°C下將該黃色曱苯溶液(〇·ΐ M)加熱2 h。蒸發 甲苯,產生紅色固體狀期望產物。1H NMR (400 MHz, DMSO,300K) δ 13·37 (1H, s),8.23 (1H,s),7.63 (1H,dd, J=8.6 Hz,J=2.5 Hz),7.48 (1H,dd,J=8.6 Hz,J=2.5 Hz), 3·66 (3H,s)· MS (ES+) C9H7FN2〇2規定值·· m,實驗值·· 195 (M+H)、 步驟2 : 5-氟-1H_吲唑·7_甲醯胺(D2) 將(Dl)/谷於一氧雜環己烧/水〇/},〇· i m)中並添加 (1 · 5 eq)。在RT下祝拌12 h後,在減壓下去除溶劑。白色 127700.doc -86 - 200836731 固體未經純化即可用於後續偶合。 將羧酸溶於DMF (0.1 M)中並在〇°C下添加TBTU (1.5 eq)。在 15 min 後,添加 DIPEA (2.0 eq·)及氨(3.0 eq.,0.5 Μ ’存於二氧雜環己烷中)並將該混合物在rt下攪拌% h。 添加EtOAc並用飽和NaHC〇3水溶液(3x)及鹽水(2x)洗滌有 機相。有機相經乾燥並於減壓下蒸發。藉由急驟層析使用 1 -2〇% MeOH/DCM純化粗製物以產生白色固體狀(D2)。 MS (ES+) C8H6FN30規定值:179,實驗值:180 (M+H)+。 步驟3 : 2-(4_溴苯基)_5_氟_2H_吲唑-7•甲醯胺(D3) 向D2 (1.0 eq)存於DMF (0.2 Μ)之溶液中添加k2C〇3 (1 3 eq)及4-溴氟苯(ι〇·〇 eq)並在Mw條件下於i80°c下將該反應 混合物加熱20 min。使該反應混合物冷卻至RT並用Et〇Ac 稀釋。有機相用鹽水洗滌,乾燥(NaeCU)。蒸發溶劑,產 生(D3),其藉由矽膠層析用50-70% EtOAc/石油醚溶洗來 純化以獲得黃色粉末狀標題化合物。1H NMR (400 ΜΗι DMS〇-d6, 3G〇K) δ 9.34 (1H,s),8·50 (1H,br· s),8.17 (2H, d,J 9.0 Hz),8.03 (1H,br. s),7.90-7.80 (4H, m)。MS (ES+) C14H9BrFN30 規定值:334/336,實驗值:335/337 (M+H)+ 〇 步驟4 ·· S-氟-2-(4-吡啶-3-基苯基)·2Η-吲唑。_甲醯胺(D4) 用Ar氣流對(D3) (1.0 eq)及吡啶-3_硼酸(13 存於 dmf (1·〇 M)以及2N Na2CCMe液(2〇 eq)中之混合物進行 脫氣,30分鐘。添加BV(〇〇5 eq)m(d叫 (〇·〇5 eq)並將该反應混合物在9〇。(;下加熱48 h。使該混合 127700.doc -87- 200836731 物冷卻至RT,添加DCM且有機相用飽和NaHC03水溶液、 鹽水洗滌,乾燥(NaeCU)。在減壓下濃縮溶液並藉由石夕膠 層析依次用50-90% EtOAc/石油醚及ΐοο/oMeOH/DCM溶洗 來純化殘留物以獲得黃色粉末狀標題化合物。iH NMr (400 MHz,DMSO-d6,300K) δ 9·40 (1H,s), 9·01 (1H,d J=1.6 Hz),8.63 (1H,dd,J=4.8,1.6 Hz),8.57 (1H,br. s), 8.32 (2H,d,J=8.8 Hz),8.20 (1H,d,J=7.8 Hz),8.10 (1H, b r · s), 8.01 (2 H,d,J - 8 · 8 H z),7 · 8 8 - 7 · 8 2 (2 H,m),7 · 5 4 (1H dd,J=7.8, 4.8 Hz). MS (ES) C19H13FN40規定值:332,實 驗值:333 (M+H+)。 步驟5 : 3-{4-[7-(胺基羰基)-5-氟-2H-吲唑-2-基]苯基}六氫 吡啶-1-曱酸苄基酯(D5) 向(D4)存於乾燥MeOH (0.2 Μ)之經攪拌溶液中加入(B2) was heated in a sealed tube at 60 ° C for 2 days in 7 Torr 3 in MeOH (0.1 M). The solvent was reduced in vacuo and the crude product was purified by trituration with Et2 to afford the desired yellow solid (B3). lH NMR (400 MHz, CDC13, 300K) δ 9.04 (1H, br. s), 8·51 (1H, s), 8.31 (1Η, d, J = 6.8 Ηζ), 7.91 (1Η, d, J=8.3 Ηζ), 7.84 (2Η, d, J - 8·2 Hz), 7.42 (2H, d, J = 8.2 Hz), 7.31-7.22 (1H, m overlaps with CDC13 signal), 5.95 (1^1, |^) 3), 4 4〇_4 〇5(211,111), 2 9〇_ 2·7〇(3H,m), 2.15-2.00 (1H,m),1.85-1.75 (1H,m)5 1.75- 1·5〇(2H,m overlaps with H20 signal), ι ·48 (9H,s)_ MS (ES) C24H28N4〇3 specified value: 420, experimental value: 421 (m+H)+. Step 4: Gasification 3_{4-[7-(Aminomethyl)-2 oxazol-2-yl]phenyl}hexahydropyridinium (B4) to (B3) (1.0 eq) in EtOAc A 4N HC1/1,4-oxohedo-sinter solution (1 〇·〇eq) was added to the stirred solution (0.2 M) and the reaction mixture was stirred at RT for 3 h. The solvent was evaporated under reduced pressure and the crude product was purified by trituration with Et20 to yield the desired yellow solid (B4). 4 NMR (400 MHz, DMSO-d6, 300K) δ 9·32 (1H, s), 9.12 (1H, br·s), 8.87 (1H, br·s), 8.55 (1H, br. s), 8.13 (2H, d, J = 8.6 Hz), 8.06 (1H, J = 7.0 Hz), 8.02 (1H, d, J = 8.4 Hz), 7.89 (1H, br. s), 7.55 (2H, d, J = 8.6 Hz), 7.27 (lH, dd, J = 8.4, 7.0 Hz), 3.43 - 3.27 (2H, m), 3.17 - 3.03 (2H, m), 3.00 - 2.85 (lH, m), 2.00 · 1.70 (4H , m)· MS (ES) C19H21C1N40 specified value: 320, experiment 127700.doc -84- 200836731 Value: 321 (M+H)+. Example 2 2-{4_[(3R)-Hexahydropyridine-3-yl]phenyl}-2H-indazole-7-carboxamide (C1) and 2-{4-[(3S)-hexahydropyridine -3-yl]phenyl}-2H-carbazole-7-carboxamide (C2) by palmitic SFC (column: Chiralpak AS-H, 1 X 25 mm, flow rate: 10 ml/min, Tcol : 35 ° C, Pc〇i : 100 bar, improver: 55% (iprOH + 4 ° / 〇 Et 2 NH)) Separation of Example 1, B4 using CO 2 as a supercritical solvent to obtain two pure enantiomers . The first eluted enantiomer (C1) was obtained as a white powder, and the residence time (SFC): 4.80 min. 4 NMR (400 MHz, DMSO-d6, 300K) δ 9.28 (s, 1 Η), 8.57 (br· s, 1 Η), 8.06 (d, 2 Η, J = 7.2 Ηζ), 8·04 (d, 2H, J =8.4 Hz), 7.88 (br. s, 1H), 7.49 (d, 2H, J = 8.4 Hz), 7.27 (dd, 1H, J=8.4, 7.2 Hz), 3.08-2.94 (m, 2H), 2.77 -2.67 (m,1H), 2.64-2.52 (m,1H), 1.98=1.90 (m,1H), 1.75] 47 (m,4H). MS (ES) C19H2〇N40 Specified value: 320, Experimental value: 321 (M+H)+. Conversion of the free base to gasified (3 and)-3-{4-[7-(aminobenzyl)-2H-indazol-2-yl]phenyl}hexahydro-pyridinium oxime and measuring the optical rotation : [ol]20d=+133.3 ( c 〇·15, MeOH). The second eluted enantiomer (C2) was obtained as a white powder, and the residence time (SFC): 6.51 min. 4 NMR (400 MHz, DMSO-d6, 3 〇〇Κ) δ 9·28 (s, 1 Η), 8·57 (br· s, 1 Η), 8·06 (d, 2 Η, J = 7.2 Ηζ), 8 〇4 (d, 2H, J=8.4 Hz), 7·88 (br· s, 1H), 7.49 (d, 2H, J = 8 4 Hz) 7.27 (dd, 1H, J=8.4, 7.2 Hz) ,3.08-2.94 (m,2H),2·77·2 67 (m,1H),2.64-2.52 (m,1H),1.98-1.90 (m,1H),〇127700.doc -85 - 200836731 (m , 4H). MS (ES) C19H2 〇 N40: 320, found: 321 (M + H) +. The free assay was converted to (35)-3-{4_[7-(aminocarbonyl)-2H-indazol-2-yl]phenyl}hexahydropyridinium chloride and the optical rotation was measured: = -137.9 ( c 0.145, MeOH). Example 3 Trifluoroacetic acid 3_{4-[7-(aminocarbonyl)-5-fluoro-2H-indazol-2-yl]phenyl} hexahydro 11 ratio bite (D4) Step 1: 5-Fluoro- 1H-carbazole_7-methyl formate (D1) To a solution of Example 4, E3 (1_0 eq) in 1,2-dioxaethane (〇.1 μ) was added AcCl (5 eq) at 55 C Heat down for 2 h. Thereafter, the solvent was removed under reduced pressure. The white solid was dissolved in toluene/water (5/1, 0.1 M). The solution was cooled to 0 ° C and HCl (10 eq·, 3 7%) was added. Subsequently, several portions of NaN02 (10 eq) were slowly added and the mixture was spoiled at 〇 ° C for 3 h. The organic phase was washed with water (3x), dried over MgSCU and solvent was removed under pressure. The yellow benzene solution (〇·ΐ M) was then heated at 90 ° C for 2 h. The toluene was evaporated to give the desired product as a red solid. 1H NMR (400 MHz, DMSO, 300K) δ 13·37 (1H, s), 8.23 (1H, s), 7.63 (1H, dd, J = 8.6 Hz, J = 2.5 Hz), 7.48 (1H, dd, J=8.6 Hz, J=2.5 Hz), 3·66 (3H, s)· MS (ES+) C9H7FN2〇2 specified value·· m, experimental value·· 195 (M+H), step 2: 5-fluorine -1H_carbazole·7-formamide (D2) (Dl)/valid in monooxane/hydroquinone/}, 〇· im) was added (1·5 eq). After stirring for 12 h at RT, the solvent was removed under reduced pressure. White 127700.doc -86 - 200836731 The solid was used for subsequent coupling without purification. The carboxylic acid was dissolved in DMF (0.1 M) and TBTU (1.5 eq) was added at EtOAc. After 15 min, DIPEA (2.0 eq.) and ammonia (3.0 eq., 0.5 Μ' in dioxane) were added and the mixture was stirred at rt. EtOAc was added and the organic phase was washed with saturated aq. NaH.sub.3 (3x) and brine (2x). The organic phase was dried and evaporated under reduced pressure. The crude was purified by flash chromatography using EtOAc (EtOAc) elute MS (ES+) C8H6FN30: 179, found: 180 (M+H)+. Step 3: 2-(4-Bromophenyl)-5-fluoro-2H-indazole-7•carboxamide (D3) Add K2C〇3 to D2 (1.0 eq) in DMF (0.2 Μ) 1 3 eq) and 4-bromofluorobenzene (ι〇·〇eq) and the reaction mixture was heated at i80 ° C for 20 min under Mw conditions. The reaction mixture was cooled to RT and diluted with EtOAc. The organic phase was washed with brine and dried (NaeCU). The solvent was evaporated to give (D3), which was purified eluting eluting eluting 1H NMR (400 ΜΗι DMS〇-d6, 3G〇K) δ 9.34 (1H, s), 8.50 (1H, br·s), 8.17 (2H, d, J 9.0 Hz), 8.03 (1H, br. s), 7.90-7.80 (4H, m). MS (ES+) C14H9BrFN30 Specified value: 334/336, Experimental value: 335/337 (M+H) + 〇 Step 4 ··S-Fluoro-2-(4-pyridin-3-ylphenyl)·2Η-吲Oxazole. _Metformamide (D4) Degassed with a mixture of (D3) (1.0 eq) and pyridine-3_boric acid (13 in dmf (1·〇M) and 2N Na2CCMe (2〇eq) with Ar gas stream , 30 minutes. Add BV (〇〇5 eq) m (d called (〇·〇5 eq) and the reaction mixture is at 9 〇.(;; heating for 48 h. Make the mixture 127700.doc -87-200836731 After cooling to RT, DCM was added and EtOAc (EtOAc m.). /DCM </ RTI> </ RTI> </ RTI> </ RTI> </ RTI> </ RTI> <RTIgt; </ RTI> </ RTI> </ RTI> <RTIgt; , 8.63 (1H, dd, J = 4.8, 1.6 Hz), 8.57 (1H, br. s), 8.32 (2H, d, J = 8.8 Hz), 8.20 (1H, d, J = 7.8 Hz), 8.10 ( 1H, br · s), 8.01 (2 H,d,J - 8 · 8 H z),7 · 8 8 - 7 · 8 2 (2 H,m),7 · 5 4 (1H dd, J=7.8 , 4.8 Hz). MS (ES) C19H13FN40: 332, 303 (M+H+). Step 5: 3-{4-[7-(aminocarbonyl)-5-fluoro-2H-carbazole -2-yl]phenyl} Benzyl hexahydropyridine-1-decanoate (D5) was added to a stirred solution of (D4) in dry MeOH (0.2 Μ).
NaBH4 (1·2 eq)且隨後於_65〇C 下逐滴加入Cbz-Cl (1.2 eq)。 使該反應物達RT O/N,且隨後用HA淬滅。在減壓下濃縮 MeOH並添力口EtOAc〇有機相用飽和NaHC03水溶液洗務, 乾燥(Na2SO〇。蒸發溶劑,產生(D5),其未經進一步純化 即用於下一步驟。MS (ES) C27H25FN403規定值:472,實 驗值:473 (M+H+)。 步驟6 :三氟乙酸3-{4-[7-(胺基羰基)_5_氟-2H_吲唑·2-基】 苯基}六氫啦啶鑌(D6) 向(D5) (1.0 eq)存於MeOH (0.2 M)之溶液中添加pd/c 10% (0.05 eq)及HC1 (1.0 eq)並將該反應混合物在h2氣氛 (1 atm)下擾拌48 h。隨後,該混合物經由石夕藻土過濾並在 127700.doc -88 - 200836731 真空中去除溶劑,獲得(D6),其藉由反相RP-HPLC (管 柱:C18),使用 H20 (0.1% TFA)及 MeCN (0.1% TFA)作為 溶洗劑來純化,將期望部分凍乾以獲得白色粉末狀標題化 合物(D6)。4 NMR (400 MHz,CD3CN,300K) δ 9·28 (1H, s),8.89 (1Η,br. s)5 8·60_8·50 (2Η,m),8·13 (2Η,d,J=8_6 Hz),8.09 (1H,br. s),7.90-7.70 (2H,m), 7·54 (2H,d,J=8.6 Hz),3.40-3.30 (2H,m),3.20-2.80 (3H,m),2.00-1.90 (2H, m),1.80-1.70 (2H,m),MS (ES) C19H19FN40規定值:338, 實驗值:339 (M+H+)。 實例4 5-氟_2-(3-氣_4·六氫吡啶-3-基苯基丨唑-7-甲醯胺 三氟乙酸鹽(E6) 步驟1 : 5-氟_3-甲基-2-硝基苯曱酸(E1) 於〇°C下’向3·氟-5_甲基苯甲酸(ι·0 eq.)存於濃H2S04之 溶液中緩慢地添加KNO3 (1.1 eq)。將該混合物在rt下授拌 1 h且隨後緩慢地注入冰水中。在攪拌直到冰完全熔化 後,過濾、出白色沈澱,用冷水洗滌並在減壓下乾燥。白色 固體未經進一步純化即用於下一步驟。iH NMR (400 MHz, DMSO, 300K) δ 14.08 (1H,br. s),7.65 (2H,m),2.30 (3H,s)。 步驟2 : 5-氟-3-甲基-2-硝基苯甲酸甲基酯(E2) 於RT下,向(El)及碳酸铯(1_5 eq·)存於DMF (〇·25 M)之 溶液中加入碘甲烷(1.0 eq·)。在將該混合物攪拌18 h後, 加入鹽水並用EtOAc萃取該混合物。乾燥(Na2S〇〇有機相 並在減壓下實施濃縮。該黃色固體未經純化即用於下一步 127700.doc -89 - 200836731 驟。1H NMR (400 MHz,DMSO, 300K) δ 7.63 (2H,m),3.83 (3H,s),2.29 (3H,s)。 步驟3: 2-胺基-5-氟-3·甲基苯甲酸甲酯(E3) 於 RT、H2 氣氛(1 atm)下將(E2) (1.〇 eq·)及 pd/C (10% w/w)存於MeOH (0·25 Μ)中之混合物攪拌3 d。該混合物經 由Celite®過遽且隨後在加壓下蒸發出溶劑。白色固體未經 進一步純化即可用於後續步驟。1H NMR (400 MHz,DMSO, 300K) δ 7·29 (1H,dd,J=9.5 Hz,J=3.0 Hz),7.12 (1H,dd,J=9.5 Hz,J=3.0 Hz),6·36 (2H,br· s),3.78 (3H,s),2.11 (3H,s)。 步驟4 : 2,5·二氟-3-曱基苯甲酸甲酯(E4) 於〇°C下,向(Ε3) (1.0 eq·)存於乾燥DCM (0.4 Μ)之溶液 中逐份添加四氟硼酸亞硝鑌(1·3 eq·)。在1 h後,於0°C下 添加乾燥一氟苯(12 0 e q ·)並將該反應物緩慢地加熱至16 〇 °C,同時蒸餾出DCM。在3 hr後,使該混合物冷卻至rt, 加入EtOAc並用鹽水(2x)洗滌有機相。在經MgS04乾燥 後,於減壓下去除溶劑。藉由急驟層析使用丨〇% EtOAc/ 石油醚純化粗製物以產生黃色油狀(E4)。iH NMR (400 MHz,CDC135 300K) δ 7·42 (1H,m),7·06 (1H,m),3.92 (3H, s),2.30 (3H,d,J=2.3 Hz)。 步驟5 : 2,5-二氟-3-甲醯基苯曱酸甲酯(E5) 按照在製備實例A,步驟2及3中所報告一般程序自E4製 備(E5)。藉由急驟層析(1-20% EtOAc/石油醚)純化粗製物 以產生白色固體。1H NMR (300 MHz,DMSO,300K) δ 10.19 (1Η,d,J=2.4 Ηζ),7·98 (1Η,m),7·86 (1Η,m),3·89 (3Η, 127700.doc -90- 200836731 S)。MS (ES+) C9H6F203規定值:200,實驗值:201 (M+H)+。 步驟6 · 5-氟-2_(3-氟-4-六氫吡啶-3-基苯基)-2H-吲唑-7-甲 醯胺三氟乙酸鹽(E6) 按照在製備實例A步驟4及5中所報告一般程序使用3_(‘ 胺基-2-氟苯基)六氫吼啶_;^甲酸第三-丁基酯將(E5)轉化成 對應σ?| ^圭。 ’ 藉由於RT下用K0H U.3 eq·)存於二氧雜環己烷/水中之 ( 混合物(0.1 M)處理12 h來將所得2-{4-[1-(第三-丁氧基羰 基)六氫吡啶-3·基卜3-氟苯基卜5-氟_2斤-吲唑-7-甲酸甲酯進 一步轉化成對應甲醯胺。在減壓下去除溶劑。將羧酸溶於 DMF (〇·ΐ M)中並添加TBTU (1 5叫·)。在15㈤匕後,加入 DIPEA (2.0 eq·)及氨(3.0 eq·,〇·5 Μ,存於THF中)並將該溶 液攪拌36 h。用EtOAc稀釋該混合物且隨後用飽和NaHe〇3 水 >谷液及鹽水洗滌有機相。在蒸發出溶劑後,該殘留物未 經純化即用於下一步驟。NaBH4 (1.2 eq) and then Cbz-Cl (1.2 eq) was added dropwise at _65 °C. The reaction was allowed to reach RT O/N and then quenched with HA. The MeOH was concentrated under EtOAc (EtOAc)EtOAcEtOAcEtOAc. C27H25FN403 specified: 472, found: 473 (M+H+). Step 6: trifluoroacetic acid 3-{4-[7-(aminocarbonyl)_5-fluoro-2H-indazole-2-yl]phenyl To the solution of (D5) (1.0 eq) in MeOH (0.2 M), add pd/c 10% (0.05 eq) and HCl (1.0 eq) and the reaction mixture in h2 The mixture was stirred for 48 h under atmosphere (1 atm). Subsequently, the mixture was filtered through celite and the solvent was removed in vacuo at 127700.doc -88 - 200836731 to obtain (D6) by reversed phase RP-HPLC (tube Column: C18), Purified using H20 (0.1% TFA) and MeCN (0.1% TFA) as solvent. The desired portion was lyophilized to give the title compound (D6) as white powder. 4 NMR (400 MHz, CD3CN, 300K) δ 9·28 (1H, s), 8.89 (1Η, br. s) 5 8·60_8·50 (2Η, m), 8·13 (2Η, d, J=8_6 Hz), 8.09 (1H, Br. s), 7.90-7.70 (2H,m), 7·54 (2H,d,J=8.6 Hz), 3.40-3.30 (2H m), 3.20-2.80 (3H, m), 2.00-1.90 (2H, m), 1.80-1.70 (2H, m), MS (ES) C19H19FN40: 338, 355 (M+H+). Example 4 5-Fluoro-2-(3- gas_4·hexahydropyridin-3-ylphenyloxazole-7-formamide trifluoroacetate (E6) Step 1: 5-fluoro-3-methyl -2-Nitrobenzoic acid (E1) slowly added KNO3 (1.1 eq) to a solution of 3·fluoro-5-methylbenzoic acid (ι·0 eq.) in concentrated H2S04 at 〇 °C The mixture was stirred at rt for 1 h and then slowly poured into ice water. After stirring until the ice was completely melted, filtered, white precipitated, washed with cold water and dried under reduced pressure. Used in the next step. iH NMR (400 MHz, DMSO, 300K) δ 14.08 (1H, br. s), 7.65 (2H, m), 2.30 (3H, s). Step 2: 5-fluoro-3- Methyl 2-nitrobenzoate (E2) Add methyl iodide (1.0 eq·) to a solution of (El) and cesium carbonate (1_5 eq·) in DMF (〇·25 M) at RT . After the mixture was stirred for 18 h, brine was added and the mixture was extracted with EtOAc. The organic phase was dried (Na2S EtOAc (EtOAc m.) m), 3.83 (3H, s), 2.29 (3H, s) Step 3: 2-Amino-5-fluoro-3-methylbenzoic acid methyl ester (E3) under RT, H2 atmosphere (1 atm) The mixture of (E2) (1.〇eq·) and pd/C (10% w/w) in MeOH (0·25 Μ) was stirred for 3 d. The mixture was passed through Celite® and then pressurized. The solvent was evaporated. The white solid was used in the next step without further purification. 1H NMR (400 MHz, DMSO, 300K) δ 7·29 (1H, dd, J=9.5 Hz, J=3.0 Hz), 7.12 (1H , dd, J = 9.5 Hz, J = 3.0 Hz), 6.36 (2H, br · s), 3.78 (3H, s), 2.11 (3H, s) Step 4: 2,5·Difluoro-3 Methyl-mercaptobenzoate (E4) To a solution of (Ε3) (1.0 eq·) in dry DCM (0.4 Μ), nitroxonium tetrafluoroborate (1·3 eq) was added at 〇°C. ·) After 1 h, dry monofluorobenzene (12 0 eq ·) was added at 0 ° C and the reaction was slowly heated to 16 ° C while distilling off DCM. After hr, the mixture was cooled to EtOAc, EtOAc (EtOAc)EtOAc.EtOAc. To produce a yellow oil (E4). iH NMR (400 MHz, CDC 135 300K) δ 7·42 (1H, m), 7·06 (1H, m), 3.92 (3H, s), 2.30 (3H, d, J = 2.3 Hz). Step 5: Methyl 2,5-difluoro-3-indolylbenzoate (E5) Prepared from E4 according to the general procedure reported in Preparation Example A, Steps 2 and 3. (E5) The crude was purified by flash chromatography (1-20%EtOAcEtOAcEtOAc) elute (1Η,m),7·86 (1Η,m),3·89 (3Η, 127700.doc -90- 200836731 S).MS (ES+) C9H6F203 specified value: 200, experimental value: 201 (M+H) +. Step 6 · 5-Fluoro-2(3-fluoro-4-hexahydropyridin-3-ylphenyl)-2H-indazole-7-carbenamide trifluoroacetate (E6) according to Step 4 in Preparation Example A And the general procedure reported in 5 uses 3_('amino-2-fluorophenyl)hexahydroacridine_;^carboxylic acid tert-butyl ester to convert (E5) to the corresponding σ?| The resulting 2-{4-[1-(third-butoxy) was obtained by treatment in a mixture of dioxane/water (mixed (0.1 M) for 12 h at RT). The carbonyl group) hexahydropyridine-3· kib 3-fluorophenyl b 5-fluoro 1-2 lb-carbazole-7-carboxylic acid methyl ester is further converted into the corresponding formamide. The solvent is removed under reduced pressure. Soluble in DMF (〇·ΐ M) and add TBTU (1 5 ··). After 15 (five) ,, add DIPEA (2.0 eq·) and ammonia (3.0 eq·, 〇·5 Μ, stored in THF) and The solution was stirred for 36 h. The mixture was diluted with EtOAc and EtOAc EtOAc EtOAc.
為了實施去保護’將該粗製物溶於TFA/DCM (0· 1 M)中 並在RT下攪拌3 h。蒸發溶劑,產生殘留物,其藉由反相 HPLC (管柱:C18)純化以獲得標題化合物(E6)。NMR ' (4〇〇 MHz,DMS0, 300K) 5 9.34 (1H,s),8·90 (1H,m),8·61 (1H,m),8.49 (1H,s),8.18 (1H,dd,J=11.6 Hz,2.0 Hz), 8.05 (2H,m),7.81 (2H,m),7.63 (1H,m),3.34 (3H,m), 3.13 (1H,m),2.94 (1H,m),1.95-1.76 (4H,m). MS (ES + )To carry out deprotection, the crude material was dissolved in TFA/DCM (0.1 M) and stirred at RT for 3 h. The solvent was evaporated to give a crystal crystal crystal crystal crystal crystal crystal crystal crystal NMR ' (4〇〇MHz, DMS0, 300K) 5 9.34 (1H, s), 8.90 (1H, m), 8.61 (1H, m), 8.49 (1H, s), 8.18 (1H, dd , J=11.6 Hz, 2.0 Hz), 8.05 (2H, m), 7.81 (2H, m), 7.63 (1H, m), 3.34 (3H, m), 3.13 (1H, m), 2.94 (1H, m ), 1.95-1.76 (4H, m). MS (ES + )
Ci9H18F2N40規定值:356,實驗值:357 (M+H)+。 實例5 127700.doc -91 - 200836731 4-曱基苯磺酸(3S)-3-{4-丨7_(胺基羰基)_2H•吲唑_2•基丨苯 基}六氫ϋ比咬鏽(F4) 步驟1 : (3S)_3-[4-({(IE)_[3-(甲氧基羰基)小硝基苯基】亞甲 基}胺基)本基】六氫啦唆_1_甲酸第三_ 丁基酯(F1) 按照在實例1,B1中所述自八3及幻_3_(4_胺基苯基)六 氫吡啶_1-甲酸第三-丁基酯(藉由利用2當量於Me〇H中之L- 二苯甲醯基酒石酸拆分3-(4_胺基苯基)_六氫吼啶并隨後 Boc-保護而製得)製備(F1)。 步驟2 ·· 2-{4-[(3S)-M第三-丁氧基羰基)六氫吼啶_3_基】苯 基}-211-吲唑_7_甲酸(F2) 將(FI) (1 eq)及疊氮化鈉u eq)在dmF (0.25M)中製成漿 液、惰性化並添加2,6-二甲基吡啶(1·〇 eq)。將該混合物加 熱至11 0 C内部溫度,達20小時。將所得褐色溶液冷卻至 20°C並添加THF及25 wt % LiCl水溶液。分離各相並再用 25 wt % LiCl水溶液將有機部分洗滌3次。向以上有機溶液 中添加2.0MNaOH (10 eq)並將該混合物加熱至35°C,達20 小時,隨後冷卻至20°C並分離各相。用2.0M HC1酸及鹽水 之混合物洗滌有機層並分離各層,有機層用鹽水進一步洗 務並農縮以獲得(F 2)’其無需進一步純化。 步驟3 : (3S)-3-{4-[7-(胺基羰基)-2H-吲唑-2-基]苯基}六氫 吡啶-1-甲酸第三-丁基酯(F3) 將F2溶於DC Μ (0.3 5 M)中並在RT下添加碳酸二第三-丁 基酯(1.3 eq)及吡啶(1·〇 eq)。在30分鐘後,添加碳酸氫銨 (1.3 eq)並持續攪拌20小時。加入1M HC1 (5 mL/g)並分離 127700.doc •92- 200836731 各相,有機層用水洗滌兩次並濃縮達較小體積。粗製化合 物(F3)經由矽藻土墊過濾且隨後自甲基第三_丁基醚結晶。 步驟4 : 4-甲基苯磺酸(3S)_3_{4_[7-(胺基羰基>211_吲唑_2_ 基]苯基}六氫响咬鑌(F4) 將F3溶於THF (0.15M)中並添加水(5%,相對於THF)。 加入對-曱苯磺酸單水合物(2·2 eq)並將該混合物加熱至% C且攪拌過夜。在冷卻後,藉由過濾分離期望固體鹽並經 證實為單水合物(F4)。iH NMR (4〇〇 mHz,DMSO, 300K) δ 9.34 (1Η,s); 9·20 (1Η,broad s),8.58 (1Η,s),8.14 (2Η,d, J—8.8 Hz), 8.05 (2H,ddd,J=1.2,7.2,16.8 Hz), 7.93 (1H, s),7·52 (4H,dd,J=8.8,16.8 Hz),7.27 (1H,dd,J=6.8,8.0 Hz),7.13 (2H, d,J=8 Hz),3.48 (3H,m),3·10 (2H,m),2.90 (1H,m); 2.30 (3H,s),1.89 (2H,m),1.75 (2H,m)。 貧例 名稱 MW M+H+ 實例之程序 6 二氟乙酸3_{4_[7_(胺基羰基)_2Η·吲唑基]苯基} 六氫吼啶鏽 320 321 1 7 5-氟·2·(4-六氫吡啶-3-基苯基)-2H-吲唑-7-甲鹂胺 338 339 3 8 氣化(3S)-3_{4_[7_(胺基幾基)_2Η-σ引σ坐基]苯基} 六氫吼啶鏽 320 321 2 9 氟化(3R)_3-{4-[7_(胺基幾基)-2Η-叫卜坐基]苯基} 六氫吼啶鑌 320 321 2 10 (R)-5-氟-2-(4-六氫吼咬-3-基苯基)-2Η-σ弓卜坐-7-甲 醯胺 338 339 2 11 (8)-5-氟-2-(4-六氫°比咬-3-基本基)-2Η-σ引嗤-7-曱醢 胺 _ 338 339 ------ 2 12 (R)-5-氣-2-{3-氟-4-六虱σ比咬-3-基苯基}-2Η-α引唾· 7-甲醯胺 356 357 2 13 7-甲醯胺 _ 356 357 2 127700.doc -93-Ci9H18F2N40 specified value: 356, experimental value: 357 (M+H)+. Example 5 127700.doc -91 - 200836731 4-Mercaptobenzenesulfonic acid (3S)-3-{4-丨7_(aminocarbonyl)_2H•carbazole_2•ylphenyl”hexahydroindole (F4) Step 1: (3S)_3-[4-({(IE)_[3-(Methoxycarbonyl)- small nitrophenyl]methylene}amino)amino]hexahydropyrene _ 1 - formic acid tert-butyl ester (F1) as described in Example 1, B1, from Oct. 3 and Magic-3-(4-aminophenyl)hexahydropyridinium 1-carboxylic acid tert-butyl ester ( (F1) was prepared by splitting 3-(4-aminophenyl)-hexahydroacridine with 2 equivalents of L-dibenyl tartaric acid in Me〇H and then Boc-protection. Step 2 ···{{[[3S)-M-t-butoxycarbonyl)hexahydroacridine_3_yl]phenyl}-211-carbazole_7_carboxylic acid (F2) (1 eq) and sodium azide u eq) were slurried in dmF (0.25 M), inertized and 2,6-lutidine (1·〇eq) was added. The mixture was heated to an internal temperature of 110 ° C for 20 hours. The resulting brown solution was cooled to 20 ° C and THF and a 25 wt% aqueous solution of LiCl were added. The phases were separated and the organic portion was washed three more times with a 25 wt% aqueous solution of LiCl. 2.0 M NaOH (10 eq) was added to the above organic solution and the mixture was heated to 35 ° C for 20 hours, then cooled to 20 ° C and the phases were separated. The organic layer was washed with a mixture of 2.0M EtOAc and brine and the layers were partitioned. Step 3: (3S)-3-{4-[7-(Aminocarbonyl)-2H-indazol-2-yl]phenyl}hexahydropyridine-1-carboxylic acid tert-butyl ester (F3) F2 was dissolved in DC Μ (0.3 5 M) and di-tert-butyl carbonate (1.3 eq) and pyridine (1·〇eq) were added at RT. After 30 minutes, ammonium bicarbonate (1.3 eq) was added and stirring was continued for 20 hours. 1 M HCl (5 mL/g) was added and the phases were separated 127700.doc • 92-200836731. The organic layer was washed twice with water and concentrated to a smaller volume. The crude compound (F3) was filtered through a pad of diatomaceous earth and subsequently crystallized from methyl third-butyl ether. Step 4: 4-methylbenzenesulfonic acid (3S)_3_{4_[7-(aminocarbonyl>211_carbazole-2-yl)phenyl}hexahydrofluorene (F4) F3 is dissolved in THF ( Water (5%, relative to THF) was added to 0.15 M). p-Toluenesulfonic acid monohydrate (2·2 eq) was added and the mixture was heated to % C and stirred overnight. The desired solid salt was isolated by filtration and confirmed to be monohydrate (F4). iH NMR (4 〇〇mHz, DMSO, 300K) δ 9.34 (1 Η, s); 9·20 (1 Η, broad s), 8.58 (1 Η, s), 8.14 (2Η, d, J—8.8 Hz), 8.05 (2H, ddd, J=1.2, 7.2, 16.8 Hz), 7.93 (1H, s), 7.52 (4H, dd, J=8.8, 16.8 Hz), 7.27 (1H, dd, J=6.8, 8.0 Hz), 7.13 (2H, d, J=8 Hz), 3.48 (3H, m), 3·10 (2H, m), 2.90 (1H, m); 2.30 (3H, s), 1.89 (2H, m), 1.75 (2H, m). Poor name MW M+H+ Example procedure 6 Difluoroacetic acid 3_{4_[7_(aminocarbonyl)_2Η· Carbazolyl]phenyl} hexahydropyridinium rust 320 321 1 7 5-fluoro·2·(4-hexahydropyridin-3-ylphenyl)-2H-carbazole-7-formamide 338 339 3 8 Gasification (3S)-3_{4_[7_(Amino-based)_2Η-σ引σ基基]Phenyl} Hexahydropurine rust 3 20 321 2 9 fluorinated (3R)_3-{4-[7-(amino aryl)-2Η-called sylylene]phenyl} hexahydroacridinium 320 321 2 10 (R)-5-fluoro- 2-(4-hexahydroindole-3-ylphenyl)-2Η-σ 弓 坐-7-carbamimid 338 339 2 11 (8)-5-fluoro-2-(4-hexahydrogen ratio Bite-3-basic)-2Η-σ 嗤-7-nonylamine _ 338 339 ------ 2 12 (R)-5-gas-2-{3-fluoro-4-hexafluorene Than -3-ylphenyl}-2Η-α-induced salivary 7-carbamimid 356 357 2 13 7-carbamamine _ 356 357 2 127700.doc -93-
Claims (1)
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
GBGB0700432.8A GB0700432D0 (en) | 2007-01-10 | 2007-01-10 | Therapeutic compounds |
US92131007P | 2007-04-02 | 2007-04-02 |
Publications (2)
Publication Number | Publication Date |
---|---|
TW200836731A true TW200836731A (en) | 2008-09-16 |
TWI528961B TWI528961B (en) | 2016-04-11 |
Family
ID=37809722
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
TW097100730A TWI528961B (en) | 2007-01-10 | 2008-01-08 | Amide substituted indazoles as poly(adp-ribose)polymerase (parp) inhibitors |
Country Status (21)
Country | Link |
---|---|
CN (1) | CN101578279B (en) |
AT (1) | ATE502933T1 (en) |
CR (1) | CR10910A (en) |
DE (1) | DE602008005711D1 (en) |
DK (2) | DK2336120T3 (en) |
ES (3) | ES2728199T3 (en) |
GB (1) | GB0700432D0 (en) |
HN (1) | HN2009001260A (en) |
HU (1) | HUE044513T2 (en) |
LT (1) | LT2805945T (en) |
LU (1) | LUC00072I2 (en) |
ME (1) | ME03481B (en) |
MY (1) | MY147789A (en) |
NI (1) | NI200900135A (en) |
NZ (1) | NZ578256A (en) |
PE (1) | PE20081558A1 (en) |
PT (3) | PT2109608E (en) |
SI (2) | SI2109608T1 (en) |
TW (1) | TWI528961B (en) |
UA (1) | UA97658C2 (en) |
ZA (1) | ZA200903898B (en) |
Families Citing this family (13)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN106496187A (en) * | 2016-09-14 | 2017-03-15 | 陕西科技大学 | A kind of synthetic method for preparing PARP inhibitor Niraparib |
CN106432187A (en) * | 2016-09-17 | 2017-02-22 | 青岛云天生物技术有限公司 | Chiral resolution method of 2-[4-(-3-piperidyl)phenyl]-2H-indazole-7-formamide |
CN106432188A (en) * | 2016-09-17 | 2017-02-22 | 青岛辰达生物科技有限公司 | Preparation method of anti-cancer drug 2-[4-((3S)-3-Piperidinyl) phenyl]-2H-Indazole-7-Formamide |
CN106632244A (en) * | 2016-09-30 | 2017-05-10 | 陕西科技大学 | A novel synthetic method for preparing an anticancer medicine Niraparib |
CN106467513A (en) * | 2016-09-30 | 2017-03-01 | 陕西科技大学 | A kind of synthetic method preparing Niraparib |
CN108201537A (en) * | 2016-12-16 | 2018-06-26 | 苏州苏融生物医药有限公司 | A kind of Ni Lapani sustained and controlled release medicaments composition and application thereof |
KR20190130625A (en) * | 2017-03-27 | 2019-11-22 | 테사로, 인코포레이티드 | Niraprip formulation |
CN109081828B (en) * | 2017-06-14 | 2021-03-26 | 上海时莱生物技术有限公司 | Poly (ADP-ribose) polymerase inhibitor, preparation method and application |
CA3076907A1 (en) | 2017-09-26 | 2019-04-04 | Tesaro, Inc. | Niraparib formulations |
CN108084157A (en) * | 2018-02-12 | 2018-05-29 | 安庆奇创药业有限公司 | A kind of synthetic method of Ni Lapani |
CN108203404A (en) * | 2018-03-02 | 2018-06-26 | 上海博邦医药科技有限公司 | (R) synthetic method of -3- Phenylpiperidines or/and the chiral intermediate of (S) -3- Phenylpiperidines and Ni Lapani |
US20240016775A1 (en) | 2020-02-24 | 2024-01-18 | Fukang (Shanghai) Health Technology Co., Ltd | Anti-coronavirus application of poly adp ribose polymerase inhibitor |
CN115611860B (en) * | 2021-07-13 | 2024-06-11 | 上海博璞诺科技发展有限公司 | Method for synthesizing nilaparib |
-
2007
- 2007-01-10 GB GBGB0700432.8A patent/GB0700432D0/en not_active Ceased
-
2008
- 2008-01-08 DK DK11157369.7T patent/DK2336120T3/en active
- 2008-01-08 HU HUE14176452 patent/HUE044513T2/en unknown
- 2008-01-08 MY MYPI20092876A patent/MY147789A/en unknown
- 2008-01-08 PT PT08702101T patent/PT2109608E/en unknown
- 2008-01-08 LT LTEP14176452.2T patent/LT2805945T/en unknown
- 2008-01-08 SI SI200830240T patent/SI2109608T1/en unknown
- 2008-01-08 PE PE2008000097A patent/PE20081558A1/en active IP Right Grant
- 2008-01-08 NZ NZ578256A patent/NZ578256A/en unknown
- 2008-01-08 UA UAA200908335A patent/UA97658C2/en unknown
- 2008-01-08 AT AT08702101T patent/ATE502933T1/en active
- 2008-01-08 ES ES14176452T patent/ES2728199T3/en active Active
- 2008-01-08 CN CN2008800019265A patent/CN101578279B/en active Active
- 2008-01-08 DE DE602008005711T patent/DE602008005711D1/en active Active
- 2008-01-08 PT PT111573697T patent/PT2336120E/en unknown
- 2008-01-08 ES ES08702101T patent/ES2362214T3/en active Active
- 2008-01-08 ME MEP-2019-147A patent/ME03481B/en unknown
- 2008-01-08 SI SI200831266T patent/SI2336120T1/en unknown
- 2008-01-08 ES ES11157369.7T patent/ES2509115T3/en active Active
- 2008-01-08 TW TW097100730A patent/TWI528961B/en active
- 2008-01-08 PT PT14176452T patent/PT2805945T/en unknown
- 2008-01-08 DK DK08702101.0T patent/DK2109608T3/en active
-
2009
- 2009-06-04 ZA ZA200903898A patent/ZA200903898B/en unknown
- 2009-07-02 CR CR10910A patent/CR10910A/en unknown
- 2009-07-02 HN HN2009001260A patent/HN2009001260A/en unknown
- 2009-07-02 NI NI200900135A patent/NI200900135A/en unknown
-
2018
- 2018-05-03 LU LU00072C patent/LUC00072I2/en unknown
Also Published As
Publication number | Publication date |
---|---|
LT2805945T (en) | 2019-06-25 |
ATE502933T1 (en) | 2011-04-15 |
PT2109608E (en) | 2011-05-30 |
NZ578256A (en) | 2011-12-22 |
GB0700432D0 (en) | 2007-02-21 |
CN101578279A (en) | 2009-11-11 |
SI2109608T1 (en) | 2011-07-29 |
DE602008005711D1 (en) | 2011-05-05 |
LUC00072I2 (en) | 2018-07-04 |
MY147789A (en) | 2013-01-31 |
CR10910A (en) | 2010-01-19 |
HUE044513T2 (en) | 2019-10-28 |
ES2509115T3 (en) | 2014-10-17 |
ME03481B (en) | 2020-01-20 |
UA97658C2 (en) | 2012-03-12 |
ZA200903898B (en) | 2010-04-28 |
PT2336120E (en) | 2014-10-01 |
DK2109608T3 (en) | 2011-07-11 |
HN2009001260A (en) | 2012-03-26 |
PE20081558A1 (en) | 2008-10-30 |
ES2728199T3 (en) | 2019-10-22 |
PT2805945T (en) | 2019-06-06 |
SI2336120T1 (en) | 2014-10-30 |
NI200900135A (en) | 2010-08-30 |
ES2362214T3 (en) | 2011-06-29 |
DK2336120T3 (en) | 2014-10-06 |
TWI528961B (en) | 2016-04-11 |
CN101578279B (en) | 2013-07-17 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
TW200836731A (en) | Amide substituted indazoles as poly (ADP-ribose) polymerase (PARP) inhibitors | |
CA2674436C (en) | Amide substituted indazoles as poly(adp-ribose)polymerase (parp) inhibitors | |
EP2240466B1 (en) | Pharmaceutically acceptable salts of 2-{4-[(3s)-piperidin-3- yl]phenyl} -2h-indazole-7-carboxamide | |
ES2524787T3 (en) | Pyridazinone derivatives as PARP inhibitors | |
TW201210592A (en) | Substituted 5-fluoro-1H-pyrazolopyridines and their use | |
TWI522352B (en) | Amids substituted indazole derivatives as poly (adp-ribose) polymerase inhibitors | |
TR201907948T4 (en) | Amide-substituted indazoles as poly (ADP-ribose) polymerase (PARP) inhibitors. |