TW201938171A - Tricyclic compounds as vasopressin V1a receptor antagonists - Google Patents

Tricyclic compounds as vasopressin V1a receptor antagonists Download PDF

Info

Publication number
TW201938171A
TW201938171A TW107144825A TW107144825A TW201938171A TW 201938171 A TW201938171 A TW 201938171A TW 107144825 A TW107144825 A TW 107144825A TW 107144825 A TW107144825 A TW 107144825A TW 201938171 A TW201938171 A TW 201938171A
Authority
TW
Taiwan
Prior art keywords
dihydro
triazolo
trans
cyclohexyl
chloro
Prior art date
Application number
TW107144825A
Other languages
Chinese (zh)
Inventor
殷雷 巴塔
梭特 澤爾克奇
克里斯蒂娜 容黛柯達斯
蓋伯 桑圖
Original Assignee
匈牙利商羅特格登公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from HU1700522A external-priority patent/HUP1700522A1/en
Priority claimed from HU1800332A external-priority patent/HUP1800332A1/en
Application filed by 匈牙利商羅特格登公司 filed Critical 匈牙利商羅特格登公司
Publication of TW201938171A publication Critical patent/TW201938171A/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/12Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains three hetero rings
    • C07D487/14Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D223/00Heterocyclic compounds containing seven-membered rings having one nitrogen atom as the only ring hetero atom
    • C07D223/02Heterocyclic compounds containing seven-membered rings having one nitrogen atom as the only ring hetero atom not condensed with other rings
    • C07D223/06Heterocyclic compounds containing seven-membered rings having one nitrogen atom as the only ring hetero atom not condensed with other rings with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D223/00Heterocyclic compounds containing seven-membered rings having one nitrogen atom as the only ring hetero atom
    • C07D223/02Heterocyclic compounds containing seven-membered rings having one nitrogen atom as the only ring hetero atom not condensed with other rings
    • C07D223/06Heterocyclic compounds containing seven-membered rings having one nitrogen atom as the only ring hetero atom not condensed with other rings with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D223/08Oxygen atoms
    • C07D223/10Oxygen atoms attached in position 2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pain & Pain Management (AREA)
  • Psychiatry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)

Abstract

The present invention relates to 5,6-dihydro-4H-[1,2,4]triazolo[4,3-a][1] benzazepine derivatives of general formula (I) and/or salts thereof and/or geometric isomers thereof and/or stereoisomers thereof and/or enantiomers thereof and/or racemates thereof and/or diastereomers thereof and/or biologically active metabolites thereof and/or prodrugs thereof and/or solvates thereof and/or hydrates thereof and/or polymorphs thereof which are centrally and/or peripherally acting V1a receptor modulators, particularly V1a receptor antagonists. Additional subject of the present invention is the process for the preparation of the compounds and the intermediates of the preparation process as well. The invention also relates to the pharmaceutical compositions containing the compounds or together with one or more other active substances, as well as to the use in the treatment and/or prophylaxis of a disease or condition associated with V1a receptor function.

Description

作為血管升壓素V1a受體拮抗劑之三環化合物Tricyclic compounds as vasopressin V1a receptor antagonists

本發明關於通式(I)之5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯衍生物類及/或彼等之鹽及/或彼等之幾何異構物及/或彼等之立體異構物及/或彼等之鏡像異構物及/或彼等之消旋物及/或彼等之非鏡像異構物及/或彼等之生物活性代謝物及/或彼等之前藥及/或彼等之溶劑合物及/或彼等之水合物及/或彼等之多形體,該等係中樞及/或周圍作用性V1a受體調節劑,特別是V1a受體拮抗劑。本發明之另一標的係製備該等化合物之方法和該製備之過程的中間體。本發明亦關於含有該等化合物之醫藥組成物和該醫藥組成物於治療及/或預防與V1a受體功能有關之疾病或病症之用途。The present invention relates to 5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazine derivatives and / or the like of the general formula (I) Salts and / or their geometric isomers and / or their stereoisomers and / or their mirror isomers and / or their racemates and / or their non-mirror isomers Structures and / or their biologically active metabolites and / or their prodrugs and / or their solvates and / or their hydrates and / or their polymorphs, these are central and And / or peripherally acting V1a receptor modulators, particularly V1a receptor antagonists. Another subject of the present invention is a method for preparing the compounds and an intermediate for the process of the preparation. The present invention also relates to a pharmaceutical composition containing the compounds and the use of the pharmaceutical composition for the treatment and / or prevention of diseases or conditions related to the function of the Via receptor.

血管升壓素(抗利尿激素;ADH; CYIQNCPRG)係9個胺基酸肽激素,其係由下視丘之室旁核(PVN)和視上核(SON)的巨大細胞神經元產製並被直接分泌至腦下垂體之後葉,且血管升壓素被儲存於該後葉直至進入血液。於周圍組織,血管升壓素之主要作用在於血管收縮、葡萄糖代謝及排泄調節。
據此,因血管升壓素之不適當分泌而缺少血管升壓素所引起之病症可導致身體之病理變化,諸如中樞性尿崩症或異常低血壓(hypotension),而於血管升壓素之濃度升高或外源性給藥之情況下,可觀察到多種型態的攻擊行為之強化(Ferris et al.,BMC Neuroscience 2008, 9:111)。
催產素(OXT; CYIQNCPLG)係血管升壓素相關肽激素,其與血管升壓素之差1個胺基酸,且催產素之受體亦與血管升壓素之受體相關。化合物對於催產素受體之作用顯示種屬特異性差異,但是於不同哺乳動物種屬體內的催產素激素本身係相同。同樣地,於所有哺乳動物(除了有袋動物和豬之外)體內的血管升壓素肽係相同且該血管升壓素肽通過彼之受體所顯現之作用亦呈種屬特異性差異。施加於中樞神經系統之催產素的抗焦慮作用係為習知(Neumann ID.J Neuroendocrinol 2008, 20(6): 858-65),因此對於中樞神經系統之催產素受體的抑制作用可引發為不欲之副作用的焦慮。
可區分3種血管升壓素受體,彼等皆為G蛋白偶合受體。其中,V1a受體(V1aR)主要表現於大腦皮層、海馬及腦下垂體且另周圍地表現於肝、血管平滑肌、肺、子宮及睪丸(Frank et al.,Eur J Pharmacol 2008, 583:226-42)。於皮質、海馬及腦下垂體亦可發現V1b受體(V1bR)且於周圍組織該V1bR於調節胰腺和腎上腺上扮演重要角色。相對地,V2受體(V2R)主要局限於周圍組織且於腎臟內增加水再吸收,因而實現血管升壓素之抗利尿作用(Robben et al.,Am J Physiol Renal Physiol 2007, 292(1): F253-60)。由於水平衡調節之改變,對於該V2受體之作用可引起不欲之副作用。
該V1a和V1b受體之次級訊號傳導途徑包括通過磷脂酸肌醇之細胞內Ca2+ 濃度的變化,而該V2受體活化腺苷酸環化酶並影響cAMP濃度(Gouzenes et al., J Physiol 1999, 517(Pt3):771-9;Tahara et al.,Pflugers Arch 1999, 437 (2):219-26)。
與該V1a受體連接之重要作用係調節晝夜節律。視交叉上核(SCN)之神經元中1/3表現血管升壓素且於該腦區域該V1a受體之mRNA顯現晝夜波動,其中可於夜晚時間觀察到該mRNA之最高值(de Vries and Miller,Prog Brain Res 1998, 119:3-20)。儘管該V1aR mRNA之分佈和量於男性和女性體內並無不同,血管升壓素於引起行為習性上顯現性別差異(Szot et al.,Brain Res Mol Brain Res 1994, 24 (1-4):1-10)。小鼠實驗業已證實:於小鼠睡眠期間之前,藉由該小鼠之體內生理時鐘而非生理需要可引起增加水吸收(Gizowski et al.,Nature 2016, 537(7622):685-8)。睡眠障礙係自閉症之主要伴隨症狀(Glickman,Neurosci Biobehav Rev 2010, 34(5):755-68)。
血管升壓素於腦內作為神經調節劑,且在壓力下於杏仁核可偵測到升高之血管升壓素濃度(Ebner et al.,Eur J Neurosci 2002, 15(2):384-8)。已知壓力/緊張生活狀態會增加發生抑鬱和焦慮之可能性(Kendler et al.,Arch Gen Psychiatry 2003, 60(8):789-96;Simon et al., Recent Pat CNS Drug Discov , 2008, 3(2):77-93;Egashira et al.,J Pharmacol Sci 2009, 109(1):44-9;Bielsky et al.,Neuropsychopharmacology 2004, 29(3):483-93)。V1aR於腦內高度表現,特別是於邊緣系統之某些部分,諸如杏仁核、側隔及海馬,該等部分於焦慮之發展上扮演重要角色。雄性V1aR基因剔除小鼠於舉臂式十字迷宮(elevated plus maze)試驗、曠野試驗及明暗箱試驗顯現焦慮之減輕,但是並未於雌性小鼠觀察到此等差異(Bielsky et al.,Behav Brain Res 2005, 164(1):132-6)。
雄性V1aR剔除小鼠於運動表現上未顯現任何表型差異。於一般光暗循環實驗,V1aR剔除小鼠與彼等之野生型同窩小鼠相比較並未顯現差異,然而於連續黑暗下進行之實驗中,該V1a剔除小鼠之晝夜節律明顯改變(Egashira et al.,Behav Brain Res 2007, 178(1):123-7)。
該V1aR剔除小鼠於前脈衝抑制(prepulse inhibition)試驗顯現改善之活性,該前脈衝抑制試驗已被接受為於大多數精神分裂症患者所觀察到之感覺性運動缺陷的動物模式。於適合測量該雄性和雌性二者之V1aR剔除小鼠的社會認知行為之社會互動試驗中,Egashira等人業已顯示功能之降低,但是於經拮抗劑治療後未觀察到該功能之降低(Bleickard et al.,Psychopharmacology (Berl) , 2009, 202: 711-18)。
於編碼V1a受體之AVPR1A基因變異體之案例中,可測量與自閉症有關之2個微小多型性(Kim et al.,Mol Psychiatry 2002, 7:503-7;Yirmiya et al.,Mol Psychiatry 2006, 11: 488-94;Yang et al.,Psychiatry Res , 2010, 178 (1):199-201;Yang et al.,Neurosci Lett 2010, 479(3):197-200)。於帶有該V1aR基因之2個危險等位基因之病患體內可偵測到的改變之杏仁核的活化亦涉及遺傳連接。業已顯示該等經修飾之受體能於情緒臉部認知過程期間改變杏仁核之活化閾值(Meyer-Lindenberg et al.,Mol Psychiatry 2009, 14: 968-75)。
臨床前實驗數據亦支持V1aR拮抗劑於自閉症之功效。廣泛被使用且被接受之自閉症的動物模式係研究於子宮內經丙戊酸鹽(VPA)處理之大鼠的行為。該經VPA處理之動物的降低之社會行為可藉由V1aR拮抗劑化合物回復至正常值。於功能性磁共振顯影研究中,亦發現藉由該V1aR拮抗劑可回復出生前經VPA處理之動物的不同腦區域中降低之灌注值。經該V1aR拮抗劑治療可增加皮質、下丘、海馬及下視丘之降低的功能,然而於腹側蓋、紋狀體及上丘,藉由該V1aR拮抗劑使增強之灌注正常化(Grundschober et al., Poster presented atAnnual Meeting of the American College of Neuropsychopharmacology , 2014, Phoenix, USA)。基於此理由,預期能顯現有利於血腦障壁穿透之V1aR拮抗劑化合物將會是有優勢的。
使用小分子拮抗劑影響V1aR係一種用於治療雌性性器官之多種病理症狀(諸如但不限於痛經和性功能障礙)、血壓控制之持久性病理症狀(諸如但不限於高血壓及/或慢性心臟衰竭)及因血管升壓素之不適當分泌所引起之症狀(諸如但不限於尿崩症、腎衰竭、腎病症候群及肝硬化)的有前景之策略。使用小分子拮抗劑影響V1aR可被視為用於治療焦慮、抑鬱、攻擊行為及中樞神經系統疾病(其中該疾病之多種症狀之一及/或多種症狀可能與前述3種疾病有關或與該前述3種疾病顯示合併症)的另一有前景之策略。該等症狀包括但不限於自閉症類群障礙(良好功能自閉症、亞斯伯格(Asperger)症候群、待分類的廣泛性發展障礙(PDD-NOS)、自閉症譜系障礙(ASD)及彼之多種不同症候型:X染色體易裂症、普瑞德威利(Prader-Willi)症候群、蕾特(Rett)氏症及結節性硬化症)、強迫症(OCD)、多種不同類型之唐(Down)氏綜合症及創傷後應激障礙(PTSD)。V1aR拮抗劑亦適用於治療攻擊性行為障礙及/或易怒(諸如但不限於罹患ASD、亨丁頓舞蹈症(Huntington’s disease;HD)或多種不同類型之精神分裂症的病患)、行為過動症(諸如但不限於注意力不足過動症(ADHD))、認知障礙(諸如但不限於癡呆、輕度認知障礙(MCI)、與精神分裂症有關之認知障礙(CIAS)及阿茲海默(Alzheimer)症)及其他神經精神疾病(諸如但不限於精神分裂症和相關疾病)。
許多專利申請案涉及V1a受體拮抗劑,例如大塚製藥株式會社(Otsuka)揭露苯並雜環衍生物(WO 95/034540 A1;WO 2009/001968 A1;WO 2011/052519 A1),安斯泰來製藥公司(Astellas Pharma;山之內)揭露稠合之苯並二吖呯和三唑衍生物(WO 95/03305 A1;WO 01/87855 A1;WO 02/44179 A1),艾伯維生物製藥公司(AbbVie)揭露羥吲哚衍生物(WO 2006/072458 A2;WO 2006/100082 A2),拜耳製藥公司(Bayer Pharma)揭露芳基或雜芳基三唑衍生物(WO 2017/191102 A1;WO 2017/191107 A1;WO 2017/191114 A1)。含有苯並薁核之多種不同衍生物(WO 2005/068466 A1;WO 2006/021213 A2;WO 2006/021882 A1;WO 2011/114109 A1;WO 2011/128265 A1;WO 2011/141396 A1;WO 2014/127350 A1)、螺吲哚啉酮及吲哚基羰基衍生物(WO 97/15556 A1;WO 2007/009906 A1;WO 2007/014851 A2)亦被描述作為V1a受體拮抗劑。
首度臨床開發考慮V1a受體作為外圍標的,因此低劣之腦穿透性有利於化合物開發。該化合物係賽諾菲製藥廠(Sanofi)之吲哚啉核化合物“瑞考伐普坦(relcovaptan)”(SR-49059, WO 93/03013 A1),其被發展至第2期臨床試驗。研究之適應症係早產、經期期間觀察到之骨盆疼痛、痛經(Brouard et al.,Br J Obstetr Gynaecol 2000, 107:614-9)、心臟衰竭、高血壓及冠狀動脈痙攣,但亦於小細胞肺癌經測試作為抗腫瘤劑直至於2003年中止最後之臨床試驗(Serradeil-Le Gal et al.,Prog Brain Res 2002, 139:197-210;AdisInsight: Relcovaptan - 2006年10月3日最後資料更新http://adisinsight.springer.com/drugs/800004942)。已自1993年起進行瑞考伐普坦之臨床開發且現今為V1aR研究中最常使用之活體外工具(Tahara et al.,Br J Pharmacol 2000, 129:131-9)。
基於作為有效之V1aR拮抗劑所測量之數據,輝瑞藥廠(Pfizer)針對痛經研究三唑衍生物PF 00738245 (WO 2005/063754 A1)和三唑並苯並二吖呯核之化合物PF-184563 (WO 2004/074291 A1)之臨床前開發(Russell et al.,Eur. J Pharmacol 2011, 670(2): 347-355;Johnson et al.,Bioorg Med Chem Lett 2011, 21:5684-7),但是彼等之開發業已中止。
藉由檢驗對於中樞神經系統所顯現之功效,亦已提出治療抑鬱和焦慮為新治療領域。強生公司(Johnson & Johnson)之螺苯並吖呯核化合物JNJ-17308616係第1個作用於中樞神經系統之V1aR拮抗劑化合物(Bleickard et al.,Psychopharmacology (Berl.), 2009, 202:711-18;WO 02/02531 A1),其經證實於用於焦慮研究的多種不同之動物模式中顯現功效:於大鼠幼崽之舉臂式十字迷宮試驗、玻璃球埋藏試驗及分離引起之超聲波發聲中顯現顯著減輕之焦慮行為。雖然該螺苯並吖呯核化合物JNJ-17308616經證實能有效地影響升高之O-迷宮和條件舔反應,但是基於彼於囓齒動物體內所測量之低劣的代謝安定性,彼之功效不佳且僅可於高劑量下被測量,因此難於進行測試。
艾日凡製藥廠(Azevan)之V1aR拮抗劑四氫吖唉酮衍生物SRX246和SRX251(亦稱為API246或API251;WO 03/031407 A2)亦達到臨床試驗階段。SRX246之臨床試驗現今亦正在進行以治療攻擊行為、及亨丁頓舞蹈症和創傷後應激障礙之間歇性暴怒和易怒以及焦慮和恐懼之人體行為模式(AdisInsight: SRX 246 - 2017年2月16日最後資料更新http://adisinsight.springer.com/drugs/800023656)。SRX251之臨床試驗治療痛經,但是第1期研究於2016年中止,且同樣地於臨床前開發研究SRX246針對攻擊行為之功效(AdisInsight: SRX 251 - 2017年11月4日最後資料更新http://adisinsight.springer.com/drugs/800025117)。SRX-246和SRX-251對人V1a受體具有活性且該二者化合物於大鼠腦部測量之濃度為彼等於結合分析所測量之有效濃度約100倍(Guillon et al.,Bioorg Med Chem 2007, 15:2054-80;Fabio et al.,J Pharm Sci 2013, 102(6):2033-43)。
凡替拉製藥廠(Vantia)之吡唑並苯並二吖呯核的V1aR拮抗劑化合物VA 111913 (WO 2010/097576 A1;AdisInsight: VA 111913-2015年8月25日最後資料更新http://adisinsight.springer.com/drugs/800028777)業已於第2期臨床試驗進行測試以治療痛經,但是自2015年以後無該化合物之開發資料。
大塚製藥株式會社之V1aR拮抗劑“喹啉酮衍生物OPC 21268”(EP 0382185 A2;AdisInsight: OPC 21268-2006年10月6日最後資料更新http://adisinsight.springer.com/drugs/800000284) 於臨床前階段測試對胃黏膜損傷適應症之功效,且於第2期臨床試驗研究對心臟衰竭和高血壓之功效,但是自2015年以後無該化合物之開發資料(Yamamura et al.,Science 1991, 252:572;Serradeil-Le Gal et al.,J Clin Invest 1993, 92(1):224)。
當於死後人體樣本檢查腦幹時,於前置核(nucleus prepositus)可偵測到與催產素受體無關之V1a受體的選擇性局部化,該前置核於視線安定性上扮演某種角色(Freeman et al.,Soc Neurosci 2017, 12(2):113-123)。人類社會行為所需要之基本技能係生物相關資料之認知和眼動追蹤(Klin et al.,Nature 2009, 459:257-63;Simion et al.,PNAS 2008, 105(2):809-13)。最為積極之V1aR研究藥廠Hoffmann-La Roche的吲哚衍生物RO5028442已進入第1期研究(RG-7713;WO 2007/006688 A1),其中於人體可偵測到對視線型態取向之正面功效(Umbricht et al.,Neuropsychopharmacology 2017, 42 (9):1914-1923;AdisInsight: RG 7713-2015年11月5日最後資料更新http://adisinsight.springer.com/drugs/800043668)。具有三唑並苯並二吖呯核之巴羅非田(balovaptan)正進行治療自閉症之第2期臨床試驗(RG-7314, RO5285119;WO 2010/060836 A1;AdisInsight: RG 7314 - 2017年9月10日最後資料更新http://adisinsight.springer.com/drugs/800035102)。
儘管許多V1aR拮抗劑化合物和臨床研究,未獲滿足的醫療需求仍然持續以需要開發V1aR拮抗劑,該V1aR拮抗劑能適合於治療及/或預防雌性性器官之多種病理症狀、血壓控制之持久性症狀、因血管升壓素之不適當分泌所引起之症狀、焦慮、抑鬱、攻擊行為、中樞神經系統疾病(其中該疾病之多種症狀之一及/或多種症狀可能與焦慮、抑鬱或攻擊行為有關或與該前述3種疾病顯示合併症(自閉症類群障礙、強迫症、多種不同類型之唐氏綜合症及創傷後應激障礙))、攻擊性行為障礙及/或易怒、行為過動症、認知障礙或其他神經精神疾病。
Angiotensin (antidiuretic hormone; ADH; CYIQNCPRG) is a 9 amino acid peptide hormone, which is produced by giant cell neurons in the paraventricular nucleus (PVN) and supraoptic nucleus (SON) of the hypothalamus. It is secreted directly into the posterior lobe of the pituitary gland, and vasopressin is stored in the posterior lobe until it enters the blood. In the surrounding tissues, vasopressin's main role is to regulate vasoconstriction, glucose metabolism and excretion.
Accordingly, conditions caused by the lack of vasopressin due to improper secretion of vasopressin can lead to pathological changes in the body, such as central diabetes insipidus or abnormal hypotension. In the case of increased concentrations or exogenous administration, a variety of forms of aggressive behavior can be observed (Ferris et al., BMC Neuroscience 2008, 9: 111).
Oxytocin (OXT; CYIQNCPLG) is a vasopressin-related peptide hormone, which differs from vasopressin by 1 amino acid, and the receptor of oxytocin is also related to the receptor of vasopressin. The compound's effect on the oxytocin receptor shows species-specific differences, but the oxytocin hormone itself is the same in different mammal species. Similarly, the vasopressin peptides are the same in all mammals (except marsupials and pigs) and the effects of the vasopressin peptide through its receptors are species-specific. The anxiolytic effect of oxytocin applied to the central nervous system is known (Neumann ID. J Neuroendocrinol 2008, 20 (6): 858-65), so the inhibitory effect on the oxytocin receptor of the central nervous system can be triggered as Unwanted side effects of anxiety.
Three types of vasopressin receptors can be distinguished, all of which are G protein-coupled receptors. Among them, the V1a receptor (V1aR) is mainly expressed in the cerebral cortex, hippocampus, and pituitary gland and other peripherally in liver, vascular smooth muscle, lung, uterus, and testes (Frank et al., Eur J Pharmacol 2008, 583: 226- 42). V1b receptors (V1bR) can also be found in the cortex, hippocampus, and pituitary gland, and this V1bR plays an important role in regulating the pancreas and adrenals in surrounding tissues. In contrast, V2 receptors (V2R) are mainly confined to surrounding tissues and increase water reabsorption in the kidney, thereby achieving the anti-diuretic effect of vasopressin (Robben et al., Am J Physiol Renal Physiol 2007, 292 (1) : F253-60). Due to changes in water balance regulation, effects on the V2 receptor can cause unwanted side effects.
The secondary signal transduction pathways of the V1a and V1b receptors include changes in intracellular Ca 2+ concentration through phosphatidylinositol, and the V2 receptor activates adenylate cyclase and affects cAMP concentration (Gouzenes et al ., J Physiol 1999, 517 (Pt3): 771-9; Tahara et al., Pflugers Arch 1999, 437 (2): 219-26).
An important role linked to this V1a receptor is to regulate circadian rhythm. One third of the neurons in the suprachiasmatic nucleus (SCN) show vasopressin and the mRNA of the V1a receptor in the brain region shows diurnal fluctuations, and the highest value of the mRNA can be observed at night time (de Vries and Miller, Prog Brain Res 1998, 119: 3-20). Although the distribution and amount of this V1aR mRNA is not different in men and women, vasopressin shows a gender difference in behavioral habits (Szot et al., Brain Res Mol Brain Res 1994, 24 (1-4): 1 -10). Experiments in mice have been shown to increase the water absorption by the mouse's internal physiological clock rather than its physiological needs before the mouse sleeps (Gizowski et al., Nature 2016, 537 (7622): 685-8). Sleep disorders are the main concomitant symptoms of autism (Glickman, Neurosci Biobehav Rev 2010, 34 (5): 755-68).
Vasopressin acts as a neuromodulator in the brain, and elevated vasopressin concentrations can be detected in the amygdala under stress (Ebner et al., Eur J Neurosci 2002, 15 (2): 384-8 ). Stress / stressful living conditions are known to increase the likelihood of depression and anxiety (Kendler et al., Arch Gen Psychiatry 2003, 60 (8): 789-96; Simon et al., R ecent Pat CNS Drug Discov , 2008, 3 (2): 77-93; Egashira et al., J Pharmacol Sci 2009, 109 (1): 44-9; Bielsky et al., Neuropsychopharmacology 2004, 29 (3): 483-93). V1aR is highly expressed in the brain, especially in parts of the limbic system, such as the amygdala, lateral septum, and hippocampus, which play an important role in the development of anxiety. Male V1aR knockout mice showed reduced anxiety in the raised plus maze test, the open field test, and the light and dark box test, but these differences were not observed in female mice (Bielsky et al., Behav Brain Res 2005, 164 (1): 132-6).
Male V1aR knockout mice showed no phenotypic differences in athletic performance. In general light-dark cycle experiments, V1aR-excluded mice did not show a difference compared to their wild-type littermates. However, in experiments performed under continuous darkness, the circadian rhythm of the V1a-excluded mice changed significantly (Egashira et al., Behav Brain Res 2007, 178 (1): 123-7).
The V1aR-excluded mice showed improved activity in a prepulse inhibition test, which has been accepted as the animal model of sensorimotor deficits observed in most patients with schizophrenia. Egashira et al. Have shown a decrease in function in a social interaction test suitable for measuring the social cognitive behavior of V1aR knockout mice in both male and female (Bleickard et al. al., Psychopharmacology (Berl) , 2009, 202: 711-18).
In the case of the AVPR1A gene variant encoding the V1a receptor, two minor polymorphisms related to autism can be measured (Kim et al., Mol Psychiatry 2002, 7: 503-7; Yirmiya et al., Mol Psychiatry 2006, 11: 488-94; Yang et al., Psychiatry Res , 2010, 178 (1): 199-201; Yang et al., Neurosci Lett 2010, 479 (3): 197-200). The detectable altered activation of the amygdala in patients with the two dangerous alleles of the V1aR gene also involves genetic linkage. These modified receptors have been shown to alter the activation threshold of the amygdala during the emotional facial recognition process (Meyer-Lindenberg et al., Mol Psychiatry 2009, 14: 968-75).
Preclinical experimental data also support the efficacy of V1aR antagonists in autism. The widely used and accepted animal model of autism is to study the behavior of valproate (VPA) treated rats in the womb. The reduced social behavior of the VPA-treated animal can be restored to normal values by the VIaR antagonist compound. In functional magnetic resonance imaging studies, it was also found that the V1aR antagonist can restore reduced perfusion values in different brain regions of VPA-treated animals before birth. Treatment with this V1aR antagonist can increase the reduced function of the cortex, inferior colliculus, hippocampus, and inferior optic cumulus, but in the ventral striatum, striatum, and superior colliculus, enhanced perfusion is normalized by the V1aR antagonist (Grundschober et al., Poster presented at Annual Meeting of the American College of Neuropsychopharmacology , 2014, Phoenix, USA). For this reason, it is expected that V1aR antagonist compounds that will appear to facilitate blood-brain barrier penetration will be advantageous.
The use of small molecule antagonists to influence V1aR is a variety of pathological symptoms (such as, but not limited to, dysmenorrhea and sexual dysfunction) for the treatment of female sexual organs, persistent pathological symptoms of blood pressure control (such as, but not limited to, hypertension and / or chronic heart failure) ) And promising strategies for symptoms caused by improper secretion of vasopressin, such as, but not limited to, diabetes insipidus, renal failure, kidney syndrome, and cirrhosis. The use of small molecule antagonists to influence V1aR can be considered for the treatment of anxiety, depression, aggressive behavior, and central nervous system diseases (wherein one of the symptoms of the disease and / or multiple symptoms may be related to the aforementioned 3 diseases or related to the aforementioned 3 diseases show comorbidities) is another promising strategy. Such symptoms include, but are not limited to, autism group disorders (good functioning autism, Asperger syndrome, generalized developmental disorder to be classified (PDD-NOS), autism spectrum disorder (ASD), and Many different syndromes: Fragile X chromosome, Prader-Willi syndrome, Rett's disease and tuberous sclerosis), OCD, many different types of Tang (Down) syndrome and post-traumatic stress disorder (PTSD). V1aR antagonists are also suitable for the treatment of aggressive behavior disorders and / or irritability (such as, but not limited to, patients with ASD, Huntington's disease (HD), or many different types of schizophrenia), behavioral hyperactivity Disorders (such as, but not limited to, ADHD), cognitive disorders (such as, but not limited to, dementia, mild cognitive impairment (MCI), schizophrenia-related cognitive impairment (CIAS), and Alzheimer's (Alzheimer's disease) and other neuropsychiatric diseases (such as, but not limited to, schizophrenia and related diseases).
Many patent applications involve V1a receptor antagonists, such as Otsuka's disclosure of benzoheterocyclic derivatives (WO 95/034540 A1; WO 2009/001968 A1; WO 2011/052519 A1), Astellas Pharmaceutical company (Astellas Pharma; Yamanouchi) reveals fused benzodiazepine and triazole derivatives (WO 95/03305 A1; WO 01/87855 A1; WO 02/44179 A1), AbbVie Biopharmaceutical Company (AbbVie) discloses oxindole derivatives (WO 2006/072458 A2; WO 2006/100082 A2), Bayer Pharma discloses aryl or heteroaryltriazole derivatives (WO 2017/191102 A1; WO 2017 / 191107 A1; WO 2017/191114 A1). Many different derivatives containing benzofluorene cores (WO 2005/068466 A1; WO 2006/021213 A2; WO 2006/021882 A1; WO 2011/114109 A1; WO 2011/128265 A1; WO 2011/141396 A1; WO 2014 / 127350 A1), spirolindolinone and indolylcarbonyl derivatives (WO 97/15556 A1; WO 2007/009906 A1; WO 2007/014851 A2) have also been described as V1a receptor antagonists.
For the first clinical development, the V1a receptor was considered as a peripheral target, so poor brain penetration is conducive to compound development. This compound is an indololine nuclear compound "relcovaptan" (SR-49059, WO 93/03013 A1) of Sanofi Pharmaceutical Factory (Sanofi), which has been developed to a Phase 2 clinical trial. The indications studied were premature delivery, pelvic pain observed during menstruation, dysmenorrhea (Brouard et al., Br J Obstetr Gynaecol 2000, 107: 614-9), heart failure, hypertension, and coronary spasm, but also in small cells Lung cancer has been tested as an anti-tumor agent until the last clinical trial was suspended in 2003 (Serradeil-Le Gal et al., Prog Brain Res 2002, 139: 197-210; AdisInsight: Relcovaptan-October 3, 2006 Final Information Update http : //adisinsight.springer.com/drugs/800004942). The clinical development of racuvaptan has been carried out since 1993 and is now the most commonly used in vitro tool in V1aR studies (Tahara et al., Br J Pharmacol 2000, 129: 131-9).
Based on data measured as a potent V1aR antagonist, Pfizer has studied triazole derivatives PF 00738245 (WO 2005/063754 A1) and triazobenzobenzodiazepine core compound PF-184563 ( WO 2004/074291 A1) preclinical development (Russell et al., Eur. J Pharmacol , 2011, 670 (2): 347-355; Johnson et al., Bioorg Med Chem Lett 2011, 21: 5684-7), However, their development has been suspended.
By examining the effects shown on the central nervous system, the treatment of depression and anxiety has also been proposed as a new therapeutic area. Johnson &Johnson's spirobenzoacridine nuclear compound JNJ-17308616 is the first V1aR antagonist compound that acts on the central nervous system (Bleickard et al., Psychopharmacology (Berl.), 2009, 202: 711- 18; WO 02/02531 A1), which has been shown to show efficacy in a variety of different animal models used for anxiety research: raised-arm cross maze test in rat pups, glass bulb burying test, and ultrasonic vocalization caused by separation Significantly reduced anxiety behavior. Although the spirobenzoacridine nuclear compound JNJ-17308616 has been proven to effectively affect the elevated O-maze and conditional licking response, it has poor efficacy due to its poor metabolic stability as measured in rodents. It can only be measured at high doses, making testing difficult.
Tetrahydroacridone derivatives SRX246 and SRX251 (also known as API246 or API251; WO 03/031407 A2), V1aR antagonists of Aivan Pharmaceutical Factory (Azevan), have also reached the clinical trial stage. Clinical trials of SRX246 are also currently underway to treat human behavior patterns of intermittent anger and irritability and anxiety and fear in the treatment of aggressive behavior and Huntington's disease and post-traumatic stress disorder (AdisInsight: SRX 246-February 2017 The last update on the 16th is http://adisinsight.springer.com/drugs/800023656). The clinical trial of SRX251 treats dysmenorrhea, but the first phase of the study was discontinued in 2016, and the efficacy of SRX246 against aggression was similarly developed in the preclinical setting (AdisInsight: SRX 251-November 4, 2017 Last update of data http: // adisinsight.springer.com/drugs/800025117). SRX-246 and SRX-251 are active at human V1a receptors and the concentrations of these two compounds measured in the rat brain are about 100 times their effective concentrations measured by binding analysis (Guillon et al., Bioorg Med Chem 2007 15: 2054-80; Fabio et al., J Pharm Sci 2013, 102 (6): 2033-43).
V1aR antagonist compound of pyrazolobenzodiazepine core of Vantila Pharmaceutical Factory (Vantia) VA 111913 (WO 2010/097576 A1; AdisInsight: VA 111913-August 25, 2015 Last update of the data http: // (adisinsight.springer.com/drugs/800028777) has been tested in a Phase 2 clinical trial to treat dysmenorrhea, but no data on the development of this compound has been available since 2015.
Otsuka Pharmaceutical Co., Ltd.'s V1aR antagonist "quinolinone derivative OPC 21268" (EP 0382185 A2; AdisInsight: OPC 21268-October 6, 2006, last data update http://adisinsight.springer.com/drugs/800000284) The efficacy of gastric mucosal injury indications was tested in the preclinical stage, and the efficacy on heart failure and hypertension was studied in a phase 2 clinical trial, but no data have been developed on the compound since 2015 (Yamamura et al., Science 1991 252: 572; Serradeil-Le Gal et al., J Clin Invest 1993, 92 (1): 224).
When the brain stem is examined in postmortem human samples, selective localization of the V1a receptor that is not related to the oxytocin receptor can be detected in the nucleus prepositus, which plays a role in stability of sight Role (Freeman et al., Soc Neurosci 2017, 12 (2): 113-123). The basic skills required for human social behavior are cognitive and eye tracking of biological-related data (Klin et al., Nature 2009, 459: 257-63; Simion et al., PNAS 2008, 105 (2): 809-13) . The most active V1aR research pharmaceutical company Hoffmann-La Roche's indole derivative RO5028442 has entered the first phase of research (RG-7713; WO 2007/006688 A1), in which positive effects on sight pattern orientation can be detected in the human body (Umbricht et al., Neuropsychopharmacology 2017, 42 (9): 1914-1923; AdisInsight: RG 7713-November 5, 2015, final data update http://adisinsight.springer.com/drugs/800043668). Balovaptan with a triazolobenzodiazepine core is undergoing a Phase 2 clinical trial to treat autism (RG-7314, RO5285119; WO 2010/060836 A1; AdisInsight: RG 7314-2017 (September 10, the latest information update (http://adisinsight.springer.com/drugs/800035102).
Despite many V1aR antagonist compounds and clinical studies, unmet medical needs continue to require the development of V1aR antagonists, which are suitable for the treatment and / or prevention of various pathological symptoms of female sex organs and persistent symptoms of blood pressure control , Symptoms caused by inappropriate secretion of vasopressin, anxiety, depression, aggressive behavior, central nervous system disease (where one of the symptoms of the disease and / or multiple symptoms may be related to anxiety, depression or aggressive behavior or Comorbidities with these three diseases (autism group disorder, obsessive-compulsive disorder, many different types of Down syndrome and post-traumatic stress disorder), aggressive behavior disorder and / or irritability, hyperactivity disorder, Cognitive impairment or other neuropsychiatric disorders.

發明簡述
本發明之目標係合成具有新穎結構之V1a受體拮抗劑,該V1a受體拮抗劑之物理化學性質(例如動力學或熱力學溶解度、離子化、親脂性或滲透性)或藥學性質(例如代謝安定性或CYP-450酶抑制)能提供有利之生物可利用性、ADME(吸收、分佈、代謝及排泄)、膜滲透或血腦障壁穿透。
令人意外地,已製備新穎的通式(I)之5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯衍生物,其顯現V1a受體拮抗活性輪廓。
本發明關於通式(I)化合物:

其中
環A係環烷基或雜環基;
若環B存在,Y係-O-、-C(O)-、-CH2 -、-NH-或鍵結;或,若環B不存在,Y係-N(C1-4 烷基)2 、C(O)OC1-4 烷基、可選擇地經鹵素取代之C1-4 烷基、C1-4 烷氧基或鹵素;
環B係可選擇地經取代之雜芳基、芳基或雜環基;
或B-Y-A-結合代表3H -螺[2-苯並呋喃-1,4’-哌啶-1’-基];或

R1 係氫、鹵素、C1-4 烷基、C1-4 烷氧基、CF3 或CN;
R2 係氫或C1-4 烷基;
R3 係(CH2 )n R4 、C(O)R5 或可選擇地經R6 取代之C1-4 烷基;
或R2 與R3 結合代表-(CH2 )p -O-(CH2 )q -或-(CH2 )r -;
R4 係CN、疊氮基或Cy1
R5 係C1-4 烷基、C1-4 烷氧基、OH或NR7 R8
R6 係OR9 、NR10 R11 、側氧基、-O(CH2 )2 O-或一或多個鹵素;
R7 和R8 獨立地氫、C1-4 烷基或Cy2 或R7 和R8 與彼等連接之N一起形成雜環;
R9 係氫、可選擇地經NH2 或可選擇地經取代之芳基取代之C1-4 烷基、Si(CH3 )3 或C(O)R12
R10 和R11 獨立地氫、可選擇地經C1-4 烷氧基取代之C1-4 烷基、Cy3 或C(O)R13 ,或R10 和R11 與彼等連接之N一起形成可選擇地經取代之雜環;
R12 係C1-5 烷基或NR14 R15
R13 係C1-4 烷基、Cy1 或NR16 R17
R14 和R15 獨立地氫或可選擇地經取代之芳基,或R14 和R15 與彼等連接之N一起形成可選擇地經取代之雜環;
R16 和R17 係C1-4 烷基,或R16 和R17 與彼等連接之N一起形成可選擇地經取代之雜環;
Cy1 係可選擇地經取代之雜芳基;
Cy2 係可選擇地經取代之芳基或雜芳基;
Cy3 係可選擇地經取代之環烷基或雜環基;
X係C1-4 烷基、芳基或雜芳基;
Z係C1-4 烷基;
n係0或1;
p係1、2或3;
q係1、2或3;
r係4、5或6;
及/或彼等之鹽及/或彼等之幾何異構物及/或彼等之立體異構物及/或彼等之鏡像異構物及/或彼等之消旋物及/或彼等之非鏡像異構物及/或彼等之生物活性代謝物及/或彼等之前藥及/或彼等之溶劑合物及/或彼等之水合物及/或彼等之多形體。
本發明亦關於醫藥組成物,其含有該通式(I)化合物及/或彼等之鹽及/或彼等之幾何異構物及/或彼等之立體異構物及/或彼等之鏡像異構物及/或彼等之消旋物及/或彼等之非鏡像異構物及/或彼等之前藥及/或彼等之溶劑合物及/或彼等之水合物及/或彼等之多形體作為活性物質。
此外,本發明亦關於該通式(I)化合物及/或彼等之鹽及/或彼等之幾何異構物及/或彼等之立體異構物及/或彼等之鏡像異構物及/或彼等之消旋物及/或彼等之非鏡像異構物及/或彼等之前藥及/或彼等之溶劑合物及/或彼等之水合物及/或彼等之多形體的製備、該製備之過程的中間體及含有該等化合物之醫藥組成物的化學和醫藥製劑。
本發明亦關於一種治療罹患其中調節且較佳地拮抗該V1a受體可能具有治療益處之中樞及/或周圍疾病的包括人之哺乳動物之方法,其中施予該通式(I)化合物及/或彼等之鹽及/或彼等之幾何異構物及/或彼等之立體異構物及/或彼等之鏡像異構物及/或彼等之消旋物及/或彼等之非鏡像異構物及/或彼等之前藥及/或彼等之溶劑合物及/或彼等之水合物及/或彼等之多形體或彼等於組成物之治療有效量。
本發明亦關於該通式(I)化合物及/或彼等之鹽及/或彼等之幾何異構物及/或彼等之立體異構物及/或彼等之鏡像異構物及/或彼等之消旋物及/或彼等之非鏡像異構物及/或彼等之前藥及/或彼等之溶劑合物及/或彼等之水合物及/或彼等之多形體於製造藥物之用途,該藥物用於治療及/或預防與V1a受體功能有關之疾病或病症。

發明詳細說明
本發明關於V1a受體調節劑,特別是V1a受體拮抗劑。本發明之另一目標係提供選擇性V1a受體抑制劑化合物,因為選擇性比較不會引起不欲之副作用。本發明之另一方面係提供具有有利的物理化學性質之化合物,如該有利的物理化學性質被預期能導致該化合物的有益之生物可利用性、ADME(吸收、分佈、代謝及排泄)、膜滲透或血腦障壁穿透。
因此,本發明之通式(I)化合物係V1a受體拮抗劑,其係中樞及/或周圍作用性治療劑以治療及/或預防雌性性器官之多種病理症狀、血壓控制之持久性症狀、因血管升壓素之不適當分泌所引起之症狀、焦慮、抑鬱、攻擊行為、中樞神經系統疾病(其中該疾病之多種症狀之一及/或多種症狀可能與焦慮、抑鬱或攻擊行為有關或與該前述3種疾病顯示合併症(自閉症類群障礙、強迫症、多種不同類型之唐氏綜合症及創傷後應激障礙))、攻擊性行為障礙及/或易怒、行為過動症、認知障礙或其他神經精神疾病。
本發明關於通式(I)化合物:

其中
環A係環烷基或雜環基;
若環B存在,Y係-O-、-C(O)-、-CH2 -、-NH-或鍵結;或,若環B不存在,Y係-N(C1-4 烷基)2 、C(O)OC1-4 烷基、可選擇地經鹵素取代之C1-4 烷基、C1-4 烷氧基或鹵素;
環B係可選擇地經取代之雜芳基、芳基或雜環基;
或B-Y-A-結合代表3H -螺[2-苯並呋喃-1,4’-哌啶-1’-基];或

R1 係氫、鹵素、C1-4 烷基、C1-4 烷氧基、CF3 或CN;
R2 係氫或C1-4 烷基;
R3 係(CH2 )n R4 、C(O)R5 或可選擇地經R6 取代之C1-4 烷基;
或R2 與R3 結合代表-(CH2 )p -O-(CH2 )q -或-(CH2 )r -;
R4 係CN、疊氮基或Cy1
R5 係C1-4 烷基、C1-4 烷氧基、OH或NR7 R8
R6 係OR9 、NR10 R11 、側氧基、-O(CH2 )2 O-或一或多個鹵素;
R7 和R8 獨立地氫、C1-4 烷基或Cy2 或R7 和R8 與彼等連接之N一起形成雜環;
R9 係氫、可選擇地經NH2 或可選擇地經取代之芳基取代之C1-4 烷基、Si(CH3 )3 或C(O)R12
R10 和R11 獨立地氫、可選擇地經C1-4 烷氧基取代之C1-4 烷基、Cy3 或C(O)R13 ,或R10 和R11 與彼等連接之N一起形成可選擇地經取代之雜環;
R12 係C1-5 烷基或NR14 R15
R13 係C1-4 烷基、Cy1 或NR16 R17
R14 和R15 獨立地氫或可選擇地經取代之芳基,或R14 和R15 與彼等連接之N一起形成可選擇地經取代之雜環;
R16 和R17 係C1-4 烷基,或R16 和R17 與彼等連接之N一起形成可選擇地經取代之雜環;
Cy1 係可選擇地經取代之雜芳基;
Cy2 係可選擇地經取代之芳基或雜芳基;
Cy3 係可選擇地經取代之環烷基或雜環基;
X係C1-4 烷基、芳基或雜芳基;
Z係C1-4 烷基;
n係0或1;
p係1、2或3;
q係1、2或3;
r係4、5或6;
及/或彼等之鹽及/或彼等之幾何異構物及/或彼等之立體異構物及/或彼等之鏡像異構物及/或彼等之消旋物及/或彼等之非鏡像異構物及/或彼等之生物活性代謝物及/或彼等之前藥及/或彼等之溶劑合物及/或彼等之水合物及/或彼等之多形體。
本文所使用之一般用語(無論該用語是否被討論)的定義係獨立地或以與其他基團之組合的方式說明如下。
“環烷基”單獨或與其他基團組合係指3至8員(較佳地3至6員)之飽和或未飽和(較佳地飽和)的碳環基。實例包括環丙基、環丁基、環戊基或環己基。於環A和Cy3 中,該“環烷基”較佳地係指4至6員飽和碳環基。實例包括環丁基、環戊基或環己基。較佳的是環丁基或環己基。特佳的是環己基。“經取代之環烷基”較佳地係指具有孿鹵素取代之環烷基。
“芳基”單獨或與其他基團組合係指6至14員(較佳地6至10員)之芳香族碳環基,其包含至少1個芳香族環或含有至少1個芳香族環之稠合環系統。實例包括但不限於苯基、苄基、萘基、聯苯基、蒽基、薁基或二氫茚基。特別較佳的是苯基。
“雜環基”單獨或與其他基團組合係指3至8員(較佳地4至7員)之飽和或未飽和(較佳地飽和)的含有1、2、3或4個選自O、S或N之雜原子的單環、雙環、稠合及/或橋連環。較佳之4至7員單環雜環含有1個N、2個N、1個O、1個N和1個O、或1個N和1個S。實例包括但不限於環氧乙烷、氧雜環丁烷、四氫呋喃、四氫哌喃、四氫吖唉(azetidine)、吡咯啶、哌啶、哌嗪、嗎啉、1,3-噁唑烷、1,3-噻唑烷、硫代嗎啉-1,1-二氧化物及類似者。“經取代之雜環基”較佳地係指經C1-4 烷基取代之雜環。
當環A係雜環基時,該雜環基較佳地係指4至7員之含有1個N或2個N的飽和雜環基,其中環A係經環氮原子連接Y或該5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯(benzazepine)核之三唑環。實例包括但不限於四氫吖唉基、1,3-二(四氫吖唉基)、吡咯啶基、吡唑啶基、咪唑啶基、哌啶基、哌嗪基、氮雜環庚烷基、1,3-或1,4-二氮雜環庚烷基。較佳的是四氫吖唉基、哌啶基或哌嗪基。
對於Cy3 ,該雜環較佳地係指4至7員之含有1個O的飽和雜環基,更佳地係指氧雜環丁烷或四氫哌喃。
當環B係雜環基時,該雜環基較佳地係指4至7員之含有1個N或1個N和1個O的飽和雜環基。實例包括但不限於四氫吖唉基、吡咯啶基、哌啶基、哌嗪基或嗎啉基。
當R7 和R8 、R10 和R11 、R14 和R15 或R16 和R17 “與彼等連接之N一起形成可選擇地經取代之雜環”時,該雜環較佳地係指4至7員之含有1個N、2個N、1個N和1個O、或1個N和1個S的飽和環。實例包括但不限於嗎啉、4-甲基-哌嗪基、吡咯啶基、哌啶基、哌嗪基、1,3-噁唑烷、1,3-噻唑烷或硫代嗎啉-1,1-二氧化物。
“雜芳基”單獨或與其他基團組合係指含有單一5至6員環之環狀芳香族基,該5至6員環含有1、2、3或4個雜原子,其中至少1個雜環係芳香族基。“6員單雜芳基”係指單環芳香族基,其係含有1、2或3個選自O、S或N之雜原子的單一6員環。實例包括但不限於吡啶基、嘧啶基、吡嗪基、噠嗪基、噻嗪基、噁嗪基及類似者。較佳之單一6員單雜芳基含有1或2個N。較佳之6員環係吡啶基,更佳地係吡啶-2-基和吡啶-3-基。特佳的是吡啶-2-基。“5員單雜芳基”表示含有1、2、3或4個選自O、S或N之雜原子的單一5員環之單環芳香族基。較佳之單一5員單雜芳基含有2個N和1個O、2個N和1個S、4個N、3個N、2個N、1個N或1個S或1個N和1個O。實例包括但不限於噻吩基、呋喃基、吡咯基、四唑基、三唑基、咪唑基、噻唑基、異噁唑基、異噻唑基、噁二唑基、噻二唑基、1H -吡唑基及類似者。較佳的是異噁唑-3-基、四唑基及1,2,4-噁二唑-5-基。“經取代之雜芳基”較佳地係指經C1-4 烷基或鹵素取代之雜芳基。
對於Cy1 ,該可選擇地經取代之雜芳基較佳地係指經C1-4 烷基(更佳地甲基)取代之含有2個N和1個O或4個N的單一5員單雜芳基。較佳的是四唑基或3-甲基-1,2,4-噁二唑-5-基。
當環B係可選擇地經取代之雜芳基時,該雜芳基係指可選擇地經鹵素(較佳地氯)或C1-4 烷基(較佳地甲基)取代之含有1個N、2個N或1個N和1個O的5或6員單雜芳基。實例包括但不限於吡啶-2-基、3-氯-吡啶-2-基、3-甲基-吡啶-2-基、吡啶-3-基、嘧啶-2-基或5-甲基-異噁唑-3-基。
“鍵結”係指單鍵,其中2個原子共有1對電子。
“C1-4 烷基”或“C1-5 烷基”,單獨或與其他基團組合,係指直鏈或支鏈、單一或多個支鏈之烴基且係由1至4個或1至5個碳原子組成。實例包括但不限於甲基、乙基、丙基、異丙基、正丁基、2-丁基(二級丁基)、三級丁基、正戊基、三級戊基、新戊基、異戊基、2-戊基(二級戊基)或3-戊基。較佳之烷基係具有1至3個碳原子者。較佳的是甲基、乙基、異丙基、三級丁基及三級戊基。特別較佳的是甲基。
“C1-4 烷氧基”,單獨或與其他基團組合,係指-O-C1-4 烷基,其中該C1-4 烷基係如上述定義者。實例包括但不限於甲氧基、乙氧基、丙氧基或三級丁氧基。較佳之烷氧基係甲氧基、丙氧基或三級丁氧基。特別地 較佳的是甲氧基和三級丁氧基。
“Boc”,單獨或與其他基團組合,係指三級丁氧羰基。
“鹵素”,單獨或與其他基團組合,係指氟、氯、溴或碘,較佳地氟、氯或溴。對於R1 和R6 ,較佳的是氟、氯或溴。對於R1 ,更佳的是氯或溴,甚佳的是氯。對於R6 ,更佳的是氟。
於相關基團之任何原子的“可選擇地經取代”係指經一或多個C1-4 烷基或鹵素取代。此處,“一或多個”表示1個至最高可能之數目的取代,即自取代1個氫至取代所有的氫。因此,較佳的是給定之原子的1、2或3個取代。甚佳的是1或2個取代或1個取代。對可選擇地經取代之環烷基、芳基、雜環基或雜芳基,特別較佳的是1個取代。
“可選擇地經鹵素取代之C1-4 烷基”較佳地係指於C1-4 烷基之任何原子上具有1、2或3個鹵素取代的C1-4 烷基,更佳地係指具有3個鹵素取代的甲基。特別較佳的是CF3
於“經R6 取代之C1-4 烷基”中,該C1-4 烷基係如上述定義者。於“經R6 取代之C1-4 烷基”中,該經取代之C1-4 烷基較佳地係由1至4個碳原子組成之支鏈、單一或多個支鏈之烴基。於“經R6 取代之C1-4 烷基”中,該“經取代”係指於相關基團之任何原子上經一或多個R6 取代。該“一或多個”表示1個至最高可能之數目的取代,即自取代1個氫至取代所有的氫。當R6 係OR9 或NR10 R11 時,該“經R6 取代之C1-4 烷基”係例如但不限於-CH2 R6 或-CH(R6 )CH3 ,較佳地-CH2 R6 。當R6 係側氧基或-O(CH2 )2 O-時,該“經R6 取代之C1-4 烷基”係例如但不限於-C(R6 )CH3 。當R6 係一或多個鹵素時,該“經R6 取代之C1-4 烷基”、“一或多個”及“鹵素”係如上述定義者。此處,該“經R6 取代之C1-4 烷基”係例如但不限於-CH2 鹵素、-CH(鹵素)CH3 或-C(鹵素)2 CH3 ,較佳地-CH2 鹵素。
“鹽”係指醫藥上可接受及/或醫藥上不可接受之鹽。該醫藥上可接受之鹽係指慣用之酸加成鹽和鹼加成鹽,該等鹽保留該通式(I)化合物之生物功效和性質且可經由適當之非毒性有機或無機酸或有機或無機鹼生成。酸加成鹽之實例包括衍生自無機酸(諸如但不限於氫氯酸、氫溴酸、氫碘酸、硫酸、胺磺酸、磷酸、硝酸及過氯酸)之鹽和衍生自多種有機酸(諸如但不限於乙酸、丙酸、苯甲酸、羥乙酸、苯乙酸、水楊酸、丙二酸、順丁烯二酸、油酸、雙羥萘酸、棕櫚酸、苯磺酸、甲苯磺酸、甲磺酸、草酸、酒石酸、琥珀酸、檸檬酸、蘋果酸、乳酸、麩胺酸、反丁烯二酸及類似者)之鹽。鹼加成鹽之實例係衍生自氫氧化銨、氫氧化鉀、氫氧化鈉及氫氧化四級銨(諸如氫氧化四甲基銨)之鹽。該等鹽通常比用於製備彼等之化合物本身具有較為有利的溶解度性質並因此更適合用於製備例如液體或乳化調製劑。該醫藥上不可接受之鹽可較佳地用於純化和分離該通式(I)化合物並因此亦落於本發明之範疇。
“前藥”係指本發明之通式(I)化合物的衍生物,其本身不具有治療功效但含有經活體內化學或代謝降解(生物轉化)後成為“生物活性代謝物”之基團,該生物活性代謝物承擔該治療功效。與本發明之該通式(I)化合物連接的該分解基團(特別是適合前藥者)係為此技術領域所習知且亦可適用於本發明之化合物(Rautio et al.,Nat Rev Drug Discov 2008, 7:255-270)。
該通式(I)化合物可存有多種幾何異構型式。此外,某些通式(I)化合物可含有一或多個不對稱中心並因此可以立體異構物和非鏡像異構物之形式存在。所有該等化合物,諸如順式異構物、反式異構物、非鏡像異構混合物、消旋物、鏡像異構物之非消旋性混合物、實質性純和純鏡像異構物亦落於本發明之範疇。該實質性純鏡像異構物佔對應之相反鏡像異構物的達5重量%,較佳地2重量%且最佳地1重量%。
藉由習知之方法(例如藉由使用光學活性酸或鹼以生成非鏡像異構性鹽或藉由生成共價非鏡像異構物)解析消旋性混合物,可製備光學異構物。適當之酸包括例如酒石酸、二乙醯基酒石酸、二苯甲醯基酒石酸、二甲苯甲醯基酒石酸及樟腦磺酸。藉由此技術領域習知之方法(諸如層析或分次結晶),基於個別非鏡像異構物之物理及/或化學差異,可將非鏡像異構混合物分離成個別之非鏡像異構物。隨後,自該等經分離之非鏡像異構性鹽釋出該光學活性鹼或酸。各種不同之分離光學異構物的方法包括手性層析(例如手性HPLC管柱),其可選擇地經使用以衍生達到最佳分離鏡像異構物之目標。適當之手性HPLC管柱係Diacel管柱,諸如CHIRALPAK或CHIRALCEL管柱,彼等可如所欲地被常規選用。如適用地,亦可使用衍生化實施之酶催化分離。藉由無消旋反應條件之手性合成且使用光學活性起始物,亦可分離光學活性之通式(I)化合物。
藉由文獻(Freedman et al.,Chirality 2003,15 (9):743-58;Stephens et al.,Chirality 2008,20 :643-663)描述之振動圓二色性光譜(VCD)方法及/或藉由自手性化合物產生之化合物的非鏡像異構物對之1 H NMR光譜分析(Seco et al.,J Org Chem 1999,64: 4669-4675;Seco et al.,Tetrahedron Asymmetry 2001,12: 2915-2925;Latypov et al.,J. Am. Chem. Soc . 1998,120 , 4741-4751),測定手性化合物之絕對構型。
該通式(I)化合物可存有多種多形體型式。如此技術領域所習知者,多形體係化合物結晶成超過一種晶型(即多形體型式)之能力。藉由相同化學式或組成可定義特定化合物之多形體型式,該等多形體型式之化學結構不同,如同2個不同化合物之結晶結構。
該通式(I)化合物和彼等之鹽亦可以溶劑合物或水合物之形式存在,該等溶劑合物和水合物亦落於本發明之範疇。該“溶劑合物”係指溶劑與溶質之非共價組合。該“水合物”係指水與溶質之非共價組合。
本發明另關於醫藥組成物,其含有該通式(I)化合物及/或彼等之鹽及/或彼等之幾何異構物及/或彼等之立體異構物及/或彼等之鏡像異構物及/或彼等之消旋物及/或彼等之非鏡像異構物及/或彼等之前藥及/或彼等之溶劑合物及/或彼等之水合物及/或彼等之多形體。
本發明亦關於醫藥組成物之化學和醫藥製劑,該醫藥組成物含有該通式(I)化合物及/或彼等之鹽及/或彼等之幾何異構物及/或彼等之立體異構物及/或彼等之鏡像異構物及/或彼等之消旋物及/或彼等之非鏡像異構物及/或彼等之前藥及/或彼等之溶劑合物及/或彼等之水合物及/或彼等之多形體。
本發明之醫藥組成物可經調製成多種不同之醫藥調製劑,彼等係諸如但不限於固體口服劑型,諸如片劑(例如經頰、舌下、發泡、可咀嚼及口服可分散)、膠囊、藥丸、丸劑、口服可分散膜、顆粒及粉末;液體調製劑,諸如溶液、乳劑、懸浮液、糖漿、酏劑及滴劑;非經腸劑型,諸如靜脈注射劑、肌肉注射劑及皮下注射劑;其他醫藥型式,諸如眼滴劑、半固體眼用製劑、半固體皮膚製劑(諸如藥膏、乳霜及糊劑)、穿皮治療系統、栓劑、直腸膠囊、直腸溶液、乳劑及懸浮液等。
本發明之一實施態樣關於供小兒科使用之醫藥組成物,其係諸如但不限於溶液、糖漿、酏劑、懸浮液、供製備懸浮液之粉末、可分散或發泡之片劑、可咀嚼片劑、可口腔分散片劑、片劑或塗覆片劑、口腔發泡粉末或顆粒、或膠囊。
藉由本身為習知之方法,諸如慣用之混合、溶解、乳化、懸浮、微膠囊化、凍乾、擠出與搓圓、層壓、薄膜塗層、顆粒化、封裝、糖衣化或壓縮,可製備本發明之醫藥組成物。
藉由使用一或多種生理上可接受的賦形劑(其包括結合劑)之慣用方式,可調製本發明之醫藥組成物,該(等)生理上可接受的賦形劑能促進活性物質併入醫藥上可接受之醫藥劑型。適當之調製劑取決於所選擇之投藥模式。可使用此技術領域習知之任何技術和賦形劑。
適用於製備之賦形劑可選自下述範圍,其係諸如但不限於片劑和膠囊之填料、片劑和膠囊之結合劑、改質藥物釋出劑、崩解劑、助流劑、潤滑劑、甜味劑、掩味劑、調味劑、塗料、表面活性劑、安定劑、防腐劑或抗氧化劑、緩衝劑、複合劑、潤濕劑或乳化劑、調整滲透壓之鹽、凍乾賦形劑、微膠囊劑、藥膏料、滲透促進劑、增溶劑、溶劑、栓劑料及懸浮劑。適當之醫藥賦形劑可為例如澱粉、微晶纖維素、滑石、葡萄糖、乳糖、明膠、二氧化矽、滑石、硬脂酸鎂、硬脂酸鈉、單硬脂酸甘油酯、纖維素衍生物、氯化鈉、甘油、丙二醇、水、乙醇及類似者。
本發明之另一實施態樣關於使用可改善活性物質之溶解度、溶解性、滲透性、吸收或生物可利用性的特定結合劑,其係諸如但不限於親水性聚合物、熱熔融擠壓賦形劑、表面活性劑、緩衝劑、複合劑、乳化劑、凍乾賦形劑、崩解劑、微膠囊劑、滲透促進劑、增溶劑、共溶劑及懸浮劑。
上述賦形劑和多種不同之製備方法僅是代表性實例。亦可使用此技術領域習知之其他材料和製程技術。
“與V1a受體功能有關之疾病或病症”或“與中樞及/或周圍調節(較佳地V1a受體拮抗作用)有關之疾病或病症”係指選自雌性性器官之多種病理症狀、血壓控制之持久性症狀、因血管升壓素之不適當分泌所引起之症狀、焦慮、抑鬱、攻擊行為、中樞神經系統疾病(其中該疾病之多種症狀之一及/或多種症狀可能與焦慮、抑鬱或攻擊行為有關或與該前述3種疾病顯示合併症(自閉症類群障礙、強迫症、多種不同類型之唐氏綜合症及創傷後應激障礙))、攻擊性行為障礙及/或易怒、行為過動症、認知障礙或其他神經精神疾病之疾病或病症。
該雌性性器官之多種病理症狀包括但不限於痛經(原始性及/或繼發性)或性功能障礙。
該血壓控制之持久性症狀包括但不限於高血壓及/或慢性心臟衰竭。
該因血管升壓素之不適當分泌所引起之症狀包括但不限於尿崩症、腎衰竭、腎病症候群或肝硬化。
該中樞神經系統疾病(其中該疾病之多種症狀之一及/或多種症狀可能與焦慮、抑鬱或攻擊行為有關或與該前述3種疾病顯示合併症)包括但不限於自閉症類群障礙(良好功能自閉症、亞斯伯格症候群、待分類的廣泛性發展障礙(PDD-NOS)、自閉症譜系障礙(ASD)及彼之多種不同症候型:X染色體易裂症、普瑞德威利症候群、蕾特氏症及結節性硬化症)、強迫症(OCD)、多種不同類型之唐氏綜合症及創傷後應激障礙(PTSD)。
該攻擊性行為障礙及/或易怒包括但不限於ASD、亨丁頓舞蹈症或不同類型之精神分裂症。
該行為過動症包括但不限於注意力不足過動症。
該認知障礙包括但不限於癡呆、輕度認知障礙、與精神分裂症有關之認知障礙或阿茲海默症。
該其他神經精神疾病包括但不限於精神分裂症和相關疾病。
於一實施態樣中,該與V1a受體功能有關之疾病或病症或與中樞及/或周圍調節(較佳地V1a受體拮抗作用)有關之疾病或病症係指自閉症類群障礙。
本發明關於一種治療及/或預防與V1a受體功能有關之疾病或病症之方法,其包含對需要該治療及/或預防之個體(較佳地哺乳動物且更佳地人)投予治療有效量的單獨通式(I)化合物及/或彼等之鹽及/或彼等之幾何異構物及/或彼等之立體異構物及/或彼等之鏡像異構物及/或彼等之消旋物及/或彼等之非鏡像異構物及/或彼等之前藥及/或彼等之溶劑合物及/或彼等之水合物及/或彼等之多形體或與至少一種醫藥上可接受之賦形劑以呈醫藥調製劑之型式。
本發明關於一種治療罹患疾病或病症之個體(較佳地哺乳動物且更佳地人)之方法,該疾病或病症選自雌性性器官之多種病理症狀、血壓控制之持久性症狀、因血管升壓素之不適當分泌所引起之症狀、焦慮、抑鬱、攻擊行為、中樞神經系統疾病(其中該疾病之多種症狀之一及/或多種症狀可能與焦慮、抑鬱或攻擊行為有關或與該前述3種疾病顯示合併症(自閉症類群障礙、強迫症、多種不同類型之唐氏綜合症及創傷後應激障礙))、攻擊性行為障礙及/或易怒、行為過動症、認知障礙或其他神經精神疾病、或該等疾病之組合。該治療方法包含對需要該治療之個體(較佳地哺乳動物且更佳地人)投予治療有效量的通式(I)化合物及/或彼等之鹽及/或彼等之幾何異構物及/或彼等之立體異構物及/或彼等之鏡像異構物及/或彼等之消旋物及/或彼等之非鏡像異構物及/或彼等之前藥及/或彼等之溶劑合物及/或彼等之水合物及/或彼等之多形體。該治療方法可包括對需要該治療之個體(較佳地哺乳動物且更佳地人)投予治療有效量的醫藥組成物,該醫藥組成物包含通式(I)化合物及/或彼等之鹽及/或彼等之幾何異構物及/或彼等之立體異構物及/或彼等之鏡像異構物及/或彼等之消旋物及/或彼等之非鏡像異構物及/或彼等之前藥及/或彼等之溶劑合物及/或彼等之水合物及/或彼等之多形體。
本發明關於該通式(I)化合物及/或彼等之鹽及/或彼等之幾何異構物及/或彼等之立體異構物及/或彼等之鏡像異構物及/或彼等之消旋物及/或彼等之非鏡像異構物及/或彼等之前藥及/或彼等之溶劑合物及/或彼等之水合物及/或彼等之多形體於製造藥物之用途,該藥物用於治療及/或預防與V1a受體功能有關之疾病或病症。
該“治療”係指已罹患或經診斷罹患疾病之病患或個體的特定病理症狀之緩和、病症之一或多種症狀之除去或減輕、疾病狀態進展之減緩或消除及病理症狀復發之預防或遲延。若藥物已給予業已發生疾病或病症之病患,一般藉由相同或相似之方法給予該藥物以實施該“預防”(或疾病發生之預防或遲延)。
“治療有效量”係指導致疾病或病理症狀或副作用之治療、治癒、預防或改善的活性物質之量,且與未接受該治療有效量之對應個體相比較,能減緩該疾病或病理症狀之進展。該治療有效量亦包括能增強正常生理功能之有效量。對用於治療,可投予治療有效量之該通式(I)化合物及/或彼等之幾何異構物及/或彼等之立體異構物及/或彼等之鏡像異構物及/或彼等之消旋物及/或彼等之非鏡像異構物及/或彼等之前藥及/或彼等之溶劑合物及/或彼等之水合物及/或彼等之多形體以及彼等之任何醫藥上可接受之鹽作為原料化學品。此外,該活性物質可以醫藥調製劑之方式提供。該通式(I)化合物及/或彼等之鹽及/或彼等之幾何異構物及/或彼等之立體異構物及/或彼等之鏡像異構物及/或彼等之消旋物及/或彼等之非鏡像異構物及/或彼等之前藥及/或彼等之溶劑合物及/或彼等之水合物及/或彼等之多形體的精確之治療有效量取決於許多因素,該等因素包括但不限於被治療之個體(病患)之年齡和體重、需要治療之疾病的確切類型和嚴重程度、醫藥品之本性和投藥途徑。
“哺乳動物”係指“哺乳類”之任何成員,其包括但不限於人。
本發明亦關於適合用於治療與中樞及/或周圍調節(較佳地V1a受體拮抗作用)有關之疾病或病症之醫藥組成物,其包含該通式(I)化合物及/或彼等之鹽及/或彼等之幾何異構物及/或彼等之立體異構物及/或彼等之鏡像異構物及/或彼等之消旋物及/或彼等之非鏡像異構物及/或彼等之前藥及/或彼等之溶劑合物及/或彼等之水合物及/或彼等之多形體。
本發明之化合物亦可與一或多種本發明之化合物或一或多種其他活性物質(例如精神阻斷劑、精神興奮劑、抗高血壓藥、解痙藥、抗癲癇藥或其他藥劑)組合以用於罹患其中V1a受體之中樞及/或周圍調節且較佳地拮抗可能具有治療益處之中樞及/或周圍疾病的哺乳動物,其包括但不限於人。
精神阻斷劑包括但不限於抗精神病藥、抗焦慮劑及鎮靜催眠藥或麻醉劑。
抗精神病藥包括但不限於典型和非典型之抗精神病藥,諸如具有脂肪族側鏈之吩噻嗪(氯丙嗪、丙嗪、甲氧異丁嗪、乙醯丙嗪、三氟拉嗪、沙美麥嗪(ciamemazine)、氯丙沙嗪、丙泰提(protipendyl))、哌嗪衍生之吩噻嗪(地西拉嗪、氟非那嗪、甲哌丙嗪、羥哌氯丙嗪、丙氯比拉嗪、硫普哌嗪、三氟吡啦嗪、乙醯非那嗪、硫丙拉嗪、布他哌嗪、甲哌丙嗪)、哌啶衍生之吩噻嗪(哌氰嗪、甲硫哌啶、美索噠嗪、哌泊噻嗪)、硫雜蒽(氯丙硫葸、環戊噻吨(clopenthixole)、氟哌噻吨、甲哌硫丙硫蒽、珠氯噻吨)、丁酸酚酮衍生物(氟哌啶醇、三氟哌啶醇、美哌隆、莫哌隆、匹泮哌隆、溴哌啶醇、苯哌唑酮、氟哌利多、硫咪哌酮、氟阿尼酮)、二苯基丁基哌啶衍生物(氟斯必靈、五氟利多、呱咪清)、二吖呯或氧吖呯或硫吖呯衍生物(氯氮平、奧氮平、氯噻平、喹硫平、洛沙平、阿塞那平)、吲哚衍生物(舍吲哚、齊拉西酮、魯拉西酮、嗎啉吲酮、噁哌亭(oxipertine))、苯甲醯胺衍生物(舒必利、舒妥必利、硫必利、瑞莫必利、胺磺必利、維拉必利、奈莫必利、維硫拉必利)或其他藥劑(利培酮、阿立哌唑、卡利拉嗪、依匹哌唑、美托拉麥、莫沙帕明、伊潘立酮、帕潘立酮、阿莫沙平、安哌匹隆、哌羅匹隆、卡巴咪嗪、氯卡帕明、丁苯那嗪、鋰)。
抗焦慮劑包括但不限於苯並二吖呯(二氮平、氯二氮平、美達氮平、去甲羥氮平、氯氮平酸鉀、樂耐平、阿地唑崙、溴氮平、甲酮氮平、凱他唑侖、環丙氮平、阿普唑他、三氟氮平、丙炔氮平、卡氮平、原氮平、氟二氮平、氯氟卓乙酯、依替唑侖、氯噻氮平、奧沙唑侖、托菲唑侖(tophizopam))、二苯基甲烷衍生物(羥嗪、丁硫二苯胺)、胺甲酸酯類(胺甲丙二酯、依咪卡美(emilcamate)、甲戊胺酯)、二苯並二環辛二烯衍生物(苯醌)、氮雜螺癸二酮類(丁螺環酮)、其他藥劑(美芬噁酮、吉多卡爾、氯乙苯嗪、菲瑪唑(fabomotizole)、三甲泰辛(trimethosine))、藉由增加GABAA 介導之抑制作用而作用之衍生物或作用於血清素受體之化合物、及其他GABA神經原性劑(諸如GABAA α5 NAMs (例如巴米沙尼)和GABAA α5 PAMs (例如RG7816))。
鎮靜催眠藥或麻醉劑包括但不限於巴比妥類藥物(戊巴比妥、異戊巴比妥、正丁巴比妥、巴比妥、烯丙異丙巴比妥、司可巴比妥、他布比妥、乙烯比妥、戊烯比妥、環巴比妥、庚巴比妥、西可巴比妥、美索比妥、環己烯巴比妥、硫噴妥鈉、依沙比妥、阿洛巴比妥、普西巴比妥(proxibarbital))、醛類(水合氯醛、氯醛已醇、乙醯基甘胺醯胺水合氯醛、二氯醛安替比林、三聚乙醛)、苯並二吖呯類(氟西泮、硝西泮、氟硝西泮、艾司唑侖、三唑侖、氯甲西泮、替馬西泮、咪達唑侖、溴替唑侖、夸西泮、氯普唑崙、度氟西泮、西諾西泮)、哌啶二酮衍生物(苯乙呱啶酮、甲乙哌酮、二乙吡啶二酮)、環吡咯酮苯並二吖呯衍生物(佐匹克隆、唑吡坦、札來普隆、艾司佐匹克隆)、退黑激素受體激動劑(退黑激素、瑞美替胺)或其他催眠藥和鎮靜劑(甲喹酮、氯美噻唑、溴米索伐、二乙溴乙醯脲、莨菪胺、丙醯馬嗪、三氯乙磷酸、乙氯戊炔醇、纈草根、己丙胺酯、溴化物、丙烯異丙乙酸尿、戊諾醯胺、甲基戊炔醇、烟胺哌嗪、右美托嘧啶)。
精神興奮劑包括但不限於精神刺激劑或抗抑鬱劑。
精神刺激劑包括但不限於中樞作用性擬交感神經劑(安非他命、右旋安非他命、甲基苯丙胺、哌甲酯、苯異妥英、芬樟胺、莫達非尼、乙苯噁唑酮、阿托莫西汀、芬奈替尼(phenetilline)、右哌甲酯、甲磺酸離胺酸安非他命)、益智劑或其他精神刺激劑(咖啡因、丙戊托菲林、甲氯芬酯、吡硫醇、必舒坦、二甲胺乙醇、非哌西特、胞磷膽鹼、奥拉西坦、吡舒達諾、利諾吡啶、尼唑苯酮、阿尼西坦、乙醯基肉鹼、艾地苯醌、丙苯乙吡咯、哌苯甲醇、普拉西坦、阿屈非尼、長春西汀、他克林、多奈哌齊、卡巴拉汀、加蘭他敏、艾匹達林(ipidachrine)、美金剛、美比卡(mebicar)、非尼布特)。
抗抑鬱劑包括但不限於非選擇性單胺再吸收抑制劑(脫甲丙咪嗪、丙咪嗪、丙咪嗪氧化物、氯米帕明、奧匹哌醇、三甲丙咪嗪、洛非帕明、地本西平、阿米替林、去甲阿米替林、普羅替林、杜使平、伊普吲哚、美利曲辛、丁替林、度硫平、阿莫沙平、二甲他林、安咪奈丁、嗎丙啶、奎紐帕明)、血清素調節劑和刺激劑(維拉唑酮、沃替西汀)、選擇性血清素再吸收抑制劑(齊美定、氟西汀、帕羅西汀、舍曲林、阿拉丙酯、氟伏沙明、依托哌酮、西酞普蘭、立普能)、非選擇性肼衍生之單胺氧化酶抑制劑(異唑肼、尼亞拉胺、苯乙肼、反苯環丙胺、異菸鹼異丙醯肼、異菸鹼環苷(iprocloside))、非肼之單胺氧化酶抑制劑(嗎氯苯甲醯胺、托洛沙酮)或其他藥劑(羥色胺酸、色胺酸、米安色林、胺苯甲異喹、曲唑酮、奈法唑酮、苯噠嗎啉、二苯美崙、氯苄吡醇、奥沙氟生、米氮平、美地沙明、噻奈普汀、匹伐加賓、文拉法辛、米那普侖、瑞波西汀、噠嗪醇(pyrazidol)、度洛西汀、阿戈美拉汀、去甲文拉法辛、安非他酮、吉哌隆、草金絲桃萃取物)。
抗高血壓藥包括但不限於β受體阻斷劑、噻嗪類利尿劑、血管收縮素轉化酶抑制劑、鈣拮抗劑、血管收縮素受體拮抗劑(氯沙坦)、蘿芙木生物鹼(桂皮利血胺、利血平、去甲氧利血平、美索舍平、比他舍平)、甲基多巴、咪唑啉受體激動劑(可樂寧、氯苯乙胍、托洛尼定、莫索尼定、利美尼定)、神經節阻斷抗腎上腺素劑(鋶衍生性咪噻芬、二級和三級胺四甲基雙環庚胺)、周圍作用性抗腎上腺素劑、α腎上腺素受體阻斷劑(哌唑嗪、吲哚拉明、曲馬唑嗪、多沙唑嗪、哌胺甲尿啶)、胍衍生物(倍他尼定、胍乙啶、胍生、異喹胍、胍氯酚、胍那佐定、胍羥苯)、小動脈平滑肌作用劑、噻嗪衍生性氯甲苯噻嗪、肼基酞嗪衍生物(肼苯噠嗪、肼屈嗪、恩屈嗪、卡屈嗪)、嘧啶衍生性米諾地爾、硝基鐵氰化物衍生性五氰基亞硝醯基鐵酸、胍衍生性吡那地爾、非蘿芙木生物鹼藜蘆、酪胺酸羥化酶抑制性甲基酪胺酸、MAO抑制劑丙炔甲基苄胺、血清素拮抗劑酮色林或其他抗高血壓藥(波生坦、安倍生坦、西他生坦、馬西替坦、利奧西呱)及該等藥劑與利尿劑之組合。
解痙藥或抗痙劑包括但不限於周圍肌肉鬆弛劑、箭毒生物鹼、膽鹼衍生物、其他四級銨肌肉鬆弛劑(潘侃朗寧、加拉碘銨、維克羅因、阿屈庫銨、六氟脲、哌庫溴銨、多西環素氯化物、法扎溴銨、羅庫溴銨、米庫溴銨、順式阿屈庫銨、肉毒桿菌毒素)、中樞神經系統肌肉鬆弛劑、胺甲酸酯(苯丙胺酯、異丙基甲西雙脲、美索巴莫、胺甲羥苯乙酯、非巴胺酯)、噁唑衍生物、噻嗪衍生物及三嗪衍生物(氯美紮酮、氯苯噁唑酮)、與抗組織胺藥有關之醚(奧菲那特林、癒瘡木酚)和其他組織胺能藥物(諸如組織胺H3 受體拮抗劑/反激動劑,例如沙芬、硫丙咪胺、哌托生特、丙酸倍氯松、ABT-239、可內新、A-349,821、倍他司汀)、其他中樞作用劑(氯苯胺丁酸、阿巴氯芬、替扎尼定、二苯哌丙醇、托哌酮、硫秋水仙苷、甲酚甘油醚、四氫西泮、胺苯庚烯、苯吡胺醇)、直接作用肌肉鬆弛劑丹曲林和彼之衍生物、藉由增加GABAA 媒介之抑制作用或減少Na+ 傳輸而作用之化合物(二苯妥因、卡巴氮平、拉莫三嗪、VPA)、γ-胺基丁酸衍生物(胺己烯酸、加巴噴丁)、其他GABA作用劑(諸如GABAB PAMs,例如ADX71441)、含有三級胺基之酯(羥苄利明、卡米羅芬、美貝維林、曲美布汀、羅西維林、雙環維林、雙己維林、雙苯美林、二苯哌酯)、四級銨化合物(苯咯銨、葡糖吡咯鎓、奧芬溴銨、噴噻銨、丙胺西林、奧替溴銨、乙胺太林、曲地銨、異丙胺、己環銨、泊利定、甲哌佐酯、苯醇哌酯、哌苯偶酯、二苯甲哌、艾美碘銨(emetonium iodide)、替莫碘銨、吡芬溴銨、噻哌溴銨及甲苯哌醯胺)、含有三級胺之醯胺(阿斯特拉1397、尼非他胺、苯醯胺桂胺)、罌粟鹼和彼之衍生物(屈他維林、莫沙維林、乙基罌粟鹼)、血清素受體作用劑(阿洛司瓊、替加色羅、西蘭司瓊、普卡比利)、官能性胃腸疾病之其他藥劑(二苯丙哌啶、地索普明、氯苯沙明、匹維銨、非諾維林、依丹帕明、普羅沙唑、阿爾維林、三乙丁酮、異美汀、卡羅維林、間苯三酚、聚矽氧、三甲基二苯基丙胺)、琥珀醯亞胺衍生物(乙琥胺、苯琥胺、甲琥胺)或顛茄生物鹼類和彼等之衍生物(阿托品、天仙子胺、丁基東莨菪鹼、甲基阿托品、甲基東莨菪鹼、芬托銨、溴氰菊酯)。
抗癲癇藥包括但不限於巴比妥類藥物和彼等之衍生物(甲基苯巴比妥、苯巴比妥、乙苯嘧啶二酮、巴比沙隆、美沙比妥)、乙內醯脲衍生物(乙苯妥英、二苯妥因、胺基(二苯妥因)戊酸、美芬妥因、磷苯妥英)、噁唑烷衍生物(甲乙雙酮帕臘二酮、三甲環二酮、乙唑二酮(ethadion))、琥珀醯亞胺衍生物(乙琥胺、苯琥胺、甲琥胺)、苯並二吖呯衍生物氯硝西泮、羧醯胺衍生物(卡巴氮平、奧卡西平、盧非醯胺)、脂肪酸衍生物(丙戊酸、丙戊醯胺、胺基丁酸、胺己烯酸、氟柳雙胺、噻加賓)及其他抗癲癇藥(舒噻嗪、苯乙醯脲、拉莫三嗪、非胺酯、托吡酯、加巴噴丁、苯丁醯脲、左乙拉西坦、唑尼沙胺、普瑞巴林、司替戊醇、拉科醯胺、卡立胺酯、瑞替加濱、布瓦西坦、氯丙醯苄胺)。
其他藥劑包括但不限於醫藥品(益生菌、消化助劑/消化劑、草藥萃取物)、維生素(水溶性和脂溶性二者,諸如但不限於維生素A、D3、E、K、B1、B5、B6、B12、C或彼等之衍生物)及營養補充品(輔酶,例如Q10;黃酮類,例如白藜蘆醇;卵磷脂;不飽和脂肪酸,其包括脂肪酸ω-3和ω-6)。
本發明之化合物亦可與磷酸二酯酶5同功酶抑制劑(PDE5)、一氧化氮供體、環氧合酶抑制劑、其他V1a受體拮抗劑(諸如巴羅非田)或L-精胺酸組合以用於治療及/或預防與V1a受體功能有關之疾病或病症。
該組合組成物可於單一劑型中或分開地包含本發明之化合物和另一活性物質。該組合組成物可經同時、分別或依序投予。
適當之劑型包括口服、直腸、黏膜、經皮或經腸投予;非經腸投予包括肌內、皮下、靜脈內和髓內注射及關節內、鞘內、直接心室內、腹膜內、鼻內或眼內注射和眼滴劑。
可替代地,該化合物可經局部且非全身地投予,例如藉由通常呈改質之釋出調製劑直接注射該化合物至腎臟或心臟。此外,可藉由標的載體系統(例如組織專一性抗體封裝之脂質體)投予藥物。該脂質體選擇性轉移該活性物質至吸收該活性物質之標的器官。
該醫藥組成物可經多種不同之方式和醫藥劑型投予。本發明之化合物可於單一或多個劑量經單獨或與醫藥上可接受之賦形劑組合投予。達到適當治療功效所需之劑量可廣泛地變化,且考量疾病階段、欲治療之病患的狀態和體重、活性物質之選擇性、劑量配方形式及每日治療次數,該劑量必須適合個體之需要。
對於簡單投予,較佳的是該醫藥組成物係由多個劑量單位組成,該劑量單位含有經一次投予之藥物劑量或少數量之該藥物劑量的多重、1/2、1/3或1/4劑量。該劑量單位係例如片劑,其可具有1/2或1/4溝槽以提供1/2或1/4分開之片劑以測定所需之藥物劑量。
含有本發明之活性物質的醫藥組成物通常含有0.01至500 mg活性物質/劑量單位。當然亦為可能的是每一調製劑之活性物質的量超出上述上限或下限。
通式(I)化合物之進一步較佳基團係其中下述環A、環B、X、Y、Z、R1 -R17 、Cy1 -Cy3 、n、p、q及r之各個實施態樣的可選擇之組合者。如下述定義之環A、環B、X、Y、Z、R1 -R17 、Cy1 -Cy3 、n、p、q及r之較佳實施態樣、更佳實施態樣或最佳實施態樣之任何組合亦為通式(I)化合物之較佳、更佳或最佳基團。
於本發明之某些實施態樣中,該通式(I)化合物中環A係4至6員飽和碳環。
於本發明之某些較佳實施態樣中,該通式(I)化合物中環A係環丁基或環己基。
於本發明之某些更佳實施態樣中,該通式(I)化合物中環A係環己基。
於本發明之某些實施態樣中,該通式(I)化合物中環A係含有1或2個N之4至7員飽和雜環基,其中環A係經由環氮與Y連接。
於本發明之某些實施態樣中,該通式(I)化合物中環A係含有1或2個N之4至7員飽和雜環基,其中環A係經由環氮與該5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯核之三唑環連接。
於本發明之某些實施態樣中,該通式(I)化合物中環A係四氫吖唉基、1,3-二(四氫吖唉基)、吡咯啶基、吡唑啶基、咪唑啶基、哌啶基、哌嗪基、氮雜環庚烷基、1,3-或1,4-二氮雜環庚烷基,其中環A係經由環氮與Y連接。
於本發明之某些實施態樣中,該通式(I)化合物中環A係四氫吖唉基、1,3-二(四氫吖唉基)、吡咯啶基、吡唑啶基、咪唑啶基、哌啶基、哌嗪基、氮雜環庚烷基、1,3-或1,4-二氮雜環庚烷基,其中環A係經由環氮與該5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯核之三唑環連接。
於本發明之某些較佳實施態樣中,該通式(I)化合物中環A係四氫吖唉-1,3-二基、哌啶-1,4-二基或哌嗪-1,4-二基,其中環A係經由環氮與Y或該5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯核之三唑環連接。
於本發明之某些實施態樣中,該通式(I)化合物中環B係可選擇地經取代之雜芳基。
於本發明之某些較佳實施態樣中,該通式(I)化合物中環B係可選擇地經取代之含有1個N、2個N或1個N和1個O之5或6員單雜芳基。
於本發明之某些更佳實施態樣中,該通式(I)化合物中環B係吡啶-2-基、3-氯-吡啶-2-基、3-甲基-吡啶-2-基、吡啶-3-基、嘧啶-2-基或5-甲基-異噁唑-3-基。
於本發明之某些最佳實施態樣中,該通式(I)化合物中環B係吡啶-2-基。
於本發明之某些實施態樣中,該通式(I)化合物中環B係可選擇地經取代之芳基。
於本發明之某些較佳實施態樣中,該通式(I)化合物中環B係可選擇地經取代之苯基。
於本發明之某些實施態樣中,該通式(I)化合物中環B係可選擇地經取代之雜環基。
於本發明之某些較佳實施態樣中,該通式(I)化合物中環B係可選擇地經取代之含有1個N或1個N和1個O之4至7員雜環基。
於本發明之某些更佳實施態樣中,該通式(I)化合物中環B係四氫吖唉-1-基、吡咯啶-1-基、哌啶基、哌嗪基或嗎啉-4-基。
於本發明之某些實施態樣中,若環B存在,該通式(I)化合物中Y係-O-。
於本發明之某些實施態樣中,若環B存在,該通式(I)化合物中Y係-C(O)-。
於本發明之某些實施態樣中,若環B存在,該通式(I)化合物中Y係-CH2 -。
於本發明之某些實施態樣中,若環B存在,該通式(I)化合物中Y係-NH-。
於本發明之某些實施態樣中,若環B存在,該通式(I)化合物中Y係單鍵。
於本發明之某些實施態樣中,若環B不存在,該通式(I)化合物中Y係-N(C1-4 烷基)2 、可選擇地經鹵素取代之C1-4 烷基、C1-4 烷氧基、C(O)OC1-4 烷基或鹵素。
於本發明之某些實施態樣中,若環B不存在,該通式(I)化合物中Y係可選擇地經鹵素取代之C1-4 烷基或C1-4 烷氧基。
於本發明之某些較佳實施態樣中,若環B不存在,該通式(I)化合物中Y係C1-3 烷基。
於本發明之某些更佳實施態樣中,若環B不存在,該通式(I)化合物中Y係甲基或乙基。
於本發明之某些較佳實施態樣中,若環B不存在,該通式(I)化合物中Y係C1-3 烷氧基。
於本發明之某些更佳實施態樣中,若環B不存在,該通式(I)化合物中Y係甲氧基或乙氧基。
於本發明之某些較佳實施態樣中,若環B不存在,該通式(I)化合物中Y係CF3
於本發明之某些實施態樣中,若環B不存在,該通式(I)化合物中Y係指選自-N(C1-4 烷基)2 、C(O)OC1-4 烷基、可選擇地經鹵素取代之C1-4 烷基、C1-4 烷氧基或鹵素中之一者。
於本發明之某些較佳實施態樣中,若環B不存在,該通式(I)化合物中Y係指選自二甲胺或CF3 中之一者。
於本發明之某些實施態樣中,若環B不存在,該通式(I)化合物中Y係指選自可選擇地經鹵素取代之C1-4 烷基、C1-4 烷氧基或鹵素中之二者。
於本發明之某些較佳實施態樣中,若環B不存在,該通式(I)化合物中Y係指選自C1-3 烷基、C1-3 烷氧基或氟中之二者。
於本發明之某些更佳實施態樣中,若環B不存在,該通式(I)化合物中Y係指選自甲基、乙基、甲氧基、乙氧基或氟中之二者。
於本發明之某些甚佳實施態樣中,若環B不存在,該通式(I)化合物中Y係指選自甲基或甲氧基中之二者。
於本發明之某些較佳實施態樣中,該通式(I)化合物中,環A係環丁基或環己基,Y係指選自-N(C1-4 烷基)2 、CF3 或鹵素中之一者,且環B不存在。
於本發明之某些較佳實施態樣中,該通式(I)化合物中,環A係環丁基或環己基,Y係指選自可選擇地經鹵素取代之C1-4 烷基、C1-4 烷氧基或鹵素中之二者,且環B不存在。
於本發明之某些較佳實施態樣中,該通式(I)化合物中,環A係環丁基或環己基,Y係-O-或單鍵且環B係苯基、2-甲基-苯基、四氫吖唉-1-基、吡咯啶-1-基、嗎啉-4-基、吡啶-2-基、3-氯-吡啶-2-基、3-甲基-吡啶-2-基、吡啶-3-基、嘧啶-2-基或5-甲基-異噁唑-3-基。
於本發明之某些更佳實施態樣中,該通式(I)化合物中,環A係環己基,Y係-O-且環B係吡啶-2-基、吡啶-3-基、3-氯-吡啶-2-基、3-甲基-吡啶-2-基、嘧啶-2-基或5-甲基-異噁唑-3-基。
於本發明之某些最佳實施態樣中,該通式(I)化合物中,環A係環己基,Y係-O-,且環B係吡啶-2-基。
於本發明之某些較佳實施態樣中,該通式(I)化合物中,環A係環己基,Y係單鍵,且環B係嗎啉-4-基。
於本發明之某些實施態樣中,該通式(I)化合物中,環A係環丁基或環己基,Y係-C(O)-,且環B係四氫吖唉-1-基、吡咯啶-1-基或嗎啉-4-基。
於本發明之某些實施態樣中,該通式(I)化合物中,環A係環丁基或環己基,Y係-CH2 -,且環B係可選擇地經取代之芳基、雜環基或雜芳基。
於本發明之某些實施態樣中,該通式(I)化合物中,環A係環丁基或環己基,Y係-CH2 -,且環B係可選擇地經取代之含有1個N或1個N和1個O之4至7員雜環基。
於本發明之某些較佳實施態樣中,該通式(I)化合物中,環A係四氫吖唉-1,3-二基、哌啶-1,4-二基或哌嗪-1,4-二基,其中環A係經由環氮與Y或該5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯核之三唑環連接,Y係-O-或單鍵,且環B係苯基、2-甲基-苯基、四氫吖唉-1-基、吡咯啶-1-基、嗎啉-4-基、吡啶-2-基、3-氯-吡啶-2-基、3-甲基-吡啶-2-基、吡啶-3-基、嘧啶-2-基或5-甲基-異噁唑-3-基。
於本發明之某些更佳實施態樣中,該通式(I)化合物中,環A係四氫吖唉-1,3-二基,其中環A係經由環氮與Y或該5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯核之三唑環連接,Y係單鍵,且環B係吡啶-2-基、嘧啶-2-基、哌啶基或可選擇地經取代之苯基。
於本發明之某些實施態樣中,該通式(I)化合物中B-Y-A-結合代表3H -螺[2-苯並呋喃-1,4’-哌啶-1’-基]、於3位置經C1-4 烷基取代之1-氧雜-3-氮雜螺[4.5]癸-2-酮-8-基或於2位置經C1-4 烷基、芳基或雜芳基取代之2-氮雜螺[4.5]癸-1-酮-8-基。
於本發明之某些實施態樣中,該通式(I)化合物中B-Y-A-結合代表3H -螺[2-苯並呋喃-1,4’-哌啶-1’-基]、(5S,8S)-3-甲基-1-氧雜-3-氮雜螺[4.5]癸-2-酮-8-基、(5R,8R)-3-甲基-1-氧雜-3-氮雜螺[4.5]癸-2-酮-8-基、(5R ,8R )-2-(丙-2-基)-2-氮雜螺[4.5]癸-1-酮或(5S ,8S )-2-(丙-2-基)-2-氮雜螺[4.5]癸-1-酮。
於本發明之某些實施態樣中,該通式(I)化合物中R1 係氫。
於本發明之某些實施態樣中,該通式(I)化合物中R1 係鹵素。
於本發明之某些較佳實施態樣中,該通式(I)化合物中R1 係氯、溴或氟。
於本發明之某些更佳實施態樣中,該通式(I)化合物中R1 係氯。
於本發明之某些實施態樣中,該通式(I)化合物中R1 係C1-4 烷基。
於本發明之某些較佳實施態樣中,該通式(I)化合物中R1 係甲基。
於本發明之某些實施態樣中,該通式(I)化合物中R1 係C1-4 烷氧基。
於本發明之某些較佳實施態樣中,該通式(I)化合物中R1 係甲氧基。
於本發明之某些實施態樣中,該通式(I)化合物中R1 係CF3
於本發明之某些實施態樣中,該通式(I)化合物中R1 係CN。
於本發明之某些實施態樣中,該通式(I)化合物中,R2 係氫或C1-4 烷基,R3 係(CH2 )n R4 、C(O)R5 或可選擇地經R6 取代之C1-4 烷基。
於本發明之某些實施態樣中,該通式(I)化合物中,R2 係氫,R3 係(CH2 )n R4 、C(O)R5 或可選擇地經R6 取代之C1-4 烷基。
於本發明之某些實施態樣中,該通式(I)化合物中,R2 係氫,R3 係(CH2 )n R4 ,其中R4 係CN、疊氮基或可選擇地經取代之含有2個N和1個O或4個N之5員單雜芳基,且n係0或1。
於本發明之某些實施態樣中,該通式(I)化合物中,R2 係氫,R3 係(CH2 )n R4 ,其中R4 係CN,且n係0或1。
於本發明之某些實施態樣中,該通式(I)化合物中,R2 係氫,R3 係(CH2 )n R4 ,其中R4 係四唑基或3-甲基-1,2,4-噁二唑-5-基,且n係0或1。
於本發明之某些實施態樣中,該通式(I)化合物中,R2 係氫,R3 係C(O)R5 ,其中R5 係C1-4 烷基。
於本發明之某些較佳實施態樣中,該通式(I)化合物中,R2 係氫,R3 係C(O)R5 ,其中R5 係甲基、乙基、異丙基或三級丁基。
於本發明之某些更佳實施態樣中,該通式(I)化合物中,R2 係氫,R3 係C(O)R5 ,其中R5 係甲基。
於本發明之某些實施態樣中,該通式(I)化合物中,R2 係氫,R3 係C(O)R5 ,其中R5 係C1-4 烷氧基。
於本發明之某些較佳實施態樣中,該通式(I)化合物中,R2 係氫,R3 係C(O)R5 ,其中R5 係甲氧基、乙氧基、丙氧基或三級丁氧基。
於本發明之某些更佳實施態樣中,該通式(I)化合物中,R2 係氫,R3 係C(O)R5 ,其中R5 係甲氧基。
於本發明之某些實施態樣中,該通式(I)化合物中,R2 係氫,R3 係C(O)R5 ,其中R5 係OH。
於本發明之某些實施態樣中,該通式(I)化合物中,R2 係氫,R3 係C(O)R5 ,其中R5 係NR7 R8 ,其中R7 和R8 獨立地氫、C1-4 烷基或Cy2
於本發明之某些較佳實施態樣中,該通式(I)化合物中,R2 係氫,R3 係C(O)R5 ,其中R5 係NR7 R8 ,其中R7 和R8 為氫。
於本發明之某些較佳實施態樣中,該通式(I)化合物中,R2 係氫,R3 係C(O)R5 ,其中R5 係NR7 R8 ,其中R7 係氫且R8 係甲基、乙基、異丙基或三級丁基。
於本發明之某些較佳實施態樣中,該通式(I)化合物中,R2 係氫,R3 係C(O)R5 ,其中R5 係NR7 R8 ,其中R7 和R8 獨立地甲基、乙基、異丙基或三級丁基。
於本發明之某些更佳實施態樣中,該通式(I)化合物中,R2 係氫,R3 係C(O)R5 ,其中R5 係NR7 R8 ,其中R7 和R8 為甲基。
於本發明之某些較佳實施態樣中,該通式(I)化合物中,R2 係氫,R3 係C(O)R5 ,其中R5 係NR7 R8 ,其中R7 係氫且R8 係可選擇地經取代之芳基或雜芳基。
於本發明之某些較佳實施態樣中,該通式(I)化合物中,R2 係氫,R3 係C(O)R5 ,其中R5 係NR7 R8 ,其中R7 和R8 與彼等連接之N一起形成可選擇地經取代之雜環。
於本發明之某些更佳實施態樣中,該通式(I)化合物中,R2 係氫,R3 係C(O)R5 ,其中R5 係NR7 R8 ,其中R7 和R8 與彼等連接之N一起形成可選擇地經取代之含有1個N、2個N、1個N和1個O或1個N和1個S之4至7員飽和雜環。
於本發明之某些實施態樣中,該通式(I)化合物中,R2 係氫,R3 係可選擇地經R6 取代之C1-4 烷基,其中R6 係OR9 、NR10 R11 、側氧基、-O(CH2 )m O-或一或多個鹵素。
於本發明之某些較佳實施態樣中,該通式(I)化合物中,R2 係氫,R3 係甲基、乙基或異丙基。
於本發明之某些更佳實施態樣中,該通式(I)化合物中,R2 係氫,R3 係異丙基。
於本發明之某些較佳實施態樣中,該通式(I)化合物中,R2 係氫,R3 係經R6 取代之單一或多個支鏈之C1-4 烷基,其中R6 係OR9 、NR10 R11 或一或多個鹵素。
於本發明之某些較佳實施態樣中,該通式(I)化合物中,R2 係氫,R3 係經R6 取代之單一或多個支鏈之C1-4 烷基,其中R6 係一或多個鹵素。
於本發明之某些實施態樣中,該通式(I)化合物中,R2 係氫,R3 係-CH2 鹵素、-CH(鹵素)CH3 或-C(鹵素)2 CH3
於本發明之某些較佳實施態樣中,該通式(I)化合物中,R2 係氫,R3 係-CH2 鹵素。
於本發明之某些更佳實施態樣中,該通式(I)化合物中,R2 係氫,R3 係-CH2 F。
於本發明之某些實施態樣中,該通式(I)化合物中,R2 係氫,R3 係經R6 取代之C1-4 烷基,其中R6 係側氧基或 -O(CH2 )2 O-。
於本發明之某些較佳實施態樣中,該通式(I)化合物中,R2 係氫,R3 係經R6 取代之單一或多個支鏈之C1-4 烷基,其中R6 係OR9 或NR10 R11
於本發明之某些較佳實施態樣中,該通式(I)化合物中,R2 係氫,R3 係-CH2 R6 或-CH(R6 )CH3 ,其中R6 係OR9 或NR10 R11
於本發明之某些更佳實施態樣中,該通式(I)化合物中,R2 係氫,R3 係-CH2 R6 ,其中R6 係OR9 或NR10 R11
於本發明之某些實施態樣中,該通式(I)化合物中,R2 係氫,R3 係經R6 取代之C1-4 烷基,其中R6 係OR9
於本發明之某些較佳實施態樣中,該通式(I)化合物中,R2 係氫,R3 係-CH2 R6 或-CH(R6 )CH3 ,R6 係OR9 ,其中R9 係氫。
於本發明之某些較佳實施態樣中,該通式(I)化合物中,R2 係氫,R3 係-CH2 R6 或-CH(R6 )CH3 ,R6 係OR9 ,其中R9 係可選擇地經NH2 或可選擇地經取代之芳基取代之C1-4 烷基。
於本發明之某些較佳實施態樣中,該通式(I)化合物中,R2 係氫,R3 係-CH2 R6 或-CH(R6 )CH3 ,R6 係OR9 ,其中R9 係經NH2 取代之乙基或異丙基。
於本發明之某些實施態樣中,該通式(I)化合物中,R2 係氫,R3 係-CH2 R6 或-CH(R6 )CH3 ,R6 係OR9 ,其中R9 係乙基或異丙基。
於本發明之某些實施態樣中,該通式(I)化合物中,R2 係氫,R3 係-CH2 R6 或-CH(R6 )CH3 ,R6 係OR9 ,其中R9 係Si(CH3 )3
於本發明之某些實施態樣中,該通式(I)化合物中,R2 係氫,R3 係-CH2 R6 或-CH(R6 )CH3 ,R6 係OR9 ,其中R9 係C(O)R12 ,R12 係C1-5 烷基。
於本發明之某些較佳實施態樣中,該通式(I)化合物中,R2 係氫,R3 係-CH2 R6 或-CH(R6 )CH3 ,R6 係OR9 ,其中R9 係C(O)R12 ,R12 係甲基。
於本發明之某些實施態樣中,該通式(I)化合物中,R2 係氫,R3 係-CH2 R6 或-CH(R6 )CH3 ,R6 係OR9 ,其中R9 係C(O)R12 ,R12 係NR14 R15
於本發明之某些較佳實施態樣中,該通式(I)化合物中,R2 係氫,R3 係-CH2 R6 或-CH(R6 )CH3 ,R6 係OR9 ,其中R9 係C(O)R12 ,R12 係NR14 R15 ,其中R14 和R15 係氫。
於本發明之某些較佳實施態樣中,該通式(I)化合物中,R2 係氫,R3 係-CH2 R6 或-CH(R6 )CH3 ,R6 係OR9 ,其中R9 係C(O)R12 ,R12 係NR14 R15 ,其中R14 和R15 獨立地氫或可選擇地經取代之芳基。
於本發明之某些較佳實施態樣中,該通式(I)化合物中,R2 係氫,R3 係-CH2 R6 ,R6 係OR9 ,其中R9 係C(O)R12 ,R12 係NR14 R15 ,其中R14 係氫,R15 係4-氟-苯基。
於本發明之某些實施態樣中,該通式(I)化合物中,R2 係氫,R3 係-CH2 R6 或-CH(R6 )CH3 ,R6 係OR9 ,其中R9 係C(O)R12 ,R12 係NR14 R15 ,其中R14 和R15 與彼等連接之N一起形成可選擇地經取代之含有1個N、2個N、1個N和1個O或1個N和1個S之4至7員飽和雜環。
於本發明之某些較佳實施態樣中,該通式(I)化合物中,R2 係氫,R3 係-CH2 R6 ,R6 係OR9 ,其中R9 係C(O)R12 ,R12 係NR14 R15 ,其中R14 和R15 與彼等連接之N一起形成嗎啉。
於本發明之某些實施態樣中,該通式(I)化合物中,R2 係氫,R3 係經R6 取代之C1-4 烷基,其中R6 係NR10 R11
於本發明之某些較佳實施態樣中,該通式(I)化合物中,R2 係氫,R3 係-CH2 R6 或-CH(R6 )CH3 ,R6 係NR10 R11 ,其中R10 和R11 係氫。
於本發明之某些實施態樣中,該通式(I)化合物中,R2 係氫,R3 係-CH2 R6 或-CH(R6 )CH3 ,R6 係NR10 R11 ,其中R10 和R11 獨立地氫或可選擇地經C1-4 烷氧基取代之C1-4 烷基。
於本發明之某些較佳實施態樣中,該通式(I)化合物中,R2 係氫,R3 係-CH2 R6 或-CH(R6 )CH3 ,R6 係NR10 R11 ,其中R10 和R11 獨立地氫、甲基、乙基、異丙基、二級丁基或三級丁基。
於本發明之某些較佳實施態樣中,該通式(I)化合物中,R2 係氫,R3 係-CH2 R6 或-CH(R6 )CH3 ,R6 係NR10 R11 ,其中R10 和R11 獨立地氫或經甲氧基或乙氧基取代之甲基、乙基或異丙基。
於本發明之某些實施態樣中,該通式(I)化合物中,R2 係氫,R3 係-CH2 R6 或-CH(R6 )CH3 ,R6 係NR10 R11 ,其中R10 和R11 獨立地氫或Cy3
於本發明之某些較佳實施態樣中,該通式(I)化合物中,R2 係氫,R3 係-CH2 R6 或-CH(R6 )CH3 ,R6 係NR10 R11 ,其中R10 係氫,R11 係含有1個O之4至7員飽和雜環基或具有孿鹵素取代之C4-6 環烷基。
於本發明之某些實施態樣中,該通式(I)化合物中,R2 係氫,R3 係經R6 取代之C1-4 烷基,其中R6 係NR10 R11 ,其中R10 和R11 獨立地氫或C(O)R13
於本發明之某些較佳實施態樣中,該通式(I)化合物中,R2 係氫,R3 係-CH2 R6 或-CH(R6 )CH3 ,R6 係NR10 R11 ,其中R10 係氫,R11 係C(O)R13 ,R13 係C1-4 烷基。
於本發明之某些更佳實施態樣中,該通式(I)化合物中,R2 係氫,R3 係-CH2 R6 ,R6 係NR10 R11 ,其中R10 係氫,R11 係C(O)R13 ,R13 係甲基。
於本發明之某些實施態樣中,該通式(I)化合物中,R2 係氫,R3 係經R6 取代之C1-4 烷基,其中R6 係NR10 R11 ,其中R10 和R11 獨立地氫或C(O)R13 ,其中R13 係Cy1
於本發明之某些實施態樣中,該通式(I)化合物中,R2 係氫,R3 係經R6 取代之C1-4 烷基,其中R6 係NR10 R11 ,其中R10 和R11 獨立地氫或C(O)R13 ,其中R13 係NR16 R17
於本發明之某些實施態樣中,該通式(I)化合物中,R2 係C1-4 烷基,R3 係(CH2 )n R4 、C(O)R5 或可選擇地經R6 取代之C1-4 烷基。
於本發明之某些實施態樣中,該通式(I)化合物中,R2 係C1-4 烷基,R3 係(CH2 )n R4
於本發明之某些實施態樣中,該通式(I)化合物中,R2 係C1-4 烷基,R3 係C(O)R5
於本發明之某些實施態樣中,該通式(I)化合物中,R2 和R3 係C1-4 烷基。
於本發明之某些較佳實施態樣中,該通式(I)化合物中,R2 和R3 係C1-3 烷基。
於本發明之某些更佳實施態樣中,該通式(I)化合物中,R2 和R3 獨立地甲基、乙基或異丙基。
於本發明之某些甚佳實施態樣中,該通式(I)化合物中,R2 和R3 係甲基。
於本發明之某些實施態樣中,該通式(I)化合物中,R2 係C1-4 烷基,R3 係經R6 取代之C1-4 烷基,其中R6 係OR9
於本發明之某些實施態樣中,該通式(I)化合物中,R2 係C1-4 烷基,R3 係經R6 取代之C1-4 烷基,其中R6 係OR9 ,其中R9 係C1-4 烷基。
於本發明之某些較佳實施態樣中,該通式(I)化合物中,R2 係甲基或乙基,R3 係-CH2 R6 或-CH(R6 )CH3 ,R6 係OR9 ,其中R9 係氫。
於本發明之某些實施態樣中,該通式(I)化合物中,R2 係C1-4 烷基,R3 係可選擇地經R6 取代之C1-4 烷基,其中R6 係NR10 R11 、側氧基、-O(CH2 )m O-或一或多個鹵素。
於本發明之某些實施態樣中,該通式(I)化合物中,R2 和R3 結合代表-(CH2 )p -O-(CH2 )q -,其中p係1、2或3且q係1、2或3。
於本發明之某些較佳實施態樣中,該通式(I)化合物中,R2 和R3 結合代表-(CH2 )p -O-(CH2 )q -,其中p和q之和係3。
於本發明之某些較佳實施態樣中,該通式(I)化合物中,R2 和R3 結合代表-(CH2 )p -O-(CH2 )q -,其中p和q之和係4。
於本發明之某些更佳實施態樣中,該通式(I)化合物中,R2 和R3 結合代表-(CH2 )p -O-(CH2 )q -,其中p係1,且q係2。
於本發明之某些更佳實施態樣中,該通式(I)化合物中,R2 和R3 結合代表-(CH2 )p -O-(CH2 )q -,其中p係1,且q係3。
於本發明之某些更佳實施態樣中,該通式(I)化合物中,R2 和R3 結合代表-(CH2 )p -O-(CH2 )q -,其中p係2,且q係2。
於本發明之某些實施態樣中,該通式(I)化合物中,R2 和R3 結合代表-(CH2 )r -,其中r係4、5或6。
於本發明之某些較佳實施態樣中,該通式(I)化合物中,R2 和R3 結合代表-(CH2 )r -,其中r係4。
雖然業已描述本發明於某些實施態樣、某些較佳實施態樣、某些更佳實施態樣或某些最佳實施態樣,但是不欲限制本發明之範圍於所揭露之特定形式,且相反地欲涵蓋如本發明之敘述界定的本發明之精神和範圍所包括的替代、修改及均等形式。本發明之化合物的可替代之請求範圍的實例可包括:
(1) 如上述或任何其他實施態樣所描述之通式(I)化合物。
(2) 如(1)或任何其他實施態樣所描述之化合物,其中R1 係氫、氟、氯、溴、甲基、甲氧基、CF3 或CN。
(3) 如(1)至(2)中任一項或任何其他實施態樣所描述之化合物,其中
環A係3至6員飽和碳環或含有1或2個N之4至7員飽和雜環;
環B係可選擇地經取代之5或6員單雜芳基、6至10員芳香族碳環或含有1、2或3個選自O、S或N的雜原子之4至7員飽和單環、雙環、稠合及/或橋連雜環;
或B-Y-A-結合代表3H -螺[2-苯並呋喃-1,4’-哌啶-1’-基];或

X係異丙基;
Z係甲基。
(4) 如(1)至(3)中任一項或任何其他實施態樣所描述之化合物,其中環B係可選擇地經取代之6員單雜芳基、苯基或含有1或2個選自O、S或N的雜原子之4至6員飽和單環雜環。
(5) 如(1)至(4)中任一項或任何其他實施態樣所描述之化合物,其中若環B存在,Y係-O-、-C(O)-、-CH2 -、-NH-或單鍵。
(6) 如(1)至(5)中任一項或任何其他實施態樣所描述之化合物,其中環A係4至6員飽和碳環基或經由環氮與Y或該5,6-二氫-4H -[1,2,4]三唑並[4,3-a][1]苯並吖呯核之三唑環連接的含有1或2個N之4至7員飽和雜環。
(7) 如(1)至(6)中任一項或任何其他實施態樣所描述之化合物,其中環A係環己基,Y係-O-,環B係吡啶-2-基,且R1 係氯。
(8) 如(1)至(6)中任一項或任何其他實施態樣所描述之化合物,其中環A係四氫吖唉、哌啶或哌嗪,Y係-O-或單鍵,環B係吡啶、嘧啶或哌啶,且R1 係氯。
(9) 如(1)至(3)中任一項或任何其他實施態樣所描述之化合物,其中Y係-N(C1-4 烷基)2 、C(O)OC1-4 烷基、可選擇地經鹵素取代之C1-4 烷基、C1-4 烷氧基或鹵素,且環B不存在。
(10) 如(9)或任何其他實施態樣所描述之化合物,其中環A係4至6員飽和碳環基。
(11) 如(10)或任何其他實施態樣所描述之化合物,其中Y係選自-N(C1-4 烷基)2 、C(O)OC1-4 烷基、可選擇地經鹵素取代之C1-4 烷基、C1-4 烷氧基及鹵素中之一者。
(12) 如(10)或任何其他實施態樣所描述之化合物,其中Y係選自可選擇地經鹵素取代之C1-4 烷基、C1-4 烷氧基及鹵素中之二者。
(13) 如(1)至(12)中任一項或任何其他實施態樣所描述之化合物,其中R2 係氫或C1-4 烷基且R3 係(CH2 )n R4
(14) 如(13)或任何其他實施態樣所描述之化合物,其中R2 係氫,R3 係(CH2 )n R4 ,R4 係CN、疊氮基或可選擇地經C1-4 烷基取代之含有2個N和1個O或4個N的5員單雜芳基。
(15) 如(1)至(12)中任一項或任何其他實施態樣所描述之化合物,其中R2 係氫或C1-4 烷基,且R3 係C(O)R5
(16) 如(15)或任何其他實施態樣所描述之化合物,其中R2 係氫。
(17) 如(16)或任何其他實施態樣所描述之化合物,其中R5 係甲基、甲氧基、OH或NR7 R8 ,其中R7 和R8 獨立地氫、甲基、乙基或異丙基。
(18) 如(1)至(12)中任一項或任何其他實施態樣所描述之化合物,其中R2 係氫或C1-4 烷基且R3 係可選擇地經R6 取代之C1-4 烷基。
(19) 如(18)或任何其他實施態樣所描述之化合物,其中R2 係氫或C1-4 烷基,且R3 係C1-4 烷基。
(20) 如(19)或任何其他實施態樣所描述之化合物,其中R2 係氫或C1-3 烷基,且R3 係C1-3 烷基。
(21) 如(18)或任何其他實施態樣所描述之化合物,其中R2 係氫或C1-4 烷基,且R3 係經R6 取代之單一或多個支鏈的C1-4 烷基。
(22) 如(21)或任何其他實施態樣所描述之化合物,其中R3 係-CH2 R6 或-CH(R6 )CH3
(23) 如(21)至(22)中任一項或任何其他實施態樣所描述之化合物,其中R2 係氫,且R6 係OR9
(24) 如(23)或任何其他實施態樣所描述之化合物,其中R9 係氫、可選擇地經苯基取代之C1-4 烷基或C(O)R12 ,其中R12 係甲基。
(25) 如(21)至(22)中任一項或任何其他實施態樣所描述之化合物,其中R2 係C1-4 烷基,且R6 係OR9
(26) 如(25)或任何其他實施態樣所描述之化合物,其中R2 係甲基或乙基,且R9 係氫。
(27) 如(21)至(22)中任一項或任何其他實施態樣所描述之化合物,其中R2 係氫,且R6 係NR10 R11
(28) 如(27)或任何其他實施態樣所描述之化合物,其中R10 和R11 獨立地氫、可選擇地經C1-4 烷氧基取代之C1-4 烷基、Cy3 或C(O)R13 ,或R16 和R17 與彼等連接之N一起形成可選擇地經取代之雜環。
(29) 如(21)至(22)中任一項或任何其他實施態樣所描述之化合物,其中R2 係C1-4 烷基,且R6 係NR10 R11
(30) 如(21)至(22)中任一項或任何其他實施態樣所描述之化合物,其中R2 係氫,且R6 係一或多個鹵素。
(31) 如(30)或任何其他實施態樣所描述之化合物,其中該鹵素係氟。
(32) 如(21)至(22)中任一項或任何其他實施態樣所描述之化合物,其中R2 係C1-4 烷基,且R6 係一或多個鹵素。
(33) 如(21)或任何其他實施態樣所描述之化合物,其中R6 係側氧基或-O(CH2 )2 O-。
(34) 如(13)至(33)中任一項或任何其他實施態樣所描述之化合物,其中該5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯核之第5位置的碳之絕對構型係(R)。
(35) 如(13)至(33)中任一項或任何其他實施態樣所描述之化合物,其中該5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯核之第5位置的碳之絕對構型係(S)。
(36) 如(1)至(12)中任一項或任何其他實施態樣所描述之化合物,其中R2 和R3 結合代表-(CH2 )p -O-(CH2 )q -,其中p係1、2或3,且q係1、2或3。
(37) 如(36)或任何其他實施態樣所描述之化合物,其中p和q之和係3或4。
(38) 如(1)至(12)中任一項或任何其他實施態樣所描述之化合物,其中R2 和R3 結合代表-(CH2 )r -,其中r係4、5或6。
本發明之較佳通式(I)化合物係例如下述化合物及/或彼等之鹽及/或溶劑合物及/或水合物及/或多形體及/或生物活性代謝物及/或前藥:
1. 8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯;
2. 8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸;
3. 8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺;
4. 8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-腈;
5. 8-氯-N,N-二甲基-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺;
6. (5S)-8-氯-N,N-二甲基-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺;
7. (5R)-8-氯-N,N-二甲基-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺;
8. 8-氯-N-(丙-2-基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺;
9. {8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}(吡咯啶-1-基)甲酮;
10. {8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}(嗎啉-4-基)甲酮;
11. {8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}(1,1-二氧基硫代嗎啉-4-基)甲酮;
12. {8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲醇;
13. {(5S)-8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲醇;
14. {(5R)-8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲醇;
15. 8-氯-5-(甲氧基甲基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯;
16. 嗎啉-4-甲酸酯{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲基;
17. (4-氟苯基)胺甲酸{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲酯;
18. 5-(疊氮基甲基)-8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯;
19. 1-{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲胺;
20. 8-氯-5-(嗎啉-4-基甲基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯;
21. 1-{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}-N,N-二甲基甲胺;
22. N-({8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲基)丙-2-胺;
23. 8-氯-1-[1-(嘧啶-2-基)四氫吖唉-3-基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯;
24. 8-氯-1-[反式-4-(二甲基胺基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯;
25. 8-氯-1-[反式-4-(三氟甲基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯;
26. 8-氯-1-[反式-4-(嗎啉-4-基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯;
27. {1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲醇;
28. 8-氯-1-[反式-4-(吡咯啶-1-基羰基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯;
29. {8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}乙腈;
30. 8-氯-5-[(4-甲基哌嗪-1-基)甲基]-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯;
31. 8-氯-5-(氟甲基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯;
32. 8-氯-5-(3-甲基-1,2,4-噁二唑-5-基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯;
33. 8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5-(2H-四唑-5-基)-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯;
34. N-({8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲基)四氫-2H-哌喃-4-胺;
35. N,N-二甲基-1-[反式-4-(吡啶-2-基氧基)環己基]-8-(三氟甲基)-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-胺;
36. 8-氯-1-[反式-3-甲基-2-側氧基-1-氧雜-3-氮雜螺[4.5]癸-8-基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯;
37. N-({8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲基)乙醯胺;
38. N-({8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲基)氧雜環丁烷-3-胺;
39. 8-氯-1-[反式-4-(嗎啉-4-基羰基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯;
40. 8-氯-1-[順式-3-甲基-2-側氧基-1-氧雜-3-氮雜螺[4.5]癸-8-基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯;
41. 8-氯-1-[1-(吡啶-2-基)四氫吖唉-3-基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯;
42. 1-[反式-4-(四氫吖唉-1-基羰基)環己基]-8-氯-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯;
43. 8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5-(吡咯啶-1-基甲基)-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯;
44. N-({8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲基)-2-甲基-N-(2-甲基丙基)丙-1-胺;
45. 8-氯-1-[反式-4-(哌啶-1-基羰基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯;
46. 1-(反式-4-{[4-(三級丁氧羰基)哌嗪-1-基]羰基}環己基)-8-氯-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯;
47. 8-氯-1-[反式-4-(哌嗪-1-基羰基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯鹽酸鹽;
48. 8-甲氧基-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯;
49. 8-甲基-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯;
50. 8-氯-N-(4-氟苯基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺;
51. 8-溴-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯;
52. 1-(1,4’-聯哌啶-1’-基)-8-氯-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺;
53. 8-氯-1-[(5r,8r)-1-側氧基-2-(丙-2-基)-2-氮雜螺[4.5]癸-8-基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯;
54. 1-{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}乙醇;
55. 1-{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}乙酮;
56. 8-氯-5-(氟甲基)-1-(反式-4-甲氧基-4-甲基環己基)-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯;
57. 1-{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}-N-(2-甲氧基乙基)乙胺;
58. 8-氟-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯;
59. 8-氯-5-(氟甲基)-1-[反式-4-(三氟甲基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯;
60. 5-(氟甲基)-1-(反式-4-甲氧基-4-甲基環己基)-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯;
61. {8-氟-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲醇;
62. 8-氟-5-(氟甲基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯;
63. 順式-(消旋)-1-{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}乙醇;
64. 反式-(消旋)-1-{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}乙醇;
65. (1R)-1-{(5R)-8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}乙醇;
66. (1S)-1-{(5S)-8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}乙醇;
67. (1S)-1-{(5R)-8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}乙醇;
68. (1R)-1-{(5S)-8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}乙醇;
69. 5-(氟甲基)-8-甲基-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯;
70. (5S)-8-氯-5-(氟甲基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯;
71. (5R)-8-氯-5-(氟甲基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯;
72. 8-氟-1-[反式-4-(三氟甲基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯;
73. 8-氯-1-[4-(2,3-二甲基苯基)哌嗪-1-基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺;
74. {8-氟-1-[反式-4-(三氟甲基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲醇;
75. {8-氯-5-甲基-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲醇;
76. {8-氯-5-乙基-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲醇;
77. (5S)-8-氟-5-(氟甲基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯;
78. (5R)-8-氟-5-(氟甲基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯;
79. 8-氯-1-[4-(3-氯苯基)哌嗪-1-基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺;
80. 8-氯-1-[4-(吡啶-2-基)哌嗪-1-基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺;
81. 8-氯-1-[4-(吡啶-2-基氧基)哌啶-1-基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺;
82. 5-[(苄氧基)甲基]-8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯;
83. 8-氯-1-(3,3-二氟環丁基)-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯;
84. 8-氯-1-[反式-4-(哌啶-1-基甲基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯;
85. 乙酸{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲酯;
86. 8-氯-1-(1’H,3H-螺[2-苯並呋喃-1,4’-哌啶]-1’-基)-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺;
87. 8-氯-N-(吡啶-2-基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺;
88. 8-氯-1-(1’H,3H-螺[2-苯並呋喃-1,4’-哌啶]-1’-基)-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-腈;
89. 8-氯-1-[4-(2,3-二甲基苯基)哌嗪-1-基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-腈;
90. 8-氯-1-[4-(吡啶-2-基)哌嗪-1-基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-腈;
91. 8-氟-5,5-二甲基-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯;
92. 8-氟-5-(丙-2-基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯;
93. 8’-溴-1’-[反式-4-(吡啶-2-基氧基)環己基]-4’H,6’H-螺[環戊烷-1,5’-[1,2,4]三唑並[4,3-a][1]苯並吖呯];
94. 8-溴-5-(丙-2-基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯;
95. 8’-溴-1’-[反式-4-(吡啶-2-基氧基)環己基]-2,3,5,6-四氫-4’H,6’H-螺[哌喃-4,5’-[1,2,4]三唑並[4,3-a][1]苯並吖呯];
96. 8-氟-5,5-二甲基-1-[反式-4-(三氟甲基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯;
97. (5R)-8-溴-5-(丙-2-基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯;
98. (5S)-8-溴-5-(丙-2-基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯;
99. 8’-溴-1’-[反式-4-(吡啶-2-基氧基)環己基]-4,5-二氫-4’H,6’H-螺[呋喃-3,5’-[1,2,4]三唑並[4,3-a][1]苯並吖呯];
100. 8’-氯-1’-[反式-4-(吡啶-2-基氧基)環己基]-2,3,5,6-四氫-4’H,6’H-螺[哌喃-4,5’-[1,2,4]三唑並[4,3-a][1]苯並吖呯];
101. 8’-氯-1’-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H,4’H,6’H-螺[哌喃-3,5’-[1,2,4]三唑並[4,3-a][1]苯並吖呯];
102. 8’-氯-1’-[反式-4-(吡啶-2-基氧基)環己基]-4,5-二氫-4’H,6’H-螺[呋喃-3,5’-[1,2,4]三唑並[4,3-a][1]苯並吖呯];
103. 8’-氯-1’-[反式-4-(三氟甲基)環己基]-2,3,5,6-四氫-4’H,6’H-螺[哌喃-4,5’-[1,2,4]三唑並[4,3-a][1]苯並吖呯];
104. (3R)-8’-溴-1’-[反式-4-(吡啶-2-基氧基)環己基]-4,5-二氫-4’H,6’H-螺[呋喃-3,5’-[1,2,4]三唑並[4,3-a][1]苯並吖呯];
105. (3S)-8’-溴-1’-[反式-4-(吡啶-2-基氧基)環己基]-4,5-二氫-4’H,6’H-螺[呋喃-3,5’-[1,2,4]三唑並[4,3-a][1]苯並吖呯];
106. (3S)-8’-氯-1’-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H,4’H,6’H-螺[哌喃-3,5’-[1,2,4]三唑並[4,3-a][1]苯並吖呯];
107. (3R)-8’-氯-1’-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H,4’H,6’H-螺[哌喃-3,5’-[1,2,4]三唑並[4,3-a][1]苯並吖呯];
108. 8-氟-1-(反式-4-甲氧基-4-甲基環己基)-5,5-二甲基-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯;
109. 8-氟-1-(順式-4-甲氧基-4-甲基環己基)-5,5-二甲基-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯;
110. 8’-氯-1’-(反式-4-甲氧基-4-甲基環己基)-2,3,5,6-四氫-4’H,6’H-螺[哌喃-4,5’-[1,2,4]三唑並[4,3-a][1]苯並吖呯];
111. 8’-氯-1’-(順式-4-甲氧基-4-甲基環己基)-2,3,5,6-四氫-4’H,6’H-螺[哌喃-4,5’-[1,2,4]三唑並[4,3-a][1]苯並吖呯];或
112. 8-氯-1-[反式-4-(吡啶-2-基胺基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯。
Brief description of the invention
The objective of the present invention is to synthesize a V1a receptor antagonist with a novel structure, the physicochemical properties (such as kinetic or thermodynamic solubility, ionization, lipophilicity or permeability) or pharmaceutical properties (such as metabolic stability Or CYP-450 enzyme inhibition) can provide favorable bioavailability, ADME (absorption, distribution, metabolism, and excretion), membrane penetration, or blood-brain barrier penetration.
Surprisingly, a novel 5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazepine derivative of the general formula (I) has been prepared , Which shows a profile of V1a receptor antagonistic activity.
The invention relates to compounds of general formula (I):

among them
Ring A is a cycloalkyl or heterocyclic group;
If ring B exists, Y is -O-, -C (O)-, -CH 2 -, -NH- or bonding; or, if ring B does not exist, Y is -N (C 1-4 alkyl) 2 , C (O) OC 1-4 Alkyl, optionally halogen-substituted C 1-4 Alkyl, C 1-4 Alkoxy or halogen;
Ring B is optionally substituted heteroaryl, aryl or heterocyclic;
Or BYA-Combined Representative 3 H -Spiro [2-benzofuran-1,4'-piperidine-1'-yl]; or

R 1 Hydrogen, halogen, C 1-4 Alkyl, C 1-4 Alkoxy, CF 3 Or CN;
R 2 Hydrogen or C 1-4 alkyl;
R 3 Department (CH 2 ) n R 4 , C (O) R 5 Or optionally via R 6 Replaced by C 1-4 alkyl;
Or R 2 With R 3 Combined Representative-(CH 2 ) p -O- (CH 2 ) q -Or- (CH 2 ) r -;
R 4 CN, azide or Cy 1 ;
R 5 Department C 1-4 Alkyl, C 1-4 Alkoxy, OH or NR 7 R 8 ;
R 6 Department of OR 9 , NR 10 R 11 , Pendant oxygen, -O (CH 2 ) 2 O- or one or more halogens;
R 7 And R 8 Independently hydrogen, C 1-4 Alkyl or Cy 2 Or R 7 And R 8 Forms a heterocyclic ring with their attached N;
R 9 Hydrogen, optionally via NH 2 Or optionally substituted C 1-4 Alkyl, Si (CH 3 ) 3 Or C (O) R 12 ;
R 10 And R 11 Independently hydrogen, optionally via C 1-4 Alkoxy-substituted C 1-4 Alkyl, Cy 3 Or C (O) R 13 , Or R 10 And R 11 Together with the N to which they are attached form an optionally substituted heterocyclic ring;
R 12 Department C 1-5 Alkyl or NR 14 R 15 ;
R 13 Department C 1-4 Alkyl, Cy 1 Or NR 16 R 17 ;
R 14 And R 15 Independently hydrogen or optionally substituted aryl, or R 14 And R 15 Together with the N to which they are attached form an optionally substituted heterocyclic ring;
R 16 And R 17 Department C 1-4 Alkyl, or R 16 And R 17 Together with the N to which they are attached form an optionally substituted heterocyclic ring;
Cy 1 Is optionally substituted heteroaryl;
Cy 2 Is optionally substituted aryl or heteroaryl;
Cy 3 Is optionally substituted cycloalkyl or heterocyclyl;
X series C 1-4 Alkyl, aryl or heteroaryl;
Z series C 1-4 alkyl;
n is 0 or 1;
p is 1, 2 or 3;
q is 1, 2 or 3;
r is 4, 5 or 6;
And / or their salts and / or their geometric isomers and / or their stereoisomers and / or their mirror isomers and / or their racemates and / or their Non-mirromeric isomers and / or their biologically active metabolites and / or their prodrugs and / or their solvates and / or their hydrates and / or their polymorphs.
The present invention also relates to a pharmaceutical composition containing the compound of general formula (I) and / or their salts and / or their geometric isomers and / or their stereoisomers and / or their Mirror isomers and / or their racemates and / or their non-mirro isomers and / or their prodrugs and / or their solvates and / or their hydrates and / Or their polymorphs as active substances.
In addition, the invention also relates to the compounds of the general formula (I) and / or their salts and / or their geometric isomers and / or their stereoisomers and / or their mirror isomers And / or their racemates and / or their non-mirromeric isomers and / or their prodrugs and / or their solvates and / or their hydrates and / or their Preparation of polymorphs, intermediates in the process of the preparation, and chemical and pharmaceutical preparations of pharmaceutical compositions containing these compounds.
The present invention also relates to a method for treating mammals, including humans, suffering from a central and / or peripheral disease that modulates and preferably antagonizes the V1a receptor, where the compound of formula (I) and / Or their salts and / or their geometric isomers and / or their stereoisomers and / or their mirror isomers and / or their racemates and / or their Non-mirromeric isomers and / or their prodrugs and / or their solvates and / or their hydrates and / or their polymorphs or their therapeutically effective amounts.
The invention also relates to the compounds of general formula (I) and / or their salts and / or their geometric isomers and / or their stereoisomers and / or their mirror isomers and / Or their racemates and / or their non-image isomers and / or their prodrugs and / or their solvates and / or their hydrates and / or their polymorphs For use in the manufacture of a medicament for the treatment and / or prevention of diseases or conditions related to the function of the Via receptor.

Detailed description of the invention
The present invention relates to V1a receptor modulators, and in particular to V1a receptor antagonists. Another object of the present invention is to provide selective Via receptor inhibitor compounds, because the selectivity is less likely to cause unwanted side effects. Another aspect of the present invention is to provide compounds having advantageous physicochemical properties, such as those that are expected to lead to beneficial bioavailability of the compound, ADME (absorption, distribution, metabolism and excretion), membranes Penetration or blood brain barrier penetration.
Therefore, the compound of general formula (I) of the present invention is a V1a receptor antagonist, which is a central and / or peripherally acting therapeutic agent for the treatment and / or prevention of various pathological symptoms of female sexual organs, persistent symptoms of blood pressure control, causes Symptoms caused by inappropriate secretion of vasopressin, anxiety, depression, aggressive behavior, central nervous system disease (wherein one of the symptoms of the disease and / or multiple symptoms may be related to or related to anxiety, depression or aggressive behavior The aforementioned 3 diseases show comorbidities (autism group disorder, obsessive-compulsive disorder, many different types of Down syndrome and post-traumatic stress disorder), aggressive behavior disorder and / or irritability, hyperactivity disorder, cognitive impairment Or other neuropsychiatric disorders.
The invention relates to compounds of general formula (I):

among them
Ring A is a cycloalkyl or heterocyclic group;
If ring B exists, Y is -O-, -C (O)-, -CH 2 -, -NH- or bonding; or, if ring B does not exist, Y is -N (C 1-4 alkyl) 2 , C (O) OC 1-4 Alkyl, optionally halogen-substituted C 1-4 Alkyl, C 1-4 Alkoxy or halogen;
Ring B is optionally substituted heteroaryl, aryl or heterocyclic;
Or BYA-Combined Representative 3 H -Spiro [2-benzofuran-1,4'-piperidine-1'-yl]; or

R 1 Hydrogen, halogen, C 1-4 Alkyl, C 1-4 Alkoxy, CF 3 Or CN;
R 2 Hydrogen or C 1-4 alkyl;
R 3 Department (CH 2 ) n R 4 , C (O) R 5 Or optionally via R 6 Replaced by C 1-4 alkyl;
Or R 2 With R 3 Combined Representative-(CH 2 ) p -O- (CH 2 ) q -Or- (CH 2 ) r -;
R 4 CN, azide or Cy 1 ;
R 5 Department C 1-4 Alkyl, C 1-4 Alkoxy, OH or NR 7 R 8 ;
R 6 Department of OR 9 , NR 10 R 11 , Pendant oxygen, -O (CH 2 ) 2 O- or one or more halogens;
R 7 And R 8 Independently hydrogen, C 1-4 Alkyl or Cy 2 Or R 7 And R 8 Forms a heterocyclic ring with their attached N;
R 9 Hydrogen, optionally via NH 2 Or optionally substituted C 1-4 Alkyl, Si (CH 3 ) 3 Or C (O) R 12 ;
R 10 And R 11 Independently hydrogen, optionally via C 1-4 Alkoxy-substituted C 1-4 Alkyl, Cy 3 Or C (O) R 13 , Or R 10 And R 11 Together with the N to which they are attached form an optionally substituted heterocyclic ring;
R 12 Department C 1-5 Alkyl or NR 14 R 15 ;
R 13 Department C 1-4 Alkyl, Cy 1 Or NR 16 R 17 ;
R 14 And R 15 Independently hydrogen or optionally substituted aryl, or R 14 And R 15 Together with the N to which they are attached form an optionally substituted heterocyclic ring;
R 16 And R 17 Department C 1-4 Alkyl, or R 16 And R 17 Together with the N to which they are attached form an optionally substituted heterocyclic ring;
Cy 1 Is optionally substituted heteroaryl;
Cy 2 Is optionally substituted aryl or heteroaryl;
Cy 3 Is optionally substituted cycloalkyl or heterocyclyl;
X series C 1-4 Alkyl, aryl or heteroaryl;
Z series C 1-4 alkyl;
n is 0 or 1;
p is 1, 2 or 3;
q is 1, 2 or 3;
r is 4, 5 or 6;
And / or their salts and / or their geometric isomers and / or their stereoisomers and / or their mirror isomers and / or their racemates and / or their Non-mirromeric isomers and / or their biologically active metabolites and / or their prodrugs and / or their solvates and / or their hydrates and / or their polymorphs.
The definitions of the general terms used herein (whether or not the term is discussed) are described below, either independently or in combination with other groups.
"Cycloalkyl" refers to a saturated or unsaturated (preferably saturated) carbocyclic group of 3 to 8 members (preferably 3 to 6 members), alone or in combination with other groups. Examples include cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl. On rings A and Cy 3 Herein, the "cycloalkyl group" preferably refers to a 4- to 6-membered saturated carbocyclic group. Examples include cyclobutyl, cyclopentyl or cyclohexyl. Preferred is cyclobutyl or cyclohexyl. Particularly preferred is cyclohexyl. "Substituted cycloalkyl" preferably refers to a cycloalkyl having a twin halogen substitution.
"Aryl" alone or in combination with other groups means an aromatic carbocyclic group of 6 to 14 members (preferably 6 to 10 members), which contains at least 1 aromatic ring or contains at least 1 aromatic ring Fused ring system. Examples include, but are not limited to, phenyl, benzyl, naphthyl, biphenyl, anthracenyl, fluorenyl, or dihydroindenyl. Particularly preferred is phenyl.
"Heterocyclyl" alone or in combination with other groups means a saturated or unsaturated (preferably saturated) 3 to 8 member (preferably 4 to 7 member) containing 1, 2, 3 or 4 members selected from A monocyclic, bicyclic, fused and / or bridged ring of a heteroatom of O, S or N. Preferred 4- to 7-membered monocyclic heterocyclic rings contain 1 N, 2 N, 1 O, 1 N and 1 O, or 1 N and 1 S. Examples include, but are not limited to, ethylene oxide, oxetane, tetrahydrofuran, tetrahydropiran, tetrahydroazetidine, pyrrolidine, piperidine, piperazine, morpholine, 1,3-oxazolidine , 1,3-thiazolidine, thiomorpholine-1,1-dioxide and the like. "Substituted heterocyclyl" preferably means C 1-4 Alkyl-substituted heterocyclic ring.
When ring A is a heterocyclic group, the heterocyclic group preferably refers to a saturated heterocyclic group containing 4 to 7 members containing 1 N or 2 N, wherein Ring A is connected to Y via a ring nitrogen atom or the 5 , 6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] triazole ring with a benzazepine core. Examples include, but are not limited to, tetrahydroazepine, 1,3-bis (tetrahydroazepine), pyrrolidinyl, pyrazolyl, imidazolidinyl, piperidinyl, piperazinyl, azacycloheptane Radical, 1,3- or 1,4-diazacycloheptyl. Preferred are tetrahydroacryl, piperidinyl or piperazinyl.
For Cy 3 The heterocyclic ring preferably refers to a saturated heterocyclic group containing 1 to 4 members, and more preferably refers to oxetane or tetrahydropiperan.
When ring B is a heterocyclic group, the heterocyclic group preferably refers to a saturated heterocyclic group of 4 to 7 members containing 1 N or 1 N and 1 O. Examples include, but are not limited to, tetrahydroazino, pyrrolidinyl, piperidinyl, piperazinyl, or morpholinyl.
When R 7 And R 8 , R 10 And R 11 , R 14 And R 15 Or R 16 And R 17 "When forming an optionally substituted heterocyclic ring with the N to which they are attached", the heterocyclic ring preferably means 4 to 7 members containing 1 N, 2 N, 1 N, and 1 O, Or a saturated ring of N and 1 S. Examples include, but are not limited to, morpholine, 4-methyl-piperazinyl, pyrrolidinyl, piperidinyl, piperazinyl, 1,3-oxazolidine, 1,3-thiazolidine, or thiomorpholine-1 , 1-dioxide.
"Heteroaryl" alone or in combination with other groups refers to a cyclic aromatic group containing a single 5- to 6-membered ring containing 1, 2, 3 or 4 heteroatoms, at least one Heterocyclic aromatic group. "6-membered monoheteroaryl" refers to a monocyclic aromatic group, which is a single 6-membered ring containing 1, 2, or 3 heteroatoms selected from O, S, or N. Examples include, but are not limited to, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, thiazinyl, oxazinyl, and the like. Preferred single 6-membered monoheteroaryl groups contain 1 or 2 N. Preferred 6-membered rings are pyridyl, more preferably pyridin-2-yl and pyridin-3-yl. Particularly preferred is pyridin-2-yl. "5-membered monoheteroaryl" means a single 5-membered ring monocyclic aromatic group containing 1, 2, 3, or 4 heteroatoms selected from O, S, or N. Preferred single 5-membered monoheteroaryls contain 2 N and 1 O, 2 N and 1 S, 4 N, 3 N, 2 N, 1 N or 1 S or 1 N and 1 O. Examples include, but are not limited to, thienyl, furyl, pyrrolyl, tetrazolyl, triazolyl, imidazolyl, thiazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, thiadiazolyl, 1 H -Pyrazolyl and the like. Preferred are isoxazol-3-yl, tetrazolyl and 1,2,4-oxadiazol-5-yl. "Substituted heteroaryl" preferably means C 1-4 Alkyl or halogen-substituted heteroaryl.
For Cy 1 , The optionally substituted heteroaryl preferably means C 1-4 Alkyl (preferably methyl) substituted single 5 membered monoheteroaryl containing 2 N and 1 O or 4 N. Preferred is tetrazolyl or 3-methyl-1,2,4-oxadiazol-5-yl.
When ring B is an optionally substituted heteroaryl, the heteroaryl refers to an optionally substituted halogen (preferably chlorine) or C 1-4 Alkyl (preferably methyl) substituted 5 or 6 membered monoheteroaryl groups containing 1 N, 2 N or 1 N and 1 O. Examples include, but are not limited to, pyridin-2-yl, 3-chloro-pyridin-2-yl, 3-methyl-pyridin-2-yl, pyridin-3-yl, pyrimidin-2-yl, or 5-methyl-iso Oxazol-3-yl.
"Bonding" refers to a single bond in which two atoms share a pair of electrons.
"C 1-4 Alkyl "or" C 1-5 "Alkyl", alone or in combination with other groups, refers to a straight or branched, single or multiple branched hydrocarbon group and is composed of 1 to 4 or 1 to 5 carbon atoms. Examples include, but are not limited to, methyl Methyl, ethyl, propyl, isopropyl, n-butyl, 2-butyl (secondary butyl), tertiary butyl, n-pentyl, tertiary pentyl, neopentyl, isopentyl, 2 -Pentyl (secondary pentyl) or 3-pentyl. Preferred alkyl groups are those having 1 to 3 carbon atoms. Preferred are methyl, ethyl, isopropyl, tertiary butyl and tertiary. Amyl. Particularly preferred is methyl.
"C 1-4 "Alkoxy", alone or in combination with other groups, means -OC 1-4 Alkyl, where the C 1-4 Alkyl is as defined above. Examples include, but are not limited to, methoxy, ethoxy, propoxy, or tertiary butoxy. Preferred alkoxy is methoxy, propoxy or tertiary butoxy. Particularly preferred are methoxy and tertiary butoxy.
"Boc", alone or in combination with other groups, refers to tertiary butoxycarbonyl.
"Halogen", alone or in combination with other groups, means fluorine, chlorine, bromine or iodine, preferably fluorine, chlorine or bromine. For R 1 And R 6 Preferred is fluorine, chlorine or bromine. For R 1 More preferred is chlorine or bromine, and even more preferred is chlorine. For R 6 More preferably, it is fluorine.
"Optionally substituted" for any atom of the relevant group means one or more C 1-4 Alkyl or halogen substituted. Here, "one or more" means 1 to the highest possible number of substitutions, that is, from one hydrogen to all hydrogens. Therefore, 1, 2 or 3 substitutions of a given atom are preferred. Very preferred are 1 or 2 substitutions or 1 substitution. For optionally substituted cycloalkyl, aryl, heterocyclyl or heteroaryl, 1 substitution is particularly preferred.
"C optionally substituted with halogen 1-4 "Alkyl" preferably refers to C 1-4 C with 1, 2 or 3 halogen substitutions on any atom of the alkyl group 1-4 Alkyl refers more preferably to methyl having 3 halogen substitutions. Especially preferred is CF 3 .
In "Through R 6 Replaced by C 1-4 Alkyl ", the C 1-4 Alkyl is as defined above. In "Through R 6 Replaced by C 1-4 Alkyl ", the substituted C 1-4 Alkyl is preferably a branched, single or multiple branched hydrocarbon group consisting of 1 to 4 carbon atoms. In "Through R 6 Replaced by C 1-4 "Alkyl" means "substituted" means that one or more R 6 To replace. The "one or more" means 1 to the highest possible number of substitutions, ie from 1 hydrogen to all hydrogens. When R 6 Department of OR 9 Or NR 10 R 11 When "The R 6 Replaced by C 1-4 "Alkyl" is, for example, but is not limited to, -CH 2 R 6 Or -CH (R 6 ) CH 3 , Preferably -CH 2 R 6 . When R 6 System side oxygen or -O (CH 2 ) 2 O-, the " 6 Replaced by C 1-4 "Alkyl" is, for example but not limited to, -C (R 6 ) CH 3 . When R 6 In the case of one or more halogens, the " 6 Replaced by C 1-4 "Alkyl", "one or more", and "halogen" are as defined above. Here, the " 6 Replaced by C 1-4 "Alkyl" is, for example, but is not limited to, -CH 2 Halogen, -CH (halogen) CH 3 Or -C (halogen) 2 CH 3 , Preferably -CH 2 halogen.
"Salt" means a pharmaceutically acceptable and / or pharmaceutically unacceptable salt. The pharmaceutically acceptable salts refer to the conventional acid addition salts and base addition salts which retain the biological efficacy and properties of the compound of the general formula (I) and can be passed through a suitable non-toxic organic or inorganic acid or organic Or inorganic base formation. Examples of acid addition salts include salts derived from inorganic acids such as, but not limited to, hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, sulfamic acid, phosphoric acid, nitric acid, and perchloric acid, and derived from various organic acids (Such as, but not limited to, acetic acid, propionic acid, benzoic acid, glycolic acid, phenylacetic acid, salicylic acid, malonic acid, maleic acid, oleic acid, binaconic acid, palmitic acid, benzenesulfonic acid, toluenesulfonic acid Acid, methanesulfonic acid, oxalic acid, tartaric acid, succinic acid, citric acid, malic acid, lactic acid, glutamic acid, fumaric acid, and the like). Examples of base addition salts are salts derived from ammonium hydroxide, potassium hydroxide, sodium hydroxide, and quaternary ammonium hydroxide, such as tetramethylammonium hydroxide. These salts generally have more favorable solubility properties than the compounds themselves used to prepare them and are therefore more suitable for use in the preparation of, for example, liquid or emulsion modulators. The pharmaceutically unacceptable salt is preferably used for the purification and isolation of the compound of the general formula (I) and therefore falls within the scope of the present invention.
"Prodrug" means a derivative of the compound of general formula (I) of the present invention, which does not have a therapeutic effect itself, but contains a group that becomes a "bioactive metabolite" after chemical or metabolic degradation (biotransformation) in vivo, The bioactive metabolite assumes the therapeutic effect. The decomposing group (especially suitable for a prodrug) connected to the compound of the general formula (I) of the present invention is well known in this technical field and also applicable to the compound of the present invention (Rautio et al., Nat Rev Drug Discov 2008, 7: 255-270).
The compound of the general formula (I) may exist in various geometrically isomeric forms. In addition, certain compounds of general formula (I) may contain one or more asymmetric centers and may therefore exist as stereoisomers and non-mirror isomers. All such compounds, such as cis isomers, trans isomers, non-mirromeric mixtures, racemates, non-racemic mixtures of mirror isomers, substantially pure and pure mirror isomers Within the scope of the present invention. The substantially pure enantiomer is up to 5% by weight of the corresponding opposite enantiomer, preferably 2% by weight and most preferably 1% by weight.
Optical isomers can be prepared by resolving racemic mixtures by conventional methods, such as by using an optically active acid or base to form a non-mirromeric isomer salt or by forming a covalent non-mirror isomer. Suitable acids include, for example, tartaric acid, diethylfluorenyl tartaric acid, dibenzyl tartaric acid, xylylene tartaric acid, and camphorsulfonic acid. By methods known in this technical field, such as chromatography or fractional crystallization, non-image-isomer mixtures can be separated into individual non-image-isomers based on the physical and / or chemical differences of the individual non-image-isomers. The optically active base or acid is then released from the isolated non-mirromeric isomers. Various methods of separating optical isomers include chiral chromatography (eg, chiral HPLC columns), which are optionally used to derive the goal of optimal separation of mirror isomers. Suitable chiral HPLC columns are Diacel columns, such as CHIRALPAK or CHIRALCEL columns, and they can be routinely selected as desired. Where applicable, catalyzed separation by derivatization can also be used. Optically active compounds of general formula (I) can also be isolated by chiral synthesis without racemic reaction conditions and using optically active starting materials.
With the literature (Freedman et al., Chirality 2003, 15 (9): 743-58; Stephens et al., Chirality 2008, 20 : 643-663) described by the Vibration Circular Dichroism Spectroscopy (VCD) method and / or the 1 H NMR spectral analysis (Seco et al., J Org Chem 1999, 64: 4669-4675; Seco et al., Tetrahedron Asymmetry 2001, 12: 2915-2925; Latypov et al., J. Am. Chem. Soc . 1998, 120 , 4741-4751), to determine the absolute configuration of the chiral compound.
The compound of the general formula (I) may exist in various polymorphic forms. As is known in the art, the ability of a polymorphic system compound to crystallize into more than one crystalline form (ie, a polymorphic form). Polymorphic forms of specific compounds can be defined by the same chemical formula or composition. The chemical structures of these polymorphic forms are different, as are the crystal structures of two different compounds.
The compounds of the general formula (I) and their salts may also exist in the form of solvates or hydrates, and these solvates and hydrates also fall within the scope of the present invention. The "solvate" refers to a non-covalent combination of a solvent and a solute. The "hydrate" refers to a non-covalent combination of water and solute.
The present invention also relates to a pharmaceutical composition containing the compound of the general formula (I) and / or their salts and / or their geometric isomers and / or their stereoisomers and / or their Mirror isomers and / or their racemates and / or their non-mirro isomers and / or their prodrugs and / or their solvates and / or their hydrates and / Or their polymorphs.
The invention also relates to chemical and pharmaceutical preparations of pharmaceutical compositions containing the compounds of the general formula (I) and / or their salts and / or their geometric isomers and / or their stereoisomers Structures and / or their mirror isomers and / or their racemates and / or their non-mirro isomers and / or their prodrugs and / or their solvates and / Or their hydrates and / or their polymorphs.
The pharmaceutical composition of the present invention can be formulated into a variety of different pharmaceutical formulations, such as, but not limited to, solid oral dosage forms such as tablets (eg, buccal, sublingual, foaming, chewable, and orally dispersible), Capsules, pills, pills, oral dispersible films, granules and powders; liquid preparations such as solutions, emulsions, suspensions, syrups, elixirs and drops; parenteral forms such as intravenous, intramuscular and subcutaneous injections; Other medical types, such as eye drops, semi-solid ophthalmic preparations, semi-solid skin preparations (such as ointments, creams and pastes), transdermal therapeutic systems, suppositories, rectal capsules, rectal solutions, emulsions and suspensions, etc.
One embodiment of the present invention relates to pharmaceutical compositions for pediatrics, such as, but not limited to, solutions, syrups, elixirs, suspensions, powders for preparing suspensions, dispersible or foamable tablets, chewable Tablets, orally dispersible tablets, tablets or coated tablets, orally foaming powders or granules, or capsules.
By methods known per se, such as conventional mixing, dissolving, emulsifying, suspending, microencapsulating, lyophilizing, extrusion and rounding, lamination, film coating, granulation, encapsulation, sugar coating or compression, The pharmaceutical composition of the present invention is prepared.
The pharmaceutical composition of the present invention can be formulated by customary means using one or more physiologically acceptable excipients (including binding agents) which promote the active substance and Into a pharmaceutically acceptable pharmaceutical dosage form. Proper formulation depends on the mode of administration chosen. Any technique and excipient known in the art can be used.
Excipients suitable for preparation can be selected from the following ranges, such as but not limited to fillers for tablets and capsules, binding agents for tablets and capsules, modified drug release agents, disintegrants, glidants, Lubricants, sweeteners, taste-masking agents, flavoring agents, coatings, surfactants, stabilizers, preservatives or antioxidants, buffers, complexing agents, wetting agents or emulsifiers, osmotic salts, lyophilization Excipients, microcapsules, salves, penetration enhancers, solubilizers, solvents, suppositories and suspending agents. Suitable pharmaceutical excipients can be, for example, starch, microcrystalline cellulose, talc, glucose, lactose, gelatin, silica, talc, magnesium stearate, sodium stearate, glyceryl monostearate, cellulose-derived Substances, sodium chloride, glycerol, propylene glycol, water, ethanol, and the like.
Another aspect of the present invention relates to the use of specific binding agents that can improve the solubility, solubility, permeability, absorption, or bioavailability of the active substance, such as but not limited to hydrophilic polymers, hot melt extrusion Forming agents, surfactants, buffering agents, complexing agents, emulsifiers, lyophilized excipients, disintegrating agents, microcapsules, penetration enhancers, solubilizers, co-solvents and suspending agents.
The above-mentioned excipients and various preparation methods are only representative examples. Other materials and process technologies known in the art can also be used.
"Disease or condition related to V1a receptor function" or "Disease or condition related to central and / or peripheral regulation (preferably V1a receptor antagonism)" means a variety of pathological symptoms, blood pressure control selected from the female sex organ Persistent symptoms, symptoms caused by inappropriate secretion of vasopressin, anxiety, depression, aggressive behavior, central nervous system disease (wherein one of the symptoms of the disease and / or multiple symptoms may be related to anxiety, depression or Aggressive behaviors or comorbidities with the aforementioned three diseases (autism group disorder, obsessive-compulsive disorder, many different types of Down syndrome and post-traumatic stress disorder), aggressive behavior disorder and / or irritability, behavior Diseases or disorders of hyperactivity, cognitive impairment, or other neuropsychiatric disorders.
Various pathological symptoms of the female sex organ include, but are not limited to, dysmenorrhea (primitive and / or secondary) or sexual dysfunction.
Persistent symptoms of this blood pressure control include, but are not limited to, hypertension and / or chronic heart failure.
Symptoms caused by the inappropriate secretion of vasopressin include, but are not limited to, diabetes insipidus, renal failure, renal syndrome, or cirrhosis.
The central nervous system diseases (wherein one of the symptoms of the disease and / or multiple symptoms may be related to anxiety, depression or aggressive behavior or show comorbidities with the aforementioned three diseases) include but are not limited to autism group disorders (good Functional Autism, Asperger's Syndrome, PWD-NOS, ASD and ASD and many different syndromes: Fragile X Chromosome, Prideway Syndrome, Leiter's disease and tuberous sclerosis), obsessive-compulsive disorder (OCD), many different types of Down syndrome and post-traumatic stress disorder (PTSD).
This aggressive behavior disorder and / or irritability includes, but is not limited to, ASD, Huntington's disease, or different types of schizophrenia.
The behavioral hyperactivity disorder includes, but is not limited to, attention deficit hyperactivity disorder.
The cognitive impairment includes, but is not limited to, dementia, mild cognitive impairment, cognitive impairment associated with schizophrenia, or Alzheimer's disease.
The other neuropsychiatric diseases include, but are not limited to, schizophrenia and related diseases.
In one embodiment, the disease or condition related to the function of the Via receptor or the disease or condition related to central and / or peripheral regulation (preferably the antagonist of the Via receptor) refers to an autism group disorder.
The present invention relates to a method for treating and / or preventing a disease or condition related to the function of a Via receptor, which comprises administering a treatment effective to an individual (preferably a mammal and more preferably a human) in need of the treatment and / or prevention. Amounts of the compounds of the general formula (I) alone and / or their salts and / or their geometric isomers and / or their stereoisomers and / or their mirror isomers and / or their And other racemates and / or their non-mirromeric isomers and / or their prodrugs and / or their solvates and / or their hydrates and / or their polymorphs or At least one pharmaceutically acceptable excipient is in the form of a pharmaceutical modulator.
The present invention relates to a method for treating an individual (preferably a mammal and more preferably a human) suffering from a disease or condition selected from a variety of pathological symptoms of female sex organs, persistent symptoms of blood pressure control, and vasopressor Symptoms, anxiety, depression, aggressive behavior, central nervous system disease (where one or more of the symptoms of the disease and / or multiple symptoms may be related to anxiety, depression or aggressive behavior or related to the aforementioned three Illnesses showing comorbidities (autism group disorder, obsessive-compulsive disorder, many different types of Down syndrome and post-traumatic stress disorder), aggressive behavior disorder and / or irritability, hyperactivity disorder, cognitive impairment or other nerves Mental illness, or a combination of these disorders. The method of treatment comprises administering a therapeutically effective amount of a compound of general formula (I) and / or their salts and / or their geometric isomerism to an individual (preferably a mammal and more preferably a human) in need of the treatment. And / or their stereoisomers and / or their mirror isomers and / or their racemates and / or their non-image isomers and / or their prodrugs and / Or their solvates and / or their hydrates and / or their polymorphs. The treatment method may include administering a therapeutically effective amount of a pharmaceutical composition to an individual (preferably a mammal and more preferably a human) in need of the treatment, the pharmaceutical composition comprising a compound of general formula (I) and / or Salts and / or their geometric isomers and / or their stereoisomers and / or their mirror isomers and / or their racemates and / or their non-image isomers And / or their prodrugs and / or their solvates and / or their hydrates and / or their polymorphs.
The invention relates to the compounds of the general formula (I) and / or their salts and / or their geometric isomers and / or their stereoisomers and / or their mirror isomers and / or Their racemates and / or their non-image isomers and / or their prodrugs and / or their solvates and / or their hydrates and / or their polymorphs are Use in the manufacture of a medicament for the treatment and / or prevention of a disease or condition related to the function of the Via receptor.
The "treatment" refers to the alleviation of specific pathological symptoms in patients or individuals who have suffered or been diagnosed with the disease, the removal or alleviation of one or more symptoms, the slowing or elimination of the progression of the disease state, and the prevention or recurrence of pathological symptoms or delay. If a drug has been administered to a patient who has developed a disease or disorder, the drug is generally administered by the same or similar method to implement the "prevention" (or prevention or delay in the occurrence of the disease).
A "therapeutically effective amount" refers to the amount of active substance that causes the treatment, cure, prevention, or amelioration of a disease or pathological symptom or side effect, and can reduce the disease or pathological symptom compared to a corresponding individual who has not received the therapeutically effective amount. progress. The therapeutically effective amount also includes an effective amount that enhances normal physiological functions. For the treatment, a therapeutically effective amount of the compound of the general formula (I) and / or their geometric isomers and / or their stereoisomers and / or their mirror isomers and / Or their racemates and / or their non-mirromeric isomers and / or their prodrugs and / or their solvates and / or their hydrates and / or as many of them Forms and any of their pharmaceutically acceptable salts are used as raw chemicals. In addition, the active substance can be provided as a pharmaceutical preparation. The compounds of general formula (I) and / or their salts and / or their geometric isomers and / or their stereoisomers and / or their mirror isomers and / or their Accurate treatment of racemates and / or their non-mirromeric isomers and / or their prodrugs and / or their solvates and / or their hydrates and / or their polymorphs The effective amount depends on many factors, including but not limited to the age and weight of the individual (patient) being treated, the exact type and severity of the disease to be treated, the nature of the drug and the route of administration.
"Mammal" means any member of the "mammalian group", including but not limited to humans.
The present invention also relates to a pharmaceutical composition suitable for treating a disease or condition related to central and / or peripheral regulation (preferably V1a receptor antagonism), which comprises the compound of the general formula (I) and / or their Salts and / or their geometric isomers and / or their stereoisomers and / or their mirror isomers and / or their racemates and / or their non-image isomers And / or their prodrugs and / or their solvates and / or their hydrates and / or their polymorphs.
The compounds of the present invention may also be combined with one or more compounds of the present invention or one or more other active substances (e.g., psycho blockers, psychostimulants, antihypertensives, antispasmodics, antiepileptics or other agents) to For mammals suffering from central and / or peripheral regulation of the V1a receptor and preferably antagonizing central and / or peripheral disease that may have therapeutic benefits, including but not limited to humans.
Psychotic agents include, but are not limited to, antipsychotics, anxiolytics, and sedative hypnotics or anesthetics.
Antipsychotics include, but are not limited to, typical and atypical antipsychotics, such as phenothiazine (chloropromazine, promethazine, methisobutazine, acepromazine, triflurazine, Salmepramine (ciamemazine, chlorpromazine, protipendyl), piperazine-derived phenothiazine (desicillazine, flufenazine, meperpromazine, oxypiperazine, propane) Clopirazine, Thioprazine, Triflurilazine, Epiphenazine, Thipromazine, Butapiperazine, Mepiperazine), Piperidine-derived phenothiazine (piperazine, Methiopiperidine, mesopyrazine, piperazine), thioanthracene (chloroprothione, cloopenthixole, flupentiaxant, meperidine, thioprene, and meclizine) , Butyric acid ketone derivatives (haloperidol, trihaloperidol, mepirone, mopirone, pipipronol, bromperidone, benziprodone, droperidol, and thimeridone , Fluanidone), diphenylbutylpiperidine derivatives (fluspirene, pentafluridol, amimidamine), diazine or oxacridine or thiazine derivatives (clozapine, Austria Azapine, clothiapine, quetiapine, losapine, asena ), Indole derivatives (serindole, ziprasidone, lurasidone, morpholininone, oxipertine), benzamidine derivatives (sulpiride, sutopril, thiopiridine) Rilimb, remabilil, amisulpride, verapride, nemabilil, vilapirab) or other agents (risperidone, aripiprazole, carbazrazine, epiprazole , Metopramine, moxapamine, iperidone, paliperidone, amozapine, ampipirone, piropiron, carbamate, chlorcarbamine, tetrabenazine, lithium).
Anxiolytics include, but are not limited to, benzodiazepine (diazepine, clodazapine, medarazapine, nordazapine, potassium clozapine acid, lenapine, azozolan, bromazepine Ping, Ketazopine, Ketazolol, Ciprodazapine, Alprazota, Trifluzapine, Propynezepine, Carzapine, Prozapine, Fludiazepine, Chlorflurane , Etizolam, clotazapine, oxazolam, tophizopam), diphenylmethane derivatives (hydroxyzine, butylthiodiphenylamine), carbamates (methalamine Esters, emilcamate, mevalyl amine esters), dibenzobicyclooctadiene derivatives (benzoquinones), azaspirodecanediones (buspirone), other agents (mefenam Oxadione, Guidocar, Chlorphenazine, Fabomotizole, Trimethosine), by increasing GABA A Derivatives or compounds that act on serotonin receptors, and other GABA neurogenic agents (such as GABA A α5 NAMs (e.g. Parmesani) and GABA A α5 PAMs (e.g. RG7816)).
Sedative hypnotics or narcotics include, but are not limited to, barbiturates (pentabarbital, isopentabarbital, n-butabarbital, barbiturate, allyl isobarbital, sercobarbital, Tarbbitol, Ethylbital, Pentenylbital, Cycbarbital, Heptobarbital, Westcobarbital, Mesopital, Cyclohexene Barbital, Thiopental Sodium, Ixabi , Alobarbital, proxibarbital), aldehydes (chloral hydrate, chloralhexanol, acetamigamine, chloral hydrate, antichlorin dichloride, Polyacetaldehyde), benzodiazepines (fluzepam, nitrazepam, flunazepam, estazolam, triazolam, clorazepam, temazepam, midazolam, bromozozolam , Quasiazepam, cloproxen, dulozepam, cinozepam), piperidine dione derivatives (acetophenone, mesperidone, diethylpyridine dione), cyclopyrrolidone benzodione Acridine derivatives (zopiclone, zolpidem, zaleplon, eszopiclone), melatonin receptor agonists (melatonin, rismetamine), or other hypnotics and sedatives (a Quinone, clomethiazole, bromisoval , Diethylbromide, sulfonamide, promethazine, trichloroethionic acid, chloropentynol, valerian root, hexyl propyl ester, bromide, propylene isopropyl acetate, pentamidine, methyl Pentynol, nicotinamide piperazine, dexmedetomidine).
Psychostimulants include, but are not limited to, psychostimulants or antidepressants.
Psychological stimulants include, but are not limited to, centrally acting sympathomimetics (amphetamine, dextroamphetamine, methamphetamine, methylphenidate, phenisotoin, fentalamine, modafinil, ethampoxone, alfa Tomoxetine, phenetilline, dextromethylphenidate, amphetamine mesylate), nootropics or other psychostimulants (caffeine, valprofen, meclofenate, pyridoxine Thiols, bisulfentan, dimethylamine ethanol, non-pisetide, citicoline, oxiracetam, pirostano, linopyridine, nizophenone, aniracetam, ethidium carnitine , Idebenone, Propylpyrrolid, Perbenmethanol, Pramiracetam, Averfinib, Vinpocetine, Tacrine, Donepezil, Carbatin, Galantamine, Ipidarin (ipidachrine ), Memantine, mebicar, fenibut).
Antidepressants include, but are not limited to, non-selective monoamine reuptake inhibitors (norpromazine, imipramine, imipramine oxide, clomipramine, opipritol, trimethoprim, rofil Palmin, dibenzepine, amitriptyline, normitamitriptyline, protiline, dushipin, ipindole, melitrazine, dintiline, dutiapine, amozapine, Metformin, aminidine, morphine, quinipramine), serotonin regulators and stimulants (verazolidone, vortioxetine), selective serotonin reuptake inhibitors (Qimimet Dioxetine, fluoxetine, paroxetine, sertraline, alapropionate, fluvoxamine, etoperidone, citalopram, lipunon), non-selective hydrazine-derived monoamine oxidase inhibitors (isoazohydrazine, nepal Yalamine, Phenylhydrazine, Trans-Phenylcypromine, Isonicotine Isoprazine, Isoproline (iprocloside), Non-hydrazine Monoamine Oxidase Inhibitors (Mortochlor, Toloxadone) Or other agents (hydroxytryptamine, tryptophan, mianserin, amine benzisoquine, trazodone, nefazodone, benmorpholine, dibenzilam, clotrimazole, oxafluoxine Mirtazapine, medesamin, tiempritine, pivagabine, venlafaxine, milnacipran, reboxetine, pyrididol, duloxetine, agomelatine , Desvenlafaxine, bupropion, gepirone, grass hypericum extract).
Antihypertensive drugs include, but are not limited to, beta-blockers, thiazide diuretics, angiotensin-converting enzyme inhibitors, calcium antagonists, angiotensin receptor antagonists (losartan), lovum bio Bases (Cinnamide, Reserpine, Norrepinepine, Mesoserpine, Bitaxerpine), Methyldopa, Imidazoline Receptor Agonists (Cronin, Chlophene, Tol Lonidine, Moxonidine, Rimenidine), Ganglion Blocking Anti-Adrenaline Agents (Gadolinium-Derived Mithifene, Secondary and Tertiary Amines Tetramethyl Dicycloheptylamine), Peripheral-acting Anti-Adrenaline Agents, alpha adrenergic receptor blockers (prazosin, indolamin, tramazosin, doxazosin, piperazine), guanidine derivatives (betanidine, guanethidine, guanidine Health, isoquinidine, guanaclofen, guanazodine, guanidinoline), arteriolar smooth muscle agents, thiazine-derived chlorotoxazine, hydrazinophthalazine derivatives (hydrazinophenidazine, hydralazine) (Entrazine, cardazine), pyrimidine-derived minoxidil, nitroferricyanide-derived pentacyannitrosammonium ferric acid, guanidine-derived pinacidil, and philophyllum alkaloids Reed, tyrosine hydroxylase inhibitory methyltyrosine, MAO inhibitor propynemethylbenzylamine, serotonin antagonist ketoserin, or other antihypertensive drugs (bosentan, ambrisentan, sitax Shengtan, Massitidine, Leosizone) and combinations of these agents and diuretics.
Antispasmodics or antispasmodics include, but are not limited to, peripheral muscle relaxants, arrow poison alkaloids, choline derivatives, and other quaternary ammonium muscle relaxants (Pankan Langning, Galatium Ammonium, Vicroine, Atracu Ammonium, hexafluorourea, pipecuronium bromide, doxycycline chloride, fazabromide, rocuronium, micuronium, cis-atracurium, botulinum toxin), central nervous system muscles Relaxants, carbamates (amphetamine, isopropylmethizuron, mesopotamone, methamphetamine, non-pamine esters), oxazole derivatives, thiazine derivatives, and triazine derivatives (Clomezadone, chloroxazolone), ethers related to antihistamines (ofinatlin, guaiacol) and other histaminergic drugs (such as histamine H 3 Receptor antagonists / inverse agonists, such as saphene, thiomipramine, pethsente, beclomethasone propionate, ABT-239, connexin, A-349,821, betahistine), other central acting agents (Chloroaniline butyric acid, abaclofen, tizanidine, diphenidalyl alcohol, topiradone, thiocolchicoside, cresol glyceryl ether, tetrahydroazepam, amine phenylheptene, benzylamine ), Directly acting muscle relaxant dantrolene and its derivatives, by increasing GABA A Inhibition of the media or reduction of Na + Transporting compounds (diphenytoin, carbazapine, lamotrigine, VPA), γ-aminobutyric acid derivatives (aminohexenoic acid, gabapentin), other GABA agents (such as GABA B PAMs, such as ADX71441), tertiary amine-containing esters (oxybenzamine, camiprofen, mebeverine, trimebutine, rosivirin, dicyclovirin, dihexaverine, diphenvirin Lin, diphenperate), quaternary ammonium compounds (benzyl ammonium, glucopyrrolium, orphenammonium bromide, pentammonium, prilamicillin, oltibromonium, ethamidine, tridemonium, isopropylamine , Hexammonium, poliridine, mepenzolate, benphenate, phenphenate, diphenidate, emetonium iodide, temoperidomide, pirfenammonium bromide, and thipemide Ammonium bromide and tolperiamide), tertiary amines (Astra 1397, nifitazamine, phenamidine ceramide), papaverine, and their derivatives (drotaverine, mosa Verin, ethyl papaverine), serotonin receptor acting agents (alosetron, tegaserod, cilansetron, pukabili), other agents for functional gastrointestinal diseases (diphenylpropidin , Desopramine, chlorpheniramine, pirmonium, fenavirin, edampamin, prozazole, alverin, triethylbutanone, isomethine, carlovirin, resorcinol, Polysiloxane, trimethyldiphenylpropane Amine), succinimine derivatives (ethosuccin, phenylsuccinamide, mesuccinine) or belladonna alkaloids and their derivatives (atropine, celastamine, butyl scopolamine, methyl atropine, methyl atropine, formazan Scopolamine, fentonium, deltamethrin).
Antiepileptic drugs include, but are not limited to, barbiturates and their derivatives (methylphenobarbital, phenobarbital, ethylpyrimidinedione, barbizarone, methapital), hydantoin Derivatives (ethylphenytoin, diphenytoin, amine (diphenytoin) valeric acid, mephenytoin, fosphenytoin), oxazolidine derivatives (methyl ethyl ketone, paradione, trimethylcyclodione, Ethadion), succinimine derivatives (ethosuccin, phenylsuccinidine, mesuccinine), benzodiazepine derivatives clonazepam, carboxamide derivatives (carbazpine , Oxcarbazepine, rufibramide), fatty acid derivatives (valproic acid, valprofen, aminobutyric acid, aminohexic acid, flusalamine, tiagabine) and other antiepileptic drugs (Shu Thiazine, phenethylcarbamuron, lamotrigine, non-amine esters, topiramate, gabapentin, phentermine, levetiracetam, zonisamide, pregabalin, stifol, lakoxamine , Carbamine, retigabine, bovaracetam, chlorpromazine).
Other agents include but are not limited to pharmaceuticals (probiotics, digestive aids / digestants, herbal extracts), vitamins (both water-soluble and fat-soluble, such as, but not limited to, vitamins A, D3, E, K, B1, B5 , B6, B12, C or their derivatives) and nutritional supplements (coenzymes such as Q10; flavones such as resveratrol; lecithin; unsaturated fatty acids including fatty acids omega-3 and omega-6) .
The compounds of the present invention can also be used with phosphodiesterase 5 isoenzyme inhibitors (PDE5), nitric oxide donors, cyclooxygenase inhibitors, other V1a receptor antagonists (such as balofeta) or L- Arginine is used in combination to treat and / or prevent diseases or conditions related to the function of the Via receptor.
The combination composition may contain the compound of the present invention and another active substance in a single dosage form or separately. The combined composition can be administered simultaneously, separately, or sequentially.
Suitable dosage forms include oral, rectal, mucosal, transdermal or enteral administration; parenteral administration includes intramuscular, subcutaneous, intravenous and intramedullary injections as well as intra-articular, intrathecal, direct intraventricular, intraperitoneal, nasal Intra- or intraocular injections and eye drops.
Alternatively, the compound may be administered locally and non-systemically, such as by injecting the compound directly into the kidney or heart by a release modifier that is usually modified. In addition, the drug can be administered via a target carrier system, such as a tissue-specific antibody-encapsulated liposome. The liposome selectively transfers the active substance to a target organ that absorbs the active substance.
The pharmaceutical composition can be administered in a number of different ways and pharmaceutical dosage forms. The compounds of the present invention may be administered in single or multiple doses alone or in combination with pharmaceutically acceptable excipients. The dosage required to achieve proper therapeutic efficacy can vary widely, and considering the stage of the disease, the state and weight of the patient to be treated, the selectivity of the active substance, the dosage formulation form and the number of daily treatments, the dosage must be suitable for the needs of the individual .
For simple administration, it is preferred that the medicinal composition is composed of multiple dosage units containing multiple, 1/2, 1/3 or 1/4 dose. The dosage unit is, for example, a tablet, which may have 1/2 or 1/4 grooves to provide 1/2 or 1/4 divided tablets to determine the required drug dose.
The pharmaceutical composition containing the active substance of the present invention usually contains 0.01 to 500 mg of active substance / dosage unit. Of course, it is also possible that the amount of active substance of each modulator exceeds the above-mentioned upper or lower limit.
A further preferred group of the compound of the general formula (I) is the following ring A, ring B, X, Y, Z, R 1 -R 17 , Cy 1 -Cy 3 , N, p, q, and r. Ring A, Ring B, X, Y, Z, R as defined below 1 -R 17 , Cy 1 -Cy 3 Any combination of the preferred embodiment, the better embodiment, or the best embodiment of, n, p, q, and r is also a preferred, better, or optimal group of a compound of general formula (I).
In certain embodiments of the present invention, the ring A in the compound of the general formula (I) is a 4- to 6-membered saturated carbocyclic ring.
In some preferred embodiments of the present invention, the ring A in the compound of the general formula (I) is cyclobutyl or cyclohexyl.
In some more preferred embodiments of the present invention, the ring A in the compound of the general formula (I) is cyclohexyl.
In certain embodiments of the present invention, in the compound of the general formula (I), ring A is a 4- to 7-membered saturated heterocyclic group containing 1 or 2 N, wherein ring A is connected to Y via a ring nitrogen.
In certain embodiments of the present invention, in the compound of the general formula (I), ring A is a 4 to 7-membered saturated heterocyclic group containing 1 or 2 N, wherein ring A is connected to the 5,6- Dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzotriazine core is attached to the triazole ring.
In certain embodiments of the present invention, the ring A of the compound of the general formula (I) is tetrahydroacryl, 1,3-bis (tetrahydroacryl), pyrrolidinyl, pyrazoridinyl, imidazole Pyridyl, piperidinyl, piperazinyl, azacycloheptyl, 1,3- or 1,4-diazacycloheptyl, wherein ring A is connected to Y via a ring nitrogen.
In certain embodiments of the present invention, the ring A of the compound of the general formula (I) is tetrahydroacryl, 1,3-bis (tetrahydroacryl), pyrrolidinyl, pyrazoridinyl, imidazole Pyridyl, piperidinyl, piperazinyl, azacycloheptyl, 1,3- or 1,4-diazacycloheptyl, wherein ring A is linked to the 5,6-dihydro via a ring nitrogen -4H- [1,2,4] triazolo [4,3-a] [1] benzotriazine nucleus is attached to the triazole ring.
In certain preferred embodiments of the present invention, the ring A of the compound of the general formula (I) is tetrahydroazepine-1,3-diyl, piperidine-1,4-diyl, or piperazine-1, 4-diyl, where ring A is via ring nitrogen with Y or the 5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoacene core The triazole ring is attached.
In certain embodiments of the present invention, the ring B in the compound of the general formula (I) is an optionally substituted heteroaryl group.
In certain preferred embodiments of the present invention, the ring B in the compound of the general formula (I) is optionally substituted and contains 5 or 6 members of 1 N, 2 N or 1 N and 1 O. Single heteroaryl.
In some preferred embodiments of the present invention, the ring B in the compound of the general formula (I) is pyridin-2-yl, 3-chloro-pyridin-2-yl, 3-methyl-pyridin-2-yl, Pyridine-3-yl, pyrimidin-2-yl or 5-methyl-isoxazol-3-yl.
In some preferred embodiments of the present invention, the ring B in the compound of the general formula (I) is pyridin-2-yl.
In certain embodiments of the present invention, the ring B in the compound of the general formula (I) is an optionally substituted aryl group.
In certain preferred embodiments of the present invention, the ring B in the compound of the general formula (I) is an optionally substituted phenyl group.
In certain embodiments of the present invention, the ring B in the compound of the general formula (I) is an optionally substituted heterocyclic group.
In certain preferred embodiments of the present invention, the ring B in the compound of the general formula (I) is optionally substituted and contains a 4- to 7-membered heterocyclic group containing 1 N or 1 N and 1 O.
In certain more preferred embodiments of the present invention, the ring B of the compound of the general formula (I) is tetrahydroazino-1-yl, pyrrolidin-1-yl, piperidinyl, piperazinyl, or morpholine- 4-based.
In certain embodiments of the present invention, if ring B is present, Y is -O- in the compound of the general formula (I).
In certain embodiments of the present invention, if ring B is present, Y in the compound of general formula (I) is -C (O)-.
In certain embodiments of the present invention, if ring B is present, Y is -CH in the compound of formula (I). 2 -.
In certain embodiments of the present invention, if ring B is present, Y in the compound of general formula (I) is -NH-.
In certain embodiments of the present invention, if ring B is present, Y is a single bond in the compound of general formula (I).
In certain embodiments of the present invention, if ring B does not exist, Y is -N (C 1-4 alkyl) 2 C, optionally substituted by halogen 1-4 Alkyl, C 1-4 Alkoxy, C (O) OC 1-4 Alkyl or halogen.
In certain embodiments of the present invention, if ring B is absent, Y in the compound of the general formula (I) may be optionally substituted with C. 1-4 Alkyl or C 1-4 Alkoxy.
In certain preferred embodiments of the present invention, if ring B does not exist, Y is C in the compound of formula (I) 1-3 alkyl.
In some preferred embodiments of the present invention, if ring B is absent, Y in the compound of general formula (I) is methyl or ethyl.
In certain preferred embodiments of the present invention, if ring B does not exist, Y is C in the compound of formula (I) 1-3 Alkoxy.
In some preferred embodiments of the present invention, if ring B is absent, Y in the compound of general formula (I) is methoxy or ethoxy.
In certain preferred embodiments of the present invention, if ring B does not exist, Y is CF in the compound of general formula (I) 3 .
In certain embodiments of the present invention, if ring B does not exist, Y in the compound of general formula (I) means selected from -N (C 1-4 alkyl) 2 , C (O) OC 1-4 Alkyl, optionally halogen-substituted C 1-4 Alkyl, C 1-4 Either alkoxy or halogen.
In certain preferred embodiments of the present invention, if ring B is absent, Y in the compound of general formula (I) means selected from dimethylamine or CF. 3 One of them.
In certain embodiments of the present invention, if ring B is absent, Y in the compound of the general formula (I) refers to a group selected from C optionally substituted with halogen. 1-4 Alkyl, C 1-4 Both alkoxy or halogen.
In certain preferred embodiments of the present invention, if ring B does not exist, Y in the compound of the general formula (I) means selected from C 1-3 Alkyl, C 1-3 Both alkoxy or fluorine.
In some preferred embodiments of the present invention, if ring B is absent, Y in the compound of the general formula (I) refers to two selected from methyl, ethyl, methoxy, ethoxy, or fluorine. By.
In some very preferred embodiments of the present invention, if ring B is absent, Y in the compound of general formula (I) refers to both selected from methyl or methoxy.
In certain preferred embodiments of the present invention, in the compound of the general formula (I), ring A is cyclobutyl or cyclohexyl, and Y is selected from -N (C 1-4 alkyl) 2 CF 3 Or halogen, and ring B is absent.
In certain preferred embodiments of the present invention, in the compound of the general formula (I), ring A is cyclobutyl or cyclohexyl, and Y is selected from C which is optionally substituted with halogen. 1-4 Alkyl, C 1-4 Either alkoxy or halogen, and ring B is absent.
In certain preferred embodiments of the present invention, in the compound of the general formula (I), ring A is cyclobutyl or cyclohexyl, Y is -O- or a single bond and ring B is phenyl, 2-methyl -Phenyl, tetrahydroacryl-1-yl, pyrrolidin-1-yl, morpholin-4-yl, pyridin-2-yl, 3-chloro-pyridin-2-yl, 3-methyl-pyridine 2-yl, pyridin-3-yl, pyrimidin-2-yl or 5-methyl-isoxazol-3-yl.
In some preferred embodiments of the present invention, in the compound of the general formula (I), ring A is cyclohexyl, Y is -O- and ring B is pyridin-2-yl, pyridin-3-yl, 3 -Chloro-pyridin-2-yl, 3-methyl-pyridin-2-yl, pyrimidin-2-yl, or 5-methyl-isoxazol-3-yl.
In some preferred embodiments of the present invention, in the compound of the general formula (I), ring A is cyclohexyl, Y is -O-, and ring B is pyridin-2-yl.
In certain preferred embodiments of the present invention, in the compound of the general formula (I), ring A is a cyclohexyl group, Y is a single bond, and ring B is a morpholin-4-yl group.
In certain embodiments of the present invention, in the compound of general formula (I), ring A is cyclobutyl or cyclohexyl, Y is -C (O)-, and ring B is tetrahydroacryl-1- Group, pyrrolidin-1-yl or morpholin-4-yl.
In certain embodiments of the present invention, in the compound of the general formula (I), ring A is cyclobutyl or cyclohexyl, and Y is -CH. 2 -, And Ring B is optionally substituted aryl, heterocyclyl or heteroaryl.
In certain embodiments of the present invention, in the compound of the general formula (I), ring A is cyclobutyl or cyclohexyl, and Y is -CH. 2 -, And Ring B is optionally substituted 4 to 7 membered heterocyclic group containing 1 N or 1 N and 1 O.
In certain preferred embodiments of the present invention, in the compound of the general formula (I), ring A is tetrahydroazepine-1,3-diyl, piperidine-1,4-diyl, or piperazine- 1,4-diyl, wherein ring A is via ring nitrogen with Y or the 5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazine The triazole ring of the fluorene core is connected, Y is -O- or a single bond, and ring B is phenyl, 2-methyl-phenyl, tetrahydroazepine-1-yl, pyrrolidin-1-yl, morpholine 4-yl, pyridin-2-yl, 3-chloro-pyridin-2-yl, 3-methyl-pyridin-2-yl, pyridin-3-yl, pyrimidin-2-yl, or 5-methyl-iso Oxazol-3-yl.
In some more preferred embodiments of the present invention, in the compound of the general formula (I), ring A is tetrahydroazine-1,3-diyl, wherein ring A is via ring nitrogen and Y or the 5, 6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzotriazine core triazole ring connection, Y is a single bond, and ring B is pyridine-2 -Yl, pyrimidin-2-yl, piperidinyl or optionally substituted phenyl.
In some embodiments of the present invention, the BYA-bond in the compound of the general formula (I) represents 3 H -Spiro [2-benzofuran-1,4'-piperidine-1'-yl], via C at the 3 position 1-4 Alkyl-substituted 1-oxa-3-azaspiro [4.5] dec-2-one-8-yl or via C at the 2 position 1-4 Alkyl, aryl or heteroaryl substituted 2-azaspiro [4.5] dec-1-one-8-yl.
In some embodiments of the present invention, the BYA-bond in the compound of the general formula (I) represents 3 H -Spiro [2-benzofuran-1,4'-piperidine-1'-yl], (5S, 8S) -3-methyl-1-oxa-3-azaspiro [4.5] dec-2 -Keto-8-yl, (5R, 8R) -3-methyl-1-oxa-3-azaspiro [4.5] dec-2-one-8-yl, (5 R ,8 R ) -2- (prop-2-yl) -2-azaspiro [4.5] dec-1-one or (5 S ,8 S ) -2- (prop-2-yl) -2-azaspiro [4.5] dec-1-one.
In certain embodiments of the present invention, R in the compound of general formula (I) 1 Department of hydrogen.
In certain embodiments of the present invention, R in the compound of general formula (I) 1 Department of halogen.
In certain preferred embodiments of the present invention, R in the compound of the general formula (I) 1 Department of chlorine, bromine or fluorine.
In some preferred embodiments of the present invention, R in the compound of the general formula (I) 1 Department of chlorine.
In certain embodiments of the present invention, R in the compound of general formula (I) 1 Department C 1-4 alkyl.
In certain preferred embodiments of the present invention, R in the compound of the general formula (I) 1 Department of methyl.
In certain embodiments of the present invention, R in the compound of general formula (I) 1 Department C 1-4 Alkoxy.
In certain preferred embodiments of the present invention, R in the compound of the general formula (I) 1 Department of methoxy.
In certain embodiments of the present invention, R in the compound of general formula (I) 1 Department of CF 3 .
In certain embodiments of the present invention, R in the compound of general formula (I) 1 Department of CN.
In certain embodiments of the present invention, in the compound of the general formula (I), R 2 Hydrogen or C 1-4 Alkyl, R 3 Department (CH 2 ) n R 4 , C (O) R 5 Or optionally via R 6 Replaced by C 1-4 alkyl.
In certain embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department (CH 2 ) n R 4 , C (O) R 5 Or optionally via R 6 Replaced by C 1-4 alkyl.
In certain embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department (CH 2 ) n R 4 Where R 4 Is CN, azide, or optionally substituted 5-membered monoheteroaryl containing 2 N and 1 O or 4 N, and n is 0 or 1.
In certain embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department (CH 2 ) n R 4 Where R 4 Is CN, and n is 0 or 1.
In certain embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department (CH 2 ) n R 4 Where R 4 Is tetrazolyl or 3-methyl-1,2,4-oxadiazol-5-yl, and n is 0 or 1.
In certain embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 C (O) R 5 Where R 5 Department C 1-4 alkyl.
In certain preferred embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 C (O) R 5 Where R 5 Methyl, ethyl, isopropyl or tertiary butyl.
In some preferred embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 C (O) R 5 Where R 5 Department of methyl.
In certain embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 C (O) R 5 Where R 5 Department C 1-4 Alkoxy.
In certain preferred embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 C (O) R 5 Where R 5 It is methoxy, ethoxy, propoxy or tertiary butoxy.
In some preferred embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 C (O) R 5 Where R 5 Department of methoxy.
In certain embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 C (O) R 5 Where R 5 Department of OH.
In certain embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 C (O) R 5 Where R 5 Department of NR 7 R 8 Where R 7 And R 8 Independently hydrogen, C 1-4 Alkyl or Cy 2 .
In certain preferred embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 C (O) R 5 Where R 5 Department of NR 7 R 8 Where R 7 And R 8 For hydrogen.
In certain preferred embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 C (O) R 5 Where R 5 Department of NR 7 R 8 Where R 7 Hydrogen and R 8 Methyl, ethyl, isopropyl or tertiary butyl.
In certain preferred embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 C (O) R 5 Where R 5 Department of NR 7 R 8 Where R 7 And R 8 Independently methyl, ethyl, isopropyl or tertiary butyl.
In some preferred embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 C (O) R 5 Where R 5 Department of NR 7 R 8 Where R 7 And R 8 Is methyl.
In certain preferred embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 C (O) R 5 Where R 5 Department of NR 7 R 8 Where R 7 Hydrogen and R 8 Is optionally substituted aryl or heteroaryl.
In certain preferred embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 C (O) R 5 Where R 5 Department of NR 7 R 8 Where R 7 And R 8 Together with the N to which they are attached forms an optionally substituted heterocyclic ring.
In some preferred embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 C (O) R 5 Where R 5 Department of NR 7 R 8 Where R 7 And R 8 Together with their linked N, a 4 to 7 membered saturated heterocyclic ring containing 1 N, 2 N, 1 N and 1 O or 1 N and 1 S is optionally substituted.
In certain embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Optionally via R 6 Replaced by C 1-4 Alkyl, where R 6 Department of OR 9 , NR 10 R 11 , Pendant oxygen, -O (CH 2 ) m O- or one or more halogens.
In certain preferred embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Methyl, ethyl or isopropyl.
In some preferred embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Is isopropyl.
In certain preferred embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department of Economics R 6 Single or multiple branched C 1-4 Alkyl, where R 6 Department of OR 9 , NR 10 R 11 Or one or more halogens.
In certain preferred embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department of Economics R 6 Single or multiple branched C 1-4 Alkyl, where R 6 One or more halogens.
In certain embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department-CH 2 Halogen, -CH (halogen) CH 3 Or -C (halogen) 2 CH 3 .
In certain preferred embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department-CH 2 halogen.
In some preferred embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department-CH 2 F.
In certain embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department of Economics R 6 Replaced by C 1-4 Alkyl, where R 6 System side oxygen or -O (CH 2 ) 2 O-.
In certain preferred embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department of Economics R 6 Single or multiple branched C 1-4 Alkyl, where R 6 Department of OR 9 Or NR 10 R 11 .
In certain preferred embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department-CH 2 R 6 Or -CH (R 6 ) CH 3 Where R 6 Department of OR 9 Or NR 10 R 11 .
In some preferred embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department-CH 2 R 6 Where R 6 Department of OR 9 Or NR 10 R 11 .
In certain embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department of Economics R 6 Replaced by C 1-4 Alkyl, where R 6 Department of OR 9 .
In certain preferred embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department-CH 2 R 6 Or -CH (R 6 ) CH 3 , R 6 Department of OR 9 Where R 9 Department of hydrogen.
In certain preferred embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department-CH 2 R 6 Or -CH (R 6 ) CH 3 , R 6 Department of OR 9 Where R 9 Optionally via NH 2 Or optionally substituted C 1-4 alkyl.
In certain preferred embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department-CH 2 R 6 Or -CH (R 6 ) CH 3 , R 6 Department of OR 9 Where R 9 Department of NH 2 Substituted ethyl or isopropyl.
In certain embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department-CH 2 R 6 Or -CH (R 6 ) CH 3 , R 6 Department of OR 9 Where R 9 Is ethyl or isopropyl.
In certain embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department-CH 2 R 6 Or -CH (R 6 ) CH 3 , R 6 Department of OR 9 Where R 9 Department of Si (CH 3 ) 3 .
In certain embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department-CH 2 R 6 Or -CH (R 6 ) CH 3 , R 6 Department of OR 9 Where R 9 C (O) R 12 , R 12 Department C 1-5 alkyl.
In certain preferred embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department-CH 2 R 6 Or -CH (R 6 ) CH 3 , R 6 Department of OR 9 Where R 9 C (O) R 12 , R 12 Department of methyl.
In certain embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department-CH 2 R 6 Or -CH (R 6 ) CH 3 , R 6 Department of OR 9 Where R 9 C (O) R 12 , R 12 Department of NR 14 R 15 .
In certain preferred embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department-CH 2 R 6 Or -CH (R 6 ) CH 3 , R 6 Department of OR 9 Where R 9 C (O) R 12 , R 12 Department of NR 14 R 15 Where R 14 And R 15 Department of hydrogen.
In certain preferred embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department-CH 2 R 6 Or -CH (R 6 ) CH 3 , R 6 Department of OR 9 Where R 9 C (O) R 12 , R 12 Department of NR 14 R 15 Where R 14 And R 15 Independently hydrogen or optionally substituted aryl.
In certain preferred embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department-CH 2 R 6 , R 6 Department of OR 9 Where R 9 C (O) R 12 , R 12 Department of NR 14 R 15 Where R 14 System hydrogen, R 15 Is 4-fluoro-phenyl.
In certain embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department-CH 2 R 6 Or -CH (R 6 ) CH 3 , R 6 Department of OR 9 Where R 9 C (O) R 12 , R 12 Department of NR 14 R 15 Where R 14 And R 15 Together with their linked N, a 4 to 7 membered saturated heterocyclic ring containing 1 N, 2 N, 1 N and 1 O or 1 N and 1 S is optionally substituted.
In certain preferred embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department-CH 2 R 6 , R 6 Department of OR 9 Where R 9 C (O) R 12 , R 12 Department of NR 14 R 15 Where R 14 And R 15 Together with their attached N, they form morpholines.
In certain embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department of Economics R 6 Replaced by C 1-4 Alkyl, where R 6 Department of NR 10 R 11 .
In certain preferred embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department-CH 2 R 6 Or -CH (R 6 ) CH 3 , R 6 Department of NR 10 R 11 Where R 10 And R 11 Department of hydrogen.
In certain embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department-CH 2 R 6 Or -CH (R 6 ) CH 3 , R 6 Department of NR 10 R 11 Where R 10 And R 11 Independently hydrogen or optionally via C 1-4 Alkoxy-substituted C 1-4 alkyl.
In certain preferred embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department-CH 2 R 6 Or -CH (R 6 ) CH 3 , R 6 Department of NR 10 R 11 Where R 10 And R 11 Independently hydrogen, methyl, ethyl, isopropyl, secondary butyl or tertiary butyl.
In certain preferred embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department-CH 2 R 6 Or -CH (R 6 ) CH 3 , R 6 Department of NR 10 R 11 Where R 10 And R 11 Independently hydrogen or methyl, ethyl or isopropyl substituted with methoxy or ethoxy.
In certain embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department-CH 2 R 6 Or -CH (R 6 ) CH 3 , R 6 Department of NR 10 R 11 Where R 10 And R 11 Independently hydrogen or Cy 3 .
In certain preferred embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department-CH 2 R 6 Or -CH (R 6 ) CH 3 , R 6 Department of NR 10 R 11 Where R 10 System hydrogen, R 11 4 to 7-membered saturated heterocyclic group containing 1 O or C substituted with a twin halogen 4-6 Cycloalkyl.
In certain embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department of Economics R 6 Replaced by C 1-4 Alkyl, where R 6 Department of NR 10 R 11 Where R 10 And R 11 Independently hydrogen or C (O) R 13 .
In certain preferred embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department-CH 2 R 6 Or -CH (R 6 ) CH 3 , R 6 Department of NR 10 R 11 Where R 10 System hydrogen, R 11 C (O) R 13 , R 13 Department C 1-4 alkyl.
In some preferred embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department-CH 2 R 6 , R 6 Department of NR 10 R 11 Where R 10 System hydrogen, R 11 C (O) R 13 , R 13 Department of methyl.
In certain embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department of Economics R 6 Replaced by C 1-4 Alkyl, where R 6 Department of NR 10 R 11 Where R 10 And R 11 Independently hydrogen or C (O) R 13 Where R 13 Department Cy 1 .
In certain embodiments of the present invention, in the compound of the general formula (I), R 2 System hydrogen, R 3 Department of Economics R 6 Replaced by C 1-4 Alkyl, where R 6 Department of NR 10 R 11 Where R 10 And R 11 Independently hydrogen or C (O) R 13 Where R 13 Department of NR 16 R 17 .
In certain embodiments of the present invention, in the compound of the general formula (I), R 2 Department C 1-4 Alkyl, R 3 Department (CH 2 ) n R 4 , C (O) R 5 Or optionally via R 6 Replaced by C 1-4 alkyl.
In certain embodiments of the present invention, in the compound of the general formula (I), R 2 Department C 1-4 Alkyl, R 3 Department (CH 2 ) n R 4 .
In certain embodiments of the present invention, in the compound of the general formula (I), R 2 Department C 1-4 Alkyl, R 3 C (O) R 5 .
In certain embodiments of the present invention, in the compound of the general formula (I), R 2 And R 3 Department C 1-4 alkyl.
In certain preferred embodiments of the present invention, in the compound of the general formula (I), R 2 And R 3 Department C 1-3 alkyl.
In some preferred embodiments of the present invention, in the compound of the general formula (I), R 2 And R 3 Independently methyl, ethyl or isopropyl.
In some very preferred embodiments of the present invention, in the compound of formula (I), R 2 And R 3 Department of methyl.
In certain embodiments of the present invention, in the compound of the general formula (I), R 2 Department C 1-4 Alkyl, R 3 Department of Economics R 6 Replaced by C 1-4 Alkyl, where R 6 Department of OR 9 .
In certain embodiments of the present invention, in the compound of the general formula (I), R 2 Department C 1-4 Alkyl, R 3 Department of Economics R 6 Replaced by C 1-4 Alkyl, where R 6 Department of OR 9 Where R 9 Department C 1-4 alkyl.
In certain preferred embodiments of the present invention, in the compound of the general formula (I), R 2 Methyl or ethyl, R 3 Department-CH 2 R 6 Or -CH (R 6 ) CH 3 , R 6 Department of OR 9 Where R 9 Department of hydrogen.
In certain embodiments of the present invention, in the compound of the general formula (I), R 2 Department C 1-4 Alkyl, R 3 Optionally via R 6 Replaced by C 1-4 Alkyl, where R 6 Department of NR 10 R 11 , Pendant oxygen, -O (CH 2 ) m O- or one or more halogens.
In certain embodiments of the present invention, in the compound of the general formula (I), R 2 And R 3 Combined Representative-(CH 2 ) p -O- (CH 2 ) q -, Where p is 1, 2 or 3 and q is 1, 2 or 3.
In certain preferred embodiments of the present invention, in the compound of the general formula (I), R 2 And R 3 Combined Representative-(CH 2 ) p -O- (CH 2 ) q -, Where the sum of p and q is 3.
In certain preferred embodiments of the present invention, in the compound of the general formula (I), R 2 And R 3 Combined Representative-(CH 2 ) p -O- (CH 2 ) q -, Where the sum of p and q is 4.
In some preferred embodiments of the present invention, in the compound of the general formula (I), R 2 And R 3 Combined Representative-(CH 2 ) p -O- (CH 2 ) q -, Where p is 1 and q is 2.
In some preferred embodiments of the present invention, in the compound of the general formula (I), R 2 And R 3 Combined Representative-(CH 2 ) p -O- (CH 2 ) q -, Where p is 1 and q is 3.
In some preferred embodiments of the present invention, in the compound of the general formula (I), R 2 And R 3 Combined Representative-(CH 2 ) p -O- (CH 2 ) q -, Where p is 2 and q is 2.
In certain embodiments of the present invention, in the compound of the general formula (I), R 2 And R 3 Combined Representative-(CH 2 ) r -, Where r is 4, 5, or 6.
In certain preferred embodiments of the present invention, in the compound of the general formula (I), R 2 And R 3 Combined Representative-(CH 2 ) r -, Where r 系 4.
Although the invention has been described in terms of certain embodiments, certain preferred embodiments, certain better embodiments, or certain best practices, it is not intended to limit the scope of the invention to the particular forms disclosed And, on the contrary, the intention is to cover alternatives, modifications, and equivalents as included in the spirit and scope of the invention as defined by the description of the invention. Examples of alternative claimed ranges of compounds of the invention may include:
(1) A compound of general formula (I) as described above or in any other embodiment.
(2) The compound as described in (1) or any other embodiment, wherein R 1 Based on hydrogen, fluorine, chlorine, bromine, methyl, methoxy, CF 3 Or CN.
(3) The compound as described in any one of (1) to (2) or any other embodiment, wherein
Ring A is a 3 to 6-membered saturated carbocyclic ring or a 4 to 7-membered saturated heterocyclic ring containing 1 or 2 N;
Ring B is optionally substituted 5 or 6 membered monoheteroaryl, 6 to 10 membered aromatic carbocyclic rings or 4 to 7 members saturated with 1, 2 or 3 heteroatoms selected from O, S or N Monocyclic, bicyclic, fused and / or bridged heterocyclic rings;
Or BYA-Combined Representative 3 H -Spiro [2-benzofuran-1,4'-piperidine-1'-yl]; or

X is isopropyl;
Z is methyl.
(4) The compound as described in any one of (1) to (3) or any other embodiment, wherein ring B is optionally substituted 6-membered monoheteroaryl, phenyl, or contains 1 or 2 4- to 6-membered saturated monocyclic heterocycles of heteroatoms selected from O, S or N.
(5) The compound as described in any one of (1) to (4) or any other embodiment, wherein if ring B exists, Y is -O-, -C (O)-, -CH 2 -, -NH- or single bond.
(6) The compound as described in any one of (1) to (5) or any other embodiment, wherein ring A is a 4- to 6-membered saturated carbocyclic group or via ring nitrogen with Y or the 5,6- Dihydro-4 H -[1,2,4] triazolo [4,3-a] [1] benzotriazine core with a triazole ring containing 1 to 2 4 to 7 membered saturated heterocyclic rings.
(7) The compound as described in any one of (1) to (6) or any other embodiment, wherein ring A is cyclohexyl, Y is -O-, ring B is pyridin-2-yl, and R 1 Department of chlorine.
(8) The compound as described in any one of (1) to (6) or any other embodiment, wherein ring A is tetrahydroacridine, piperidine or piperazine, Y is -O- or a single bond, Ring B is pyridine, pyrimidine or piperidine, and R 1 Department of chlorine.
(9) The compound as described in any one of (1) to (3) or any other embodiment, wherein Y is -N (C 1-4 alkyl) 2 , C (O) OC 1-4 Alkyl, optionally halogen-substituted C 1-4 Alkyl, C 1-4 Alkoxy or halogen, and ring B is absent.
(10) The compound as described in (9) or any other embodiment, wherein ring A is a 4- to 6-membered saturated carbocyclic group.
(11) The compound as described in (10) or any other embodiment, wherein Y is selected from -N (C 1-4 alkyl) 2 , C (O) OC 1-4 Alkyl, optionally halogen-substituted C 1-4 Alkyl, C 1-4 One of alkoxy and halogen.
(12) A compound as described in (10) or any other embodiment, wherein Y is selected from C optionally substituted with halogen 1-4 Alkyl, C 1-4 Both alkoxy and halogen.
(13) The compound as described in any one of (1) to (12) or any other embodiment, wherein R 2 Hydrogen or C 1-4 Alkyl and R 3 Department (CH 2 ) n R 4 .
(14) The compound as described in (13) or any other embodiment, wherein R 2 System hydrogen, R 3 Department (CH 2 ) n R 4 , R 4 Is CN, azide, or optionally C 1-4 Alkyl substituted 5-membered monoheteroaryl containing 2 N and 1 O or 4 N.
(15) The compound as described in any one of (1) to (12) or any other embodiment, wherein R 2 Hydrogen or C 1-4 Alkyl, and R 3 C (O) R 5 .
(16) The compound as described in (15) or any other embodiment, wherein R 2 Department of hydrogen.
(17) The compound as described in (16) or any other embodiment, wherein R 5 Methyl, methoxy, OH or NR 7 R 8 Where R 7 And R 8 Independently hydrogen, methyl, ethyl or isopropyl.
(18) The compound as described in any one of (1) to (12) or any other embodiment, wherein R 2 Hydrogen or C 1-4 Alkyl and R 3 Optionally via R 6 Replaced by C 1-4 alkyl.
(19) The compound as described in (18) or any other embodiment, wherein R 2 Hydrogen or C 1-4 Alkyl, and R 3 Department C 1-4 alkyl.
(20) The compound as described in (19) or any other embodiment, wherein R 2 Hydrogen or C 1-3 Alkyl, and R 3 Department C 1-3 alkyl.
(21) The compound as described in (18) or any other embodiment, wherein R 2 Hydrogen or C 1-4 Alkyl, and R 3 Department of Economics R 6 Single or multiple branched C 1-4 alkyl.
(22) A compound as described in (21) or any other embodiment, wherein R 3 Department-CH 2 R 6 Or -CH (R 6 ) CH 3 .
(23) The compound as described in any one of (21) to (22) or any other embodiment, wherein R 2 Is hydrogen, and R 6 Department of OR 9 .
(24) A compound as described in (23) or any other embodiment, wherein R 9 Hydrogen, optionally C substituted with phenyl 1-4 Alkyl or C (O) R 12 Where R 12 Department of methyl.
(25) The compound as described in any one of (21) to (22) or any other embodiment, wherein R 2 Department C 1-4 Alkyl, and R 6 Department of OR 9 .
(26) The compound as described in (25) or any other embodiment, wherein R 2 Methyl or ethyl, and R 9 Department of hydrogen.
(27) The compound as described in any one of (21) to (22) or any other embodiment, wherein R 2 Is hydrogen, and R 6 Department of NR 10 R 11 .
(28) The compound as described in (27) or any other embodiment, wherein R 10 And R 11 Independently hydrogen, optionally via C 1-4 Alkoxy-substituted C 1-4 Alkyl, Cy 3 Or C (O) R 13 , Or R 16 And R 17 Together with the N to which they are attached forms an optionally substituted heterocyclic ring.
(29) The compound as described in any one of (21) to (22) or any other embodiment, wherein R 2 Department C 1-4 Alkyl, and R 6 Department of NR 10 R 11 .
(30) The compound as described in any one of (21) to (22) or any other embodiment, wherein R 2 Is hydrogen, and R 6 One or more halogens.
(31) The compound as described in (30) or any other embodiment, wherein the halogen is fluorine.
(32) The compound as described in any one of (21) to (22) or any other embodiment, wherein R 2 Department C 1-4 Alkyl, and R 6 One or more halogens.
(33) A compound as described in (21) or any other embodiment, wherein R 6 System side oxygen or -O (CH 2 ) 2 O-.
(34) The compound as described in any one of (13) to (33) or any other embodiment, wherein the 5,6-dihydro-4H- [1,2,4] triazolo [4, 3-a] [1] Absolute configuration of the carbon at position 5 of the benzoacridine core (R).
(35) The compound as described in any one of (13) to (33) or any other embodiment, wherein the 5,6-dihydro-4H- [1,2,4] triazolo [4, 3-a] [1] Absolute configuration of the carbon at position 5 of the benzoacridine core (S).
(36) The compound as described in any one of (1) to (12) or any other embodiment, wherein R 2 And R 3 Combined Representative-(CH 2 ) p -O- (CH 2 ) q -, Where p is 1, 2 or 3 and q is 1, 2 or 3.
(37) A compound as described in (36) or any other embodiment, wherein the sum of p and q is 3 or 4.
(38) A compound as described in any one of (1) to (12) or any other embodiment, wherein R 2 And R 3 Combined Representative-(CH 2 ) r -, Where r is 4, 5, or 6.
Preferred compounds of the general formula (I) according to the invention are, for example, the following compounds and / or their salts and / or solvates and / or hydrates and / or polymorphs and / or bioactive metabolites and / or medicine:
1. 8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3 -a] [1] methyl benzoazine-5-carboxylic acid;
2. 8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3 -a] [1] benzoazine-5-carboxylic acid;
3. 8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3 -a] [1] benzoazine-5-carboxamide;
4. 8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3 -a] [1] benzoazine-5-carbonitrile;
5. 8-chloro-N, N-dimethyl-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4 ] Triazolo [4,3-a] [1] benzoazepine-5-carboxamide;
6. (5S) -8-chloro-N, N-dimethyl-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1 , 2,4] triazolo [4,3-a] [1] benzoazine-5-carboxamidine;
7. (5R) -8-chloro-N, N-dimethyl-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1 , 2,4] triazolo [4,3-a] [1] benzoazine-5-carboxamidine;
8. 8-chloro-N- (prop-2-yl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2 , 4] triazolo [4,3-a] [1] benzoazepine-5-carboxamide;
9. {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4, 3-a] [1] benzoazepine-5-yl} (pyrrolidin-1-yl) methanone;
10. {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4, 3-a] [1] benzoazin-5-yl} (morpholin-4-yl) methanone;
11. {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4, 3-a] [1] benzoazin-5-yl} (1,1-dioxythiomorpholin-4-yl) methanone;
12. {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4, 3-a] [1] benzoazepine-5-yl} methanol;
13. {(5S) -8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazole Ac [4,3-a] [1] benzoazepine-5-yl} methanol;
14. {(5R) -8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazole Ac [4,3-a] [1] benzoazepine-5-yl} methanol;
15. 8-chloro-5- (methoxymethyl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2 , 4] triazolo [4,3-a] [1] benzoazine;
16. Morpholine-4-formate {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2, 4] triazolo [4,3-a] [1] benzoazin-5-yl} methyl;
17. (4-fluorophenyl) carbamic acid {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2 , 4] triazolo [4,3-a] [1] benzoazin-5-yl} methyl ester;
18. 5- (azidomethyl) -8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2 , 4] triazolo [4,3-a] [1] benzoazine;
19. 1- {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [ 4,3-a] [1] benzoazin-5-yl} methylamine;
20. 8-chloro-5- (morpholin-4-ylmethyl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [ 1,2,4] triazolo [4,3-a] [1] benzoazine;
21. 1- {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [ 4,3-a] [1] benzoazin-5-yl} -N, N-dimethylmethylamine;
22. N-({8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazepine-5-yl} methyl) propan-2-amine;
23. 8-chloro-1- [1- (pyrimidin-2-yl) tetrahydroazepine-3-yl] -5,6-dihydro-4H- [1,2,4] triazolo [4, 3-a] [1] Methylbenzoazine-5-carboxylic acid;
24. 8-Chloro-1- [trans-4- (dimethylamino) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a ] [1] Methyl benzoazine-5-carboxylic acid;
25. 8-chloro-1- [trans-4- (trifluoromethyl) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] methyl benzoazine-5-carboxylic acid;
26. 8-chloro-1- [trans-4- (morpholin-4-yl) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3- a] [1] Methylbenzoazine-5-carboxylic acid;
27. {1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazepine-5-yl} methanol;
28. 8-chloro-1- [trans-4- (pyrrolidin-1-ylcarbonyl) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3 -a] [1] methyl benzoazine-5-carboxylic acid;
29. {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4, 3-a] [1] benzoazin-5-yl} acetonitrile;
30. 8-chloro-5-[(4-methylpiperazin-1-yl) methyl] -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6- Dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazine;
31. 8-chloro-5- (fluoromethyl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4 ] Triazolo [4,3-a] [1] benzoazine;
32. 8-chloro-5- (3-methyl-1,2,4-oxadiazol-5-yl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl]- 5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazine;
33. 8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5- (2H-tetrazol-5-yl) -5,6-dihydro-4H- [ 1,2,4] triazolo [4,3-a] [1] benzoazine;
34. N-({8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazepine-5-yl} methyl) tetrahydro-2H-piperan-4-amine;
35. N, N-dimethyl-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -8- (trifluoromethyl) -5,6-dihydro-4H- [ 1,2,4] triazolo [4,3-a] [1] benzoazine-5-amine;
36. 8-Chloro-1- [trans-3-methyl-2-oxo-1-oxa-3-azaspiro [4.5] dec-8-yl] -5,6-dihydro- 4H- [1,2,4] triazolo [4,3-a] [1] benzoazine-5-carboxylic acid methyl ester;
37. N-({8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazepine-5-yl} methyl) acetamide;
38. N-({8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazepine-5-yl} methyl) oxetan-3-amine;
39. 8-chloro-1- [trans-4- (morpholin-4-ylcarbonyl) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3 -a] [1] methyl benzoazine-5-carboxylic acid;
40. 8-Chloro-1- [cis-3-methyl-2-oxo-1-oxa-3-azaspiro [4.5] dec-8-yl] -5,6-dihydro- 4H- [1,2,4] triazolo [4,3-a] [1] benzoazine-5-carboxylic acid methyl ester;
41. 8-chloro-1- [1- (pyridin-2-yl) tetrahydroazepine-3-yl] -5,6-dihydro-4H- [1,2,4] triazolo [4, 3-a] [1] Methylbenzoazine-5-carboxylic acid;
42. 1- [trans-4- (tetrahydroazepine-1-ylcarbonyl) cyclohexyl] -8-chloro-5,6-dihydro-4H- [1,2,4] triazolo [4 , 3-a] [1] Methylbenzoazine-5-carboxylic acid;
43. 8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5- (pyrrolidin-1-ylmethyl) -5,6-dihydro-4H- [ 1,2,4] triazolo [4,3-a] [1] benzoazine;
44. N-({8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazepine-5-yl} methyl) -2-methyl-N- (2-methylpropyl) propan-1-amine;
45. 8-chloro-1- [trans-4- (piperidin-1-ylcarbonyl) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3 -a] [1] methyl benzoazine-5-carboxylic acid;
46. 1- (trans-4-{[4- (tertiary butoxycarbonyl) piperazin-1-yl] carbonyl} cyclohexyl) -8-chloro-5,6-dihydro-4H- [1, 2,4] triazolo [4,3-a] [1] benzoazine-5-carboxylic acid methyl ester;
47. 8-Chloro-1- [trans-4- (piperazin-1-ylcarbonyl) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3 -a] [1] Methylbenzoazine-5-carboxylic acid hydrochloride;
48. 8-methoxy-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4 , 3-a] [1] Methylbenzoazine-5-carboxylic acid;
49. 8-methyl-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4, 3-a] [1] Methylbenzoazine-5-carboxylic acid;
50. 8-Chloro-N- (4-fluorophenyl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2 , 4] triazolo [4,3-a] [1] benzoazepine-5-carboxamide;
51. 8-bromo-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3 -a] [1] methyl benzoazine-5-carboxylic acid;
52. 1- (1,4'-bipiperidin-1'-yl) -8-chloro-5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] Benzacene-5-carboxamide;
53. 8-chloro-1-[(5r, 8r) -1-oxo-2- (prop-2-yl) -2-azaspiro [4.5] dec-8-yl] -5,6- Dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazine-5-carboxylic acid methyl ester;
54. 1- {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [ 4,3-a] [1] benzoazepine-5-yl} ethanol;
55. 1- {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [ 4,3-a] [1] benzoazin-5-yl} ethanone;
56. 8-chloro-5- (fluoromethyl) -1- (trans-4-methoxy-4-methylcyclohexyl) -5,6-dihydro-4H- [1,2,4] Triazolo [4,3-a] [1] benzoazine;
57. 1- {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [ 4,3-a] [1] benzoazin-5-yl} -N- (2-methoxyethyl) ethylamine;
58. 8-fluoro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3 -a] [1] methyl benzoazine-5-carboxylic acid;
59. 8-chloro-5- (fluoromethyl) -1- [trans-4- (trifluoromethyl) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazole Ac [4,3-a] [1] benzoazine;
60. 5- (fluoromethyl) -1- (trans-4-methoxy-4-methylcyclohexyl) -5,6-dihydro-4H- [1,2,4] triazolo [ 4,3-a] [1] benzoazine;
61. {8-fluoro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4, 3-a] [1] benzoazepine-5-yl} methanol;
62. 8-fluoro-5- (fluoromethyl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4 ] Triazolo [4,3-a] [1] benzoazine;
63. cis- (racemic) -1- {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1, 2,4] triazolo [4,3-a] [1] benzoazin-5-yl} ethanol;
64. trans- (racemic) -1- {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1, 2,4] triazolo [4,3-a] [1] benzoazin-5-yl} ethanol;
65. (1R) -1-{(5R) -8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1, 2,4] triazolo [4,3-a] [1] benzoazin-5-yl} ethanol;
66. (1S) -1-{(5S) -8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1, 2,4] triazolo [4,3-a] [1] benzoazin-5-yl} ethanol;
67. (1S) -1-{(5R) -8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1, 2,4] triazolo [4,3-a] [1] benzoazin-5-yl} ethanol;
68. (1R) -1-{(5S) -8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1, 2,4] triazolo [4,3-a] [1] benzoazin-5-yl} ethanol;
69. 5- (fluoromethyl) -8-methyl-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2, 4] triazolo [4,3-a] [1] benzoazine;
70. (5S) -8-chloro-5- (fluoromethyl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1 , 2,4] triazolo [4,3-a] [1] benzoazine;
71. (5R) -8-chloro-5- (fluoromethyl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1 , 2,4] triazolo [4,3-a] [1] benzoazine;
72. 8-fluoro-1- [trans-4- (trifluoromethyl) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] methyl benzoazine-5-carboxylic acid;
73. 8-chloro-1- [4- (2,3-dimethylphenyl) piperazin-1-yl] -5,6-dihydro-4H- [1,2,4] triazolo [ 4,3-a] [1] benzoazepine-5-carboxamide;
74. {8-fluoro-1- [trans-4- (trifluoromethyl) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a ] [1] Benzacridin-5-yl} methanol;
75. {8-chloro-5-methyl-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] tri Zolo [4,3-a] [1] benzoazepine-5-yl} methanol;
76. {8-chloro-5-ethyl-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] tri Zolo [4,3-a] [1] benzoazepine-5-yl} methanol;
77. (5S) -8-fluoro-5- (fluoromethyl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1 , 2,4] triazolo [4,3-a] [1] benzoazine;
78. (5R) -8-fluoro-5- (fluoromethyl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1 , 2,4] triazolo [4,3-a] [1] benzoazine;
79. 8-chloro-1- [4- (3-chlorophenyl) piperazin-1-yl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3- a] [1] Benzacridin-5-carboxamide;
80. 8-chloro-1- [4- (pyridin-2-yl) piperazin-1-yl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3- a] [1] Benzacridin-5-carboxamide;
81. 8-chloro-1- [4- (pyridin-2-yloxy) piperidin-1-yl] -5,6-dihydro-4H- [1,2,4] triazolo [4, 3-a] [1] benzoazine-5-carboxamide;
82. 5-[(Benzyloxy) methyl] -8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1 , 2,4] triazolo [4,3-a] [1] benzoazine;
83. 8-chloro-1- (3,3-difluorocyclobutyl) -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzene And methyl acridine-5-carboxylate;
84. 8-chloro-1- [trans-4- (piperidin-1-ylmethyl) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4, 3-a] [1] Methylbenzoazine-5-carboxylic acid;
85. Acetic acid {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4 , 3-a] [1] benzoazine-5-yl} methyl ester;
86. 8-chloro-1- (1'H, 3H-spiro [2-benzofuran-1,4'-piperidine] -1'-yl) -5,6-dihydro-4H- [1, 2,4] triazolo [4,3-a] [1] benzoazine-5-carboxamide;
87. 8-Chloro-N- (pyridin-2-yl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2 , 4] triazolo [4,3-a] [1] benzoazepine-5-carboxamide;
88. 8-Chloro-1- (1'H, 3H-spiro [2-benzofuran-1,4'-piperidine] -1'-yl) -5,6-dihydro-4H- [1, 2,4] triazolo [4,3-a] [1] benzoazine-5-carbonitrile;
89. 8-chloro-1- [4- (2,3-dimethylphenyl) piperazin-1-yl] -5,6-dihydro-4H- [1,2,4] triazolo [ 4,3-a] [1] benzoazine-5-carbonitrile;
90. 8-chloro-1- [4- (pyridin-2-yl) piperazin-1-yl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3- a] [1] Benzacene-5-carbonitrile;
91. 8-fluoro-5,5-dimethyl-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4 ] Triazolo [4,3-a] [1] benzoazine;
92. 8-fluoro-5- (prop-2-yl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2 , 4] triazolo [4,3-a] [1] benzoazine;
93.8'-Bromo-1 '-[trans-4- (pyridin-2-yloxy) cyclohexyl] -4'H, 6'H-spiro [cyclopentane-1,5'-[1 , 2,4] triazolo [4,3-a] [1] benzoazine];
94. 8-bromo-5- (prop-2-yl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2 , 4] triazolo [4,3-a] [1] benzoazine;
95.8'-bromo-1 '-[trans-4- (pyridin-2-yloxy) cyclohexyl] -2,3,5,6-tetrahydro-4'H, 6'H-spiro [ Piperan-4,5 '-[1,2,4] triazolo [4,3-a] [1] benzoazine];
96. 8-fluoro-5,5-dimethyl-1- [trans-4- (trifluoromethyl) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazole Ac [4,3-a] [1] benzoazine;
97. (5R) -8-bromo-5- (prop-2-yl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazine;
98. (5S) -8-bromo-5- (prop-2-yl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazine;
99. 8'-Bromo-1 '-[trans-4- (pyridin-2-yloxy) cyclohexyl] -4,5-dihydro-4'H, 6'H-spiro [furan-3, 5 '-[1,2,4] triazolo [4,3-a] [1] benzoazine];
100. 8'-Chloro-1 '-[trans-4- (pyridin-2-yloxy) cyclohexyl] -2,3,5,6-tetrahydro-4'H, 6'H-spiro [ Piperan-4,5 '-[1,2,4] triazolo [4,3-a] [1] benzoazine];
101. 8'-Chloro-1 '-[trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H, 4'H, 6'H-spiro [piperan -3,5 '-[1,2,4] triazolo [4,3-a] [1] benzoazine];
102. 8'-Chloro-1 '-[trans-4- (pyridin-2-yloxy) cyclohexyl] -4,5-dihydro-4'H, 6'H-spiro [furan-3, 5 '-[1,2,4] triazolo [4,3-a] [1] benzoazine];
103. 8'-Chloro-1 '-[trans-4- (trifluoromethyl) cyclohexyl] -2,3,5,6-tetrahydro-4'H, 6'H-spiro [piperan- 4,5 '-[1,2,4] triazolo [4,3-a] [1] benzoazine];
104. (3R) -8'-bromo-1 '-[trans-4- (pyridin-2-yloxy) cyclohexyl] -4,5-dihydro-4'H, 6'H-spiro [ Furan-3,5 '-[1,2,4] triazolo [4,3-a] [1] benzoazine];
105. (3S) -8'-bromo-1 '-[trans-4- (pyridin-2-yloxy) cyclohexyl] -4,5-dihydro-4'H, 6'H-spiro [ Furan-3,5 '-[1,2,4] triazolo [4,3-a] [1] benzoazine];
106. (3S) -8'-chloro-1 '-[trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H, 4'H, 6'H- Spiro [piperan-3,5 '-[1,2,4] triazolo [4,3-a] [1] benzoazepine];
107. (3R) -8'-chloro-1 '-[trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H, 4'H, 6'H- Spiro [piperan-3,5 '-[1,2,4] triazolo [4,3-a] [1] benzoazepine];
108. 8-fluoro-1- (trans-4-methoxy-4-methylcyclohexyl) -5,5-dimethyl-5,6-dihydro-4H- [1,2,4] Triazolo [4,3-a] [1] benzoazine;
109. 8-fluoro-1- (cis-4-methoxy-4-methylcyclohexyl) -5,5-dimethyl-5,6-dihydro-4H- [1,2,4] Triazolo [4,3-a] [1] benzoazine;
110. 8'-Chloro-1 '-(trans-4-methoxy-4-methylcyclohexyl) -2,3,5,6-tetrahydro-4'H, 6'H-spiro [pipe Ran-4,5 '-[1,2,4] triazolo [4,3-a] [1] benzoazine];
111. 8'-Chloro-1 '-(cis-4-methoxy-4-methylcyclohexyl) -2,3,5,6-tetrahydro-4'H, 6'H-spiro [pipe Ran-4,5 '-[1,2,4] triazolo [4,3-a] [1] benzoazine]; or
112. 8-chloro-1- [trans-4- (pyridin-2-ylamino) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3 -a] [1] Benzoazine-5-carboxylic acid methyl ester.

本發明亦關於通式(I)化合物之合成。於是,可藉由下述方法中一者製備本發明之通式(I)化合物。
至此,於通式(I)化合物中,R2 係氫,R3 係C(O)R5 ,R5 係C1-4 烷氧基且環A係環烷基或含有1個N之4至7員飽和雜環,其中環A係經由環氮與Y連接,藉由令通式(II)化合物與通式(III)化合物反應以製備本發明之通式(I)化合物,其中該通式(II)化合物係

其中環B和Y係如上述通式(I)定義者且環A係環烷基或含有1個N之4至7員飽和雜環,其中環A係經由環氮與Y連接,且該通式(III)化合物係

其中R1 係如上述通式(I)定義者,R3 係C(O)R5 且R5 係C1-4 烷氧基。
該反應詳細地示於圖1:

於圖1之步驟a)中,通式(II)之醯肼係與通式(III)之苯並吖呯-硫酮反應。較佳地於適當之溶劑中且於該溶劑之沸點下並以所需4至120小時之反應時間進行該反應。適當之溶劑包括例如二甲苯、正丁醇及1,4-二噁烷。
較佳之實施態樣係例如下述者:
i) 式(II)與式(III)之反應,於二甲苯中且於140℃下進行20至120小時;或
ii) 式(II)與式(III)之反應,於正丁醇中且於110℃下進行20至50小時。
可藉由多種不同之方法進行合成通式(II)之醯肼(圖2):

於圖2之步驟a)中,通式(IV)之甲酸酯係於適當之醇中且於該溶劑之沸點下與水合肼反應以獲得通式(II)之醯肼,或於步驟c)中,通式(V)之甲酸係與肼基甲酸三級丁酯反應且使用適當之酸(步驟d))除去經保護之通式(VI)之甲醯肼衍生物的保護基。
較佳之實施態樣係例如下述者:
步驟a) 甲醇或乙醇;水合肼;回流溫度;4至50小時;
步驟b) 甲醇;亞硫醯氯;0至25℃;4至24小時;
步驟c) 肼基甲酸三級丁酯;N,N-二甲基甲醯胺;N,N-二異丙基乙胺;N-(3-二甲基胺基丙基)-N’-乙基碳二亞胺鹽酸鹽;1-羥基苯並三唑水合物;室溫;4至20小時;
步驟d) 溶於乙酸乙酯之鹽酸;室溫;4至20小時。
該通式(IV)之甲酸酯和通式(V)之甲酸係可購得者或可依據實施例描述之方法加以製備。
依據圖3所示之方法,可製備通式(III-a)之主要中間體苯並吖呯-硫酮衍生物(其中R1 係如上述通式(I)定義者):

通式(VII)之酸衍生物(其中R1 係如上述通式(I)定義者)係經酯化(步驟a))以生成通式(VIII)之酯衍生物,且自該通式(VIII)之酯衍生物製備通式(IX)之肟衍生物(步驟b))。藉由貝克曼(Beckmann)重排,將該通式(IX)之肟衍生物轉化為通式(X)之苯並吖呯化合物(步驟c)),且自該通式(X)之苯並吖呯化合物製備該通式(III-a)之苯並吖呯-硫酮衍生物(步驟d))。
較佳之實施態樣係例如下述者:
步驟a) 甲醇;濃硫酸;回流溫度;4至20小時;
步驟b) 甲醇;乙酸鈉;羥基胺鹽酸鹽;回流溫度;2至6小時;
步驟c) 多磷酸;100至120℃;15至60分鐘;
步驟d) i) 勞森(Lawesson)反應劑;吡啶;回流溫度;2至10小時;或
ii) 勞森反應劑;四氫呋喃;室溫;6至20小時。
可自該通式(X)之苯並吖呯衍生物(其中R1 係如上述通式(I)定義者)製備圖4至圖6所示之其他中間體。

於圖4之步驟a)中,藉由1個步驟或3個連續步驟可製得通式(XI)之乙醯基衍生物。該通式(XI)化合物係主要中間體,且自該通式(XI)化合物可製備通式(XII)之二噁茂烷化合物(步驟b))、通式(XIII)之二氟衍生物(步驟c))、通式(XIV)之經保護之化合物(步驟d))(其中PG1 係保護基(Peter G. M. Wuts: Greene’s Protective Groups in Organic Synthesis: Fifth Edition, Chapter 7. Protection for the Amino Group, pages 895-1193),較佳地4-甲氧基苄基-)及通式(XV-a)之羥基化合物(步驟e-1))。藉由使通式(XIV)化合物還原(步驟e-2))以製備通式(XV)之羥基衍生物,且自該通式(XV)之羥基衍生物可製備通式(XVI)之單氟化合物(步驟f))。藉由除去該通式(XVI)之單氟化合物的保護基可獲得通式(XVII)化合物(步驟g))。藉由保護自由羥基,可將該式(XV-a)之未經保護之羥基衍生物轉化為通式(XIX)化合物(步驟h)),其中PG2 係與PG1 不同之保護基(Peter G. M. Wuts: Greene’s Protective Groups in Organic Synthesis: Fifth Edition, Chapter 2. Protection for the Hydroxyl Group, Including 1,2- and 1,3-Diols, pages 17-471),較佳地矽基保護基。
較佳之實施態樣係例如下述者:
步驟a) 甲基鋰溶液;四氫呋喃;二乙醚;(-10℃)至5℃;2至3小時;或
步驟i) 氫氧化鈉水溶液;甲醇;室溫;0.5至3小時;隨後
步驟ii) N-甲氧基甲胺鹽酸鹽;N -(3-二甲基胺基丙基)-N’ -乙基碳二亞胺鹽酸鹽;N,N-二甲基甲醯胺;N,N-二異丙基乙胺或三乙胺;1-羥基苯並三唑水合物;室溫;2至20小時;隨後
步驟iii) 甲基溴化鎂之二乙醚溶液;四氫呋喃;(-10℃)至0℃;2至12小時;
步驟b) 乙二醇;對甲苯磺酸;甲苯;回流溫度;6至12小時;
步驟c)和f) 二乙基胺基三氟化硫;二氯甲烷;(-78)至10℃;4至12小時;
步驟d) 4-甲氧基苄基氯;氫化鈉;N,N-二甲基甲醯胺;0至25℃;3至6小時;
步驟e-1)和e-2) 氫硼化鈉;乙醇或甲醇;室溫;0.5至2小時;
步驟g) i) 硝酸鈰銨;水;乙腈;0至25℃;6至18小時;或
ii) 三氟乙酸;二氯甲烷;室溫;12至24小時;或
iii) 三氟甲烷磺酸;二氯甲烷;室溫;2至12小時;
步驟h) 三氟甲烷磺酸三級丁基二甲基矽烷酯;2,6-二甲基吡啶;二氯甲烷;N,N- 二甲基甲醯胺;0至25℃;6至18小時。
於圖5中,對該通式(XV)之醇衍生物提供保護基(步驟a)),其中PG1 係保護基,較佳地4-甲氧基苄基,且PG2 係與PG1 不同之保護基,較佳地矽基保護基(Peter G. M. Wuts: Greene’s Protective Groups in Organic Synthesis: Fifth Edition, Chapter 2. Protection for the Hydroxyl Group, Including 1,2- and 1,3-Diols, pages 17-471),藉以獲得通式(XVIII)化合物。自該通式(XVIII)化合物除去該保護基PG1 (步驟b))以獲得通式(XIX)化合物。藉由使該通式(XV)之羥基化合物烷基化,可製備通式(XX)化合物(其中烷基表示C1-4 烷基)(步驟c)),且隨後藉由除去該通式(XX)化合物之PG1 保護基,可製備通式(XXI)化合物。

較佳之實施態樣係例如下述者:
步驟a) 三甲基氯矽烷; 咪唑;N,N-二甲基甲醯胺;室溫;4至12小時;
步驟b)和d) i) 硝酸鈰銨;水;乙腈;0至25℃;6至18小時;或
ii) 三氟乙酸;二氯甲烷;室溫;12至24小時;或
iii) 三氟甲烷磺酸;二氯甲烷;室溫;2至12小時;
步驟c) i) 氫化鈉;鹵烷;N,N-二甲基甲醯胺;0至100℃;12至48小時;或
ii) 氫化鈉;磺酸二烷酯;N,N-二甲基甲醯胺;0至100℃;12至48小時;或
iii) 氧化銀;鹵烷;N,N-二甲基甲醯胺;室溫;48至120小時。
如圖6所示,藉由保護基保護通式(X)之苯並吖呯衍生物的氮原子(步驟a))以獲得通式(XXII)之經保護之苯並吖呯衍生物(其中PG1 係保護基(Peter G. M. Wuts: Greene’s Protective Groups in Organic Synthesis: Fifth Edition, Chapter 7. Protection for the Amino Group, pages 895-1193),較佳地4-甲氧基苄基),且隨後使酯基還原(步驟b))以獲得通式(XXIII)之羥基化合物。藉由使該通式(XXIII)之羥基化合物烷基化以製備通式(XXV)化合物(其中烷基表示C1-4 烷基)(步驟c)),且隨後藉由除去保護基PG1 (步驟d))以獲得通式(XXIV)化合物。於步驟e)製備通式(III-b)之苯並吖呯-硫酮衍生物。

較佳之實施態樣係例如下述者:
步驟a) 4-甲氧基苄基氯;氫化鈉;N,N-二甲基甲醯胺;0至25℃;3至6小時;
步驟b) 氫化鋁鋰;四氫呋喃;(-20)至0℃;0.1至1小時;
步驟c) i) 氫化鈉;鹵烷;N,N-二甲基甲醯胺;0至100℃;12至48小時;或
ii) 氫化鈉;磺酸二烷酯;N,N-二甲基甲醯胺;0至100℃;12至48小時;或
iii) 氧化銀;鹵烷;N,N-二甲基甲醯胺;室溫;48至120小時;
步驟d) i) 硝酸鈰銨;水;乙腈;0至25℃;6至18小時;或
ii) 三氟乙酸;二氯甲烷;室溫;12至24小時;或
iii) 三氟甲烷磺酸;二氯甲烷;室溫;2至12小時;
步驟e) i) 勞森反應劑;吡啶;回流溫度;2至10小時;或
ii) 勞森反應劑;四氫呋喃;室溫;6至20小時。
藉由令通式(II)化合物與通式(III-a)化合物反應,可製備通式(I-b)化合物(圖7)。

其中環B、Y及R1 係如上述通式(I)定義者,環A係環烷基或含有1個N之4至7員飽和雜環,其中環A係經由環氮與Y連接。
較佳之實施態樣係例如下述者:
i) 式(II)與式(III-a)之反應:二甲苯;140℃;20至120小時;或
ii) 式(II)與式(III-a)之反應:正丁醇;110℃;20至50小時;或
iii) 式(II)與式(III-a)之反應:1,4-二噁烷;110℃;20至50小時。
如所欲地,藉由習知之方法,經由導入新取代基及/或修飾或除去存在之取代基及/或形成鹽及/或自鹽釋出鹼及/或自消旋性混合物製備鏡像異構物,亦可將因此所獲得之通式(I-b)化合物轉化為另一該通式(I)化合物。該反應流程詳細地描述於圖8和圖9。

藉由使通式(I-b)化合物水解(步驟a)),可製備通式(I-c)化合物(其中環B、Y及R1 係如上述通式(I)定義者,環A係環烷基或含有1個N之4至7員飽和雜環,其中環A係經由環氮與Y連接),且隨後自該通式(I-c)化合物可製備通式(I-d)之醯胺(步驟b))或通式(I-g)之醯胺(步驟e))(其中環B、Y及R1 係如上述通式(I)定義者,環A係環烷基或含有1個N之4至7員飽和雜環,其中環A係經由環氮與Y連接,R’代表氫,R”代表C1-4 烷基或Cy2 ,或R’和R”與彼等連接之N一起形成雜環)。自該通式(I-d)之醯胺可製備通式(I-e)之腈(步驟c)),且自該通式(I-e)之腈可製備通式(I-f)之四唑(步驟d))。自該通式(I-c)之酸亦可製備通式(I-h)之噁二唑衍生物(步驟f))。
較佳之實施態樣係例如下述者:
步驟a) 甲醇;氫氧化鈉;室溫;1至20小時;
步驟b) 氯化銨;N-(3-二甲基胺基丙基)-N’-乙基碳二亞胺鹽酸鹽;N,N-二甲基甲醯胺;N,N-二異丙基乙胺或三乙胺;1-羥基苯並三唑水合物;室溫;4至20小時;
步驟c) 吡啶;磷醯氯;室溫;2至20小時;
步驟d) 疊氮化鈉;氯化銨;N,N-二甲基甲醯胺;100至130℃;2至8小時;
步驟e) i) 適當之胺衍生物;N-(3-二甲基胺基丙基)-N’-乙基碳二亞胺鹽酸鹽;N,N-二甲基甲醯胺;N,N-二異丙基乙胺或三乙胺;1-羥基苯並三唑水合物;室溫;4至20小時;或
ii) 草醯氯;二氯甲烷;室溫;0.5至2小時;隨後吡啶和適當之胺衍生物;室溫;1至5小時;
步驟f) N-(3-二甲基胺基丙基)-N’-乙基碳二亞胺鹽酸鹽;N,N-二甲基甲醯胺;1-羥基苯並三唑水合物;N-羥基乙醯胺;80至120℃;4至20小時。
此外,藉由使該通式(I-b)化合物還原,可製備通式(I-i)之羥基甲基衍生物(圖9;步驟a))。

其中環B、Y及R1 係如上述通式(I)定義者,環A係環烷基或含有1個N之4至7員飽和雜環,其中環A係經由環氮與Y連接。
如所欲地,藉由習知之方法,經由導入新取代基及/或修飾或除去存在之取代基及/或形成鹽及/或自鹽釋出鹼及/或自消旋性混合物製備鏡像異構物,亦可將該通式(I-i)化合物轉化為另一該通式(I)化合物。例如,藉由催化氫化反應,可製備化合物,其中R1 係氫原子(未示於圖)。藉由烷基化或醯化反應,可製備其他衍生物以獲得通式(I-j)化合物(步驟b)),其中環B、Y及R1 係如上述通式(I)定義者,環A係環烷基或含有1個N之4至7員飽和雜環,其中環A係經由環氮與Y連接,且R係烷基或醯基。
自藉由該通式(I-i)化合物之甲烷磺醯化所獲得之該通式(I-k)化合物(步驟c)),亦可製備通式(I-l)之腈衍生物(步驟d))、通式(I-m)之氟化合物(步驟e))及通式(I-n)之胺衍生物(步驟f)),其中環B、Y及R1 係如上述通式(I)定義者,環A係環烷基或含有1個N之4至7員飽和雜環,其中環A係經由環氮與Y連接,R10 和R11 係如上述通式(I)定義之可選擇地經取代之C1-4 烷基,或R10 和R11 與彼等連接之N一起形成可選擇地經取代之雜環。於通式(I-j)中,當OR代表胺甲酸酯時,依據步驟g),可自該通式(I-k)化合物製備化合物。
較佳之實施態樣係例如下述者:
步驟a) 二氯甲烷;二乙醚;氫化鋁鋰;(-30)至(-10) ℃;15至60分鐘;
步驟b) i) 氫化鈉;鹵烷;N,N-二甲基甲醯胺或四氫呋喃;(-5)至40℃;2至10小時;或
ii) 氫化鈉;乙腈;異氰酸酯衍生物;室溫;4至20小時;或
iii) 醯基氯;吡啶;室溫;4至20小時;或
iv) 醯基氯;二氯甲烷;三乙胺或N,N-二異丙基乙胺;室溫;4至20小時;或
v) 酸酐;吡啶;室溫;4至20小時;或
vi) 酸;六氟磷酸N,N,N’,N’-四甲基-O-(1H-苯並三唑-1-基)脲鎓;N,N-二甲基甲醯胺;N,N-二異丙基乙胺;室溫;4至20小時;或
vii) 酸;N-(3-二甲基胺基丙基)-N’-乙基碳二亞胺鹽酸鹽;N,N-二異丙基乙胺;N,N-二甲基甲醯胺;1-羥基苯並三唑水合物;室溫;4至20小時;
步驟c) 甲烷磺醯氯;二氯甲烷;三乙胺;4-二甲基胺基吡啶;室溫;2至20小時;
步驟d) 氰化鈉;N,N-二甲基甲醯胺;70至100℃;3至20小時;
步驟e) 四丁基氟化銨;四氫呋喃;50至70℃;4至20小時;
步驟f) 胺;N,N-二甲基甲醯胺;N,N-二異丙基乙胺;80至120℃;3至20小時;
步驟g) 二級胺;N,N-二甲基甲醯胺;三乙胺;碳酸銫;100℃;12至48小時。
自該通式(I-k)化合物可製備圖10所示之其他衍生物。

步驟a)生成通式(I-o)之疊氮化合物,其中環B、Y及R1 係如上述通式(I)定義者,環A係環烷基或含有1個N之4至7員飽和雜環,其中環A係經由環氮與Y連接,且自該通式(I-o)之疊氮化合物藉由還原反應可製備通式(I-p)之胺衍生物(步驟b))。該通式(I-p)之胺衍生物係通式(I-q)之烷基化合物(步驟c))和通式(I-r)之醯基化合物(步驟d))之起始物,其中環B、Y及R1 係如上述通式(I)定義者,環A係環烷基或含有1個N之4至7員飽和雜環,其中環A係經由環氮與Y連接,R”’代表可選擇地經取代之C1-4 烷基或如上述通式(I)之R10 和R11 所定義之Cy3 ,且醯基係如上述通式(I)之R10 和R11 所定義之C(O)R13 。依據步驟e),亦可自該通式(I-p)化合物製備通式(I-n)化合物,其中R10 和R11 係可選擇地經取代之C1-4 烷基。
較佳之實施態樣係例如下述者:
步驟a) 疊氮化鈉;N,N-二甲基甲醯胺;60至100℃;2至8小時;
步驟b) 三苯膦;四氫呋喃;水;室溫;4至20小時;
步驟c) i) 氫化鈉;鹵烷;N,N-二甲基甲醯胺或四氫呋喃;(-5)至40℃;2至10小時;或
ii) 酮;1,2-二氯乙烷;乙酸;三乙醯氧基氫硼化鈉;室溫;4至20小時;
步驟d) i) 醯基氯;吡啶;室溫;4至20小時;或
ii) 醯基氯;二氯甲烷;三乙胺或N,N-二異丙基乙胺;室溫;4至20小時;或
iii) 酸酐;吡啶;室溫;4至20小時;或
iv) 酸;六氟磷酸N,N,N’,N’-四甲基-O-(1H-苯並三唑-1-基)脲鎓;N,N-二甲基甲醯胺;N,N-二異丙基乙胺;室溫;4至20小時;或
v) 酸;N-(3-二甲基胺基丙基)-N’-乙基碳二亞胺鹽酸鹽;N,N-二異丙基乙胺;N,N-二甲基甲醯胺;1-羥基苯並三唑水合物;室溫;4至20小時;
步驟e) 醛;1,2-二氯乙烷;乙酸;三乙醯氧基氫硼化鈉;室溫;4至20小時。
依據圖11,自通式(XII)化合物之甲基化(步驟a))製備通式(XXVI)之甲氧基苯並吖呯衍生物,其中R1 係如上述通式(I)定義者,且於未經分離下令該通式(XXVI)之甲氧基苯並吖呯衍生物進一步與通式(II)之醯肼反應(步驟b))以生成通式(I-s)化合物,其中環B、Y及R1 係如上述通式(I)定義者,環A係環烷基或含有1個N之4至7員飽和雜環,其中環A係經由環氮與Y連接。經除去縮酮保護基(步驟c))後,得到通式(I-t)之側氧基化合物,其中環B、Y及R1 係如上述通式(I)定義者,環A係環烷基或含有1個N之4至7員飽和雜環,其中環A係經由環氮與Y連接。還原該通式(I-t)之化合物生成通式(I-u)之羥基化合物(步驟d))。使用適當之氧化劑可使該通式(I-u)之化合物氧化(步驟e))以生成該通式(I-t)之化合物,且該通式(I-t)之化合物可經進一步反應以得到通式(I-aa)之胺衍生物,其中R表示可選擇地經烷氧基取代之C1-4 烷基。

較佳之實施態樣係例如下述者:
步驟a) 二氯甲烷;三氟乙酸或碳酸鉀;四氟硼酸三甲基氧鎓;室溫;20至25小時;
步驟b) 通式(II)之醯肼;二氯甲烷或乙腈;50℃;6至20小時;
步驟c) 甲醇;濃氫氯酸;70℃;2至6小時;
步驟d) 氫硼化鈉;乙醇或甲醇;室溫;1至2小時;
步驟e) 戴斯-馬丁碘劑(Dess-Martin periodinane);二氯甲烷;0℃至室溫;1至5小時;
步驟f) 適當之胺;1,2-二氯乙烷;三乙醯氧基氫硼化鈉;乙酸;室溫;4至20小時。
藉由令通式(II)化合物與通式(III-b)化合物反應,可製備通式(I-j)之化合物,其中烷基代表C1-4 烷基且環B、Y及R1 係如上述通式(I)定義者,環A係環烷基或含有1個N之4至7員飽和雜環,其中環A係經由環氮與Y連接(圖12)。

較佳之實施態樣係例如下述者:
i) 式(II)與式(III-b)之反應:二甲苯;140℃;20至120小時;或
ii) 式(II)與式(III-b)之反應:正丁醇;110℃;20至50小時;或
iii) 式(II)與式(III-b)之反應:1,4-二噁烷;110℃;20至50小時。
自該通式(XIII)之二氟衍生物製備通式(III-c)之苯並吖呯-硫酮衍生物(圖13;步驟a)),其中R1 係如上述通式(I)定義者,且令該通式(III-c)之苯並吖呯-硫酮衍生物與通式(II)之醯肼反應(步驟b))以得到通式(I-v)化合物,其中環B、Y及R1 係如上述通式(I)定義者,環A係環烷基或含有1個N之4至7員飽和雜環,其中環A係經由環氮與Y連接。

較佳之實施態樣係例如下述者:
步驟a) i) 勞森反應劑(Lawesson reagent);吡啶;回流溫度;2至10小時;或
ii) 勞森反應劑;四氫呋喃;室溫;6至20小時;
步驟b) i) 式(II)與式(III-c)之反應:二甲苯;140℃;20至120小時;或
ii) 式(II)與式(III-c)之反應:正丁醇;110℃;20至50小時;或
iii) 式(II)與式(III-c)之反應:1,4-二噁烷;110℃;20至50小時。
自通式(XVII)之單氟衍生物製備通式(III-d)之苯並吖呯-硫酮衍生物,其中R1 係如上述通式(I)定義者(圖14;步驟a))且令該通式(III-d)之苯並吖呯-硫酮衍生物與該通式(II)之醯肼反應(步驟b))以得到通式(I-w)化合物,其中環B、Y及R1 係如上述通式(I)定義者,環A係環烷基或含有1個N之4至7員飽和雜環,其中環A係經由環氮與Y連接。

較佳之實施態樣係例如下述者:
步驟a) i) 勞森反應劑;吡啶;回流溫度;2至10小時;或
ii) 勞森反應劑;四氫呋喃;室溫;6至20小時;
步驟b) i) 式(II)與式(III-d)之反應:二甲苯;140℃;20至120小時;或
ii) 式(II)與式(III-d)之反應:正丁醇;110℃;20至50小時;或
iii) 式(II)與式(III-d)之反應:1,4-二噁烷;110℃;20至50小時。
自通式(XIX)之經保護之羥基化合物製備通式(III-e)之苯並吖呯-硫酮衍生物(圖15;步驟a)),其中R1 係如上述通式(I)定義者,PG1 係保護基(Peter G. M. Wuts: Greene’s Protective Groups in Organic Synthesis: Fifth Edition, Chapter 2. Protection for the Hydroxyl Group, Including 1,2- and 1,3-Diols, pages 17-471),較佳地矽基保護基,且令該通式(III-e)之苯並吖呯-硫酮衍生物與通式(II)之醯肼反應(步驟b))以得到通式(I-x)化合物,其中環B、Y及R1 係如上述通式(I)定義者,環A係環烷基或含有1個N之4至7員飽和雜環,其中環A係經由環氮與Y連接。

藉由此合成途徑,經除去通式(I-x)化合物之保護基,亦可製備通式(I-u)之羥基衍生物。
較佳之實施態樣係例如下述者:
步驟a) i) 勞森反應劑;吡啶;回流溫度;2至10小時;或
ii) 勞森反應劑;四氫呋喃;室溫;6至20小時;
步驟b) i) 式(II)與式(III-e)之反應:二甲苯;140℃;20至120小時;或
ii) 式(II)與式(III-e)之反應:正丁醇;110℃;20至50小時;或
iii) 式(II)與式(III-e)之反應:1,4-二噁烷;110℃;20至50小時;
步驟c) 四丁基氟化銨;四氫呋喃;室溫;3至10小時。
自通式(XXI)之烷基衍生物製備通式(III-f)之苯並吖呯-硫酮衍生物,其中R1 係如上述通式(I)定義者且烷基代表C1-4 烷基(圖16;步驟a)),且令該通式(III-f)之苯並吖呯-硫酮衍生物與通式(II)之醯肼反應(步驟b))以得到通式(I-y)化合物,其中環B、Y及R1 係如上述通式(I)定義者,環A係環烷基或含有1個N之4至7員飽和雜環,其中環A係經由環氮與Y連接。

較佳之實施態樣係例如下述者:
步驟a) i) 勞森反應劑;吡啶;回流溫度;2至10小時;或
ii) 勞森反應劑;四氫呋喃;室溫;6至20小時;
步驟b) i) 式(II)與式(III-f)之反應:二甲苯;140℃;20至120小時;或
ii) 式(II)與式(III-f)之反應:正丁醇;110℃;20至50小時;或
iii) 式(II)與式(III-f)之反應:1,4-二噁烷;110℃;20至50小時。
只要於通式(I)化合物中,R2 係C1-4 烷基,R3 係C(O)R5 ,R5 係C1-4 烷氧基且環A係環烷基或含有1個N之4至7員飽和雜環,其中環A係經由環氮與Y連接,藉由令通式(II)化合物與通式(XXVII)化合物反應以製備本發明之通式(I)化合物,其中該通式(II)化合物係

其中環B和Y係如上述通式(I)定義者,環A係環烷基或含有1個N之4至7員飽和雜環,其中環A係經由環氮與Y連接,且該通式(XXVII)化合物係

其中R1 係如上述通式(I)定義者,R2 係C1-4 烷基,R3 係C(O)R5 且R5 係C1-4 烷氧基。
該反應詳細地示於圖17:

圖17之步驟a)的較佳實施態樣係例如下述者:
i) 二甲苯;140℃;20至120小時;或
ii) 正丁醇;110℃;20至50小時;或
iii) 1,4二噁烷;110℃;20至50小時。
藉由下述合成中一者,可製備用於合成通式(XXVII)之苯並吖呯-硫酮衍生物的主要中間體:

方法A (圖18):
藉由二噁茂烷環保護通式(VIII)化合物之酮基(步驟a))且使所生成之通式(XXVIII)化合物烷基化(步驟b))以得到通式(XXIX)化合物,其中R1 係如上述通式(I)定義者且R2 係C1-4 烷基。令該通式(XXIX)化合物去保護(步驟c))且自所生成之通式(XXX)衍生物製備通式(XXXI)之肟化合物(步驟d))。藉由貝克曼重排將該通式(XXXI)之肟轉化為通式(XXXII)之苯並吖呯化合物(步驟e))。

較佳之實施態樣係例如下述者:
步驟a) 乙二醇;甲苯;對甲苯磺酸;回流溫度;2至12小時;
步驟b) 二異丙基胺化鋰;烷基碘;四氫呋喃;(-78) ℃;0.5至2小時;
步驟c) i) 甲醇;濃氫氯酸;70℃;2至6小時;或
ii) 乙酸水溶液;110℃;5至20小時;
步驟d) 甲醇;乙酸鈉;羥基胺鹽酸鹽;回流溫度;3至6小時;
步驟e) 多磷酸;100至120℃;15至60分鐘。

方法B (圖19):
令該經保護之苯並吖呯衍生物(XXII)經烷基化(步驟a)),其中R1 係如上述通式(I)定義者且PG1 係保護基(Peter G. M. Wuts: Greene’s Protective Groups in Organic Synthesis: Fifth Edition, Chapter 7. Protection for the Amino Group, pages 895-1193),較佳地4-甲氧基苄基,且隨後令通式(XXXIII)化合物經去保護(步驟b))以得到通式(XXXII)化合物。

較佳之實施態樣係例如下述者:
步驟a) i) 氫化鈉;N,N-二甲基甲醯胺;烷基碘;0至100℃;1至12小時;或
ii) 二異丙基胺化鋰;烷基碘;四氫呋喃;(-78) ℃;0.5至2小時;
步驟b) i) 硝酸鈰銨;水;乙腈;0至25℃;6至18小時;或
ii) 三氟乙酸;二氯甲烷;室溫;12至24小時;或
iii) 三氟甲烷磺酸;二氯甲烷;室溫;2至12小時。

方法C (圖20):
自通式(XXXIV)化合物製備通式(XXXV)化合物(步驟a)),其中R1 係如上述通式(I)定義者且R2 係C1-4 烷基,並令所生成之通式(XXXV)化合物經水解(步驟b))以得到通式(XXXVI)化合物。令該通式(XXXVI)化合物閉環(步驟c))且令所生成之通式(XXXVII)化合物經酯化(步驟d))。藉由圖18之步驟將所生成之通式(XXX)化合物轉化為通式(XXXII)化合物。

較佳之實施態樣係例如下述者:
步驟a) 二異丙基胺化鋰;四氫呋喃;溴乙酸三級丁酯;0至20℃;3至6小時;
步驟b) 甲醇;氫氧化鈉;室溫;3至6小時;
步驟c) 硫酸;室溫;0.5至1.5小時;
步驟d) 亞硫醯氯;甲醇;60℃;6至8小時。
通式(XXXIV)之胺衍生物係可購得者或可依據實施例描述之方法加以製備。
自通式(XXXII)之主要中間體製備通式(XXVII-a)之苯並吖呯-硫酮衍生物(圖21;步驟a))且令該通式(XXVII-a)之苯並吖呯-硫酮衍生物與通式(II)之醯肼反應以得到通式(I-z)化合物(步驟b)),其中環B、Y及R1 係如上述通式(I)定義者,環A係環烷基或含有1個N之4至7員飽和雜環,其中環A係經由環氮與Y連接且R2 係C1-4 烷基。亦可自通式(XL-a)化合物合成通式(I-z)化合物(步驟d)),且可藉由甲基化反應自通式(XXVII-a)化合物得到該通式(XL-a)化合物(步驟c))。

較佳之實施態樣係例如下述者:
步驟a) i) 勞森反應劑;吡啶;回流溫度;2至10小時;或
ii) 勞森反應劑;四氫呋喃;室溫;6至20小時;
步驟b) i) 式(II)與式(XXVII-a)之反應:二甲苯;140℃;20至120小時;或
ii) 式(II)與式(XXVII-a)之反應:正丁醇;110℃;20至50小時;或
iii) 式(II)與式(XXVII-a)之反應:1,4-二噁烷;110℃;20至50小時;
步驟c) 碘甲烷;碳酸鉀;丙酮;室溫;4至24小時;
步驟d) i) 式(II)與式(XXVII-a)之反應:二甲苯;於催化性鹽酸之存在下;140℃;4至20小時;或
ii) 式(II)與式(XXVII-a)之反應:1,4-二噁烷;於催化性鹽酸之存在下;110℃;4至20小時。
類似於通式(I-b)或(I-i)化合物,如所欲地,藉由之習知方法,經由導入新取代基及/或修飾或除去存在之取代基及/或形成鹽及/或自鹽釋出鹼及/或自消旋性混合物製備鏡像異構物,例如圖8至圖10所示之任何方法,亦可將通式(I-z)化合物轉化為另一該通式(I)化合物。

圖10
只要於該通式(I)化合物中,R2 係氫,R3 係C(O)NH2 且環A係含有1或2個N之4至7員飽和雜環,其中環A係經由環氮與該5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯核之三唑環連接,藉由令通式(XXXVIII)化合物與通式(XXXIX)化合物反應以製備本發明之通式(I)化合物,其中該通式(XXXVIII)化合物係

其中環B和Y係如上述通式(I)定義者,環A係含有1或2個N之4至7員飽和雜環,其中環A係經由環氮與該5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯核之三唑環連接,且該通式(XXXIX)化合物係

其中R1 係如上述通式(I)定義者,R2 係氫且R3 係C(O)NH2
該反應詳細地示於圖22:

圖22之步驟a)之較佳實施態樣係例如下述者:
i) 熔融體(無溶劑);120至150℃;3至72小時;或
ii) 環丁碸;140至180℃;3至72小時。
通式(XXXVIII)之胺衍生物係可購得者或可依據實施例描述之方法加以製備。
藉由圖23之方法可製備通式(XXXIX)之三唑並-苯並吖呯衍生物,其中R1 係如上述通式(I)定義者。

令通式(III-a)化合物經甲基化(步驟a))並令所生成之通式(XL-b)化合物與甲醯肼反應(步驟b))以得到通式(XLI)化合物,其中R1 係如上述通式(I)定義者。令該通式(XLI)化合物經溴化(步驟c))以得到通式(XLII)之溴衍生物,其中R1 係如上述通式(I)定義者。藉由水解自所生成之該通式(XLII)之溴衍生物製備通式(XLIII)化合物(步驟d))並隨後於步驟e)生成通式(XXXIX)之醯胺。亦可自該通式(XXXIX)之醯胺合成通式(LII)之腈衍生物(步驟f))且該通式(LII)之腈衍生物可與下述通式(XXXVIII)化合物反應以生成通式(I-ac)化合物:

其中環B和Y係如上述通式(I)定義者,環A係含有1或2個N之4至7員飽和雜環,其中環A係經由環氮與該5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯核之三唑環連接。
較佳之實施態樣係例如下述者:
步驟a) 碘甲烷;碳酸鉀;丙酮;室溫;4至24小時;
步驟b) 甲醯基肼;1,4-二噁烷;90℃;3至10小時;
步驟c) N-溴琥珀醯亞胺;四氫呋喃;70℃;10至60分鐘;
步驟d) 甲醇;氫氧化鈉;室溫;1至20小時;
步驟e) 氯化銨;N-(3-二甲基胺基丙基)-N’-乙基碳二亞胺鹽酸鹽;N,N-二甲基甲醯胺;N,N-二異丙基乙胺或三乙胺;1-羥基苯並三唑水合物;室溫;4至20小時;
步驟f) 三氟乙酸酐;二氯甲烷;三乙胺;0℃至室溫;1至5小時;
步驟g) i) 熔融體(無溶劑);120至150℃;3至72小時;或
ii) 環丁碸;140至180℃;3至72小時。
如所欲地,藉由習知之方法,經由導入新取代基及/或修飾或除去存在之取代基及/或形成鹽及/或自鹽釋出鹼及/或自消旋性混合物製備鏡像異構物,例如使用圖8所示之步驟c)和d),亦可將依據圖22製備之通式(I-aa)化合物轉化為另一該通式(I)化合物。
藉由詳細描述於圖24之方法,可製備該通式(I)化合物,其中R1 係如上述通式(I)定義者,R2 係氫或C1-4 烷基,R3 係C1-4 烷基,或R2 和R3 結合代表-(CH2 )p -O-(CH2 )q -或 -(CH2 )r -,其中p係1、2或3;q係1、2或3;r係4、5或6。

藉由格任亞(Grignard)反應(步驟a))將通式(XLIV)之丙二酸二乙酯衍生物(其中R2 係氫或C1-4 烷基,R3 係C1-4 烷基,或R2 和R3 結合代表-(CH2 )p -O-(CH2 )q -或-(CH2 )r -,其中p係1、2或3;q係1、2或3;r係4、5或6)轉化為通式(XLV)之苄基衍生物,其中R1 代表氫或氟。於鹼性條件下,藉由水解通式(XLV)化合物以製備通式(XLVI)之自由酸(步驟b))。通式(XLVI)化合物之去羧基化(步驟c))生成通式(XLVII)化合物,且令該通式(XLVII)化合物經濃硫酸處理以製備通式(XLVIII)化合物(步驟d))。隨後自該通式(XLVIII)化合物製備通式(XLIX)之肟衍生物(步驟e)),且藉由貝克曼重排(步驟f))將該通式(XLIX)之肟衍生物轉化為通式(L)之苯並吖呯化合物。若R3 係氫,令通式(L)化合物經氯化或溴化(步驟g))以得到通式(LI)化合物。將通式(LI)或(L)化合物轉化為通式(I-ad)化合物(步驟h)),其中R1 係如上述通式(I)定義者,R2 係氫或C1-4 烷基,R3 係C1-4 烷基,或R2 和R3 結合代表-(CH2 )p -O-(CH2 )q -或-(CH2 )r -,其中p係1、2或3;q係1、2或3;r係4、5或6,環B和Y係如上述通式(I)定義者且環A係環烷基或含有1個N之4至7員飽和雜環,其中環A係經由環氮與Y連接。
圖24之合成之較佳實施態樣係例如下述者:
步驟a) 苄基氯化鎂或3-氟苄基氯化鎂;二乙醚;室溫;2至12小時;
步驟b) 2M氫氧化鈉水溶液;乙醇;回流溫度;2小時;
步驟c) 於180至200℃下加熱3至5分鐘;
步驟d) 硫酸;35至40℃;60分鐘;
步驟e) 甲醇;乙酸鈉;羥基胺鹽酸鹽;回流溫度;2至6小時;
步驟f) 多磷酸;100至120℃;15至60分鐘;
步驟g) 溴;乙酸/水;室溫;30至90分鐘;或,N-氯琥珀醯亞胺;DMF;室溫;30至90分鐘;
步驟h) i) 勞森反應劑;吡啶;回流溫度;2至10小時;
ii) 碘甲烷;碳酸鉀;丙酮;40℃;1至3小時;
iii) 通式(II)之醯肼;濃氫氯酸(數滴);1,4-二噁烷;回流;1至8小時;

i) 四氟硼酸三甲基氧鎓;三氟乙酸或碳酸鉀;二氯甲烷;室溫;2至4小時;
ii) 通式(II)之醯肼;三氟乙酸(數滴);1,4-二噁烷或乙腈;回流;1至8小時。
藉由圖2描述之不同方法,可合成通式(II)之醯肼,其中環B和Y係如上述通式(I)定義者且環A係環烷基或含有1個N之4至7員飽和雜環,其中環A係經由環氮與Y連接。
依據圖25所示之方法,亦可合成通式(I-m)化合物。

令通式(X)化合物(其中R1 係如上述通式(I)定義者)經還原(步驟a))並將所得之通式(LIII)之羥基化合物轉化為通式(LIV)之氟衍生物(步驟b))。直接自通式(LV)之苯並吖呯-硫酮(其係得自於步驟c)之通式(LIV)化合物)(步驟e))或自通式(LVII)之甲硫基衍生物(步驟h)),可合成通式(I-m)化合物。藉由甲基化(步驟f))可自該通式(LV)之苯並吖呯-硫酮製備該通式(LVII)之甲硫基衍生物。可替代之合成途徑係使該通式(LIV)化合物甲基化(步驟d))並於原位(in situ)令所得之通式(LVI)之甲氧基衍生物與通式(II)化合物反應(步驟g))以生成該通式(I-m)化合物。
圖25之合成之較佳實施態樣係例如下述者:
步驟a) 氫化鋁鋰;二乙醚;四氫呋喃;(-40)至0℃;0.1至2小時;
步驟b) 二乙基胺基三氟化硫;二氯甲烷;(-78)至10℃;4至12小時;
步驟c) i) 勞森反應劑;吡啶;回流溫度;2至10小時;或
ii) 勞森反應劑;四氫呋喃;室溫;6至20小時;
步驟d) 四氟硼酸三甲基氧鎓;三氟乙酸或碳酸鉀;二氯甲烷;室溫;2至4小時;
步驟e) i) 通式(II)之醯肼與通式(LV)反應;二甲苯;140℃;20至120小時;或
ii) 通式(II)之醯肼與通式(LV)反應;正丁醇;110℃;20至50小時;或
iii) 通式(II)之醯肼與通式(LV)反應;1,4-二噁烷;110℃;20至50小時;
步驟f) 碘甲烷;碳酸鉀;丙酮;室溫;4至24小時;
步驟g) 通式(II)之醯肼;二氯甲烷或乙腈;50℃;6至20小時;
步驟h) 通式(II)之醯肼與通式(LVII)反應;1,4-二噁烷;110℃;2至20小時。
依據圖26所示之方法,可製備本發明之通式(I-ad)化合物,其中R1 係如上述通式(I)定義者,R2 係氫或C1-4 烷基,R3 係C1-4 烷基,或R2 和R3 結合代表-(CH2 )p -O-(CH2 )q -或-(CH2 )r -,其中p係1、2或3;q係1、2或3;r係4、5或6,且環A係含有1或2個N之4至7員飽和雜環,其中環A係經由環氮與該5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯核之三唑環連接。

令通式(LI)化合物(R1 係如上述通式(I)定義者,R2 係氫或C1-4 烷基,R3 係C1-4 烷基,或R2 和R3 結合代表-(CH2 )p -O-(CH2 )q -或 -(CH2 )r -,其中p係1、2或3;q係1、2或3;r係4、5或6)與勞森反應劑反應(步驟a))以生成通式(LVIII)之苯並吖呯-硫酮衍生物,並令該通式(LVIII)之苯並吖呯-硫酮衍生物經甲基化(步驟b))且令所得之通式(LIX)化合物與甲醯肼反應(步驟c))以生成通式(LX)化合物。令該通式(LX)化合物經溴化(步驟d))以得到通式(LXI)之溴衍生物,該通式(LXI)之溴衍生物與下述通式(XXXVIII)化合物反應(步驟e))以生成通式(I-ae)化合物:

其中環B和Y係如上述通式(I)定義者,環A係含有1或2個N之4至7員飽和雜環,其中環A係經由環氮與該5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯核之三唑環連接。
圖26之合成之較佳實施態樣係例如下述者:
步驟a) i) 勞森反應劑;吡啶;回流溫度;2至10小時;或
ii) 勞森反應劑;四氫呋喃;室溫;6至20小時;
步驟b) 碘甲烷;碳酸鉀;丙酮;室溫;4至24小時;
步驟c) 甲醯基肼;1,4-二噁烷;90℃;3至10小時;
步驟d) N-溴琥珀醯亞胺;四氫呋喃;70℃;10至60分鐘;
步驟e) i) 熔融體(無溶劑);120至150℃;3至72小時;或
ii) 環丁碸;140至180℃;3至72小時。
上述反應所需之反應劑和詳細製程步驟係描述於實施例。
本發明之一方面關於製備通式(I)之化合物的過程所合成的通式(III-a)、(X)、(XLII)、(LI)及(LIV)所示之新穎中間體,其中R1 係如上述通式(I)定義者,特別是7-氯-2-側氧基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯(中間體3)、7-氯-2-側硫基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯(中間體4)、1-溴-8-氯-5,6-二氫-4H -[1,2,4]三唑並[4,3-a ][1]苯並吖呯-5-甲酸甲酯(中間體24)、7-氯-4-(氟甲基)-1,3,4,5-四氫-2H -1-苯並吖呯-2-酮(中間體36),或其中R2 和R3 結合代表-(CH2 )p -O-(CH2 )q -或-(CH2 )r -,其中p係1、2或3;q係1、2或3;r係4、5或6。
藉由下述之方法活體外或活體內測定本發明之每一通式(I)化合物的活性數據。

人血管升壓素V1a受體結合分析
細胞和放射性配體
使用結構性且穩定表現人血管升壓素V1a受體之永生化1321N1細胞株(Perkin Elmer, ES-361-M400-UA)和作為放射性配體之血管升壓素(8-L-精胺酸)的經[苯基丙胺醯基-3,4,5-3 H(N)]標記之化合物(Perkin Elmer Life and Analytical Sciences)以測定所製備之化合物的親和性。

方法
細胞膜製備:依據Jarvis氏方法進行表現經增殖之人血管升壓素V1a受體的永生化1321N1細胞之細胞膜製備(Jarvis et al.,J Pharmacol Exp Ther 2004, 310:407-16)。令細胞懸浮於製備性緩衝液(50 mM Tris,1 mM EDTA,0.1 mM PMSF)並經玻璃均質器均勻化。為分離粗細胞膜餾分,進行2次連續離心操作(40,000 g,20分鐘,4℃)且隨後於最終沖洗步驟將該細胞膜置入該製備性緩衝液,將所得之細胞膜製劑分為數份且儲存於-80℃下直至測量時使用。
依據Lowry氏方法,使用牛血清白蛋白(BSA)之標準稀釋系列,測量所製備之細胞膜的蛋白含量(Lowry et al.,J Biol Chem 1951, 193:265-75)。
受體結合試驗:於受體結合分析中,使用至少8種不同濃度且每種濃度為3個並行之未知親和性的物質。為測定最終親和性數值,至少2個獨立實驗之結果列入考慮。分析混合物包括培育緩衝液(50 mM Tris-HCl,pH 7.4+3% BSA)、表現人血管升壓素V1a受體之1321N1細胞的細胞膜製劑(167 µg/ml)及作為放射性配體之血管升壓素(8-L-精胺酸)[苯基丙胺醯基-3,4,5-3 H(N)](1 nM)。
於1.2x10-6 M未經標記之(Arg8 )-血管升壓素的存在下測量非專一性結合值。令總體積為0.33 ml之樣品於27℃下經培育60分鐘。藉由通過經0.5%聚乙烯亞胺浸漬之UniFilter® GF/BTM 過濾以分離與細胞膜結合之配體和游離配體。經乾燥濾盤後,對該樣品加入40 μl Microscint-20 (Packard)閃爍液。最終,使用MicroBeta2 微盤計數儀(Perkin Elmer)測量放射性。
使用S形擬合數學方法y=(A1-A2)/(1+(x/x0 )p)+A2和Origin 7.5.軟體(OriginLab Corporation, Northampton, USA)濃度位移曲線計算IC50 值(即替代50%專一性結合之放射性配體的該未知物質之濃度)。於擬合時,不固定漸近線。使用Cheng-Prusoff方程式Ki= IC50 /[1+(L/KD )]計算抑制常數(Ki )值,其中[L]係實驗所使用之放射性配體濃度且[KD ]係經放射性標記之配體對給予受體之親和性。使用Scatchard曲線預先測定KD

測試化合物對表現人血管升壓素V1a受體之細胞株的功能性分析
細胞
使用結構性且穩定表現人血管升壓素V1a受體之永生化1321N1細胞株(Perkin Elmer, ES-361-M400-UA)以測量所製備之化合物。使用待測之GPCR受體的一般二級信使途徑,即內源性Gq 相關系統。

方法
使用96孔槽盤(30,000細胞/盤)測量化合物。該測量使用之緩衝液組成(以mM表示)係如下:140 NaCl,5 KCl,2 CaCl2 ,2 MgCl2 ,10葡萄糖,10 HEPES (4-(2-羥基乙基)-1-哌嗪-乙烷磺酸),2丙磺舒(probenecid),pH 7.4。使用FLIPR Calcium 5套組(Molecular Devices)作為螢光染料,於填充該染料之前未除去基質且其中前後皆未清洗細胞。於室溫下進行培育且最終DMSO濃度為1%。於15至20分鐘預處理投予待測之物質且對每一化合物之每一濃度進行至少2個平行測量。
使用螢光訊號以測定細胞內Ca2+ 濃度且讀數計係FlexStation II96。藉由螢光測定法且使用FlexStation II96盤讀數計(激發:485 nm;發射:525 nm)測量細胞質Ca2+ 濃度。每1.4秒記錄螢光訊號達1分鐘。使用之參考化合物係如下:對每一盤於EC80 濃度下測定之(Arg8 )-血管升壓素作為激動劑,且於1 μM下測定之瑞考伐普坦作為拮抗劑。測定化合物的每一濃度之抑制%和IC50 值,其中亦測定濃度線。
記錄每盤之總AVP濃度-反應曲線。所測量之化合物的功效係以相對於對照組反應之相對抑制%表示。對於數據之圖示,使用SoftMaxPro軟體且依據下述公式:y=A-D/(1+(x/C)^B)+D,其中A=0且D=100-下方/上方固定漸近線,y=抑制%,x=測試化合物濃度之對數值,B=曲線陡度且C=IC50 (對照組反應之50%抑制的濃度),進行非線性4參數並列。對於所有化合物,自至少3次獨立測量計算平均IC50 值。





小鼠血管升壓素V1a受體結合分析
細胞和放射性配體
使用結構性且穩定表現小鼠血管升壓素V1a受體之永生化1321N1細胞株(B9/1321N1選殖株)和作為放射性配體之血管升壓素(8-L-精胺酸)的經[苯基丙胺醯基-3,4,5-3 H(N)]標記之化合物(Perkin Elmer Life and Analytical Sciences)以測定所製備之化合物的親和性。

方法
細胞膜製備:依據Jarvis氏方法進行表現經增殖之小鼠血管升壓素V1a受體的永生化1321N1細胞之細胞膜製備(Jarvis et al.,J Pharmacol Exp Ther 2004, 310:407-16)。令細胞懸浮於製備性緩衝液(50 mM Tris,1 mM EDTA,0.1 mM PMSF)並經玻璃均質器均勻化。為分離粗細胞膜餾分,進行2次連續離心操作(40,000 g,20分鐘,4℃)且隨後於最終沖洗步驟將該細胞膜置入該製備性緩衝液,將所得之細胞膜製劑分為數份且儲存於-80℃下直至測量時使用。
依據Lowry氏方法,使用牛血清白蛋白(BSA)之標準稀釋系列,測量所製備之細胞膜的蛋白含量(Lowry et al.,J Biol Chem 1951, 193:265-75)。
受體結合試驗:於受體結合分析中,使用至少8種不同濃度且每種濃度為3個並行之未知親和性的物質。為測定最終親和性數值,至少2個獨立實驗之結果列入考慮。分析混合物包括培育緩衝液(50 mM Tris-HCl,pH 7.4+3% BSA)、表現小鼠血管升壓素V1a受體之1321N1細胞的細胞膜製劑(152 µg/ml)及作為放射性配體之血管升壓素(8-L-精胺酸)[苯基丙胺醯基-3,4,5-3 H(N)](約KD 之35至50%濃度)。
於1.2x10-6 M未經標記之(Arg8 )-血管升壓素的存在下測量非專一性結合值。令總體積為0.33 ml之樣品於27℃下經培育60分鐘。藉由通過經0.5%聚乙烯亞胺浸漬之UniFilter® GF/BTM 過濾以分離與細胞膜結合之配體和游離配體。經乾燥濾盤後,對該樣品加入40 μl Microscint-20 (Packard)閃爍液。最終,使用MicroBeta2 微盤計數儀(Perkin Elmer)測量放射性。
藉由至少2個獨立實驗測量物質之放射性配體箝夾能力。專一性放射性配體結合可被定義為於飽和量之未經標記配體或不同濃度之待測物的存在下全部結合與非專一性結合之差異。結果係以於待測物的存在下所達到之專一性結合的抑制%表示。
使用S形擬合數學方法y=(A1-A2)/(1+(x/x0 )p)+A2和Origin 7.5.軟體(OriginLab Corporation, Northampton, USA) 濃度位移曲線計算IC50 值(即替代50%專一性結合之放射性配體的該未知物質之濃度)。於擬合時,不固定漸近線。使用Cheng-Prusoff方程式Ki= IC50 /[1+(L/KD )]計算抑制常數(Ki )值,其中[L]係實驗所使用之放射性配體濃度且[KD ]係經放射性標記之配體對給予受體之親和性。使用Scatchard曲線預先測定KD


人血管升壓素V2受體結合分析
細胞和放射性配體
使用穩定且結構性表現人血管升壓素V2受體之永生化1321N1細胞株(Perkin Elmer, ES-363-M400-UA)(批號1765208)、表現人血管升壓素V2受體之CHO-K1細胞膜(Perkin Elmer, 6110541400UA)及作為放射性配體之血管升壓素(8-L-精胺酸)的經[苯基丙胺醯基-3,4,5-3 H(N)]標記之化合物(Perkin Elmer Life and Analytical Sciences)以測定所製備之化合物的親和性。

方法
受體結合試驗:於受體結合分析中,使用至少8種不同濃度且每種濃度為3個並行之未知親和性的物質。為測定最終親和性數值,至少2個獨立實驗之結果列入考慮。分析混合物包括培育緩衝液(50 mM Tris-HCl,pH 7.4+3% BSA)、表現人血管升壓素V2受體之1321N1細胞的細胞膜製劑(1.82 µg/ml)及作為放射性配體之血管升壓素(8-L-精胺酸)[苯基丙胺醯基-3,4,5-3 H(N)](約KD 之濃度)。
於1.2x10-6 M未經標記之(Arg8 )-血管升壓素的存在下測量非專一性結合值。令總體積為0.55 ml之樣品於27℃下經培育90分鐘。藉由通過經0.5%聚乙烯亞胺浸漬之UniFilter® GF/BTM 過濾以分離與細胞膜結合之配體和游離配體。經乾燥濾盤後,對該樣品加入40 μl Microscint-20 (Packard)閃爍液。最終,使用MicroBeta2 微盤計數儀(Perkin Elmer)測量放射性。
藉由至少2個獨立實驗測量物質之放射性配體替代能力。專一性放射性配體結合可被定義為於飽和量之未經標記配體或不同濃度之待測物的存在下全部結合與非專一性結合之差異。結果係以於待測物的存在下所達到之專一性結合的抑制%表示。
使用S形擬合數學方法y=(A1-A2)/(1+(x/x0 )p)+A2和Origin 7.5.軟體(OriginLab Corporation, Northampton, USA)由濃度位移曲線計算IC50 值(即替代50%專一性結合之放射性配體的該未知物質之濃度)。於擬合時,不固定漸近線。使用Cheng-Prusoff方程式Ki= IC50 /[1+(L/KD )]計算抑制常數(Ki )值,其中[L]係實驗所使用之放射性配體濃度且[KD ]係經放射性標記之配體對給予受體之親和性。使用Scatchard曲線預先測定KD

方法
於至少2個使用培育緩衝液(50 mM Tris-HCl,5 mM MgCl2 ,pH 7.4+0.1% BSA)、表現人血管升壓素V2受體之CHO-K1細胞膜製劑(7 µg/µl)(Perkin Elmer, 6110541400UA)及作為放射性配體之血管升壓素(8-L-精胺酸)[苯基丙胺醯基-3,4,5-3 H(N)](約KD 之濃度)的獨立實驗,使用至少8種濃度且每種濃度為2或3個並行樣品以實施受體結合分析。
於1.2x10-6 M未經標記之(Arg8 )-血管升壓素的存在下可測量非專一性結合值。令總體積為0.55 ml之樣品於27℃下經培育90分鐘。藉由通過經聚乙烯亞胺浸漬之UniFilter® GF/BTM 過濾以分離與細胞膜結合之配體和游離配體。使用0.5 ml冰冷沖洗緩衝液(50 mM Tris-HCl,pH 7.4)沖洗濾盤3次。經乾燥該濾盤後,對每個孔槽加入40 μl Microscint-20 (Packard)閃爍液。最終,使用Tri-Carb 2900TR液體閃爍分析儀(Perkin Elmer)測量放射性。
藉由至少2個獨立實驗測量物質之放射性配體替代能力。專一性放射性配體結合可被定義為於飽和量之未經標記配體或不同濃度之待測物的存在下全部結合與非專一性結合之差異。結果係以於待測物的存在下所達到之專一性結合的抑制%表示。
使用S形擬合數學方法y=(A1-A2)/(1+(x/x0 )p)+A2和Origin 7.5.軟體(OriginLab Corporation, Northampton, USA)由濃度位移曲線計算IC50 值(即替代50%專一性結合之放射性配體的該未知物質之濃度)。於擬合時,不固定漸近線。使用Cheng-Prusoff方程式Ki= IC50 /[1+(L/KD )]計算抑制常數(Ki )值,其中[L]係實驗所使用之放射性配體濃度且[KD ]係經放射性標記之配體對給予受體之親和性。使用Scatchard曲線預先測定KD
於該表現人血管升壓素V2受體之1321N1細胞株所測量之親和性數據(Ki )係與表現人血管升壓素V2受體之CHO-K1細胞株所得之Ki 結果極為緊密相關。

功能性V1a活體內試驗
動物
使用重18至40 g雄性小鼠(NMRI, ToxiCoop)。自送達後飼養動物至少5天,且於圈養和測量期間令動物自由攝食和飲水。實驗經由地區動物保護委員會批准且依據歐洲動物保護指南(EU Directive 2010/63/EU )實施。

方法
藉由自動化行為分析系統(LABORASTM )測量動物行為。位於平板下方之感測器偵測動物運動所產生之機械振動並轉換為電子訊號(Quinn et al.,J Neurosci Methods 2003, 130:83-92)。經分析訊號後,該系統分析下列行為參數耗費之時間:運動、靜止不動、攀爬及梳洗。定義之梳洗算法能測量搔抓行為反應。於實驗期間,令小鼠經測試物質或載體預處理,且經該預處理期間後,投予引起搔抓之化合物(皮下0.3 mg/kg催產素),並隨後將該動物獨立地置入測量籠。觀察該等小鼠之行為達1小時。為減少探索活動,於該籠內圈養1小時後測量該等動物。令該等行為參數與對照組動物之並行測量的參數相比較。
藉由經並行測量的載體處理組之平均值計算測試物質之行為抑制功效並以抑制%表示,其中0%表示載體預處理動物(和載體預處理之皮下生理食鹽水者)之搔抓行為的平均值,且100%表示皮下接受催產素之載體預處理動物的搔抓平均值。針對統計分析,使用Tukey事後比較檢定之單因子變異數分析(ANOVA)。
令人意外地,經發現本發明之某些化合物於活體內功能性試驗對小鼠V1a受體能產生顯著之功效。

本發明將藉由下述之實施態樣進一步加以說明,但是本發明之範圍不限於該等實施態樣。藉由上述說明和下述實施例,熟習此技術之人士可確定本發明之實質特徵,且未偏離本發明之實質和範圍下可作某些改變和修飾以使本發明能適於多種不同之應用和條件。於是,本發明不限於下述說明性實施例,而是限於所附之申請專利範圍所判定之範圍。
通常,依據熟習此技術之人士的一般知識及/或製備實施例化合物及/或中間體之方法可製備該通式(I)化合物。熟習此技術之人士可輕易地選擇溶劑、溫度、壓力及其他反應條件。起始物係可購得者及/或可藉由熟習此技術之人士輕易地製備。於製備化合物時,可使用例如組合技術,其中存在之中間體基團係適合用於該等方法。
於合成中,使用下述之術語和簡稱:
dry=無水
Boc=三級丁氧羰基
戴斯-馬丁碘劑(Dess-Martin periodinane)=1,1,1-三(乙醯氧基)-1,1-二氫-1,2-苯並碘雜噁唑-3-(1H)-酮
DIPEA=N,N-二異丙基-乙胺
DMAP=4-二甲基胺基-吡啶
DMF=N,N-二甲基甲醯胺
EDC=N-(3-二甲基胺基丙基)-N’-乙基碳二亞胺鹽酸鹽
HOBt=1-羥基苯並三唑水合物
HBTU=六氟磷酸N,N,N’,N’-四甲基-O-(1H-苯並三唑-1-基)脲鎓
K2 CO3 =碳酸鉀
勞森反應劑(Lawesson reagent)=2,4-二(4-甲氧基苯基)-1,3,2,4-二硫雜二磷雜環丁烷-2,4-二硫化物
米氏酸(Meldrum’s acid)=2,2-二甲基-1,3-二噁烷-4,6-二酮
MgSO4 =硫酸鎂
NaBH4 =氫硼化鈉
NaBH(OAc)3 =三乙醯氧基氫硼化鈉
NaCl=氯化鈉
Na2 CO3 =碳酸鈉
NaHCO3 =碳酸氫鈉
NaOAc=乙酸鈉
NaOH=氫氧化鈉
Na2 SO4 =硫酸鈉
Pd/C=碳上鈀
Pt/C=碳上鉑
THF=四氫呋喃

中間體1
7-氯-4-側氧基-1,2,3,4-四氫萘-2-甲酸甲酯

令7-氯-4-側氧基-1,2,3,4-四氫萘-2-甲酸(6.0 g,26.7毫莫耳)(R. D. Haworth et al.,J Chem Soc 1943, 10)溶解於甲醇(100 ml)並加入濃硫酸(0.5 ml)且令混合物於回流溫度下經隔夜攪拌並經濃縮。令殘餘物溶解於二乙醚和乙酸乙酯(1:1)混合物並經10% Na2 CO3 溶液和水沖洗。令水相經乙酸乙酯萃取。令合併之有機相經飽和NaCl溶液沖洗,置於無水硫酸鈉上乾燥,且經過濾和濃縮。因此,得到呈米色結晶之標的產物(6.27 g,98.4%)。MS (ESI) m/z 239.1 (M+H)+

中間體2
7-氯-4-(羥基亞胺基)-1,2,3,4-四氫萘-2-甲酸甲酯

令7-氯-4-側氧基-1,2,3,4-四氫萘-2-甲酸甲酯(中間體1)(6.27 g,26.3毫莫耳)溶解於甲醇(220 ml),且隨後加入無水NaOAc(6.57 g,80毫莫耳)和羥基胺鹽酸鹽(5.55 g,80毫莫耳)。令混合物經回流3小時並隨後於室溫下經隔夜攪拌。令反應混合物經濃縮,加入水,經乙酸乙酯萃取,令合併之有機相經飽和NaCl溶液沖洗,置於無水硫酸鈉上乾燥,且經過濾和濃縮。因此,得到呈微黃色結晶之標的產物(6.63 g,99.4%)。MS (ESI) m/z 254.1 (M+H)+

中間體3
7-氯-2-側氧基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯

將7-氯-4-(羥基亞胺基)-1,2,3,4-四氫萘-2-甲酸甲酯(中間體2)(6.57 g,25.9毫莫耳)加入至多磷酸(60.0 g)並於110℃下經攪拌20分鐘。將冰加入至反應混合物並經攪拌10分鐘。隨後經乙酸乙酯萃取,令合併之有機相經飽和NaCl溶液沖洗,置於無水硫酸鈉上乾燥,且經過濾和濃縮。令殘餘物經管柱層析(使用環己烷:乙酸乙酯=2:1作為溶析液)純化。因此,得到呈白色結晶之標的產物(3.03 g,46.2%)。MS (ESI) m/z 254.1 (M+H)+

中間體4
7-氯-2-側硫基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯

令7-氯-2-側氧基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯(中間體3)(1.50 g,5.9毫莫耳)溶解於吡啶(80 ml),加入勞森反應劑(3.11 g,7.7毫莫耳)並令反應混合物於回流溫度下經攪拌2小時,隨後經濃縮。將水(15 ml)和飽和NaHCO3 溶液(60 ml)加入至殘餘物並令高度沉澱之懸浮液經攪拌30分鐘。過濾出沉澱之黃色產物並於真空和50℃下置於五氧化二磷上乾燥。因此,得到呈黃色結晶之標的產物(1.58 g,99.9%)。MS (ESI) m/z 270.1 (M+H)+

中間體5
2-{[反式-4-(三氟甲基)環己基]羰基}肼甲酸三級丁酯

令反式-4-(三氟甲基)環己烷甲酸(Manchester Organics Ltd)(2.13 g,10.9毫莫耳)溶解於DMF (50 ml)。對該溶液加入肼甲酸三級丁酯(1.44 g,10.9毫莫耳)、DIPEA (4.75 ml,27.3毫莫耳)、HOBt (2.00 g,13.10毫莫耳)及EDC (2.51 g,13.1毫莫耳)。令反應混合物於室溫下經攪拌36小時並隨後經濃縮。將飽和NaHCO3 溶液(40 ml)加入至殘餘物並經短暫攪拌後令沉澱物經過濾,經水沖洗且於真空烘箱中置於五氧化二磷上乾燥。因此,得到呈白色粉末之標的產物(3.35 g,99%)。GC-MS (EI) m/z 310.1。

中間體6
反式-4-(三氟甲基)環己烷甲醯肼

令2-{[反式-4-(三氟甲基)環己基]羰基}肼甲酸三級丁酯(中間體5)(3.35 g,10.79毫莫耳)溶解於乙酸乙酯(50 ml)和乙醇(20 ml)之混合物並隨後對該溶液加入2.5M鹽酸之乙酸乙酯溶液(30 ml)。令反應混合物於室溫下經攪拌16小時,隨後加入二乙醚(150 ml)並於冰水浴中經冷卻。令沉澱之產物經過濾並經二乙醚沖洗。令過濾物經飽和NaHCO3 溶液(pH約8)(100 ml)攪拌,經過濾,經水沖洗且於真空烘箱中置於五氧化二磷上乾燥。因此,得到呈白色粉末之標的產物(1.77 g,78%)。GC-MS (EI) m/z 210.1。

中間體7
反式-4-(哌啶-1-基甲基)環己烷甲酸甲酯

令反式-4-甲醯基環己烷甲酸甲酯(Synthonix)(0.30 g,1.8毫莫耳)溶解於1,2-二氯乙烷(10 ml)並對該溶液加入哌啶(0.52 ml,5.3毫莫耳)和乙酸(0.19 ml,3.4毫莫耳)。令所生成之混合物冷卻至0℃並加入NaBH(OAc)3 (1.16 g,5.5毫莫耳)且令反應混合物於室溫下經攪拌16小時。隨後對該反應混合物加入水(30 ml)並使用Na2 CO3 溶液調整該混合物之pH至約9。令該混合物經二氯甲烷(20 ml)萃取2次,令合併之有機相置於無水硫酸鈉上乾燥並經過濾和濃縮。因此,得到標的產物(0.40 g,95%),其未經進一步純化而使用。

中間體8
反式-4-(哌啶-1-基甲基)環己烷甲醯肼

令反式-4-(哌啶-1-基甲基)環己烷甲酸甲酯(中間體7)(0.40 g,1.68毫莫耳)溶解於甲醇(5 ml)並將溶液倒入抗壓性玻璃反應器。加入水合肼(5 ml,100毫莫耳)並令反應混合物於75℃下經攪拌16小時。令該反應混合物經濃縮並使環己烷和無水甲苯蒸發以得到殘餘物。因此,得到呈白色粉末之標的產物(0.39 g,97%)。GC-MS (EI) m/z 239.2。

中間體9
反式-4-(吡咯啶-1-基羰基)環己烷甲醯肼

a) 反式-4-(吡咯啶-1-基羰基)環己烷甲酸甲酯

令反式-4-(甲氧基羰基)環己烷甲酸(Combi-Blocks Inc.)(186 mg,1毫莫耳)、吡咯啶(83.5 µl,1毫莫耳)、無水DMF (5 ml)、DIPEA (348 µl,2毫莫耳)、EDC (230 mg,1.2毫莫耳)及HOBt (162 mg,1.2毫莫耳)之混合物於室溫下經攪拌24小時。對反應混合物加入乙酸乙酯和NaHCO3 水溶液並分離相層且令水相經乙酸乙酯萃取1次。令合併之有機相經1N氫氯酸和水沖洗,置於無水硫酸鈉上乾燥,且經過濾和濃縮。因此,得到標的產物(180 mg,75%)。GC-MS (EI) m/z 239。

b) 反式-4-(吡咯啶-1-基羰基)環己烷甲醯肼
令反式-4-(吡咯啶-1-基羰基)環己烷甲酸甲酯(180 mg,0.75毫莫耳)、甲醇(1.1 ml)及水合肼(1.1 ml)於抗壓性玻璃反應器中且於75℃下經攪拌24小時。令反應混合物經濃縮並加入環己烷且隨後進行蒸發。因此,得到標的產物(183 mg,76%)。GC-MS (EI) m/z 239。

中間體10
反式-4-(嗎啉-1-基羰基)環己烷甲醯肼

a) 反式-4-(嗎啉-4-基羰基)環己烷甲酸甲酯

依據中間體9之步驟a)描述之方法,自反式-4-(甲氧基羰基)環己烷甲酸(Combi-Blocks Inc.)和嗎啉製備標的產物。GC-MS (EI) m/z 255。

b) 反式-4-(嗎啉-1-基羰基)環己烷甲醯肼
依據中間體9之步驟b)描述之方法,自反式-4-(嗎啉-4-基羰基)環己烷甲酸甲酯製備標的產物。GC-MS (EI) m/z 255。

中間體11
反式-4-(二甲基胺基)環己烷甲醯肼

依據中間體8描述之方法,自反式-4-(二甲基胺基)環己烷甲酸甲酯(EP 1 582 521 A1 (05.10.2005), TANABE SEIYAKU CO.)製備標的產物。MS (ESI) m/z 186.3 (M+H)+

中間體12
反式-4-(嗎啉-4-基)環己烷甲醯肼

依據中間體8描述之方法,自反式-4-(嗎啉-4-基)環己烷甲酸甲酯(EP 1 582 521 A1 (05.10.2005) TANABE SEIYAKU CO.)製備標的產物。GC-MS (EI) m/z 227。

中間體13
1-(嘧啶-2-基)四氫吖唉-3-甲醯肼

依據中間體8描述之方法,自1-(嘧啶-2-基)四氫吖唉-3-甲酸甲酯(WO 2006/124748 A2 (23.11.2006) LEXICON GENETICS INCORP.)製備標的產物。MS (ESI) m/z 194.2 (M+H)+

中間體14
1-(吡啶-2-基)四氫吖唉-3-甲醯肼

依據中間體8描述之方法,自1-(吡啶-2-基)四氫吖唉-3-甲酸甲酯(WO 2017/007756 A1 (12.01.2017) RODIN THERAPEUTICS INC.)製備標的產物。GC-MS (EI) m/z 192。

中間體15
(反式)-3-甲基-2-側氧基-1-氧雜-3-氮雜螺[4.5]癸烷-8-甲酸乙酯


中間體16
(順式)-3-甲基-2-側氧基-1-氧雜-3-氮雜螺[4.5]癸烷-8-甲酸乙酯

令60%氫化鈉之油分散液(1.8 g,45.0毫莫耳)懸浮於無水DMF (60 ml),經冷卻至0至5℃,且隨後逐滴加入溶解於DMF (60 ml)之(順式)-2-側氧基-1-氧雜-3-氮雜螺[4.5]癸烷-8-甲酸乙酯和(反式)-2-側氧基-1-氧雜-3-氮雜螺[4.5]癸烷-8-甲酸乙酯(WO 2008/092887 A1 (07.08.2008) GLAXO GROUP LTD.)的約1:1混合物(6.00 g,26.4毫莫耳),使得該混合物之溫度維持介於0至5℃之間。令該反應混合物於此溫度下經攪拌20分鐘,且隨後經20分鐘逐滴加入碘甲烷(2.46 ml,39.5毫莫耳)。令混合物於0至5℃下再經攪拌1小時,回溫至室溫且於該溫度下經攪拌3小時。隨後經10分鐘逐滴加入乙酸(1.8 ml,31毫莫耳),經攪拌15分鐘後,令反應混合物經濃縮且蒸發正庚烷(90 ml)2次以得到殘餘物。對該殘餘物加入乙酸乙酯(180 ml)、飽和NaHCO3 溶液(90 ml)及水(90 ml),分離相層,令有機相經NaCl (90 ml)沖洗,置於硫酸鈉上乾燥,且經過濾和濃縮。令殘餘物經管柱層析(使用甲苯:異丙醇=93:7作為溶析液)純化。令適當之餾分經濃縮並令殘餘物經二異丙醚結晶。因此,得到呈白色粉末之(反式)-3-甲基-2-側氧基-1-氧雜-3-氮雜螺[4.5]癸烷-8-甲酸乙酯(中間體15)(1.38 g,22%)和(順式)-3-甲基-2-側氧基-1-氧雜-3-氮雜螺[4.5]癸烷-8-甲酸乙酯(中間體16)(2.45 g,39%)。GC-MS (EI) m/z 241。

中間體17
(反式)-3-甲基-2-側氧基-1-氧雜-3-氮雜螺[4.5]癸烷-8-甲醯肼

依據中間體8描述之方法,自(反式)-3-甲基-2-側氧基-1-氧雜-3-氮雜螺[4.5]癸烷-8-甲酸乙酯(中間體15)製備標的產物。GC-MS (EI) m/z 227。

中間體18
(順式)-3-甲基-2-側氧基-1-氧雜-3-氮雜螺[4.5]癸烷-8-甲醯肼

依據中間體8描述之方法,自(順式)-3-甲基-2-側氧基-1-氧雜-3-氮雜螺[4.5]癸烷-8-甲酸乙酯(中間體16)製備標的產物。GC-MS (EI) m/z 227。

中間體19
反式-4-(四氫吖唉-1-基羰基)環己烷甲醯肼


a) 反式-4-(四氫吖唉-1-基羰基)環己烷甲酸甲酯

依據中間體9之步驟a)描述之方法,自反式-4-(甲氧基羰基)環己烷甲酸(Combi-Blocks Inc.)和四氫吖唉製備標的產物。GC-MS (EI) m/z 225。

b) 反式-4-(四氫吖唉-1-基羰基)環己烷甲醯肼
依據中間體9之步驟b)描述之方法,自反式-4-(四氫吖唉-1-基羰基)環己烷甲酸甲酯製備標的產物。GC-MS (EI) m/z 225。

中間體20
反式-4-(哌啶-1-基羰基)環己烷甲醯肼

a) 反式-4-(哌啶-1-基羰基)環己烷甲酸甲酯

依據中間體9之步驟a)描述之方法,自反式-4-(甲氧基羰基)環己烷甲酸(Combi-Blocks Inc.)和哌啶製備標的產物。GC-MS (EI) m/z 253。

b) 反式-4-(哌啶-1-基羰基)環己烷甲醯肼
依據中間體9之步驟b)描述之方法,自反式-4-(哌啶-1-基羰基)環己烷甲酸甲酯製備標的產物。GC-MS (EI) m/z 253。

中間體21
4-{[反式-4-(肼基羰基)環己基]羰基}哌嗪-1-甲酸三級丁酯

a) 4-{[反式-4-(甲氧基羰基)環己基]羰基}哌嗪-1-甲酸三級丁酯

依據中間體9之步驟a)描述之方法,自反式-4-(甲氧基羰基)環己烷甲酸(Combi-Blocks Inc.)和哌嗪-1-甲酸三級丁酯製備標的產物,其未經進一步純化而使用。

b) 反式-4-(嗎啉-1-基羰基)環己烷甲醯肼
依據中間體9之步驟b)描述之方法,自4-{[反式-4-(甲氧基羰基)-環己基]羰基}哌嗪-1-甲酸三級丁酯製備標的產物,其未經進一步純化而使用。

中間體22
7-氯-2-(甲硫基)-4,5-二氫-3H-1-苯並吖呯-4-甲酸甲酯

令7-氯-2-側硫基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯(中間體4)(250 mg,0.93毫莫耳)、丙酮(18.5 ml)、K2 CO3 (259 mg,1.87毫莫耳)及碘甲烷(0.18 ml,2.89毫莫耳)之溶液於室溫下經攪拌20小時。令反應混合物經濃縮,對殘餘物加入水並經乙酸乙酯萃取,令有機相置於無水硫酸鈉上乾燥且經過濾和濃縮。因此,得到標的產物(261 mg,99%)。MS (ESI) m/z 284.1 (M+H)+

中間體23
8-氯-5,6-二氫-4H -[1,2,4]三唑並[4,3-a ][1]苯並吖呯-5-甲酸甲酯

令7-氯-2-(甲硫基)-4,5-二氫-3H-1-苯並吖呯-4-甲酸甲酯(中間體22)(261 mg,0.92毫莫耳)溶解於1,4-二噁烷(6 ml)並令溶液溫熱至90℃。於氬氣下經4小時加入甲醯肼(277 mg,4.61毫莫耳)。隨後令反應混合物於90℃下經攪拌8小時並經冷卻至室溫後,於真空下蒸發溶劑。令殘餘物經管柱層析(使用二氯甲烷:甲醇=95:5作為溶析液)純化。因此,得到標的產物(188 mg,74%)。MS (ESI) m/z 278.1 (M+H)+

中間體24
1-溴-8-氯-5,6-二氫-4H -[1,2,4]三唑並[4,3-a ][1]苯並吖呯-5-甲酸甲酯

令8-氯-5,6-二氫-4H -[1,2,4]三唑並[4,3-a ][1]苯並吖呯-5-甲酸甲酯(中間體23)(187 mg,0.67毫莫耳)溶解於THF (15 ml)。加入N-溴琥珀醯亞胺(120 mg,0.68毫莫耳)並令所生成之淡黃色溶液於回流溫度且經RH-500型鹵素燈(Tracon Electric)照射下經攪拌60分鐘。此時,該溶液之顏色起初呈黑暗且隨後逐漸褪色。經冷卻至室溫後,於真空下蒸發溶劑。令殘餘物經管柱層析(使用環己烷:乙酸乙酯=1;1至1:4作為溶析液)純化。因此,得到標的化合物(226 mg,94%)。MS (ESI) m/z 358.0 (M+H)+

中間體25
7-甲基-2-側硫基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯

令7-甲基-2-側氧基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯(100 mg,0.43毫莫耳)(其係依據中間體3描述之方法製備)溶解於THF (10 ml),隨後加入勞森反應劑(105 mg,0.26毫莫耳)並令混合物於室溫下經攪拌20小時。令反應混合物首先經濃縮,隨後對殘餘物加入飽和NaHCO3 溶液且經攪拌60分鐘。令沉澱之產物經過濾,經水沖洗且經乾燥。因此,得到標的產物(80 mg,75%)。MS (ESI) m/z 250.2 (M+H)+

中間體26
7-甲氧基-2-側硫基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯

依據中間體25描述之方法,自7-甲氧基-2-側氧基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯(其係依據中間體3描述之方法製備)製備標的產物。MS (ESI) m/z 266.1 (M+H)+

中間體27
7-溴-2-側硫基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯

依據中間體25描述之方法,自7-溴-2-側氧基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯(其係依據中間體3描述之方法製備)製備標的產物。MS (ESI) m/z 316.0 (M+H)+

中間體28
7-溴-2-(甲硫基)-4,5-二氫-3H -1-苯並吖呯-4-甲酸甲酯

依據中間體22描述之方法,自7-溴-2-側硫基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯(中間體27)製備標的產物。MS (ESI) m/z 330.1 (M+H)+

中間體29
1-溴-8-氯-5,6-二氫-4H -[1,2,4]三唑並[4,3-a ][1]苯並吖呯-5-羧醯胺

a) 1-溴-8-氯-5,6-二氫-4H -[1,2,4]三唑並[4,3-a ][1]苯並吖呯-5-甲酸

令1-溴-8-氯-5,6-二氫-4H -[1,2,4]三唑並[4,3-a ][1]苯並吖呯-5-甲酸甲酯(中間體24)(631 mg,1.77毫莫耳)、THF (13 ml)、甲醇(6.5 ml)、水(6.5 ml)及氫氧化鋰水合物(379 mg,9.03毫莫耳)之混合物於室溫下經攪拌2.5小時,隨後蒸發溶劑且令殘留之水溶液經水(10 ml)稀釋。令溶液之pH經1M氫氯酸調整至5,令沉澱之產物經過濾且經水沖洗。令母液經乙酸乙酯萃取,令結合之有機層經鹽水沖洗,置於硫酸鈉上乾燥且經過濾和濃縮。令所得之殘餘物與過濾產物結合以生成標的化合物(433 mg,71%)。MS (ESI) m/z 343.9 (M+H)+

b) 1-溴-8-氯-5,6-二氫-4H -[1,2,4]三唑並[4,3-a ][1]苯並吖呯-5-羧醯胺
對1-溴-8-氯-5,6-二氫-4H -[1,2,4]三唑並[4,3-a ][1]苯並吖呯-5-甲酸(中間體29之步驟a))(620 mg,1.8毫莫耳)、DMF (8 ml)、HOBt (498 mg,3.25毫莫耳)、DIPEA (0.88 ml,5.06毫莫耳)及EDC (623 mg,3.25毫莫耳)之冰冷混合物加入氯化銨(290 mg,5.48毫莫耳)並令反應混合物於5℃下經攪拌0.5小時且隨後於室溫下再經攪拌20小時。經反應完成後,令混合物經水和鹽水稀釋並經乙酸乙酯萃取。令結合之有機層置於硫酸鈉上乾燥並經過濾和濃縮。令殘餘物自乙酸乙酯結晶析出以生成標的化合物(428 mg,69%)。MS (ESI) m/z 343.0 (M+H)+

中間體30
(5s ,8s )-1-側氧基-2-(丙-2-基)-2-氮雜螺[4.5]癸烷-8-甲酸甲酯


中間體31
(5r ,8r )-1-側氧基-2-(丙-2-基)-2-氮雜螺[4.5]癸烷-8-甲酸甲酯

令反式-1-(2-側氧基乙基)環己烷-1,4-二甲酸二甲酯(WO 2011/143150 A1, (05.10.2011) SANOFI)(0.9 g,4.0毫莫耳)、1,2-二氯乙烷(40 ml)、異丙胺(316 µl,3.71毫莫耳)及乙酸(637 µl,11.1毫莫耳)之攪拌混合物冷卻至5℃並對反應混合物加入三乙醯氧基氫硼化鈉(2.36 g,11.1毫莫耳),其中該加入速率使內部溫度維持低於5℃。經加入完成後,令反應混合物於室溫下經攪拌2小時並隨後經水稀釋。藉由加入10% K2 CO3 溶液將混合物之pH調整至8,分離相層且令水相經二氯甲烷萃取。令合併之有機相連續經10% K2 CO3 溶液、水及鹽水沖洗,置於硫酸鈉上乾燥且經過濾和濃縮。令殘餘物溶解於無水THF (40 ml)並加入三級丁氧化鉀(330 mg,2.94毫莫耳)。令反應混合物於室溫下經攪拌3小時並隨後加入CO2 固體進行中和。經加入水後,蒸發THF並令水相經乙酸乙酯萃取。令有機相置於硫酸鈉上乾燥並經過濾和濃縮。令殘餘物經快速管柱層析(使用環己烷:乙酸乙酯=45:55之混合物作為溶析液)純化以生成為第一餾分之(5s ,8s )-1-側氧基-2-(丙-2-基)-2-氮雜螺[4.5]癸烷-8-甲酸甲酯(中間體30)(56 mg,6%)和為第二餾分之(5r ,8r )-1-側氧基-2-(丙-2-基)-2-氮雜螺[4.5]癸烷-8-甲酸甲酯(中間體31)(172 mg,19%)。GC-MS (EI) m/z 253。

中間體32
(5r ,8r )-1-側氧基-2-(丙-2-基)-2-氮雜螺[4.5]癸烷-8-甲醯肼

依據中間體9之步驟b)描述之方法,自(5r ,8r )-1-側氧基-2-(丙-2-基)-2-氮雜螺[4.5]癸烷-8-甲酸甲酯(中間體31)製備標的化合物。GC-MS (EI) m/z 253。

中間體33
4-(1-{[三級丁基(二甲基)矽基]氧}乙基)-7-氯-1,3,4,5-四氫-2H -1-苯並吖呯-2-酮


a) 7-氯-2-側氧基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸

令7-氯-2-側氧基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯(中間體3)(12.38 g,48.8毫莫耳)、甲醇(55 ml)及50% NaOH溶液(12 ml)之混合物於室溫下經攪拌0.5小時並隨後冷卻至0℃且加入5%氫氯酸溶液(105 ml)。於0℃下經攪拌10分鐘後,令沉澱之產物經過濾,經水沖洗且經乾燥以生成標的化合物(11.1 g,95%)。MS (ESI) m/z 240.2 (M+H)+

b) 7-氯-N -甲氧基-N -甲基-2-側氧基-2,3,4,5-四氫-1H -1-苯並吖呯-4-羧醯胺

依據中間體9之步驟a)描述之方法,自7-氯-2-側氧基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸(中間體33之步驟a))和N-甲氧基甲胺鹽酸鹽製備標的化合物,其未經進一步純化而用於下一個步驟。

c) 4-乙醯基-7-氯-1,3,4,5-四氫-2H -1-苯並吖呯-2-酮

於氬氣下令7-氯-N -甲氧基-N -甲基-2-側氧基-2,3,4,5-四氫-1H -1-苯並吖呯-4-羧醯胺(中間體33之步驟b))(2.5 g,8.84毫莫耳)於無水THF (100 ml)之混合物冷卻至-15℃並加入3M甲基溴化鎂之二乙醚溶液(14.7 ml,44.1毫莫耳)。令反應混合物於-10℃至0℃下經攪拌3小時,隨後經藉由加入飽和NH4 Cl溶液驟冷且經乙酸乙酯萃取。令結合之有機層經鹽水沖洗,置於硫酸鈉上乾燥且經過濾和濃縮。令殘餘物經快速管柱層析(使用二氯甲烷:甲醇=96:4作為溶析液)純化以生成標的化合物(1.6 g,76%)。MS (ESI) m/z 238.2 (M+H)+

d) 7-氯-4-(1-羥基乙基)-1,3,4,5-四氫-2H -1-苯並吖呯-2-酮

令4-乙醯基-7-氯-1,3,4,5-四氫-2H -1-苯並吖呯-2-酮(中間體33之步驟c))(300 mg,1.26毫莫耳)、乙醇(50 ml)及NaBH4 (96 mg,2.5毫莫耳)之混合物於室溫下經攪拌0.5小時,隨後經藉由加入1M氫氯酸溶液驟冷且蒸發乙醇。令水相經乙酸乙酯萃取,令結合之有機層經鹽水沖洗,置於硫酸鈉上乾燥且經過濾和濃縮。令殘餘物經快速管柱層析(使用二氯甲烷:甲醇=95:5作為溶析液)純化以生成呈63:37非鏡像異構物的混合物之標的化合物(235 mg,78%)。MS (ESI) m/z 240.2 (M+H)+

e) 4-(1-{[三級丁基(二甲基)矽基]氧}乙基)-7-氯-1,3,4,5-四氫-2H -1-苯並吖呯-2-酮
令7-氯-4-(1-羥基乙基)-1,3,4,5-四氫-2H -1-苯並吖呯-2-酮(中間體33之步驟d))(665 mg,2.77毫莫耳)、DMF (5 ml)、二氯甲烷(50 ml)及2,6-二甲基吡啶(0.8 ml,6.94毫莫耳)之攪拌混合物冷卻至0℃並於氬氣下加入三氟甲烷磺酸三級丁基二甲基矽烷酯(792 mg,3.0毫莫耳)。令反應混合物回溫至室溫並於該溫度下經攪拌16小時。蒸發二氯甲烷,加入水(10 ml)和飽和NaHCO3 溶液(10 ml)並令混合物經乙酸乙酯萃取。令結合之有機層經鹽水沖洗,置於硫酸鈉上乾燥且經過濾和濃縮。令殘餘物經快速管柱層析(使用環己烷:乙酸乙酯=75:25作為溶析液)純化。第一餾分係非鏡像異構物之一(200 mg,20%),第二餾分係非鏡像異構物之混合物(191 mg,19%)且第三餾分係其他非鏡像異構物(326 mg,33%)。MS (ESI) m/z 354.2 (M+H)+

中間體34
4-(1-{[三級丁基(二甲基)矽基]氧}乙基)-7-氯-1,3,4,5-四氫-2H -1-苯並吖呯-2-硫酮

依據中間體4描述之方法,自4-(1-{[三級丁基(二甲基)矽基]氧}乙基)-7-氯-1,3,4,5-四氫-2H -1-苯並吖呯-2-酮(中間體33之步驟e))製備標的化合物,其未經進一步純化而用於下一個步驟。對於非鏡像異構物之混合物(中間體33之步驟e)的第二餾分)或獨立地非鏡像異構物(中間體33之步驟e)的第一餾分和第三餾分)使用相同之方法。

中間體35
4-甲氧基-4-甲基環己烷甲醯肼

a) 4-羥基-4-甲基環己烷甲酸乙酯

於氬氣下對2M三甲基鋁之甲苯攪拌溶液(100 ml,220毫莫耳)於-60℃下經2.5小時加入4-側氧基環己烷甲酸乙酯(8.7 ml,55毫莫耳)之甲苯(50 ml)溶液。經該加入完成後,令混合物於-60℃下經攪拌0.5小時且隨後經2小時回溫至-20℃。於同時維持內部溫度低於10℃下,藉由插管經25至30分鐘將反應混合物轉移至乙酸乙酯(180 ml)、水(425 ml)、濃氫氯酸(75 ml)及碎冰(100 g)之冰冷混合物。分離相層,令有機相連續經水(400 ml)和鹽水(400 ml)沖洗,置於硫酸鈉上乾燥且經過濾和濃縮以生成標的化合物(5.44 g,53%)。依據1 HNMR光譜,該化合物係順式和反式異構物之28:72混合物。該混合物未經進一步純化而用於下一個步驟。

b) 4-甲氧基-4-甲基環己烷甲酸乙酯

於氬氣下對60%氫化鈉之礦物油溶液(2.16 g,54毫莫耳)、無水THF (34 ml)、四丁基碘化銨(200 mg,0.54毫莫耳)、咪唑(49 mg,0.72毫莫耳)及碘甲烷(3.36 ml,54毫莫耳)之攪拌溶液於20至25℃下經30至40分鐘加入4-羥基-4-甲基環己烷甲酸乙酯(中間體35之步驟a))(3.36 g,18毫莫耳)之無水THF (21 ml)溶液。令反應混合物於室溫下經攪拌3小時,隨後冷卻至0至5℃且經10分鐘加入乙酸(2.28 ml,40毫莫耳)。令混合物經攪拌15分鐘,隨後倒入至二乙醚(280 ml)和飽和NaHCO3 溶液(120 ml)之混合物。分離相層,令有機相經鹽水沖洗,置於硫酸鈉上乾燥且經過濾和濃縮。令殘餘物經管柱層析(使用正己烷:乙酸乙酯=85:15作為溶析液)純化以生成標的化合物(2.3 g,64%)。依據1 HNMR光譜,該化合物係順式和反式異構物之21:79混合物。

c) 4-甲氧基-4-甲基環己烷甲醯肼
依據中間體9之步驟b)描述之方法,自4-甲氧基-4-甲基環己烷甲酸乙酯(中間體35之步驟b))製備標的化合物。依據1 HNMR光譜,該化合物係順式和反式異構物之21:79混合物。

中間體36
7-氯-4-(氟甲基)-1,3,4,5-四氫-2H -1-苯並吖呯-2-酮

a) 7-氯-4-(羥基甲基)-1,3,4,5-四氫-2H -1-苯並吖呯-2-酮

於氬氣下令7-氯-2-側氧基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯(中間體3)(500 mg,1.97毫莫耳)、無水二乙醚(15 ml)及無水THF (20 ml)之攪拌混合物冷卻至-40℃並加入1M氫化鋁鋰之二乙醚溶液(4.33 ml,4.33毫莫耳)。令反應混合物於-20℃下經攪拌1小時且隨後回溫至0℃並小心地加入水。蒸發THF並令水相經乙酸乙酯萃取,令合併之有機相經鹽水沖洗,置於硫酸鈉上乾燥且經過濾和濃縮以生成標的化合物(422 mg,95%)。MS (ESI) m/z 226.1 (M+H)+

b) 7-氯-4-(氟甲基)-1,3,4,5-四氫-2H -1-苯並吖呯-2-酮
於氬氣下令7-氯-4-(羥基甲基)-1,3,4,5-四氫-2H -1-苯並吖呯-2-酮(中間體36之步驟a))(532 mg,2.36毫莫耳)及無水二氯甲烷(50 ml)之攪拌混合物冷卻至-78℃並逐滴加入(二乙基胺基)三氟化硫(950 mg,5.89毫莫耳)之二氯甲烷(1 ml)溶液。令反應混合物經4小時回溫至0℃,於該溫度下經攪拌45分鐘且隨後加入飽和NaHCO3 溶液。分離相層,令水相經二氯甲烷萃取,令合併之有機相經鹽水沖洗,置於硫酸鈉上乾燥且經過濾和濃縮。令殘餘物經逆相快速管柱層析(使用水(含有0.1%三氟乙酸):乙腈=60:40作為溶析液)純化以生成標的化合物(222 mg,41%)。MS (ESI) m/z 228.1 (M+H)+

中間體37
7-氯-4-(氟甲基)-1,3,4,5-四氫-2H -1-苯並吖呯-2-硫酮

依據中間體4描述之方法,自7-氯-4-(氟甲基)-1,3,4,5-四氫-2H -1-苯並吖呯-2-酮(中間體36之步驟b))製備標的化合物,其未經進一步純化而用於下一個步驟。

中間體38
7-氟-2-側氧基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯

a) 7-氟-4-(羥基亞胺基)-1,2,3,4-四氫萘-2-甲酸甲酯

依據中間體2描述之方法,自7-氟-4-側氧基-1,2,3,4-四氫萘-2-甲酸甲酯(US5595872 (03.09.1993) Bristol-Myers Squibb Company)製備標的化合物,其未經進一步純化而用於下一個步驟。

b) 7-氟-2-側氧基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯
依據中間體3描述之方法,自7-氟-4-(羥基亞胺基)-1,2,3,4-四氫萘-2-甲酸甲酯(中間體38之步驟a))製備標的化合物。MS (ESI) m/z 238.2 (M+H)+

中間體39
7-氟-2-側硫基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯

依據中間體4描述之方法,自7-氟-2-側氧基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯(中間體38之步驟b))製備標的化合物,其未經進一步純化而用於下一個步驟。

中間體40
7-氟-4-(氟甲基)-1,3,4,5-四氫-2H -1-苯並吖呯-2-酮

a) 7-氟-4-(羥基甲基)-1,3,4,5-四氫-2H -1-苯並吖呯-2-酮

依據中間體36之步驟a)描述之方法,自7-氟-2-側氧基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯(中間體38之步驟b))製備標的化合物,其未經進一步純化而用於下一個步驟。

b) 7-氟-4-(氟甲基)-1,3,4,5-四氫-2H -1-苯並吖呯-2-酮
依據中間體36之步驟b)描述之方法,自7-氟-4-(羥基甲基)-1,3,4,5-四氫-2H -1-苯並吖呯-2-酮(中間體40之步驟a))製備標的化合物,其未經進一步純化而用於下一個步驟。

中間體41
7-氟-4-(氟甲基)-1,3,4,5-四氫-2H -1-苯並吖呯-2-硫酮

依據中間體4描述之方法,自7-氟-4-(氟甲基)-1,3,4,5-四氫-2H -1-苯並吖呯-2-酮(中間體40之步驟b))製備標的化合物,其未經進一步純化而用於下一個步驟。

中間體42
4-(氟甲基)-7-甲基-1,3,4,5-四氫-2H -1-苯並吖呯-2-酮

a) 4-(羥基甲基)-7-甲基-1,3,4,5-四氫-2H -1-苯並吖呯-2-酮

依據中間體36之步驟a)描述之方法,自7-甲基-2-側氧基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯(依據中間體3描述之方法製備)製備標的化合物,其未經進一步純化而用於下一個步驟。

b) 4-(氟甲基)-7-甲基-1,3,4,5-四氫-2H -1-苯並吖呯-2-酮
依據中間體36之步驟b)描述之方法,自4-(羥基甲基)-7-甲基-1,3,4,5-四氫-2H -1-苯並吖呯-2-酮(中間體42之步驟a))製備標的化合物。MS (ESI) m/z 208.2 (M+H)+

中間體43
4-(氟甲基)-7-甲基-1,3,4,5-四氫-2H -1-苯並吖呯-2-硫酮

依據中間體4描述之方法,自4-(氟甲基)-7-甲基-1,3,4,5-四氫-2H -1-苯並吖呯-2-酮(中間體42之步驟b))製備標的化合物,其未經進一步純化而用於下一個步驟。

中間體44
7-氟-2-(甲硫基)-4,5-二氫-3H -1-苯並吖呯-4-甲酸甲酯

依據中間體22描述之方法,自7-氟-2-側硫基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯(中間體39)製備標的化合物。MS (ESI) m/z 268.1 (M+H)+

中間體45
7-氯-4-甲基-2-側氧基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯

a) 7-氯-1-(4-甲氧基苄基)-2-側氧基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯

令7-氯-2-側氧基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯(中間體3)(1.0 g,3.94毫莫耳)於無水DMF (10 ml)之攪拌混合物冷卻至0℃並於氬氣下加入60%氫化鈉(205 mg,5.12毫莫耳)之礦物油溶液。令混合物於此溫度下經攪拌1小時,隨後加入4-甲氧基苄基氯(0.8 ml,5.9毫莫耳)且令反應混合物於室溫下經攪拌20小時。經反應完成後,令混合物經水和鹽水稀釋,經乙酸乙酯萃取,令結合之有機層經鹽水沖洗,置於硫酸鈉上乾燥且經過濾和濃縮。令殘餘物經快速管柱層析(使用環己烷:乙酸乙酯=4:1作為溶析液)純化以生成標的化合物(820 mg,56%)。MS (ESI) m/z 374.1 (M+H)+

b) 7-氯-1-(4-甲氧基苄基)-4-甲基-2-側氧基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯

令7-氯-1-(4-甲氧基苄基)-2-側氧基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯(中間體45之步驟a))(600 mg,1.61毫莫耳)於無水THF (30 ml)之攪拌溶液冷卻至-70℃並於氬氣下加入2.0 M二異丙基胺化鋰之THF/庚烷/乙基苯溶液(1.2 ml,2.4毫莫耳)。令混合物於此溫度下經攪拌2小時,隨後加入碘甲烷(0.2 ml,3.2毫莫耳)且令反應混合物經2小時回溫至0℃。經反應完成後,令混合物經藉由加入飽和氯化銨驟冷,經乙酸乙酯萃取,令結合之有機層經鹽水沖洗,置於硫酸鈉上乾燥且經過濾和濃縮。令殘餘物經快速管柱層析(使用環己烷:乙酸乙酯=4:1作為溶析液)純化以生成標的化合物(460 mg,74%)。MS (ESI) m/z 388.1 (M+H)+

c) 7-氯-4-甲基-2-側氧基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯
令7-氯-1-(4-甲氧基苄基)-4-甲基-2-側氧基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯(中間體45之步驟b))(460 mg,1.2毫莫耳)、乙腈(20 ml)、水(5 ml)及硝酸鈰(IV)銨(1.95 g,3.56毫莫耳)之混合物於室溫下經攪拌2小時且隨後經水稀釋。藉由加入飽和NaHCO3 溶液將混合物之pH調整至8並隨後經乙酸乙酯萃取。令結合之有機層經鹽水沖洗,置於硫酸鈉上乾燥且經過濾和濃縮。令殘餘物經快速管柱層析(使用環己烷:乙酸乙酯=4:1作為溶析液)純化以生成標的化合物(190 mg,60%)。MS (ESI) m/z 268.1 (M+H)+

中間體46
7-氯-4-甲基-2-側硫基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯

依據中間體25描述之方法,自7-氯-4-甲基-2-側氧基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯(中間體45之步驟c))製備標的化合物,其未經進一步純化而用於下一個步驟。

中間體47
7-氯-4-乙基-2-側氧基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯


a) 7-氯-1-(4-甲氧基苄基)-4-乙基-2-側氧基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯

依據中間體45之步驟b)描述之方法,自7-氯-1-(4-甲氧基苄基)-2-側氧基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯(中間體45之步驟a))和碘乙烷製備標的化合物,其未經進一步純化而用於下一個步驟。

b) 7-氯-4-乙基-2-側氧基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯
依據中間體45之步驟c)描述之方法,自7-氯-1-(4-甲氧基苄基)-4-乙基-2-側氧基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯(中間體47之步驟a))製備標的化合物。MS (ESI) m/z 282.1 (M+H)+

中間體48
7-氯-4-乙基-2-側硫基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯

依據中間體25描述之方法,自7-氯-4-乙基-2-側氧基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯(中間體47之步驟b))製備標的化合物,其未經進一步純化而用於下一個步驟。

中間體49
7-氯-4-乙基-2-(甲硫基)-4,5-二氫-3H -1-苯並吖呯-4-甲酸甲酯

依據中間體22描述之方法,自7-氯-4-乙基-2-側硫基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯(中間體48)製備標的化合物,其未經進一步純化而用於下一個步驟。

中間體50
2-[(3,3-二氟環丁基)羰基]肼甲酸三級丁酯

依據中間體5描述之方法,自3,3-二氟環丁烷甲酸(Combi-Blocks Inc.)製備標的化合物。GC-MS (EI) m/z 250.1。

中間體51
3,3-二氟環丁烷甲醯肼

依據中間體6描述之方法,自2-[(3,3-二氟環丁基)羰基]肼甲酸三級丁酯(中間體50)製備標的化合物。GC-MS (EI) m/z 150.1。

中間體52
1-溴-8-氯-5,6-二氫-4H -[1,2,4]三唑並[4,3-a ][1]苯並吖呯-5-腈

令1-溴-8-氯-5,6-二氫-4H -[1,2,4]三唑並[4,3-a ][1]苯並吖呯-5-羧醯胺(中間體29之步驟b))(155 mg,0.45毫莫耳)、無水二氯甲烷(77 ml)及三乙胺(400 µl,2.86毫莫耳)之攪拌溶液冷卻至0℃並加入三氟乙酸酐(105 µl,0.73毫莫耳)。令混合物之溫度回溫至室溫並令反應混合物於此溫度下經攪拌1小時,隨後冷卻至0℃並加入三乙胺(400 µl,2.86毫莫耳)和三氟乙酸酐(105 µl,0.73毫莫耳)。令混合物於室溫下經攪拌2.5小時且隨後經飽和NaHCO3 溶液稀釋。分離相層並令水相經二氯甲烷萃取。令合併之有機相經水和鹽水沖洗,置於硫酸鈉上乾燥且經過濾和濃縮。令殘餘物經管柱層析(使用二氯甲烷:甲醇:氫氧化銨=180:10:1作為溶析液)純化以生成標的化合物(125 mg,85%)。MS (ESI) m/z 323.0 (M+H)+

中間體53
7-氟-4,4-二甲基-1,3,4,5-四氫-2H -1-苯並吖呯-2-酮

a) [1-(3-氟苯基)-2-甲基丙-2-基]丙二酸二乙酯

於氬氣下藉由滴漏斗經4小時將0.5M 3-氟苄基氯化鎂(Alfa Aesar)之2-Me-THF溶液(33 ml,16.5毫莫耳)緩慢加入至(丙-2-亞基)丙二酸二乙酯(Aldrich)(3.0 g,15.0毫莫耳)之二乙醚(150 ml)溶液。隨後於室溫下持續攪拌18小時。隨後令反應物經1M氫氯酸溶液驟冷,經乙酸乙酯萃取,令合併之有機相經飽和NaCl溶液沖洗,置於無水硫酸鈉上乾燥且經過濾和濃縮。因此,得到呈黃色油之標的產物(4.30 g,92.5%)。MS (ESI) m/z 311.1 (M+H)+

b) [1-(3-氟苯基)-2-甲基丙-2-基]丙二酸

令[1-(3-氟苯基)-2-甲基丙-2-基]丙二酸二乙酯(4.30 g,13.9毫莫耳)、2M NaOH水溶液(60 ml,120毫莫耳)及乙醇(60 ml)之溶液於50℃下經隔夜攪拌。真空下除去有機溶劑,令殘餘物經3M氫氯酸酸化且經二氯甲烷萃取,置於無水硫酸鈉上乾燥且經過濾和濃縮。因此,得到呈微褐色油之標的產物(3.43 g,97%)。MS (ESI) m/z 255.1 (M+H)+

c) 4-(3-氟苯基)-3,3-二甲基丁酸

令[1-(3-氟苯基)-2-甲基丙-2-基]丙二酸(3.43 g,13.5毫莫耳)於210℃下經加熱直至氣體產生中止且隨後冷卻至室溫。因此,得到呈油狀之標的產物(2.35 g,83%)。MS (ESI) m/z 211.1 (M+H)+

d) 6-氟-3,3-二甲基-3,4-二氫萘-1(2H)-酮

將濃硫酸(8 ml)加入至4-(3-氟苯基)-3,3-二甲基丁酸(2.35 g,11.2毫莫耳)並令所生成之混合物於40℃下經攪拌1小時。隨後倒至冰上,經乙酸乙酯萃取,經飽和NaCl溶液沖洗,置於無水硫酸鈉上乾燥且經過濾和濃縮。因此,得到呈微褐色油之標的產物(1.51 g,70%)。MS (ESI) m/z 193.4 (M+H)+

e) N-(6-氟-3,3-二甲基-3,4-二氫萘-1(2H)-亞基)羥基胺

令6-氟-3,3-二甲基-3,4-二氫萘-1(2H)-酮(1.51 g,7.86毫莫耳)、羥基胺鹽酸鹽(Aldrich)(3.01 g,43.3毫莫耳)、無水乙酸鈉(Aldrich)(3.61 g,44.0毫莫耳)及甲醇(55 ml)之溶液經隔夜回流。隨後真空下除去溶劑,加入水和乙酸乙酯,分離相層,令水相經乙酸乙酯萃取,令合併之有機相經飽和NaCl溶液沖洗,置於無水硫酸鈉上乾燥且經過濾和濃縮。因此,得到呈米色固體之標的產物(1.45 g,89%)。MS (ESI) m/z 208.1 (M+H)+

f) 7-氟-4,4-二甲基-1,3,4,5-四氫-2H-1-苯並吖呯-2-酮
將N-(6-氟-3,3-二甲基-3,4-二氫萘-1(2H )-亞基)羥基胺(1.45 g,7.00毫莫耳)加入至多磷酸(15.0 g)並於130℃下經攪拌20分鐘。將冰加入至反應混合物並經攪拌10分鐘。隨後經氯仿萃取,置於無水硫酸鈉上乾燥且經過濾和濃縮。令殘餘物經二乙醚磨碎並藉由抽氣收集沉澱物。因此,得到呈白色晶體之標的產物(1.10 g,76%)。MS (ESI) m/z 208.2 (M+H)+

中間體54
7-氟-4-(丙-2-基)-1,3,4,5-四氫-2H -1-苯並吖呯-2-酮

依據中間體53描述之方法,自2-(2-甲基亞丙基)丙二酸二乙酯(Combi Blocks)製備標的產物。MS (ESI) m/z 222.1 (M+H)+

中間體55
1,5-二氫螺[1-苯並吖呯-4,1’-環戊烷]-2(3H)-酮

a) (1-苄基環戊基)丙二酸二乙酯

於氬氣下藉由滴漏斗經4小時將2M苄基氯化鎂之THF (Aldrich)溶液(12 ml,24毫莫耳)緩慢加入至2-亞環戊基丙二酸二乙酯(Lehnert, W.,Tetrahedron 1973,29 :635-638) (5.0 g,22.1毫莫耳)之二乙醚(100 ml)溶液。隨後於室溫下持續攪拌18小時。隨後令反應物經1M氫氯酸溶液驟冷,經乙酸乙酯萃取,令合併之有機相經飽和NaCl溶液沖洗,置於無水硫酸鈉上乾燥且經過濾和濃縮。因此,得到呈油狀之標的產物(5.62 g,80%)。MS (ESI) m/z 319.4 (M+H)+
b) (1-苄基環戊基)丙二酸

令(1-苄基環戊基)丙二酸二乙酯(中間體55之步驟a))(5.62 g,17.7毫莫耳)、2M NaOH水溶液(60 ml,120毫莫耳)及乙醇(30 ml)之溶液經回流2小時。真空下除去有機溶劑,令殘餘物經3M氫氯酸酸化並經乙酸乙酯萃取,令合併之有機相經飽和NaCl溶液沖洗,置於無水硫酸鈉上乾燥且經過濾和濃縮。因此,得到呈油狀之標的產物(3.52 g,76%)。MS (ESI) m/z 263.3 (M+H)+

c) (1-苄基環戊基)乙酸

令(1-苄基環戊基)丙二酸(中間體55之步驟b))(3.52 g,13.4毫莫耳)於180℃下經加熱直至氣體產生中止並隨後冷卻至室溫。因此,得到呈油狀之標的產物(2.90 g,99%)。MS (ESI) m/z 219.1 (M+H)+

d) 1’H -螺[環戊烷-1,2’-萘]-4’(3’H )-酮

將濃硫酸(Aldrich)(20 ml)加入至(1-苄基環戊基)乙酸(中間體55之步驟c))(2.90 g,13.3毫莫耳)並令所生成之混合物於35℃下經攪拌1小時。隨後倒至冰上,經乙酸乙酯萃取,令合併之有機相經飽和NaCl溶液沖洗,置於無水硫酸鈉上乾燥且經過濾和濃縮。將殘餘物置入二氯甲烷並經二氧化矽墊過濾以除去有色雜質。因此,得到呈油狀之標的產物(1.41 g,53%)。MS (ESI) m/z 201.2 (M+H)+

e) N-1’H -螺[環戊烷-1,2’-萘]-4’(3’H )-亞基羥基胺

令1’H -螺[環戊烷-1,2’-萘]-4’(3’H )-酮(中間體55之步驟d))(1.41 g,7.05毫莫耳)、羥基胺鹽酸鹽(Aldrich)(1.95 g,28毫莫耳)、無水乙酸鈉(Aldrich)(2.30 g,28毫莫耳)及甲醇(40 ml)之溶液經回流2小時。隨後真空下除去溶劑,加入水和乙酸乙酯,分離相層,令水相經乙酸乙酯萃取,令合併之有機相經飽和NaCl溶液沖洗,置於無水硫酸鈉上乾燥且經過濾和濃縮。因此,得到呈米色固體之標的產物(1.33 g,88%)。MS (ESI) m/z 216.1 (M+H)+

f) 1,5-二氫螺[1-苯並吖呯-4,1’-環戊烷]-2(3H)-酮
將N-1’H -螺[環戊烷-1,2’-萘]-4’(3’H )-亞基羥基胺(中間體55之步驟e))(1.33 g,6.19毫莫耳)加入至多磷酸(20.0 g)並於110℃下經攪拌20分鐘。將冰加入至反應混合物並經攪拌10分鐘。隨後經乙酸乙酯萃取,令合併之有機相經飽和NaCl溶液沖洗,置於無水硫酸鈉上乾燥且經過濾和濃縮。令殘餘物經管柱層析(使用環己烷:乙酸乙酯=2:1作為溶析液)純化。因此,得到呈白色晶體之標的產物(0.51 g,38%)。MS (ESI) m/z 216.1 (M+H)+

中間體56
4-(丙-2-基)-1,3,4,5-四氫-2H -1-苯並吖呯-2-酮

依據中間體53描述之方法,自2-(2-甲基亞丙基)丙二酸二乙酯(Combi Blocks)製備標的產物。MS (ESI) m/z 204.1 (M+H)+

中間體57
1,5-二氫螺[1-苯並吖呯-4,4’-環氧乙烷]-2(3H )-酮

依據中間體53描述之方法,自四氫-4H -哌喃-4-亞基丙二酸二乙酯(Griffiths et al.,HeIv Chim Acta 1991,74 (2): 09-314)製備標的產物。MS (ESI) m/z 232.2 (M+H)+

中間體58
1,4’,5,5’-四氫螺[1-苯並吖呯-4,3’-呋喃]-2(3H )-酮

依據中間體53描述之方法,自二氫呋喃-3(2H)-亞基丙二酸二乙酯(Griffiths et al.,HeIv Chim Acta 1991,74 (2): 09-314)製備標的產物。MS (ESI) m/z 218.2 (M+H)+
中間體59
1,5,5’,6’-四氫-4’H -螺[1-苯並吖呯-4,3’-哌喃]-2(3H )-酮

依據中間體53描述之方法,自二氫-2H-哌喃-3(4H)-亞基丙二酸二乙酯(Griffiths et al.,HeIv Chim Acta 1991,74 2): 09-314)製備標的產物。MS (ESI) m/z 232.1 (M+H)+

中間體60
7-溴-1,5-二氫螺[1-苯並吖呯-4,1’-環戊烷]-2(3H )-酮

對1,5-二氫螺[1-苯並吖呯-4,1’-環戊烷]-2(3H )-酮(中間體55)(0.51 g,2.37毫莫耳)、乙酸(10 ml)及水(2 ml)之溶液緩慢加入溴(Aldrich)(0.121 ml,2.37毫莫耳)之乙酸(3 ml)溶液。經於室溫下攪拌反應混合物1小時後,倒至冰上,藉由抽氣過濾沉澱物且經乾燥。因此,得到呈米色晶體之標的產物(0.50 g,72%)。MS (ESI) m/z 294.2 & 296.1 (M+H)+

中間體61
7-溴-4-(丙-2-基)-1,3,4,5-四氫-2H -1-苯並吖呯-2-酮

依據中間體60描述之方法,自4-(丙-2-基)-1,3,4,5-四氫-2H -1-苯並吖呯-2-酮(中間體56)製備標的產物。MS (ESI) m/z 282.1 & 284.1 (M+H)+

中間體62
7-溴-1,5-二氫螺[1-苯並吖呯-4,4’-環氧乙烷]-2(3H )-酮

依據中間體60描述之方法,自1,5-二氫螺[1-苯並吖呯-4,4’-環氧乙烷]-2(3H )-酮(中間體57)製備標的產物。MS (ESI) m/z 310.1 & 312.1 (M+H)+

中間體63
7-溴-1,5-二氫螺[1-苯並吖呯-4,3’-四氫呋喃]-2(3H )-酮

依據中間體60描述之方法,自1,4’,5,5’-四氫螺[1-苯並吖呯-4,3’-呋喃]-2(3H )-酮(中間體58)製備標的產物。MS (ESI) m/z 296.2 & 298.2 (M+H)+

中間體64
7-氯-1,5-二氫螺[1-苯並吖呯-4,4’-環氧乙烷]-2(3H )-酮

對1,5-二氫螺[1-苯並吖呯-4,4’-環氧乙烷]-2(3H )-酮(中間體57)(0.73 g,3.16毫莫耳)之DMF (20 ml)溶液加入N-氯琥珀醯亞胺(0.59 g,4.42毫莫耳)。於室溫下經攪拌反應混合物1小時後,倒至冰上,藉由抽氣過濾沉澱物且經乾燥。因此,得到呈黃色晶體之標的產物(0.58 g,69%)。MS (ESI) m/z 266.1 (M+H)+

中間體65
7-氯-1,5-二氫螺[1-苯並吖呯-4,3’-環氧乙烷]-2(3H )-酮

依據中間體64描述之方法,自1,5,5’,6’-四氫-4’H -螺[1-苯並吖呯-4,3’-哌喃]-2(3H )-酮(中間體59)製備標的產物。MS (ESI) m/z 266.2 (M+H)+

中間體66
7-氯-1,5-二氫螺[1-苯並吖呯-4,3’-四氫呋喃]-2(3H )-酮

依據中間體64描述之方法,自1,4’,5,5’-四氫螺[1-苯並吖呯-4,3’-呋喃]-2(3H )-酮(中間體58)製備標的產物。MS (ESI) m/z 252.2 (M+H)+

中間體67
反式-4-(吡啶-2-基胺基)環己烷甲醯肼

a) 反式-4-(吡啶-2-基胺基)環己烷甲酸甲酯

令反式-4-胺基環己烷甲酸甲酯鹽酸鹽(1.5 g,7.745毫莫耳)、2-氟吡啶(4 ml,46.5毫莫耳)及DIPEA (1.35 ml,7.75毫莫耳)之混合物於抗壓性玻璃反應器中且於125℃下經攪拌20小時並隨後冷卻至室溫。令反應混合物經乙酸乙酯(20 ml)稀釋,經水(2 x 30 ml)和飽和NaCl溶液(2 x 30 ml)沖洗,令有機相置於無水硫酸鈉上乾燥且經過濾和濃縮以生成標的化合物(325 mg,18%)。MS (ESI) m/z 235.1 (M+H)+

b) 反式-4-(吡啶-2-基胺基)環己烷甲醯肼
依據中間體9之步驟b)描述之方法,自反式-4-(吡啶-2-基胺基)環己烷甲酸甲酯(中間體67之步驟a))製備標的化合物,其未經進一步純化而用於下一個步驟。

實施例1
8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯

令7-氯-2-側硫基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯(中間體4)(92 mg,0.34毫莫耳)、反式-4-(吡啶-2-基氧基)環己烷甲醯肼(WO2010/060836 A1 (03.06.2010) F. HOFFMANN-LA ROCHE AG.)(96.5 mg,0.41毫莫耳)及二甲苯(10 ml)之溶液於回流溫度下經攪拌3天。令反應混合物經濃縮並令殘餘物經管柱層析(使用二氯甲烷:甲醇=20:1作為溶析液)純化。因此,得到標的產物(68 mg,44.0%)。MS (ESI) m/z 454.2 (M+H)+

實施例2
8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸

令8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯(實施例1) (0.85 g,1.88毫莫耳)溶解於甲醇(60 ml),隨後加入2M NaOH溶液(20 ml)且令混合物經隔夜攪拌。自反應混合物蒸發甲醇,令水相之pH經1M氫氯酸溶液調整至3,且隨後經乙酸乙酯萃取。令合併之有機相經飽和NaCl溶液沖洗,置於無水硫酸鈉上乾燥且經過濾和濃縮。因此,得到標的產物(0.83 g,99.9%)。MS (ESI) m/z 439.1 (M+H)+

實施例3
8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺

令8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸(實施例2)(0.4 g,0.91毫莫耳)、HOBt (0.25 g,1.64毫莫耳)及EDC (0.314 g,1.64毫莫耳)溶解於無水DMF (3 ml)。令反應混合物於冰水浴中冷卻並於氬氣下加入氯化銨(0.146 g,2.73毫莫耳)和DIPEA (0.443 ml,2.55毫莫耳)。令反應混合物於0℃下經攪拌2小時並隨後於室溫下經隔夜攪拌。對反應混合物加入水,經乙酸乙酯萃取,令合併之有機相經飽和NaCl溶液沖洗,置於無水硫酸鈉上乾燥且經過濾和真空下濃縮。因此,得到標的產物(0.34 g,85.4%)。MS (ESI) m/z 438.2 (M+H)+

實施例4
8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-腈

令8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺(實施例3)(0.26 g,0.60毫莫耳)溶解於吡啶(4.4 ml)。隨後冷卻至5℃並將磷醯氯(56 μl,0.60毫莫耳)加入至反應混合物。令該混合物於室溫下經隔夜攪拌。令反應混合物隨後經濃縮,對殘餘物加入水且令水相之pH經1M NaOH溶液調整至9。令水相經乙酸乙酯萃取。令合併之有機相經飽和NaCl溶液沖洗,置於無水硫酸鈉上乾燥且經過濾和濃縮。令殘餘物經管柱層析(使用二氯甲烷:甲醇=20:1作為溶析液)純化。因此,得到標的產物(0.177 g,70.2%)。MS (ESI) m/z 420.2 (M+H)+

實施例5
8-氯-N,N-二甲基-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺

依據實施例3描述之方法,自8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸(實施例2)和二甲胺(2M THF溶液)製備標的產物。MS (ESI) m/z 466.2 (M+H)+

實施例6
(5S)-8-氯-N,N-二甲基-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺


實施例7
(5R)-8-氯-N,N-二甲基-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺

藉由手性製備性HPLC (CHIRALPAK IA (含有製備性20 μm固定相) 2.5 x 20 cm;F=15 ml/分鐘;溶析液:正己烷:EtOH=75:25+0.3%二乙胺;等度;溫度=25℃),自消旋性8-氯-N,N-二甲基-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺(實施例5)製備標的產物。第一溶析化合物之滯留時間係11.3分鐘;=-8.3˚ (c=0.1;甲醇);第二溶析化合物之滯留時間係19.7分鐘;=+4.9˚ (c=0.1;甲醇)。未測定該等化合物之絕對構型。

實施例8
8-氯-N-(丙-2-基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺

依據實施例3描述之方法,自8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸(實施例2)和異丙胺製備標的產物。MS (ESI) m/z 480.2 (M+H)+

實施例9
{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}(吡咯啶-1-基)甲酮

依據實施例3描述之方法,自8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸(實施例2)和吡咯啶製備標的產物。MS (ESI) m/z 492.2 (M+H)+

實施例10
{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}(嗎啉-4-基)甲酮

依據實施例3描述之方法,自8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸(實施例2)和嗎啉製備標的產物。MS (ESI) m/z 508.2 (M+H)+

實施例11
{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}(1,1-二氧基硫代嗎啉-4-基)甲酮

依據實施例3描述之方法,自8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸(實施例2)和硫代嗎啉-1,1-二氧化物製備標的產物。MS (ESI) m/z 556.2 (M+H)+

實施例12
{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲醇

令8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯(實施例1)(1.11 g,2.51毫莫耳)溶解於無水二乙醚(60 ml)與無水二氯甲烷(60 ml)之混合物並令溶液冷卻至-20℃。對混合物逐滴加入1M氫化鋁鋰之二乙醚溶液(5.5 ml)並於-20℃下經攪拌20分鐘。對反應混合物小心地加入水並隨後於真空下蒸發二乙醚。隨後令水相經乙酸乙酯萃取。令合併之有機相經飽和NaCl溶液沖洗,置於無水硫酸鈉上乾燥且經過濾和真空下濃縮。因此,得到標的產物(0.955 g,85.4%)。MS (ESI) m/z 425.2 (M+H)+

實施例13
{(5R)-8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲醇


實施例14
{(5S)-8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲醇

藉由手性製備性HPLC (CHIRALPAK IC (含有製備性20 μm固定相) 2.5 x 20 cm;F=15 ml/分鐘;溶析液:正己烷:EtOH=75:25;等度;溫度=25℃),自消旋性{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲醇(實施例12)製備標的產物。第一溶析化合物(滯留時間7.0分鐘)係{(5R)-8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲醇(實施例13);=+11.9° (c=0.1;氯仿);第二溶析化合物(滯留時間7.8分鐘)係{(5S)-8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲醇(實施例14);=-11.1˚ (c=0.1;氯仿)。藉由VCD方法和由彼合成之非鏡像異構物對的1 H NMR光譜測定該等化合物之絕對構型。

實施例15
8-氯-5-(甲氧基甲基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯

令{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲醇(實施例12)(70 mg,0.16毫莫耳)溶解於無水THF (2 ml)並隨後令溶液冷卻至-5℃。加入60%氫化鈉(13 mg,0.32毫莫耳)之油分散液並於冷卻下經攪拌90分鐘,隨後將碘甲烷(12 μl,0.2毫莫耳)加入至反應混合物並於室溫下經攪拌2小時。隨後再次加入碘甲烷(12 μl,0.2毫莫耳)並令混合物於40℃下經加熱2小時。令反應物經水(30 ml)稀釋並隨後經乙酸乙酯萃取。令合併之有機相經飽和NaCl溶液沖洗,置於無水硫酸鈉上乾燥且經過濾和濃縮。令殘餘物經管柱層析(使用二氯甲烷:甲醇=50:1作為溶析液)純化。因此,得到標的產物(20 mg,28%)。MS (ESI) m/z 439.2 (M+H)+

實施例16
嗎啉-4-甲酸{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲酯

a) 甲烷磺酸{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲酯

令{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲醇(實施例12)(0.855 g,2.01毫莫耳)溶解於無水二氯甲烷(80 ml)並隨後冷卻至0℃。加入三乙胺(0.785 ml,5.63毫莫耳)、甲烷磺醯氯(0.258 ml,3.38毫莫耳)及DMAP (34 mg,0.28毫莫耳)。令反應混合物於0℃下經攪拌1小時並隨後於室溫下經隔夜攪拌。令反應混合物經飽和NaHCO3 溶液沖洗並令水相經二氯甲烷萃取。令合併之有機相經飽和NaCl溶液沖洗,置於無水硫酸鈉上乾燥且經過濾和濃縮。因此,得到標的產物(1.0 g,99%)。MS (ESI) m/z 503.1 (M+H)+

b) 嗎啉-4-甲酸{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲酯
令甲烷磺酸{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲酯(實施例16之步驟a))(0.053 g,0.106毫莫耳)溶解於無水DMF (5 ml)。加入三乙胺(0.030 ml,0.212毫莫耳)、碳酸銫(0.038 g,0.117毫莫耳)及嗎啉(0.050 ml,0.573毫莫耳)並令混合物於100℃下經攪拌2天。隨後令反應混合物經冷卻,加入水且經乙酸乙酯萃取。令合併之有機相經飽和NaCl溶液沖洗,置於無水硫酸鈉上乾燥且經過濾和濃縮。令殘餘物經管柱層析(使用二氯甲烷:甲醇=20:1作為溶析液)純化。因此,得到標的產物(0.013 g,23.0%)。MS (ESI) m/z 538.2 (M+H)+

實施例17
(4-氟苯基)胺甲酸{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲酯

令{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲醇(實施例12)(40 mg,0.09毫莫耳)溶解於無水THF (3 ml),經冷卻至5℃並於導入氬氣同時加入三乙胺(24 μl,0.17毫莫耳)。將異氰酸4-氟苯酯(14 mg,0.10毫莫耳)之無水THF (1 ml)溶液加入至反應混合物。該反應甚至於回流溫度下未發生,因此經濃縮。令生成物溶解於乙腈(5 ml)並加入60%氫化鈉(4 mg,0.09毫莫耳)之油分散液。令反應混合物於室溫下經隔夜攪拌並隨後經濃縮。令殘餘物經管柱層析(使用二氯甲烷:甲醇=20:1作為溶析液)純化。因此,得到標的產物(35 mg,66%)。MS (ESI) m/z 562.2 (M+H)+

實施例18
5-(疊氮基甲基)-8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯

將甲烷磺酸{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲酯(實施例16之步驟a))(0.418 g,0.83毫莫耳)、無水DMF (10 ml)及疊氮化鈉(0.162 g,2.49毫莫耳)填充至抗壓性玻璃反應器並隨後於80℃下經攪拌4小時。隨後對反應混合物加入水並經乙酸乙酯萃取。令合併之有機相經飽和NaCl溶液沖洗,置於無水硫酸鈉上乾燥且經過濾和濃縮。令殘餘物經管柱層析(使用二氯甲烷:甲醇=20:1作為溶析液)純化。因此,得到標的產物(0.26 g,69.5%)。MS (ESI) m/z 450.2 (M+H)+

實施例19
1-{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲胺

令5-(疊氮基甲基)-8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯(實施例18)(0.242 g,0.54毫莫耳)溶解於THF (20 ml)並隨後加入水(1 ml)和三苯膦(0.423 g,1.61毫莫耳)。令反應混合物於氬氣和室溫下經隔夜攪拌,隨後經濃縮且令殘餘物經管柱層析(使用二氯甲烷:甲醇:氫氧化銨=10:1:0.1作為溶析液)純化。因此,得到標的產物(0.183 g,80%)。MS (ESI) m/z 424.2 (M+H)+

實施例20
8-氯-5-(嗎啉-4-基甲基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯

將甲烷磺酸{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲酯(實施例16之步驟a))(100 mg,0.20毫莫耳)、無水DMF (3 ml)、DIPEA (0.42 ml,2.39毫莫耳)及嗎啉(0.21 ml,2.39毫莫耳)填充至抗壓性玻璃反應器並隨後於110℃下經攪拌1天。隨後令反應混合物經冷卻,加入水且經乙酸乙酯萃取。令合併之有機相經飽和NaCl溶液沖洗,置於無水硫酸鈉上乾燥且經過濾和濃縮。令殘餘物經管柱層析(使用二氯甲烷:甲醇=50:1作為溶析液)純化。因此,得到標的產物(59 mg,60%)。MS (ESI) m/z 494.2 (M+H)+

實施例21
1-{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}-N,N-二甲基甲胺

依據實施例20描述之方法,自甲烷磺酸{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲酯(實施例16之步驟a))和二甲胺(2M THF溶液)製備標的產物。MS (ESI) m/z 452,2 (M+H)+

實施例22
N-({8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲基)丙-2-胺

令1-{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲胺(實施例19)(50 mg,0.12毫莫耳)溶解於1,2-二氯乙烷(5 ml)並隨後加入乙酸(14 µl,0.25毫莫耳)、丙酮(45 µl,0.61毫莫耳)及三乙醯氧基氫硼化鈉(77 mg,0.37毫莫耳)。令反應混合物於室溫下經攪拌1天並隨後加入水。令水相之pH經1M NaOH溶液調整至9並分離有機相。令水相經二氯甲烷萃取並令合併之有機相經飽和NaCl溶液沖洗,置於無水硫酸鈉上乾燥且經過濾和濃縮。令殘餘物經管柱層析(使用二氯甲烷:甲醇:氫氧化銨=10:1:0.1作為溶析液)純化。因此,得到標的產物(46 mg,83.6%)。MS (ESI) m/z 466.2 (M+H)+

實施例23
8-氯-1-[1-(嘧啶-2-基)四氫吖唉-3-基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯

依據實施例1描述之方法,自7-氯-2-側硫基-2,3,4,5-四氫-1H-1-苯並吖呯-4-甲酸甲酯(中間體4)和1-(嘧啶-2-基)四氫吖唉-3-甲醯肼(中間體13)製備標的產物。MS (ESI) m/z 411.1 (M+H)+

實施例24
8-氯-1-[反式-4-(二甲基胺基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯

依據實施例1描述之方法,自7-氯-2-側硫基-2,3,4,5-四氫-1H-1-苯並吖呯-4-甲酸甲酯(中間體4)和反式-4-(二甲基胺基)環己烷甲醯肼(中間體11)製備標的產物。MS (ESI) m/z 403.2 (M+H)+

實施例25
8-氯-1-[反式-4-(三氟甲基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯

依據實施例1描述之方法,自7-氯-2-側硫基-2,3,4,5-四氫-1H-1-苯並吖呯-4-甲酸甲酯(中間體4)和反式-4-(三氟甲基)環己烷甲醯肼(中間體6)製備標的產物。MS (ESI) m/z 428.1 (M+H)+

實施例26
8-氯-1-[反式-4-(嗎啉-4-基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯

依據實施例1描述之方法,自7-氯-2-側硫基-2,3,4,5-四氫-1H-1-苯並吖呯-4-甲酸甲酯(中間體4)和反式-4-(嗎啉-4-基)環己烷甲醯肼(中間體12)製備標的產物。MS (ESI) m/z 445.2 (M+H)+

實施例27
{1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲醇

令{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲醇(實施例12)(63 mg,0.15毫莫耳)溶解於乙醇(15 ml)並隨後將10% Pd/C (65 mg)加入至溶液且於室溫下進行氫化。經反應完成後,過濾除去觸媒,經乙醇沖洗並令濾液經濃縮。令殘餘物經管柱層析(使用二氯甲烷:甲醇:氫氧化銨=10:1:0.1作為溶析液)純化。因此,得到標的產物(23 mg,39%)。MS (ESI) m/z 391.2 (M+H)+

實施例28
8-氯-1-[反式-4-(吡咯啶-1-基羰基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯

令7-氯-2-側硫基-2,3,4,5-四氫-1H-1-苯並吖呯-4-甲酸甲酯(中間體4)(56.8 mg,0.21毫莫耳)、反式-4-(吡咯啶-1-基羰基)環己烷甲醯肼(中間體9)(75.6 mg,0.32毫莫耳)及正丁醇(2 ml)之溶液經回流28小時並隨後令反應混合物經真空下濃縮。令殘餘物經管柱層析(使用二氯甲烷:甲醇=9:1作為溶析液)純化。因此,得到標的產物(29.6 mg,31%)。MS (ESI) m/z 457.2 (M+H)+

實施例29
{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}乙腈

令甲烷磺酸{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲酯(實施例16之步驟a))(100 mg,0.20毫莫耳)、無水DMF (10 ml)及氰化鈉(25 mg,0.52毫莫耳)之混合物於抗壓性玻璃反應器中且於80℃下經攪拌5小時。隨後將水加入至反應混合物並經乙酸乙酯萃取。令合併之有機相經飽和NaCl溶液沖洗,置於無水硫酸鈉上乾燥且經過濾和濃縮。令殘餘物經管柱層析(使用二氯甲烷:甲醇=20:1作為溶析液)純化。因此,得到標的產物(62 mg,71.8%)。MS (ESI) m/z 434.2 (M+H)+

實施例30
8-氯-5-[(4-甲基哌嗪-1-基)甲基]-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯

依據實施例20描述之方法,自甲烷磺酸{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲酯(實施例16之步驟a))和1-甲基-哌嗪製備標的產物。MS (ESI) m/z 507.2 (M+H)+

實施例31
8-氯-5-(氟甲基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯

令甲烷磺酸{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲酯(實施例16之步驟a))(100 mg,0.20毫莫耳)和1M四丁基氟化銨之THF溶液(1 ml)於氬氣和回流溫度下經隔夜攪拌。隨後對反應混合物加入水並經乙酸乙酯萃取。令合併之有機相經飽和NaCl溶液沖洗,置於無水硫酸鈉上乾燥且經過濾和濃縮。令殘餘物經製備性HPLC (Kinetex EVO C18;5 μm負載;150 x 21.2 mm;F=20 ml/分鐘;溶析液:A:水;B:乙腈,梯度:B% 15→70;溫度=40℃)純化。因此,得到標的產物(21 mg,24.7%)。MS (ESI) m/z 427.2 (M+H)+

實施例32
8-氯-5-(3-甲基-1,2,4-噁二唑-5-基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯

令8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸(實施例2)(50 mg,0.11毫莫耳)、HOBt (21 mg,0.13毫莫耳)、EDC (26 mg,0.13毫莫耳)及DMF (1 ml)之混合物於氬氣和室溫下經攪拌15分鐘並隨後加入N-羥基乙脒(10 mg,0.13毫莫耳)和額外DMF (1 ml)。令反應混合物於100℃下經隔夜攪拌。隨後令反應混合物經冷卻至室溫,加入水且經乙酸乙酯萃取。令合併之有機相經飽和NaCl溶液沖洗,置於無水硫酸鈉上乾燥且經過濾和濃縮。令殘餘物經管柱層析(使用二氯甲烷:甲醇=50:1作為溶析液)純化。因此,得到標的產物(29 mg,53.7%)。MS (ESI) m/z 477.2 (M+H)+

實施例33
8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5-(2H-四唑-5-基)-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯

令8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-腈(實施例4)(90 mg,0.21毫莫耳)、DMF (2.5 ml)、疊氮化鈉(133 mg,2.04毫莫耳)及氯化銨(111 mg,2.08毫莫耳)之混合物於抗壓性玻璃反應器中且於115℃下經攪拌4小時。令反應混合物經冷卻,加入水且經乙酸乙酯萃取。令合併之有機相經飽和NaCl溶液沖洗,置於無水硫酸鈉上乾燥且經過濾和濃縮。令殘餘物經管柱層析(使用二氯甲烷:甲醇=3:1作為溶析液)純化。因此,得到標的產物(54 mg,54.5%)。MS (ESI) m/z 463.2 (M+H)+

實施例34
N-({8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲基)四氫-2H-哌喃-4-胺

依據實施例22描述之方法,自1-{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲胺(實施例19)和四氫-4H -哌喃-4-酮製備標的產物。MS (ESI) m/z 508.2 (M+H)+

實施例35
N,N-二甲基-1-[反式-4-(吡啶-2-基氧基)環己基]-8-(三氟甲基)-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-胺

依據實施例22描述之方法,自1-{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲胺(實施例19)和4,4-二氟環己酮製備標的產物。MS (ESI) m/z 542.2 (M+H)+

實施例36
8-氯-1-[反式-3-甲基-2-側氧基-1-氧雜-3-氮雜螺[4.5]癸-8-基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯

依據實施例1描述之方法,自7-氯-2-側硫基-2,3,4,5-四氫-1H-1-苯並吖呯-4-甲酸甲酯(中間體4)和(反式)-3-甲基-2-側氧基-1-氧雜-3-氮雜螺[4.5]癸烷-8-甲醯肼(中間體17)製備標的產物。MS (ESI) m/z 445.2 (M+H)+

實施例37
N-({8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲基)乙醯胺

令1-{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲胺(實施例19)(45 mg,0.12毫莫耳)溶解於吡啶(5 ml)並隨後加入乙酸酐(0.10 ml,1.06毫莫耳)。令反應混合物於室溫下經攪拌4小時且隨後經濃縮。令殘餘物溶解於水。令水相經二氯甲烷萃取並令合併之有機相經飽和NaCl溶液沖洗,置於無水硫酸鈉上乾燥且經過濾和濃縮。因此,得到標的產物(44 mg,89.0%)。MS (ESI) m/z 466.2 (M+H)+

實施例38
N-({8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲基)氧雜環丁烷-3-胺

依據實施例22描述之方法,自1-{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲胺(實施例19)和3-氧雜環丁酮製備標的產物。MS (ESI) m/z 480.2 (M+H)+

實施例39
8-氯-1-[反式-4-(嗎啉-4-基羰基)環己基]-5,6-二氫-4H -[1,2,4]三唑並[4,3-a ][1]苯並吖呯-5-甲酸甲酯

依據實施例28描述之方法,自7-氯-2-側硫基-2,3,4,5-四氫-1H-1-苯並吖呯-4-甲酸甲酯(中間體4)和反式-4-(嗎啉-1-基羰基)環己烷甲醯肼(中間體10)製備標的產物。MS (ESI) m/z 473.2 (M+H)+

實施例40
8-氯-1-[順式-3-甲基-2-側氧基-1-氧雜-3-氮雜螺[4.5]癸-8-基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯

依據實施例1描述之方法,自7-氯-2-側硫基-2,3,4,5-四氫-1H-1-苯並吖呯-4-甲酸甲酯(中間體4)和(順式)-3-甲基-2-側氧基-1-氧雜-3-氮雜螺[4.5]癸烷-8-甲醯肼(中間體18)製備標的產物。MS (ESI) m/z 445.2 (M+H)+

實施例41
8-氯-1-[1-(吡啶-2-基)四氫吖唉-3-基]-5,6-二氫-4H -[1,2,4]三唑並[4,3-a ][1]苯並吖呯-5-甲酸甲酯

依據實施例28描述之方法,自7-氯-2-側硫基-2,3,4,5-四氫-1H-1-苯並吖呯-4-甲酸甲酯(中間體4)和1-(吡啶-2-基)四氫吖唉-3-甲醯肼(中間體14)製備標的產物。MS (ESI) m/z 410.1 (M+H)+

實施例42
1-[反式-4-(四氫吖唉-1-基羰基)環己基]-8-氯-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯

依據實施例1描述之方法,自7-氯-2-側硫基-2,3,4,5-四氫-1H-1-苯並吖呯-4-甲酸甲酯(中間體4)和反式-4-(四氫吖唉-1-基羰基)環己烷甲醯肼(中間體19)製備標的產物。MS (ESI) m/z 443.2 (M+H)+

實施例43
8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5-(吡咯啶-1-基甲基)-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯

依據實施例20描述之方法,自甲烷磺酸{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲酯(實施例16之步驟a))和吡咯啶製備標的產物。MS (ESI) m/z 478.2 (M+H)+

實施例44
N-({8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲基)-2-甲基-N-(2-甲基丙基)丙-1-胺

依據實施例22描述之方法,自1-{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲胺(實施例19)和2-甲基丙醛製備標的產物。MS (ESI) m/z 536.3 (M+H)+

實施例45
8-氯-1-[反式-4-(哌啶-1-基羰基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯

依據實施例28描述之方法,自7-氯-2-側硫基-2,3,4,5-四氫-1H-1-苯並吖呯-4-甲酸甲酯(中間體4)和反式-4-(哌啶-1-基羰基)環己烷甲醯肼(中間體20)製備標的產物。MS (ESI) m/z 471.2 (M+H)+

實施例46
1-(反式-4-{[4-(三級丁氧羰基)哌嗪-1-基]羰基}環己基)-8-氯-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯

依據實施例28描述之方法,自7-氯-2-側硫基-2,3,4,5-四氫-1H-1-苯並吖呯-4-甲酸甲酯(中間體4)和4-{[反式-4-(肼基羰基)環己基]羰基}哌嗪-1-甲酸三級丁酯(中間體21)製備標的產物。MS (ESI) m/z 572.2 (M+H)+

實施例47
8-氯-1-[反式-4-(哌嗪-1-基羰基)環己基]-5,6-二氫-4H -[1,2,4]三唑並[4,3-a ][1]苯並吖呯-5-甲酸甲酯鹽酸鹽

令1-(反式-4-{[4-(三級丁氧羰基)哌嗪-1-基]羰基}環己基)-8-氯-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯(實施例46)(41 mg,0.07毫莫耳)溶解於乙酸乙酯(1 ml)並對溶液加入2.5M鹽酸之乙酸乙酯溶液(1 ml)。令反應混合物於室溫下經攪拌1小時並隨後加入二乙醚。令沉澱之產物經過濾並經二乙醚沖洗。因此,得到標的產物(30.5 mg,84%)。MS (ESI) m/z 472.2 (M+H)+

實施例48
8-甲氧基-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯

依據實施例1描述之方法,自7-甲氧基-2-側硫基-2,3,4,5-四氫-1H-1-苯並吖呯-4-甲酸甲酯(中間體26)製備標的化合物。MS (ESI) m/z 449.3 (M+H)+

實施例49
8-甲基-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯

依據實施例1描述之方法,自7-甲基-2-側硫基-2,3,4,5-四氫-1H-1-苯並吖呯-4-甲酸甲酯(中間體25)製備標的化合物。MS (ESI) m/z 433.2 (M+H)+

實施例50
8-氯-N-(4-氟苯基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺

對8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸(實施例2)(100 mg,0.23毫莫耳)、DMF (15 ml)、HOBt (84 mg,0.55毫莫耳)、DIPEA (151 µl,0.87毫莫耳)及EDC (105 mg,0.55毫莫耳)之冰冷混合物加入4-氟苯胺(25 mg,0.23毫莫耳)並令反應混合物於5℃下經攪拌0.5小時且隨後於室溫下再經攪拌20小時。經反應完成後,令混合物經水和鹽水稀釋並經乙酸乙酯萃取。令結合之有機層置於硫酸鈉上乾燥且經過濾和濃縮。令殘餘物經快速管柱層析(使用二氯甲烷:甲醇=95:5作為溶析液)純化以生成標的化合物(105 mg,87%)。MS (ESI) m/z 532.1 (M+H)+

實施例51
8-溴-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯

依據實施例1描述之方法,自7-溴-2-側硫基-2,3,4,5-四氫-1H-1-苯並吖呯-4-甲酸甲酯(中間體27)製備標的化合物。MS (ESI) m/z 499.1 (M+H)+

實施例52
1-(1,4’-聯哌啶-1’-基)-8-氯-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺

令1-溴-8-氯-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺(中間體29)(34 mg,0.1毫莫耳)、環丁碸(0.3 ml)及4-哌啶並哌啶(218 mg,1.3毫莫耳)之混合物於180℃下經攪拌3小時並隨後冷卻至室溫。令反應混合物經二氯甲烷稀釋並經鹽水沖洗,令有機相置於硫酸鈉上乾燥且經過濾和濃縮。令殘餘物經快速管柱層析(使用二氯甲烷:甲醇:氫氧化銨=9:1:0.1作為溶析液)純化以生成標的化合物(13 mg,30%)。MS (ESI) m/z 429.2 (M+H)+

實施例53
8-氯-1-[(5r,8r)-1-側氧基-2-(丙-2-基)-2-氮雜螺[4.5]癸-8-基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯

依據實施例28描述之方法,自7-氯-2-側硫基-2,3,4,5-四氫-1H-1-苯並吖呯-4-甲酸甲酯(中間體4)和(5r,8r)-1-側氧基-2-(丙-2-基)-2-氮雜螺[4.5]癸烷-8-甲醯肼(中間體32)製備標的化合物。MS (ESI) m/z 471.2 (M+H)+

實施例54
1-{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}乙醇

a) 5-(1-{[三級丁基(二甲基)矽基]氧}乙基)-8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯

依據實施例1描述之方法,自4-(1-{[三級丁基(二甲基)矽基]氧}乙基)-7-氯-1,3,4,5-四氫-2H-1-苯並吖呯-2-硫酮(中間體34)製備標的化合物。產物係非鏡像異構物之55:45混合物。MS (ESI) m/z 553.2 (M+H)+

b) 1-{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}乙醇
令5-(1-{[三級丁基(二甲基)矽基]氧}乙基)-8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯(實施例54之步驟a))(222 mg,0.4毫莫耳)和無水THF (40 ml)之攪拌混合物冷卻至0℃並加入1M四丁基氟化銨(0.8 ml,0.8毫莫耳)之THF溶液且令反應混合物於室溫下經攪拌20小時並隨後經濃縮。對殘餘物加入水並經乙酸乙酯萃取。令結合之有機層經鹽水沖洗,置於硫酸鈉上乾燥且經過濾和濃縮。令殘餘物經快速管柱層析(使用二氯甲烷:甲醇=9:1作為溶析液)純化以生成標的化合物(172 mg,98%)。產物係非鏡像異構物之54:46混合物。MS (ESI) m/z 439.1 (M+H)+

實施例55
1-{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}乙酮

令1-{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}乙醇(實施例54)(142 mg,0.32毫莫耳)和二氯甲烷(10 ml)之攪拌混合物冷卻至0℃並加入戴斯-馬丁碘劑(206 mg,0.48毫莫耳)。令反應混合物於室溫下經攪拌3小時並隨後經過濾。令濾液經水稀釋,分離相層且令水相經二氯甲烷萃取。令結合之有機層經鹽水沖洗,置於硫酸鈉上乾燥且經過濾和濃縮。令殘餘物經快速管柱層析(使用二氯甲烷:甲醇=95:5作為溶析液)純化以生成標的化合物(111 mg,79%)。MS (ESI) m/z 437.2 (M+H)+

實施例56
8-氯-5-(氟甲基)-1-(反式-4-甲氧基-4-甲基環己基)-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯

依據實施例1描述之方法,自7-氯-4-(氟甲基)-1,3,4,5-四氫-2H-1-苯並吖呯-2-硫酮(中間體37)和4-甲氧基-4-甲基環己烷甲醯肼(中間體35之步驟c))製備標的化合物。MS (ESI) m/z 378.2 (M+H)+

實施例57
1-{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}-N-(2-甲氧基乙基)乙胺

令1-{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}乙酮(實施例55)(30 mg,0.07毫莫耳)、1,2-二氯甲烷(5 ml)、2-甲氧基乙基胺(24 µl,0.275毫莫耳)、乙酸(30 µl)及NaBH(OAc)3 (29 mg,0.137毫莫耳)之混合物於室溫下經攪拌4小時並隨後經濃縮。藉由加入1M NaOH將殘餘物之pH調整至9並隨後令混合物經二氯甲烷萃取。令結合之有機層經鹽水沖洗,置於硫酸鈉上乾燥且經過濾和濃縮。令殘餘物經快速管柱層析(使用二氯甲烷:甲醇=97:3作為溶析液)純化以生成標的化合物(12 mg,35%)。產物係非鏡像異構物之58:42混合物。MS (ESI) m/z 496.3 (M+H)+

實施例58
8-氟-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯

依據實施例1描述之方法,自7-氟-2-側硫基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯(中間體39)製備標的化合物。MS (ESI) m/z 437.4 (M+H)+

實施例59
8-氯-5-(氟甲基)-1-[反式-4-(三氟甲基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯

依據實施例1描述之方法,自7-氯-4-(氟甲基)-1,3,4,5-四氫-2H-1-苯並吖呯-2-硫酮(中間體37)和反式-4-(三氟甲基)環己烷甲醯肼(中間體6)製備標的化合物。MS (ESI) m/z 402.3 (M+H)+

實施例60
5-(氟甲基)-1-(反式-4-甲氧基-4-甲基環己基)-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯

依據實施例27描述之方法,自8-氯-5-(氟甲基)-1-(反式-4-甲氧基-4-甲基環己基)-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯(實施例56)製備標的化合物。MS (ESI) m/z 344.3 (M+H)+

實施例61
{8-氟-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲醇

依據實施例12描述之方法,自8-氟-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯(實施例58)製備標的化合物。MS (ESI) m/z 409.3 (M+H)+

實施例62
8-氟-5-(氟甲基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯

依據實施例1描述之方法,自7-氟-4-(氟甲基)-1,3,4,5-四氫-2H-1-苯並吖呯-2-硫酮(中間體41)製備標的化合物。MS (ESI) m/z 411.3 (M+H)+

實施例63
順式-(消旋)-1-{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}乙醇


實施例64
反式-(消旋)-1-{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}乙醇

a) 5-[順式-(消旋)-1-{[三級丁基(二甲基)矽基]氧}乙基]-8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯和5-[反式-(消旋)-1-{[三級丁基(二甲基)矽基]氧}乙基]-8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯

依據實施例1描述之方法,自4-(1-{[三級丁基(二甲基)矽基]氧}乙基)-7-氯-1,3,4,5-四氫-2H-1-苯並吖呯-2-硫酮(中間體34)之經分離的單一非鏡像異構物製備標的化合物。產物係單一非鏡像異構物但未測定彼等之構型。MS (ESI) m/z 553.3 (M+H)+

b) 順式-(消旋)-1-{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}乙醇和反式-(消旋)-1-{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}乙醇
依據實施例54之步驟b)描述之方法,自5-[順式-(消旋)-1-{[三級丁基(二甲基)矽基]氧}乙基]-8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯和5-[反式-(消旋)-1-{[三級丁基(二甲基)矽基]氧}乙基]-8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯(實施例63和64之步驟a))製備標的化合物。產物係單一非鏡像異構物但未測定彼等之構型。MS (ESI) m/z 439.4 (M+H)+

實施例65
(1R)-1-{(5R)-8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}乙醇


實施例66
(1S)-1-{(5S)-8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}乙醇


實施例67
(1S)-1-{(5R)-8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}乙醇


實施例68
(1R)-1-{(5S)-8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}乙醇

藉由手性製備性HPLC (CHIRALPAK IG製備性20 µm固定相;5 x 30 cm;F=50 ml/分鐘;溶析液:正庚烷:異丙醇=8:2;等度;溫度=25℃),自經分離之非鏡像異構物:順式-(消旋)-1-{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}乙醇和反式-(消旋)-1-{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}乙醇(實施例63和64)得到標的化合物。分離消旋性混合物之一者產生2個化合物:第一溶析化合物(Tr 13.9分鐘)之旋光度係=+14˚ (c=0.1;氯仿)且第二溶析化合物(Tr 16.7分鐘)之旋光度係=-12˚ (c=0.1;氯仿)。分離消旋性混合物之另一者產生另2個化合物:第一溶析化合物(Tr 19.0分鐘)之旋光度係=+21˚ (c=0.1;氯仿)且第二溶析化合物(Tr 21.5分鐘)之旋光度係=-16˚ (c=0.1;氯仿)。未測定該等化合物之絕對構型。

實施例69
5-(氟甲基)-8-甲基-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯

依據實施例1描述之方法,自4-(氟甲基)-7-甲基-1,3,4,5-四氫-2H-1-苯並吖呯-2-硫酮(中間體43)製備標的化合物。MS (ESI) m/z 407.3 (M+H)+

實施例70
(5S)-8-氯-5-(氟甲基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯


實施例71
(5R)-8-氯-5-(氟甲基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯

藉由手性製備性HPLC (CHIRALPAK IG製備性20 µm固定相;5 x 30 cm;F=50 ml/分鐘;溶析液:三級丁基甲基醚:二氯甲烷:EtOH=90:8:2;等度;溫度=25℃),自消旋性8-氯-5-(氟甲基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a ][1]苯並吖呯(實施例31)得到標的化合物。第一溶析化合物(滯留時間20.2分鐘)係(5S)-8-氯-5-(氟甲基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯(實施例70),=+6.3° (c=0.1;二氯甲烷);且,第二溶析化合物(滯留時間22.9分鐘)係(5R)-8-氯-5-(氟甲基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯(實施例71),=-8.9° (c=0.1;二氯甲烷)。藉由VCD方法測定該等化合物之構型。

實施例72
8-氟-1-[反式-4-(三氟甲基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯

令7-氟-2-(甲硫基)-4,5-二氫-3H-1-苯並吖呯-4-甲酸甲酯(中間體44)(270 mg,0.1毫莫耳)、1,4-二噁烷(20 ml)、反式-4-(三氟甲基)環己烷甲醯肼(中間體6)(210 mg,0.1毫莫耳)及濃氫氯酸(10 µl)之混合物經回流1小時並隨後經濃縮。令殘餘物經快速管柱層析(使用二氯甲烷:甲醇=95:5作為溶析液)純化以生成標的化合物(145 mg,35%)。MS (ESI) m/z 412.1 (M+H)+

實施例73
8-氯-1-[4-(2,3-二甲基苯基)哌嗪-1-基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺

依據實施例52描述之方法,自1-溴-8-氯-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺(中間體29)和1-(2,3-二甲基苯基)哌嗪製備標的化合物。MS (ESI) m/z 451.2 (M+H)+

實施例74
{8-氟-1-[反式-4-(三氟甲基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲醇

依據實施例12描述之方法,自8-氟-1-[反式-4-(三氟甲基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯(實施例72)製備標的化合物。MS (ESI) m/z 384.2 (M+H)+

實施例75
{8-氯-5-甲基-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲醇

a) 8-氯-5-甲基-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯

依據實施例1描述之方法,自7-氯-4-甲基-2-側硫基-2,3,4,5-四氫-1H-1-苯並吖呯-4-甲酸甲酯(中間體46)製備標的化合物。MS (ESI) m/z 467.1 (M+H)+

b) {8-氯-5-甲基-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲醇
依據實施例12描述之方法,自8-氯-5-甲基-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯(實施例75之步驟a))製備標的化合物。MS (ESI) m/z 439.1 (M+H)+

實施例76
{8-氯-5-乙基-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲醇

a) 8-氯-5-乙基-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯

依據實施例72描述之方法,自7-氯-4-乙基-2-(甲硫基)-4,5-二氫-3H-1-苯並吖呯-4-甲酸甲酯(中間體49)製備標的化合物。MS (ESI) m/z 481.2 (M+H)+

b) {8-氯-5-乙基-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲醇
依據實施例12描述之方法,自8-氯-5-乙基-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯(實施例76之步驟a))製備標的化合物。MS (ESI) m/z 453.2 (M+H)+

實施例77
(5S)-8-氟-5-(氟甲基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯


實施例78
(5R)-8-氟-5-(氟甲基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯

藉由手性製備性HPLC (CHIRALPAK IA製備性20 µm固定相;2.5 x 20 cm;F=15 ml/分鐘;溶析液:三級丁基甲基醚:二氯甲烷=75:25;等度;溫度=25℃),自消旋性8-氟-5-(氟甲基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯(實施例62)得到標的化合物。第一溶析化合物(Tr 16.6分鐘)之旋光度係=+9° (c=0.1;二氯甲烷)且第二溶析化合物(Tr 19.0分鐘)之旋光度係=-10° (c=0.1;二氯甲烷)。未測定該等化合物之絕對構型。

實施例79
8-氯-1-[4-(3-氯苯基)哌嗪-1-基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺

依據實施例52描述之方法,自1-溴-8-氯-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺(中間體29)和1-(3-氯苯基)哌嗪製備標的化合物。MS (ESI) m/z 457.2 (M+H)+

實施例80
8-氯-1-[4-(吡啶-2-基)哌嗪-1-基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺

依據實施例52描述之方法,自1-溴-8-氯-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺(中間體29)和1-(吡啶-2-基)哌嗪製備標的化合物。MS (ESI) m/z 424.2 (M+H)+

實施例81
8-氯-1-[4-(吡啶-2-基氧基)哌啶-1-基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺

依據實施例52描述之方法,自1-溴-8-氯-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺(中間體29)和2-(哌啶-4-基氧基)吡啶製備標的化合物。MS (ESI) m/z 439.2 (M+H)+

實施例82
5-[(苄氧基)甲基]-8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯

令{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲醇(實施例12)(100 mg,0.24毫莫耳)之無水DMF (3 ml)攪拌混合物冷卻至0℃並於氬氣下加入60%氫化鈉(24 mg,0.59毫莫耳)之礦物油溶液。令混合物於此溫度下經攪拌0.5小時,隨後加入苄基溴(70 µl,0.59毫莫耳)和四丁基碘化銨(27 mg,0.07毫莫耳)且令反應混合物於室溫下經攪拌3小時。經反應完成後,令混合物經飽和氯化銨溶液稀釋,經乙酸乙酯萃取,令結合之有機層經鹽水沖洗,置於硫酸鈉上乾燥且經過濾和濃縮。令殘餘物經快速管柱層析(使用二氯甲烷:甲醇=95:5作為溶析液)純化以生成標的化合物(44 mg,36%)。MS (ESI) m/z 515.2 (M+H)+

實施例83
8-氯-1-(3,3-二氟環丁基)-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯

依據實施例1描述之方法,自7-氯-2-側硫基-2,3,4,5-四氫-1H-1-苯並吖呯-4-甲酸甲酯(中間體4)和3,3-二氟環丁烷甲醯肼(中間體51)製備標的化合物。MS (ESI) m/z 368.1 (M+H)+

實施例84
8-氯-1-[反式-4-(哌啶-1-基甲基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯

依據實施例1描述之方法,自7-氯-2-側硫基-2,3,4,5-四氫-1H-1-苯並吖呯-4-甲酸甲酯(中間體4)和反式-4-(哌啶-1-基甲基)環己烷甲醯肼(中間體8)製備標的化合物。MS (ESI) m/z 457.3 (M+H)+

實施例85
{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}乙酸甲酯

令{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲醇(實施例12)(100 mg,0.24毫莫耳)、二氯甲烷(5 ml)、吡啶(57 µl,0.7毫莫耳)、乙酸酐(67 µl,0.7毫莫耳)及DMAP (3 mg,0.02毫莫耳)之混合物於室溫下經攪拌20小時,隨後經水稀釋且經二氯甲烷萃取。令結合之有機層經鹽水沖洗,置於硫酸鈉上乾燥且經過濾和濃縮。令殘餘物經快速管柱層析(使用二氯甲烷:甲醇=97:3作為溶析液)純化以生成標的化合物(38 mg,35%)。MS (ESI) m/z 467.1 (M+H)+

實施例86
8-氯-1-(1’H,3H-螺[2-苯並呋喃-1,4’-哌啶]-1’-基)-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺

依據實施例52描述之方法,自1-溴-8-氯-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺(中間體29)和3H-螺[2-苯並呋喃-1,4’-哌啶](Combi-Blocks)製備標的化合物。MS (ESI) m/z 450.2 (M+H)+

實施例87
8-氯-N-(吡啶-2-基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺

對8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸(實施例2)(361 mg,0.82毫莫耳)之二氯甲烷(25 ml)攪拌溶液加入草醯氯(446 µl,5.3毫莫耳)和DMF (50 µl)。令反應混合物於室溫下經攪拌30分鐘並隨後經濃縮。令殘餘物溶解於吡啶(5 ml)並加入2-胺基吡啶(155 mg,1.64毫莫耳)。令所得之反應混合物於室溫下經攪拌2小時,隨後經濃縮且令殘餘物經製備性HPLC (KINETEX EVO 5 µm固定相;150 x 21.2 mm;F=20 ml/分鐘;溶析液:(水+0.1%三氟乙酸):(甲醇:乙腈=2:1)=52:48;等度;溫度=40℃)純化以生成標的化合物(12.7 mg,3%)。MS (ESI) m/z 515.2 (M+H)+

實施例88
8-氯-1-(1’H,3H-螺[2-苯並呋喃-1,4’-哌啶]-1’-基)-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-腈

令8-氯-1-(1’H,3H-螺[2-苯並呋喃-1,4’-哌啶]-1’-基)-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺(實施例86)(78.6 mg,0.175毫莫耳)、無水二氯甲烷(30 ml)及三乙胺(153 µl,1.1毫莫耳)之攪拌溶液冷卻至0℃並加入三氟乙酸酐(40 µl,0.28毫莫耳)。令混合物之溫度回溫至室溫並令反應混合物於此溫度下經攪拌1小時,隨後加入三乙胺(153 µl,1.1毫莫耳)和三氟乙酸酐(40 µl,0.28毫莫耳)。令混合物於室溫下經隔夜攪拌,隨後加入三乙胺(153 µl,1.1毫莫耳)和三氟乙酸酐(80 µl,0.56毫莫耳)且於室溫下持續隔夜攪拌。令反應混合物經飽和NaHCO3 溶液稀釋,分離相層且令水相經二氯甲烷萃取。令合併之有機相經水和鹽水沖洗,置於硫酸鈉上乾燥且經過濾和濃縮。令殘餘物經管柱層析(使用二氯甲烷:甲醇:氫氧化銨=180:10:1作為溶析液)純化以生成標的化合物(11.5 mg,15%)。MS (ESI) m/z 432.1 (M+H)+

實施例89
8-氯-1-[4-(2,3-二甲基苯基)哌嗪-1-基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-腈

令1-溴-8-氯-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-腈(中間體52)(100 mg,0.31毫莫耳)和1-(2,3-二甲基苯基)哌嗪(412 mg,2.16毫莫耳)之混合物於氬氣和180℃下經攪拌4小時並隨後冷卻至室溫。令殘餘物經管柱層析(使用二氯甲烷:甲醇:氫氧化銨=180:10:1作為溶析液)純化且自二乙醚結晶後生成標的化合物(28 mg,21%)。MS (ESI) m/z 433.1 (M+H)+

實施例90
8-氯-1-[4-(吡啶-2-基)哌嗪-1-基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-腈

依據實施例89描述之方法,自1-溴-8-氯-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-腈(中間體52)和1-(吡啶-2-基)哌嗪製備標的化合物。MS (ESI) m/z 406.1 (M+H)+

實施例91
8-氟-5,5-二甲基-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯

a) 7-氟-4,4-二甲基-1,3,4,5-四氫-2H-1-苯並吖呯-2-硫酮

對7-氟-4,4-二甲基-1,3,4,5-四氫-2H-1-苯並吖呯-2-酮(中間體53)(1.10 g,5.31毫莫耳)和吡啶(10 ml)之溶液加入勞森反應劑(2.79 g,6.90毫莫耳)並令反應混合物經回流2小時。隨後,真空下除去溶劑,令殘餘物經飽和NaHCO3 磨碎且藉由過濾收集沉澱物。因此,得到標的產物(1.00 g,84%),其未經進一步純化而使用。

b) 7-氟-4,4-二甲基-2-(甲硫基)-4,5-二氫-3H-1-苯並吖呯

對7-氟-4,4-二甲基-1,3,4,5-四氫-2H-1-苯並吖呯-2-硫酮(實施例91之步驟a))(0.40 g,1.68毫莫耳)、碳酸鉀(6.60 g,4.78毫莫耳)及丙酮(30 ml)之混合物加入碘甲烷(0.297 ml,4.78毫莫耳)並令反應混合物於40℃下經攪拌1小時。經蒸發溶劑後,將水和乙酸乙酯加入至殘餘物,分離相層且令水相經乙酸乙酯萃取。令合併之有機相經飽和NaCl溶液沖洗,置於無水硫酸鈉上乾燥且經過濾和濃縮。因此,得到標的產物(0.40 g,99%),其未經進一步純化而使用。

c) 8-氟-5,5-二甲基-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯
對7-氟-4,4-二甲基-2-(甲硫基)-4,5-二氫-3H-1-苯並吖呯(實施例91之步驟b))(0.30 g,1.26毫莫耳)、反式-4-(吡啶-2-基氧基)環己烷甲醯肼(WO2010/060836 A1 (2010.06.03) F. HOFFMANN-LA ROCHE AG.)(0.30 g,1.26毫莫耳)及1,4-二噁烷(20 ml)之混合物加入濃氫氯酸(0.010 ml)並令反應混合物經回流3小時。經蒸發溶劑後,令殘餘物經管柱層析(使用二氯甲烷:甲醇=20:1作為溶析液)純化。因此,得到標的產物(0.108 mg,21%)。MS (ESI) m/z 407.2 (M+H)+

實施例92
8-氟-5-(丙-2-基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯

依據實施例91描述之方法,自7-氟-4-(丙-2-基)-1,3,4,5-四氫-2H-1-苯並吖呯-2-酮(中間體54)製備標的產物。MS (ESI) m/z 421.2 (M+H)+

實施例93
8’-溴-1’-[反式-4-(吡啶-2-基氧基)環己基]-4’H,6’H-螺[環戊烷-1,5’-[1,2,4]三唑並[4,3-a][1]苯並吖呯]

依據實施例91描述之方法,自7-溴-1,5-二氫螺[1-苯並吖呯-4,1’-環戊烷]-2(3H)-酮(中間體60)製備標的產物。MS (ESI) m/z 493.1和495.1 (M+H)+

實施例94
8-溴-5-(丙-2-基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯

依據實施例91描述之方法,自7-溴-4-(丙-2-基)-1,3,4,5-四氫-2H-1-苯並吖呯-2-酮(中間體61)製備標的產物。MS (ESI) m/z 481.1和483.1 (M+H)+

實施例95
8’-溴-1’-[反式-4-(吡啶-2-基氧基)環己基]-2,3,5,6-四氫-4’H,6’H-螺[哌喃-4,5’-[1,2,4]三唑並[4,3-a][1]苯並吖呯]

a) 7-溴-2-甲氧基-3,5-二氫螺[1-苯並吖呯-4,4’-環氧乙烷]

令7-溴-1,5-二氫螺[1-苯並吖呯-4,4’-環氧乙烷]-2(3H )-酮(中間體62)(0.193 g,062毫莫耳)、四氟硼酸三甲基氧鎓(0.183 g,1.24毫莫耳)、碳酸鉀(0.343 g,2.48毫莫耳)及二氯甲烷(30 ml)之混合物於室溫下經攪拌3小時。隨後令反應混合物經過濾並令濾液經濃縮。因此,得到標的產物(0.190 g,98%),其未經進一步純化而使用。

b) 8’-溴-1’-[反式-4-(吡啶-2-基氧基)環己基]-2,3,5,6-四氫-4’H,6’H-螺[哌喃-4,5’-[1,2,4]三唑並[4,3-a][1]苯並吖呯]
對7-溴-2-甲氧基-3,5-二氫螺[1-苯並吖呯-4,4’-環氧乙烷](0.190 g,0.62毫莫耳)、反式-4-(吡啶-2-基氧基)環己烷甲醯肼(WO2010/060836 A1 (03.06.2010) F. HOFFMANN-LA ROCHE AG.)(0.161 g,0.68毫莫耳)及1,4-二噁烷(20 ml)之混合物加入三氟乙酸(0.010 ml)並令反應混合物經回流3小時。經蒸發溶劑後,令殘餘物經管柱層析(使用二氯甲烷:甲醇=20:1作為溶析液)純化。因此,得到標的產物(0.144 mg,45%)。MS (ESI) m/z 509.2和511.2 (M+H)+

實施例96
8-氟-5,5-二甲基-1-[反式-4-(三氟甲基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯

依據實施例91之步驟c)描述之方法,自7-氟-4,4-二甲基-2-(甲硫基)-4,5-二氫-3H-1-苯並吖呯(實施例91之步驟b))和反式-4-(三氟甲基)環己烷甲醯肼(中間體6)製備標的產物。MS (ESI) m/z 382.2 (M+H)+

實施例97
(5R)-8-溴-5-(丙-2-基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯


實施例98
(5S)-8-溴-5-(丙-2-基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯

藉由手性製備性HPLC (CHIRALPAK IC製備性20 μm固定相;2.5 x 20 cm;F=20 ml/分鐘;溶析液:三級丁基甲基醚:二氯甲烷:乙醇=85:13:2;等度;溫度=25℃),自消旋性8-溴-5-(丙-2-基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯(實施例94)製備標的產物。藉由分析性HPLC (CHIRALPAK IA 5 μm固定相;250 x 4.6 mm;F=1 ml/分鐘;溶析液:三級丁基甲基醚:二氯甲烷:乙醇=85:10:5;等度;溫度=35℃)測定滯留時間:第一溶析物係15.0分鐘;=-5.0˚ (c=0.1;二氯甲烷);MS (ESI) m/z 481.2和483.2 (M+H)+ ;第二溶析物係18.5分鐘;=+10.0˚ (c=0.1;二氯甲烷);MS (ESI) m/z 481.2和483.2 (M+H)+ 。未測定該等化合物之絕對構型。

實施例99
8’-溴-1’-[反式-4-(吡啶-2-基氧基)環己基]-4,5-二氫-4’H,6’H-螺[呋喃-3,5’-[1,2,4]三唑並[4,3-a][1]苯並吖呯]

依據實施例95描述之方法,自7-溴-1,5-二氫螺[1-苯並吖呯-4,3’-四氫呋喃]-2(3H)-酮(中間體63)製備標的產物。MS (ESI) m/z 495.1和497.2 (M+H)+

實施例100
8’-氯-1’-[反式-4-(吡啶-2-基氧基)環己基]-2,3,5,6-四氫-4’H,6’H-螺[哌喃-4,5’-[1,2,4]三唑並[4,3-a][1]苯並吖呯]

依據實施例95描述之方法,自7-氯-1,5-二氫螺[1-苯並吖呯-4,4’-環氧乙烷]-2(3H)-酮(中間體64)製備標的產物。MS (ESI) m/z 465.2 (M+H)+

實施例101
8’-氯-1’-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H,4’H,6’H-螺[哌喃-3,5’-[1,2,4]三唑並[4,3-a][1]苯並吖呯]

依據實施例95描述之方法,自7-氯-1,5-二氫螺[1-苯並吖呯-4,3’-環氧乙烷]-2(3H)-酮(中間體65)製備標的產物。MS (ESI) m/z 465.3 (M+H)+
實施例102
8’-氯-1’-[反式-4-(吡啶-2-基氧基)環己基]-4,5-二氫-4’H,6’H-螺[呋喃-3,5’-[1,2,4]三唑並[4,3-a][1]苯並吖呯]

依據實施例95描述之方法,自7-氯-1,5-二氫螺[1-苯並吖呯-4,3’-四氫呋喃]-2(3H)-酮(中間體66)製備標的產物。MS (ESI) m/z 451.2 (M+H)+
實施例103
8’-氯-1’-[反式-4-(三氟甲基)環己基]-2,3,5,6-四氫-4’H,6’H-螺[哌喃-4,5’-[1,2,4]三唑並[4,3-a][1]苯並吖呯]

依據實施例91描述之方法,自7-氯-1,5-二氫螺[1-苯並吖呯-4,4’-環氧乙烷]-2(3H)-酮(中間體64)和反式-4-(三氟甲基)環己烷甲醯肼(中間體6)製備標的產物。MS (ESI) m/z 440.2 (M+H)+

實施例104
(3R)-8’-溴-1’-[反式-4-(吡啶-2-基氧基)環己基]-4,5-二氫-4’H,6’H-螺[呋喃-3,5’-[1,2,4]三唑並[4,3-a][1]苯並吖呯]


實施例105
(3S)-8’-溴-1’-[反式-4-(吡啶-2-基氧基)環己基]-4,5-二氫-4’H,6’H-螺[呋喃-3,5’-[1,2,4]三唑並[4,3-a][1]苯並吖呯]

藉由手性製備性HPLC (CHIRALPAK IG製備性20 μm固定相;5 x 30 cm;F=50 ml/分鐘;溶析液:正庚烷:乙醇=6:4;等度;溫度=25℃),自消旋性8’-溴-1’-[反式-4-(吡啶-2-基氧基)環己基]-4,5-二氫-4’H,6’H-螺[呋喃-3,5’-[1,2,4]三唑並[4,3-a][1]苯並吖呯](實施例99)製備標的產物。藉由分析性HPLC (CHIRALPAK IG 5 μm固定相;250 x 4.6 mm;F=1 ml/分鐘;溶析液:正庚烷:乙醇=6:4;等度;溫度=25℃)測定滯留時間:第一溶析物係18.6分鐘;=+10.0˚(c=0.11;CHCl3 );MS (ESI) m/z 495.2和497.2 (M+H)+ ;第二溶析物係23.0分鐘;=-3.8˚ (c=0.105;CHCl3 );MS (ESI) m/z 495.2和497.2 (M+H)+ 。未測定該等化合物之絕對構型。

實施例106
(3S)-8’-氯-1’-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H,4’H,6’H-螺[哌喃-3,5’-[1,2,4]三唑並[4,3-a][1]苯並吖呯]


實施例107
(3R)-8’-氯-1’-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H,4’H,6’H-螺[哌喃-3,5’-[1,2,4]三唑並[4,3-a][1]苯並吖呯]

藉由手性製備性HPLC (CHIRALPAK IG製備性20 μm固定相;5 x 30 cm;F=50 ml/分鐘;溶析液:正庚烷:乙醇=6:4;等度;溫度=25℃),自消旋性8’-氯-1’-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H,4’H,6’H-螺[哌喃-3,5’-[1,2,4]三唑並[4,3-a][1]苯並吖呯](實施例101)製備標的產物。藉由分析性HPLC (CHIRALPAK IG 5 μm固定相;250 x 4.6 mm;F=1 ml/分鐘;溶析液:正庚烷:乙醇=6:4;等度;溫度=25℃)測定滯留時間:第一溶析物係20.2分鐘;=+19.0˚ (c=0.11;CHCl3 );MS (ESI) m/z 465.2 (M+H)+ ;第二溶析物係26.5分鐘;=-7.6˚ (c= 0.105;CHCl3 );MS (ESI) m/z 465.2 (M+H)+ 。未測定該等化合物之絕對構型。

實施例108
8-氟-1-(反式-4-甲氧基-4-甲基環己基)-5,5-二甲基-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯


實施例109
8-氟-1-(順式-4-甲氧基-4-甲基環己基)-5,5-二甲基-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯

對7-氟-4,4-二甲基-2-(甲硫基)-4,5-二氫-3H-1-苯並吖呯(實施例91之步驟b))(0.15 g,0.63毫莫耳)、4-甲氧基-4-甲基環己烷-1-甲醯肼(中間體35之步驟c))(0.18 g,0.95毫莫耳)及1,4-二噁烷(20 ml)之混合物加入濃氫氯酸(0.010 ml)並令反應混合物經回流3小時。經蒸發溶劑後,令殘餘物經製備性HPLC (Chromolith製備性RP18管柱;100 × 25 mm;25 ml/分鐘;A:水+0.1%三氟乙酸;B:乙腈+0.1%三氟乙酸;梯度:0至12分鐘;B:0至49%;12至24分鐘;B:49至90%;溫度=40℃)純化。藉由分析性HPLC (Chromolith效能RP18管柱;100 × 4.6 mm;1 ml/分鐘;溶析液:A:水+0.1%三氟乙酸;B:乙腈+0.1%三氟乙酸;梯度:0至10分鐘;B:0至90%;10至15分鐘;B:90%;溫度=40℃)測定滯留時間:第一溶析物係8.62分鐘;反式異構物;0.088 g (39%);MS (ESI) m/z 358.3 (M+H)+ ;第二溶析物係8.95分鐘;順式異構物;0.021 g (9%);MS (ESI) m/z 358.2 (M+H)+

實施例110
8’-氯-1’-(反式-4-甲氧基-4-甲基環己基)-2,3,5,6-四氫-4’H,6’H-螺[哌喃-4,5’-[1,2,4]三唑並[4,3-a][1]苯並吖呯]


實施例111
8’-氯-1’-(順式-4-甲氧基-4-甲基環己基)-2,3,5,6-四氫-4’H,6’H-螺[哌喃-4,5’-[1,2,4]三唑並[4,3-a][1]苯並吖呯]

a) 7-氯-2-甲氧基-3,5-二氫螺[1-苯並吖呯-4,4’-環氧乙烷]

令7-氯-1,5-二氫螺[1-苯並吖呯-4,4’-環氧乙烷]-2(3H)-酮(中間體54)(0.35 g,1.32毫莫耳)、四氟硼酸三甲基氧鎓(0.583 g,3.95毫莫耳)、碳酸鉀(0.73 g,5.27毫莫耳)及二氯甲烷(25 ml)之混合物於室溫下經攪拌3小時。隨後令反應混合物經過濾並令有機相經濃縮。因此,得到標的產物(0.35 g,98%),其未經進一步純化和性質界定而用於下一個反應步驟。

b) 8’-氯-1’-(4-甲氧基-4-甲基環己基)-2,3,5,6-四氫-4’H,6’H-螺[哌喃-4,5’-[1,2,4]三唑並[4,3-a][1]苯並吖呯]
對7-氯-2-甲氧基-3,5-二氫螺[1-苯並吖呯-4,4’-環氧乙烷](0.35 g,1.31毫莫耳)、4-甲氧基-4-甲基環己烷-1-甲醯肼(中間體35之步驟c))(0.74 g,3.95毫莫耳)及1,4-二噁烷(20 ml)之混合物加入三氟乙酸(0.030 ml)並令反應混合物於75℃下經隔夜攪拌。經蒸發溶劑後,令殘餘物經製備性HPLC (Kinetex EVO管柱;5 μm負載;150 × 21.2 mm;20 ml/分鐘;溶析液:20毫莫耳/L (NH4 )2 CO3 :乙腈=65:35;等度;溫度=40℃)純化。藉由分析性HPLC (Kinetex EVO管柱;5 μm負載;150 × 4.6 mm;1 ml/分鐘;溶析液:20毫莫耳/L (NH4 )2 CO3 :乙腈=65:35;等度;溫度=40℃)測量滯留時間:第一溶析物係5.53分鐘;反式異構物;0.056 g (10%);MS (ESI) m/z 416.2 (M+H)+ ;第二溶析物係8.06分鐘;順式異構物;0.016 g (3%);MS (ESI) m/z 416.3 (M+H)+

實施例112
8-氯-1-[反式-4-(吡啶-2-基胺基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯

依據實施例1描述之方法,自7-氯-2-側硫基-2,3,4,5-四氫-1H -1-苯並吖呯-4-甲酸甲酯(中間體4)和反式-4-(吡啶-2-基胺基)環己烷甲醯肼(中間體67)製備標的產物。MS (ESI) m/z 452.1 (M+H)+
下述調製劑實施例說明本發明之代表性醫藥組成物。然而,本發明不受限於下述醫藥組成物。

The invention also relates to the synthesis of compounds of general formula (I). Thus, the compound of the general formula (I) of the present invention can be prepared by one of the following methods.
So far, in the compound of general formula (I), R 2 System hydrogen, R 3 C (O) R 5 , R 5 Department C 1-4 Alkoxy and ring A is a cycloalkyl or a 4- to 7-membered saturated heterocyclic ring containing 1 N, in which ring A is connected to Y via a ring nitrogen, by combining a compound of general formula (II) with general formula (III) Compounds are reacted to prepare compounds of general formula (I) of the present invention, wherein the compounds of general formula (II) are

Wherein ring B and Y are as defined in the general formula (I) above and ring A is a cycloalkyl group or a 4- to 7-membered saturated heterocyclic ring containing one N, wherein ring A is connected to Y via a ring nitrogen, and the general Compound of formula (III)

Where R 1 Is defined by the general formula (I) above, R 3 C (O) R 5 And R 5 Department C 1-4 Alkoxy.
The reaction is shown in detail in Figure 1:

In step a) of FIG. 1, the hydrazine of the general formula (II) is reacted with a benzoazepine-thione of the general formula (III). The reaction is preferably carried out in a suitable solvent and at the boiling point of the solvent with a required reaction time of 4 to 120 hours. Suitable solvents include, for example, xylene, n-butanol and 1,4-dioxane.
Preferred implementations are, for example, the following:
i) the reaction of formula (II) and formula (III) in xylene at 140 ° C for 20 to 120 hours; or
ii) The reaction of formula (II) and formula (III) is carried out in n-butanol at 110 ° C for 20 to 50 hours.
The hydrazine of general formula (II) can be synthesized by a number of different methods (Figure 2):

In step a) of FIG. 2, the formate of the general formula (IV) is in a suitable alcohol and reacted with the hydrazine hydrate at the boiling point of the solvent to obtain the hydrazine of the general formula (II), or in step c ), The formic acid of the general formula (V) is reacted with tert-butyl hydrazinoformate and the protecting group of the protected formamidine hydrazine derivative of the general formula (VI) is removed using an appropriate acid (step d)).
Preferred implementations are, for example, the following:
Step a) methanol or ethanol; hydrazine hydrate; reflux temperature; 4 to 50 hours;
Step b) methanol; thionyl chloride; 0 to 25 ° C; 4 to 24 hours;
Step c) tert-butyl hydrazinecarboxylic acid; N, N-dimethylformamide; N, N-diisopropylethylamine; N- (3-dimethylaminopropyl) -N'- Ethylcarbodiimide hydrochloride; 1-hydroxybenzotriazole hydrate; room temperature; 4 to 20 hours;
Step d) Hydrochloric acid in ethyl acetate; room temperature; 4 to 20 hours.
The formate of the general formula (IV) and the formic acid of the general formula (V) are commercially available or can be prepared according to the method described in the examples.
According to the method shown in FIG. 3, the main intermediate benzoazepine-thione derivative (wherein R is 1 (As defined by the general formula (I) above):

Acid derivatives of general formula (VII) (where R 1 (As defined by the general formula (I) above) is esterified (step a)) to form an ester derivative of the general formula (VIII), and the general formula (IX) is prepared from the ester derivative of the general formula (VIII) Oxime derivative (step b)). By Beckmann rearrangement, the oxime derivative of the general formula (IX) is converted into a benzoacridine compound of the general formula (X) (step c)), and from the benzene of the general formula (X) The acridine compound is used to prepare a benzoacridine-thione derivative of the general formula (III-a) (step d)).
Preferred implementations are, for example, the following:
Step a) methanol; concentrated sulfuric acid; reflux temperature; 4 to 20 hours;
Step b) methanol; sodium acetate; hydroxylamine hydrochloride; reflux temperature; 2 to 6 hours;
Step c) polyphosphoric acid; 100 to 120 ° C; 15 to 60 minutes;
Step d) i) Lawesson's reagent; pyridine; reflux temperature; 2 to 10 hours; or
ii) Lawson's reagent; tetrahydrofuran; room temperature; 6 to 20 hours.
Benzacridine derivatives of general formula (X) 1 (As defined by the general formula (I) above) to prepare other intermediates shown in FIG. 4 to FIG. 6.

In step a) of FIG. 4, an ethylamidine derivative of the general formula (XI) can be prepared by one step or three consecutive steps. The compound of the general formula (XI) is a main intermediate, and from the compound of the general formula (XI), a dioxane compound of the general formula (XII) (step b)) and a difluoro derivative of the general formula (XIII) can be prepared. (Step c)), protected compounds of the general formula (XIV) (step d)) (where PG 1 Is a protective group (Peter GM Wuts: Greene's Protective Groups in Organic Synthesis: Fifth Edition, Chapter 7. Protection for the Amino Group, pages 895-1193), preferably 4-methoxybenzyl-) and the general formula (XV -a) a hydroxy compound (step e-1)). A hydroxy derivative of the general formula (XV) is prepared by reducing the compound of the general formula (XIV) (step e-2)), and a unit of the general formula (XVI) can be prepared from the hydroxy derivative of the general formula (XV) Fluorine compound (step f)). A compound of the general formula (XVII) can be obtained by removing the protecting group of the monofluoro compound of the general formula (XVI) (step g)). By protecting the free hydroxyl group, this unprotected hydroxyl derivative of formula (XV-a) can be converted into a compound of formula (XIX) (step h)), where PG 2 Department and PG 1 Different protective groups (Peter GM Wuts: Greene's Protective Groups in Organic Synthesis: Fifth Edition, Chapter 2. Protection for the Hydroxyl Group, Including 1,2- and 1,3-Diols, pages 17-471), preferably silicon Radical protection radical.
Preferred implementations are, for example, the following:
Step a) methyl lithium solution; tetrahydrofuran; diethyl ether; (-10 ° C) to 5 ° C; 2 to 3 hours; or
Step i) aqueous sodium hydroxide solution; methanol; room temperature; 0.5 to 3 hours;
Step ii) N-methoxymethylamine hydrochloride; N -(3-dimethylaminopropyl)- N ' -Ethylcarbodiimide hydrochloride; N, N-dimethylformamide; N, N-diisopropylethylamine or triethylamine; 1-hydroxybenzotriazole hydrate; room temperature; 2 to 20 hours; then
Step iii) Diethyl ether solution of methyl magnesium bromide; tetrahydrofuran; (-10 ° C) to 0 ° C; 2 to 12 hours;
Step b) ethylene glycol; p-toluenesulfonic acid; toluene; reflux temperature; 6 to 12 hours;
Steps c) and f) diethylaminosulfur trifluoride; dichloromethane; (-78) to 10 ° C; 4 to 12 hours;
Step d) 4-methoxybenzyl chloride; sodium hydride; N, N-dimethylformamide; 0 to 25 ° C; 3 to 6 hours;
Steps e-1) and e-2) sodium borohydride; ethanol or methanol; room temperature; 0.5 to 2 hours;
Step g) i) ammonium cerium nitrate; water; acetonitrile; 0 to 25 ° C; 6 to 18 hours; or
ii) trifluoroacetic acid; dichloromethane; room temperature; 12 to 24 hours; or
iii) trifluoromethanesulfonic acid; dichloromethane; room temperature; 2 to 12 hours;
Step h) tert-butyldimethylsilyl trifluoromethanesulfonate; 2,6-dimethylpyridine; dichloromethane; N, N- Dimethylformamide; 0 to 25 ° C; 6 to 18 hours.
In FIG. 5, a protective group is provided for the alcohol derivative of the general formula (XV) (step a)), where PG 1 Is a protecting group, preferably 4-methoxybenzyl, and PG 2 Department and PG 1 Different protection groups, preferably silicon protection groups (Peter GM Wuts: Greene's Protective Groups in Organic Synthesis: Fifth Edition, Chapter 2. Protection for the Hydroxyl Group, Including 1, 2- and 1, 3-Diols, pages 17 -471) to obtain a compound of formula (XVIII). Removal of the protecting group PG from the compound of formula (XVIII) 1 (Step b)) to obtain a compound of general formula (XIX). By alkylating the hydroxy compound of the general formula (XV), a compound of the general formula (XX) (wherein the alkyl group represents C 1-4 Alkyl) (step c)), and then by removing the PG of the compound of formula (XX) 1 A protecting group can prepare a compound of the general formula (XXI).

Preferred implementations are, for example, the following:
Step a) trimethylchlorosilane; imidazole; N, N-dimethylformamide; room temperature; 4 to 12 hours;
Steps b) and d) i) cerium ammonium nitrate; water; acetonitrile; 0 to 25 ° C; 6 to 18 hours; or
ii) trifluoroacetic acid; dichloromethane; room temperature; 12 to 24 hours; or
iii) trifluoromethanesulfonic acid; dichloromethane; room temperature; 2 to 12 hours;
Step c) i) sodium hydride; haloalkane; N, N-dimethylformamide; 0 to 100 ° C; 12 to 48 hours; or
ii) sodium hydride; dialkyl sulfonate; N, N-dimethylformamide; 0 to 100 ° C; 12 to 48 hours; or
iii) silver oxide; halothane; N, N-dimethylformamide; room temperature; 48 to 120 hours.
As shown in FIG. 6, the nitrogen atom of the benzoazine derivative of the general formula (X) is protected by a protecting group (step a)) to obtain the protected benzoazine derivative of the general formula (XXII) (wherein PG 1 Is a protective group (Peter GM Wuts: Greene's Protective Groups in Organic Synthesis: Fifth Edition, Chapter 7. Protection for the Amino Group, pages 895-1193), preferably 4-methoxybenzyl), and then the ester group Reduction (step b)) to obtain a hydroxy compound of the general formula (XXIII). A compound of the general formula (XXV) is prepared by alkylating the hydroxy compound of the general formula (XXIII) (wherein the alkyl group represents C 1-4 Alkyl) (step c)), and then by removing the protecting group PG 1 (Step d)) to obtain a compound of the general formula (XXIV). In step e), a benzoazine-thione derivative of the general formula (III-b) is prepared.

Preferred implementations are, for example, the following:
Step a) 4-methoxybenzyl chloride; sodium hydride; N, N-dimethylformamide; 0 to 25 ° C; 3 to 6 hours;
Step b) lithium aluminum hydride; tetrahydrofuran; (-20) to 0 ° C; 0.1 to 1 hour;
Step c) i) sodium hydride; haloalkane; N, N-dimethylformamide; 0 to 100 ° C; 12 to 48 hours; or
ii) sodium hydride; dialkyl sulfonate; N, N-dimethylformamide; 0 to 100 ° C; 12 to 48 hours; or
iii) silver oxide; halothane; N, N-dimethylformamide; room temperature; 48 to 120 hours;
Step d) i) ammonium cerium nitrate; water; acetonitrile; 0 to 25 ° C; 6 to 18 hours; or
ii) trifluoroacetic acid; dichloromethane; room temperature; 12 to 24 hours; or
iii) trifluoromethanesulfonic acid; dichloromethane; room temperature; 2 to 12 hours;
Step e) i) Lawson's reagent; pyridine; reflux temperature; 2 to 10 hours; or
ii) Lawson's reagent; tetrahydrofuran; room temperature; 6 to 20 hours.
By reacting a compound of the general formula (II) with a compound of the general formula (III-a), a compound of the general formula (Ib) can be prepared (FIG. 7).

Where rings B, Y, and R 1 As defined in the above general formula (I), ring A is a cycloalkyl group or a 4- to 7-membered saturated heterocyclic ring containing 1 N, wherein ring A is connected to Y via a ring nitrogen.
Preferred implementations are, for example, the following:
i) the reaction of formula (II) and formula (III-a): xylene; 140 ° C; 20 to 120 hours; or
ii) the reaction of formula (II) and formula (III-a): n-butanol; 110 ° C; 20 to 50 hours; or
iii) Reaction of formula (II) with formula (III-a): 1,4-dioxane; 110 ° C; 20 to 50 hours.
As desired, mirror image isomers are prepared by conventional methods by introducing new substituents and / or modifying or removing existing substituents and / or forming a salt and / or liberating a base from a salt and / or a racemic mixture. The compound of the general formula (Ib) thus obtained can also be converted into another compound of the general formula (I). This reaction scheme is described in detail in FIGS. 8 and 9.

By hydrolyzing the compound of the general formula (Ib) (step a)), the compound of the general formula (Ic) (wherein rings B, Y and R 1 Is as defined in the above general formula (I), ring A is a cycloalkyl group or a 4- to 7-membered saturated heterocyclic ring containing 1 N, wherein ring A is connected to Y via a ring nitrogen), and then from this general formula ( Ic) Compounds can be prepared from amidines of the general formula (Id) (step b)) or amidines of the general formula (Ig) (step e)) (wherein rings B, Y and R 1 As defined by the general formula (I) above, ring A is a cycloalkyl group or a 4- to 7-membered saturated heterocyclic ring containing one N, wherein ring A is connected to Y via a ring nitrogen, R 'represents hydrogen, and R "represents C 1-4 Alkyl or Cy 2 , Or R 'and R "together with the N to which they are attached form a heterocycle). Nitriles of the general formula (Id) can be prepared from nitriles of the general formula (Ie) (step c)), and from the general formula ( Tetrazoles of general formula (If) can be prepared from nitriles of Ie) (step d)). Oxadiazole derivatives of general formula (Ih) can also be prepared from steps (f) of acids of the general formula (Ic).
Preferred implementations are, for example, the following:
Step a) methanol; sodium hydroxide; room temperature; 1 to 20 hours;
Step b) ammonium chloride; N- (3-dimethylaminopropyl) -N'-ethylcarbodiimide hydrochloride; N, N-dimethylformamide; N, N-di Isopropylethylamine or triethylamine; 1-hydroxybenzotriazole hydrate; room temperature; 4 to 20 hours;
Step c) pyridine; phosphorous chloride; room temperature; 2 to 20 hours;
Step d) sodium azide; ammonium chloride; N, N-dimethylformamide; 100 to 130 ° C; 2 to 8 hours;
Step e) i) Appropriate amine derivative; N- (3-dimethylaminopropyl) -N'-ethylcarbodiimide hydrochloride; N, N-dimethylformamide; N , N-diisopropylethylamine or triethylamine; 1-hydroxybenzotriazole hydrate; room temperature; 4 to 20 hours; or
ii) chloramphenicol; dichloromethane; room temperature; 0.5 to 2 hours; subsequent pyridine and appropriate amine derivatives; room temperature; 1 to 5 hours;
Step f) N- (3-dimethylaminopropyl) -N'-ethylcarbodiimide hydrochloride; N, N-dimethylformamide; 1-hydroxybenzotriazole hydrate N-hydroxyacetamidamine; 80 to 120 ° C; 4 to 20 hours.
In addition, by reducing the compound of the general formula (Ib), a hydroxymethyl derivative of the general formula (Ii) can be prepared (FIG. 9; step a)).

Where rings B, Y, and R 1 As defined in the above general formula (I), ring A is a cycloalkyl group or a 4- to 7-membered saturated heterocyclic ring containing 1 N, wherein ring A is connected to Y via a ring nitrogen.
As desired, mirror image isomers are prepared by conventional methods by introducing new substituents and / or modifying or removing existing substituents and / or forming a salt and / or liberating a base from a salt and / or a racemic mixture. Structure, the compound of general formula (Ii) can also be converted into another compound of general formula (I). For example, compounds can be prepared by catalytic hydrogenation reactions where R 1 A hydrogen atom (not shown). By alkylation or halogenation reaction, other derivatives can be prepared to obtain compounds of general formula (Ij) (step b)), in which ring B, Y and R 1 As defined in the above general formula (I), ring A is a cycloalkyl group or a 4- to 7-membered saturated heterocyclic ring containing 1 N, wherein ring A is connected to Y via a ring nitrogen, and R is an alkyl group or a fluorenyl group. .
From the compound of the general formula (Ik) obtained by the methane sulfonation of the compound of the general formula (Ii) (step c)), a nitrile derivative of the general formula (Il) (step d)), A fluorine compound of formula (Im) (step e)) and an amine derivative of formula (In) (step f)), wherein rings B, Y and R 1 As defined in the above general formula (I), ring A is a cycloalkyl group or a 4- to 7-membered saturated heterocyclic ring containing 1 N, wherein ring A is connected to Y via a ring nitrogen, and R 10 And R 11 Is optionally substituted C as defined by the general formula (I) above 1-4 Alkyl, or R 10 And R 11 Together with the N to which they are attached forms an optionally substituted heterocyclic ring. In the general formula (Ij), when OR represents a carbamate, a compound can be prepared from the compound of the general formula (Ik) according to step g).
Preferred implementations are, for example, the following:
Step a) methylene chloride; diethyl ether; lithium aluminum hydride; (-30) to (-10) ° C; 15 to 60 minutes;
Step b) i) sodium hydride; haloalkane; N, N-dimethylformamide or tetrahydrofuran; (-5) to 40 ° C; 2 to 10 hours; or
ii) sodium hydride; acetonitrile; isocyanate derivatives; room temperature; 4 to 20 hours; or
iii) fluorenyl chloride; pyridine; room temperature; 4 to 20 hours; or
iv) fluorenyl chloride; methylene chloride; triethylamine or N, N-diisopropylethylamine; room temperature; 4 to 20 hours; or
v) anhydride; pyridine; room temperature; 4 to 20 hours; or
vi) acid; hexafluorophosphate N, N, N ', N'-tetramethyl-O- (1H-benzotriazol-1-yl) urenium; N, N-dimethylformamide; N , N-diisopropylethylamine; room temperature; 4 to 20 hours; or
vii) acid; N- (3-dimethylaminopropyl) -N'-ethylcarbodiimide hydrochloride; N, N-diisopropylethylamine; N, N-dimethylformamide Amidine; 1-hydroxybenzotriazole hydrate; room temperature; 4 to 20 hours;
Step c) methanesulfonyl chloride; dichloromethane; triethylamine; 4-dimethylaminopyridine; room temperature; 2 to 20 hours;
Step d) sodium cyanide; N, N-dimethylformamide; 70 to 100 ° C; 3 to 20 hours;
Step e) tetrabutylammonium fluoride; tetrahydrofuran; 50 to 70 ° C; 4 to 20 hours;
Step f) amine; N, N-dimethylformamide; N, N-diisopropylethylamine; 80 to 120 ° C; 3 to 20 hours;
Step g) secondary amine; N, N-dimethylformamide; triethylamine; cesium carbonate; 100 ° C; 12 to 48 hours.
From this compound of general formula (Ik), other derivatives shown in FIG. 10 can be prepared.

Step a) generates an azide compound of general formula (Io), wherein rings B, Y and R 1 As defined in the above general formula (I), ring A is a cycloalkyl group or a 4- to 7-membered saturated heterocyclic ring containing 1 N, wherein ring A is connected to Y via a ring nitrogen, and from this general formula (Io) The azide compound can be reduced to prepare an amine derivative of the general formula (Ip) (step b)). The amine derivative of the general formula (Ip) is a starting material of an alkyl compound of the general formula (Iq) (step c)) and a fluorenyl compound of the general formula (Ir) (step d)), in which ring B, Y And R 1 As defined in the general formula (I) above, ring A is a cycloalkyl group or a 4- to 7-membered saturated heterocyclic ring containing 1 N, wherein ring A is connected to Y via a ring nitrogen, and R "'stands for optionally via Replaced by C 1-4 Alkyl or R as in the general formula (I) above 10 And R 11 Defined Cy 3 And the fluorene group is as R in the general formula (I) 10 And R 11 C (O) R as defined 13 . According to step e), the compound of general formula (In) can also be prepared from the compound of general formula (Ip), where R 10 And R 11 Is optionally substituted C 1-4 alkyl.
Preferred implementations are, for example, the following:
Step a) sodium azide; N, N-dimethylformamide; 60 to 100 ° C; 2 to 8 hours;
Step b) triphenylphosphine; tetrahydrofuran; water; room temperature; 4 to 20 hours;
Step c) i) sodium hydride; haloalkane; N, N-dimethylformamide or tetrahydrofuran; (-5) to 40 ° C; 2 to 10 hours; or
ii) ketones; 1,2-dichloroethane; acetic acid; sodium triethoxylate borohydride; room temperature; 4 to 20 hours;
Step d) i) fluorenyl chloride; pyridine; room temperature; 4 to 20 hours; or
ii) fluorenyl chloride; dichloromethane; triethylamine or N, N-diisopropylethylamine; room temperature; 4 to 20 hours; or
iii) acid anhydride; pyridine; room temperature; 4 to 20 hours; or
iv) acid; hexafluorophosphate N, N, N ', N'-tetramethyl-O- (1H-benzotriazol-1-yl) urenium; N, N-dimethylformamide; N , N-diisopropylethylamine; room temperature; 4 to 20 hours; or
v) acid; N- (3-dimethylaminopropyl) -N'-ethylcarbodiimide hydrochloride; N, N-diisopropylethylamine; N, N-dimethylformamide Amidine; 1-hydroxybenzotriazole hydrate; room temperature; 4 to 20 hours;
Step e) Aldehyde; 1,2-dichloroethane; acetic acid; sodium triethoxyalkoxyborohydride; room temperature; 4 to 20 hours.
According to FIG. 11, a methoxybenzoazine derivative of the general formula (XXVI) is prepared from methylation of a compound of the general formula (XII) (step a)), where R 1 Is as defined in the above general formula (I), and the methoxybenzoacridine derivative of the general formula (XXVI) is further reacted with the hydrazine of the general formula (II) without isolation (step b)) to Generates compounds of general formula (Is), in which rings B, Y, and R 1 As defined in the above general formula (I), ring A is a cycloalkyl group or a 4- to 7-membered saturated heterocyclic ring containing 1 N, wherein ring A is connected to Y via a ring nitrogen. After removing the ketal protecting group (step c)), a pendant oxygen compound of the general formula (It) is obtained, in which the rings B, Y and R 1 As defined in the above general formula (I), ring A is a cycloalkyl group or a 4- to 7-membered saturated heterocyclic ring containing 1 N, wherein ring A is connected to Y via a ring nitrogen. Reduction of the compound of general formula (It) produces a hydroxy compound of general formula (Iu) (step d)). The compound of the general formula (Iu) can be oxidized (step e)) using an appropriate oxidant to form the compound of the general formula (It), and the compound of the general formula (It) can be further reacted to obtain the general formula (I) -aa), wherein R represents C optionally substituted with alkoxy 1-4 alkyl.

Preferred implementations are, for example, the following:
Step a) methylene chloride; trifluoroacetic acid or potassium carbonate; trimethyloxonium tetrafluoroborate; room temperature; 20 to 25 hours;
Step b) hydrazine of general formula (II); dichloromethane or acetonitrile; 50 ° C; 6 to 20 hours;
Step c) methanol; concentrated hydrochloric acid; 70 ° C; 2 to 6 hours;
Step d) sodium borohydride; ethanol or methanol; room temperature; 1 to 2 hours;
Step e) Dess-Martin periodinane; methylene chloride; 0 ° C to room temperature; 1 to 5 hours;
Step f) Appropriate amine; 1,2-dichloroethane; sodium triethoxyhoxyborohydride; acetic acid; room temperature;
A compound of the general formula (Ij) can be prepared by reacting a compound of the general formula (II) with a compound of the general formula (III-b), wherein the alkyl group represents C 1-4 Alkyl and rings B, Y, and R 1 As defined in the above general formula (I), ring A is a cycloalkyl group or a 4- to 7-membered saturated heterocyclic ring containing 1 N, wherein ring A is connected to Y via a ring nitrogen (FIG. 12).

Preferred implementations are, for example, the following:
i) the reaction of formula (II) and formula (III-b): xylene; 140 ° C; 20 to 120 hours; or
ii) the reaction of formula (II) and formula (III-b): n-butanol; 110 ° C; 20 to 50 hours; or
iii) Reaction of formula (II) with formula (III-b): 1,4-dioxane; 110 ° C; 20 to 50 hours.
A benzoazine-thione derivative of the general formula (III-c) is prepared from the difluoro derivative of the general formula (XIII) (Figure 13; step a)), where R 1 Is as defined in the above general formula (I), and the benzoazepine-thione derivative of the general formula (III-c) is reacted with hydrazine of the general formula (II) (step b)) to obtain the general formula (Iv) a compound in which Rings B, Y and R 1 As defined in the above general formula (I), ring A is a cycloalkyl group or a 4- to 7-membered saturated heterocyclic ring containing 1 N, wherein ring A is connected to Y via a ring nitrogen.

Preferred implementations are, for example, the following:
Step a) i) Lawesson reagent; pyridine; reflux temperature; 2 to 10 hours; or
ii) Lawson's reagent; tetrahydrofuran; room temperature; 6 to 20 hours;
Step b) i) the reaction of formula (II) and formula (III-c): xylene; 140 ° C; 20 to 120 hours; or
ii) the reaction of formula (II) and formula (III-c): n-butanol; 110 ° C; 20 to 50 hours; or
iii) Reaction of formula (II) with formula (III-c): 1,4-dioxane; 110 ° C; 20 to 50 hours.
A benzoazine-thione derivative of the general formula (III-d) is prepared from a monofluoro derivative of the general formula (XVII), where R 1 Is as defined in the above general formula (I) (FIG. 14; step a)) and the benzoazepine-thione derivative of the general formula (III-d) is reacted with the hydrazine of the general formula (II) ( Step b)) to obtain a compound of general formula (Iw), wherein rings B, Y and R 1 As defined in the above general formula (I), ring A is a cycloalkyl group or a 4- to 7-membered saturated heterocyclic ring containing 1 N, wherein ring A is connected to Y via a ring nitrogen.

Preferred implementations are, for example, the following:
Step a) i) Lawson's reagent; pyridine; reflux temperature; 2 to 10 hours; or
ii) Lawson's reagent; tetrahydrofuran; room temperature; 6 to 20 hours;
Step b) i) the reaction of formula (II) and formula (III-d): xylene; 140 ° C; 20 to 120 hours; or
ii) the reaction of formula (II) and formula (III-d): n-butanol; 110 ° C; 20 to 50 hours; or
iii) Reaction of formula (II) with formula (III-d): 1,4-dioxane; 110 ° C; 20 to 50 hours.
A benzoazine-thione derivative of general formula (III-e) is prepared from a protected hydroxy compound of general formula (XIX) (Figure 15; step a)), where R 1 As defined by the general formula (I) above, PG 1 Department of protection (Peter GM Wuts: Greene's Protective Groups in Organic Synthesis: Fifth Edition, Chapter 2. Protection for the Hydroxyl Group, Including 1,2- and 1,3-Diols, pages 17-471), preferably silicon-based A protecting group, and reacting the benzoazine-thione derivative of the general formula (III-e) with the hydrazine of the general formula (II) (step b)) to obtain a compound of the general formula (Ix), wherein the ring B , Y and R 1 As defined in the above general formula (I), ring A is a cycloalkyl group or a 4- to 7-membered saturated heterocyclic ring containing 1 N, wherein ring A is connected to Y via a ring nitrogen.

Through this synthetic route, a hydroxyl derivative of the general formula (Iu) can also be prepared by removing the protective group of the compound of the general formula (Ix).
Preferred implementations are, for example, the following:
Step a) i) Lawson's reagent; pyridine; reflux temperature; 2 to 10 hours; or
ii) Lawson's reagent; tetrahydrofuran; room temperature; 6 to 20 hours;
Step b) i) the reaction of formula (II) and formula (III-e): xylene; 140 ° C; 20 to 120 hours; or
ii) the reaction of formula (II) and formula (III-e): n-butanol; 110 ° C; 20 to 50 hours; or
iii) reaction of formula (II) and formula (III-e): 1,4-dioxane; 110 ° C; 20 to 50 hours;
Step c) tetrabutylammonium fluoride; tetrahydrofuran; room temperature; 3 to 10 hours.
A benzoazepine-thione derivative of general formula (III-f) is prepared from an alkyl derivative of general formula (XXI), where R 1 Is as defined in the general formula (I) above and alkyl represents C 1-4 Alkyl group (Figure 16; step a)), and the benzoazepine-thione derivative of the general formula (III-f) is reacted with the hydrazine of the general formula (II) (step b)) to obtain the general formula (Iy) a compound in which Rings B, Y, and R 1 As defined in the above general formula (I), ring A is a cycloalkyl group or a 4- to 7-membered saturated heterocyclic ring containing 1 N, wherein ring A is connected to Y via a ring nitrogen.

Preferred implementations are, for example, the following:
Step a) i) Lawson's reagent; pyridine; reflux temperature; 2 to 10 hours; or
ii) Lawson's reagent; tetrahydrofuran; room temperature; 6 to 20 hours;
Step b) i) the reaction of formula (II) and formula (III-f): xylene; 140 ° C; 20 to 120 hours; or
ii) the reaction of formula (II) and formula (III-f): n-butanol; 110 ° C; 20 to 50 hours; or
iii) Reaction of formula (II) with formula (III-f): 1,4-dioxane; 110 ° C; 20 to 50 hours.
As long as in the compound of general formula (I), R 2 Department C 1-4 Alkyl, R 3 C (O) R 5 , R 5 Department C 1-4 Alkoxy and ring A is a cycloalkyl or a 4- to 7-membered saturated heterocyclic ring containing 1 N, in which ring A is connected to Y via a ring nitrogen, by combining a compound of general formula (II) with general formula (XXVII) Compounds are reacted to prepare compounds of general formula (I) of the present invention, wherein the compounds of general formula (II) are
,
Wherein ring B and Y are as defined in the general formula (I) above, ring A is a cycloalkyl group or a 4- to 7-membered saturated heterocyclic ring containing 1 N, wherein ring A is connected to Y via a ring nitrogen, and the general Compound of formula (XXVII)
,
Where R 1 Is defined by the general formula (I) above, R 2 Department C 1-4 Alkyl, R 3 C (O) R 5 And R 5 Department C 1-4 Alkoxy.
The reaction is shown in detail in Figure 17:

A preferred embodiment of step a) in FIG. 17 is, for example, the following:
i) xylene; 140 ° C; 20 to 120 hours; or
ii) n-butanol; 110 ° C; 20 to 50 hours; or
iii) 1,4 dioxane; 110 ° C; 20 to 50 hours.
The main intermediates for the synthesis of benzoazine-thione derivatives of general formula (XXVII) can be prepared by one of the following synthesis:

Method A (Figure 18):
Protecting the keto group of the compound of general formula (VIII) by a dioxane ring (step a)) and alkylating the resulting compound of general formula (XXVIII) (step b)) to obtain a compound of general formula (XXIX), Where R 1 Is as defined by the general formula (I) above and R 2 Department C 1-4 alkyl. The compound of the general formula (XXIX) is deprotected (step c)) and an oxime compound of the general formula (XXXI) is prepared from the resulting derivative of the general formula (XXX) (step d)). This oxime of the general formula (XXXI) is converted to a benzoazepine compound of the general formula (XXXII) by a Beckman rearrangement (step e)).

Preferred implementations are, for example, the following:
Step a) ethylene glycol; toluene; p-toluenesulfonic acid; reflux temperature; 2 to 12 hours;
Step b) lithium diisopropylamide; alkyl iodide; tetrahydrofuran; (-78) ° C; 0.5 to 2 hours;
Step c) i) methanol; concentrated hydrochloric acid; 70 ° C; 2 to 6 hours; or
ii) aqueous acetic acid; 110 ° C; 5 to 20 hours;
Step d) methanol; sodium acetate; hydroxylamine hydrochloride; reflux temperature; 3 to 6 hours;
Step e) Polyphosphoric acid; 100 to 120 ° C; 15 to 60 minutes.

Method B (Figure 19):
The protected benzoazine derivative (XXII) is alkylated (step a)), where R 1 Is as defined in the general formula (I) above and PG 1 Is a protective group (Peter GM Wuts: Greene's Protective Groups in Organic Synthesis: Fifth Edition, Chapter 7. Protection for the Amino Group, pages 895-1193), preferably 4-methoxybenzyl, and then the general formula ( The compound of XXXIII) is deprotected (step b)) to obtain a compound of general formula (XXXII).

Preferred implementations are, for example, the following:
Step a) i) sodium hydride; N, N-dimethylformamide; alkyl iodide; 0 to 100 ° C; 1 to 12 hours; or
ii) lithium diisopropylamide; alkyl iodide; tetrahydrofuran; (-78) ° C; 0.5 to 2 hours;
Step b) i) cerium ammonium nitrate; water; acetonitrile; 0 to 25 ° C; 6 to 18 hours; or
ii) trifluoroacetic acid; dichloromethane; room temperature; 12 to 24 hours; or
iii) trifluoromethanesulfonic acid; dichloromethane; room temperature; 2 to 12 hours.

Method C (Figure 20):
A compound of formula (XXXV) is prepared from a compound of formula (XXXIV) (step a)), where R 1 Is as defined by the general formula (I) above and R 2 Department C 1-4 An alkyl group, and subjecting the resulting compound of general formula (XXXV) to hydrolysis (step b)) to obtain a compound of general formula (XXXVI). The compound of the general formula (XXXVI) is ring-closed (step c)) and the compound of the general formula (XXXVII) is esterified (step d)). The generated compound of the general formula (XXX) is converted into a compound of the general formula (XXXII) by the steps of FIG. 18.

Preferred implementations are, for example, the following:
Step a) lithium diisopropylamide; tetrahydrofuran; tert-butyl bromoacetate; 0 to 20 ° C; 3 to 6 hours;
Step b) methanol; sodium hydroxide; room temperature; 3 to 6 hours;
Step c) sulfuric acid; room temperature; 0.5 to 1.5 hours;
Step d) sulfenyl chloride; methanol; 60 ° C; 6 to 8 hours.
Amine derivatives of general formula (XXXIV) are commercially available or can be prepared according to the methods described in the examples.
A benzoazine-thione derivative of the general formula (XXVII-a) is prepared from a main intermediate of the general formula (XXXII) (FIG. 21; step a)) and the benzoazine of the general formula (XXVII-a) is made The hydrazone-thione derivative is reacted with hydrazine of the general formula (II) to obtain a compound of the general formula (Iz) (step b)), wherein the rings B, Y and R 1 As defined in the above general formula (I), ring A is a cycloalkyl group or a 4- to 7-membered saturated heterocyclic ring containing 1 N, wherein ring A is connected to Y via a ring nitrogen and R 2 Department C 1-4 alkyl. The compound of general formula (Iz) can also be synthesized from the compound of general formula (XL-a) (step d)), and the general formula (XL-a) can be obtained from the compound of general formula (XXVII-a) by a methylation reaction. Compound (step c)).

Preferred implementations are, for example, the following:
Step a) i) Lawson's reagent; pyridine; reflux temperature; 2 to 10 hours; or
ii) Lawson's reagent; tetrahydrofuran; room temperature; 6 to 20 hours;
Step b) i) the reaction of formula (II) and formula (XXVII-a): xylene; 140 ° C; 20 to 120 hours; or
ii) the reaction of formula (II) and formula (XXVII-a): n-butanol; 110 ° C; 20 to 50 hours; or
iii) reaction of formula (II) with formula (XXVII-a): 1,4-dioxane; 110 ° C; 20 to 50 hours;
Step c) methyl iodide; potassium carbonate; acetone; room temperature; 4 to 24 hours;
Step d) i) the reaction of formula (II) and formula (XXVII-a): xylene; in the presence of catalytic hydrochloric acid; 140 ° C; 4 to 20 hours; or
ii) Reaction of formula (II) with formula (XXVII-a): 1,4-dioxane; in the presence of catalytic hydrochloric acid; 110 ° C; 4 to 20 hours.
Similar to compounds of general formula (Ib) or (Ii), as desired, by the introduction of new substituents and / or modification or removal of existing substituents and / or formation of salts and / or self-salts by conventional methods The base and / or the racemic mixture is released to prepare a mirror image isomer. For example, any method shown in FIG. 8 to FIG. 10 can also convert a compound of the general formula (Iz) into another compound of the general formula (I).

Figure 10
As long as the compound of the general formula (I), R 2 System hydrogen, R 3 C (O) NH 2 And ring A contains 1 or 2 4 to 7 membered saturated heterocyclic rings of N, wherein ring A is connected to the 5,6-dihydro-4H- [1,2,4] triazolo [4, 3-a] [1] The benzoazine nucleus is connected with a triazole ring, and a compound of the general formula (I) is prepared by reacting a compound of the general formula (XXXVIII) with a compound of the general formula (XXXIX), wherein the general Compound of formula (XXXVIII)
,
Wherein ring B and Y are as defined in the above general formula (I), ring A contains 1 to 2 4 to 7 membered saturated heterocyclic rings of N, and ring A is connected to the 5,6-dihydro through the ring nitrogen- 4H- [1,2,4] triazolo [4,3-a] [1] benzotriazine core triazole ring is connected, and the compound of general formula (XXXIX) is
,
Where R 1 Is defined by the general formula (I) above, R 2 Hydrogen and R 3 C (O) NH 2 .
The reaction is shown in detail in Figure 22:

A preferred embodiment of step a) in FIG. 22 is, for example, the following:
i) melt (solvent-free); 120 to 150 ° C; 3 to 72 hours; or
ii) Cyclopyridine; 140 to 180 ° C; 3 to 72 hours.
Amine derivatives of general formula (XXXVIII) are commercially available or can be prepared according to the methods described in the examples.
By the method of FIG. 23, a triazolo-benzoazine derivative of the general formula (XXXIX) can be prepared, in which R 1 It is as defined in the above general formula (I).

Methylating the compound of the general formula (III-a) (step a)) and reacting the resulting compound of the general formula (XL-b) with formamidine (step b)) to obtain a compound of the general formula (XLI), Where R 1 It is as defined in the above general formula (I). The compound of general formula (XLI) is brominated (step c)) to obtain a bromine derivative of general formula (XLII), where R 1 It is as defined in the above general formula (I). A compound of the general formula (XLIII) is prepared by hydrolysis of the bromine derivative of the general formula (XLII) produced (step d)) and then amidamine of the general formula (XXXIX) is formed in step e). A nitrile derivative of the general formula (LII) can also be synthesized from the amidine of the general formula (XXXIX) (step f)) and the nitrile derivative of the general formula (LII) can be reacted with a compound of the following general formula (XXXVIII) to Formation of compounds of general formula (I-ac):
,
Wherein ring B and Y are as defined in the above general formula (I), ring A contains 1 to 2 4 to 7 membered saturated heterocyclic rings of N, and ring A is connected to the 5,6-dihydro through the ring nitrogen- The 4H- [1,2,4] triazolo [4,3-a] [1] benzoazine nucleus is attached to the triazole ring.
Preferred implementations are, for example, the following:
Step a) methyl iodide; potassium carbonate; acetone; room temperature; 4 to 24 hours;
Step b) Formamylhydrazine; 1,4-dioxane; 90 ° C; 3 to 10 hours;
Step c) N-bromosuccinimide; tetrahydrofuran; 70 ° C; 10 to 60 minutes;
Step d) methanol; sodium hydroxide; room temperature; 1 to 20 hours;
Step e) Ammonium chloride; N- (3-dimethylaminopropyl) -N'-ethylcarbodiimide hydrochloride; N, N-dimethylformamide; N, N-di Isopropylethylamine or triethylamine; 1-hydroxybenzotriazole hydrate; room temperature; 4 to 20 hours;
Step f) trifluoroacetic anhydride; dichloromethane; triethylamine; 0 ° C to room temperature; 1 to 5 hours;
Step g) i) melt (solvent-free); 120 to 150 ° C; 3 to 72 hours; or
ii) Cyclopyridine; 140 to 180 ° C; 3 to 72 hours.
As desired, mirror image isomers are prepared by conventional methods by introducing new substituents and / or modifying or removing existing substituents and / or forming a salt and / or liberating a base from a salt and / or a racemic mixture. For example, using steps c) and d) shown in FIG. 8, the compound of general formula (I-aa) prepared according to FIG. 22 can also be converted into another compound of general formula (I).
By the method described in detail in FIG. 24, the compound of the general formula (I) can be prepared, in which R 1 Is defined by the general formula (I) above, R 2 Hydrogen or C 1-4 Alkyl, R 3 Department C 1-4 Alkyl, or R 2 And R 3 Combined Representative-(CH 2 ) p -O- (CH 2 ) q -Or- (CH 2 ) r -, Where p is 1, 2, or 3; q is 1, 2, or 3; and r is 4, 5, or 6.

By a Grignard reaction (step a)), a diethyl malonate derivative of the general formula (XLIV) (where R 2 Hydrogen or C 1-4 Alkyl, R 3 Department C 1-4 Alkyl, or R 2 And R 3 Combined Representative-(CH 2 ) p -O- (CH 2 ) q -Or- (CH 2 ) r -, Wherein p is 1, 2, or 3; q is 1, 2, or 3; r is 4, 5, or 6) is converted to a benzyl derivative of the general formula (XLV), where R 1 Represents hydrogen or fluorine. Under basic conditions, a free acid of the general formula (XLVI) is prepared by hydrolyzing a compound of the general formula (XLV) (step b)). Decarboxylation of the compound of the general formula (XLVI) (step c)) generates a compound of the general formula (XLVII), and the compound of the general formula (XLVII) is treated with concentrated sulfuric acid to prepare a compound of the general formula (XLVIII) (step d)). An oxime derivative of the general formula (XLIX) is then prepared from the compound of the general formula (XLVIII) (step e)), and the oxime derivative of the general formula (XLIX) is converted to Benzacridine compounds of general formula (L). If R 3 Based on hydrogen, the compound of general formula (L) is chlorinated or brominated (step g)) to obtain the compound of general formula (LI). Convert a compound of general formula (LI) or (L) into a compound of general formula (I-ad) (step h)), where R 1 Is defined by the general formula (I) above, R 2 Hydrogen or C 1-4 Alkyl, R 3 Department C 1-4 Alkyl, or R 2 And R 3 Combined Representative-(CH 2 ) p -O- (CH 2 ) q -Or- (CH 2 ) r -, Wherein p is 1, 2, or 3; q is 1, 2, or 3; r is 4, 5, or 6, ring B and Y are as defined in the above general formula (I) and ring A is cycloalkyl or contains A 4- to 7-membered saturated heterocyclic ring of N, wherein ring A is connected to Y via a ring nitrogen.
A preferred embodiment of the synthesis of FIG. 24 is, for example, the following:
Step a) benzyl magnesium chloride or 3-fluorobenzyl magnesium chloride; diethyl ether; room temperature; 2 to 12 hours;
Step b) 2M aqueous sodium hydroxide solution; ethanol; reflux temperature; 2 hours;
Step c) heating at 180 to 200 ° C for 3 to 5 minutes;
Step d) sulfuric acid; 35 to 40 ° C; 60 minutes;
Step e) methanol; sodium acetate; hydroxylamine hydrochloride; reflux temperature; 2 to 6 hours;
Step f) polyphosphoric acid; 100 to 120 ° C; 15 to 60 minutes;
Step g) bromine; acetic acid / water; room temperature; 30 to 90 minutes; or, N-chlorosuccinimide; DMF; room temperature; 30 to 90 minutes;
Step h) i) Lawson's reagent; pyridine; reflux temperature; 2 to 10 hours;
ii) methyl iodide; potassium carbonate; acetone; 40 ° C; 1 to 3 hours;
iii) hydrazine of general formula (II); concentrated hydrochloric acid (a few drops); 1,4-dioxane; reflux; 1 to 8 hours;
or
i) trimethyloxonium tetrafluoroborate; trifluoroacetic acid or potassium carbonate; dichloromethane; room temperature; 2 to 4 hours;
ii) hydrazine of general formula (II); trifluoroacetic acid (several drops); 1,4-dioxane or acetonitrile; reflux; 1 to 8 hours.
By different methods described in FIG. 2, hydrazine of general formula (II) can be synthesized, in which ring B and Y are as defined in general formula (I) above and ring A is cycloalkyl or 4 to 4 containing 1 N. A 7-membered saturated heterocyclic ring in which ring A is connected to Y via a ring nitrogen.
According to the method shown in FIG. 25, a compound of the general formula (Im) can also be synthesized.

Let the compound of formula (X) (where R 1 (As defined in the above general formula (I)) is reduced (step a)) and the obtained hydroxy compound of general formula (LIII) is converted into a fluorine derivative of general formula (LIV) (step b)). Directly from a benzoazepine-thione of the general formula (LV) (which is derived from a compound of the general formula (LIV) from step c)) (step e)) or a methylthio derivative from the general formula (LVII) (Step h)), a compound of the general formula (Im) can be synthesized. The methylthio derivative of the general formula (LVII) can be prepared from the benzoazepine-thione of the general formula (LV) by methylation (step f)). An alternative synthetic route is to methylate the compound of general formula (LIV) (step d)) and in situ obtain the methoxy derivative of general formula (LVI) and general formula (II) The compound is reacted (step g)) to produce the compound of the general formula (Im).
A preferred embodiment of the synthesis of FIG. 25 is, for example, the following:
Step a) lithium aluminum hydride; diethyl ether; tetrahydrofuran; (-40) to 0 ° C; 0.1 to 2 hours;
Step b) diethylaminosulfur trifluoride; dichloromethane; (-78) to 10 ° C; 4 to 12 hours;
Step c) i) Lawson's reagent; pyridine; reflux temperature; 2 to 10 hours; or
ii) Lawson's reagent; tetrahydrofuran; room temperature; 6 to 20 hours;
Step d) trimethyloxonium tetrafluoroborate; trifluoroacetic acid or potassium carbonate; dichloromethane; room temperature; 2 to 4 hours;
Step e) i) reacting hydrazine of general formula (II) with general formula (LV); xylene; 140 ° C; 20 to 120 hours; or
ii) reaction of hydrazine of general formula (II) with general formula (LV); n-butanol; 110 ° C; 20 to 50 hours; or
iii) reaction of hydrazine of general formula (II) with general formula (LV); 1,4-dioxane; 110 ° C; 20 to 50 hours;
Step f) methyl iodide; potassium carbonate; acetone; room temperature; 4 to 24 hours;
Step g) hydrazine of general formula (II); dichloromethane or acetonitrile; 50 ° C; 6 to 20 hours;
Step h) The hydrazine of the general formula (II) is reacted with the general formula (LVII); 1,4-dioxane; 110 ° C; 2 to 20 hours.
According to the method shown in FIG. 26, the compound of the general formula (I-ad) of the present invention can be prepared, in which R 1 Is defined by the general formula (I) above, R 2 Hydrogen or C 1-4 Alkyl, R 3 Department C 1-4 Alkyl, or R 2 And R 3 Combined Representative-(CH 2 ) p -O- (CH 2 ) q -Or- (CH 2 ) r -, Wherein p is 1, 2, or 3; q is 1, 2, or 3; r is 4, 5, or 6, and ring A contains 1 or 2 4 to 7-membered saturated heterocyclic rings of N, wherein ring A is The triazole ring of the 5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazine core is connected via a ring nitrogen.

Let the compound of general formula (LI) (R 1 Is defined by the general formula (I) above, R 2 Hydrogen or C 1-4 Alkyl, R 3 Department C 1-4 Alkyl, or R 2 And R 3 Combined Representative-(CH 2 ) p -O- (CH 2 ) q -Or- (CH 2 ) r -, Wherein p is 1, 2, or 3; q is 1, 2, or 3; r is 4, 5, or 6) and a Lawson reactant (step a)) to form a benzoacene of the general formula (LVIII) -A thioketone derivative, and the benzoazine-thione derivative of the general formula (LVIII) is methylated (step b)) and the obtained compound of the general formula (LIX) is reacted with formamidine hydrazine (step c)) to form a compound of general formula (LX). The compound of general formula (LX) is brominated (step d)) to obtain a bromine derivative of general formula (LXI), and the bromine derivative of general formula (LXI) is reacted with a compound of general formula (XXXVIII) e)) to form compounds of general formula (I-ae):
,
Wherein ring B and Y are as defined in the above general formula (I), ring A contains 1 to 2 4 to 7 membered saturated heterocyclic rings of N, and ring A is connected to the 5,6-dihydro through the ring nitrogen- The 4H- [1,2,4] triazolo [4,3-a] [1] benzoazine nucleus is attached to the triazole ring.
A preferred embodiment of the synthesis of FIG. 26 is, for example, the following:
Step a) i) Lawson's reagent; pyridine; reflux temperature; 2 to 10 hours; or
ii) Lawson's reagent; tetrahydrofuran; room temperature; 6 to 20 hours;
Step b) methyl iodide; potassium carbonate; acetone; room temperature; 4 to 24 hours;
Step c) Formamylhydrazine; 1,4-dioxane; 90 ° C; 3 to 10 hours;
Step d) N-bromosuccinimide; tetrahydrofuran; 70 ° C; 10 to 60 minutes;
Step e) i) melt (solvent-free); 120 to 150 ° C; 3 to 72 hours; or
ii) Cyclopyridine; 140 to 180 ° C; 3 to 72 hours.
The reagents and detailed process steps required for the above reactions are described in the examples.
One aspect of the present invention is a novel intermediate represented by the general formula (III-a), (X), (XLII), (LI), and (LIV) synthesized by a process for preparing a compound of general formula (I), wherein R 1 As defined by the general formula (I) above, especially 7-chloro-2- pendantoxy-2,3,4,5-tetrahydro-1 H Methyl-1-benzoazine-4-carboxylic acid (intermediate 3), 7-chloro-2- pendant thio-2,3,4,5-tetrahydro-1 H Methyl-1-benzoazine-4-carboxylic acid (intermediate 4), 1-bromo-8-chloro-5,6-dihydro-4 H -[1,2,4] triazolo [4,3- a ] [1] Methylbenzoazine-5-carboxylic acid (intermediate 24), 7-chloro-4- (fluoromethyl) -1,3,4,5-tetrahydro-2 H -1-benzoazin-2-one (Intermediate 36), or R 2 And R 3 Combined Representative-(CH 2 ) p -O- (CH 2 ) q -Or- (CH 2 ) r -, Where p is 1, 2, or 3; q is 1, 2, or 3; and r is 4, 5, or 6.
The activity data of each compound of the general formula (I) of the present invention was measured in vitro or in vivo by the following method.

Human Vasopressin V1a Receptor Binding Analysis
Cells and radioligands
Immortalized 1321N1 cell line (Perkin Elmer, ES-361-M400-UA) that structurally and stably expresses human vasopressin V1a receptor and vasopressin (8-L-arginine) as a radioligand ) Via [phenylpropylaminomethyl-3,4,5- 3 H (N)]-labeled compounds (Perkin Elmer Life and Analytical Sciences) to determine the affinity of the compounds prepared.

method
Cell membrane preparation: Cell membrane preparation of immortalized 1321N1 cells expressing proliferated human vasopressin V1a receptors according to the Jarvis method (Jarvis et al., J Pharmacol Exp Ther 2004, 310: 407-16). The cells were suspended in a preparative buffer (50 mM Tris, 1 mM EDTA, 0.1 mM PMSF) and homogenized by a glass homogenizer. In order to separate the crude cell membrane fraction, two continuous centrifugation operations (40,000 g, 20 minutes, 4 ° C.) were performed, and then the cell membrane was placed in the preparative buffer in the final washing step. The resulting cell membrane preparation was divided into several portions and stored in Use at -80 ° C until measurement.
According to the Lowry's method, a standard dilution series of bovine serum albumin (BSA) was used to measure the protein content of the prepared cell membrane (Lowry et al., J Biol Chem 1951, 193: 265-75).
Receptor binding assay: In receptor binding analysis, at least 8 different concentrations of each substance with 3 parallel unknown affinities are used. To determine the final affinity value, the results of at least two independent experiments were taken into account. The analysis mixture included incubation buffer (50 mM Tris-HCl, pH 7.4 + 3% BSA), a cell membrane preparation of 1321N1 cells expressing human vasopressin V1a receptor (167 µg / ml), and vascular as a radioligand. (8-L-arginine) [phenylalaninino-3,4,5- 3 H (N)] (1 nM).
At 1.2x10 -6 M unlabeled (Arg 8 )-Non-specific binding values are measured in the presence of vasopressin. A sample with a total volume of 0.33 ml was incubated at 27 ° C for 60 minutes. By passing UniFilter® GF / B impregnated with 0.5% polyethyleneimine TM Filtration to separate cell-bound and free ligands. After drying the filter pan, 40 μl of Microscint-20 (Packard) scintillation liquid was added to the sample. Finally, use MicroBeta 2 Microdisk counter (Perkin Elmer) measures radioactivity.
Use S-shape fitting mathematical method y = (A1-A2) / (1+ (x / x 0 ) p) + A2 and Origin 7.5. Software (OriginLab Corporation, Northampton, USA) concentration shift curve calculation IC 50 Value (ie, the concentration of the unknown substance that replaces 50% of the specifically bound radioligand). When fitting, the asymptotes are not fixed. Use Cheng-Prusoff equation K i = IC 50 / [1+ (L / K D )] Calculate the inhibition constant (K i ) Value, where [L] is the concentration of radioligand used in the experiment and [K D ] Is the affinity of the radiolabeled ligand to the recipient. Pre-determining K using a Scatchard curve D .

Functional analysis of test compounds on cell lines expressing human vasopressin V1a receptor
cell
The immortalized 1321N1 cell line (Perkin Elmer, ES-361-M400-UA) that structurally and stably expresses human vasopressin V1a receptor was used to measure the compounds prepared. General secondary messenger pathway using the GPCR receptor to be tested, endogenous G q Related systems.

method
Compounds were measured using a 96-well trough plate (30,000 cells / plate). The composition of the buffer used for this measurement (expressed in mM) is as follows: 140 NaCl, 5 KCl, 2 CaCl 2 , 2 MgCl 2 , 10 glucose, 10 HEPES (4- (2-hydroxyethyl) -1-piperazine-ethanesulfonic acid), 2 probenecid, pH 7.4. The FLIPR Calcium 5 kit (Molecular Devices) was used as the fluorescent dye, and the matrix was not removed before the dye was filled and the cells were not washed before and after. Incubate at room temperature and the final DMSO concentration is 1%. The substances to be tested are pretreated for 15 to 20 minutes and at least 2 parallel measurements are performed for each concentration of each compound.
Use of fluorescent signals to determine intracellular Ca 2+ Concentration and reading meter is FlexStation II96. Cytoplasmic Ca was measured by fluorometry and using a FlexStation II 96 disk reader (excitation: 485 nm; emission: 525 nm) 2+ concentration. Records fluorescent signal for 1 minute every 1.4 seconds. The reference compounds used are as follows: 80 (Arg 8 )-Vasopressin as an agonist, and rivasuvaptan measured at 1 μM as an antagonist. Determination of% inhibition and IC for each concentration of compound 50 Value, where the concentration line is also determined.
The total AVP concentration-response curve was recorded for each plate. The measured efficacy of a compound is expressed as a relative inhibition% response relative to a control group. For the graph of the data, use SoftMaxPro software and according to the following formula: y = AD / (1+ (x / C) ^ B) + D, where A = 0 and D = 100-fixed asymptote below / above, y =% Inhibition, x = logarithmic concentration of test compound, B = steepness of curve and C = IC 50 (Concentration of 50% inhibition of the response of the control group), a non-linear 4 parameter juxtaposition was performed. For all compounds, calculate the average IC from at least 3 independent measurements 50 value.





Mouse Vasopressin V1a Receptor Binding Analysis
Cells and radioligands
Using the immortalized 1321N1 cell line (B9 / 1321N1 selective strain) which structurally and stably expresses the vasopressin V1a receptor in mice and the vasopressin (8-L-arginine) as a radioligand [Phenylaminopropyl-3,4,5- 3 H (N)]-labeled compounds (Perkin Elmer Life and Analytical Sciences) to determine the affinity of the compounds prepared.

method
Cell membrane preparation: Cell membrane preparation of immortalized 1321N1 cells expressing proliferating mouse vasopressin V1a receptors according to the Jarvis method (Jarvis et al., J Pharmacol Exp Ther 2004, 310: 407-16). The cells were suspended in a preparative buffer (50 mM Tris, 1 mM EDTA, 0.1 mM PMSF) and homogenized by a glass homogenizer. In order to separate the crude cell membrane fraction, two continuous centrifugation operations (40,000 g, 20 minutes, 4 ° C.) were performed, and then the cell membrane was placed in the preparative buffer in the final washing step. The resulting cell membrane preparation was divided into several portions and stored in Use at -80 ° C until measurement.
According to the Lowry's method, a standard dilution series of bovine serum albumin (BSA) was used to measure the protein content of the prepared cell membrane (Lowry et al., J Biol Chem 1951, 193: 265-75).
Receptor binding assay: In receptor binding analysis, at least 8 different concentrations of each substance with 3 parallel unknown affinities are used. To determine the final affinity value, the results of at least two independent experiments were taken into account. The analysis mixture included an incubation buffer (50 mM Tris-HCl, pH 7.4 + 3% BSA), a cell membrane preparation (152 µg / ml) of 1321N1 cells expressing mouse vasopressin V1a receptor, and blood vessels as radioligands Vasopressin (8-L-arginine) [phenylalaninino-3,4,5- 3 H (N)) (about K D (35 to 50% concentration).
At 1.2x10 -6 M unlabeled (Arg 8 )-Non-specific binding values are measured in the presence of vasopressin. A sample with a total volume of 0.33 ml was incubated at 27 ° C for 60 minutes. By passing UniFilter® GF / B impregnated with 0.5% polyethyleneimine TM Filtration to separate cell-bound and free ligands. After drying the filter pan, 40 μl of Microscint-20 (Packard) scintillation liquid was added to the sample. Finally, use MicroBeta 2 Microdisk counter (Perkin Elmer) measures radioactivity.
The radioligand clamping ability of the substance was measured by at least 2 independent experiments. Specific radioligand binding can be defined as the difference between total binding and non-specific binding in the presence of saturated amounts of unlabeled ligand or different concentrations of the test substance. Results are expressed as% inhibition of specific binding achieved in the presence of the analyte.
Use S-shape fitting mathematical method y = (A1-A2) / (1+ (x / x 0 ) p) + A2 and Origin 7.5. Software (OriginLab Corporation, Northampton, USA) Concentration shift curve calculation IC 50 Value (ie, the concentration of the unknown substance that replaces 50% of the specifically bound radioligand). When fitting, the asymptotes are not fixed. Use Cheng-Prusoff equation K i = IC 50 / [1+ (L / K D )] Calculate the inhibition constant (K i ) Value, where [L] is the concentration of radioligand used in the experiment and [K D ] Is the affinity of the radiolabeled ligand to the recipient. Pre-determining K using a Scatchard curve D .


Human Vasopressin V2 Receptor Binding Analysis
Cells and radioligands
Immortalized 1321N1 cell line (Perkin Elmer, ES-363-M400-UA) (lot 1765208), which expresses human vasopressin V2 receptor, and CHO-K1, which express human vasopressin V2 receptor, were used Cell membrane (Perkin Elmer, 6110541400UA) and vasopressin (8-L-arginine) as a radioligand via [phenylalanine-3,4,5- 3 H (N)]-labeled compounds (Perkin Elmer Life and Analytical Sciences) to determine the affinity of the compounds prepared.

method
Receptor binding assay: In receptor binding analysis, at least 8 different concentrations of each substance with 3 parallel unknown affinities are used. To determine the final affinity value, the results of at least two independent experiments were taken into account. The analysis mixture included an incubation buffer (50 mM Tris-HCl, pH 7.4 + 3% BSA), a cell membrane preparation (1.82 µg / ml) of 1321N1 cells expressing human vasopressin V2 receptor, and a vasoliter as radioligand (8-L-arginine) [phenylalaninino-3,4,5- 3 H (N)) (about K D Concentration).
At 1.2x10 -6 M unlabeled (Arg 8 )-Non-specific binding values are measured in the presence of vasopressin. A sample with a total volume of 0.55 ml was incubated at 27 ° C for 90 minutes. By passing UniFilter® GF / B impregnated with 0.5% polyethyleneimine TM Filtration to separate cell-bound and free ligands. After drying the filter pan, 40 μl of Microscint-20 (Packard) scintillation liquid was added to the sample. Finally, use MicroBeta 2 Microdisk counter (Perkin Elmer) measures radioactivity.
The radioligand substitution capacity of a substance is measured by at least 2 independent experiments. Specific radioligand binding can be defined as the difference between total binding and non-specific binding in the presence of saturated amounts of unlabeled ligand or different concentrations of the test substance. Results are expressed as% inhibition of specific binding achieved in the presence of the analyte.
Use S-shape fitting mathematical method y = (A1-A2) / (1+ (x / x 0 ) p) + A2 and Origin 7.5. Software (OriginLab Corporation, Northampton, USA) calculates IC from concentration shift curve 50 Value (ie, the concentration of the unknown substance that replaces 50% of the specifically bound radioligand). When fitting, the asymptotes are not fixed. Use Cheng-Prusoff equation K i = IC 50 / [1+ (L / K D )] Calculate the inhibition constant (K i ) Value, where [L] is the concentration of radioligand used in the experiment and [K D ] Is the affinity of the radiolabeled ligand to the recipient. Pre-determining K using a Scatchard curve D .

method
Use at least 2 incubation buffers (50 mM Tris-HCl, 5 mM MgCl 2 , PH 7.4 + 0.1% BSA), CHO-K1 cell membrane preparation (7 µg / µl) (Perkin Elmer, 6110541400UA) showing human vasopressin V2 receptor and vasopressin (8-L) as a radioligand -Arginine) (phenylalaninino-3,4,5- 3 H (N)) (about K D Concentration)), independent receptor experiments were performed using at least 8 concentrations and 2 or 3 parallel samples each.
At 1.2x10 -6 M unlabeled (Arg 8 )-Non-specific binding values can be measured in the presence of vasopressin. A sample with a total volume of 0.55 ml was incubated at 27 ° C for 90 minutes. By passing through UniFilter® GF / B impregnated with polyethyleneimine TM Filtration to separate cell-bound and free ligands. Rinse the filter plate 3 times with 0.5 ml ice-cold wash buffer (50 mM Tris-HCl, pH 7.4). After drying the filter disc, 40 μl of Microscint-20 (Packard) scintillation liquid was added to each well. Finally, the radioactivity was measured using a Tri-Carb 2900TR liquid scintillation analyzer (Perkin Elmer).
The radioligand substitution capacity of a substance is measured by at least 2 independent experiments. Specific radioligand binding can be defined as the difference between total binding and non-specific binding in the presence of saturated amounts of unlabeled ligand or different concentrations of the test substance. Results are expressed as% inhibition of specific binding achieved in the presence of the analyte.
Use S-shape fitting mathematical method y = (A1-A2) / (1+ (x / x 0 ) p) + A2 and Origin 7.5. Software (OriginLab Corporation, Northampton, USA) calculates IC from concentration shift curve 50 Value (ie, the concentration of the unknown substance that replaces 50% of the specifically bound radioligand). When fitting, the asymptotes are not fixed. Use Cheng-Prusoff equation K i = IC 50 / [1+ (L / K D )] Calculate the inhibition constant (K i ) Value, where [L] is the concentration of radioligand used in the experiment and [K D ] Is the affinity of the radiolabeled ligand to the recipient. Pre-determining K using a Scatchard curve D .
Affinity data measured in this 1321N1 cell line expressing human vasopressin V2 receptor i K) and K obtained from CHO-K1 cell line expressing human vasopressin V2 receptor i The results are extremely closely related.

Functional V1a in vivo test
animal
Male mice weighing 18 to 40 g (NMRI, ToxiCoop) were used. Animals have been kept for at least 5 days since delivery, and animals are allowed to eat and drink freely during captivity and measurement. The experiments were approved by the Regional Animal Protection Committee and in accordance with European Animal Protection Guidelines ( EU Directive 2010/63 / EU ) Implementation.

method
With automated behavior analysis system (LABORAS TM ) Measure animal behavior. Sensors located under the plate detect mechanical vibrations generated by animal movements and convert them into electronic signals (Quinn et al., J Neurosci Methods 2003, 130: 83-92). After analyzing the signal, the system analyzes the time spent on the following behavioral parameters: movement, standing still, climbing and grooming. The defined grooming algorithm measures the response to scratching behavior. During the experimental period, the mice were pretreated with the test substance or carrier, and after this pretreatment period, the compounds causing the scratching (0.3 mg / kg oxytocin subcutaneously) were administered, and then the animals were individually placed in the measurement cage. The behavior of these mice was observed for 1 hour. To reduce exploration activities, the animals were measured after 1 hour in the cage. These behavioral parameters were compared to those measured in parallel in control animals.
The behavior inhibitory effect of the test substance was calculated from the mean value of the carrier treatment group measured in parallel and expressed as% inhibition, where 0% represents the scratching behavior of the carrier-pretreated animals (and the carrier-pretreated subcutaneous saline). The average value, and 100% represents the average scratching value of the vehicle pretreated with subcutaneously received oxytocin. For statistical analysis, a single factor analysis of variance (ANOVA) using Tukey's post hoc comparison test was used.
Surprisingly, in vivo functional tests of certain compounds of the present invention have been found to produce significant effects on mouse Vla receptors.

The present invention will be further explained by the following embodiments, but the scope of the present invention is not limited to these embodiments. With the above description and the following examples, those skilled in the art can determine the essential features of the invention, and without departing from the spirit and scope of the invention, certain changes and modifications can be made to make the invention suitable for many different Applications and conditions. Therefore, the present invention is not limited to the following illustrative embodiments, but is limited to the scope determined by the scope of the attached patent application.
In general, the compound of general formula (I) can be prepared according to the general knowledge of those skilled in the art and / or methods of preparing the compounds of the examples and / or intermediates. Those skilled in the art can easily select solvents, temperatures, pressures, and other reaction conditions. The starting materials are commercially available and / or can be easily prepared by those skilled in the art. In preparing the compounds, for example, combinatorial techniques can be used, and the intermediate groups present therein are suitable for use in these methods.
In the synthesis, the following terms and abbreviations are used:
dry = No water
Boc = tertiary butoxycarbonyl
Dess-Martin periodinane = 1,1,1-tris (ethoxyl) -1,1-dihydro-1,2-benzoiodoxazole-3- (1H) -ketone
DIPEA = N, N-diisopropyl-ethylamine
DMAP = 4-dimethylamino-pyridine
DMF = N, N-dimethylformamide
EDC = N- (3-dimethylaminopropyl) -N'-ethylcarbodiimide hydrochloride
HOBt = 1-hydroxybenzotriazole hydrate
HBTU = N, N, N ', N'-tetramethyl-O- (1H-benzotriazol-1-yl) urenium hexafluorophosphate
K 2 CO 3 = Potassium carbonate
Lawesson reagent = 2,4-bis (4-methoxyphenyl) -1,3,2,4-dithiadiphosphacyclobutane-2,4-disulfide
Meldrum's acid = 2,2-dimethyl-1,3-dioxane-4,6-dione
MgSO 4 = Magnesium sulfate
NaBH 4 = Sodium borohydride
NaBH (OAc) 3 = Sodium triethylsulfoxyborohydride
NaCl = Sodium chloride
Na 2 CO 3 = Sodium carbonate
NaHCO 3 = Sodium bicarbonate
NaOAc = Sodium acetate
NaOH = Sodium hydroxide
Na 2 SO 4 = Sodium sulfate
Pd / C = palladium on carbon
Pt / C = platinum on carbon
THF = tetrahydrofuran

Intermediate 1
7-Chloro-4-lanthoxy-1,2,3,4-tetrahydronaphthalene-2-carboxylic acid methyl ester

Let 7-chloro-4- pendantoxy-1,2,3,4-tetrahydronaphthalene-2-carboxylic acid (6.0 g, 26.7 mmol) (RD Haworth et al., J Chem Soc 1943, 10) was dissolved in methanol (100 ml) and concentrated sulfuric acid (0.5 ml) was added and the mixture was stirred overnight at reflux temperature and concentrated. The residue was dissolved in a mixture of diethyl ether and ethyl acetate (1: 1) and subjected to 10% Na 2 CO 3 Rinse with solution and water. The aqueous phase was extracted with ethyl acetate. The combined organic phases were washed with a saturated NaCl solution, dried over anhydrous sodium sulfate, filtered and concentrated. Thus, the target product (6.27 g, 98.4%) was obtained as beige crystals. MS (ESI) m / z 239.1 (M + H) + .

Intermediate 2
7-chloro-4- (hydroxyimino) -1,2,3,4-tetrahydronaphthalene-2-carboxylic acid methyl ester

Dissolve methyl 7-chloro-4-oxo-1,2,3,4-tetrahydronaphthalene-2-carboxylic acid (Intermediate 1) (6.27 g, 26.3 mmol) in methanol (220 ml), And then anhydrous NaOAc (6.57 g, 80 mmol) and hydroxylamine hydrochloride (5.55 g, 80 mmol) were added. The mixture was refluxed for 3 hours and then stirred overnight at room temperature. The reaction mixture was concentrated, water was added, and the mixture was extracted with ethyl acetate. The combined organic phases were washed with a saturated NaCl solution, dried over anhydrous sodium sulfate, and filtered and concentrated. Thus, the target product was obtained as slightly yellow crystals (6.63 g, 99.4%). MS (ESI) m / z 254.1 (M + H) + .

Intermediate 3
7-chloro-2- pendantoxy-2,3,4,5-tetrahydro-1 H Methyl-1-benzoazine-4-carboxylic acid

Add 7-chloro-4- (hydroxyimino) -1,2,3,4-tetrahydronaphthalene-2-carboxylic acid methyl ester (Intermediate 2) (6.57 g, 25.9 mmol) to the polyphosphoric acid (60.0 g) and stirred at 110 ° C for 20 minutes. Ice was added to the reaction mixture and stirred for 10 minutes. It was then extracted with ethyl acetate, and the combined organic phases were washed with a saturated NaCl solution, dried over anhydrous sodium sulfate, and filtered and concentrated. The residue was purified by column chromatography (using cyclohexane: ethyl acetate = 2: 1 as eluent). Thus, the target product (3.03 g, 46.2%) was obtained as white crystals. MS (ESI) m / z 254.1 (M + H) + .

Intermediate 4
7-chloro-2- pendant thio-2,3,4,5-tetrahydro-1 H Methyl-1-benzoazine-4-carboxylic acid

Let 7-chloro-2- pendantoxy-2,3,4,5-tetrahydro-1 H Methyl-1-benzoazine-4-carboxylic acid (Intermediate 3) (1.50 g, 5.9 mmol) was dissolved in pyridine (80 ml), Lawson's reagent (3.11 g, 7.7 mmol) was added and The reaction mixture was allowed to stir at reflux temperature for 2 hours and then concentrated. Water (15 ml) and saturated NaHCO 3 The solution (60 ml) was added to the residue and the highly precipitated suspension was stirred for 30 minutes. The precipitated yellow product was filtered off and dried under vacuum at 50 ° C over phosphorus pentoxide. Thus, the target product (1.58 g, 99.9%) was obtained as yellow crystals. MS (ESI) m / z 270.1 (M + H) + .

Intermediate 5
2-{[trans-4- (trifluoromethyl) cyclohexyl] carbonyl} tert-butyl hydrazine formate

Trans-4- (trifluoromethyl) cyclohexanecarboxylic acid (Manchester Organics Ltd) (2.13 g, 10.9 mmol) was dissolved in DMF (50 ml). To this solution was added tert-butyl hydrazineformate (1.44 g, 10.9 mmol), DIPEA (4.75 ml, 27.3 mmol), HOBt (2.00 g, 13.10 mmol), and EDC (2.51 g, 13.1 mmol) ear). The reaction mixture was allowed to stir at room temperature for 36 hours and then concentrated. Saturated NaHCO 3 The solution (40 ml) was added to the residue and the precipitate was filtered after brief stirring, rinsed with water and dried on phosphorus pentoxide in a vacuum oven. Thus, the target product was obtained as a white powder (3.35 g, 99%). GC-MS (EI) m / z 310.1.

Intermediate 6
Trans-4- (trifluoromethyl) cyclohexanemethanehydrazine

Soluble tert-butyl 2-{[trans-4- (trifluoromethyl) cyclohexyl] carbonyl} hydrazine formate (Intermediate 5) (3.35 g, 10.79 mmol) in ethyl acetate (50 ml) And ethanol (20 ml) and then 2.5 M hydrochloric acid in ethyl acetate (30 ml) was added to the solution. The reaction mixture was allowed to stir at room temperature for 16 hours, then diethyl ether (150 ml) was added and cooled in an ice-water bath. The precipitated product was filtered and washed with diethyl ether. Pass the filter through saturated NaHCO 3 The solution (pH about 8) (100 ml) was stirred, filtered, washed with water and dried in a vacuum oven over phosphorus pentoxide. Thus, the target product (1.77 g, 78%) was obtained as a white powder. GC-MS (EI) m / z 210.1.

Intermediate 7
Methyl trans-4- (piperidin-1-ylmethyl) cyclohexanecarboxylate

Dissolve methyl trans-4-formylcyclohexanecarboxylate (Synthonix) (0.30 g, 1.8 mmol) in 1,2-dichloroethane (10 ml) and add piperidine (0.52) to the solution. ml, 5.3 mmoles) and acetic acid (0.19 ml, 3.4 mmoles). The resulting mixture was cooled to 0 ° C and NaBH (OAc) was added. 3 (1.16 g, 5.5 mmol) and the reaction mixture was stirred at room temperature for 16 hours. The reaction mixture was then added with water (30 ml) and Na 2 CO 3 The solution adjusted the pH of the mixture to about 9. The mixture was extracted twice with dichloromethane (20 ml), and the combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated. Thus, the target product (0.40 g, 95%) was obtained, which was used without further purification.

Intermediate 8
Trans-4- (piperidin-1-ylmethyl) cyclohexanemethanehydrazine

Dissolve methyl trans-4- (piperidin-1-ylmethyl) cyclohexanecarboxylate (Intermediate 7) (0.40 g, 1.68 mmol) in methanol (5 ml) and pour the solution under pressure Glass reactor. Hydrazine hydrate (5 ml, 100 mmol) was added and the reaction mixture was stirred at 75 ° C for 16 hours. The reaction mixture was concentrated and cyclohexane and anhydrous toluene were evaporated to give a residue. Thus, the target product (0.39 g, 97%) was obtained as a white powder. GC-MS (EI) m / z 239.2.

Intermediate 9
Trans-4- (pyrrolidin-1-ylcarbonyl) cyclohexanemethanehydrazine

a) Methyl trans-4- (pyrrolidin-1-ylcarbonyl) cyclohexanecarboxylate

Let trans-4- (methoxycarbonyl) cyclohexanecarboxylic acid (Combi-Blocks Inc.) (186 mg, 1 mmol), pyrrolidine (83.5 µl, 1 mmol), anhydrous DMF (5 ml ), A mixture of DIPEA (348 µl, 2 mmol), EDC (230 mg, 1.2 mmol) and HOBt (162 mg, 1.2 mmol) at room temperature with stirring for 24 hours. Add ethyl acetate and NaHCO to the reaction mixture 3 The aqueous layer was separated and the layers were separated and the aqueous phase was extracted once with ethyl acetate. The combined organic phases were washed with 1N hydrochloric acid and water, dried over anhydrous sodium sulfate, filtered and concentrated. Thus, the target product was obtained (180 mg, 75%). GC-MS (EI) m / z 239.

b) trans-4- (pyrrolidin-1-ylcarbonyl) cyclohexanemethanehydrazine
Put trans-4- (pyrrolidin-1-ylcarbonyl) cyclohexanecarboxylic acid methyl ester (180 mg, 0.75 mmol), methanol (1.1 ml) and hydrazine hydrate (1.1 ml) in a pressure-resistant glass reactor The mixture was stirred at 75 ° C for 24 hours. The reaction mixture was concentrated and cyclohexane was added and then evaporated. Thus, the target product was obtained (183 mg, 76%). GC-MS (EI) m / z 239.

Intermediate 10
Trans-4- (morpholin-1-ylcarbonyl) cyclohexanemethanehydrazine

a) Methyl trans-4- (morpholin-4-ylcarbonyl) cyclohexanecarboxylate

The target product was prepared from trans-4- (methoxycarbonyl) cyclohexanecarboxylic acid (Combi-Blocks Inc.) and morpholine according to the method described in step a) of Intermediate 9. GC-MS (EI) m / z 255.

b) trans-4- (morpholin-1-ylcarbonyl) cyclohexanemethanehydrazine
The target product was prepared from trans-4- (morpholin-4-ylcarbonyl) cyclohexanecarboxylic acid methyl ester according to the method described in step 9) of intermediate 9. GC-MS (EI) m / z 255.

Intermediate 11
Trans-4- (dimethylamino) cyclohexanemethylhydrazine

The target product was prepared from trans-4- (dimethylamino) cyclohexanecarboxylic acid methyl ester (EP 1 582 521 A1 (05.10.2005), TANABE SEIYAKU CO.) According to the method described in Intermediate 8. MS (ESI) m / z 186.3 (M + H) + .

Intermediate 12
Trans-4- (morpholin-4-yl) cyclohexanemethanehydrazine

The target product was prepared according to the method described in Intermediate 8 from methyl trans-4- (morpholin-4-yl) cyclohexanecarboxylate (EP 1 582 521 A1 (05.10.2005) TANABE SEIYAKU CO.). GC-MS (EI) m / z 227.

Intermediate 13
1- (pyrimidin-2-yl) tetrahydroazepine-3-formamidine

The target product was prepared according to the method described in Intermediate 8 from methyl 1- (pyrimidin-2-yl) tetrahydroazepine-3-carboxylic acid (WO 2006/124748 A2 (23.11.2006) LEXICON GENETICS INCORP.). MS (ESI) m / z 194.2 (M + H) + .

Intermediate 14
1- (pyridin-2-yl) tetrahydroazepine-3-formamidine

The target product was prepared according to the method described in Intermediate 8 from methyl 1- (pyridin-2-yl) tetrahydroazepine-3-carboxylic acid (WO 2017/007756 A1 (12.01.2017) RODIN THERAPEUTICS INC.). GC-MS (EI) m / z 192.

Intermediate 15
(Trans) -3-methyl-2-oxo-1-oxo-3-azaspiro [4.5] decane-8-carboxylic acid ethyl ester

with
Intermediate 16
(Cis) -3-methyl-2-oxo-1-oxo-3-azaspiro [4.5] decane-8-carboxylic acid ethyl ester

A 60% sodium hydride oil dispersion (1.8 g, 45.0 mmol) was suspended in anhydrous DMF (60 ml), cooled to 0 to 5 ° C, and then dissolved in DMF (60 ml) (cis. (Formula) -2-Phenoxy-1-oxa-3-azaspiro [4.5] decane-8-carboxylic acid ethyl ester and (trans) -2-Phenoxy-1-oxa-3-nitro Approximately 1: 1 mixture of heterospiro [4.5] decane-8-carboxylic acid ethyl ester (WO 2008/092887 A1 (07.08.2008) GLAXO GROUP LTD.) (6.00 g, 26.4 mmol) so that the temperature of the mixture Maintain between 0 and 5 ° C. The reaction mixture was allowed to stir at this temperature for 20 minutes, and then methyl iodide (2.46 ml, 39.5 mmol) was added dropwise over 20 minutes. The mixture was allowed to stir for an additional hour at 0 to 5 ° C, returned to room temperature and stirred for 3 hours at this temperature. Acetic acid (1.8 ml, 31 mmol) was then added dropwise over 10 minutes, and after stirring for 15 minutes, the reaction mixture was concentrated and n-heptane (90 ml) was evaporated twice to obtain a residue. To the residue was added ethyl acetate (180 ml), saturated NaHCO 3 The solution (90 ml) and water (90 ml) were separated into layers, and the organic phase was washed with NaCl (90 ml), dried over sodium sulfate, filtered and concentrated. The residue was purified by column chromatography using toluene: isopropanol = 93: 7 as the eluent. The appropriate fractions were concentrated and the residue was crystallized from diisopropyl ether. Thus, (trans) -3-methyl-2-oxo-1-oxo-3-azaspiro [4.5] decane-8-carboxylic acid ethyl ester (Intermediate 15) was obtained as a white powder (Intermediate 15) ( 1.38 g, 22%) and (cis) -3-methyl-2-oxo-1-oxa-3-azaspiro [4.5] decane-8-carboxylic acid ethyl ester (Intermediate 16) ( 2.45 g, 39%). GC-MS (EI) m / z 241.

Intermediate 17
(Trans) -3-methyl-2-oxo-1-oxa-3-azaspiro [4.5] decane-8-formamidine

According to the method described for intermediate 8, ) Preparation of the target product. GC-MS (EI) m / z 227.

Intermediate 18
(Cis) -3-methyl-2-oxo-1-oxa-3-azaspiro [4.5] decane-8-formamidine

According to the method described in Intermediate 8, from (cis) -3-methyl-2-oxo-1-oxa-3-azaspiro [4.5] decane-8-carboxylic acid ethyl ester (Intermediate 16 ) Preparation of the target product. GC-MS (EI) m / z 227.

Intermediate 19
Trans-4- (tetrahydroazepine-1-ylcarbonyl) cyclohexanemethylhydrazine


a) Methyl trans-4- (tetrahydroazepine-1-ylcarbonyl) cyclohexanecarboxylate

The target product was prepared from trans-4- (methoxycarbonyl) cyclohexanecarboxylic acid (Combi-Blocks Inc.) and tetrahydroacridine according to the method described in step a) of Intermediate 9. GC-MS (EI) m / z 225.

b) trans-4- (tetrahydroacryl-1-ylcarbonyl) cyclohexanemethylhydrazine
The title product was prepared from methyl trans-4- (tetrahydroacryl-1-ylcarbonyl) cyclohexanecarboxylate according to the method described in Step 9) of Intermediate 9. GC-MS (EI) m / z 225.

Intermediate 20
Trans-4- (piperidin-1-ylcarbonyl) cyclohexanemethanehydrazine

a) Methyl trans-4- (piperidin-1-ylcarbonyl) cyclohexanecarboxylate

The target product was prepared from trans-4- (methoxycarbonyl) cyclohexanecarboxylic acid (Combi-Blocks Inc.) and piperidine according to the method described in step a) of Intermediate 9. GC-MS (EI) m / z 253.

b) trans-4- (piperidin-1-ylcarbonyl) cyclohexanemethylhydrazine
The target product was prepared from trans-4- (piperidin-1-ylcarbonyl) cyclohexanecarboxylic acid methyl ester according to the method described in Intermediate 9 step b). GC-MS (EI) m / z 253.

Intermediate 21
4-{[trans-4- (hydrazinocarbonyl) cyclohexyl] carbonyl} piperazine-1-carboxylic acid tert-butyl ester

a) 4-{[trans-4- (methoxycarbonyl) cyclohexyl] carbonyl} piperazine-1-carboxylic acid tert-butyl ester

Prepare the target product from trans-4- (methoxycarbonyl) cyclohexanecarboxylic acid (Combi-Blocks Inc.) and piperazine-1-carboxylic acid tert-butyl ester according to the method described in step a) of Intermediate 9, It was used without further purification.

b) trans-4- (morpholin-1-ylcarbonyl) cyclohexanemethanehydrazine
The target product was prepared from 4-{[trans-4- (methoxycarbonyl) -cyclohexyl] carbonyl} piperazine-1-carboxylic acid tert-butyl ester according to the method described in step 9) of Intermediate 9. Used with further purification.

Intermediate 22
7-chloro-2- (methylthio) -4,5-dihydro-3H-1-benzoazine-4-carboxylic acid methyl ester

Let 7-chloro-2- pendant thio-2,3,4,5-tetrahydro-1 H Methyl-1-benzoazine-4-carboxylic acid (Intermediate 4) (250 mg, 0.93 mmol), acetone (18.5 ml), K 2 CO 3 (259 mg, 1.87 mmol) and iodomethane (0.18 ml, 2.89 mmol) were stirred at room temperature for 20 hours. The reaction mixture was concentrated, water was added to the residue and extracted with ethyl acetate, and the organic phase was dried over anhydrous sodium sulfate and filtered and concentrated. Thus, the target product was obtained (261 mg, 99%). MS (ESI) m / z 284.1 (M + H) + .

Intermediate 23
8-chloro-5,6-dihydro-4 H -[1,2,4] triazolo [4,3- a ] [1] Methylbenzoazine-5-carboxylic acid

7-Chloro-2- (methylthio) -4,5-dihydro-3H-1-benzoazine-4-carboxylic acid methyl ester (Intermediate 22) (261 mg, 0.92 mmol) was dissolved in 1,4-dioxane (6 ml) and warm the solution to 90 ° C. Methylhydrazine (277 mg, 4.61 mmol) was added over 4 hours under argon. After the reaction mixture was subsequently stirred at 90 ° C for 8 hours and cooled to room temperature, the solvent was evaporated under vacuum. The residue was purified by column chromatography using dichloromethane: methanol = 95: 5 as the eluent. Thus, the target product was obtained (188 mg, 74%). MS (ESI) m / z 278.1 (M + H) + .

Intermediate 24
1-bromo-8-chloro-5,6-dihydro-4 H -[1,2,4] triazolo [4,3- a ] [1] Methylbenzoazine-5-carboxylic acid

Let 8-chloro-5,6-dihydro-4 H -[1,2,4] triazolo [4,3- a ] [1] Benzoazine-5-carboxylic acid methyl ester (Intermediate 23) (187 mg, 0.67 mmol) was dissolved in THF (15 ml). N-Bromosuccinimide (120 mg, 0.68 mmol) was added and the resulting pale yellow solution was stirred at reflux temperature and irradiated by a RH-500 halogen lamp (Tracon Electric) for 60 minutes. At this time, the color of the solution was initially dark and then gradually faded. After cooling to room temperature, the solvent was evaporated under vacuum. The residue was purified by column chromatography (using cyclohexane: ethyl acetate = 1; 1 to 1: 4 as eluent). Thus, the target compound (226 mg, 94%) was obtained. MS (ESI) m / z 358.0 (M + H) + .

Intermediate 25
7-methyl-2- pendant thio-2,3,4,5-tetrahydro-1 H Methyl-1-benzoazine-4-carboxylic acid

Let 7-methyl-2- pendantoxy-2,3,4,5-tetrahydro-1 H Methyl-1-benzoacryl-4-carboxylic acid (100 mg, 0.43 mmol), which was prepared according to the method described in Intermediate 3, was dissolved in THF (10 ml), followed by the addition of a Lawson reaction (105 mg, 0.26 mmol) and the mixture was stirred at room temperature for 20 hours. The reaction mixture was first concentrated, and the residue was then added with saturated NaHCO 3 The solution was stirred for 60 minutes. The precipitated product was filtered, washed with water and dried. Thus, the target product was obtained (80 mg, 75%). MS (ESI) m / z 250.2 (M + H) + .

Intermediate 26
7-methoxy-2- pendant thio-2,3,4,5-tetrahydro-1 H Methyl-1-benzoazine-4-carboxylic acid

According to the method described in Intermediate 25, from 7-methoxy-2- pendantoxy-2,3,4,5-tetrahydro-1 H Methyl-1-benzoazine-4-carboxylic acid (which was prepared according to the method described in Intermediate 3) was used to prepare the target product. MS (ESI) m / z 266.1 (M + H) + .

Intermediate 27
7-bromo-2- pendant thio-2,3,4,5-tetrahydro-1 H Methyl-1-benzoazine-4-carboxylic acid

According to the method described in Intermediate 25, from 7-bromo-2- pendantoxy-2,3,4,5-tetrahydro-1 H Methyl-1-benzoazine-4-carboxylic acid (which was prepared according to the method described in Intermediate 3) was used to prepare the target product. MS (ESI) m / z 316.0 (M + H) + .

Intermediate 28
7-bromo-2- (methylthio) -4,5-dihydro-3 H Methyl-1-benzoazine-4-carboxylic acid

According to the method described in Intermediate 22, from 7-bromo-2- pendant thio-2,3,4,5-tetrahydro-1 H Methyl-1-benzoazine-4-carboxylic acid (Intermediate 27) was used to prepare the target product. MS (ESI) m / z 330.1 (M + H) + .

Intermediate 29
1-bromo-8-chloro-5,6-dihydro-4 H -[1,2,4] triazolo [4,3- a ] [1] Benzacridin-5-carboxamide

a) 1-Bromo-8-chloro-5,6-dihydro-4 H -[1,2,4] triazolo [4,3- a ] [1] Benzacene-5-carboxylic acid

Let 1-bromo-8-chloro-5,6-dihydro-4 H -[1,2,4] triazolo [4,3- a ] [1] Methyl benzoazine-5-carboxylate (Intermediate 24) (631 mg, 1.77 mmol), THF (13 ml), methanol (6.5 ml), water (6.5 ml) and lithium hydroxide A mixture of hydrates (379 mg, 9.03 mmol) was stirred at room temperature for 2.5 hours, then the solvent was evaporated and the remaining aqueous solution was diluted with water (10 ml). The pH of the solution was adjusted to 5 with 1M hydrochloric acid, and the precipitated product was filtered and washed with water. The mother liquor was extracted with ethyl acetate, and the combined organic layers were washed with brine, dried over sodium sulfate, filtered and concentrated. The resulting residue was combined with the filtered product to give the target compound (433 mg, 71%). MS (ESI) m / z 343.9 (M + H) + .

b) 1-bromo-8-chloro-5,6-dihydro-4 H -[1,2,4] triazolo [4,3- a ] [1] Benzacridin-5-carboxamide
P-Bromo-8-chloro-5,6-dihydro-4 H -[1,2,4] triazolo [4,3- a ] [1] Benzacene-5-carboxylic acid (Intermediate 29 step a)) (620 mg, 1.8 mmol), DMF (8 ml), HOBt (498 mg, 3.25 mmol), DIPEA ( 0.88 ml, 5.06 mmoles) and EDC (623 mg, 3.25 mmoles) in an ice-cold mixture was added ammonium chloride (290 mg, 5.48 mmoles) and the reaction mixture was stirred at 5 ° C for 0.5 hours and then Stir for another 20 hours at room temperature. After completion of the reaction, the mixture was diluted with water and brine and extracted with ethyl acetate. The combined organic layers were dried over sodium sulfate and filtered and concentrated. The residue was crystallized from ethyl acetate to give the target compound (428 mg, 69%). MS (ESI) m / z 343.0 (M + H) + .

Intermediate 30
(5 s ,8 s ) -1-Pentoxy-2- (prop-2-yl) -2-azaspiro [4.5] decane-8-carboxylic acid methyl ester

with
Intermediate 31
(5 r ,8 r ) -1-Pentoxy-2- (prop-2-yl) -2-azaspiro [4.5] decane-8-carboxylic acid methyl ester

Let trans-1- (2-oxoethyl) cyclohexane-1,4-dicarboxylic acid dimethyl ester (WO 2011/143150 A1, (05.10.2011) SANOFI) (0.9 g, 4.0 mmol) ), 1,2-dichloroethane (40 ml), isopropylamine (316 µl, 3.71 mmol) and acetic acid (637 µl, 11.1 mmol) were cooled to 5 ° C and the reaction mixture was added with three Acetyloxyborohydride (2.36 g, 11.1 mmol), where the addition rate maintains the internal temperature below 5 ° C. After the addition was complete, the reaction mixture was allowed to stir at room temperature for 2 hours and then diluted with water. By adding 10% K 2 CO 3 The solution adjusted the pH of the mixture to 8, the phases were separated and the aqueous phase was extracted with dichloromethane. Let the combined organic phase undergo continuous 10% K 2 CO 3 The solution, water and brine were rinsed, dried over sodium sulfate and filtered and concentrated. The residue was dissolved in anhydrous THF (40 ml) and tertiary potassium butoxide (330 mg, 2.94 mmol) was added. The reaction mixture was allowed to stir at room temperature for 3 hours and then CO was added. 2 The solid was neutralized. After adding water, the THF was evaporated and the aqueous phase was extracted with ethyl acetate. The organic phase was dried over sodium sulfate and filtered and concentrated. The residue was purified by flash column chromatography (using a mixture of cyclohexane: ethyl acetate = 45: 55 as the eluent) to produce the first fraction (5 s ,8 s ) -1-Pentoxy-2- (prop-2-yl) -2-azaspiro [4.5] decane-8-carboxylic acid methyl ester (Intermediate 30) (56 mg, 6%) and Distillate (5 r ,8 r ) -1-Phenoxy-2- (prop-2-yl) -2-azaspiro [4.5] decane-8-carboxylic acid methyl ester (Intermediate 31) (172 mg, 19%). GC-MS (EI) m / z 253.

Intermediate 32
(5 r ,8 r ) -1-Pentoxy-2- (prop-2-yl) -2-azaspiro [4.5] decane-8-formamidine

According to the method described in step b) of intermediate 9, from (5 r ,8 r ) -1-Pentoxy-2- (prop-2-yl) -2-azaspiro [4.5] decane-8-carboxylic acid methyl ester (Intermediate 31) to prepare the target compound. GC-MS (EI) m / z 253.

Intermediate 33
4- (1-([tertiarybutyl (dimethyl) silyl] oxy} ethyl) -7-chloro-1,3,4,5-tetrahydro-2 H -1-benzoazepine-2-one


a) 7-Chloro-2-sideoxy-2,3,4,5-tetrahydro-1 H -1-Benzazine-4-carboxylic acid

Let 7-chloro-2- pendantoxy-2,3,4,5-tetrahydro-1 H A mixture of methyl-1-benzoazine-4-carboxylate (Intermediate 3) (12.38 g, 48.8 mmol), methanol (55 ml) and 50% NaOH solution (12 ml) was stirred at room temperature 0.5 hours and then cooled to 0 ° C and 5% hydrochloric acid solution (105 ml) was added. After stirring for 10 minutes at 0 ° C, the precipitated product was filtered, washed with water and dried to give the target compound (11.1 g, 95%). MS (ESI) m / z 240.2 (M + H) + .

b) 7-chloro- N -Methoxy- N -Methyl-2- pendantoxy-2,3,4,5-tetrahydro-1 H -1-benzoazepine-4-carboxamide

According to the method described in step a) of Intermediate 9, from 7-chloro-2- pendantoxy-2,3,4,5-tetrahydro-1 H 1-Benzacne-4-carboxylic acid (Intermediate 33 step a)) and N-methoxymethylamine hydrochloride were used to prepare the target compound, which was used in the next step without further purification.

c) 4-Ethyl-7-chloro-1,3,4,5-tetrahydro-2 H -1-benzoazepine-2-one

Order 7-chloro- under argon N -Methoxy- N -Methyl-2- pendantoxy-2,3,4,5-tetrahydro-1 H A mixture of -1-benzoazepine-4-carboxamidine (Intermediate 33 step b)) (2.5 g, 8.84 mmol) in anhydrous THF (100 ml) was cooled to -15 ° C and 3M methyl was added Diethyl ether solution of magnesium bromide (14.7 ml, 44.1 mmol). The reaction mixture was allowed to stir at -10 ° C to 0 ° C for 3 hours, and then by adding saturated NH 4 The Cl solution was quenched and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate and filtered and concentrated. The residue was purified by flash column chromatography (using dichloromethane: methanol = 96: 4 as the eluent) to give the target compound (1.6 g, 76%). MS (ESI) m / z 238.2 (M + H) + .

d) 7-chloro-4- (1-hydroxyethyl) -1,3,4,5-tetrahydro-2 H -1-benzoazepine-2-one

Let 4-ethenyl-7-chloro-1,3,4,5-tetrahydro-2 H -1-Benzacridin-2-one (Intermediate 33 step c)) (300 mg, 1.26 mmol), ethanol (50 ml), and NaBH 4 The mixture (96 mg, 2.5 mmol) was stirred at room temperature for 0.5 hours, then quenched by the addition of a 1M solution of hydrochloric acid and the ethanol was evaporated. The aqueous phase was extracted with ethyl acetate, and the combined organic layers were washed with brine, dried over sodium sulfate, filtered and concentrated. The residue was purified by flash column chromatography (using dichloromethane: methanol = 95: 5 as the eluent) to give the target compound (235 mg, 78%) as a mixture of 63:37 non-mirror isomers. MS (ESI) m / z 240.2 (M + H) + .

e) 4- (1-([tertiary butyl (dimethyl) silyl] oxy} ethyl) -7-chloro-1,3,4,5-tetrahydro-2 H -1-benzoazepine-2-one
Let 7-chloro-4- (1-hydroxyethyl) -1,3,4,5-tetrahydro-2 H -1-Benzacridin-2-one (Step d) of Intermediate 33) (665 mg, 2.77 mmol), DMF (5 ml), methylene chloride (50 ml), and 2,6-dimethylamine The stirred mixture of pyridine (0.8 ml, 6.94 millimoles) was cooled to 0 ° C and tributylmethane sulfonyl trifluoromethanesulfonate (792 mg, 3.0 millimoles) was added under argon. The reaction mixture was allowed to warm to room temperature and was stirred at this temperature for 16 hours. Evaporate the dichloromethane, add water (10 ml) and saturated NaHCO 3 Solution (10 ml) and the mixture was extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate and filtered and concentrated. The residue was purified by flash column chromatography (using cyclohexane: ethyl acetate = 75: 25 as eluent). The first fraction is one of the non-mirro isomers (200 mg, 20%), the second fraction is a mixture of non-mirro isomers (191 mg, 19%) and the third fraction is the other non-mirro isomer (326 mg, 33%). MS (ESI) m / z 354.2 (M + H) + .

Intermediate 34
4- (1-([tertiarybutyl (dimethyl) silyl] oxy} ethyl) -7-chloro-1,3,4,5-tetrahydro-2 H -1-benzoazepine-2-thione

According to the method described in Intermediate 4, from 4- (1-{[Tri-butyl (dimethyl) silyl] oxy} ethyl) -7-chloro-1,3,4,5-tetrahydro-2 H 1-Benzacridin-2-one (Intermediate 33 step e)) prepares the target compound, which is used in the next step without further purification. Use the same method for a mixture of non-image isomers (second fraction of step e) of intermediate 33) or independently non-image isomers (first and third fractions of step 33 of intermediate 33) .

Intermediate 35
4-methoxy-4-methylcyclohexanemethanehydrazine

a) Ethyl 4-hydroxy-4-methylcyclohexanecarboxylate

To a stirred solution of 2M trimethylaluminum in toluene (100 ml, 220 mmol) under argon was added 4-lanthoxycyclohexanecarboxylic acid ethyl ester (8.7 ml, 55 mmol) at -60 ° C over 2.5 hours. Ear) in toluene (50 ml). After the addition was completed, the mixture was allowed to stir at -60 ° C for 0.5 hours and then warmed to -20 ° C over 2 hours. While maintaining the internal temperature below 10 ° C, the reaction mixture was transferred to ethyl acetate (180 ml), water (425 ml), concentrated hydrochloric acid (75 ml), and crushed ice by intubation over 25 to 30 minutes. (100 g) of ice-cold mixture. The layers were separated and the organic phase was washed successively with water (400 ml) and brine (400 ml), dried over sodium sulfate, filtered and concentrated to give the target compound (5.44 g, 53%). in accordance with 1 HNMR spectrum, the compound is a 28:72 mixture of cis and trans isomers. This mixture was used in the next step without further purification.

b) Ethyl 4-methoxy-4-methylcyclohexanecarboxylate

Under argon, a solution of 60% sodium hydride in mineral oil (2.16 g, 54 mmol), anhydrous THF (34 ml), tetrabutylammonium iodide (200 mg, 0.54 mmol), imidazole (49 mg , 0.72 millimolar) and a solution of methyl iodide (3.36 ml, 54 millimolar) at 20 to 25 ° C over 30 to 40 minutes. Ethyl 4-hydroxy-4-methylcyclohexanecarboxylic acid (intermediate Step a)) of 35 (3.36 g, 18 mmol) in anhydrous THF (21 ml). The reaction mixture was allowed to stir at room temperature for 3 hours, then cooled to 0 to 5 ° C and acetic acid (2.28 ml, 40 mmol) was added over 10 minutes. Allow the mixture to stir for 15 minutes, then pour into diethyl ether (280 ml) and saturated NaHCO 3 A mixture of solutions (120 ml). The layers were separated and the organic phase was washed with brine, dried over sodium sulfate and filtered and concentrated. The residue was purified by column chromatography (using n-hexane: ethyl acetate = 85: 15 as the eluent) to give the target compound (2.3 g, 64%). in accordance with 1 HNMR spectrum, the compound is a 21:79 mixture of cis and trans isomers.

c) 4-methoxy-4-methylcyclohexanemethanehydrazine
The subject compound was prepared from 4-methoxy-4-methylcyclohexanecarboxylic acid ethyl ester (intermediate 35 step b)) according to the method described in intermediate step 9). in accordance with 1 HNMR spectrum, the compound is a 21:79 mixture of cis and trans isomers.

Intermediate 36
7-chloro-4- (fluoromethyl) -1,3,4,5-tetrahydro-2 H -1-benzoazepine-2-one

a) 7-Chloro-4- (hydroxymethyl) -1,3,4,5-tetrahydro-2 H -1-benzoazepine-2-one

Order 7-chloro-2-lanthoxy-2,3,4,5-tetrahydro-1 under argon H A stirred mixture of methyl-1-benzoazine-4-carboxylic acid methyl ester (Intermediate 3) (500 mg, 1.97 mmol), anhydrous diethyl ether (15 ml) and anhydrous THF (20 ml) was cooled to -40 ° C. A 1 M solution of lithium aluminum hydride in diethyl ether (4.33 ml, 4.33 mmol) was added. The reaction mixture was allowed to stir at -20 ° C for 1 hour and then warmed to 0 ° C and water was carefully added. The THF was evaporated and the aqueous phase was extracted with ethyl acetate. The combined organic phases were washed with brine, dried over sodium sulfate and filtered and concentrated to give the title compound (422 mg, 95%). MS (ESI) m / z 226.1 (M + H) + .

b) 7-chloro-4- (fluoromethyl) -1,3,4,5-tetrahydro-2 H -1-benzoazepine-2-one
Order 7-chloro-4- (hydroxymethyl) -1,3,4,5-tetrahydro-2 under argon H A stirred mixture of -1-benzoazepine-2-one (Intermediate 36 step a)) (532 mg, 2.36 mmol) and anhydrous dichloromethane (50 ml) was cooled to -78 ° C and added dropwise (Diethylamino) sulfur trifluoride (950 mg, 5.89 mmol) in dichloromethane (1 ml). The reaction mixture was allowed to warm to 0 ° C over 4 hours, stirred at this temperature for 45 minutes and then saturated NaHCO was added. 3 Solution. The layers were separated, the aqueous phase was extracted with dichloromethane, the combined organic phases were washed with brine, dried over sodium sulfate, filtered and concentrated. The residue was purified by reverse-phase flash column chromatography (using water (containing 0.1% trifluoroacetic acid): acetonitrile = 60:40 as the eluent) to give the target compound (222 mg, 41%). MS (ESI) m / z 228.1 (M + H) + .

Intermediate 37
7-chloro-4- (fluoromethyl) -1,3,4,5-tetrahydro-2 H -1-benzoazepine-2-thione

According to the method described in Intermediate 4, from 7-chloro-4- (fluoromethyl) -1,3,4,5-tetrahydro-2 H 1-Benzacridin-2-one (step b) of intermediate 36) prepares the target compound, which is used in the next step without further purification.

Intermediate 38
7-fluoro-2-oxo-2,3,4,5-tetrahydro-1 H Methyl-1-benzoazine-4-carboxylic acid

a) 7-Fluoro-4- (hydroxyimino) -1,2,3,4-tetrahydronaphthalene-2-carboxylic acid methyl ester

Prepared according to the method described in Intermediate 2 from methyl 7-fluoro-4-oxo-1,2,3,4-tetrahydronaphthalene-2-carboxylic acid (US5595872 (03.09.1993) Bristol-Myers Squibb Company) The target compound was used in the next step without further purification.

b) 7-fluoro-2-oxo-2,3,4,5-tetrahydro-1 H Methyl-1-benzoazine-4-carboxylic acid
Prepare the target compound from 7-fluoro-4- (hydroxyimino) -1,2,3,4-tetrahydronaphthalene-2-carboxylic acid methyl ester (Intermediate 38 step a)) according to the method described in Intermediate 3. Compound. MS (ESI) m / z 238.2 (M + H) + .

Intermediate 39
7-fluoro-2- pendant thio-2,3,4,5-tetrahydro-1 H Methyl-1-benzoazine-4-carboxylic acid

According to the method described in Intermediate 4, from 7-fluoro-2- pendantoxy-2,3,4,5-tetrahydro-1 H Methyl-1-benzoazine-4-carboxylic acid (Intermediate 38, step b)) prepared the target compound, which was used in the next step without further purification.

Intermediate 40
7-fluoro-4- (fluoromethyl) -1,3,4,5-tetrahydro-2 H -1-benzoazepine-2-one

a) 7-Fluoro-4- (hydroxymethyl) -1,3,4,5-tetrahydro-2 H -1-benzoazepine-2-one

According to the method described in step a) of Intermediate 36, from 7-fluoro-2- pendantoxy-2,3,4,5-tetrahydro-1 H Methyl-1-benzoazine-4-carboxylic acid (Intermediate 38, step b)) prepared the target compound, which was used in the next step without further purification.

b) 7-fluoro-4- (fluoromethyl) -1,3,4,5-tetrahydro-2 H -1-benzoazepine-2-one
According to the method described in step b) of Intermediate 36, from 7-fluoro-4- (hydroxymethyl) -1,3,4,5-tetrahydro-2 H 1-Benzacridin-2-one (Intermediate 40, step a)) prepares the target compound, which is used in the next step without further purification.

Intermediate 41
7-fluoro-4- (fluoromethyl) -1,3,4,5-tetrahydro-2 H -1-benzoazepine-2-thione

According to the method described in Intermediate 4, from 7-fluoro-4- (fluoromethyl) -1,3,4,5-tetrahydro-2 H 1-Benzacridin-2-one (Intermediate 40, step b)) prepares the target compound, which is used in the next step without further purification.

Intermediate 42
4- (fluoromethyl) -7-methyl-1,3,4,5-tetrahydro-2 H -1-benzoazepine-2-one

a) 4- (hydroxymethyl) -7-methyl-1,3,4,5-tetrahydro-2 H -1-benzoazepine-2-one

According to the method described in step a) of intermediate 36, from 7-methyl-2- pendantoxy-2,3,4,5-tetrahydro-1 H Methyl-1-benzoazine-4-carboxylic acid (prepared according to the method described in Intermediate 3) was used to prepare the target compound, which was used in the next step without further purification.

b) 4- (fluoromethyl) -7-methyl-1,3,4,5-tetrahydro-2 H -1-benzoazepine-2-one
From 4- (hydroxymethyl) -7-methyl-1,3,4,5-tetrahydro-2 according to the method described in step b) of intermediate 36 H 1-Benzacridin-2-one (Intermediate 42 step a)) to prepare the target compound. MS (ESI) m / z 208.2 (M + H) + .

Intermediate 43
4- (fluoromethyl) -7-methyl-1,3,4,5-tetrahydro-2 H -1-benzoazepine-2-thione

From 4- (fluoromethyl) -7-methyl-1,3,4,5-tetrahydro-2 according to the method described in Intermediate 4 H 1-Benzacridin-2-one (Intermediate 42 step b)) prepares the target compound, which is used in the next step without further purification.

Intermediate 44
7-fluoro-2- (methylthio) -4,5-dihydro-3 H Methyl-1-benzoazine-4-carboxylic acid

According to the method described in Intermediate 22, from 7-fluoro-2- pendant thio-2,3,4,5-tetrahydro-1 H Methyl-1-benzoazine-4-carboxylic acid (Intermediate 39) prepared the target compound. MS (ESI) m / z 268.1 (M + H) + .

Intermediate 45
7-chloro-4-methyl-2-oxo-2,3,4,5-tetrahydro-1 H Methyl-1-benzoazine-4-carboxylic acid

a) 7-Chloro-1- (4-methoxybenzyl) -2-oxo-2,3,4,5-tetrahydro-1 H Methyl-1-benzoazine-4-carboxylic acid

Let 7-chloro-2- pendantoxy-2,3,4,5-tetrahydro-1 H A stirred mixture of methyl-1-benzoacryl-4-carboxylate (Intermediate 3) (1.0 g, 3.94 mmol) in anhydrous DMF (10 ml) was cooled to 0 ° C and 60% hydrogenated under argon. A solution of sodium (205 mg, 5.12 mmol) in mineral oil. The mixture was allowed to stir at this temperature for 1 hour, then 4-methoxybenzyl chloride (0.8 ml, 5.9 mmol) was added and the reaction mixture was stirred at room temperature for 20 hours. After the reaction was completed, the mixture was diluted with water and brine, extracted with ethyl acetate, and the combined organic layers were washed with brine, dried over sodium sulfate, filtered and concentrated. The residue was purified by flash column chromatography (using cyclohexane: ethyl acetate = 4: 1 as the eluent) to give the target compound (820 mg, 56%). MS (ESI) m / z 374.1 (M + H) + .

b) 7-Chloro-1- (4-methoxybenzyl) -4-methyl-2- pendantoxy-2,3,4,5-tetrahydro-1 H Methyl-1-benzoazine-4-carboxylic acid

Let 7-chloro-1- (4-methoxybenzyl) -2- pendantoxy-2,3,4,5-tetrahydro-1 H Methyl-1-benzoazine-4-carboxylic acid methyl ester (Intermediate 45, step a)) (600 mg, 1.61 mmol) was stirred in anhydrous THF (30 ml) and cooled to -70 ° C under argon. A 2.0 M solution of lithium diisopropylamide in THF / heptane / ethylbenzene (1.2 ml, 2.4 mmol) was added. The mixture was allowed to stir at this temperature for 2 hours, then methyl iodide (0.2 ml, 3.2 mmol) was added and the reaction mixture was allowed to warm to 0 ° C over 2 hours. After the reaction was completed, the mixture was quenched by the addition of saturated ammonium chloride, extracted with ethyl acetate, the combined organic layers were washed with brine, dried over sodium sulfate, filtered and concentrated. The residue was purified by flash column chromatography (using cyclohexane: ethyl acetate = 4: 1 as the eluent) to give the target compound (460 mg, 74%). MS (ESI) m / z 388.1 (M + H) + .

c) 7-chloro-4-methyl-2-oxo-2,3,4,5-tetrahydro-1 H Methyl-1-benzoazine-4-carboxylic acid
Let 7-chloro-1- (4-methoxybenzyl) -4-methyl-2- pendantoxy-2,3,4,5-tetrahydro-1 H Methyl-1-benzoazine-4-carboxylate (Intermediate 45 step b)) (460 mg, 1.2 mmol), acetonitrile (20 ml), water (5 ml), and cerium (IV) ammonium nitrate (1.95 g, 3.56 mmol) was stirred at room temperature for 2 hours and then diluted with water. By adding saturated NaHCO 3 The solution adjusted the pH of the mixture to 8 and then extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate and filtered and concentrated. The residue was purified by flash column chromatography (using cyclohexane: ethyl acetate = 4: 1 as the eluent) to give the target compound (190 mg, 60%). MS (ESI) m / z 268.1 (M + H) + .

Intermediate 46
7-chloro-4-methyl-2- pendant thio-2,3,4,5-tetrahydro-1 H Methyl-1-benzoazine-4-carboxylic acid

According to the method described in Intermediate 25, from 7-chloro-4-methyl-2- pendantoxy-2,3,4,5-tetrahydro-1 H Methyl-1-benzoazine-4-carboxylic acid (Intermediate 45, step c)) The title compound was prepared and used in the next step without further purification.

Intermediate 47
7-chloro-4-ethyl-2-oxo-2,3,4,5-tetrahydro-1 H Methyl-1-benzoazine-4-carboxylic acid


a) 7-Chloro-1- (4-methoxybenzyl) -4-ethyl-2- pendantoxy-2,3,4,5-tetrahydro-1 H Methyl-1-benzoazine-4-carboxylic acid

According to the method described in step b) of Intermediate 45, from 7-chloro-1- (4-methoxybenzyl) -2- pendantoxy-2,3,4,5-tetrahydro-1 H Methyl-1-benzoazine-4-carboxylic acid (Intermediate 45, step a)) and iodoethane prepared the target compound, which was used in the next step without further purification.

b) 7-chloro-4-ethyl-2-oxo-2,3,4,5-tetrahydro-1 H Methyl-1-benzoazine-4-carboxylic acid
According to the method described in step c) of intermediate 45, from 7-chloro-1- (4-methoxybenzyl) -4-ethyl-2- pendantoxy-2,3,4,5-tetrahydro -1 H Methyl-1-benzoazine-4-carboxylic acid (Intermediate 47, step a)) to prepare the target compound. MS (ESI) m / z 282.1 (M + H) + .

Intermediate 48
7-chloro-4-ethyl-2-sulfanyl-2,3,4,5-tetrahydro-1 H Methyl-1-benzoazine-4-carboxylic acid

According to the method described in Intermediate 25, from 7-chloro-4-ethyl-2- pendantoxy-2,3,4,5-tetrahydro-1 H Methyl-1-benzoazine-4-carboxylic acid (step b) of intermediate 47) prepared the target compound, which was used in the next step without further purification.

Intermediate 49
7-chloro-4-ethyl-2- (methylthio) -4,5-dihydro-3 H Methyl-1-benzoazine-4-carboxylic acid

According to the method described in Intermediate 22, from 7-chloro-4-ethyl-2- pendant thio-2,3,4,5-tetrahydro-1 H Methyl-1-benzoazine-4-carboxylic acid (Intermediate 48) prepared the title compound, which was used in the next step without further purification.

Intermediate 50
Tert-butyl 2-[(3,3-difluorocyclobutyl) carbonyl] hydrazine formate

The target compound was prepared from 3,3-difluorocyclobutanecarboxylic acid (Combi-Blocks Inc.) according to the method described in Intermediate 5. GC-MS (EI) m / z 250.1.

Intermediate 51
3,3-difluorocyclobutanemethanehydrazine

The title compound was prepared from tert-butyl 2-[(3,3-difluorocyclobutyl) carbonyl] hydrazinecarboxylic acid (intermediate 50) according to the method described in Intermediate 6. GC-MS (EI) m / z 150.1.

Intermediate 52
1-bromo-8-chloro-5,6-dihydro-4 H -[1,2,4] triazolo [4,3- a ] [1] Benzacridin-5-carbonitrile

Let 1-bromo-8-chloro-5,6-dihydro-4 H -[1,2,4] triazolo [4,3- a ] [1] Benzacridine-5-carboxamide (Intermediate 29 step b)) (155 mg, 0.45 mmol), anhydrous dichloromethane (77 ml), and triethylamine (400 µl, 2.86 The stirred solution was cooled to 0 ° C and trifluoroacetic anhydride (105 µl, 0.73 mmol) was added. The mixture was allowed to warm to room temperature and the reaction mixture was stirred at this temperature for 1 hour, then cooled to 0 ° C and triethylamine (400 µl, 2.86 mmol) and trifluoroacetic anhydride (105 µl, 0.73 millimolar). The mixture was allowed to stir at room temperature for 2.5 hours and then saturated NaHCO 3 The solution was diluted. The phases were separated and the aqueous phase was extracted with dichloromethane. The combined organic phases were washed with water and brine, dried over sodium sulfate and filtered and concentrated. The residue was purified by column chromatography (using dichloromethane: methanol: ammonium hydroxide = 180: 10: 1 as eluent) to give the target compound (125 mg, 85%). MS (ESI) m / z 323.0 (M + H) + .

Intermediate 53
7-fluoro-4,4-dimethyl-1,3,4,5-tetrahydro-2 H -1-benzoazepine-2-one

a) [1- (3-Fluorophenyl) -2-methylprop-2-yl] diethyl malonate

Slowly add 0.5M 3-fluorobenzylmagnesium chloride (Alfa Aesar) in 2-Me-THF (33 ml, 16.5 mmol) to (propan-2-yl) via a dropping funnel under argon over 4 hours ) A solution of diethyl malonate (Aldrich) (3.0 g, 15.0 mmol) in diethyl ether (150 ml). Stirring was then continued at room temperature for 18 hours. The reaction was then quenched with a 1M hydrochloric acid solution and extracted with ethyl acetate. The combined organic phases were washed with a saturated NaCl solution, dried over anhydrous sodium sulfate, filtered and concentrated. Thus, the target product was obtained as a yellow oil (4.30 g, 92.5%). MS (ESI) m / z 311.1 (M + H) + .

b) [1- (3-Fluorophenyl) -2-methylprop-2-yl] malonate

Let [1- (3-fluorophenyl) -2-methylprop-2-yl] diethyl malonate (4.30 g, 13.9 mmol), 2M aqueous NaOH (60 ml, 120 mmol) And ethanol (60 ml) solution was stirred overnight at 50 ° C. The organic solvent was removed under vacuum, the residue was acidified with 3M hydrochloric acid and extracted with dichloromethane, dried over anhydrous sodium sulfate, filtered and concentrated. Thus, the target product was obtained as a slightly brown oil (3.43 g, 97%). MS (ESI) m / z 255.1 (M + H) + .

c) 4- (3-fluorophenyl) -3,3-dimethylbutanoic acid

[1- (3-Fluorophenyl) -2-methylprop-2-yl] malonic acid (3.43 g, 13.5 mmol) was heated at 210 ° C until gas generation ceased and then cooled to room temperature . Thus, the target product was obtained as an oil (2.35 g, 83%). MS (ESI) m / z 211.1 (M + H) + .

d) 6-fluoro-3,3-dimethyl-3,4-dihydronaphthalene-1 (2H) -one

Concentrated sulfuric acid (8 ml) was added to 4- (3-fluorophenyl) -3,3-dimethylbutanoic acid (2.35 g, 11.2 mmol) and the resulting mixture was stirred at 40 ° C for 1 hour. It was then poured onto ice, extracted with ethyl acetate, washed with saturated NaCl solution, dried over anhydrous sodium sulfate, filtered and concentrated. Thus, the target product was obtained as a slightly brown oil (1.51 g, 70%). MS (ESI) m / z 193.4 (M + H) + .

e) N- (6-fluoro-3,3-dimethyl-3,4-dihydronaphthalene-1 (2H) -subunit) hydroxylamine

Let 6-fluoro-3,3-dimethyl-3,4-dihydronaphthalene-1 (2H) -one (1.51 g, 7.86 mmol), hydroxylamine hydrochloride (Aldrich) (3.01 g, 43.3 A solution of sodium mol), anhydrous sodium acetate (Aldrich) (3.61 g, 44.0 mmol) and methanol (55 ml) were refluxed overnight. The solvent was then removed under vacuum, water and ethyl acetate were added, the layers were separated, the aqueous phase was extracted with ethyl acetate, the combined organic phases were washed with saturated NaCl solution, dried over anhydrous sodium sulfate, filtered and concentrated. Thus, the target product was obtained as a beige solid (1.45 g, 89%). MS (ESI) m / z 208.1 (M + H) + .

f) 7-fluoro-4,4-dimethyl-1,3,4,5-tetrahydro-2H-1-benzoazepine-2-one
N- (6-fluoro-3,3-dimethyl-3,4-dihydronaphthalene-1 (2 H ) -Subunit) hydroxylamine (1.45 g, 7.00 mmol) was added to the polyphosphoric acid (15.0 g) and stirred at 130 ° C for 20 minutes. Ice was added to the reaction mixture and stirred for 10 minutes. It was then extracted with chloroform, dried over anhydrous sodium sulfate and filtered and concentrated. The residue was triturated with diethyl ether and the precipitate was collected by suction. Thus, the target product was obtained as white crystals (1.10 g, 76%). MS (ESI) m / z 208.2 (M + H) + .

Intermediate 54
7-fluoro-4- (prop-2-yl) -1,3,4,5-tetrahydro-2 H -1-benzoazepine-2-one

The target product was prepared from diethyl 2- (2-methylpropylene) malonate (Combi Blocks) according to the method described in Intermediate 53. MS (ESI) m / z 222.1 (M + H) + .

Intermediate 55
1,5-dihydrospiro [1-benzoazine-4,1'-cyclopentane] -2 (3H) -one

a) (1-Benzylcyclopentyl) diethylmalonate

A 2M solution of benzyl magnesium chloride in THF (Aldrich) (12 ml, 24 mmol) was slowly added to the diethyl 2-cyclopentylmalonate (Lehnert, W. ., Tetrahedron 1973, 29 : 635-638) (5.0 g, 22.1 mmol) in diethyl ether (100 ml). Stirring was then continued at room temperature for 18 hours. The reaction was then quenched with a 1M hydrochloric acid solution and extracted with ethyl acetate. The combined organic phases were washed with a saturated NaCl solution, dried over anhydrous sodium sulfate, filtered and concentrated. Thus, the target product was obtained as an oil (5.62 g, 80%). MS (ESI) m / z 319.4 (M + H) + .
b) (1-Benzylcyclopentyl) malonic acid

Let (1-benzylcyclopentyl) diethyl malonate (Step a) of Intermediate 55) (5.62 g, 17.7 mmol), 2M aqueous NaOH (60 ml, 120 mmol) and ethanol ( 30 ml) of the solution was refluxed for 2 hours. The organic solvent was removed under vacuum, the residue was acidified with 3M hydrochloric acid and extracted with ethyl acetate. The combined organic phases were washed with saturated NaCl solution, dried over anhydrous sodium sulfate, filtered and concentrated. Thus, the target product was obtained as an oil (3.52 g, 76%). MS (ESI) m / z 263.3 (M + H) + .

c) (1-Benzylcyclopentyl) acetic acid

(1-Benzylcyclopentyl) malonic acid (Step b) of Intermediate 55) (3.52 g, 13.4 mmol) was heated at 180 ° C until gas evolution ceased and then cooled to room temperature. Thus, the target product was obtained as an oil (2.90 g, 99%). MS (ESI) m / z 219.1 (M + H) + .

d) 1 ' H -Spiro [cyclopentane-1,2'-naphthalene] -4 '(3' H )-ketone

Concentrated sulfuric acid (Aldrich) (20 ml) was added to (1-benzylcyclopentyl) acetic acid (Step c) of Intermediate 55) (2.90 g, 13.3 mmol) and the resulting mixture was allowed to stand at 35 ° C. Stir for 1 hour. It was then poured onto ice and extracted with ethyl acetate. The combined organic phases were washed with saturated NaCl solution, dried over anhydrous sodium sulfate, filtered and concentrated. The residue was placed in dichloromethane and filtered through a pad of silica to remove colored impurities. Thus, the target product was obtained as an oil (1.41 g, 53%). MS (ESI) m / z 201.2 (M + H) + .

e) N-1 ' H -Spiro [cyclopentane-1,2'-naphthalene] -4 '(3' H ) -Subunit hydroxylamine

Order 1 ' H -Spiro [cyclopentane-1,2'-naphthalene] -4 '(3' H ) -Ketone (step d)) of Intermediate 55) (1.41 g, 7.05 mmol), hydroxylamine hydrochloride (Aldrich) (1.95 g, 28 mmol), anhydrous sodium acetate (Aldrich) (2.30 g, 28 mmol) and methanol (40 ml) were refluxed for 2 hours. The solvent was then removed under vacuum, water and ethyl acetate were added, the layers were separated, the aqueous phase was extracted with ethyl acetate, the combined organic phases were washed with saturated NaCl solution, dried over anhydrous sodium sulfate, filtered and concentrated. Thus, the target product was obtained as a beige solid (1.33 g, 88%). MS (ESI) m / z 216.1 (M + H) + .

f) 1,5-Dihydrospiro [1-benzoazine-4,1'-cyclopentane] -2 (3H) -one
Will N-1 ' H -Spiro [cyclopentane-1,2'-naphthalene] -4 '(3' H ) -Subunit hydroxyamine (Intermediate 55 step e)) (1.33 g, 6.19 mmol) was added to the polyphosphoric acid (20.0 g) and stirred at 110 ° C for 20 minutes. Ice was added to the reaction mixture and stirred for 10 minutes. It was then extracted with ethyl acetate, and the combined organic phases were washed with saturated NaCl solution, dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by column chromatography (using cyclohexane: ethyl acetate = 2: 1 as eluent). Thus, the target product was obtained as white crystals (0.51 g, 38%). MS (ESI) m / z 216.1 (M + H) + .

Intermediate 56
4- (prop-2-yl) -1,3,4,5-tetrahydro-2 H -1-benzoazepine-2-one

The target product was prepared from diethyl 2- (2-methylpropylene) malonate (Combi Blocks) according to the method described in Intermediate 53. MS (ESI) m / z 204.1 (M + H) + .

Intermediate 57
1,5-dihydrospiro [1-benzoazine-4,4'-ethylene oxide] -2 (3 H )-ketone

According to the method described in Intermediate 53, from tetrahydro-4 H -Piperan-4-ylidene malonate (Griffiths et al., HeIv Chim Acta 1991, 74 (2): 09-314) Prepare the target product. MS (ESI) m / z 232.2 (M + H) + .

Intermediate 58
1,4 ', 5,5'-tetrahydrospiro [1-benzoazine-4,3'-furan] -2 (3 H )-ketone

According to the method described in Intermediate 53, from dihydrofuran-3 (2H) -diylmalonate (Griffiths et al., HeIv Chim Acta 1991, 74 (2): 09-314) Prepare the target product. MS (ESI) m / z 218.2 (M + H) + .
Intermediate 59
1,5,5 ', 6'-tetrahydro-4' H -Spiro [1-benzoazine-4,3'-piperan] -2 (3 H )-ketone

According to the method described in Intermediate 53, from dihydro-2H-piperan-3 (4H) -diylmalonate (Griffiths et al., HeIv Chim Acta 1991, 74 2): 09-314) Prepare the target product. MS (ESI) m / z 232.1 (M + H) + .

Intermediate 60
7-bromo-1,5-dihydrospiro [1-benzoazine-4,1'-cyclopentane] -2 (3 H )-ketone

P-1,5-Dihydrospiro [1-benzoazine-4,1'-cyclopentane] -2 (3 H ) -Ketone (Intermediate 55) (0.51 g, 2.37 mmol), acetic acid (10 ml) and water (2 ml) was slowly added to the solution of Aldrich (0.121 ml, 2.37 mmol) in acetic acid (3 ml) solution. After stirring the reaction mixture at room temperature for 1 hour, it was poured onto ice, and the precipitate was filtered by suction and dried. Thus, the target product was obtained as beige crystals (0.50 g, 72%). MS (ESI) m / z 294.2 & 296.1 (M + H) + .

Intermediate 61
7-bromo-4- (prop-2-yl) -1,3,4,5-tetrahydro-2 H -1-benzoazepine-2-one

From 4- (prop-2-yl) -1,3,4,5-tetrahydro-2 according to the method described in Intermediate 60 H 1-Benzacridin-2-one (Intermediate 56) prepared the target product. MS (ESI) m / z 282.1 & 284.1 (M + H) + .

Intermediate 62
7-bromo-1,5-dihydrospiro [1-benzoazine-4,4'-ethylene oxide] -2 (3 H )-ketone

According to the method described in Intermediate 60, from 1,5-Dihydrospiro [1-benzoazine-4,4'-ethylene oxide] -2 (3 H ) -One (Intermediate 57) to prepare the target product. MS (ESI) m / z 310.1 & 312.1 (M + H) + .

Intermediate 63
7-bromo-1,5-dihydrospiro [1-benzoazine-4,3'-tetrahydrofuran] -2 (3 H )-ketone

According to the method described in intermediate 60, from 1,4 ', 5,5'-tetrahydrospiro [1-benzoazine-4,3'-furan] -2 (3 H ) -One (Intermediate 58) to prepare the target product. MS (ESI) m / z 296.2 & 298.2 (M + H) + .

Intermediate 64
7-chloro-1,5-dihydrospiro [1-benzoazine-4,4'-ethylene oxide] -2 (3 H )-ketone

P-1,5-Dihydrospiro [1-benzoazine-4,4'-ethylene oxide] -2 (3 H ) -Ketone (Intermediate 57) (0.73 g, 3.16 mmol) in DMF (20 ml) was added with N-chlorosuccinimide (0.59 g, 4.42 mmol). After stirring the reaction mixture at room temperature for 1 hour, it was poured onto ice, and the precipitate was filtered by suction and dried. Thus, the target product was obtained as yellow crystals (0.58 g, 69%). MS (ESI) m / z 266.1 (M + H) + .

Intermediate 65
7-chloro-1,5-dihydrospiro [1-benzoazine-4,3'-ethylene oxide] -2 (3 H )-ketone

According to the method described in Intermediate 64, from 1,5,5 ', 6'-tetrahydro-4' H -Spiro [1-benzoazine-4,3'-piperan] -2 (3 H ) -One (Intermediate 59) to prepare the target product. MS (ESI) m / z 266.2 (M + H) + .

Intermediate 66
7-chloro-1,5-dihydrospiro [1-benzoazine-4,3'-tetrahydrofuran] -2 (3 H )-ketone

According to the method described in Intermediate 64, from 1,4 ', 5,5'-tetrahydrospiro [1-benzoazine-4,3'-furan] -2 (3 H ) -One (Intermediate 58) to prepare the target product. MS (ESI) m / z 252.2 (M + H) + .

Intermediate 67
Trans-4- (pyridin-2-ylamino) cyclohexanemethylhydrazine

a) Methyl trans-4- (pyridin-2-ylamino) cyclohexanecarboxylate

Let trans-4-aminocyclohexanecarboxylic acid methyl ester hydrochloride (1.5 g, 7.745 mmol), 2-fluoropyridine (4 ml, 46.5 mmol) and DIPEA (1.35 ml, 7.75 mmol) The mixture) was stirred in a pressure-resistant glass reactor at 125 ° C. for 20 hours and then cooled to room temperature. The reaction mixture was diluted with ethyl acetate (20 ml), washed with water (2 x 30 ml) and saturated NaCl solution (2 x 30 ml), and the organic phase was dried over anhydrous sodium sulfate and filtered and concentrated to produce The target compound (325 mg, 18%). MS (ESI) m / z 235.1 (M + H) + .

b) trans-4- (pyridin-2-ylamino) cyclohexanemethylhydrazine
The title compound was prepared from trans-4- (pyridin-2-ylamino) cyclohexanecarboxylic acid methyl ester (intermediate 67 step a)) according to the method described in intermediate step 9) without further purification. Purified and used in the next step.

Example 1
8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a ] [1] Methylbenzoazine-5-carboxylic acid

Let 7-chloro-2- pendant thio-2,3,4,5-tetrahydro-1 H Methyl-1-benzoazine-4-carboxylic acid (Intermediate 4) (92 mg, 0.34 mmol), trans-4- (pyridin-2-yloxy) cyclohexanemethanehydrazine (WO2010 / 060836 A1 (03.06.2010) A solution of F. HOFFMANN-LA ROCHE AG.) (96.5 mg, 0.41 mmol) and xylene (10 ml) was stirred at reflux temperature for 3 days. The reaction mixture was concentrated and the residue was purified by column chromatography using dichloromethane: methanol = 20: 1 as the eluent. Thus, the target product was obtained (68 mg, 44.0%). MS (ESI) m / z 454.2 (M + H) + .

Example 2
8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a ] [1] Benzacene-5-carboxylic acid

Let 8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3- a] [1] Methyl benzoazine-5-carboxylic acid (Example 1) (0.85 g, 1.88 mmol) was dissolved in methanol (60 ml), followed by the addition of a 2M NaOH solution (20 ml) and the mixture was Stir overnight. Methanol was evaporated from the reaction mixture, the pH of the aqueous phase was adjusted to 3 with 1M hydrochloric acid solution, and then extracted with ethyl acetate. The combined organic phases were washed with a saturated NaCl solution, dried over anhydrous sodium sulfate and filtered and concentrated. Thus, the target product was obtained (0.83 g, 99.9%). MS (ESI) m / z 439.1 (M + H) + .

Example 3
8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a ] [1] Benzacridin-5-carboxamide

Let 8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3- a] [1] Benzacridine-5-carboxylic acid (Example 2) (0.4 g, 0.91 mmol), HOBt (0.25 g, 1.64 mmol) and EDC (0.314 g, 1.64 mmol) were dissolved In anhydrous DMF (3 ml). The reaction mixture was cooled in an ice water bath and ammonium chloride (0.146 g, 2.73 mmol) and DIPEA (0.443 ml, 2.55 mmol) were added under argon. The reaction mixture was allowed to stir at 0 ° C for 2 hours and then overnight at room temperature. Water was added to the reaction mixture, and the mixture was extracted with ethyl acetate. The combined organic phases were washed with a saturated NaCl solution, dried over anhydrous sodium sulfate, filtered and concentrated under vacuum. Thus, the target product was obtained (0.34 g, 85.4%). MS (ESI) m / z 438.2 (M + H) + .

Example 4
8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a ] [1] Benzacridin-5-carbonitrile

Let 8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3- a] [1] Benzoazine-5-carboxamide (Example 3) (0.26 g, 0.60 mmol) was dissolved in pyridine (4.4 ml). It was then cooled to 5 ° C and phosphonium chloride (56 μl, 0.60 mmol) was added to the reaction mixture. The mixture was allowed to stir overnight at room temperature. The reaction mixture was then concentrated, water was added to the residue and the pH of the aqueous phase was adjusted to 9 with a 1M NaOH solution. The aqueous phase was extracted with ethyl acetate. The combined organic phases were washed with a saturated NaCl solution, dried over anhydrous sodium sulfate and filtered and concentrated. The residue was purified by column chromatography using dichloromethane: methanol = 20: 1 as the eluent. Thus, the target product was obtained (0.177 g, 70.2%). MS (ESI) m / z 420.2 (M + H) + .

Example 5
8-chloro-N, N-dimethyl-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] Zolo [4,3-a] [1] benzoazine-5-carboxamide

According to the method described in Example 3, from 8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] Triazolo [4,3-a] [1] benzoazine-5-carboxylic acid (Example 2) and dimethylamine (2M THF solution) were used to prepare the target product. MS (ESI) m / z 466.2 (M + H) + .

Example 6
(5S) -8-chloro-N, N-dimethyl-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2 , 4] triazolo [4,3-a] [1] benzoazepine-5-carboxamide

with
Example 7
(5R) -8-chloro-N, N-dimethyl-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2 , 4] triazolo [4,3-a] [1] benzoazepine-5-carboxamide

By chiral preparative HPLC (CHIRALPAK IA (containing preparative 20 μm stationary phase) 2.5 x 20 cm; F = 15 ml / min; eluent: n-hexane: EtOH = 75: 25 + 0.3% diethylamine; Isocratic; temperature = 25 ℃), self-racemic 8-chloro-N, N-dimethyl-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6- Dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazine-5-carboxamide (Example 5) was used to prepare the target product. The residence time of the first eluted compound was 11.3 minutes; = -8.3˚ (c = 0.1; methanol); the residence time of the second eluting compound is 19.7 minutes; = + 4.9˚ (c = 0.1; methanol). The absolute configuration of these compounds has not been determined.

Example 8
8-chloro-N- (prop-2-yl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4 ] Triazolo [4,3-a] [1] benzoazine-5-carboxamide

According to the method described in Example 3, from 8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] Triazolo [4,3-a] [1] benzoazine-5-carboxylic acid (Example 2) and isopropylamine were used to prepare the target product. MS (ESI) m / z 480.2 (M + H) + .

Example 9
{8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3- a] [1] Benzacridin-5-yl} (pyrrolidin-1-yl) methanone

According to the method described in Example 3, from 8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] Triazolo [4,3-a] [1] benzoazine-5-carboxylic acid (Example 2) and pyrrolidine were used to prepare the target product. MS (ESI) m / z 492.2 (M + H) + .

Example 10
{8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3- a] [1] Benzacridin-5-yl} (morpholin-4-yl) methanone

According to the method described in Example 3, from 8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] Triazolo [4,3-a] [1] benzoazine-5-carboxylic acid (Example 2) and morpholine prepared the target product. MS (ESI) m / z 508.2 (M + H) + .

Example 11
{8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3- a] [1] Benzacridin-5-yl} (1,1-dioxythiomorpholin-4-yl) methanone

According to the method described in Example 3, from 8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] Triazolo [4,3-a] [1] benzoazine-5-carboxylic acid (Example 2) and thiomorpholine-1,1-dioxide were used to prepare the target product. MS (ESI) m / z 556.2 (M + H) + .

Example 12
{8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3- a] [1] Benzacridin-5-yl} methanol

Let 8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3- a] [1] Methyl benzoazine-5-carboxylate (Example 1) (1.11 g, 2.51 mmol) was dissolved in a mixture of anhydrous diethyl ether (60 ml) and anhydrous dichloromethane (60 ml) and Allow the solution to cool to -20 ° C. To the mixture was added dropwise a 1 M lithium aluminum hydride diethyl ether solution (5.5 ml) and stirred at -20 ° C for 20 minutes. The reaction mixture was carefully added with water and then diethyl ether was evaporated under vacuum. The aqueous phase was then extracted with ethyl acetate. The combined organic phases were washed with saturated NaCl solution, dried over anhydrous sodium sulfate and filtered and concentrated under vacuum. Thus, the target product was obtained (0.955 g, 85.4%). MS (ESI) m / z 425.2 (M + H) + .

Example 13
{(5R) -8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [ 4,3-a] [1] benzoazepine-5-yl} methanol

with
Example 14
{(5S) -8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [ 4,3-a] [1] benzoazepine-5-yl} methanol

By chiral preparative HPLC (CHIRALPAK IC (containing preparative 20 μm stationary phase) 2.5 x 20 cm; F = 15 ml / min; eluent: n-hexane: EtOH = 75: 25; isocratic; temperature = 25 ℃), self-racemic {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] Zolo [4,3-a] [1] benzoazin-5-yl} methanol (Example 12) prepared the target product. The first eluting compound (retention time 7.0 minutes) is {(5R) -8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H -[1,2,4] triazolo [4,3-a] [1] benzoazin-5-yl} methanol (Example 13); = + 11.9 ° (c = 0.1; chloroform); the second elutable compound (retention time: 7.8 minutes) is {(5S) -8-chloro-1- [trans-4- (pyridin-2-yloxy) Cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazin-5-yl} methanol (Example 14); = -11.1˚ (c = 0.1; chloroform). By the VCD method and the 1 H NMR spectroscopy determined the absolute configuration of these compounds.

Example 15
8-chloro-5- (methoxymethyl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4 ] Triazolo [4,3-a] [1] benzoazine

Let {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3 -a] [1] Benzacridin-5-yl} methanol (Example 12) (70 mg, 0.16 mmol) was dissolved in anhydrous THF (2 ml) and the solution was then cooled to -5 ° C. A 60% sodium hydride (13 mg, 0.32 mmol) oil dispersion was added and stirred under cooling for 90 minutes, and then methyl iodide (12 μl, 0.2 mmol) was added to the reaction mixture and the mixture was stirred at room temperature. Stir for 2 hours. Subsequently, methyl iodide (12 μl, 0.2 mmol) was added again and the mixture was heated at 40 ° C. for 2 hours. The reaction was diluted with water (30 ml) and then extracted with ethyl acetate. The combined organic phases were washed with a saturated NaCl solution, dried over anhydrous sodium sulfate and filtered and concentrated. The residue was purified by column chromatography using dichloromethane: methanol = 50: 1 as the eluent. Thus, the target product was obtained (20 mg, 28%). MS (ESI) m / z 439.2 (M + H) + .

Example 16
Morpholine-4-carboxylic acid {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazole Benzo [4,3-a] [1] benzoazin-5-yl} methyl ester

a) Methanesulfonic acid {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] Benzacridin-5-yl} methyl ester

Let {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3 -a] [1] Benzacridin-5-yl} methanol (Example 12) (0.855 g, 2.01 mmol) was dissolved in anhydrous dichloromethane (80 ml) and then cooled to 0 ° C. Triethylamine (0.785 ml, 5.63 mmol), methanesulfonyl chloride (0.258 ml, 3.38 mmol) and DMAP (34 mg, 0.28 mmol) were added. The reaction mixture was allowed to stir at 0 ° C for 1 hour and then overnight at room temperature. Pass the reaction mixture over saturated NaHCO 3 The solution was rinsed and the aqueous phase was extracted with dichloromethane. The combined organic phases were washed with a saturated NaCl solution, dried over anhydrous sodium sulfate and filtered and concentrated. Thus, the target product was obtained (1.0 g, 99%). MS (ESI) m / z 503.1 (M + H) + .

b) Morpholine-4-carboxylic acid {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] Triazolo [4,3-a] [1] benzoazin-5-yl} methyl ester
Let methanesulfonic acid {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [ 4,3-a] [1] benzoazino-5-yl} methyl ester (step a) of Example 16) (0.053 g, 0.106 mmol) was dissolved in anhydrous DMF (5 ml). Triethylamine (0.030 ml, 0.212 mmol), cesium carbonate (0.038 g, 0.117 mmol) and morpholine (0.050 ml, 0.573 mmol) were added and the mixture was stirred at 100 ° C for 2 days. The reaction mixture was then cooled, water was added and extracted with ethyl acetate. The combined organic phases were washed with a saturated NaCl solution, dried over anhydrous sodium sulfate and filtered and concentrated. The residue was purified by column chromatography using dichloromethane: methanol = 20: 1 as the eluent. Thus, the target product was obtained (0.013 g, 23.0%). MS (ESI) m / z 538.2 (M + H) + .

Example 17
(4-fluorophenyl) carbamic acid {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4 ] Triazolo [4,3-a] [1] benzoazin-5-yl} methyl ester

Let {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3 -a] [1] Benzacridin-5-yl} methanol (Example 12) (40 mg, 0.09 mmol) was dissolved in anhydrous THF (3 ml), cooled to 5 ° C and introduced with argon Triethylamine (24 μl, 0.17 mmol) was added. A solution of 4-fluorophenyl isocyanate (14 mg, 0.10 mmol) in anhydrous THF (1 ml) was added to the reaction mixture. The reaction did not occur even at reflux temperature and was therefore concentrated. The product was dissolved in acetonitrile (5 ml) and an oil dispersion of 60% sodium hydride (4 mg, 0.09 mmol) was added. The reaction mixture was allowed to stir overnight at room temperature and then concentrated. The residue was purified by column chromatography using dichloromethane: methanol = 20: 1 as the eluent. Thus, the target product was obtained (35 mg, 66%). MS (ESI) m / z 562.2 (M + H) + .

Example 18
5- (azidomethyl) -8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4 ] Triazolo [4,3-a] [1] benzoazine

Methanesulfonic acid {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [ 4,3-a] [1] benzoazin-5-yl} methyl ester (step a) of Example 16) (0.418 g, 0.83 mmol), anhydrous DMF (10 ml), and sodium azide (0.162 g, 2.49 mmol) was filled into a pressure-resistant glass reactor and then stirred at 80 ° C. for 4 hours. The reaction mixture was then added with water and extracted with ethyl acetate. The combined organic phases were washed with a saturated NaCl solution, dried over anhydrous sodium sulfate and filtered and concentrated. The residue was purified by column chromatography using dichloromethane: methanol = 20: 1 as the eluent. Thus, the target product was obtained (0.26 g, 69.5%). MS (ESI) m / z 450.2 (M + H) + .

Example 19
1- {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4, 3-a] [1] benzoazin-5-yl} methylamine

Let 5- (azidomethyl) -8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2, 4] Triazolo [4,3-a] [1] benzoazine (Example 18) (0.242 g, 0.54 mmol) was dissolved in THF (20 ml) and then water (1 ml) and three Phenylphosphine (0.423 g, 1.61 mmol). The reaction mixture was allowed to stir overnight under argon and room temperature, then concentrated and the residue was purified by column chromatography using dichloromethane: methanol: ammonium hydroxide = 10: 1: 0.1 as the eluent. Thus, the target product was obtained (0.183 g, 80%). MS (ESI) m / z 424.2 (M + H) + .

Example 20
8-chloro-5- (morpholin-4-ylmethyl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1, 2,4] triazolo [4,3-a] [1] benzoazine

Methanesulfonic acid {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [ 4,3-a] [1] benzoazine-5-yl} methyl ester (step a) of Example 16) (100 mg, 0.20 mmol), anhydrous DMF (3 ml), DIPEA (0.42 ml , 2.39 mmoles) and morpholine (0.21 ml, 2.39 mmoles) were filled into a pressure-resistant glass reactor and then stirred at 110 ° C. for 1 day. The reaction mixture was then cooled, water was added and extracted with ethyl acetate. The combined organic phases were washed with a saturated NaCl solution, dried over anhydrous sodium sulfate and filtered and concentrated. The residue was purified by column chromatography using dichloromethane: methanol = 50: 1 as the eluent. Thus, the target product was obtained (59 mg, 60%). MS (ESI) m / z 494.2 (M + H) + .

Example 21
1- {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4, 3-a] [1] benzoazepine-5-yl} -N, N-dimethylmethylamine

According to the method described in Example 20, from methanesulfonic acid {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1, 2,4] triazolo [4,3-a] [1] benzoazin-5-yl} methyl ester (step a) of Example 16) and dimethylamine (2M THF solution) to prepare the target product. MS (ESI) m / z 452,2 (M + H) + .

Example 22
N-({8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4 , 3-a] [1] benzoazin-5-yl} methyl) propan-2-amine

Let 1- {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4 , 3-a] [1] Benzacridin-5-yl} methylamine (Example 19) (50 mg, 0.12 mmol) was dissolved in 1,2-dichloroethane (5 ml) and subsequently added Acetic acid (14 µl, 0.25 millimoles), acetone (45 µl, 0.61 millimoles), and sodium triacetoxyborohydride (77 mg, 0.37 millimoles). The reaction mixture was allowed to stir at room temperature for 1 day and then water was added. The pH of the aqueous phase was adjusted to 9 with a 1M NaOH solution and the organic phase was separated. The aqueous phase was extracted with dichloromethane and the combined organic phases were washed with a saturated NaCl solution, dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by column chromatography using dichloromethane: methanol: ammonium hydroxide = 10: 1: 0.1 as the eluent. Thus, the target product was obtained (46 mg, 83.6%). MS (ESI) m / z 466.2 (M + H) + .

Example 23
8-chloro-1- [1- (pyrimidin-2-yl) tetrahydroazepine-3-yl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3- a] [1] Methylbenzoazine-5-carboxylic acid

According to the method described in Example 1, from 1- (Pyrimidin-2-yl) tetrahydroazepine-3-formamidine (Intermediate 13) prepared the target product. MS (ESI) m / z 411.1 (M + H) + .

Example 24
8-chloro-1- [trans-4- (dimethylamino) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [ 1] Methyl benzoazine-5-carboxylate

According to the method described in Example 1, from 7-chloro-2- pendant thio-2,3,4,5-tetrahydro-1H-1-benzoazine-4-carboxylic acid methyl ester (intermediate 4) and The trans-4- (dimethylamino) cyclohexanemethylhydrazine (Intermediate 11) prepared the target product. MS (ESI) m / z 403.2 (M + H) + .

Example 25
8-chloro-1- [trans-4- (trifluoromethyl) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1 ] Methyl benzoazine-5-carboxylate

According to the method described in Example 1, from 7-chloro-2- pendant thio-2,3,4,5-tetrahydro-1H-1-benzoazine-4-carboxylic acid methyl ester (intermediate 4) and Trans-4- (trifluoromethyl) cyclohexanemethanehydrazine (Intermediate 6) prepared the target product. MS (ESI) m / z 428.1 (M + H) + .

Example 26
8-chloro-1- [trans-4- (morpholin-4-yl) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] Methyl benzoazine-5-carboxylate

According to the method described in Example 1, from 7-chloro-2- pendant thio-2,3,4,5-tetrahydro-1H-1-benzoazine-4-carboxylic acid methyl ester (intermediate 4) and The trans-4- (morpholin-4-yl) cyclohexanemethanehydrazine (Intermediate 12) prepared the target product. MS (ESI) m / z 445.2 (M + H) + .

Example 27
{1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1 ] Benzacridin-5-yl} methanol

Let {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3 -a] [1] Benzacridin-5-yl} methanol (Example 12) (63 mg, 0.15 mmol) was dissolved in ethanol (15 ml) and then 10% Pd / C (65 mg) was added To solution and hydrogenation at room temperature. After the reaction was completed, the catalyst was removed by filtration, rinsed with ethanol and the filtrate was concentrated. The residue was purified by column chromatography using dichloromethane: methanol: ammonium hydroxide = 10: 1: 0.1 as the eluent. Thus, the target product was obtained (23 mg, 39%). MS (ESI) m / z 391.2 (M + H) + .

Example 28
8-chloro-1- [trans-4- (pyrrolidin-1-ylcarbonyl) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a ] [1] Methylbenzoazine-5-carboxylic acid

Let 7-chloro-2- pendant thio-2,3,4,5-tetrahydro-1H-1-benzoazine-4-carboxylic acid methyl ester (Intermediate 4) (56.8 mg, 0.21 mmol) A solution of trans-4- (pyrrolidin-1-ylcarbonyl) cyclohexanemethylhydrazine (Intermediate 9) (75.6 mg, 0.32 mmol) and n-butanol (2 ml) was refluxed for 28 hours and The reaction mixture was then concentrated under vacuum. The residue was purified by column chromatography (using dichloromethane: methanol = 9: 1 as eluent). Thus, the target product was obtained (29.6 mg, 31%). MS (ESI) m / z 457.2 (M + H) + .

Example 29
{8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3- a] [1] Benzacridin-5-yl} acetonitrile

Let methanesulfonic acid {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [ 4,3-a] [1] benzoazine-5-yl} methyl ester (step a) of Example 16) (100 mg, 0.20 mmol), anhydrous DMF (10 ml), and sodium cyanide ( 25 mg, 0.52 mmol) in a pressure-resistant glass reactor and stirred at 80 ° C. for 5 hours. Water was then added to the reaction mixture and extracted with ethyl acetate. The combined organic phases were washed with a saturated NaCl solution, dried over anhydrous sodium sulfate and filtered and concentrated. The residue was purified by column chromatography using dichloromethane: methanol = 20: 1 as the eluent. Thus, the target product was obtained (62 mg, 71.8%). MS (ESI) m / z 434.2 (M + H) + .

Example 30
8-chloro-5-[(4-methylpiperazin-1-yl) methyl] -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro -4H- [1,2,4] triazolo [4,3-a] [1] benzoazine

According to the method described in Example 20, from methanesulfonic acid {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1, 2,4] triazolo [4,3-a] [1] benzoazin-5-yl} methyl ester (step a) of Example 16) and 1-methyl-piperazine to prepare the target product. MS (ESI) m / z 507.2 (M + H) + .

Example 31
8-chloro-5- (fluoromethyl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] tris Zolo [4,3-a] [1] benzoazine

Let methanesulfonic acid {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [ 4,3-a] [1] Benzacridin-5-yl} methyl ester (step a) of Example 16) (100 mg, 0.20 mmol) and a 1M solution of tetrabutylammonium fluoride in THF ( 1 ml) was stirred overnight under argon and reflux temperature. The reaction mixture was then added with water and extracted with ethyl acetate. The combined organic phases were washed with a saturated NaCl solution, dried over anhydrous sodium sulfate and filtered and concentrated. The residue was subjected to preparative HPLC (Kinetex EVO C18; 5 μm load; 150 x 21.2 mm; F = 20 ml / min; eluent: A: water; B: acetonitrile, gradient: B% 15 → 70; temperature = 40 ° C). Thus, the target product was obtained (21 mg, 24.7%). MS (ESI) m / z 427.2 (M + H) + .

Example 32
8-chloro-5- (3-methyl-1,2,4-oxadiazol-5-yl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5, 6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazine

Let 8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3- a] [1] Benzoazine-5-carboxylic acid (Example 2) (50 mg, 0.11 mmol), HOBt (21 mg, 0.13 mmol), EDC (26 mg, 0.13 mmol) and A mixture of DMF (1 ml) was stirred under argon and room temperature for 15 minutes and then N-hydroxyacetamidine (10 mg, 0.13 mmol) and additional DMF (1 ml) were added. The reaction mixture was allowed to stir overnight at 100 ° C. The reaction mixture was then cooled to room temperature, water was added and extracted with ethyl acetate. The combined organic phases were washed with a saturated NaCl solution, dried over anhydrous sodium sulfate and filtered and concentrated. The residue was purified by column chromatography using dichloromethane: methanol = 50: 1 as the eluent. Thus, the target product was obtained (29 mg, 53.7%). MS (ESI) m / z 477.2 (M + H) + .

Example 33
8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5- (2H-tetrazol-5-yl) -5,6-dihydro-4H- [1, 2,4] triazolo [4,3-a] [1] benzoazine

Let 8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3- a] [1] Benzoazine-5-carbonitrile (Example 4) (90 mg, 0.21 mmol), DMF (2.5 ml), sodium azide (133 mg, 2.04 mmol) and chloride A mixture of ammonium (111 mg, 2.08 mmol) was placed in a pressure resistant glass reactor and stirred at 115 ° C for 4 hours. The reaction mixture was allowed to cool, water was added and extracted with ethyl acetate. The combined organic phases were washed with a saturated NaCl solution, dried over anhydrous sodium sulfate and filtered and concentrated. The residue was purified by column chromatography (using dichloromethane: methanol = 3: 1 as eluent). Thus, the target product was obtained (54 mg, 54.5%). MS (ESI) m / z 463.2 (M + H) + .

Example 34
N-({8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4 , 3-a] [1] benzoazepine-5-yl} methyl) tetrahydro-2H-piperan-4-amine

According to the method described in Example 22, from 1- {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2 , 4] triazolo [4,3-a] [1] benzoazin-5-yl} methylamine (Example 19) and tetrahydro-4 H -Piperan-4-one to prepare the target product. MS (ESI) m / z 508.2 (M + H) + .

Example 35
N, N-dimethyl-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -8- (trifluoromethyl) -5,6-dihydro-4H- [1, 2,4] triazolo [4,3-a] [1] benzoazepine-5-amine

According to the method described in Example 22, from 1- {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2 , 4] triazolo [4,3-a] [1] benzoazin-5-yl} methylamine (Example 19) and 4,4-difluorocyclohexanone to prepare the target product. MS (ESI) m / z 542.2 (M + H) + .

Example 36
8-Chloro-1- [trans-3-methyl-2-oxo-1-oxa-3-azaspiro [4.5] dec-8-yl] -5,6-dihydro-4H- [1,2,4] Triazolo [4,3-a] [1] benzoazine-5-carboxylic acid methyl ester

According to the method described in Example 1, from 7-chloro-2- pendant thio-2,3,4,5-tetrahydro-1H-1-benzoazine-4-carboxylic acid methyl ester (intermediate 4) and (Trans) -3-methyl-2-oxo-1-oxan-3-azaspiro [4.5] decane-8-formamidine (Intermediate 17) The title product was prepared. MS (ESI) m / z 445.2 (M + H) + .

Example 37
N-({8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4 , 3-a] [1] benzoazepine-5-yl} methyl) acetamide

Let 1- {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4 , 3-a] [1] benzoazin-5-yl} methylamine (Example 19) (45 mg, 0.12 mmol) was dissolved in pyridine (5 ml) and then acetic anhydride (0.10 ml, 1.06 Mol). The reaction mixture was allowed to stir at room temperature for 4 hours and then concentrated. The residue was dissolved in water. The aqueous phase was extracted with dichloromethane and the combined organic phases were washed with a saturated NaCl solution, dried over anhydrous sodium sulfate, filtered and concentrated. Thus, the target product was obtained (44 mg, 89.0%). MS (ESI) m / z 466.2 (M + H) + .

Example 38
N-({8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4 , 3-a] [1] benzoazepine-5-yl} methyl) oxetan-3-amine

According to the method described in Example 22, from 1- {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2 , 4] triazolo [4,3-a] [1] benzoazin-5-yl} methylamine (Example 19) and 3-oxetanone to prepare the target product. MS (ESI) m / z 480.2 (M + H) + .

Example 39
8-chloro-1- [trans-4- (morpholin-4-ylcarbonyl) cyclohexyl] -5,6-dihydro-4 H -[1,2,4] triazolo [4,3- a ] [1] Methylbenzoazine-5-carboxylic acid

According to the method described in Example 28, from the Trans-4- (morpholin-1-ylcarbonyl) cyclohexanemethanehydrazine (Intermediate 10) prepared the target product. MS (ESI) m / z 473.2 (M + H) + .

Example 40
8-chloro-1- [cis-3-methyl-2-oxo-1-oxa-3-azaspiro [4.5] dec-8-yl] -5,6-dihydro-4H- [1,2,4] Triazolo [4,3-a] [1] benzoazine-5-carboxylic acid methyl ester

According to the method described in Example 1, from 7-chloro-2- pendant thio-2,3,4,5-tetrahydro-1H-1-benzoazine-4-carboxylic acid methyl ester (intermediate 4) and (Cis) -3-methyl-2-oxo-1-oxa-3-azaspiro [4.5] decane-8-formamidine (Intermediate 18) The title product was prepared. MS (ESI) m / z 445.2 (M + H) + .

Example 41
8-chloro-1- [1- (pyridin-2-yl) tetrahydroazepine-3-yl] -5,6-dihydro-4 H -[1,2,4] triazolo [4,3- a ] [1] Methylbenzoazine-5-carboxylic acid

According to the method described in Example 28, starting from 7-chloro-2- pendant thio-2,3,4,5-tetrahydro-1H-1-benzoazine-4-carboxylic acid methyl ester (Intermediate 4) and 1- (Pyridin-2-yl) tetrahydroazepine-3-formamidine (Intermediate 14) prepared the target product. MS (ESI) m / z 410.1 (M + H) + .

Example 42
1- [trans-4- (tetrahydroazepine-1-ylcarbonyl) cyclohexyl] -8-chloro-5,6-dihydro-4H- [1,2,4] triazolo [4,3 -a] [1] Benzacne-5-carboxylic acid methyl ester

According to the method described in Example 1, from 7-chloro-2- pendant thio-2,3,4,5-tetrahydro-1H-1-benzoazine-4-carboxylic acid methyl ester (intermediate 4) and Trans-4- (tetrahydroacryl-1-ylcarbonyl) cyclohexanemethanehydrazine (Intermediate 19) prepared the target product. MS (ESI) m / z 443.2 (M + H) + .

Example 43
8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5- (pyrrolidin-1-ylmethyl) -5,6-dihydro-4H- [1, 2,4] triazolo [4,3-a] [1] benzoazine

According to the method described in Example 20, from methanesulfonic acid {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1, 2,4] triazolo [4,3-a] [1] benzoazin-5-yl} methyl ester (step a) of Example 16) and pyrrolidine to prepare the target product. MS (ESI) m / z 478.2 (M + H) + .

Example 44
N-({8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4 , 3-a] [1] benzoazin-5-yl} methyl) -2-methyl-N- (2-methylpropyl) propan-1-amine

According to the method described in Example 22, from 1- {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2 , 4] triazolo [4,3-a] [1] benzoazin-5-yl} methylamine (Example 19) and 2-methylpropanal to prepare the target product. MS (ESI) m / z 536.3 (M + H) + .

Example 45
8-chloro-1- [trans-4- (piperidin-1-ylcarbonyl) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a ] [1] Methylbenzoazine-5-carboxylic acid

According to the method described in Example 28, starting from 7-chloro-2- pendant thio-2,3,4,5-tetrahydro-1H-1-benzoazine-4-carboxylic acid methyl ester (Intermediate 4) and Trans-4- (piperidin-1-ylcarbonyl) cyclohexanemethanehydrazine (Intermediate 20) prepared the target product. MS (ESI) m / z 471.2 (M + H) + .

Example 46
1- (trans-4-{[4- (tertiary butoxycarbonyl) piperazin-1-yl] carbonyl} cyclohexyl) -8-chloro-5,6-dihydro-4H- [1,2, 4] Triazolo [4,3-a] [1] benzoazine-5-carboxylic acid methyl ester

According to the method described in Example 28, starting from 7-chloro-2- pendant thio-2,3,4,5-tetrahydro-1H-1-benzoazine-4-carboxylic acid methyl ester (Intermediate 4) and 4-{[trans-4- (hydrazinocarbonyl) cyclohexyl] carbonyl] piperazine-1-carboxylic acid tert-butyl ester (Intermediate 21) The target product was prepared. MS (ESI) m / z 572.2 (M + H) + .

Example 47
8-chloro-1- [trans-4- (piperazin-1-ylcarbonyl) cyclohexyl] -5,6-dihydro-4 H -[1,2,4] triazolo [4,3- a ] [1] Benzacne-5-carboxylic acid methyl ester hydrochloride

Let 1- (trans-4-{[4- (tertiary butoxycarbonyl) piperazin-1-yl] carbonyl} cyclohexyl) -8-chloro-5,6-dihydro-4H- [1,2 , 4] triazolo [4,3-a] [1] benzoazine-5-carboxylic acid methyl ester (Example 46) (41 mg, 0.07 mmol) was dissolved in ethyl acetate (1 ml) and To the solution was added a 2.5 M solution of hydrochloric acid in ethyl acetate (1 ml). The reaction mixture was allowed to stir at room temperature for 1 hour and then diethyl ether was added. The precipitated product was filtered and washed with diethyl ether. Thus, the target product was obtained (30.5 mg, 84%). MS (ESI) m / z 472.2 (M + H) + .

Example 48
8-methoxy-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3 -a] [1] Benzacne-5-carboxylic acid methyl ester

According to the method described in Example 1, from 7-methoxy-2- pendant thio-2,3,4,5-tetrahydro-1H-1-benzoazine-4-carboxylic acid methyl ester (Intermediate 26 ) Preparation of the target compound. MS (ESI) m / z 449.3 (M + H) + .

Example 49
8-methyl-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3- a] [1] Methylbenzoazine-5-carboxylic acid

According to the method described in Example 1, from 7-methyl-2- pendant thio-2,3,4,5-tetrahydro-1H-1-benzoazine-4-carboxylic acid methyl ester (Intermediate 25) Prepare the target compound. MS (ESI) m / z 433.2 (M + H) + .

Example 50
8-chloro-N- (4-fluorophenyl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4 ] Triazolo [4,3-a] [1] benzoazine-5-carboxamide

P-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3- a] [1] Benzoazine-5-carboxylic acid (Example 2) (100 mg, 0.23 mmol), DMF (15 ml), HOBt (84 mg, 0.55 mmol), DIPEA (151 µl, 0.87 mmoles) and EDC (105 mg, 0.55 mmoles) in an ice-cold mixture was added 4-fluoroaniline (25 mg, 0.23 mmoles) and the reaction mixture was stirred at 5 ° C for 0.5 hours and then at room temperature After stirring for another 20 hours. After completion of the reaction, the mixture was diluted with water and brine and extracted with ethyl acetate. The combined organic layers were dried over sodium sulfate and filtered and concentrated. The residue was purified by flash column chromatography (using dichloromethane: methanol = 95: 5 as the eluent) to give the target compound (105 mg, 87%). MS (ESI) m / z 532.1 (M + H) + .

Example 51
8-bromo-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a ] [1] Methylbenzoazine-5-carboxylic acid

Prepared according to the method described in Example 1 from 7-bromo-2- pendant thio-2,3,4,5-tetrahydro-1H-1-benzoazine-4-carboxylic acid methyl ester (Intermediate 27) Underlying compound. MS (ESI) m / z 499.1 (M + H) + .

Example 52
1- (1,4'-bipiperidin-1'-yl) -8-chloro-5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1 ] Benzacridin-5-carboxamide

Let 1-bromo-8-chloro-5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazepine-5-carboxamide (middle Body 29) (34 mg, 0.1 mmol), cyclobutane (0.3 ml) and 4-piperidinepiperidine (218 mg, 1.3 mmol) were stirred at 180 ° C for 3 hours and then cooled To room temperature. The reaction mixture was diluted with dichloromethane and washed with brine, and the organic phase was dried over sodium sulfate and filtered and concentrated. The residue was purified by flash column chromatography (using dichloromethane: methanol: ammonium hydroxide = 9: 1: 0.1 as the eluent) to give the target compound (13 mg, 30%). MS (ESI) m / z 429.2 (M + H) + .

Example 53
8-chloro-1-[(5r, 8r) -1-oxo-2- (prop-2-yl) -2-azaspiro [4.5] dec-8-yl] -5,6-dihydro -4H- [1,2,4] triazolo [4,3-a] [1] benzoazine-5-carboxylic acid methyl ester

According to the method described in Example 28, from the (5r, 8r) -1-Pentoxy-2- (prop-2-yl) -2-azaspiro [4.5] decane-8-formamidine (Intermediate 32) The title compound was prepared. MS (ESI) m / z 471.2 (M + H) + .

Example 54
1- {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4, 3-a] [1] benzoazepine-5-yl} ethanol

a) 5- (1-{[Tri-butyl (dimethyl) silyl] oxy} ethyl) -8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl ] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazine

According to the method described in Example 1, from 4- (1-{[tertiary butyl (dimethyl) silyl] oxy} ethyl) -7-chloro-1,3,4,5-tetrahydro-2H -1-Benzazine-2-thione (Intermediate 34) prepared the target compound. The product was a 55:45 mixture of non-mirromeric isomers. MS (ESI) m / z 553.2 (M + H) + .

b) 1- {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [ 4,3-a] [1] benzoazepine-5-yl} ethanol
Let 5- (1-{[tertiary butyl (dimethyl) silyl] oxy} ethyl) -8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazine (step a) of Example 54) (222 mg, 0.4 mmol Ear) and anhydrous THF (40 ml) was cooled to 0 ° C and 1M tetrabutylammonium fluoride (0.8 ml, 0.8 mmol) in THF was added and the reaction mixture was stirred at room temperature for 20 hours and It was then concentrated. Water was added to the residue and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate and filtered and concentrated. The residue was purified by flash column chromatography (using dichloromethane: methanol = 9: 1 as the eluent) to give the target compound (172 mg, 98%). The product is a 54:46 mixture of non-mirromeric isomers. MS (ESI) m / z 439.1 (M + H) + .

Example 55
1- {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4, 3-a] [1] benzoazin-5-yl} ethanone

Let 1- {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4 , 3-a] [1] Benzacridin-5-yl} ethanol (Example 54) (142 mg, 0.32 mmol) and dichloromethane (10 ml) were stirred and cooled to 0 ° C and added to Stewart-Martin iodine (206 mg, 0.48 mmol). The reaction mixture was allowed to stir at room temperature for 3 hours and then filtered. The filtrate was diluted with water, the layers were separated and the aqueous phase was extracted with dichloromethane. The combined organic layers were washed with brine, dried over sodium sulfate and filtered and concentrated. The residue was purified by flash column chromatography (using dichloromethane: methanol = 95: 5 as the eluent) to give the target compound (111 mg, 79%). MS (ESI) m / z 437.2 (M + H) + .

Example 56
8-chloro-5- (fluoromethyl) -1- (trans-4-methoxy-4-methylcyclohexyl) -5,6-dihydro-4H- [1,2,4] triazole Ac [4,3-a] [1] benzoacene

According to the method described in Example 1, from 7-chloro-4- (fluoromethyl) -1,3,4,5-tetrahydro-2H-1-benzoazine-2-thione (Intermediate 37) And 4-methoxy-4-methylcyclohexanemethanehydrazine (Intermediate 35, step c)) to prepare the target compound. MS (ESI) m / z 378.2 (M + H) + .

Example 57
1- {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4, 3-a] [1] benzoazin-5-yl} -N- (2-methoxyethyl) ethylamine

Let 1- {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4 , 3-a] [1] benzoazin-5-yl} ethanone (Example 55) (30 mg, 0.07 mmol), 1,2-dichloromethane (5 ml), 2-methoxy Ethylamine (24 µl, 0.275 mmol), acetic acid (30 µl), and NaBH (OAc) 3 (29 mg, 0.137 mmol) was stirred at room temperature for 4 hours and then concentrated. The pH of the residue was adjusted to 9 by adding 1M NaOH and the mixture was then extracted with dichloromethane. The combined organic layers were washed with brine, dried over sodium sulfate and filtered and concentrated. The residue was purified by flash column chromatography (using dichloromethane: methanol = 97: 3 as the eluent) to give the target compound (12 mg, 35%). The product was a 58:42 mixture of non-mirromeric isomers. MS (ESI) m / z 496.3 (M + H) + .

Example 58
8-fluoro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a ] [1] Methylbenzoazine-5-carboxylic acid

According to the method described in Example 1, from 7-fluoro-2- pendant thio-2,3,4,5-tetrahydro-1 H Methyl-1-benzoazine-4-carboxylic acid (Intermediate 39) prepared the target compound. MS (ESI) m / z 437.4 (M + H) + .

Example 59
8-chloro-5- (fluoromethyl) -1- [trans-4- (trifluoromethyl) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [ 4,3-a] [1] benzoazine

According to the method described in Example 1, from 7-chloro-4- (fluoromethyl) -1,3,4,5-tetrahydro-2H-1-benzoazine-2-thione (Intermediate 37) And trans-4- (trifluoromethyl) cyclohexanemethanehydrazine (Intermediate 6) to prepare the target compound. MS (ESI) m / z 402.3 (M + H) + .

Example 60
5- (fluoromethyl) -1- (trans-4-methoxy-4-methylcyclohexyl) -5,6-dihydro-4H- [1,2,4] triazolo [4, 3-a] [1] benzoazine

According to the method described in Example 27, from 8-chloro-5- (fluoromethyl) -1- (trans-4-methoxy-4-methylcyclohexyl) -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazine (Example 56) The target compound was prepared. MS (ESI) m / z 344.3 (M + H) + .

Example 61
{8-fluoro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3- a] [1] Benzacridin-5-yl} methanol

According to the method described in Example 12, from 8-fluoro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] Triazolo [4,3-a] [1] benzoazine-5-carboxylic acid methyl ester (Example 58) prepared the target compound. MS (ESI) m / z 409.3 (M + H) + .

Example 62
8-fluoro-5- (fluoromethyl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] tris Zolo [4,3-a] [1] benzoazine

According to the method described in Example 1, from 7-fluoro-4- (fluoromethyl) -1,3,4,5-tetrahydro-2H-1-benzoazine-2-thione (intermediate 41) Prepare the target compound. MS (ESI) m / z 411.3 (M + H) + .

Example 63
Cis- (racemic) -1- {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2, 4] triazolo [4,3-a] [1] benzoazin-5-yl} ethanol

with
Example 64
Trans- (racemic) -1- {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2, 4] triazolo [4,3-a] [1] benzoazin-5-yl} ethanol

a) 5- [cis- (Meth) -1-{[tertiary butyl (dimethyl) silyl] oxy} ethyl] -8-chloro-1- [trans-4- (pyridine- 2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazine and 5- [trans- (Racemic) -1-{[tertiary butyl (dimethyl) silyl] oxy} ethyl] -8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl ] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazine

According to the method described in Example 1, from 4- (1-{[tertiary butyl (dimethyl) silyl] oxy} ethyl) -7-chloro-1,3,4,5-tetrahydro-2H The isolated single non-mirror isomer of 1-benzoazepin-2-thione (Intermediate 34) prepared the target compound. The products are single non-mirromeric isomers but their configuration has not been determined. MS (ESI) m / z 553.3 (M + H) + .

b) cis- (racemic) -1- {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1, 2,4] triazolo [4,3-a] [1] benzoazin-5-yl} ethanol and trans- (racemic) -1- {8-chloro-1- [trans-4 -(Pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazepine-5- Methyl} ethanol
According to the method described in step b) of Example 54, from 5- [cis- (racemic) -1-{[tertiary butyl (dimethyl) silyl] oxy} ethyl] -8-chloro- 1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] Benzacridine and 5- [trans- (racemic) -1-{[tertiary butyl (dimethyl) silyl] oxy} ethyl] -8-chloro-1- [trans-4- (Pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazine (Example 63 And step a)) of 64 to prepare the target compound. The products are single non-mirromeric isomers but their configuration has not been determined. MS (ESI) m / z 439.4 (M + H) + .

Example 65
(1R) -1-{(5R) -8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2, 4] triazolo [4,3-a] [1] benzoazin-5-yl} ethanol

with
Example 66
(1S) -1-{(5S) -8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2, 4] triazolo [4,3-a] [1] benzoazin-5-yl} ethanol

with
Example 67
(1S) -1-{(5R) -8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2, 4] triazolo [4,3-a] [1] benzoazin-5-yl} ethanol

with
Example 68
(1R) -1-{(5S) -8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2, 4] triazolo [4,3-a] [1] benzoazin-5-yl} ethanol

By chiral preparative HPLC (CHIRALPAK IG preparative 20 µm stationary phase; 5 x 30 cm; F = 50 ml / min; eluent: n-heptane: isopropanol = 8: 2; isocratic; temperature = 25 ° C), from the isolated non-image isomer: cis- (racemic) -1- {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl]- 5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazin-5-yl} ethanol and trans- (racemic) -1- {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3- a] [1] Benzacridin-5-yl} ethanol (Examples 63 and 64) gave the target compound. Separation of one of the racemic mixtures produces 2 compounds: the first elutable compound (T r 13.9 minutes) = + 14˚ (c = 0.1; chloroform) and the second eluting compound (T r 16.7 minutes) = -12˚ (c = 0.1; chloroform). Separation of the other of the racemic mixtures yields another 2 compounds: the first elutable compound (T r 19.0 minutes) = + 21˚ (c = 0.1; chloroform) and the second eluting compound (T r 21.5 minutes) = -16˚ (c = 0.1; chloroform). The absolute configuration of these compounds has not been determined.

Example 69
5- (fluoromethyl) -8-methyl-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] Triazolo [4,3-a] [1] benzoazine

According to the method described in Example 1, from 4- (fluoromethyl) -7-methyl-1,3,4,5-tetrahydro-2H-1-benzoazine-2-thione (Intermediate 43 ) Preparation of the target compound. MS (ESI) m / z 407.3 (M + H) + .

Example 70
(5S) -8-chloro-5- (fluoromethyl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2 , 4] triazolo [4,3-a] [1] benzoazine

with
Example 71
(5R) -8-chloro-5- (fluoromethyl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2 , 4] triazolo [4,3-a] [1] benzoazine

By chiral preparative HPLC (CHIRALPAK IG preparative 20 µm stationary phase; 5 x 30 cm; F = 50 ml / min; eluent: tertiary butyl methyl ether: methylene chloride: EtOH = 90: 8: 2 Isocratic; temperature = 25 ℃), self-racemic 8-chloro-5- (fluoromethyl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6 -Dihydro-4H- [1,2,4] triazolo [4,3- a ] [1] Benzoazine (Example 31) gave the target compound. The first eluted compound (retention time 20.2 minutes) is (5S) -8-chloro-5- (fluoromethyl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5 , 6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazine (Example 70), = + 6.3 ° (c = 0.1; dichloromethane); and the second elutable compound (retention time 22.9 minutes) is (5R) -8-chloro-5- (fluoromethyl) -1- [trans- 4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazepine (implementation Example 71), = -8.9 ° (c = 0.1; dichloromethane). The configuration of these compounds was determined by the VCD method.

Example 72
8-fluoro-1- [trans-4- (trifluoromethyl) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1 ] Methyl benzoazine-5-carboxylate

Let 7-fluoro-2- (methylthio) -4,5-dihydro-3H-1-benzoazine-4-carboxylic acid methyl ester (Intermediate 44) (270 mg, 0.1 mmol), 1 , 4-dioxane (20 ml), trans-4- (trifluoromethyl) cyclohexanemethanehydrazine (Intermediate 6) (210 mg, 0.1 mmol) and concentrated hydrochloric acid (10 µl The mixture) was refluxed for 1 hour and then concentrated. The residue was purified by flash column chromatography (using dichloromethane: methanol = 95: 5 as the eluent) to give the target compound (145 mg, 35%). MS (ESI) m / z 412.1 (M + H) + .

Example 73
8-chloro-1- [4- (2,3-dimethylphenyl) piperazin-1-yl] -5,6-dihydro-4H- [1,2,4] triazolo [4, 3-a] [1] Benzacene-5-carboxamide

According to the method described in Example 52, 5-Carboxamidamine (Intermediate 29) and 1- (2,3-dimethylphenyl) piperazine were used to prepare the target compounds. MS (ESI) m / z 451.2 (M + H) + .

Example 74
{8-fluoro-1- [trans-4- (trifluoromethyl) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [ 1] Benzacridin-5-yl} methanol

According to the method described in Example 12, from 8-fluoro-1- [trans-4- (trifluoromethyl) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] Benzacne-5-carboxylic acid methyl ester (Example 72) The target compound was prepared. MS (ESI) m / z 384.2 (M + H) + .

Example 75
{8-chloro-5-methyl-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazepine-5-yl} methanol

a) 8-Chloro-5-methyl-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazole Benzo [4,3-a] [1] benzoazine-5-carboxylic acid methyl ester

According to the method described in Example 1, from 7-chloro-4-methyl-2- pendant thio-2,3,4,5-tetrahydro-1H-1-benzoazine-4-carboxylic acid methyl ester ( Intermediate 46) The target compound was prepared. MS (ESI) m / z 467.1 (M + H) + .

b) {8-chloro-5-methyl-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] tri Zolo [4,3-a] [1] benzoazepine-5-yl} methanol
According to the method described in Example 12, from 8-chloro-5-methyl-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1 , 2,4] triazolo [4,3-a] [1] benzoazine-5-carboxylic acid methyl ester (step a) of Example 75) to prepare the target compound. MS (ESI) m / z 439.1 (M + H) + .

Example 76
{8-chloro-5-ethyl-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazepine-5-yl} methanol

a) 8-Chloro-5-ethyl-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazole Benzo [4,3-a] [1] benzoazine-5-carboxylic acid methyl ester

According to the method described in Example 72, from 7-chloro-4-ethyl-2- (methylthio) -4,5-dihydro-3H-1-benzoazine-4-carboxylic acid methyl ester (intermediate 49) Prepare the target compound. MS (ESI) m / z 481.2 (M + H) + .

b) {8-chloro-5-ethyl-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] tri Zolo [4,3-a] [1] benzoazepine-5-yl} methanol
According to the method described in Example 12, from 8-chloro-5-ethyl-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1 , 2,4] triazolo [4,3-a] [1] benzoazine-5-carboxylic acid methyl ester (step a) of Example 76) to prepare the target compound. MS (ESI) m / z 453.2 (M + H) + .

Example 77
(5S) -8-fluoro-5- (fluoromethyl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2 , 4] triazolo [4,3-a] [1] benzoazine

with
Example 78
(5R) -8-fluoro-5- (fluoromethyl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2 , 4] triazolo [4,3-a] [1] benzoazine

By chiral preparative HPLC (CHIRALPAK IA preparative 20 µm stationary phase; 2.5 x 20 cm; F = 15 ml / min; eluent: tertiary butyl methyl ether: dichloromethane = 75: 25; isocratic; Temperature = 25 ℃), self-racemic 8-fluoro-5- (fluoromethyl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro- 4H- [1,2,4] triazolo [4,3-a] [1] benzoazine (Example 62) gave the target compound. First eluting compound (T r 16.6 minutes) = + 9 ° (c = 0.1; dichloromethane) and the second eluting compound (T r 19.0 minutes) = -10 ° (c = 0.1; dichloromethane). The absolute configuration of these compounds has not been determined.

Example 79
8-chloro-1- [4- (3-chlorophenyl) piperazin-1-yl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] Benzacene-5-carboxamide

According to the method described in Example 52, from 1-bromo-8-chloro-5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazine 5-Carboxamidamine (Intermediate 29) and 1- (3-chlorophenyl) piperazine were used to prepare the target compounds. MS (ESI) m / z 457.2 (M + H) + .

Example 80
8-chloro-1- [4- (pyridin-2-yl) piperazin-1-yl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] Benzacene-5-carboxamide

According to the method described in Example 52, from 1-bromo-8-chloro-5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazine 5-Carboxamidamine (Intermediate 29) and 1- (pyridin-2-yl) piperazine prepared the target compounds. MS (ESI) m / z 424.2 (M + H) + .

Example 81
8-chloro-1- [4- (pyridin-2-yloxy) piperidin-1-yl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3- a] [1] Benzacene-5-carboxamide

According to the method described in Example 52, from 1-bromo-8-chloro-5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazine 5-Carboxamidamine (Intermediate 29) and 2- (piperidin-4-yloxy) pyridine prepared the target compounds. MS (ESI) m / z 439.2 (M + H) + .

Example 82
5-[(benzyloxy) methyl] -8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2 , 4] triazolo [4,3-a] [1] benzoazine

Let {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3 -a] [1] Benzacridin-5-yl} methanol (Example 12) (100 mg, 0.24 mmol) in anhydrous DMF (3 ml), the mixture was cooled to 0 ° C and added under argon to 60 % Sodium hydride (24 mg, 0.59 mmol) in mineral oil. The mixture was allowed to stir at this temperature for 0.5 hours, then benzyl bromide (70 µl, 0.59 mmol) and tetrabutylammonium iodide (27 mg, 0.07 mmol) were added and the reaction mixture was allowed to pass at room temperature. Stir for 3 hours. After the reaction was completed, the mixture was diluted with saturated ammonium chloride solution, extracted with ethyl acetate, and the combined organic layer was washed with brine, dried over sodium sulfate, filtered and concentrated. The residue was purified by flash column chromatography (using dichloromethane: methanol = 95: 5 as the eluent) to give the target compound (44 mg, 36%). MS (ESI) m / z 515.2 (M + H) + .

Example 83
8-chloro-1- (3,3-difluorocyclobutyl) -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazine甲酯 -5-carboxylic acid methyl ester

According to the method described in Example 1, from 7-chloro-2- pendant thio-2,3,4,5-tetrahydro-1H-1-benzoazine-4-carboxylic acid methyl ester (intermediate 4) and 3,3-Difluorocyclobutanemethanehydrazine (Intermediate 51) prepared the target compound. MS (ESI) m / z 368.1 (M + H) + .

Example 84
8-chloro-1- [trans-4- (piperidin-1-ylmethyl) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3- a] [1] Methylbenzoazine-5-carboxylic acid

According to the method described in Example 1, from 7-chloro-2- pendant thio-2,3,4,5-tetrahydro-1H-1-benzoazine-4-carboxylic acid methyl ester (intermediate 4) and Trans-4- (piperidin-1-ylmethyl) cyclohexanemethylhydrazine (Intermediate 8) prepared the title compound. MS (ESI) m / z 457.3 (M + H) + .

Example 85
{8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3- a] [1] Benzacridin-5-yl} methyl acetate

Let {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3 -a] [1] benzoazepine-5-yl} methanol (Example 12) (100 mg, 0.24 mmol), dichloromethane (5 ml), pyridine (57 µl, 0.7 mmol), A mixture of acetic anhydride (67 µl, 0.7 mmol) and DMAP (3 mg, 0.02 mmol) was stirred at room temperature for 20 hours, then diluted with water and extracted with dichloromethane. The combined organic layers were washed with brine, dried over sodium sulfate and filtered and concentrated. The residue was purified by flash column chromatography (using dichloromethane: methanol = 97: 3 as the eluent) to give the target compound (38 mg, 35%). MS (ESI) m / z 467.1 (M + H) + .

Example 86
8-chloro-1- (1'H, 3H-spiro [2-benzofuran-1,4'-piperidine] -1'-yl) -5,6-dihydro-4H- [1,2, 4] triazolo [4,3-a] [1] benzoazine-5-carboxamide

According to the method described in Example 52, from 1-bromo-8-chloro-5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazine 5-Carboxamidamine (Intermediate 29) and 3H-spiro [2-benzofuran-1,4'-piperidine] (Combi-Blocks) prepared the target compound. MS (ESI) m / z 450.2 (M + H) + .

Example 87
8-chloro-N- (pyridin-2-yl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4 ] Triazolo [4,3-a] [1] benzoazine-5-carboxamide

P-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3- a] [1] A stirred solution of benzoazine-5-carboxylic acid (Example 2) (361 mg, 0.82 mmol) in dichloromethane (25 ml) was added chloramphenicol (446 µl, 5.3 mmol) And DMF (50 µl). The reaction mixture was allowed to stir at room temperature for 30 minutes and then concentrated. The residue was dissolved in pyridine (5 ml) and 2-aminopyridine (155 mg, 1.64 mmol) was added. The resulting reaction mixture was stirred at room temperature for 2 hours, then concentrated and the residue was subjected to preparative HPLC (KINETEX EVO 5 µm stationary phase; 150 x 21.2 mm; F = 20 ml / min; eluate: ( Water + 0.1% trifluoroacetic acid): (methanol: acetonitrile = 2: 1) = 52: 48; isocratic; temperature = 40 ° C) was purified to give the target compound (12.7 mg, 3%). MS (ESI) m / z 515.2 (M + H) + .

Example 88
8-chloro-1- (1'H, 3H-spiro [2-benzofuran-1,4'-piperidine] -1'-yl) -5,6-dihydro-4H- [1,2, 4] triazolo [4,3-a] [1] benzoazine-5-carbonitrile

Let 8-chloro-1- (1'H, 3H-spiro [2-benzofuran-1,4'-piperidine] -1'-yl) -5,6-dihydro-4H- [1,2 , 4] triazolo [4,3-a] [1] benzoazepine-5-carboxamide (Example 86) (78.6 mg, 0.175 mmol), anhydrous dichloromethane (30 ml), and A stirred solution of triethylamine (153 µl, 1.1 mmol) was cooled to 0 ° C and trifluoroacetic anhydride (40 µl, 0.28 mmol) was added. The mixture was allowed to warm to room temperature and the reaction mixture was stirred at this temperature for 1 hour. Triethylamine (153 µl, 1.1 mmol) and trifluoroacetic anhydride (40 µl, 0.28 mmol) were then added. . The mixture was allowed to stir overnight at room temperature, then triethylamine (153 µl, 1.1 mmol) and trifluoroacetic anhydride (80 µl, 0.56 mmol) were added and stirring was continued overnight at room temperature. Pass the reaction mixture over saturated NaHCO 3 The solution was diluted, the phases were separated and the aqueous phase was extracted with dichloromethane. The combined organic phases were washed with water and brine, dried over sodium sulfate and filtered and concentrated. The residue was purified by column chromatography (using dichloromethane: methanol: ammonium hydroxide = 180: 10: 1 as the eluent) to give the target compound (11.5 mg, 15%). MS (ESI) m / z 432.1 (M + H) + .

Example 89
8-chloro-1- [4- (2,3-dimethylphenyl) piperazin-1-yl] -5,6-dihydro-4H- [1,2,4] triazolo [4, 3-a] [1] benzoazine-5-carbonitrile

Let 1-bromo-8-chloro-5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazine-5-carbonitrile (Intermediate 52 ) (100 mg, 0.31 mmol) and 1- (2,3-dimethylphenyl) piperazine (412 mg, 2.16 mmol) under argon and 180 ° C with stirring for 4 hours and then Cool to room temperature. The residue was purified by column chromatography (using dichloromethane: methanol: ammonium hydroxide = 180: 10: 1 as eluent) and crystallized from diethyl ether to give the title compound (28 mg, 21%). MS (ESI) m / z 433.1 (M + H) + .

Example 90
8-chloro-1- [4- (pyridin-2-yl) piperazin-1-yl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] Benazine-5-nitrile

According to the method described in Example 89, from 1-bromo-8-chloro-5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazine -5-Nitrile (Intermediate 52) and 1- (pyridin-2-yl) piperazine prepared the target compounds. MS (ESI) m / z 406.1 (M + H) + .

Example 91
8-fluoro-5,5-dimethyl-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] tris Zolo [4,3-a] [1] benzoazine

a) 7-Fluoro-4,4-dimethyl-1,3,4,5-tetrahydro-2H-1-benzoazine-2-thione

P-Fluoro-4,4-dimethyl-1,3,4,5-tetrahydro-2H-1-benzoazepine-2-one (Intermediate 53) (1.10 g, 5.31 mmol) And a solution of pyridine (10 ml) was added to Lawson's reagent (2.79 g, 6.90 mmol) and the reaction mixture was refluxed for 2 hours. The solvent was then removed in vacuo and the residue was subjected to saturated NaHCO 3 Triturate and collect the precipitate by filtration. Thus, the target product (1.00 g, 84%) was obtained, which was used without further purification.

b) 7-fluoro-4,4-dimethyl-2- (methylthio) -4,5-dihydro-3H-1-benzoazepine

P-Fluoro-4,4-dimethyl-1,3,4,5-tetrahydro-2H-1-benzoazepine-2-thione (step a) of Example 91) (0.40 g, 1.68 mmol, potassium carbonate (6.60 g, 4.78 mmol) and acetone (30 ml) was added to methyl iodide (0.297 ml, 4.78 mmol) and the reaction mixture was stirred at 40 ° C for 1 hour. After evaporation of the solvent, water and ethyl acetate were added to the residue, the layers were separated and the aqueous phase was extracted with ethyl acetate. The combined organic phases were washed with a saturated NaCl solution, dried over anhydrous sodium sulfate and filtered and concentrated. Thus, the target product (0.40 g, 99%) was obtained, which was used without further purification.

c) 8-fluoro-5,5-dimethyl-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4 ] Triazolo [4,3-a] [1] benzoazine
7-fluoro-4,4-dimethyl-2- (methylthio) -4,5-dihydro-3H-1-benzoazine (step b) of Example 91) (0.30 g, 1.26 Mol), trans-4- (pyridin-2-yloxy) cyclohexanemethylhydrazine (WO2010 / 060836 A1 (2010.06.03) F. HOFFMANN-LA ROCHE AG.) A mixture of mol) and 1,4-dioxane (20 ml) was added with concentrated hydrochloric acid (0.010 ml) and the reaction mixture was refluxed for 3 hours. After the solvent was evaporated, the residue was purified by column chromatography using dichloromethane: methanol = 20: 1 as the eluent. Thus, the target product was obtained (0.108 mg, 21%). MS (ESI) m / z 407.2 (M + H) + .

Example 92
8-fluoro-5- (prop-2-yl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4 ] Triazolo [4,3-a] [1] benzoazine

According to the method described in Example 91, starting from 7-fluoro-4- (prop-2-yl) -1,3,4,5-tetrahydro-2H-1-benzoazin-2-one (Intermediate 54 ) Preparation of the target product. MS (ESI) m / z 421.2 (M + H) + .

Example 93
8'-bromo-1 '-[trans-4- (pyridin-2-yloxy) cyclohexyl] -4'H, 6'H-spiro [cyclopentane-1,5'-[1,2 , 4] triazolo [4,3-a] [1] benzoazine]

Prepared from 7-bromo-1,5-dihydrospiro [1-benzoazine-4,1'-cyclopentane] -2 (3H) -one (intermediate 60) according to the method described in Example 91 Underlying products. MS (ESI) m / z 493.1 and 495.1 (M + H) + .

Example 94
8-bromo-5- (prop-2-yl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4 ] Triazolo [4,3-a] [1] benzoazine

According to the method described in Example 91, from 7-bromo-4- (prop-2-yl) -1,3,4,5-tetrahydro-2H-1-benzoazin-2-one (Intermediate 61 ) Preparation of the target product. MS (ESI) m / z 481.1 and 483.1 (M + H) + .

Example 95
8'-bromo-1 '-[trans-4- (pyridin-2-yloxy) cyclohexyl] -2,3,5,6-tetrahydro-4'H, 6'H-spiro [piperan -4,5 '-[1,2,4] triazolo [4,3-a] [1] benzoazepine]

a) 7-bromo-2-methoxy-3,5-dihydrospiro [1-benzoazine-4,4'-ethylene oxide]

Let 7-bromo-1,5-dihydrospiro [1-benzoazine-4,4'-ethylene oxide] -2 (3 H ) -Ketone (Intermediate 62) (0.193 g, 062 mmol), trimethyloxonium tetrafluoroborate (0.183 g, 1.24 mmol), potassium carbonate (0.343 g, 2.48 mmol) and dichloride A mixture of methane (30 ml) was stirred at room temperature for 3 hours. The reaction mixture was then filtered and the filtrate was concentrated. Thus, the target product was obtained (0.190 g, 98%), which was used without further purification.

b) 8'-bromo-1 '-[trans-4- (pyridin-2-yloxy) cyclohexyl] -2,3,5,6-tetrahydro-4'H, 6'H-spiro [ Piperan-4,5 '-[1,2,4] triazolo [4,3-a] [1] benzoazepine]
7-bromo-2-methoxy-3,5-dihydrospiro [1-benzoazine-4,4'-ethylene oxide] (0.190 g, 0.62 mmol), trans-4 -(Pyridin-2-yloxy) cyclohexanemethylhydrazine (WO2010 / 060836 A1 (03.06.2010) F. HOFFMANN-LA ROCHE AG.) (0.161 g, 0.68 mmol) and 1,4-bis A mixture of oxane (20 ml) was added with trifluoroacetic acid (0.010 ml) and the reaction mixture was refluxed for 3 hours. After the solvent was evaporated, the residue was purified by column chromatography using dichloromethane: methanol = 20: 1 as the eluent. Thus, the target product was obtained (0.144 mg, 45%). MS (ESI) m / z 509.2 and 511.2 (M + H) + .

Example 96
8-fluoro-5,5-dimethyl-1- [trans-4- (trifluoromethyl) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [ 4,3-a] [1] benzoazine

According to the method described in step c) of Example 91, from 7-fluoro-4,4-dimethyl-2- (methylthio) -4,5-dihydro-3H-1-benzoazine (implemented Step b)) of Example 91 and trans-4- (trifluoromethyl) cyclohexanemethanehydrazine (Intermediate 6) prepared the target product. MS (ESI) m / z 382.2 (M + H) + .

Example 97
(5R) -8-bromo-5- (prop-2-yl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1 , 2,4] triazolo [4,3-a] [1] benzoazine

with
Example 98
(5S) -8-bromo-5- (prop-2-yl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1 , 2,4] triazolo [4,3-a] [1] benzoazine

By chiral preparative HPLC (CHIRALPAK IC preparative 20 μm stationary phase; 2.5 x 20 cm; F = 20 ml / min; eluent: tertiary butyl methyl ether: dichloromethane: ethanol = 85: 13: 2 Isocratic; temperature = 25 ℃), self-racemic 8-bromo-5- (prop-2-yl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5 , 6-Dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazine (Example 94) The target product was prepared. By analytical HPLC (CHIRALPAK IA 5 μm stationary phase; 250 x 4.6 mm; F = 1 ml / min; eluent: tertiary butyl methyl ether: dichloromethane: ethanol = 85: 10: 5; isocratic; Temperature = 35 ° C) Determination of retention time: 15.0 minutes for the first eluate; = -5.0˚ (c = 0.1; dichloromethane); MS (ESI) m / z 481.2 and 483.2 (M + H) + ; The second eluate is 18.5 minutes; = + 10.0˚ (c = 0.1; dichloromethane); MS (ESI) m / z 481.2 and 483.2 (M + H) + . The absolute configuration of these compounds has not been determined.

Example 99
8'-bromo-1 '-[trans-4- (pyridin-2-yloxy) cyclohexyl] -4,5-dihydro-4'H, 6'H-spiro [furan-3,5' -[1,2,4] triazolo [4,3-a] [1] benzoazine]

The titled product was prepared from 7-bromo-1,5-dihydrospiro [1-benzoazine-4,3'-tetrahydrofuran] -2 (3H) -one (intermediate 63) according to the method described in Example 95 . MS (ESI) m / z 495.1 and 497.2 (M + H) + .

Example 100
8'-chloro-1 '-[trans-4- (pyridin-2-yloxy) cyclohexyl] -2,3,5,6-tetrahydro-4'H, 6'H-spiro [piperan -4,5 '-[1,2,4] triazolo [4,3-a] [1] benzoazepine]

Following the method described in Example 95, Prepare the target product. MS (ESI) m / z 465.2 (M + H) + .

Example 101
8'-chloro-1 '-[trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H, 4'H, 6'H-spiro [piperan-3 , 5 '-[1,2,4] triazolo [4,3-a] [1] benzoazepine]

According to the method described in Example 95, from 7-Chloro-1,5-dihydrospiro [1-benzoazine-4,3'-oxirane] -2 (3H) -one (Intermediate 65) Prepare the target product. MS (ESI) m / z 465.3 (M + H) + .
Example 102
8'-Chloro-1 '-[trans-4- (pyridin-2-yloxy) cyclohexyl] -4,5-dihydro-4'H, 6'H-spiro [furan-3,5' -[1,2,4] triazolo [4,3-a] [1] benzoazine]

The title product was prepared according to the method described in Example 95 from 7-chloro-1,5-dihydrospiro [1-benzoazine-4,3'-tetrahydrofuran] -2 (3H) -one (intermediate 66) . MS (ESI) m / z 451.2 (M + H) + .
Example 103
8'-chloro-1 '-[trans-4- (trifluoromethyl) cyclohexyl] -2,3,5,6-tetrahydro-4'H, 6'H-spiro [piperan-4, 5 '-[1,2,4] triazolo [4,3-a] [1] benzoazine]

According to the method described in Example 91, from 7-Chloro-1,5-dihydrospiro [1-benzoazine-4,4'-ethylene oxide] -2 (3H) -one (Intermediate 64) And trans-4- (trifluoromethyl) cyclohexanemethanehydrazine (Intermediate 6) to prepare the target product. MS (ESI) m / z 440.2 (M + H) + .

Synthesis Example 104
(3R) -8'-bromo-1 '-[trans-4- (pyridin-2-yloxy) cyclohexyl] -4,5-dihydro-4'H, 6'H-spiro [furan- 3,5 '-[1,2,4] triazolo [4,3-a] [1] benzoazepine]

with
Example 105
(3S) -8'-bromo-1 '-[trans-4- (pyridin-2-yloxy) cyclohexyl] -4,5-dihydro-4'H, 6'H-spiro [furan- 3,5 '-[1,2,4] triazolo [4,3-a] [1] benzoazepine]

By chiral preparative HPLC (CHIRALPAK IG preparative 20 μm stationary phase; 5 x 30 cm; F = 50 ml / min; eluent: n-heptane: ethanol = 6: 4; isocratic; temperature = 25 ℃ ), Self-racemic 8'-bromo-1 '-[trans-4- (pyridin-2-yloxy) cyclohexyl] -4,5-dihydro-4'H, 6'H-spiro [ Furan-3,5 '-[1,2,4] triazolo [4,3-a] [1] benzoazine] (Example 99) The target product was prepared. Determination of retention time by analytical HPLC (CHIRALPAK IG 5 μm stationary phase; 250 x 4.6 mm; F = 1 ml / min; eluent: n-heptane: ethanol = 6: 4; isocratic; temperature = 25 ° C) : The first eluate is 18.6 minutes; = + 10.0˚ (c = 0.11; CHCl 3 ); MS (ESI) m / z 495.2 and 497.2 (M + H) + ; The second lysate is 23.0 minutes; = -3.8˚ (c = 0.105; CHCl 3 ); MS (ESI) m / z 495.2 and 497.2 (M + H) + . The absolute configuration of these compounds has not been determined.

Example 106
(3S) -8'-chloro-1 '-[trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H, 4'H, 6'H-spiro [ Piperan-3,5 '-[1,2,4] triazolo [4,3-a] [1] benzoazepine]

with
Example 107
(3R) -8'-chloro-1 '-[trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H, 4'H, 6'H-spiro [ Piperan-3,5 '-[1,2,4] triazolo [4,3-a] [1] benzoazepine]

By chiral preparative HPLC (CHIRALPAK IG preparative 20 μm stationary phase; 5 x 30 cm; F = 50 ml / min; eluent: n-heptane: ethanol = 6: 4; isocratic; temperature = 25 ℃ ), Self-racemic 8'-chloro-1 '-[trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H, 4'H, 6'H- Spiro [piperan-3,5 '-[1,2,4] triazolo [4,3-a] [1] benzoazine] (Example 101) prepared the target product. Determination of retention time by analytical HPLC (CHIRALPAK IG 5 μm stationary phase; 250 x 4.6 mm; F = 1 ml / min; eluent: n-heptane: ethanol = 6: 4; isocratic; temperature = 25 ° C) : The first eluate is 20.2 minutes; = + 19.0˚ (c = 0.11; CHCl 3 ); MS (ESI) m / z 465.2 (M + H) + ; The second lysate is 26.5 minutes; = -7.6˚ (c = 0.105; CHCl 3 ); MS (ESI) m / z 465.2 (M + H) + . The absolute configuration of these compounds has not been determined.

Example 108
8-fluoro-1- (trans-4-methoxy-4-methylcyclohexyl) -5,5-dimethyl-5,6-dihydro-4H- [1,2,4] triazole Ac [4,3-a] [1] benzoacene

with
Example 109
8-fluoro-1- (cis-4-methoxy-4-methylcyclohexyl) -5,5-dimethyl-5,6-dihydro-4H- [1,2,4] triazole Ac [4,3-a] [1] benzoacene

P-Fluoro-4,4-dimethyl-2- (methylthio) -4,5-dihydro-3H-1-benzoazine (step b) of Example 91) (0.15 g, 0.63 Mmol), 4-methoxy-4-methylcyclohexane-1-methylhydrazine (Intermediate 35 step c)) (0.18 g, 0.95 mmol) and 1,4-dioxane (20 ml) of the mixture was added concentrated hydrochloric acid (0.010 ml) and the reaction mixture was refluxed for 3 hours. After evaporation of the solvent, the residue was subjected to preparative HPLC (Chromolith preparative RP18 column; 100 × 25 mm; 25 ml / min; A: water + 0.1% trifluoroacetic acid; B: acetonitrile + 0.1% trifluoroacetic acid; Gradient: 0 to 12 minutes; B: 0 to 49%; 12 to 24 minutes; B: 49 to 90%; temperature = 40 ° C). By analytical HPLC (Chromolith performance RP18 column; 100 × 4.6 mm; 1 ml / min; eluent: A: water + 0.1% trifluoroacetic acid; B: acetonitrile + 0.1% trifluoroacetic acid; gradient: 0 to 10 minutes; B: 0 to 90%; 10 to 15 minutes; B: 90%; temperature = 40 ° C) Determination of retention time: first eluate 8.62 minutes; trans isomer; 0.088 g (39%) ; MS (ESI) m / z 358.3 (M + H) + ; Second eluate 8.95 minutes; cis isomer; 0.021 g (9%); MS (ESI) m / z 358.2 (M + H) + .

Example 110
8'-Chloro-1 '-(trans-4-methoxy-4-methylcyclohexyl) -2,3,5,6-tetrahydro-4'H, 6'H-spiro [piperan- 4,5 '-[1,2,4] triazolo [4,3-a] [1] benzoazine]

with
Example 111
8'-Chloro-1 '-(cis-4-methoxy-4-methylcyclohexyl) -2,3,5,6-tetrahydro-4'H, 6'H-spiro [piperan- 4,5 '-[1,2,4] triazolo [4,3-a] [1] benzoazine]

a) 7-Chloro-2-methoxy-3,5-dihydrospiro [1-benzoazine-4,4'-ethylene oxide]

Let 7-chloro-1,5-dihydrospiro [1-benzoazine-4,4'-ethylene oxide] -2 (3H) -one (intermediate 54) (0.35 g, 1.32 mmol) ), A mixture of trimethyloxonium tetrafluoroborate (0.583 g, 3.95 mmol), potassium carbonate (0.73 g, 5.27 mmol) and dichloromethane (25 ml) were stirred at room temperature for 3 hours. The reaction mixture was then filtered and the organic phase was concentrated. Thus, the target product (0.35 g, 98%) was obtained, which was used in the next reaction step without further purification and characterization.

b) 8'-Chloro-1 '-(4-methoxy-4-methylcyclohexyl) -2,3,5,6-tetrahydro-4'H, 6'H-spiro [piperan-4 , 5 '-[1,2,4] triazolo [4,3-a] [1] benzoazepine]
P-Chloro-2-methoxy-3,5-dihydrospiro [1-benzoazine-4,4'-ethylene oxide] (0.35 g, 1.31 mmol), 4-methoxy A mixture of methyl-4-methylcyclohexane-1-formamidine (Intermediate 35 step c)) (0.74 g, 3.95 mmol) and 1,4-dioxane (20 ml) was added to trifluoro Acetic acid (0.030 ml) and the reaction mixture was stirred overnight at 75 ° C. After evaporation of the solvent, the residue was subjected to preparative HPLC (Kinetex EVO column; 5 μm load; 150 × 21.2 mm; 20 ml / min; eluate: 20 mmol / L (NH 4 ) 2 CO 3 : Acetonitrile = 65:35; isocratic; temperature = 40 ° C). By analytical HPLC (Kinetex EVO column; 5 μm load; 150 × 4.6 mm; 1 ml / min; eluent: 20 mmol / L (NH 4 ) 2 CO 3 : Acetonitrile = 65: 35; isocratic; temperature = 40 ° C) measurement retention time: first eluate 5.53 minutes; trans isomer; 0.056 g (10%); MS (ESI) m / z 416.2 ( M + H) + ; Second eluate system 8.06 minutes; cis isomer; 0.016 g (3%); MS (ESI) m / z 416.3 (M + H) + .

Synthesis Example 112
8-chloro-1- [trans-4- (pyridin-2-ylamino) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a ] [1] Methylbenzoazine-5-carboxylic acid

According to the method described in Example 1, from 7-chloro-2- pendant thio-2,3,4,5-tetrahydro-1 H Methyl-1-benzoazine-4-carboxylic acid methyl ester (Intermediate 4) and trans-4- (pyridin-2-ylamino) cyclohexanemethylhydrazine (Intermediate 67) were used to prepare the target product. MS (ESI) m / z 452.1 (M + H) + .
The following formulation examples illustrate representative pharmaceutical compositions of the present invention. However, the present invention is not limited to the following pharmaceutical composition.

Claims (53)

一種通式(I)化合物, 其中 環A係環烷基或雜環基; 若環B存在,Y係-O-、-C(O)-、-CH2 -、-NH-或鍵結;或,若環B不存在,Y係-N(C1-4 烷基)2 、C(O)OC1-4 烷基、可選擇地經鹵素取代之C1-4 烷基、C1-4 烷氧基或鹵素; 環B係可選擇地經取代之芳基、雜環基或雜芳基; 或B-Y-A-結合代表3H -螺[2-苯並呋喃-1,4’-哌啶-1’-基];或 R1 係氫、鹵素、C1-4 烷基、C1-4 烷氧基、CF3 或CN; R2 係氫或C1-4 烷基; R3 係(CH2 )n R4 、C(O)R5 或可選擇地經R6 取代之C1-4 烷基; 或R2 與R3 結合代表-(CH2 )p -O-(CH2 )q -或-(CH2 )r -; R4 係CN、疊氮基或Cy1 ; R5 係C1-4 烷基、C1-4 烷氧基、OH或NR7 R8 ; R6 係OR9 、NR10 R11 、側氧基、-O(CH2 )2 O-或一或多個鹵素; R7 和R8 獨立地氫、C1-4 烷基或Cy2 ,或R7 和R8 與彼等連接之N一起形成雜環; R9 係氫、可選擇地經NH2 或可選擇地經取代之芳基取代之C1-4 烷基、Si(CH3 )3 或C(O)R12 ; R10 和R11 獨立地氫、可選擇地經C1-4 烷氧基取代之C1-4 烷基、Cy3 或C(O)R13 ,或R10 和R11 與彼等連接之N一起形成可選擇地經取代之雜環; R12 係C1-5 烷基或NR14 R15 ; R13 係C1-4 烷基、Cy1 或NR16 R17 ; R14 和R15 獨立地氫或可選擇地經取代之芳基,或R14 和R15 與彼等連接之N一起形成可選擇地經取代之雜環; R16 和R17 係C1-4 烷基,或R16 和R17 與彼等連接之N一起形成可選擇地經取代之雜環; Cy1 係可選擇地經取代之雜芳基; Cy2 係可選擇地經取代之芳基或雜芳基; Cy3 係可選擇地經取代之環烷基或雜環基; X係C1-4 烷基、芳基或雜芳基; Z係C1-4 烷基; n係0或1; p係1、2或3; q係1、2或3; r係4、5或6; 及/或彼等之鹽及/或彼等之幾何異構物及/或彼等之立體異構物及/或彼等之鏡像異構物及/或彼等之消旋物及/或彼等之非鏡像異構物及/或彼等之生物活性代謝物及/或彼等之前藥及/或彼等之溶劑合物及/或彼等之水合物及/或彼等之多形體。A compound of general formula (I), Wherein ring A is a cycloalkyl or heterocyclic group; if ring B is present, Y is -O-, -C (O)-, -CH 2- , -NH- or a bond; or, if ring B is not present, Y-N (C 1-4 alkyl) 2 , C (O) OC 1-4 alkyl, C 1-4 alkyl optionally substituted with halogen, C 1-4 alkoxy or halogen; ring B is optionally substituted aryl, heterocyclyl or heteroaryl; or BYA-bonded represents 3H -spiro [2-benzofuran-1,4'-piperidine-1'-yl]; or R 1 is hydrogen, halogen, C 1-4 alkyl, C 1-4 alkoxy, CF 3 or CN; R 2 is hydrogen or C 1-4 alkyl; R 3 is (CH 2 ) n R 4 , C (O) R 5 or C 1-4 alkyl optionally substituted by R 6 ; or R 2 in combination with R 3 represents-(CH 2 ) p -O- (CH 2 ) q - or- (CH 2 ) r- ; R 4 is CN, azide or Cy 1 ; R 5 is C 1-4 alkyl, C 1-4 alkoxy, OH or NR 7 R 8 ; R 6 is OR 9 , NR 10 R 11 , pendant oxy, -O (CH 2 ) 2 O- or one or more halogens; R 7 and R 8 are independently hydrogen, C 1-4 alkyl or Cy 2 , or R 7 and R 8 are with them The linked N together form a heterocyclic ring; R 9 is hydrogen, C 1-4 alkyl optionally substituted with NH 2 or optionally substituted aryl, Si (CH 3 ) 3 or C (O) R 12 R 10 and R 11 are independently hydrogen, C 1-4 alkyl optionally substituted with C 1-4 alkoxy, Cy 3 or C (O) R 13 , or R 10 and R 11 are connected to each other And N together form an optionally substituted heterocyclic ring; R 12 is C 1-5 alkyl or NR 14 R 15 ; R 13 is C 1-4 alkyl, Cy 1 or NR 16 R 17 ; R 14 and R 15 is independently hydrogen or optionally substituted aryl, or R 14 and R 15 together with the N to which they are attached form an optionally substituted hetero Rings; R 16 and R 17 are C 1-4 alkyl, or R 16 and R 17 together with the N to which they are attached form an optionally substituted heterocyclic ring; Cy 1 is an optionally substituted heteroaryl Cy 2 is optionally substituted aryl or heteroaryl; Cy 3 is optionally substituted cycloalkyl or heteroaryl; X is C 1-4 alkyl, aryl or heteroaryl; Z is C 1-4 alkyl; n is 0 or 1; p is 1, 2 or 3; q is 1, 2 or 3; r is 4, 5 or 6; and / or their salts and / or Geometric isomers and / or their stereoisomers and / or their mirror isomers and / or their racemates and / or their non-image isomers and / or their Bioactive metabolites and / or their prodrugs and / or their solvates and / or their hydrates and / or their polymorphs. 如請求項1之化合物,其中R1 係氫、氟、氯、溴、甲基、甲氧基、CF3 或CN。A compound as claimed in claim 1, wherein R 1 is hydrogen, fluorine, chlorine, bromine, methyl, methoxy, CF 3 or CN. 如請求項1之化合物,其中 環A係3至6員飽和碳環、或含有1或2個N之4至7員飽和雜環; 環B係可選擇地經取代之6或5員單雜芳基、6至10員芳香族碳環或含有1、2或3個選自O、S或N的雜原子之4至7員飽和單環、雙環、稠合及/或橋連雜環; 或B-Y-A-結合代表3H -螺[2-苯並呋喃-1,4’-哌啶-1’-基];或 X係異丙基; Z係甲基。A compound as claimed in claim 1, wherein ring A is a 3 to 6 membered saturated carbocyclic ring, or a 4 to 7 membered saturated heterocyclic ring containing 1 or 2 N; ring B is optionally substituted 6 or 5 membered monocyclic Aryl, 6 to 10 membered aromatic carbocyclic rings or 4 to 7 membered saturated monocyclic, bicyclic, fused and / or bridged heterocyclic rings containing 1, 2 or 3 heteroatoms selected from O, S or N; Or BYA-binding represents 3H -spiro [2-benzofuran-1,4'-piperidine-1'-yl]; or X is isopropyl; Z is methyl. 如請求項1至3中任一項之化合物,其中環B係可選擇地經取代之6員單雜芳基、苯基、或含有1或2個選自O、S或N的雜原子之4至6員飽和單環雜環。A compound according to any one of claims 1 to 3, wherein ring B is optionally substituted 6-membered monoheteroaryl, phenyl, or a group containing 1 or 2 heteroatoms selected from O, S or N 4 to 6 membered saturated monocyclic heterocyclic ring. 如請求項1至3中任一項之化合物,其中若環B存在,Y係-O-、-C(O)-、-CH2 -、-NH-或單鍵。The compound according to any one of claims 1 to 3, wherein if ring B is present, Y is -O-, -C (O)-, -CH 2- , -NH- or a single bond. 如請求項1至3中任一項之化合物,其中環A係4至6員飽和碳環基、或經由環氮與Y或該5,6-二氫-4H -[1,2,4]三唑並[4,3-a][1]苯並吖呯(benzazepine)核之三唑環連接的含有1或2個N之4至7員飽和雜環。The compound according to any one of claims 1 to 3, wherein ring A is a 4- to 6-membered saturated carbocyclic group, or via ring nitrogen and Y or the 5,6-dihydro-4 H- [1,2,4 ] Triazolo [4,3-a] [1] Benzazepine nucleus is connected by a triazole ring containing 1 or 2 N 4 to 7 membered saturated heterocyclic ring. 如請求項1至3中任一項之化合物,其中環A係環己基,Y係-O-,環B係吡啶-2-基且R1 係氯。The compound according to any one of claims 1 to 3, wherein ring A is cyclohexyl, Y is -O-, ring B is pyridin-2-yl and R 1 is chlorine. 如請求項1至3中任一項之化合物,其中環A係四氫吖唉(azetidine)、哌啶或哌嗪,Y係-O-或單鍵,環B係吡啶、嘧啶或哌啶,且R1 係氯。A compound according to any one of claims 1 to 3, wherein ring A is azetidine, piperidine or piperazine, Y is -O- or a single bond, and ring B is pyridine, pyrimidine or piperidine, And R 1 is chlorine. 如請求項1至3中任一項之化合物,其中Y係-N(C1-4 烷基)2 、C(O)OC1-4 烷基、可選擇地經鹵素取代之C1-4 烷基、C1-4 烷氧基或鹵素,且環B不存在。A compound according to any one of claims 1 to 3, wherein Y is -N (C 1-4 alkyl) 2 , C (O) OC 1-4 alkyl, optionally substituted C 1-4 with halogen Alkyl, C 1-4 alkoxy or halogen, and ring B is absent. 如請求項3之化合物,其中環A係4至6員飽和碳環基。A compound as claimed in claim 3, wherein ring A is a 4- to 6-membered saturated carbocyclic group. 如請求項10之化合物,其中Y係選自-N(C1-4 烷基)2 、C(O)OC1-4 烷基、可選擇地經鹵素取代之C1-4 烷基、C1-4 烷氧基及鹵素中之一者。A compound as claimed in claim 10, wherein Y is selected from the group consisting of -N (C 1-4 alkyl) 2 , C (O) OC 1-4 alkyl, optionally substituted C 1-4 alkyl, C One of 1-4 alkoxy and halogen. 如請求項10之化合物,其中Y係選自可選擇地經鹵素取代之C1-4 烷基、C1-4 烷氧基及鹵素中之二者。A compound as claimed in claim 10, wherein Y is selected from the group consisting of C 1-4 alkyl, C 1-4 alkoxy, and halogen optionally substituted with halogen. 如請求項1之化合物,其中R2 係氫或C1-4 烷基,且R3 係(CH2 )n R4A compound as claimed in claim 1, wherein R 2 is hydrogen or C 1-4 alkyl, and R 3 is (CH 2 ) n R 4 . 如請求項13之化合物,其中R2 係氫,R3 係(CH2 )n R4 ,R4 係CN、疊氮基或可選擇地經C1-4 烷基取代之含有2個N和1個O或4個N的5員單雜芳基。A compound as claimed in claim 13, wherein R 2 is hydrogen, R 3 is (CH 2 ) n R 4 , R 4 is CN, azido or optionally substituted with C 1-4 alkyl containing 2 N and 1 O or 4 N 5-membered monoheteroaryl. 如請求項1之化合物,其中R2 係氫或C1-4 烷基,且R3 係C(O)R5A compound as claimed in claim 1, wherein R 2 is hydrogen or C 1-4 alkyl, and R 3 is C (O) R 5 . 如請求項15之化合物,其中R2 係氫。A compound as claimed in claim 15 wherein R 2 is hydrogen. 如請求項15之化合物,其中R5 係甲基、甲氧基、OH或NR7 R8 ,其中R7 和R8 獨立地氫、甲基、乙基或異丙基。A compound as claimed in claim 15, wherein R 5 is methyl, methoxy, OH or NR 7 R 8 , wherein R 7 and R 8 are independently hydrogen, methyl, ethyl or isopropyl. 如請求項1之化合物,其中R2 係氫或C1-4 烷基,且R3 係可選擇地經R6 取代之C1-4 烷基。The compound of 1, wherein R 2 system hydrogen or C 1-4 alkyl, and R 3 is optionally substituted with R 6 lines of C 1-4 alkyl substituted with request entry. 如請求項18之化合物,其中R2 係氫或C1-4 烷基,且R3 係C1-4 烷基。A compound as claimed in claim 18, wherein R 2 is hydrogen or C 1-4 alkyl, and R 3 is C 1-4 alkyl. 如請求項19之化合物,其中R2 係氫或C1-3 烷基,且R3 係C1-3 烷基。A compound as claimed in claim 19, wherein R 2 is hydrogen or C 1-3 alkyl, and R 3 is C 1-3 alkyl. 如請求項18之化合物,其中R3 係經R6 取代之單一或多個支鏈的C1-4 烷基。A compound as claimed in claim 18, wherein R 3 is a single or multiple branched C 1-4 alkyl substituted with R 6 . 如請求項21之化合物,其中R3 係-CH2 R6 或-CH(R6 )CH3A compound as claimed in claim 21, wherein R 3 is -CH 2 R 6 or -CH (R 6 ) CH 3 . 如請求項21之化合物,其中R2 係氫,且R6 係OR9A compound as claimed in claim 21, wherein R 2 is hydrogen and R 6 is OR 9 . 如請求項23之化合物,其中R9 係氫、可選擇地經苯基取代之C1-4 烷基、或C(O)R12 ,其中R12 係甲基。A compound as claimed in claim 23, wherein R 9 is hydrogen, optionally substituted C 1-4 alkyl, or C (O) R 12 , wherein R 12 is methyl. 如請求項21之化合物,其中R2 係C1-4 烷基,且R6 係OR9A compound as claimed in claim 21, wherein R 2 is C 1-4 alkyl and R 6 is OR 9 . 如請求項24之化合物,其中R2 係甲基或乙基,且R9 係氫。A compound as claimed in claim 24, wherein R 2 is methyl or ethyl and R 9 is hydrogen. 如請求項21之化合物,其中R2 係氫,且R6 係NR10 R11A compound as claimed in claim 21, wherein R 2 is hydrogen and R 6 is NR 10 R 11 . 如請求項27之化合物,其中R10 和R11 獨立地氫、可選擇地經C1-4 烷氧基取代之C1-4 烷基、Cy3 或C(O)R13 ,或R16 和R17 與彼等連接之N一起形成可選擇地經取代之雜環。A compound as claimed in claim 27, wherein R 10 and R 11 are independently hydrogen, C 1-4 alkyl, optionally substituted with C 1-4 alkoxy, Cy 3 or C (O) R 13 , or R 16 And R 17 together with the N to which they are attached form an optionally substituted heterocyclic ring. 如請求項21之化合物,其中R2 係C1-4 烷基,且R6 係NR10 R11A compound as claimed in claim 21, wherein R 2 is C 1-4 alkyl and R 6 is NR 10 R 11 . 如請求項21之化合物,其中R2 係氫,且R6 係一或多個鹵素。A compound as claimed in claim 21, wherein R 2 is hydrogen and R 6 is one or more halogens. 如請求項30之化合物,其中該鹵素係氟。The compound of claim 30, wherein the halogen is fluorine. 如請求項21之化合物,其中R2 係C1-4 烷基,且R6 係一或多個鹵素。A compound as claimed in claim 21, wherein R 2 is C 1-4 alkyl and R 6 is one or more halogens. 如請求項21之化合物,其中R6 係側氧基或-O(CH2 )2 O-。A compound as claimed in claim 21, wherein R 6 is pendant oxygen or -O (CH 2 ) 2 O-. 如請求項13、15及18中任一項之化合物,其中該5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯核之第5位置的碳之絕對構型係(R)。The compound according to any one of claims 13, 15 and 18, wherein the 5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazepine The absolute configuration of the carbon at position 5 of the nucleus (R). 如請求項13、15及18中任一項之化合物,其中該5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯核之第5位置的碳之絕對構型係(S)。The compound according to any one of claims 13, 15 and 18, wherein the 5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazepine The absolute configuration of the carbon at the 5th position of the nucleus (S). 如請求項1之化合物,其中R2 和R3 結合代表-(CH2 )p -O-(CH2 )q -,其中p係1、2或3,且q係1、2或3。A compound as claimed in claim 1, wherein R 2 and R 3 are combined to represent-(CH 2 ) p -O- (CH 2 ) q- , wherein p is 1, 2 or 3, and q is 1, 2 or 3. 如請求項35之化合物,其中p和q之和係3或4。A compound as claimed in claim 35, wherein the sum of p and q is 3 or 4. 如請求項1之化合物,其中R2 和R3 結合代表-(CH2 )r -,其中r係4、5或6。A compound as claimed in claim 1, wherein R 2 and R 3 in combination represent-(CH 2 ) r- , wherein r is 4, 5 or 6. 如請求項1之化合物,其中該化合物選自: 8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯; 8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸; 8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲醯胺; 8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-腈; 8-氯-N,N-二甲基-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺; (5S)-8-氯-N,N-二甲基-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺; (5R)-8-氯-N,N-二甲基-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺; 8-氯-N-(丙-2-基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺; {8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}(吡咯啶-1-基)甲酮; {8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}(嗎啉-4-基)甲酮; {8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}(1,1-二氧基硫代嗎啉-4-基)甲酮; {8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲醇; {(5S)-8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲醇; {(5R)-8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲醇; 8-氯-5-(甲氧基甲基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯; 嗎啉-4-甲酸{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲酯; (4-氟苯基)胺甲酸{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲酯; 5-(疊氮基甲基)-8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯; 1-{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲胺; 8-氯-5-(嗎啉-4-基甲基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯; 1-{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}-N,N-二甲基甲胺; N-({8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲基)丙-2-胺; 8-氯-1-[1-(嘧啶-2-基)四氫吖唉-3-基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯; 8-氯-1-[反式-4-(二甲基胺基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯; 8-氯-1-[反式-4-(三氟甲基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯; 8-氯-1-[反式-4-(嗎啉-4-基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯; {1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲醇; 8-氯-1-[反式-4-(吡咯啶-1-基羰基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯; {8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}乙腈; 8-氯-5-[(4-甲基哌嗪-1-基)甲基]-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯; 8-氯-5-(氟甲基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯; 8-氯-5-(3-甲基-1,2,4-噁二唑-5-基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯; 8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5-(2H-四唑-5-基)-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯; N-({8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲基)四氫-2H-哌喃-4-胺; N,N-二甲基-1-[反式-4-(吡啶-2-基氧基)環己基]-8-(三氟甲基)-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-胺; 8-氯-1-[反式-3-甲基-2-側氧基-1-氧雜-3-氮雜螺[4.5]癸-8-基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯; N-({8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲基)乙醯胺; N-({8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲基)氧雜環丁烷-3-胺; 8-氯-1-[反式-4-(嗎啉-4-基羰基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯; 8-氯-1-[順式-3-甲基-2-側氧基-1-氧雜-3-氮雜螺[4.5]癸-8-基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯; 8-氯-1-[1-(吡啶-2-基)四氫吖唉-3-基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯; 1-[反式-4-(四氫吖唉-1-基羰基)環己基]-8-氯-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯; 8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5-(吡咯啶-1-基甲基)-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯; N-({8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲基)-2-甲基-N-(2-甲基丙基)丙-1-胺; 8-氯-1-[反式-4-(哌啶-1-基羰基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯; 1-(反式-4-{[4-(三級丁氧羰基)哌嗪-1-基]羰基}環己基)-8-氯-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯; 8-氯-1-[反式-4-(哌嗪-1-基羰基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯鹽酸鹽; 8-甲氧基-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯; 8-甲基-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯; 8-氯-N-(4-氟苯基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺; 8-溴-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯; 1-(1,4'-聯哌啶-1’-基)-8-氯-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺; 8-氯-1-[(5r,8r)-1-側氧基-2-(丙-2-基)-2-氮雜螺[4.5]癸-8-基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯; 1-{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}乙醇; 1-{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}乙酮; 8-氯-5-(氟甲基)-1-(反式-4-甲氧基-4-甲基環己基)-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯; 1-{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}-N-(2-甲氧基乙基)乙胺; 8-氟-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯; 8-氯-5-(氟甲基)-1-[反式-4-(三氟甲基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯; 5-(氟甲基)-1-(反式-4-甲氧基-4-甲基環己基)-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯; {8-氟-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲醇; 8-氟-5-(氟甲基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯; 順式-(消旋)-1-{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}乙醇; 反式-(消旋)-1-{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}乙醇; (1R)-1-{(5R)-8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}乙醇; (1S)-1-{(5S)-8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}乙醇; (1S)-1-{(5R)-8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}乙醇; (1R)-1-{(5S)-8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}乙醇; 5-(氟甲基)-8-甲基-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯; (5S)-8-氯-5-(氟甲基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯; (5R)-8-氯-5-(氟甲基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯; 8-氟-1-[反式-4-(三氟甲基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯; 8-氯-1-[4-(2,3-二甲基苯基)哌嗪-1-基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺; {8-氟-1-[反式-4-(三氟甲基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲醇; {8-氯-5-甲基-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲醇; {8-氯-5-乙基-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲醇; (5S)-8-氟-5-(氟甲基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯; (5R)-8-氟-5-(氟甲基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯; 8-氯-1-[4-(3-氯苯基)哌嗪-1-基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺; 8-氯-1-[4-(吡啶-2-基)哌嗪-1-基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺; 8-氯-1-[4-(吡啶-2-基氧基)哌啶-1-基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺; 5-[(苄氧基)甲基]-8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯; 8-氯-1-(3,3-二氟環丁基)-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯; 8-氯-1-[反式-4-(哌啶-1-基甲基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯; 乙酸{8-氯-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-基}甲酯; 8-氯-1-(1'H,3H-螺[2-苯並呋喃-1,4'-哌啶]-1'-基)-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺; 8-氯-N-(吡啶-2-基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-羧醯胺; 8-氯-1-(1'H,3H-螺[2-苯並呋喃-1,4'-哌啶]-1'-基)-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-腈; 8-氯-1-[4-(2,3-二甲基苯基)哌嗪-1-基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-腈; 8-氯-1-[4-(吡啶-2-基)哌嗪-1-基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-腈; 8-氟-5,5-二甲基-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯; 8-氟-5-(丙-2-基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯; 8'-溴-1'-[反式-4-(吡啶-2-基氧基)環己基]-4'H,6'H-螺[環戊烷-1,5'-[1,2,4]三唑並[4,3-a][1]苯並吖呯]; 8-溴-5-(丙-2-基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯; 8'-溴-1'-[反式-4-(吡啶-2-基氧基)環己基]-2,3,5,6-四氫-4'H,6'H-螺[哌喃-4,5'-[1,2,4]三唑並[4,3-a][1]苯並吖呯]; 8-氟-5,5-二甲基-1-[反式-4-(三氟甲基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯; (5R)-8-溴-5-(丙-2-基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯; (5S)-8-溴-5-(丙-2-基)-1-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯; 8'-溴-1'-[反式-4-(吡啶-2-基氧基)環己基]-4,5-二氫-4'H,6'H-螺[呋喃-3,5'-[1,2,4]三唑並[4,3-a][1]苯並吖呯]; 8'-氯-1'-[反式-4-(吡啶-2-基氧基)環己基]-2,3,5,6-四氫-4'H,6'H-螺[哌喃-4,5'-[1,2,4]三唑並[4,3-a][1]苯並吖呯]; 8'-氯-1'-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H,4'H,6'H-螺[哌喃-3,5'-[1,2,4]三唑並[4,3-a][1]苯並吖呯]; 8'-氯-1'-[反式-4-(吡啶-2-基氧基)環己基]-4,5-二氫-4'H,6'H-螺[呋喃-3,5'-[1,2,4]三唑並[4,3-a][1]苯並吖呯]; 8'-氯-1'-[反式-4-(三氟甲基)環己基]-2,3,5,6-四氫-4'H,6'H-螺[哌喃-4,5'-[1,2,4]三唑並[4,3-a][1]苯並吖呯]; (3R)-8'-溴-1'-[反式-4-(吡啶-2-基氧基)環己基]-4,5-二氫-4'H,6'H-螺[呋喃-3,5'-[1,2,4]三唑並[4,3-a][1]苯並吖呯]; (3S)-8'-溴-1'-[反式-4-(吡啶-2-基氧基)環己基]-4,5-二氫-4'H,6'H-螺[呋喃-3,5'-[1,2,4]三唑並[4,3-a][1]苯並吖呯]; (3S)-8'-氯-1'-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H,4'H,6'H-螺[哌喃-3,5'-[1,2,4]三唑並[4,3-a][1]苯並吖呯]; (3R)-8'-氯-1'-[反式-4-(吡啶-2-基氧基)環己基]-5,6-二氫-4H,4'H,6'H-螺[哌喃-3,5'-[1,2,4]三唑並[4,3-a][1]苯並吖呯]; 8-氟-1-(反式-4-甲氧基-4-甲基環己基)-5,5-二甲基-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯; 8-氟-1-(順式-4-甲氧基-4-甲基環己基)-5,5-二甲基-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯; 8'-氯-1'-(反式-4-甲氧基-4-甲基環己基)-2,3,5,6-四氫-4'H,6'H-螺[哌喃-4,5'-[1,2,4]三唑並[4,3-a][1]苯並吖呯]; 8'-氯-1'-(順式-4-甲氧基-4-甲基環己基)-2,3,5,6-四氫-4'H,6'H-螺[哌喃-4,5'-[1,2,4]三唑並[4,3-a][1]苯並吖呯];及 8-氯-1-[反式-4-(吡啶-2-基胺基)環己基]-5,6-二氫-4H-[1,2,4]三唑並[4,3-a][1]苯並吖呯-5-甲酸甲酯。The compound of claim 1, wherein the compound is selected from: 8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a ] [1] Methyl benzoazine-5-carboxylic acid; 8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a ] [1] Benzacene-5-carboxylic acid; 8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a ] [1] Benzacene-5-carboxamide; 8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a ] [1] Benzacene-5-carbonitrile; 8-chloro-N, N-dimethyl-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] Zolo [4,3-a] [1] benzoazine-5-carboxamide; (5S) -8-chloro-N, N-dimethyl-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2 , 4] triazolo [4,3-a] [1] benzoazepine-5-carboxamide; (5R) -8-chloro-N, N-dimethyl-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2 , 4] triazolo [4,3-a] [1] benzoazepine-5-carboxamide; 8-chloro-N- (prop-2-yl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4 ] Triazolo [4,3-a] [1] benzoazepine-5-carboxamide; {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3- a] [1] Benzacridin-5-yl} (pyrrolidin-1-yl) methanone; {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3- a] [1] Benzacridin-5-yl} (morpholin-4-yl) methanone; {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3- a] [1] Benzacridin-5-yl} (1,1-dioxythiomorpholin-4-yl) methanone; {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3- a] [1] Benzacridin-5-yl} methanol; {(5S) -8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [ 4,3-a] [1] benzoazepine-5-yl} methanol; {(5R) -8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [ 4,3-a] [1] benzoazepine-5-yl} methanol; 8-chloro-5- (methoxymethyl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4 ] Triazolo [4,3-a] [1] benzoazine; Morpholine-4-carboxylic acid {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazole Benzo [4,3-a] [1] benzoazin-5-yl} methyl ester; (4-fluorophenyl) carbamic acid {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4 ] Triazolo [4,3-a] [1] benzoazin-5-yl} methyl ester; 5- (azidomethyl) -8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4 ] Triazolo [4,3-a] [1] benzoazine; 1- {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4, 3-a] [1] benzoazin-5-yl} methylamine; 8-chloro-5- (morpholin-4-ylmethyl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1, 2,4] triazolo [4,3-a] [1] benzoazine; 1- {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4, 3-a] [1] benzoazin-5-yl} -N, N-dimethylmethylamine; N-({8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4 , 3-a] [1] benzoazepine-5-yl} methyl) propan-2-amine; 8-chloro-1- [1- (pyrimidin-2-yl) tetrahydroazepine-3-yl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3- a] [1] Methylbenzoazine-5-carboxylic acid; 8-chloro-1- [trans-4- (dimethylamino) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [ 1] methyl benzoazine-5-carboxylic acid; 8-chloro-1- [trans-4- (trifluoromethyl) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1 ] Methyl benzoazine-5-carboxylic acid; 8-chloro-1- [trans-4- (morpholin-4-yl) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] methyl benzoazine-5-carboxylic acid; {1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1 ] Benzacridin-5-yl} methanol; 8-chloro-1- [trans-4- (pyrrolidin-1-ylcarbonyl) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a ] [1] Methyl benzoazine-5-carboxylic acid; {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3- a] [1] benzoazin-5-yl} acetonitrile; 8-chloro-5-[(4-methylpiperazin-1-yl) methyl] -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro -4H- [1,2,4] triazolo [4,3-a] [1] benzoazine; 8-chloro-5- (fluoromethyl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] tris Zolo [4,3-a] [1] benzoazine; 8-chloro-5- (3-methyl-1,2,4-oxadiazol-5-yl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5, 6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazine; 8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5- (2H-tetrazol-5-yl) -5,6-dihydro-4H- [1, 2,4] triazolo [4,3-a] [1] benzoazine; N-({8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4 , 3-a] [1] benzoazin-5-yl} methyl) tetrahydro-2H-piperan-4-amine; N, N-dimethyl-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -8- (trifluoromethyl) -5,6-dihydro-4H- [1, 2,4] triazolo [4,3-a] [1] benzoazepine-5-amine; 8-Chloro-1- [trans-3-methyl-2-oxo-1-oxa-3-azaspiro [4.5] dec-8-yl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazine-5-carboxylic acid methyl ester; N-({8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4 , 3-a] [1] benzoazepine-5-yl} methyl) acetamide; N-({8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4 , 3-a] [1] benzoazepin-5-yl} methyl) oxetan-3-amine; 8-chloro-1- [trans-4- (morpholin-4-ylcarbonyl) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a ] [1] Methyl benzoazine-5-carboxylic acid; 8-chloro-1- [cis-3-methyl-2-oxo-1-oxa-3-azaspiro [4.5] dec-8-yl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazine-5-carboxylic acid methyl ester; 8-chloro-1- [1- (pyridin-2-yl) tetrahydroazepine-3-yl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3- a] [1] Methylbenzoazine-5-carboxylic acid; 1- [trans-4- (tetrahydroazepine-1-ylcarbonyl) cyclohexyl] -8-chloro-5,6-dihydro-4H- [1,2,4] triazolo [4,3 -a] [1] methyl benzoazine-5-carboxylic acid; 8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5- (pyrrolidin-1-ylmethyl) -5,6-dihydro-4H- [1, 2,4] triazolo [4,3-a] [1] benzoazine; N-({8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4 , 3-a] [1] benzoazin-5-yl} methyl) -2-methyl-N- (2-methylpropyl) propan-1-amine; 8-chloro-1- [trans-4- (piperidin-1-ylcarbonyl) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a ] [1] Methyl benzoazine-5-carboxylic acid; 1- (trans-4-{[4- (tertiary butoxycarbonyl) piperazin-1-yl] carbonyl} cyclohexyl) -8-chloro-5,6-dihydro-4H- [1,2, 4] triazolo [4,3-a] [1] benzoazine-5-carboxylic acid methyl ester; 8-chloro-1- [trans-4- (piperazin-1-ylcarbonyl) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a ] [1] Benzoazine-5-carboxylic acid methyl ester hydrochloride; 8-methoxy-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3 -a] [1] methyl benzoazine-5-carboxylic acid; 8-methyl-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3- a] [1] Methylbenzoazine-5-carboxylic acid; 8-chloro-N- (4-fluorophenyl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4 ] Triazolo [4,3-a] [1] benzoazepine-5-carboxamide; 8-bromo-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a ] [1] Methyl benzoazine-5-carboxylic acid; 1- (1,4'-bipiperidin-1'-yl) -8-chloro-5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1 ] Benzoazepine-5-carboxamide; 8-chloro-1-[(5r, 8r) -1-oxo-2- (prop-2-yl) -2-azaspiro [4.5] dec-8-yl] -5,6-dihydro -4H- [1,2,4] triazolo [4,3-a] [1] benzoazine-5-carboxylic acid methyl ester; 1- {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4, 3-a] [1] benzoazepine-5-yl} ethanol; 1- {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4, 3-a] [1] benzoazin-5-yl} ethanone; 8-chloro-5- (fluoromethyl) -1- (trans-4-methoxy-4-methylcyclohexyl) -5,6-dihydro-4H- [1,2,4] triazole Ac [4,3-a] [1] benzoazine; 1- {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4, 3-a] [1] benzoazin-5-yl} -N- (2-methoxyethyl) ethylamine; 8-fluoro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a ] [1] Methyl benzoazine-5-carboxylic acid; 8-chloro-5- (fluoromethyl) -1- [trans-4- (trifluoromethyl) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [ 4,3-a] [1] benzoazine; 5- (fluoromethyl) -1- (trans-4-methoxy-4-methylcyclohexyl) -5,6-dihydro-4H- [1,2,4] triazolo [4, 3-a] [1] benzoazine; {8-fluoro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3- a] [1] Benzacridin-5-yl} methanol; 8-fluoro-5- (fluoromethyl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] tris Zolo [4,3-a] [1] benzoazine; Cis- (racemic) -1- {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2, 4] triazolo [4,3-a] [1] benzoazin-5-yl} ethanol; Trans- (racemic) -1- {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2, 4] triazolo [4,3-a] [1] benzoazin-5-yl} ethanol; (1R) -1-{(5R) -8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2, 4] triazolo [4,3-a] [1] benzoazin-5-yl} ethanol; (1S) -1-{(5S) -8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2, 4] triazolo [4,3-a] [1] benzoazin-5-yl} ethanol; (1S) -1-{(5R) -8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2, 4] triazolo [4,3-a] [1] benzoazin-5-yl} ethanol; (1R) -1-{(5S) -8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2, 4] triazolo [4,3-a] [1] benzoazin-5-yl} ethanol; 5- (fluoromethyl) -8-methyl-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] Triazolo [4,3-a] [1] benzoazine; (5S) -8-chloro-5- (fluoromethyl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2 , 4] triazolo [4,3-a] [1] benzoazine; (5R) -8-chloro-5- (fluoromethyl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2 , 4] triazolo [4,3-a] [1] benzoazine; 8-fluoro-1- [trans-4- (trifluoromethyl) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1 ] Methyl benzoazine-5-carboxylic acid; 8-chloro-1- [4- (2,3-dimethylphenyl) piperazin-1-yl] -5,6-dihydro-4H- [1,2,4] triazolo [4, 3-a] [1] benzoazine-5-carboxamide; {8-fluoro-1- [trans-4- (trifluoromethyl) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [ 1] benzoazepine-5-yl} methanol; {8-chloro-5-methyl-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazepine-5-yl} methanol; {8-chloro-5-ethyl-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazepine-5-yl} methanol; (5S) -8-fluoro-5- (fluoromethyl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2 , 4] triazolo [4,3-a] [1] benzoazine; (5R) -8-fluoro-5- (fluoromethyl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2 , 4] triazolo [4,3-a] [1] benzoazine; 8-chloro-1- [4- (3-chlorophenyl) piperazin-1-yl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] Benzacene-5-carboxamide; 8-chloro-1- [4- (pyridin-2-yl) piperazin-1-yl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] Benzacene-5-carboxamide; 8-chloro-1- [4- (pyridin-2-yloxy) piperidin-1-yl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3- a] [1] Benzacridin-5-carboxamide; 5-[(benzyloxy) methyl] -8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2 , 4] triazolo [4,3-a] [1] benzoazine; 8-chloro-1- (3,3-difluorocyclobutyl) -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] benzoazine呯 -5-carboxylic acid methyl ester; 8-chloro-1- [trans-4- (piperidin-1-ylmethyl) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3- a] [1] Methylbenzoazine-5-carboxylic acid; Acetic acid {8-chloro-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3 -a] [1] benzoazin-5-yl} methyl ester; 8-chloro-1- (1'H, 3H-spiro [2-benzofuran-1,4'-piperidine] -1'-yl) -5,6-dihydro-4H- [1,2, 4] triazolo [4,3-a] [1] benzoazine-5-carboxamide; 8-chloro-N- (pyridin-2-yl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4 ] Triazolo [4,3-a] [1] benzoazepine-5-carboxamide; 8-chloro-1- (1'H, 3H-spiro [2-benzofuran-1,4'-piperidine] -1'-yl) -5,6-dihydro-4H- [1,2, 4] triazolo [4,3-a] [1] benzoazine-5-carbonitrile; 8-chloro-1- [4- (2,3-dimethylphenyl) piperazin-1-yl] -5,6-dihydro-4H- [1,2,4] triazolo [4, 3-a] [1] benzoazine-5-carbonitrile; 8-chloro-1- [4- (pyridin-2-yl) piperazin-1-yl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a] [1] Benazine-5-carbonitrile; 8-fluoro-5,5-dimethyl-1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4] tris Zolo [4,3-a] [1] benzoazine; 8-fluoro-5- (prop-2-yl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4 ] Triazolo [4,3-a] [1] benzoazine; 8'-bromo-1 '-[trans-4- (pyridin-2-yloxy) cyclohexyl] -4'H, 6'H-spiro [cyclopentane-1,5'-[1,2 , 4] triazolo [4,3-a] [1] benzoazine]; 8-bromo-5- (prop-2-yl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1,2,4 ] Triazolo [4,3-a] [1] benzoazine; 8'-bromo-1 '-[trans-4- (pyridin-2-yloxy) cyclohexyl] -2,3,5,6-tetrahydro-4'H, 6'H-spiro [piperan -4,5 '-[1,2,4] triazolo [4,3-a] [1] benzoazine]; 8-fluoro-5,5-dimethyl-1- [trans-4- (trifluoromethyl) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [ 4,3-a] [1] benzoazine; (5R) -8-bromo-5- (prop-2-yl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1 , 2,4] triazolo [4,3-a] [1] benzoazine; (5S) -8-bromo-5- (prop-2-yl) -1- [trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H- [1 , 2,4] triazolo [4,3-a] [1] benzoazine; 8'-bromo-1 '-[trans-4- (pyridin-2-yloxy) cyclohexyl] -4,5-dihydro-4'H, 6'H-spiro [furan-3,5' -[1,2,4] triazolo [4,3-a] [1] benzoazine]; 8'-chloro-1 '-[trans-4- (pyridin-2-yloxy) cyclohexyl] -2,3,5,6-tetrahydro-4'H, 6'H-spiro [piperan -4,5 '-[1,2,4] triazolo [4,3-a] [1] benzoazine]; 8'-chloro-1 '-[trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H, 4'H, 6'H-spiro [piperan-3 , 5 '-[1,2,4] triazolo [4,3-a] [1] benzoazine]; 8'-Chloro-1 '-[trans-4- (pyridin-2-yloxy) cyclohexyl] -4,5-dihydro-4'H, 6'H-spiro [furan-3,5' -[1,2,4] triazolo [4,3-a] [1] benzoazine]; 8'-chloro-1 '-[trans-4- (trifluoromethyl) cyclohexyl] -2,3,5,6-tetrahydro-4'H, 6'H-spiro [piperan-4, 5 '-[1,2,4] triazolo [4,3-a] [1] benzoazine]; (3R) -8'-bromo-1 '-[trans-4- (pyridin-2-yloxy) cyclohexyl] -4,5-dihydro-4'H, 6'H-spiro [furan- 3,5 '-[1,2,4] triazolo [4,3-a] [1] benzoazine]; (3S) -8'-bromo-1 '-[trans-4- (pyridin-2-yloxy) cyclohexyl] -4,5-dihydro-4'H, 6'H-spiro [furan- 3,5 '-[1,2,4] triazolo [4,3-a] [1] benzoazine]; (3S) -8'-chloro-1 '-[trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H, 4'H, 6'H-spiro [ Piperan-3,5 '-[1,2,4] triazolo [4,3-a] [1] benzoazine]; (3R) -8'-chloro-1 '-[trans-4- (pyridin-2-yloxy) cyclohexyl] -5,6-dihydro-4H, 4'H, 6'H-spiro [ Piperan-3,5 '-[1,2,4] triazolo [4,3-a] [1] benzoazine]; 8-fluoro-1- (trans-4-methoxy-4-methylcyclohexyl) -5,5-dimethyl-5,6-dihydro-4H- [1,2,4] triazole Ac [4,3-a] [1] benzoazine; 8-fluoro-1- (cis-4-methoxy-4-methylcyclohexyl) -5,5-dimethyl-5,6-dihydro-4H- [1,2,4] triazole Ac [4,3-a] [1] benzoazine; 8'-Chloro-1 '-(trans-4-methoxy-4-methylcyclohexyl) -2,3,5,6-tetrahydro-4'H, 6'H-spiro [piperan- 4,5 '-[1,2,4] triazolo [4,3-a] [1] benzoazine]; 8'-Chloro-1 '-(cis-4-methoxy-4-methylcyclohexyl) -2,3,5,6-tetrahydro-4'H, 6'H-spiro [piperan- 4,5 '-[1,2,4] triazolo [4,3-a] [1] benzoazine]; and 8-chloro-1- [trans-4- (pyridin-2-ylamino) cyclohexyl] -5,6-dihydro-4H- [1,2,4] triazolo [4,3-a ] [1] Methyl benzoazine-5-carboxylic acid. 一種醫藥組成物,其包含治療有效量之如請求項1之通式(I)化合物及/或彼等之鹽及/或彼等之幾何異構物及/或彼等之立體異構物及/或彼等之鏡像異構物及/或彼等之消旋物及/或彼等之非鏡像異構物及/或彼等之生物活性代謝物及/或彼等之前藥及/或彼等之溶劑合物及/或彼等之水合物及/或彼等之多形體作為活性物質和醫藥上可接受之賦形劑。A pharmaceutical composition comprising a therapeutically effective amount of a compound of general formula (I) as claimed in claim 1 and / or their salts and / or their geometric isomers and / or their stereoisomers and / Or their mirror image isomers and / or their racemates and / or their non-image mirror isomers and / or their bioactive metabolites and / or their prodrugs and / or their Etc. solvates and / or their hydrates and / or their polymorphs serve as active substances and pharmaceutically acceptable excipients. 一種組合組成物,其包含治療有效量之如請求項1之通式(I)化合物及/或彼等之鹽及/或彼等之幾何異構物及/或彼等之立體異構物及/或彼等之鏡像異構物及/或彼等之消旋物及/或彼等之非鏡像異構物及/或彼等之生物活性代謝物及/或彼等之前藥及/或彼等之溶劑合物及/或彼等之水合物及/或彼等之多形體和一或多種其他活性物質。A combination composition comprising a therapeutically effective amount of a compound of general formula (I) as claimed in claim 1 and / or their salts and / or their geometric isomers and / or their stereoisomers and / Or their mirror image isomers and / or their racemates and / or their non-image mirror isomers and / or their bioactive metabolites and / or their prodrugs and / or their Etc. solvates and / or their hydrates and / or their polymorphs and one or more other active substances. 如請求項41之組合組成物,其中該一或多種其他活性物質選自精神阻斷劑、精神興奮劑、抗高血壓藥、解痙藥、抗癲癇藥或其他藥劑。The composition of claim 41, wherein the one or more other active substances are selected from the group consisting of a psychoblocker, a psychostimulant, an antihypertensive agent, an antispasmodic agent, an antiepileptic drug or other agents. 如請求項40至42中任一項之組成物,其係用於治療及/或預防與V1a受體之中樞及/或周圍拮抗作用有關之疾病或病症。A composition according to any one of claims 40 to 42, for use in the treatment and / or prevention of diseases or conditions associated with central and / or peripheral antagonistic effects of the V1a receptor. 一種製備具有V1a受體拮抗劑活性之醫藥組成物之方法,該醫藥組成物包含治療有效量之如請求項1至39中任一項之通式(I)化合物及/或彼等之鹽及/或彼等之幾何異構物及/或彼等之立體異構物及/或彼等之鏡像異構物及/或彼等之消旋物及/或彼等之非鏡像異構物及/或彼等之生物活性代謝物及/或彼等之前藥及/或彼等之溶劑合物及/或彼等之水合物及/或彼等之多形體作為活性物質和醫藥上可接受之賦形劑。A method for preparing a medicinal composition having V1a receptor antagonist activity, the medicinal composition comprising a therapeutically effective amount of a compound of the general formula (I) according to any one of claims 1 to 39 and / or their salts, and / Or their geometric isomers and / or their stereoisomers and / or their mirror isomers and / or their racemates and / or their non-mirro isomers and / Or their biologically active metabolites and / or their prodrugs and / or their solvates and / or their hydrates and / or their polymorphs as active substances and pharmaceutically acceptable excipient. 如請求項1之通式(I)化合物,及/或彼等之鹽及/或彼等之幾何異構物及/或彼等之立體異構物及/或彼等之鏡像異構物及/或彼等之消旋物及/或彼等之非鏡像異構物及/或彼等之生物活性代謝物及/或彼等之前藥及/或彼等之溶劑合物及/或彼等之水合物及/或彼等之多形體,其係用於治療及/或預防與V1a受體之中樞及/或周圍拮抗作用有關之疾病或病症。Compounds of general formula (I) as claimed in claim 1, and / or their salts and / or their geometric isomers and / or their stereoisomers and / or their mirror isomers and / Or their racemates and / or their non-image isomers and / or their biologically active metabolites and / or their prodrugs and / or their solvates and / or their Hydrates and / or their polymorphs are used to treat and / or prevent diseases or conditions associated with central and / or peripheral antagonistic effects of the V1a receptor. 如請求項45之化合物,其中該與V1a受體之中樞及/或周圍拮抗作用有關之疾病或病症是選自雌性性器官之多種病理症狀、血壓控制之持久性症狀、因血管升壓素之不適當分泌所引起之症狀、焦慮、抑鬱、攻擊行為、中樞神經系統疾病(其中該疾病之多種症狀之一及/或多種症狀可能與焦慮、抑鬱或攻擊行為有關或與該前述3種疾病顯示合併症(自閉症類群障礙、強迫症、多種不同類型之唐氏綜合症及創傷後應激障礙))、攻擊性行為障礙及/或易怒、行為過動症、認知障礙或其他神經精神疾病。The compound according to claim 45, wherein the disease or condition related to the central and / or peripheral antagonist of the Via receptor is selected from a variety of pathological symptoms of female sex organs, persistent symptoms of blood pressure control, and Symptoms caused by proper secretion, anxiety, depression, aggressive behavior, central nervous system disease (wherein one of the symptoms of the disease and / or multiple symptoms may be related to anxiety, depression or aggressive behavior or show a combination of the aforementioned three diseases Disease (autism group disorder, obsessive-compulsive disorder, many different types of Down's syndrome and post-traumatic stress disorder)), aggressive behavior disorder and / or irritability, hyperactivity disorder, cognitive impairment or other neuropsychiatric disorders. 一種如請求項1之通式(I)化合物及/或彼等之鹽及/或彼等之幾何異構物及/或彼等之立體異構物及/或彼等之鏡像異構物及/或彼等之消旋物及/或彼等之非鏡像異構物及/或彼等之生物活性代謝物及/或彼等之前藥及/或彼等之溶劑合物及/或彼等之水合物及/或彼等之多形體於製造藥物之用途,該藥物係用於治療及/或預防與V1a受體之中樞及/或周圍拮抗作用有關之疾病或病症。A compound of general formula (I) as claimed in claim 1 and / or their salts and / or their geometric isomers and / or their stereoisomers and / or their mirror isomers and / Or their racemates and / or their non-image isomers and / or their biologically active metabolites and / or their prodrugs and / or their solvates and / or their Hydrates and / or their polymorphs for use in the manufacture of a medicament for the treatment and / or prevention of diseases or conditions associated with central and / or peripheral antagonistic effects of the Via receptor. 如請求項47之用途,其中該與V1a受體之中樞及/或周圍拮抗作用有關之疾病或病症是選自雌性性器官之多種病理症狀、血壓控制之持久性症狀、因血管升壓素之不適當分泌所引起之症狀、焦慮、抑鬱、攻擊行為、中樞神經系統疾病(其中該疾病之多種症狀之一及/或多種症狀可能與焦慮、抑鬱或攻擊行為有關或與該前述3種疾病顯示合併症(自閉症類群障礙、強迫症、多種不同類型之唐氏綜合症及創傷後應激障礙))、攻擊性行為障礙及/或易怒、行為過動症、認知障礙或其他神經精神疾病。The use according to claim 47, wherein the disease or condition related to the central and / or peripheral antagonistic action of the V1a receptor is selected from a variety of pathological symptoms of female sex organs, persistent symptoms of blood pressure control, and Symptoms caused by proper secretion, anxiety, depression, aggressive behavior, central nervous system disease (wherein one of the symptoms of the disease and / or multiple symptoms may be related to anxiety, depression or aggressive behavior or show a combination of the aforementioned three diseases Disease (autism group disorder, obsessive-compulsive disorder, many different types of Down's syndrome and post-traumatic stress disorder)), aggressive behavior disorder and / or irritability, hyperactivity disorder, cognitive impairment or other neuropsychiatric disorders. 一種式(III-a)化合物, 其中R1 係氯、溴、氟、甲基或甲氧基。A compound of formula (III-a), Wherein R 1 is chlorine, bromine, fluorine, methyl or methoxy. 一種式(X)化合物, 其中R1 係氯、溴、氟、甲基或甲氧基。A compound of formula (X), Wherein R 1 is chlorine, bromine, fluorine, methyl or methoxy. 一種式(XLII)化合物,其係1-溴-8-氯-5,6-二氫-4H -[1,2,4]三唑並[4,3-a ][1]苯並吖呯-5-甲酸甲酯A compound of formula (XLII), which is 1-bromo-8-chloro-5,6-dihydro-4 H- [1,2,4] triazolo [4,3- a ] [1] benzoazine甲酯 -5-carboxylic acid methyl ester . 一種式(LI)化合物, 其中R1 係如通式(I)定義者,R2 和R3 一起形成-(CH2 )p -O-(CH2 )q -或-(CH2 )r -。A compound of formula (LI), Wherein R 1 is as defined by the general formula (I), R 2 and R 3 together form-(CH 2 ) p -O- (CH 2 ) q - or- (CH 2 ) r- . 一種式(LIV)化合物, 其中R1 係氯、氟或甲基。A compound of formula (LIV), Wherein R 1 is chlorine, fluorine or methyl.
TW107144825A 2017-12-15 2018-12-12 Tricyclic compounds as vasopressin V1a receptor antagonists TW201938171A (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
HUP1700522 2017-12-15
HU1700522A HUP1700522A1 (en) 2017-12-15 2017-12-15 Tricyclic compounds
HU1800332A HUP1800332A1 (en) 2018-09-28 2018-09-28 Tricyclic compounds ii
HUP1800332 2018-09-28

Publications (1)

Publication Number Publication Date
TW201938171A true TW201938171A (en) 2019-10-01

Family

ID=89992764

Family Applications (1)

Application Number Title Priority Date Filing Date
TW107144825A TW201938171A (en) 2017-12-15 2018-12-12 Tricyclic compounds as vasopressin V1a receptor antagonists

Country Status (2)

Country Link
TW (1) TW201938171A (en)
WO (1) WO2019116325A1 (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TW202202495A (en) 2020-03-26 2022-01-16 匈牙利商羅特格登公司 Naphthyridine and pyrido〔3,4-c〕pyridazine derivatives as gamma-aminobutyric acid a receptor subunit alpha 5 receptor modulators
CN114644635B (en) * 2020-12-21 2023-02-03 上海济煜医药科技有限公司 Triazole tricyclic derivative and preparation method and application thereof
HUP2100338A1 (en) 2021-09-29 2023-04-28 Richter Gedeon Nyrt Bicyclic amine derivatives as gabaa alfa5 receptor modulators

Family Cites Families (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE69009785T2 (en) 1989-02-10 1994-11-03 Otsuka Pharma Co Ltd Carbostyril derivatives.
FR2679903B1 (en) 1991-08-02 1993-12-03 Elf Sanofi DERIVATIVES OF N-SULFONYL INDOLINE CARRYING AN AMIDIC FUNCTION, THEIR PREPARATION, THE PHARMACEUTICAL COMPOSITIONS CONTAINING SAME.
US5595872A (en) 1992-03-06 1997-01-21 Bristol-Myers Squibb Company Nucleic acids encoding microsomal trigyceride transfer protein
CA2167673C (en) 1993-07-21 2004-09-21 Akihiro Tanaka Condensed benzazepine derivative and pharmaceutical composition thereof
ES2284741T3 (en) 1994-06-15 2007-11-16 Otsuka Pharmaceutical Company, Limited BENZOHETEROCICLIC DERIVATIVES USED AS OXITOCINE VASOPRSINAO MODULATORS.
FR2740136B1 (en) 1995-10-24 1998-01-09 Sanofi Sa INDOLIN-2-ONE DERIVATIVES, PROCESS FOR THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
CA2409819C (en) 2000-05-19 2009-09-15 Yamanouchi Pharmaceutical Co., Ltd. Triazole derivatives
NZ523450A (en) 2000-07-05 2004-11-26 Ortho Mcneil Pharm Inc Nonpeptide substituted spirobenzoazepines as vasopressin receptor antagonists
US7056910B2 (en) 2000-11-28 2006-06-06 Astellas Pharma Inc. 1,4,5.6-tetrahydroimidazo[4,5-d]diazepine derivatives or salts thereof
EP1434763B1 (en) 2001-10-12 2018-01-17 Azevan Pharmaceuticals, Inc. Beta-lactamyl vasopressin v1a antagonists
JP2005104838A (en) 2003-01-09 2005-04-21 Tanabe Seiyaku Co Ltd Condensed furan compound
GB0303852D0 (en) 2003-02-19 2003-03-26 Pfizer Ltd Triazole compounds useful in therapy
EA010132B1 (en) 2003-12-22 2008-06-30 Пфайзер Инк. Triazole derivatives as vasopressin antagonists
GB0400700D0 (en) 2004-01-13 2004-02-18 Pfizer Ltd Compounds useful in therapy
EP1632494A1 (en) 2004-08-24 2006-03-08 Ferring B.V. Vasopressin v1a antagonists
US7456170B2 (en) 2004-08-25 2008-11-25 Pfizer Inc. Triazolobenzodiazepines and their use as vasopressin antagonists
CA2593044A1 (en) 2004-12-31 2006-07-13 Abbott Gmbh & Co. Kg Substituted oxindole derivatives, medicaments containing said derivatives and use thereof
CA2602194A1 (en) 2005-03-24 2006-09-28 Abbott Gmbh & Co. Kg Substituted oxindol derivatives, drugs containing said derivatives and the use thereof
ZA200709626B (en) 2005-05-13 2009-02-25 Lexicon Pharmaceuticals Inc Multicyclic compounds and methods of their use
DK1904477T3 (en) 2005-07-14 2009-03-16 Hoffmann La Roche Indole-3-carbonyl-spiro-piperodome derivatives as V1A receptor antagonists
PL1912976T3 (en) 2005-07-21 2009-04-30 Hoffmann La Roche Indol-3-yl-carbonyl-piperidin-benzoimidazole derivatives as v1a receptor antagonists
EP1917255A2 (en) 2005-07-29 2008-05-07 F. Hoffmann-Roche AG Indol-3-yl-carbonyl-piperidin and piperazin derivatives
DE102006051796A1 (en) * 2006-11-03 2008-05-08 Merck Patent Gmbh Triaza-benzo [e] azulene derivatives
JP2010517966A (en) 2007-02-01 2010-05-27 グラクソ グループ リミテッド 1-oxa-3-azaspiro (4.5) decan-2-one and 1-oxa-3,8-diazaspiro (4.5) decan-2-one derivatives for the treatment of eating disorders
TWI459947B (en) 2007-06-26 2014-11-11 Otsuka Pharma Co Ltd Benzazepine compound and pharmaceutical preparation
MX2011005596A (en) 2008-11-28 2011-06-16 Hoffmann La Roche Arylcyclohexylethers of dihydrotetraazabenzoazulenes for use as vasopressin via receptor antagonists.
GB0903493D0 (en) 2009-02-27 2009-04-08 Vantia Ltd New compounds
SG10201406839VA (en) 2009-10-26 2014-12-30 Otsuka Pharma Co Ltd Benzazepine compound
GB201004677D0 (en) 2010-03-19 2010-05-05 Vantia Ltd New salt
US8461151B2 (en) 2010-04-13 2013-06-11 Hoffmann-La Roche Inc. Aryl-/heteroaryl-cyclohexenyl-tetraazabenzo[e]azulenes
US8492376B2 (en) * 2010-04-21 2013-07-23 Hoffmann-La Roche Inc. Heteroaryl-cyclohexyl-tetraazabenzo[e]azulenes
US8513238B2 (en) 2010-05-10 2013-08-20 Hoffmann-La Roche Inc. Heteroaryl-cyclohexyl-tetraazabenzo[E]azulenes
JP5968307B2 (en) 2010-05-11 2016-08-10 サノフイ Substituted phenylcycloalkylpyrrolidine (piperidine) spirolactams and amides, their preparation and therapeutic use
CN104781259B (en) * 2012-09-28 2018-06-01 拜耳制药股份公司 Inhibit the 5- aryl triazoles and azepine * of BET albumen
EP2956441A4 (en) 2013-02-18 2016-11-02 Scripps Research Inst Modulators of vasopressin receptors with therapeutic potential
RS62639B1 (en) 2015-07-06 2021-12-31 Alkermes Inc Hetero-halo inhibitors of histone deacetylase
CN106349241B (en) * 2015-07-15 2020-04-21 上海翰森生物医药科技有限公司 Triazole derivative with HSP90 inhibitory activity and preparation method and application thereof
US9988367B2 (en) 2016-05-03 2018-06-05 Bayer Pharma Aktiengesellschaft Amide-substituted pyridinyltriazole derivatives and uses thereof
AR108263A1 (en) 2016-05-03 2018-08-01 Bayer Pharma AG PROCEDURE FOR THE PREPARATION OF DERIVATIVES OF 1-PHENYL-1,2,4-TRIAZOL REPLACED BY 5-HYDROXIALQUIL
WO2017191114A1 (en) 2016-05-03 2017-11-09 Bayer Aktiengesellschaft Hydroxyalkyl-substituted heteroaryltriazole derivatives and uses thereof

Also Published As

Publication number Publication date
WO2019116325A1 (en) 2019-06-20

Similar Documents

Publication Publication Date Title
CA2997382C (en) Substituted amino triazoles useful as human chitinase inhibitors
TWI389893B (en) Di (arylamino) ary1 compound
TWI826406B (en) Triazolobenzazepines as vasopressin v1a receptor antagonists
TW201825472A (en) Novel compounds
KR20140138865A (en) Imidazo[1,2-b]pyridazine-based compounds, compositions comprising them, and uses thereof
AU2014358766B2 (en) Use of benzimidazole-proline derivatives
WO2010098488A1 (en) Aryl imidazole compounds and their use as beta amyloid production inhibitors
JP2020526543A (en) Inhibitor of leucine-rich repeat kinase 2
JP2009541463A (en) Piperidine or pyrrolidine urea derivatives, their preparation and their therapeutic use
KR20090008335A (en) Trisubstituted 1,2,4-triazoles
TW201938171A (en) Tricyclic compounds as vasopressin V1a receptor antagonists
US20230374143A1 (en) A sortilin antagonist for use in the prevention or treatment of hearing loss
JP2020505399A (en) Compound
CN112313220A (en) PD-L1 antagonist compounds
TW202035403A (en) Bicyclic derivatives as gabaa a5 receptor modulators
WO2021078227A1 (en) Fused heteroaryl derivative, preparation method therefor, and application thereof in medicine
CN115348962A (en) As GABA A Naphthyridines and pyrido [3,4-c ] of alpha 5 receptor modulators]Pyridazine derivatives
RU2527106C2 (en) IMIDAZOLE DERIVATIVES AS mGluR5 ANTAGONISTS
EA042383B1 (en) Triazolobenzazepines as V1A Vasopressin Receptor Antagonists
TW202330518A (en) BICYCLIC AMINE DERIVATIVES AS GABAA α5 RECEPTOR MODULATORS
OA19571A (en) Triazolobenzazepines as vasopressin V1a receptor antagonists.
AU2015370697A1 (en) Novel heterocyclic compounds and the use thereof in medicine and in cosmetics