TWI812677B - Novel compounds for the treatment, alleviation or prevention of disorders associated with tau aggregates - Google Patents

Novel compounds for the treatment, alleviation or prevention of disorders associated with tau aggregates Download PDF

Info

Publication number
TWI812677B
TWI812677B TW108107135A TW108107135A TWI812677B TW I812677 B TWI812677 B TW I812677B TW 108107135 A TW108107135 A TW 108107135A TW 108107135 A TW108107135 A TW 108107135A TW I812677 B TWI812677 B TW I812677B
Authority
TW
Taiwan
Prior art keywords
group
mmol
compound
tau
alkyl
Prior art date
Application number
TW108107135A
Other languages
Chinese (zh)
Other versions
TW202017923A (en
Inventor
史林尼伐沙雀利 南帕利
艾瑪紐 葛畢利瑞
傑洛米 莫里特
Original Assignee
瑞士商Ac免疫公司
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
Application filed by 瑞士商Ac免疫公司 filed Critical 瑞士商Ac免疫公司
Publication of TW202017923A publication Critical patent/TW202017923A/en
Application granted granted Critical
Publication of TWI812677B publication Critical patent/TWI812677B/en

Links

Abstract

The present invention relates to novel compounds that can be employed in the treatment, alleviation or prevention of a group of disorders and abnormalities associated with Tau (Tubulin associated unit) protein aggregates including, but not limited to, Neurofibrillary Tangles (NFTs), such as Alzheimer’s disease (AD).

Description

用於治療、改善或預防與TAU聚集體有關的病症之新穎化合物Novel compounds for treating, ameliorating or preventing conditions associated with TAU aggregates

本發明係關於新穎化合物,其可用於治療、改善或預防與Tau (微管蛋白相關單元)蛋白質聚集體(包括但不限於神經纖維纏結(NFT)有關的一組病症及異常,例如阿茲海默氏病(Alzheimer’s disease,AD)。The present invention relates to novel compounds useful in the treatment, amelioration or prevention of a group of conditions and abnormalities associated with Tau (tubulin-associated unit) protein aggregates, including but not limited to neurofibrillary tangles (NFTs), such as Alzheimer's disease Alzheimer's disease (AD).

許多老化疾病係基於類澱粉或類澱粉樣蛋白之細胞外或細胞內沈積或與其有關,該等類澱粉或類澱粉樣蛋白促成發病機制以及疾病之進展。形成細胞外聚集體之最佳表徵之類澱粉蛋白係類澱粉β (Aβ)。形成細胞外聚集體之類澱粉蛋白之其他實例係普里昂蛋白(prion)、ATTR (轉甲狀腺素)或ADan (ADanPP)。主要形成細胞內聚集體之類澱粉樣蛋白包括(但不限於) Tau、α-突觸核蛋白、TAR DNA結合蛋白43 (TDP-43)及亨庭頓蛋白(huntingtin,htt)。涉及Tau聚集體之疾病通常列示為tau蛋白病變(例如AD)。Many diseases of aging are based on or related to extracellular or intracellular deposition of amyloid or amyloid-like proteins that contribute to pathogenesis and disease progression. The best characterized amyloid protein that forms extracellular aggregates is amyloid beta (Aβ). Other examples of amyloid proteins that form extracellular aggregates are prion, ATTR (transthyretin) or ADan (ADanPP). Amyloid proteins that mainly form intracellular aggregates include (but are not limited to) Tau, α-synuclein, TAR DNA-binding protein 43 (TDP-43), and huntingtin (htt). Diseases involving tau aggregates are often classified as tauopathies (eg, AD).

類澱粉或類澱粉樣沈積物係由蛋白質之錯誤摺疊、之後聚集從而產生其中多個肽或蛋白質藉由分子間氫鍵保持在一起之β褶板總成而造成。雖然類澱粉或類澱粉樣蛋白具有不同的一級胺基酸序列,但其沈積物通常含有許多共享分子成分、尤其β褶板四級結構之存在。在很大程度上仍不清楚類澱粉沈積物與疾病之間的關聯。已發現各種各樣之蛋白質聚集體(包括與疾病病理學相關及不相關之彼等蛋白質聚集體二者)係有毒的,此表明類澱粉之共同分子特徵與疾病發病有關或導致疾病發病(Bucciantini等人,Nature, 2002, 416, 507-11)。β褶板聚集之肽或蛋白質之各種多聚體亦與範圍為二聚體直至可溶性低分子量寡聚物、原纖維或不溶性纖維狀沈積物之不同肽或蛋白質之毒性有關。Amyloid or amyloid-like deposits result from the misfolding and subsequent aggregation of proteins to produce beta-pleated plate assemblies in which multiple peptides or proteins are held together by intermolecular hydrogen bonds. Although amyloid or amyloid-like proteins have different primary amino acid sequences, their deposits usually contain many shared molecular components, especially the presence of β-pleated plate quaternary structures. The link between amyloid deposits and disease remains largely unclear. A wide variety of protein aggregates, both those related to disease pathology and those unrelated to disease pathology, have been found to be toxic, suggesting that common molecular features of amyloid species are associated with or contribute to disease pathogenesis (Bucciantini et al., Nature, 2002, 416, 507-11). Various polymers of β-pleated plate aggregates of peptides or proteins are also associated with the toxicity of different peptides or proteins ranging from dimers to soluble low molecular weight oligomers, fibrils or insoluble fibrillar deposits.

阿茲海默氏病(AD)係神經病症,主要認為其係由類澱粉斑塊所引起,即腦中(類澱粉-β) Aβ聚集體之異常沈積物之細胞外累積。AD之其他主要神經病理學標誌係細胞內神經纖維纏結(NFT),其源自高度磷酸化Tau蛋白、錯誤摺疊之Tau或病理性Tau及其構形異構物之聚集。AD與許多神經退化性tau蛋白病變、尤其與指定類型之額顳葉失智症(FTD)共享其發病原理。Tau蛋白係易溶解之「天然未摺疊」蛋白質,其緊密結合至微管(MT)以促進其組裝及穩定性。MT對於神經元之細胞支架完整性至關重要,且由此對於神經元迴路之適當形成及功能、因此對於學習及記憶至關重要。Tau與MT之結合係由動態磷酸化及去磷酸化控制,如主要在活體外及非神經元細胞中所展示。在AD腦中,Tau病理學(tau蛋白病變)之發生晚於類澱粉病理學,但業內對Aβ蛋白是否係AD之致病物(此構成所謂的類澱粉級聯假說之本質)之討論仍頗有爭議(Hardy等人,Science 1992, 256, 184-185;Musiek等人,Nature Neurosciences 2015, 18(6), 800-806)。將類澱粉與Tau病理學聯繫起來之確切機制在很大程度上仍係未知的,但提出涉及神經元信號傳導路徑之活化,該等路徑作用於作為主要「Tau-激酶」之GSK3及cdk5或藉由其起作用(Muyllaert等人,Rev. Neurol. (Paris), 2006, 162, 903-7;Muyllaert等人,Genes Brain and Behav. 2008,增刊1,57-66)。即使tau蛋白病變之發生晚於類澱粉,但其並非僅係無害的副作用,而係AD之主要病理執行者。在實驗小鼠模型中,由類澱粉病理學所引起之認知缺陷因不存在Tau蛋白而幾乎完全改善(Roberson等人,Science, 2007, 316(5825), 750-4),且認知功能障礙及失智症之嚴重程度與tau蛋白病變相關,而與類澱粉病理學不相關。Alzheimer's disease (AD) is a neurological disorder thought to be caused by amyloid plaques, the extracellular accumulation of abnormal deposits of (amyloid-β) Aβ aggregates in the brain. Other major neuropathological hallmarks of AD are intracellular neurofibrillary tangles (NFTs), which originate from the aggregation of hyperphosphorylated Tau protein, misfolded Tau, or pathological Tau and its conformational isomers. AD shares its pathogenesis with many neurodegenerative tauopathies, particularly with certain types of frontotemporal dementia (FTD). Tau protein is a readily soluble "naturally unfolded" protein that tightly binds to microtubules (MTs) to promote their assembly and stability. MTs are essential for the integrity of the cellular scaffold of neurons and thus for the proper formation and function of neuronal circuits and therefore for learning and memory. The association of Tau with MT is controlled by dynamic phosphorylation and dephosphorylation, as shown primarily in vitro and in non-neuronal cells. In the AD brain, tau pathology (tau pathology) occurs later than amyloid pathology, but there is still discussion within the industry as to whether Aβ protein is the causative agent of AD (this constitutes the essence of the so-called amyloid cascade hypothesis). Quite controversial (Hardy et al., Science 1992, 256, 184-185; Musiek et al., Nature Neurosciences 2015, 18(6), 800-806). The exact mechanisms linking amyloid and tau pathology remain largely unknown, but are proposed to involve activation of neuronal signaling pathways acting on GSK3 and cdk5 as major "Tau-kinases" or By which it acts (Muyllaert et al., Rev. Neurol. (Paris), 2006, 162, 903-7; Muyllaert et al., Genes Brain and Behav. 2008, Suppl. 1, 57-66). Even though tauopathy occurs later than amyloid, it is not just a harmless side effect, but the main pathological executor of AD. In experimental mouse models, cognitive deficits caused by amyloid pathology are almost completely ameliorated by the absence of Tau protein (Roberson et al., Science, 2007, 316(5825), 750-4), and cognitive dysfunction and Dementia severity correlates with tauopathy but not amyloid pathology.

涉及Tau聚集體之疾病通常列示為tau蛋白病變,且其包括(但不限於)阿茲海默氏病(AD)、家族性AD、PART (原發性年齡相關性Tau蛋白病變)、庫賈氏病(Creutzfeldt-Jacob disease)、拳擊手型失智症、唐氏症候群(Down’s Syndrome)、傑茨曼-斯脫司勒-史茵克病(Gerstmann-Sträussler-Scheinker disease,GSS)、包涵體肌炎、普里昂蛋白腦類澱粉血管病變、創傷性腦損傷(TBI)、肌肉萎縮性脊髓側索硬化症(ALS)、關島帕金森氏症-失智複合症(Parkinsonism-dementia complex of Guam)、非關島運動神經元病伴神經纖維纏結(non-Guamanian motor neuron disease with neurofibrillary tangles)、嗜銀顆粒病、皮質基底核退化症(CBD)、瀰漫性神經纖維纏結伴鈣化、與染色體17相關之額顳葉失智症伴帕金森氏症(FTDP-17)、哈勒沃登-施帕茨病(Hallervorden-Spatz disease)、多系統萎縮(MSA)、C型尼曼匹克症(Niemann-Pick disease type C)、蒼白球-腦橋-黑質退化、匹克氏病(Pick’s disease,PiD)、進行性皮質下神經膠瘤病、進行性核上性麻痺(PSP)、亞急性硬化性泛腦炎、纏結主導型失智症、腦炎後帕金森氏症、肌強直性營養不良、亞急性硬化性泛腦病變、LRRK2突變、慢性創傷性腦病變(CTE)、家族性英國型失智症(familial British dementia)、家族性丹麥型失智症(familial Danish dementia)、其他額顳葉退化、瓜德羅普帕金森氏症(Guadeloupean Parkinsonism)、神經退化伴腦內鐵累積、SLC9A6相關性智力遲鈍、白質tau蛋白病變伴球狀膠質細胞包涵體、癲癇、路易氏體失智症(Lewy body dementia,LBD)、輕度認知損害(MCI)、多發性硬化、帕金森氏病(Parkinson's disease)、HIV相關性失智症、成人發作型糖尿病、老年性心臟類澱粉變性、青光眼、缺血性中風、AD中之精神病及亨庭頓氏病(Huntington's disease)。(Williams等人,Intern. Med. J., 2006, 36, 652-60;Kovacs等人,J Neuropathol Exp Neurol. 2008; 67(10): 963-975;Higuchi等人,Neuropsychopharmacology - 5th Generation of Progress, 2002,部分9,第94章:1339-1354;Hilton等人,Acta Neuropathol. 1995;90(1):101-6;Iqbal等人,Biochimica et Biophysica Acta 1739 (2005) 198- 210;McQuaid等人,Neuropathol Appl Neurobiol. 1994年4月;20(2):103-10;Vossel等人,Lancet Neurol 2017; 16: 311-22;Stephan等人,Molecular Psychiatry (2012) 17, 1056-1076;Anderson等人,Brain (2008), 131, 1736-1748;Savica等人,JAMA Neurol. 2013;70(7):859-866;Brown等人,Molecular Neurodegeneration 2014, 9:40;El Khoury等人,Front. Cell. Neurosci., 2014,第8卷,文章號22: 1-18;Tanskanen等人,Ann. Med. 2008;40(3):232-9;Gupta等人,CAN J OPHTHALMOL,第43卷,第1期,2008: 53-60;Dickson等人,Int J Clin Exp Pathol 2010;3(1):1-23;Fernández-Nogales等人,Nature Medicine, 20, 881-885 (2014);Bi等人,Nature Communications第8卷,文章號:473 (2017);Murray等人,Biol Psychiatry. 2014年4月1日;75(7): 542-552)。Diseases involving tau aggregates are generally classified as tauopathies and include, but are not limited to, Alzheimer's disease (AD), familial AD, PART (primary age-related tauopathy), library Creutzfeldt-Jacob disease, boxer dementia, Down's Syndrome, Gerstmann-Sträussler-Scheinker disease (GSS), inclusion bodies Myositis, prion protein cerebral amyloid angiopathy, traumatic brain injury (TBI), amyotrophic lateral sclerosis (ALS), Parkinsonism-dementia complex of Guam , non-Guamanian motor neuron disease with neurofibrillary tangles, argyrophilic granulopathy, corticobasal degeneration (CBD), diffuse neurofibrillary tangles with calcification, associated with chromosome 17 Frontotemporal dementia with Parkinson's disease (FTDP-17), Hallervorden-Spatz disease (Hallervorden-Spatz disease), multiple system atrophy (MSA), Niemann-Pick disease type C (Niemann- Pick disease type C), pallidum-pontine-substantia nigra degeneration, Pick's disease (PiD), progressive subcortical gliomatosis, progressive supranuclear palsy (PSP), subacute sclerosing panencephalitis inflammation, tangle-dominant dementia, postencephalitic Parkinson's disease, myotonic dystrophy, subacute sclerosing panencephalopathy, LRRK2 mutations, chronic traumatic encephalopathy (CTE), familial British dementia familial British dementia, familial Danish dementia, other frontotemporal lobar degeneration, Guadeloupean Parkinsonism, neurodegeneration with iron accumulation in the brain, SLC9A6 correlation Mental retardation, white matter tauopathy with glial inclusions, epilepsy, Lewy body dementia (LBD), mild cognitive impairment (MCI), multiple sclerosis, Parkinson's disease ), HIV-related dementia, adult-onset diabetes, senile cardiac amyloidosis, glaucoma, ischemic stroke, psychosis in AD, and Huntington's disease. (Williams et al., Intern. Med. J., 2006, 36, 652-60; Kovacs et al., J Neuropathol Exp Neurol. 2008; 67(10): 963-975; Higuchi et al., Neuropsychopharmacology - 5th Generation of Progress , 2002, Part 9, Chapter 94:1339-1354; Hilton et al., Acta Neuropathol. 1995;90(1):101-6; Iqbal et al., Biochimica et Biophysica Acta 1739 (2005) 198-210; McQuaid et al. Humans, Neuropathol Appl Neurobiol. 1994 Apr;20(2):103-10; Vossel et al., Lancet Neurol 2017; 16: 311-22; Stephan et al., Molecular Psychiatry (2012) 17, 1056-1076; Anderson et al., Brain (2008), 131, 1736-1748; Savica et al., JAMA Neurol. 2013;70(7):859-866; Brown et al., Molecular Neurodegeneration 2014, 9:40; El Khoury et al., Front . Cell. Neurosci., 2014, Vol. 8, Article No. 22: 1-18; Tanskanen et al., Ann. Med. 2008;40(3):232-9; Gupta et al., CAN J OPHTHALMOL, Vol. 43 , Issue 1, 2008: 53-60; Dickson et al., Int J Clin Exp Pathol 2010;3(1):1-23; Fernández-Nogales et al., Nature Medicine, 20, 881-885 (2014); Bi et al., Nature Communications Volume 8, Article Number: 473 (2017); Murray et al., Biol Psychiatry. 2014 April 1; 75(7): 542-552).

在2017年用於治療阿茲海默氏病之臨床試驗中之所有藥劑中,靶向Tau之藥劑極稀缺且僅佔II期臨床試驗之8% (Cummings等人,Alzheimer’s & Dementia: Translational Research & Clinical Interventions 3 (2017) 367-384)。目前靶向Tau蛋白之治療方法主要包含基於抗體之方法,其中主要限制為僅靶向細胞外Tau。在使用小分子之方法中,已開發出若干種Tau激酶抑制劑,儘管就毒性及特異性而言極具有挑戰性。然而,目前僅一種激酶抑制劑(即尼羅替尼(Nilotinib))在臨床試驗中進行測試。最後,在Tau聚集抑制劑中,僅一種LMTX目前處於臨床試驗中(Cummings等人,2017)。儘管近年來,基於Tau之治療已成為日益關注之焦點,但業內仍對於靶向已知或假定引起tau蛋白病變之病理性Tau構形異構物之其他治療劑存在較大需求。Of all agents in clinical trials for the treatment of Alzheimer's disease in 2017, agents targeting tau were extremely scarce and accounted for only 8% of phase II clinical trials (Cummings et al., Alzheimer's & Dementia: Translational Research & Clinical Interventions 3 (2017) 367-384). Current treatments targeting Tau protein mainly include antibody-based methods, which are mainly limited to targeting only extracellular Tau. Using a small molecule approach, several Tau kinase inhibitors have been developed, although they are extremely challenging in terms of toxicity and specificity. However, only one kinase inhibitor, Nilotinib, is currently being tested in clinical trials. Finally, among Tau aggregation inhibitors, only one, LMTX, is currently in clinical trials (Cummings et al., 2017). Although Tau-based therapies have become the focus of increasing attention in recent years, there is still a great need in the industry for other therapeutic agents targeting pathological Tau conformational isomers known or presumed to cause tauopathies.

WO2011/128455係關於適於治療與類澱粉蛋白或類澱粉樣蛋白有關的病症之特定化合物。WO2011/128455 relates to specific compounds suitable for the treatment of amyloid or amyloid-related conditions.

WO2010/080253係關於二吡啶基-吡咯衍生物化合物,其可用於治療適於蛋白激酶信號轉導抑制、調控及/或調節之疾病。WO2010/080253 relates to dipyridyl-pyrrole derivative compounds which can be used to treat diseases suitable for inhibition, regulation and/or modulation of protein kinase signal transduction.

本發明之目標係提供可用於治療、改善或預防與Tau蛋白聚集體有關的一組病症及異常之化合物,該等病症及異常包括(但不限於) NFT,例如阿茲海默氏病(AD)。此外,業內需要可用作治療劑之化合物,其用於:(a) 藉由識別聚集之Tau且使Tau解聚(例如藉由改變Tau聚集體分子構形)來減少Tau聚集體/NFT,及/或(b) 防止Tau聚集體之形成,及/或(c) 細胞內干擾Tau聚集體,及/或(d) 降低活體內Tau錯誤摺疊及過度磷酸化及/或(f) 減少神經發炎標記物。本發明者已驚訝地發現,該等目標可由如下文中所闡述之式(I)化合物來達成。It is an object of the present invention to provide compounds that can be used to treat, ameliorate or prevent a group of disorders and abnormalities associated with Tau protein aggregates, including (but not limited to) NFT, such as Alzheimer's disease (AD). ). Additionally, there is a need in the industry for compounds that can be used as therapeutic agents for: (a) reducing Tau aggregates/NFTs by recognizing aggregated Tau and depolymerizing Tau (e.g., by changing the molecular conformation of the Tau aggregates), and/or (b) prevent the formation of Tau aggregates, and/or (c) interfere with Tau aggregates intracellularly, and/or (d) reduce Tau misfolding and hyperphosphorylation in vivo and/or (f) reduce neuronal Inflammation markers. The inventors have surprisingly found that these objectives can be achieved by compounds of formula (I) as set out below.

該等式(I)化合物(a) 展示藉由識別聚集之Tau且使Tau解聚(例如藉由改變Tau聚集體分子構形)來減少Tau聚集體之高能力,及/或(b) 防止Tau聚集體之形成,及/或(c) 細胞內干擾Tau聚集體,及/或(d) 降低活體內Tau錯誤摺疊及過度磷酸化及/或(f) 減少神經發炎標記物。儘管不希望受限於理論,但假定式(I)化合物抑制Tau聚集或使預先形成之Tau聚集體解聚,包括在細胞內存在時。由於其獨特之設計特徵,該等化合物展示諸如適當親脂性及分子量、腦攝取及藥物動力學、細胞滲透性、溶解性及代謝穩定性等性質,以成為用於治療、改善或預防tau蛋白病變之成功藥劑。The compounds of formula (I) (a) exhibit a high ability to reduce Tau aggregates by recognizing aggregated Tau and deaggregating Tau (e.g., by changing the molecular configuration of the Tau aggregates), and/or (b) preventing Formation of Tau aggregates, and/or (c) intracellular interference with Tau aggregates, and/or (d) reduction of Tau misfolding and hyperphosphorylation in vivo and/or (f) reduction of neuroinflammatory markers. Although not wishing to be bound by theory, it is postulated that compounds of formula (I) inhibit Tau aggregation or deaggregate preformed Tau aggregates, including when present within cells. Due to their unique design features, these compounds exhibit properties such as appropriate lipophilicity and molecular weight, brain uptake and pharmacokinetics, cell permeability, solubility and metabolic stability, making them useful candidates for the treatment, amelioration or prevention of tauopathies. The potion of success.

經由組織病理學分析以及經由活體內Tau PET成像二者,已顯示Tau NFT病灶之累積與AD中之認知缺陷充分相關。本發明之化合物可防止Tau聚集體之形成或使預先存在之Tau聚集體解聚,且因此可預期預防或降低AD中之相關認知缺陷。The accumulation of Tau NFT lesions has been shown to be well correlated with cognitive deficits in AD, both through histopathological analysis and through in vivo Tau PET imaging. Compounds of the present invention may prevent the formation of Tau aggregates or deaggregate pre-existing Tau aggregates, and thus may be expected to prevent or reduce associated cognitive deficits in AD.

超微結構分析已顯示,Tau包涵體係由成對螺旋絲(PHF)或直絲(SF)構成。高解析度結構分析已顯示,該等絲係由包含Tau之胺基酸306-378之核心區構成,其採取交叉β/β-螺旋結構。本發明之化合物可識別聚集之Tau且使Tau解聚(例如藉由改變Tau聚集體分子構形),且因此可預期促進Tau清除。Ultrastructural analysis has shown that the Tau inclusion system consists of paired helical filaments (PHF) or straight filaments (SF). High-resolution structural analysis has shown that these silk systems are composed of a core region containing amino acids 306-378 of Tau, which adopts a crossed β/β-helical structure. Compounds of the present invention can recognize aggregated Tau and deaggregate Tau (eg, by changing the molecular conformation of Tau aggregates), and thus are expected to promote Tau clearance.

另外,已顯示,Tau能夠自細胞至細胞傳播且某些形式之Tau (作為種子起作用)能夠誘導健康細胞內天然Tau蛋白之結構變化以經歷錯誤摺疊及聚集。認為聚集之Tau負責播種(seeding)且因此導致Tau病理學擴散。本發明之化合物可細胞內干擾聚集之Tau且因此可預期降低Tau病理學擴散並最終預防或降低AD中之相關認知缺陷。Additionally, it has been shown that Tau can spread from cell to cell and that certain forms of Tau (which act as seeds) can induce structural changes in the native Tau protein within healthy cells to undergo misfolding and aggregation. Aggregated Tau is thought to be responsible for seeding and thus causing the spread of Tau pathology. Compounds of the present invention may interfere with aggregated Tau intracellularly and thus may be expected to reduce the spread of Tau pathology and ultimately prevent or reduce associated cognitive deficits in AD.

Tau聚集級聯起始於Tau錯誤摺疊及過度磷酸化。據信該等事件係在細胞內Tau神經元包涵體之形成之前,且因此係在Tau病理學之確立及擴散之前。本發明之化合物可降低活體內Tau錯誤摺疊及過度磷酸化,且因此可預期有益於治療、改善或預防與Tau病理學有關的疾病。The Tau aggregation cascade begins with Tau misfolding and hyperphosphorylation. These events are believed to precede the formation of intracellular Tau neuronal inclusions and, therefore, the establishment and spread of Tau pathology. The compounds of the present invention can reduce Tau misfolding and hyperphosphorylation in vivo, and thus are expected to be useful in the treatment, amelioration or prevention of diseases associated with Tau pathology.

最後,現已充分確立Tau病理學與神經發炎之間的關聯。神經發炎係已在早期AD階段中之關鍵事件,且據信係觸發PHF中Tau聚集之原因之一。此外,一旦腦中充分確立Tau病理學,則若干種tau蛋白病變小鼠模型顯示顯著之神經發炎,此指示Tau病理學亦可誘導神經發炎過程。該兩個發現指示,Tau病理學及神經發炎在正反饋迴路中相關聯。在Tau病理學中,本發明之化合物減少神經發炎標記物。Finally, the link between Tau pathology and neuroinflammation is now well established. Neuroinflammation is a critical event already in the early stages of AD and is believed to be one of the triggers for Tau accumulation in PHF. Furthermore, once Tau pathology is well established in the brain, several mouse models of tauopathy display significant neuroinflammation, indicating that Tau pathology can also induce neuroinflammatory processes. These two findings indicate that tau pathology and neuroinflammation are linked in a positive feedback loop. In Tau pathology, compounds of the invention reduce markers of neuroinflammation.

本發明揭示具有以下能力之新穎式(I)化合物:(a) 減少Tau聚集體、識別聚集之Tau且使Tau解聚(例如藉由改變Tau聚集體分子構形),及/或(b) 防止Tau聚集體之形成,及/或(c) 細胞內干擾Tau聚集體,及/或(d) 降低活體內Tau錯誤摺疊及過度磷酸化及/或(f) 減少神經發炎標記物。本發明提供使用式(I)化合物或其醫藥組合物治療與Tau蛋白聚集體有關的病症及異常之方法,該等病症及異常包括(但不限於) NFT。本發明進一步提供醫藥組合物,其包含式(I)化合物及醫藥上可接受之載劑或賦形劑。The present invention discloses novel compounds of formula (I) that have the ability to: (a) reduce Tau aggregates, recognize aggregated Tau, and deaggregate Tau (e.g., by changing the molecular configuration of Tau aggregates), and/or (b) Prevent the formation of Tau aggregates, and/or (c) interfere with Tau aggregates within cells, and/or (d) reduce Tau misfolding and hyperphosphorylation in vivo and/or (f) reduce neuroinflammatory markers. The present invention provides methods of using compounds of formula (I) or pharmaceutical compositions thereof to treat conditions and abnormalities associated with Tau protein aggregates, including (but not limited to) NFTs. The present invention further provides pharmaceutical compositions comprising a compound of formula (I) and a pharmaceutically acceptable carrier or excipient.

本發明匯總於以下條目中: 1.     一種式(I)化合物, 及其所有立體異構物、外消旋混合物、互變異構物、醫藥上可接受之鹽、前藥、水合物、溶劑合物及多形體; 其中A 係選自由以下組成之群: ,其中 可在任一可用位置處連接至Q,其中 經一或多個取代基Rj 取代,且其中可視情況經一或多個取代基R 取代;B 係選自由O及NRa 組成之群;EV 獨立地選自由以下組成之群:N、NR5 、O及S;G 係選自由以下組成之群:苯環、嘧啶環及吡啶環;J 係選自由O及N-R1 組成之群;Q 係選自由N及C-R1 組成之群;Y 係選自由CZ及N組成之群,條件係當Y 係N且Y1 Y2 Y3 係CZ時,B 係N-烷基或O;Y1 係選自由CZ及N組成之群;Y2 係選自由CZ及N組成之群;Y3 係選自由CZ及N組成之群;Z 獨立地選自由以下組成之群:H、鹵素、O-烷基、烷基及CN;R 獨立地選自由及-NR3 R4 組成之群;Ra 係選自由H及烷基組成之群;Rb Rc Rd Re Rf Rg 獨立地選自由H及烷基組成之群,或Rb Rc Rd Re Rf Rg 中之任兩者可連結以形成3員至8員環;Rj 獨立地選自由以下組成之群:-鹵素、-O-烷基、-CF3 、-CN、-NR3 R4,其中含C1-2 碳原子之橋可存在於a碳原子與c或d碳原子之間或其中含C1-2 碳原子之橋可存在於b碳原子與c或d碳原子之間;R1 係選自由H及烷基組成之群;R2 獨立地選自由烷基、F及=O組成之群,其中該烷基可視情況經鹵素、-OH或-O-烷基取代,且其中若兩個R2 係孿位的,則其可連結以形成3員至6員環;R3 R4 獨立地選自由H及烷基組成之群,其中該烷基可視情況經鹵素、-OH或-O-烷基取代;R5 係選自由H及烷基組成之群;n 係0、1、2、3或4。rs 獨立地係0、1、2或3;且tu 獨立地係1、2或3。 2.     如條目1之化合物,其係式(Ia)化合物: 其中ABRb Rc Rd Re Rf Rg YZ 係如條目1中所定義。 3.     如條目1之化合物,其係式(Ib)化合物: 其中ABRb Rc Rd Re Rf Rg Z 係如條目1中所定義。 4.     如條目1至3中任一條目之化合物,其中A,其中可在任一可用位置處連接至Q或N,其中經一或多個取代基Rj 取代,且其中可視情況經一或多個取代基R 取代。 5.     如條目1至3中任一條目之化合物,其中A,其中可在任一可用位置處連接至Q或N,且其中可視情況經一或多個取代基R 取代。 6.     如條目1至5中任一條目之化合物,其係式(Ic)化合物: 其中ERVZ 係如條目1中所定義。 7.     如條目1至4中任一條目之化合物,其係式(Id)化合物: 其中Ra Rj Z 係如條目1中所定義且p 係1或2。 8.     如條目1之化合物,其中該化合物係選自由以下組成之群: 。 9.     一種醫藥組合物,其包含如條目1至8中任一條目之化合物及視情況醫藥上可接受之載劑或賦形劑。 10.   如條目1至8中任一條目之化合物,其用作藥劑。 11.    一種式(I)化合物, 及其所有立體異構物、外消旋混合物、互變異構物、醫藥上可接受之鹽、前藥、水合物、溶劑合物及多形體; 其用於治療、改善或預防與Tau蛋白聚集體有關的病症或異常, 其中A 係選自由以下組成之群: ,其中 可在任一可用位置處連接至Q,其中 經一或多個取代基Rj 取代,且其中可視情況經一或多個取代基R 取代;B 係選自由O及NRa 組成之群;EV 獨立地選自由以下組成之群:N、NR5 、O及S;G 係選自由以下組成之群:苯環、嘧啶環及吡啶環;J 係選自由O及N-R1 組成之群;Q 係選自由N及C-R1 組成之群;Y 係選自由CZ及N組成之群;Y1 係選自由CZ及N組成之群;Y2 係選自由CZ及N組成之群;Y3 係選自由CZ及N組成之群;Z 獨立地選自由以下組成之群:H、鹵素、O-烷基、烷基及CN;R 獨立地選自由及-NR3 R4 組成之群;Ra 係選自由H及烷基組成之群;Rb Rc Rd Re Rf Rg 獨立地選自由H及烷基組成之群,或Rb Rc Rd Re Rf Rg 中之任兩者可連結以形成3員至8員環;Rj 獨立地選自由以下組成之群:-鹵素、-O-烷基、-CF3 、-CN、-NR3 R4,其中含C1-2 碳原子之橋可存在於a碳原子與c或d碳原子之間或其中含C1-2 碳原子之橋可存在於b碳原子與c或d碳原子之間;R1 係選自由H及烷基組成之群;R2 獨立地選自由烷基、F及=O組成之群,其中該烷基可視情況經鹵素、-OH或-O-烷基取代,且其中若兩個R2 係孿位的,則其可連結以形成3員至6員環;R3 R4 獨立地選自由H及烷基組成之群,其中該烷基可視情況經鹵素、-OH或-O-烷基取代;R5 係選自由H及烷基組成之群;n 係0、1、2、3或4;rs 獨立地係0、1、2或3;且tu 獨立地係1、2或3。 12.   如條目11所用之化合物,其中該式(I)化合物係如條目1至8中任一條目中所定義。 13.   如條目1至8或11中任一條目之化合物,其用於降低Tau聚集。 14.   如條目1至8或11中任一條目之化合物,其用於防止Tau聚集體之形成及/或用於抑制Tau聚集。 15.   如條目1至8或11中任一條目之化合物,其用於細胞內干擾Tau聚集體。 16.   如條目1至8或11中任一條目之化合物,其用於降低活體內Tau錯誤摺疊及過度磷酸化。 17.   如條目1至8或11中任一條目之化合物,其用於減少神經發炎標記物。 18.   一種治療、預防或改善與Tau蛋白聚集體有關的病症或異常之方法,該方法包含向有需要之個體投與有效量之如條目1至8或11中任一條目之化合物。 19.   一種降低Tau聚集之方法,該方法包含向有需要之個體投與有效量之如條目1至8或11中任一條目之化合物。 20.   一種防止Tau聚集體之形成及/或抑制Tau聚集之方法,該方法包含向有需要之個體投與有效量之如條目1至8或11中任一條目之化合物。 21.   一種細胞內干擾Tau聚集體之方法,該方法包含向有需要之個體投與有效量之如條目1至8或11中任一條目之化合物。 22.   一種降低活體內Tau錯誤摺疊及過度磷酸化之方法,該方法包含向有需要之個體投與有效量之如條目1至8或11中任一條目之化合物。 23.   一種減少神經發炎標記物之方法,該方法包含向有需要之個體投與有效量之如條目1至8或11中任一條目之化合物。 24.   一種如條目1至8或11中任一條目之化合物之用途,其用於製造用於治療、預防或改善與Tau蛋白聚集體有關的病症或異常之藥劑。 25.   一種如條目1至8或11中任一條目之化合物之用途,其用於製造用於降低Tau聚集之藥劑。 26.   一種如條目1至8或11中任一條目之化合物之用途,其用於製造用於防止Tau聚集體之形成及/或用於抑制Tau聚集之藥劑。 27.   一種如條目1至8或11中任一條目之化合物之用途,其用於製造用於細胞內干擾Tau聚集體之藥劑。 28.   一種如條目1至8或11中任一條目之化合物之用途,其用於製造用於降低活體內Tau錯誤摺疊及過度磷酸化之藥劑。 29.   一種如條目1至8或11中任一條目之化合物之用途,其用於製造用於減少神經發炎標記物之藥劑。 29.   一種混合物,其包含如條目1至8中任一條目之化合物及至少一種其他生物活性化合物,該至少一種其他生物活性化合物係選自與如條目1至8中任一條目之化合物不同之治療劑, 其中該混合物進一步包含醫藥上可接受之載劑、稀釋劑及賦形劑中之至少一者。 30.   如條目29之混合物,其中該其他生物活性化合物係用於治療類澱粉變性之化合物。 31.   如條目29或30之混合物,其中該化合物及/或該其他生物活性化合物係以治療有效量存在。 32.   如條目29至31中任一條目之混合物,其中該其他生物活性化合物係選自由以下組成之群:抗氧化壓力之化合物;抗細胞凋亡化合物;金屬螯合劑;DNA修復抑制劑,例如哌侖西平(pirenzepine)及代謝物;3-胺基-1-丙烷磺酸(3APS);1,3-丙烷二磺酸鹽(1,3PDS);α-分泌酶活化劑;β-分泌酶及γ-分泌酶抑制劑;肝醣合酶激酶3抑制劑;O連接之N-乙醯葡糖胺水解酶(O-glcnacase,OGA)抑制劑;神經傳遞質;β褶板碎裂劑;類澱粉β清除/消除細胞組分引誘劑;N末端截短之類澱粉β (包括焦麩胺酸化之類澱粉β 3-42 )之抑制劑,抗發炎分子,或膽鹼酯酶抑制劑(ChEI),例如塔克寧(tacrine)、利凡斯的明(rivastigmine)、多奈派齊(donepezil)及/或加蘭他敏(galantamine);M1激動劑;其他藥物,包括任何類澱粉或Tau改質藥物及營養補充劑;抗體,包括任何功能等效抗體或其功能部分;或疫苗。 33.   如條目32之混合物,其中該其他生物活性化合物係膽鹼酯酶抑制劑(ChEI)。 34.   如條目之混合物32,其中該其他生物活性化合物係選自由以下組成之群:塔克寧、利凡斯的明、多奈派齊、加蘭他敏、菸鹼酸及美金剛(memantine)。 35.   如條目32之混合物,其中該其他生物活性化合物係抗體、具體而言單株抗體,包括任何功能等效抗體或其功能部分。 36.   如條目29至35中任一條目之混合物,其中該化合物及/或該其他生物活性化合物係以治療有效量存在。 37.   如條目11所用之化合物、如條目18之方法或如條目24之用途,其中該病症係選自阿茲海默氏病(AD)、家族性AD、原發性年齡相關性Tau蛋白病變(PART)、庫賈氏病、拳擊手型失智症、唐氏症候群、傑茨曼-斯脫司勒-史茵克病(GSS)、包涵體肌炎、普里昂蛋白腦類澱粉血管病變、創傷性腦損傷(TBI)、肌肉萎縮性脊髓側索硬化症(ALS)、關島帕金森氏症-失智複合症、非關島運動神經元病伴神經纖維纏結、嗜銀顆粒病、皮質基底核退化症(CBD)、瀰漫性神經纖維纏結伴鈣化、與染色體17相關之額顳葉失智症伴帕金森氏症(FTDP-17)、哈勒沃登-施帕茨病、多系統萎縮(MSA)、C型尼曼匹克症、蒼白球-腦橋-黑質退化、匹克氏病(PiD)、進行性皮質下神經膠瘤病、進行性核上性麻痺(PSP)、亞急性硬化性泛腦炎、纏結主導型失智症、腦炎後帕金森氏症、肌強直性營養不良、亞急性硬化性泛腦病變、LRRK2突變、慢性創傷性腦病變(CTE)、家族性英國型失智症、家族性丹麥型失智症、其他額顳葉退化、瓜德羅普帕金森氏症、神經退化伴腦內鐵累積、SLC9A6相關性智力遲鈍、白質tau蛋白病變伴球狀膠質細胞包涵體、癲癇、路易氏體失智症(LBD)、輕度認知損害(MCI)、多發性硬化、帕金森氏病、HIV相關性失智症、成人發作型糖尿病、老年性心臟類澱粉變性、青光眼、缺血性中風、AD中之精神病及亨庭頓氏病,較佳地阿茲海默氏病(AD)、皮質基底核退化症(CBD)、匹克氏病(PiD)及進行性核上性麻痺(PSP)。 38.   一種如條目1至8或11中任一條目之化合物之用途,其用作分析參考或活體外篩選工具。The invention is summarized under the following entries: 1. A compound of formula (I), and all stereoisomers, racemic mixtures, tautomers, pharmaceutically acceptable salts, prodrugs, hydrates, solvates and polymorphs thereof; wherein A is selected from the group consisting of: and ,in and Can be connected to Q at any available location, where and Substituted with one or more substituents Rj , and wherein and Optionally substituted by one or more substituents R ; B is selected from the group consisting of O and NR a ; E and V are independently selected from the group consisting of: N, NR 5 , O and S; G is selected from the following The group consisting of: benzene ring, pyrimidine ring and pyridine ring; J is selected from the group consisting of O and NR 1 ; Q is selected from the group consisting of N and CR 1 ; Y is selected from the group consisting of CZ and N, the condition is When Y is N and Y 1 , Y 2 and Y 3 are CZ, B is N-alkyl or O; Y 1 is selected from the group consisting of CZ and N; Y 2 is selected from the group consisting of CZ and N; Y 3 is selected from the group consisting of CZ and N; Z is independently selected from the group consisting of: H, halogen, O-alkyl, alkyl and CN; R is independently selected from the group consisting of and -NR 3 R 4 ; R a is selected from the group consisting of H and alkyl groups; R b , R c , R d , R e , R f and R g are independently selected from the group consisting of H and alkyl groups group, or any two of R b , R c , R d , Re , R f and R g may be linked to form a 3- to 8-membered ring; R j is independently selected from the group consisting of: - halogen, -O-alkyl, -CF 3 , -CN, -NR 3 R 4 , and , where a bridge containing C 1-2 carbon atoms can exist between a carbon atom and c or d carbon atom or where a bridge containing C 1-2 carbon atoms can exist between b carbon atom and c or d carbon atom ; R 1 is selected from the group consisting of H and alkyl; R 2 is independently selected from the group consisting of alkyl, F and =O, wherein the alkyl is optionally substituted by halogen, -OH or -O-alkyl, And if two R 2 are twin-positioned, they can be connected to form a 3- to 6-membered ring; R 3 and R 4 are independently selected from the group consisting of H and an alkyl group, wherein the alkyl group may be optionally treated with halogen , -OH or -O-alkyl substitution; R 5 is selected from the group consisting of H and alkyl; n is 0, 1, 2, 3 or 4. r and s are independently 0, 1, 2, or 3; and t and u are independently 1, 2, or 3. 2. For example, the compound of item 1 is a compound of formula (Ia): Among them, A , B , Rb , Rc , Rd , Re , Rf , Rg , Y and Z are as defined in item 1. 3. For example, the compound of item 1 is a compound of formula (Ib): Where A , B , Rb , Rc , Rd , Re , Rf , Rg and Z are as defined in item 1. 4. A compound as in any one of items 1 to 3, wherein A is and ,in and Can be connected to Q or N at any available location, where Substituted with one or more substituents Rj , and wherein Optionally substituted with one or more substituents R. 5. A compound as in any one of items 1 to 3, wherein A is ,in Can be connected to Q or N at any available location, and where Optionally substituted with one or more substituents R. 6. A compound of any one of items 1 to 5, which is a compound of formula (Ic): Where E , R , V and Z are as defined in item 1. 7. A compound of any one of items 1 to 4, which is a compound of formula (Id): where R a , R j and Z are as defined in item 1 and p is 1 or 2. 8. The compound of item 1, wherein the compound is selected from the group consisting of: . 9. A pharmaceutical composition comprising a compound as in any one of items 1 to 8 and optionally a pharmaceutically acceptable carrier or excipient. 10. A compound according to any one of items 1 to 8 for use as a medicament. 11. A compound of formula (I), and all stereoisomers, racemic mixtures, tautomers, pharmaceutically acceptable salts, prodrugs, hydrates, solvates and polymorphs thereof; for the treatment, improvement or prevention of aggregation with Tau protein A disease or abnormality related to the body, wherein series A is selected from the group consisting of: and ,in and Can be connected to Q at any available location, where and Substituted with one or more substituents Rj , and wherein and Optionally substituted by one or more substituents R ; B is selected from the group consisting of O and NR a ; E and V are independently selected from the group consisting of: N, NR 5 , O and S; G is selected from the following The group consisting of: benzene ring, pyrimidine ring and pyridine ring; J is selected from the group consisting of O and NR 1 ; Q is selected from the group consisting of N and CR 1 ; Y is selected from the group consisting of CZ and N; Y 1 Y 2 is selected from the group consisting of CZ and N; Y 3 is selected from the group consisting of CZ and N; Z is independently selected from the group consisting of: H, halogen, O- Alkyl, alkyl and CN; R is independently selected from and -NR 3 R 4 ; R a is selected from the group consisting of H and alkyl groups; R b , R c , R d , R e , R f and R g are independently selected from the group consisting of H and alkyl groups group, or any two of R b , R c , R d , Re , R f and R g may be linked to form a 3- to 8-membered ring; R j is independently selected from the group consisting of: - halogen, -O-alkyl, -CF 3 , -CN, -NR 3 R 4 , and , where a bridge containing C 1-2 carbon atoms can exist between a carbon atom and c or d carbon atom or where a bridge containing C 1-2 carbon atoms can exist between b carbon atom and c or d carbon atom ; R 1 is selected from the group consisting of H and alkyl; R 2 is independently selected from the group consisting of alkyl, F and =O, wherein the alkyl is optionally substituted by halogen, -OH or -O-alkyl, And if two R 2 are twin-positioned, they can be connected to form a 3- to 6-membered ring; R 3 and R 4 are independently selected from the group consisting of H and an alkyl group, wherein the alkyl group may be optionally treated with halogen , -OH or -O-alkyl substitution; R 5 is selected from the group consisting of H and alkyl; n is 0, 1, 2, 3 or 4; r and s are independently 0, 1, 2 or 3; And t and u are independently 1, 2 or 3. 12. A compound as used in entry 11, wherein the compound of formula (I) is as defined in any of entries 1 to 8. 13. A compound according to any one of items 1 to 8 or 11 for reducing Tau aggregation. 14. A compound according to any one of items 1 to 8 or 11 for preventing the formation of Tau aggregates and/or for inhibiting Tau aggregation. 15. A compound according to any one of items 1 to 8 or 11 for intracellular interference with Tau aggregates. 16. A compound according to any one of items 1 to 8 or 11, which is used to reduce Tau misfolding and hyperphosphorylation in vivo. 17. A compound according to any one of items 1 to 8 or 11 for reducing markers of neuroinflammation. 18. A method of treating, preventing or ameliorating a condition or abnormality associated with Tau protein aggregates, the method comprising administering an effective amount of a compound of any one of items 1 to 8 or 11 to an individual in need thereof. 19. A method of reducing Tau aggregation, the method comprising administering to an individual in need thereof an effective amount of a compound of any one of items 1 to 8 or 11. 20. A method of preventing the formation of Tau aggregates and/or inhibiting Tau aggregation, the method comprising administering an effective amount of a compound of any one of items 1 to 8 or 11 to an individual in need thereof. 21. A method of intracellularly interfering with Tau aggregates, the method comprising administering to an individual in need thereof an effective amount of a compound of any one of items 1 to 8 or 11. 22. A method of reducing Tau misfolding and hyperphosphorylation in vivo, the method comprising administering an effective amount of a compound of any one of items 1 to 8 or 11 to an individual in need thereof. 23. A method of reducing markers of neuroinflammation, the method comprising administering to an individual in need thereof an effective amount of a compound of any one of items 1 to 8 or 11. 24. Use of a compound according to any one of items 1 to 8 or 11 for the manufacture of a medicament for the treatment, prevention or amelioration of a condition or abnormality associated with Tau protein aggregates. 25. Use of a compound according to any one of items 1 to 8 or 11 for the manufacture of a medicament for reducing Tau aggregation. 26. Use of a compound according to any one of items 1 to 8 or 11 for the manufacture of a medicament for preventing the formation of Tau aggregates and/or for inhibiting Tau aggregation. 27. Use of a compound according to any one of items 1 to 8 or 11 for the manufacture of a medicament for intracellular interference with Tau aggregates. 28. Use of a compound according to any one of items 1 to 8 or 11 for the manufacture of a medicament for reducing Tau misfolding and hyperphosphorylation in vivo. 29. Use of a compound according to any one of items 1 to 8 or 11 for the manufacture of a medicament for reducing markers of neuroinflammation. 29. A mixture comprising a compound as in any one of items 1 to 8 and at least one other biologically active compound selected from a compound different from any one of items 1 to 8 Therapeutic agent, wherein the mixture further comprises at least one of a pharmaceutically acceptable carrier, diluent and excipient. 30. A mixture as in item 29, wherein the other biologically active compound is a compound used to treat amyloidosis. 31. A mixture as in item 29 or 30, wherein the compound and/or the other biologically active compound is present in a therapeutically effective amount. 32. A mixture as in any one of items 29 to 31, wherein the other biologically active compound is selected from the group consisting of: compounds that resist oxidative stress; anti-apoptotic compounds; metal chelators; DNA repair inhibitors, for example Pirenzepine and metabolites; 3-amino-1-propanesulfonic acid (3APS); 1,3-propanedisulfonate (1,3PDS); α-secretase activator; β-secretase And gamma-secretase inhibitors; glycogen synthase kinase 3 inhibitors; O-linked N-acetyl glucosamine hydrolase (O-glcnacase, OGA) inhibitors; neurotransmitters; β-pleat plate disruptors; Amyloid β scavenging/eliminating cellular component attractant; inhibitor of N-terminally truncated amyloid β-like (including pyroglutamylated amyloid β-like 3-42), anti-inflammatory molecule, or cholinesterase inhibitor ( ChEI), such as tacrine, rivastigmine, donepezil and/or galantamine; M1 agonists; other drugs, including any amyloid or Tau-modified drugs and nutritional supplements; antibodies, including any functionally equivalent antibodies or functional portions thereof; or vaccines. 33. A mixture as in item 32, wherein the other biologically active compound is a cholinesterase inhibitor (ChEI). 34. For example, mixture 32 of item 32, wherein the other biologically active compound is selected from the group consisting of: tacnin, rivastigmine, donepezil, galantamine, nicotinic acid and memantine ). 35. A mixture as in item 32, wherein the other biologically active compound is an antibody, in particular a monoclonal antibody, including any functionally equivalent antibody or functional part thereof. 36. A mixture as in any one of items 29 to 35, wherein the compound and/or the other biologically active compound is present in a therapeutically effective amount. 37. A compound as used in item 11, a method as in item 18 or a use as in item 24, wherein the disorder is selected from the group consisting of Alzheimer's disease (AD), familial AD, primary age-related tauopathy (PART), Creutzfeldt-Jakob disease, boxer's dementia, Down syndrome, Gertzmann-Steusler-Steinke disease (GSS), inclusion body myositis, prion protein cerebral amyloid angiopathy, Traumatic brain injury (TBI), amyotrophic lateral sclerosis (ALS), Guam Parkinson's disease-dementia complex, non-Guam motor neuron disease with neurofibrillary tangles, argyrophilic granulopathy, corticobasal CBD, diffuse neurofibrillary tangles with calcification, frontotemporal dementia associated with chromosome 17 and parkinsonism (FTDP-17), Hallerwarden-Spatz disease, multiple system atrophy (MSA), Niemann-Pick disease type C, pallidum-pontine-nigra degeneration, Pick's disease (PiD), progressive subcortical gliomatosis, progressive supranuclear palsy (PSP), subacute sclerosing Panencephalitis, tangle-dominant dementia, postencephalitic parkinsonism, myotonic dystrophy, subacute sclerosing panencephalopathy, LRRK2 mutations, chronic traumatic encephalopathy (CTE), familial British type Dementia, familial Danish dementia, other frontotemporal degenerations, Guadeloupe Parkinson's disease, neurodegeneration with iron accumulation in the brain, SLC9A6-related mental retardation, white matter tauopathy with glomerulocytes Inclusion bodies, epilepsy, Lewy body dementia (LBD), mild cognitive impairment (MCI), multiple sclerosis, Parkinson's disease, HIV-related dementia, adult-onset diabetes, senile cardiac amyloidosis , glaucoma, ischemic stroke, psychosis in AD and Huntington's disease, preferably Alzheimer's disease (AD), corticobasal degeneration (CBD), Pick's disease (PiD) and progressive Supranuclear palsy (PSP). 38. Use of a compound according to any one of items 1 to 8 or 11 as an analytical reference or as an in vitro screening tool.

定義 在本申請案之含義內,以下定義適用: 「烷基」係指由碳及氫原子組成之飽和直鏈或具支鏈有機部分。適宜烷基之實例具有1至6個碳原子、較佳地1至4個碳原子,且包括甲基、乙基、丙基、異丙基、正丁基、第三丁基及異丁基。烷基可視情況經鹵素(較佳地F)、-OH或-O烷基(較佳地-OMe)取代。 「Hal」或「鹵素」係指F、Cl、Br及I。 「3員至8員環」係指3員、4員、5員、6員、7員或8員環,其中該環中無碳原子、一或多個碳原子已經1或2個(對於3員環)、1、2或3個(對於4員環)、1、2、3或4個(對於5員環)或1、2、3、4或5個(對於6員環)、1、2、3、4、5或6個(對於7員環)或1、2、3、4、5、6或7個(對於8員環)相同或不同之雜原子替代,其中該等雜原子係選自O、N及S。 Definitions Within the meaning of this application, the following definitions apply: "Alkyl" means a saturated linear or branched organic moiety consisting of carbon and hydrogen atoms. Examples of suitable alkyl groups have 1 to 6 carbon atoms, preferably 1 to 4 carbon atoms, and include methyl, ethyl, propyl, isopropyl, n-butyl, tert-butyl and isobutyl . Alkyl groups are optionally substituted with halogen (preferably F), -OH or -O alkyl (preferably -OMe). "Hal" or "halogen" refers to F, Cl, Br and I. "3- to 8-membered ring" means a 3-, 4-, 5-, 6-, 7- or 8-membered ring in which there are no carbon atoms in the ring and one or more carbon atoms have 1 or 2 (for 3-member ring), 1, 2 or 3 (for 4-member ring), 1, 2, 3 or 4 (for 5-member ring) or 1, 2, 3, 4 or 5 (for 6-member ring), 1, 2, 3, 4, 5 or 6 (for a 7-membered ring) or 1, 2, 3, 4, 5, 6 or 7 (for an 8-membered ring) the same or different heteroatoms are substituted, where these The heteroatoms are selected from O, N and S.

具有一或多個光學活性碳之本發明之化合物可作為以下存在:外消旋物及外消旋混合物(包括呈所有比率之混合物)、立體異構物(包括非鏡像異構混合物及個別非鏡像異構物、鏡像異構混合物及單一鏡像異構物、構形異構物之混合物及單一構形異構物)、互變異構物、阻轉異構物及旋轉異構物。所有異構形式均包括在本發明中。本發明中所闡述含有烯烴雙鍵之化合物包括E及Z幾何異構物。本發明中亦包括所有醫藥上可接受之鹽、前藥、多形體、水合物及溶劑合物。Compounds of the present invention having one or more optically active carbons may exist as: racemates and racemic mixtures (including mixtures in all ratios), stereoisomers (including diastereomeric mixtures and individual non-enantiomeric mixtures). Enantiomers, enantiomer mixtures and single enantiomers, mixtures of configuration isomers and single configuration isomers), tautomers, atropisomers and rotamers. All isomeric forms are included in the present invention. Compounds containing olefin double bonds described in this invention include E and Z geometric isomers. Also included in the present invention are all pharmaceutically acceptable salts, prodrugs, polymorphs, hydrates and solvates.

術語「多形體」係指本發明之化合物之各種結晶結構。此可包括(但不限於)晶體形態(及非晶形材料)及所有晶格形式。本發明之鹽可係結晶且可作為一種以上多形體存在。The term "polymorph" refers to the various crystalline structures of the compounds of the invention. This may include, but is not limited to, crystalline forms (and amorphous materials) and all lattice forms. The salts of the present invention may be crystalline and may exist as more than one polymorph.

本發明亦涵蓋鹽之溶劑合物、水合物以及無水形式。溶劑合物中所包括之溶劑並無具體限制且可係任一醫藥上可接受之溶劑。實例包括水及C1-4 醇(例如甲醇或乙醇)。The invention also encompasses solvates, hydrates and anhydrous forms of the salts. The solvent included in the solvate is not specifically limited and can be any pharmaceutically acceptable solvent. Examples include water and C 1-4 alcohols (such as methanol or ethanol).

「醫藥上可接受之鹽」係定義為所揭示化合物之衍生物,其中母體化合物係藉由製備其酸式或鹼式鹽來修飾。醫藥上可接受之鹽之實例包括(但不限於)鹼性殘基(例如胺)之無機酸鹽或有機酸鹽、酸性殘基(例如羧酸)之鹼性鹽或有機鹽及諸如此類。醫藥上可接受之鹽包括自(例如)無毒無機酸或有機酸形成之母體化合物之習用無毒鹽或四級銨鹽。舉例而言,此等習用無毒鹽包括源自無機酸之彼等,該等無機酸例如(但不限於)鹽酸、氫溴酸、硫酸、胺基磺酸、磷酸、硝酸及諸如此類;及自有機酸製備之鹽,該等有機酸例如(但不限於)乙酸、丙酸、琥珀酸、乙醇酸、硬脂酸、乳酸、蘋果酸、酒石酸、檸檬酸、抗壞血酸、巴莫酸、馬來酸、羥基馬來酸、苯乙酸、麩胺酸、苯甲酸、柳酸、磺胺酸、2-乙醯氧基苯甲酸、富馬酸、甲苯磺酸、甲烷磺酸、乙烷二磺酸、草酸、羥乙磺酸及諸如此類。本發明之醫藥上可接受之鹽可藉由習用化學方法自含有鹼性或酸性部分之母體化合物來合成。通常,此等鹽可藉由使該等化合物之游離酸或鹼形式與化學計算量之適當鹼或酸於水或有機溶劑中或於該兩者之混合物中進行反應來製備。有機溶劑包括(但不限於)非水性介質,如醚、乙酸乙酯、乙醇、異丙醇或乙腈。適宜鹽之列表可參見Remington’s Pharmaceutical Sciences,第18版,Mack Publishing Company, Easton, PA, 1990,第1445頁,其揭示內容係以引用的方式併入本文中。"Pharmaceutically acceptable salts" are defined as derivatives of the disclosed compounds wherein the parent compound is modified by preparing acid or base salts thereof. Examples of pharmaceutically acceptable salts include, but are not limited to, inorganic or organic acid salts of basic residues such as amines, basic or organic salts of acidic residues such as carboxylic acids, and the like. Pharmaceutically acceptable salts include the customary non-toxic salts or quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. By way of example, such customary nontoxic salts include those derived from inorganic acids such as, but not limited to, hydrochloric acid, hydrobromic acid, sulfuric acid, sulfamic acid, phosphoric acid, nitric acid, and the like; and from organic acids. Salts prepared from acids such as (but not limited to) acetic acid, propionic acid, succinic acid, glycolic acid, stearic acid, lactic acid, malic acid, tartaric acid, citric acid, ascorbic acid, parmoic acid, maleic acid, Hydroxymaleic acid, phenylacetic acid, glutamic acid, benzoic acid, salicylic acid, sulfamic acid, 2-acetyloxybenzoic acid, fumaric acid, toluenesulfonic acid, methane sulfonic acid, ethane disulfonic acid, oxalic acid, Isethionic acid and the like. The pharmaceutically acceptable salts of the present invention can be synthesized by conventional chemical methods from parent compounds containing basic or acidic moieties. Generally, such salts may be prepared by reacting the free acid or base form of the compounds with a stoichiometric amount of the appropriate base or acid in water or an organic solvent, or in a mixture of the two. Organic solvents include, but are not limited to, non-aqueous media such as ether, ethyl acetate, ethanol, isopropanol or acetonitrile. A list of suitable salts can be found in Remington's Pharmaceutical Sciences, 18th Edition, Mack Publishing Company, Easton, PA, 1990, page 1445, the disclosure of which is incorporated herein by reference.

本發明之化合物亦可以前藥形式來提供,即在活體內代謝為活性代謝物之化合物。如下文在闡述本發明及申請專利範圍中所使用,術語「前藥」意指由於活體內生物轉化而釋放活性母體醫藥之任何共價鍵結之化合物。Goodman及Gilman闡述前藥之參考文獻(The Pharmacological Basis of Therapeutics,第8版,McGraw-Hill, Int. Ed. 1992, 「Biotransformation of Drugs」,第13-15頁)通常係以引用的方式併入本文中。The compounds of the present invention may also be provided in the form of prodrugs, ie, compounds that are metabolized in vivo to active metabolites. As used below in describing and patenting the invention, the term "prodrug" means any covalently bonded compound that releases the active parent drug as a result of biotransformation in vivo. References to Goodman and Gilman describing prodrugs (The Pharmacological Basis of Therapeutics, 8th ed., McGraw-Hill, Int. Ed. 1992, "Biotransformation of Drugs", pp. 13-15) are generally incorporated by reference. in this article.

「醫藥上可接受」係定義為在合理醫學判斷範圍內適於與人類及動物之組織接觸使用而無過度毒性、刺激性、過敏反應或其他問題或併發症且與合理益處/風險比相稱之彼等化合物、材料、組合物及/或劑型。"Pharmaceutically acceptable" is defined as suitable for use in contact with human and animal tissue within the scope of reasonable medical judgment without undue toxicity, irritation, allergic reactions or other problems or complications and proportional to a reasonable benefit/risk ratio Those compounds, materials, compositions and/or dosage forms.

本發明之患者或個體通常係動物、具體而言哺乳動物、更具體而言人類。The patient or individual of the present invention is generally an animal, specifically a mammal, and more specifically a human being.

如本文所使用之「Tau」係指主要發現於神經元中之高度可溶性微管結合蛋白,且包括主要6種同種型、裂解或截短形式及其他經修飾形式,例如源自磷酸化、醣基化、醣化、脯胺醯基異構化、硝化、乙醯化、聚胺化、泛素化、小泛素化(sumoylation)及氧化。"Tau" as used herein refers to the highly soluble microtubule-binding protein found primarily in neurons and includes the main 6 isoforms, cleaved or truncated forms and other modified forms, such as those derived from phosphorylation, sugar Sylation, glycation, prolyl isomerization, nitration, acetylation, polyamination, ubiquitination, small ubiquitination (sumoylation) and oxidation.

「聚集之Tau」係指摺疊成寡聚或聚合結構之Tau肽或蛋白質之聚集單體。"Aggregated Tau" refers to aggregated monomers of Tau peptide or protein that fold into oligomeric or polymeric structures.

如本文所使用之「神經纖維纏結」(NFT)係指含有成對螺旋絲(PHF)及直絲之高度磷酸化Tau蛋白之不溶性聚集體。其存在係AD及稱為tau蛋白病變之其他疾病之標誌。"Neurofibrillary tangles" (NFTs) as used herein refer to insoluble aggregates of highly phosphorylated Tau protein containing paired helical filaments (PHF) and straight filaments. Its presence is a hallmark of AD and other diseases called tauopathies.

如本文所使用之術語「抗體(antibody或antibodies)」係業內公認之術語且應理解係指結合至已知抗原之分子或分子之活性片段,或具體而言係指免疫球蛋白分子及免疫球蛋白分子之抗原結合部分。特定而言,本發明之混合物包括本發明之化合物及抗Tau或抗Abeta抗體。The term "antibody or antibodies" as used herein is an industry-recognized term and should be understood to refer to a molecule or active fragment of a molecule that binds to a known antigen, or specifically to immunoglobulin molecules and immunoglobulins. The antigen-binding portion of a protein molecule. In particular, mixtures of the invention include a compound of the invention and an anti-Tau or anti-Abeta antibody.

如本文所使用之術語「功能等效抗體或其功能部分」應理解為係指結合至已知抗原之等效分子或分子之活性片段,或具體而言係指免疫球蛋白分子及免疫球蛋白分子之抗原結合部分,且與其所源自之抗體具有基本上相同之(生物)活性。The term "functionally equivalent antibody or functional portion thereof" as used herein shall be understood to refer to an equivalent molecule or active fragment of a molecule that binds to a known antigen, or in particular to immunoglobulin molecules and immunoglobulins The antigen-binding portion of a molecule that has substantially the same (biological) activity as the antibody from which it is derived.

本發明混合物中所報導之「疫苗(vaccine或vaccines)」特定而言係Tau或Abeta疫苗。The "vaccines" reported in the mixtures of the invention are in particular Tau or Abeta vaccines.

除非另有說明,否則「定義」部分中所給出之定義及較佳定義適用於下文所闡述之所有實施例。Unless otherwise stated, the definitions and preferred definitions given in the "Definitions" section apply to all embodiments set forth below.

下文將闡述本發明之化合物。應理解,亦設想以下定義之所有可能組合。The compounds of the present invention are described below. It is understood that all possible combinations of the following definitions are also contemplated.

在一個實施例中,本發明係關於式(I)化合物: In one embodiment, the invention relates to compounds of formula (I):

及其所有立體異構物、外消旋混合物、互變異構物、醫藥上可接受之鹽、前藥、水合物、溶劑合物及多形體。and all stereoisomers, racemic mixtures, tautomers, pharmaceutically acceptable salts, prodrugs, hydrates, solvates and polymorphs thereof.

式(I)化合物之較佳實施例係 The preferred embodiment of the compound of formula (I) is

式(I)化合物之另一較佳實施例係 Another preferred embodiment of the compound of formula (I) is

式(I)化合物之另一較佳實施例係 Another preferred embodiment of the compound of formula (I) is

在另一較佳實施例中,式(I)化合物係 In another preferred embodiment, the compound of formula (I) is

甚至更佳地,式(I)化合物係 Even more preferably, the compound of formula (I) is or

甚至更佳地, Even better,

A 之以下定義適用於式(I)化合物及其較佳實施例。The following definition of A applies to the compounds of formula (I) and their preferred embodiments.

A 係選自由以下組成之群: ,其中G 係選自苯環、嘧啶環及吡啶環。因此涵蓋以下較佳實施例: 。 且因此,涵蓋以下實施例: 。在較佳實施例中,係選自。 可設想以下各環:。較佳實施例包括 。在該等結構式中,環G 未完全顯示,但僅由部分鍵指示。Series A is selected from the group consisting of: and , where G is selected from benzene ring, pyrimidine ring and pyridine ring. therefore Covers the following preferred embodiments: and . and therefore, The following examples are covered: and . In a preferred embodiment, Department selected from and . The following rings can be imagined: and . Preferred embodiments include and . In these structural formulas, ring G is not shown completely, but only consists of part of the bond instruct.

在一個較佳實施例中,A 係選自In a preferred embodiment, Series A is selected from and .

在另一較佳實施例中,A。在一個較佳實施例中,AIn another preferred embodiment, A series and . In a preferred embodiment, A series .

在更佳實施例中,A 係選自。在更佳實施例中,A 係選自In a more preferred embodiment, Series A is selected from and . In a more preferred embodiment, Series A is selected from and .

在另一較佳實施例中,A 係選自 In another preferred embodiment, A is selected from and .

在甚至更佳實施例中,AIn an even more preferred embodiment, Series A .

A 之上文定義及其較佳實施例中, 可在任一可用位置處連接至Q。In the above definition of A and its preferred embodiments, and Connect to Q at any available location.

A 之上文定義及其較佳實施例中, 經一或多個取代基Rj 取代。In the above definition of A and its preferred embodiments, and Substituted with one or more substituents Rj .

A 之上文定義及其較佳實施例中, 可視情況經一或多個取代基R 取代。In the above definition of A and its preferred embodiments, and Optionally substituted with one or more substituents R.

若適當,以下定義適用於式(I)及其較佳實施例。Where appropriate, the following definitions apply to formula (I) and its preferred embodiments.

B 係選自由O及NRa 組成之群。更佳地,B 係NRa 、最佳地NH。 System B is selected from the group consisting of O and NR a . More preferably, B is NR a , most preferably NH.

EV 獨立地選自由以下組成之群:N、NR5 、O及S。由於含有EV 之環係不飽和的,因此EV 中之至少一者係N。 E and V are independently selected from the group consisting of: N, NR 5 , O and S. Since the ring system containing E and V is unsaturated, at least one of E and V is N.

J 係選自由O及N-R1 組成之群,更佳為O。 J is selected from the group consisting of O and NR 1 , preferably O.

Q 係選自由N及C-R1 組成之群。較佳地,Q 係選自由N及CH組成之群,更佳地Q 係N。 Q is selected from the group consisting of N and CR 1 . Preferably, Q is selected from the group consisting of N and CH, and more preferably, Q is N.

Y 係選自由CZ及N組成之群。更佳地,Y 係選自由CH及N組成之群。甚至更佳地,Y 係CH。在一個實施例中,若Y 係N且Y1 Y2 Y3 係CZ,則B 係N-烷基或O,較佳地B 係N-烷基。 Y is selected from the group consisting of CZ and N. More preferably, Y is selected from the group consisting of CH and N. Even better, Y is CH. In one embodiment, if Y is N and Y 1 , Y 2 and Y 3 are CZ, then B is N-alkyl or O, preferably B is N-alkyl.

Y1 係選自由CZ及N組成之群。較佳地,Y1 係CZ。 Y 1 is selected from the group consisting of CZ and N. Preferably, Y 1 is CZ.

Y2 係選自由CZ及N組成之群。較佳地,Y2 係CZ。 Y 2 is selected from the group consisting of CZ and N. Preferably, Y 2 is CZ.

Y3 係選自由CZ及N組成之群。較佳地,Y3 係CZ。 Y 3 is selected from the group consisting of CZ and N. Preferably, Y 3 is CZ.

Z 獨立地選自由以下組成之群:H、鹵素(較佳地F)、O-烷基、烷基及CN,較佳選自H、鹵素(較佳地F)及O-烷基。在一個較佳實施例中,一個Z 獨立地係鹵素(較佳地F)或O-烷基,且另一Z 係H。在更佳實施例中,一個Z 係鹵素(較佳地F),且另一Z 係H。 Z is independently selected from the group consisting of: H, halogen (preferably F), O-alkyl, alkyl and CN, preferably selected from the group consisting of H, halogen (preferably F) and O-alkyl. In a preferred embodiment, one Z is independently halogen (preferably F) or O-alkyl, and the other Z is H. In a more preferred embodiment, one Z is halogen (preferably F) and the other Z is H.

R 獨立地選自由及-NR3 R4 以下組成之群,較佳地R 係選自由及-NR3 R4 組成之群,更佳地R,例如。在該等實施例中,較佳係 R is independently chosen from and -NR 3 R 4 or less, preferably the R series is free. And - a group composed of NR 3 R 4 , better R series ,For example . In these embodiments, Better series .

Ra 係選自由H及烷基組成之群,更佳係H及Me,甚至更佳係H。 R a is selected from the group consisting of H and alkyl groups, more preferably H and Me, even more preferably H.

Rb Rc Rd Re Rf Rg 獨立地選自由H及烷基組成之群,或Rb Rc Rd Re Rf Rg 中之任兩者(例如,其連接至相同或毗鄰環原子)可連結以形成3員至8員環。更佳地,Rb Rc Rd Re Rf Rg 獨立地係H或烷基,甚至更佳地係H。 R b , R c , R d , Re , R f , and R g are independently selected from the group consisting of H and alkyl, or any of R b , R c , R d , Re , R f , and R g The two (eg, which are attached to the same or adjacent ring atoms) can be linked to form a 3- to 8-membered ring. More preferably, R b , R c , R d , Re , R f , R g are independently H or alkyl, even more preferably H.

Rj 獨立地選自由以下組成之群:-鹵素、-O-烷基、-CF3 、-CN、-NR3 R4,其中含C1-2 碳原子之橋可存在於a碳原子與c或d碳原子之間或其中含C1-2 碳原子之橋可存在於b碳原子與c或d碳原子之間。更佳地,Rj 係選自由以下組成之群:-鹵素、-O-烷基、-NR3 R4,甚至更佳地Rj R j is independently selected from the group consisting of: -halogen, -O-alkyl, -CF 3 , -CN, -NR 3 R 4 , and , where a bridge containing C 1-2 carbon atoms can exist between a carbon atom and c or d carbon atom or where a bridge containing C 1-2 carbon atoms can exist between b carbon atom and c or d carbon atom . More preferably, R j is selected from the group consisting of: -halogen, -O-alkyl, -NR 3 R 4 and , or even better R j system .

R1 係選自由H及烷基組成之群,較佳係烷基,更佳係CH3 R 1 is selected from the group consisting of H and alkyl groups, preferably an alkyl group, more preferably CH 3 .

R2 獨立地選自由烷基、F及=O組成之群,其中該烷基可視情況經鹵素、-OH或-O-烷基取代,且其中若兩個R2 係孿位的,則其可連結以形成3員至6員環。在一個實施例中,R2 係視情況經取代之烷基,在另一實施例中,R2 係F,在另一實施例中,R2 係=O。 R 2 is independently selected from the group consisting of alkyl, F, and =O, wherein the alkyl group is optionally substituted by halogen, -OH or -O-alkyl, and where two R 2 are geminal, then Can be linked to form a 3 to 6 member ring. In one embodiment, R 2 is optionally substituted alkyl, in another embodiment, R 2 is F, in another embodiment, R 2 is =O.

R3 R4 獨立地選自由H及烷基組成之群,其中該烷基可視情況經鹵素、-OH或-O-烷基取代。在一個實施例中,R3 R4 係視情況經取代之烷基且另一者係H。在另一實施例中,R3 係烷基且R4 係視情況經取代之烷基。在另一實施例中,R3 R4 係H。 R 3 and R 4 are independently selected from the group consisting of H and alkyl, wherein the alkyl is optionally substituted with halogen, -OH or -O-alkyl. In one embodiment, R 3 or R 4 is optionally substituted alkyl and the other is H. In another embodiment, R 3 is alkyl and R 4 is optionally substituted alkyl. In another embodiment, R 3 and R 4 are H.

R5 係選自由H及烷基組成之群,在一個實施例中,R5 係H,在另一實施例中,R5 係烷基。 R 5 is selected from the group consisting of H and alkyl. In one embodiment, R 5 is H. In another embodiment, R 5 is alkyl.

n 係0、1、2、3或4,較佳地n 係0或1,更佳地n 係0。 n is 0, 1, 2, 3 or 4, preferably n is 0 or 1, more preferably n is 0.

p 係1或2,更佳係1。 p represents 1 or 2, preferably 1.

rs 獨立地係0、1、2或3。 r and s are independently 0, 1, 2 or 3.

tu 獨立地係1、2或3。 t and u are independently 1, 2 or 3.

較佳之式(I)化合物係 Preferred compounds of formula (I) are

較佳化合物亦說明於實例中。Preferred compounds are also illustrated in the Examples.

本發明中亦設想本文所揭示之實施例、較佳實施例及更佳實施例之任一組合。Any combination of the embodiments, preferred embodiments and further preferred embodiments disclosed herein is also contemplated by the present invention.

醫藥組合物 儘管本發明之化合物可單獨投與,但較佳根據標準醫藥實踐將其調配成醫藥組合物。因此,本發明亦提供醫藥組合物,其包含治療有效量之式(I)化合物視情況與醫藥上可接受之載劑、稀釋劑、佐劑或賦形劑之混合物。 Pharmaceutical Compositions Although the compounds of the present invention may be administered alone, they are preferably formulated into pharmaceutical compositions in accordance with standard pharmaceutical practice. Accordingly, the present invention also provides pharmaceutical compositions comprising a therapeutically effective amount of a compound of formula (I), optionally in admixture with a pharmaceutically acceptable carrier, diluent, adjuvant or excipient.

醫藥上可接受之賦形劑為醫藥技術中所熟知,且闡述於(例如) Remington's Pharmaceutical Sciences,第15版,Mack Publishing Co., New Jersey (1975)中。可根據預期投與途徑及標準醫藥實踐來選擇醫藥賦形劑。賦形劑在不對其接受者有害之意義上必須係可接受的。Pharmaceutically acceptable excipients are well known in the pharmaceutical art and are described, for example, in Remington's Pharmaceutical Sciences, 15th ed., Mack Publishing Co., New Jersey (1975). Pharmaceutical excipients can be selected based on the intended route of administration and standard pharmaceutical practice. The excipient must be acceptable in the sense that it is not harmful to the recipient.

可用於調配本發明之醫藥組合物之醫藥上可用之賦形劑可包含(例如)載劑;媒劑;稀釋劑;溶劑,例如一元醇(例如乙醇、異丙醇)及多元醇(例如二醇)及可食用油(例如大豆油、椰子油、橄欖油、紅花油、棉籽油)、油性酯(例如油酸乙酯、肉豆蔻酸異丙酯)、黏合劑;佐劑;增溶劑;增稠劑;穩定劑;崩解劑;助流劑;潤滑劑;緩衝劑;乳化劑;潤濕劑;懸浮劑;甜味劑;著色劑;矯味劑;包衣劑;防腐劑;抗氧化劑;處理劑;藥物遞送改質劑及增強劑,例如磷酸鈣、硬脂酸鎂、滑石、單醣、二醣、澱粉、明膠、纖維素、甲基纖維素、羧甲基纖維素鈉、右旋糖、羥丙基-β-環糊精、聚乙烯基吡咯啶酮、低熔點蠟及離子交換樹脂。Pharmaceutically acceptable excipients that can be used to formulate the pharmaceutical compositions of the present invention may include, for example, carriers; vehicles; diluents; solvents, such as monohydric alcohols (such as ethanol, isopropyl alcohol) and polyhydric alcohols (such as dihydric alcohols). alcohol) and edible oils (such as soybean oil, coconut oil, olive oil, safflower oil, cottonseed oil), oily esters (such as ethyl oleate, isopropyl myristate), binders; adjuvants; solubilizers; Thickener; stabilizer; disintegrant; glidant; lubricant; buffer; emulsifier; wetting agent; suspending agent; sweetener; colorant; flavoring agent; coating agent; preservative; antioxidant ; Processing agents; drug delivery modifiers and enhancers, such as calcium phosphate, magnesium stearate, talc, monosaccharides, disaccharides, starch, gelatin, cellulose, methylcellulose, sodium carboxymethylcellulose, right Spinose, hydroxypropyl-β-cyclodextrin, polyvinylpyrrolidone, low melting point wax and ion exchange resin.

本發明化合物之投與(遞送)途徑包括(但不限於)以下中之一或多者:經口(例如作為錠劑、膠囊或作為可攝取溶液)、局部、經黏膜(例如作為吸入用經鼻噴霧或氣溶膠)、經鼻、非經腸(例如藉由可注射形式)、經胃腸、脊椎內、腹膜內、肌內、靜脈內、子宮內、眼內、真皮內、顱內、氣管內、陰道內、腦室內、大腦內、皮下、經眼(包括玻璃體內或前房內)、經皮、直腸、經頰、硬膜外及舌下。Routes of administration (delivery) of the compounds of the invention include, but are not limited to, one or more of the following: oral (e.g., as a lozenge, capsule, or as an ingestible solution), topical, transmucosal (e.g., as an inhalation) nasal spray or aerosol), nasal, parenteral (e.g., by injectable form), gastrointestinal, intraspinal, intraperitoneal, intramuscular, intravenous, intrauterine, intraocular, intradermal, intracranial, tracheal Internal, intravaginal, intracerebroventricular, intracerebral, subcutaneous, transocular (including intravitreal or intraanterior chamber), transcutaneous, rectal, transbuccal, epidural and sublingual.

舉例而言,化合物可以含有矯味劑或著色劑之錠劑、膠囊、陰道栓劑(ovule)、酏劑、溶液或懸浮液形式經口投與,以用於立即、延遲、改良、持續、脈衝或受控釋放應用。For example, the compounds may be administered orally in the form of tablets, capsules, ovules, elixirs, solutions or suspensions containing flavoring or coloring agents for immediate, delayed, modified, continuous, pulsed or Controlled release applications.

錠劑可含有賦形劑,例如微晶纖維素、乳糖、檸檬酸鈉、碳酸鈣、磷酸氫鈣及甘胺酸;崩解劑,例如澱粉(較佳地玉米、馬鈴薯或木薯澱粉)、羥乙酸澱粉鈉、交聯羧甲基纖維素鈉及某些複合矽酸鹽;及粒化黏合劑,例如聚乙烯基吡咯啶酮、羥丙基甲基纖維素(HPMC)、羥丙基纖維素(HPC)、蔗糖、明膠及阿拉伯樹膠。另外,可納入諸如硬脂酸鎂、硬脂酸、山崳酸甘油基酯及滑石等潤滑劑。類似類型之固體組合物亦可作為填充劑用於明膠膠囊中。就此而言,較佳賦形劑包括乳糖、澱粉、纖維素、乳糖或高分子量聚乙二醇。對於水性懸浮液及/或酏劑而言,藥劑可與以下組合:各種調味劑或矯味劑、著色物質或染料、乳化劑及/或懸浮劑及稀釋劑(例如水、乙醇、丙二醇及甘油)及其組合。Tablets may contain excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and glycine; disintegrants such as starch (preferably corn, potato or tapioca starch), hydroxyl Sodium starch acetate, croscarmellose sodium and certain complex silicates; and granulated binders, such as polyvinylpyrrolidone, hydroxypropylmethylcellulose (HPMC), hydroxypropylcellulose (HPC), sucrose, gelatin and gum arabic. Additionally, lubricants such as magnesium stearate, stearic acid, glyceryl behenate, and talc may be incorporated. Solid compositions of a similar type may also be used as fillers in gelatin capsules. In this regard, preferred excipients include lactose, starch, cellulose, lactose or high molecular weight polyethylene glycols. For aqueous suspensions and/or elixirs, the agents may be combined with various flavorings or correctives, coloring substances or dyes, emulsifying and/or suspending agents and diluents such as water, ethanol, propylene glycol and glycerol. and combinations thereof.

若本發明之化合物係以非經腸方式投與,則此投與之實例包括以下中之一或多者:靜脈內、動脈內、腹膜內、鞘內、室內、尿道內、胸骨內、顱內、肌內或皮下投與該等化合物;及/或藉由使用輸注技術。對於非經腸投與而言,最佳使用呈無菌水溶液形式之化合物,該無菌水溶液可含有其他物質,例如足夠的鹽或葡萄糖以使得溶液與血液等滲。水溶液應視需要經適當地緩衝(較佳地至3至9之pH)。藉由熟習此項技術者熟知之標準醫藥技術易於在無菌條件下製備適宜非經腸調配物。If a compound of the invention is administered parenterally, examples of such administration include one or more of the following: intravenous, intraarterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, cranial The compounds may be administered intramuscularly, intramuscularly or subcutaneously; and/or by using infusion techniques. For parenteral administration, the compounds are preferably used in the form of sterile aqueous solutions, which may contain other substances, such as sufficient salt or glucose to render the solution isotonic with blood. Aqueous solutions should be appropriately buffered if necessary (preferably to a pH of 3 to 9). Suitable parenteral formulations are readily prepared under sterile conditions by standard pharmaceutical techniques well known to those skilled in the art.

如所指示,本發明之化合物可以鼻內方式或藉由吸入來投與,且在使用諸如二氯二氟甲烷、三氯氟甲烷、二氯四氟乙烷、氫氟代烷(例如1,1,1,2-四氟乙烷(HFA134AT)或1,1,1,2,3,3,3-七氟丙烷(HFA 227EA))、二氧化碳或其他適宜氣體等適宜推進劑之情形下,自加壓容器、幫浦、噴霧或霧化器以乾粉吸入劑或氣溶膠噴霧投遞之形式便捷地遞送。在加壓氣溶膠之情形下,劑量單位可藉由提供閥以遞送計量來確定。加壓容器、幫浦、噴霧或霧化器可含有活性化合物之溶液或懸浮液,例如使用乙醇及推進劑之混合物作為溶劑,其可另外含有潤滑劑,例如去水山梨醇三油酸酯。用於吸入器或吹入器中之膠囊及藥筒(例如,自明膠製得)可經調配以含有化合物與適宜粉末基質(例如乳糖或澱粉)之粉末混合物。The compounds of the present invention may be administered intranasally or by inhalation, as indicated, and are administered using solvents such as dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, hydrofluoroalkane (e.g., 1, In the case of suitable propellants such as 1,1,2-tetrafluoroethane (HFA134AT) or 1,1,1,2,3,3,3-heptafluoropropane (HFA 227EA)), carbon dioxide or other suitable gases, self-adding Pressurized containers, pumps, sprays or nebulizers are conveniently delivered in the form of dry powder inhalers or aerosol spray delivery. In the case of pressurized aerosols, the dosage unit may be determined by providing a valve to deliver the dose. Pressurized containers, pumps, sprays or atomizers may contain a solution or suspension of the active compound, for example using a mixture of ethanol and propellant as solvent, which may additionally contain a lubricant, such as sorbitan trioleate. Capsules and cartridges (eg, made from gelatin) for use in inhalers or insufflators may be formulated to contain a powder mixture of the compound and a suitable powder base (eg, lactose or starch).

或者,本發明之化合物可以栓劑或子宮托之形式投與,或其可以凝膠、水凝膠、洗劑、溶液、乳霜、軟膏或撒粉之形式局部施用。本發明之化合物亦可經皮或透過皮膚投與,例如藉由使用皮膚貼劑。Alternatively, the compounds of the present invention may be administered in the form of a suppository or pessary, or they may be administered topically in the form of a gel, hydrogel, lotion, solution, cream, ointment, or dusting. The compounds of the present invention may also be administered transdermally or transdermally, for example, through the use of transdermal patches.

其亦可經肺或經直腸途徑投與。其亦可經眼途徑投與。對於眼部使用,可將化合物於等滲、pH經調整之無菌鹽水中調配成微粉化懸浮液,或較佳地於等滲、pH經調整之無菌鹽水中調配成溶液,視情況與防腐劑(例如苯紮氯銨)組合。或者,其可調配於軟膏(例如石蠟脂)中。It may also be administered via the pulmonary or rectal route. It can also be administered via the eye route. For ocular use, the compounds may be formulated as a micronized suspension in isotonic, pH-adjusted sterile saline, or preferably as a solution in isotonic, pH-adjusted sterile saline, with a preservative as appropriate. (e.g. benzalkonium chloride) combination. Alternatively, it can be formulated in an ointment such as paraffin grease.

對於局部施用至皮膚而言,可將本發明之化合物調配為含有活性化合物懸浮或溶解於例如以下一或多者之混合物中之適合軟膏:礦物油、液體石蠟脂、白凡士林、丙二醇、乳化蠟及水。或者,可將其調配為適宜洗劑或乳霜,懸浮或溶解於例如以下之一或多者之混合物中:礦物油、去水山梨醇單硬脂酸酯、聚乙二醇、液體石蠟、聚山梨醇酯60、鯨蠟酯蠟、鯨蠟硬脂醇(cetearyl alcohol)、2-辛基十二烷醇、苄醇及水。For topical application to the skin, the compounds of the invention may be formulated as a suitable ointment containing the active compound suspended or dissolved in a mixture such as one or more of: mineral oil, liquid paraffin, white petrolatum, propylene glycol, emulsifying wax And water. Alternatively, it may be formulated as a suitable lotion or cream, suspended or dissolved in a mixture of, for example, one or more of the following: mineral oil, sorbitan monostearate, polyethylene glycol, liquid paraffin, Polysorbate 60, cetyl ester wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.

通常,醫師將確定最適於個別個體之實際劑量。對於任一特定個體之特定劑量及劑量頻率可變化且將取決於多種因素,包括所採用特定化合物之活性、該化合物之代謝穩定性及作用時間長度、年齡、體重、整體健康狀況、性別、飲食、投與方式及時間、排泄速率、藥物組合、特定病狀之嚴重程度及經歷療法之個體。Generally, the physician will determine the actual dosage that is most appropriate for the individual individual. The specific dosage and dosage frequency for any particular individual may vary and will depend on a variety of factors, including the activity of the particular compound employed, the metabolic stability and length of action of the compound, age, weight, general health, sex, diet , mode and timing of administration, rate of excretion, drug combinations, severity of the specific condition and individuals undergoing treatment.

本發明之化合物投與人類(大約70 kg體重)之建議劑量係每單位劑量0.1 mg至3 g、0.1 mg至2 g、0.1 mg至1 g、較佳地1 mg至500 mg之活性成分。單位劑量可例如每天投與1至4次。劑量將取決於投與途徑。應瞭解,可能需要視患者之年齡及體重以及欲治療病狀之嚴重程度對劑量作常規變化。精確劑量及投與途徑最終將由主治醫師或獸醫師判定。Recommended dosages of the compounds of the invention for administration to humans (approximately 70 kg body weight) are 0.1 mg to 3 g, 0.1 mg to 2 g, 0.1 mg to 1 g, preferably 1 mg to 500 mg of active ingredient per unit dose. Unit doses may be administered, for example, from 1 to 4 times per day. Dosage will depend on the route of administration. It should be understood that routine changes in dosage may be necessary depending on the age and weight of the patient and the severity of the condition to be treated. The precise dosage and route of administration will ultimately be determined by the attending physician or veterinarian.

本發明之化合物亦可與其他治療劑組合使用。當本發明之化合物與具有對抗相同疾病之活性之第二治療劑組合使用時,每一化合物之劑量可與單獨使用該化合物時之劑量不同。The compounds of the invention may also be used in combination with other therapeutic agents. When a compound of the present invention is used in combination with a second therapeutic agent active against the same disease, the dosage of each compound may be different than the dosage of that compound alone.

上文所提及之組合可便捷地以醫藥調配物之形式呈現使用。此等組合之個別組分可藉由任一便捷途徑以單獨或組合醫藥調配物依序或同時投與。當依序投與時,可首先投與本發明之化合物或第二治療劑。當同時投與時,該組合可以相同或不同醫藥組合物投與。當組合於同一調配物中時,應瞭解,該兩種化合物必須係穩定的且彼此相容且與調配物之其他組分相容。當單獨調配時,其可以任一便捷調配物、便捷地以諸如業內用於此等化合物之已知方式提供。The combinations mentioned above can be conveniently presented and used in the form of pharmaceutical preparations. The individual components of these combinations may be administered sequentially or simultaneously in separate or combined pharmaceutical formulations by any convenient route. When administered sequentially, the compound of the invention or the second therapeutic agent can be administered first. When administered simultaneously, the combination may be administered as the same or different pharmaceutical compositions. When combined in the same formulation, it is understood that the two compounds must be stable and compatible with each other and with the other components of the formulation. When formulated separately, they may be provided in any convenient formulation, conveniently, in a manner such as is known in the art for such compounds.

本發明之醫藥組合物可以如(例如) Remington's Pharmaceutical Sciences,第15版,Mack Publishing Co., New Jersey (1975)中所闡述之對熟習此項技術者本身已知之方式來產生。Pharmaceutical compositions of the present invention may be produced in a manner known per se to those skilled in the art, as set forth, for example, in Remington's Pharmaceutical Sciences, 15th Edition, Mack Publishing Co., New Jersey (1975).

可利用本發明之化合物治療、改善或預防之疾病或病狀係與Tau蛋白聚集體有關的病症或異常,例如神經退化病症。可治療、改善或預防之疾病及病狀之實例係由神經纖維病灶之形成引起或與其有關。此係tau蛋白病變之主要腦病理學。該等疾病及病狀包含異質群體之神經退化性疾病或病狀,包括顯示共存在Tau及類澱粉病理學之疾病或病狀。Diseases or conditions that may be treated, ameliorated, or prevented using the compounds of the invention are those associated with Tau protein aggregates, such as neurodegenerative disorders. Examples of diseases and conditions that can be treated, ameliorated or prevented are caused by or related to the formation of nerve fiber lesions. This is the main brain pathology of tauopathies. Such diseases and conditions include a heterogeneous population of neurodegenerative diseases or conditions, including those showing the coexistence of Tau and amyloid pathology.

可治療、改善或預防之疾病及病狀之實例包括(但不限於)阿茲海默氏病(AD)、家族性AD、PART (原發性年齡相關性Tau蛋白病變)、庫賈氏病、拳擊手型失智症、唐氏症候群、傑茨曼-斯脫司勒-史茵克病(GSS)、包涵體肌炎、普里昂蛋白腦類澱粉血管病變、創傷性腦損傷(TBI)、肌肉萎縮性脊髓側索硬化症(ALS)、關島帕金森氏症-失智複合症、非關島運動神經元病伴神經纖維纏結、嗜銀顆粒病、皮質基底核退化症(CBD)、瀰漫性神經纖維纏結伴鈣化、與染色體17相關之額顳葉失智症伴帕金森氏症(FTDP-17)、哈勒沃登-施帕茨病、多系統萎縮(MSA)、C型尼曼匹克症、蒼白球-腦橋-黑質退化、匹克氏病(PiD)、進行性皮質下神經膠瘤病、進行性核上性麻痺(PSP)、亞急性硬化性泛腦炎、纏結主導型失智症、腦炎後帕金森氏症、肌強直性營養不良、亞急性硬化性泛腦病變、LRRK2突變、慢性創傷性腦病變(CTE)、家族性英國型失智症、家族性丹麥型失智症、其他額顳葉退化、瓜德羅普帕金森氏症、神經退化伴腦內鐵累積、SLC9A6相關性智力遲鈍、白質tau蛋白病變伴球狀膠質細胞包涵體、癲癇、路易氏體失智症(LBD)、輕度認知損害(MCI)、多發性硬化、帕金森氏病、HIV相關性失智症、成人發作型糖尿病、老年性心臟類澱粉變性、青光眼、缺血性中風、AD中之精神病及亨庭頓氏病。較佳地,可治療、改善或預防之疾病及病狀包括阿茲海默氏病(AD),以及其他神經退化性tau蛋白病變,例如庫賈氏病、拳擊手型失智症、肌肉萎縮性脊髓側索硬化症(ALS)、嗜銀顆粒病、皮質基底核退化症(CBD)、與染色體17相關之額顳葉失智症伴帕金森氏症(FTDP-17)、匹克氏病(PiD)、進行性核上性麻痺(PSP)、纏結主導型失智症、關島帕金森氏症失智複合症、哈勒沃登-施帕茨病、慢性創傷性腦病變(CTE)、創傷性腦損傷(TBI)及其他額顳葉退化。更佳為阿茲海默氏病(AD)、皮質基底核退化症(CBD)、匹克氏病(PiD)及進行性核上性麻痺(PSP)。Examples of diseases and conditions that can be treated, ameliorated or prevented include (but are not limited to) Alzheimer's disease (AD), familial AD, PART (primary age-related tauopathy), Creutzfeldt-Jakob disease, Boxer dementia, Down syndrome, Gertzmann-Steusler-Steinke disease (GSS), inclusion body myositis, prion cerebral amyloid vasculopathy, traumatic brain injury (TBI), Amyotrophic lateral sclerosis (ALS), Guam Parkinson's disease-dementia complex, non-Guam motor neuron disease with neurofibrillary tangles, argyrophilic granulopathy, corticobasal degeneration (CBD), diffuse Neurofibrillary tangles with calcification, frontotemporal dementia with Parkinson's disease associated with chromosome 17 (FTDP-17), Hallerwarden-Spatz disease, multiple system atrophy (MSA), Niemann type C Pick's disease, pallidal-pontine-nigra degeneration, Pick's disease (PiD), progressive subcortical gliomatosis, progressive supranuclear palsy (PSP), subacute sclerosing panencephalitis, tangle-predominant form Dementia, postencephalitic parkinsonism, myotonic dystrophy, subacute sclerosing panencephalopathy, LRRK2 mutations, chronic traumatic encephalopathy (CTE), familial British dementia, familial Danish form Dementia, other frontotemporal degenerations, Guadeloupe Parkinson's disease, neurodegeneration with iron accumulation in the brain, SLC9A6-related mental retardation, white matter tauopathy with glial inclusions, epilepsy, Lewy bodies Dementia (LBD), mild cognitive impairment (MCI), multiple sclerosis, Parkinson's disease, HIV-related dementia, adult-onset diabetes, senile cardiac amyloidosis, glaucoma, ischemic stroke, Psychosis and Huntington's disease in AD. Preferably, diseases and conditions that can be treated, improved or prevented include Alzheimer's disease (AD), and other neurodegenerative tauopathies, such as Creutzfeldt-Jakob disease, Boxer's dementia, muscular dystrophy Lateral sclerosis (ALS), argyrophilic granulopathy, corticobasal degeneration (CBD), frontotemporal dementia with Parkinson's disease associated with chromosome 17 (FTDP-17), Pick's disease (PiD) ), progressive supranuclear palsy (PSP), tangle-dominant dementia, Guam Parkinson's disease dementia complex, Hallwarden-Spatz disease, chronic traumatic encephalopathy (CTE), trauma brain injury (TBI) and other frontotemporal lobar degeneration. More preferably, Alzheimer's disease (AD), corticobasal degeneration (CBD), Pick's disease (PiD) and progressive supranuclear palsy (PSP).

本發明之化合物亦可用於降低蛋白質聚集、特定而言Tau聚集。化合物降低Tau聚集之能力可(例如)使用ThT分析來測定(Hudson等人,FEBS J., 2009, 5960-72)。The compounds of the invention may also be used to reduce protein aggregation, in particular Tau aggregation. The ability of a compound to reduce Tau aggregation can be determined, for example, using a ThT assay (Hudson et al., FEBS J., 2009, 5960-72).

本發明之化合物可用於治療神經發炎過程與Tau蛋白之錯誤摺疊及/或病理性聚集有關的各種各樣之病症。The compounds of the present invention can be used to treat a variety of disorders in which neuroinflammatory processes are associated with misfolding and/or pathological aggregation of Tau protein.

本發明之化合物可用作用於表徵具有Tau病理學之組織及用於在此組織上測試靶向Tau病理學之化合物之分析參考或活體外篩選工具。The compounds of the invention can be used as analytical references or in vitro screening tools for characterizing tissues with Tau pathology and for testing compounds targeting Tau pathology on such tissues.

本發明之化合物可用作光探針以用於將化合物與靶標交聯、用於活體外篩選分析中以鑑別並表徵化合物之作用模式,包括定位結合位點。下文報導本發明化合物之光探針之一些實例: The compounds of the present invention can be used as photoprobes for cross-linking compounds to targets, and in in vitro screening assays to identify and characterize the mode of action of compounds, including localizing binding sites. Some examples of photoprobes for the compounds of the present invention are reported below:

本發明之化合物亦可以與至少一種其他生物活性化合物及/或醫藥上可接受之載劑及/或稀釋劑及/或賦形劑之混合物之形式提供。該等化合物及/或其他生物活性化合物較佳以治療有效量存在。The compounds of the invention may also be provided in the form of mixtures with at least one other biologically active compound and/or pharmaceutically acceptable carriers and/or diluents and/or excipients. These compounds and/or other biologically active compounds are preferably present in therapeutically effective amounts.

其他生物活性化合物之性質將取決於混合物之預期用途。其他生物活性物質或化合物可藉由與本發明之化合物相同或類似之機制或藉由一種不相關之作用機制或藉由多種相關及/或不相關之作用機制來發揮其生物效應。The nature of the other biologically active compounds will depend on the intended use of the mixture. Other biologically active substances or compounds may exert their biological effects through the same or similar mechanism as the compounds of the present invention, or through an unrelated mechanism of action, or through multiple related and/or unrelated mechanisms of action.

通常,其他生物活性化合物可包括神經傳遞增強劑、精神治療藥物、乙醯基膽鹼酯酶抑制劑、鈣通道阻斷劑、生物胺、苯并二氮呯鎮靜劑、乙醯基膽鹼合成、儲存或釋放增強劑、乙醯基膽鹼突觸後受體激動劑、單胺氧化酶-A或單胺氧化酶-B抑制劑、N-甲基-D-天冬胺酸鹽麩胺酸受體拮抗劑、非類固醇消炎藥、抗氧化劑及血清素能受體拮抗劑。特定而言,其他生物活性化合物可選自由以下組成之群:用於治療類澱粉變性之化合物;抗氧化壓力之化合物;抗細胞凋亡化合物;金屬螯合劑;DNA修復抑制劑,例如哌侖西平及代謝物、3-胺基-1-丙烷磺酸(3APS)、1,3-丙烷二磺酸鹽(1,3PDS);α-分泌酶活化劑;β-分泌酶及γ-分泌酶抑制劑;肝醣合酶激酶3抑制劑;O連接之N-乙醯葡糖胺水解酶(OGA)抑制劑;神經傳遞質;β褶板碎裂劑;類澱粉β清除/消除細胞組分引誘劑;N末端截短之類澱粉β (包括焦麩胺酸化之類澱粉β 3-42 )之抑制劑;抗發炎分子,或膽鹼酯酶抑制劑(ChEI),例如塔克寧、利凡斯的明、多奈派齊及/或加蘭他敏;M1激動劑;其他藥物,包括任何類澱粉或Tau改質藥物及營養補充劑;抗體,包括任何功能等效抗體或其功能部分;或疫苗。In general, other bioactive compounds may include neurotransmission enhancers, psychotherapeutic drugs, acetylcholinesterase inhibitors, calcium channel blockers, biogenic amines, benzodiazepine sedatives, acetylcholine synthesis, Storage or release enhancer, acetylcholine postsynaptic receptor agonist, monoamine oxidase-A or monoamine oxidase-B inhibitor, N-methyl-D-aspartate glutamate receptor antagonist, non- Steroid anti-inflammatory drugs, antioxidants and serotonergic receptor antagonists. In particular, other bioactive compounds may be selected from the group consisting of: compounds useful in treating amyloidosis; compounds that resist oxidative stress; anti-apoptotic compounds; metal chelators; DNA repair inhibitors, such as pirenzepine and metabolites, 3-amino-1-propanesulfonic acid (3APS), 1,3-propanedisulfonate (1,3PDS); α-secretase activator; β-secretase and γ-secretase inhibitors Agent; glycogen synthase kinase 3 inhibitor; O-linked N-acetylglucosamine hydrolase (OGA) inhibitor; neurotransmitter; beta pleated plate disruptor; amyloid beta scavenging/eliminating cellular component attractant Agents; inhibitors of N-terminally truncated starch-like β (including pyroglutamic acid-like starch β 3-42); anti-inflammatory molecules, or cholinesterase inhibitors (ChEI), such as tacnin, rivan Stimamine, donepezil and/or galantamine; M1 agonists; other drugs, including any starch-like or Tau-modified drugs and nutritional supplements; antibodies, including any functionally equivalent antibodies or functional parts thereof; or vaccine.

在另一實施例中,本發明之混合物可包含菸鹼酸或美金剛以及本發明之化合物及視情況醫藥上可接受之載劑及/或稀釋劑及/或賦形劑。In another embodiment, the mixture of the invention may comprise niacin or memantine together with a compound of the invention and optionally a pharmaceutically acceptable carrier and/or diluent and/or excipient.

在本發明之另一實施例中,提供混合物,其包含(例如)用於治療陽性及陰性精神病性症狀,包括幻覺、妄想、思維障礙(體現為明顯不連貫、思維脫軌(derailment)、言語岔開(tangentiality))及怪異或解構行為以及失樂症、情緒淡然(flattened affect)、冷漠及社會退縮之氯氮平(clozapine)、齊拉西酮(ziprasidone)、利培酮(risperidone)、阿立哌唑(aripiprazole)或奧氮平(olanzapine)作為另一生物活性化合物「非典型抗精神病藥」,以及本發明之化合物及視情況醫藥上可接受之載劑及/或稀釋劑及/或賦形劑。In another embodiment of the invention, there is provided a mixture comprising, for example, for the treatment of positive and negative psychotic symptoms, including hallucinations, delusions, thought disorders manifested by marked incoherence, derailment, and speech disturbances. tangentiality) and bizarre or deconstructive behavior, as well as amusia, flattened affect, apathy, and social withdrawal with clozapine, ziprasidone, risperidone, and Aripiprazole or olanzapine as another biologically active compound "atypical antipsychotic", as well as the compound of the present invention and optionally pharmaceutically acceptable carriers and/or diluents and/or Excipients.

可適宜地與本發明之化合物組合用於混合物中之其他化合物闡述於(例如) WO 2004/058258 (尤其參見第16及17頁)中,其包括治療性藥物靶標(第36至39頁)、烷烴磺酸及烷醇硫酸(第39至51頁)、膽鹼酯酶抑制劑(第51至56頁)、NMDA受體拮抗劑(第56至58頁)、雌激素(第58至59頁)、非類固醇消炎藥(第60及61頁)、抗氧化劑(第61及62頁)、過氧化體增殖劑活化受體(PPAR)激動劑(第63至67頁)、降膽固醇劑(第68至75頁)、類澱粉抑制劑(第75至77頁)、類澱粉形成抑制劑(第77至78頁)、金屬螯合劑(第78及79頁)、抗精神病藥及抗抑鬱藥(第80至82頁)、營養補充劑(第83至89頁)及增加腦中生物活性物質之可用性之化合物(參見第89至93頁)及前藥(第93及94頁),該文件係以引用的方式併入本文中。Other compounds which may suitably be combined with the compounds of the invention for use in mixtures are described, for example, in WO 2004/058258 (see in particular pages 16 and 17), including therapeutic drug targets (pages 36 to 39), Alkanesulfonic acids and alkanol sulfates (pages 39 to 51), cholinesterase inhibitors (pages 51 to 56), NMDA receptor antagonists (pages 56 to 58), estrogens (pages 58 to 59 ), nonsteroidal anti-inflammatory drugs (pages 60 and 61), antioxidants (pages 61 and 62), peroxisome proliferator-activated receptor (PPAR) agonists (pages 63 to 67), cholesterol-lowering agents (pages 63 to 67) Pages 68 to 75), amyloid inhibitors (pages 75 to 77), inhibitors of amyloid formation (pages 77 to 78), metal chelators (pages 78 and 79), antipsychotics and antidepressants ( pages 80 to 82), nutritional supplements (pages 83 to 89) and compounds that increase the availability of bioactive substances in the brain (see pages 89 to 93) and prodrugs (pages 93 and 94), the document is Incorporated herein by reference.

本發明亦包括本發明化合物之所有適宜同位素變化形式。本發明化合物之同位素變化形式定義為其中至少一個原子經具有相同原子序數但原子質量不同於通常或在自然界中發現之原子質量之原子替代者。可併入至本發明化合物中之同位素之實例包括氫、碳、氮、氧、硫、氟及氯之同位素,例如分別為2 H、3 H、13 C、14 C、15 N、17 O、18 O、35 S、18 F及36 Cl。本發明之某些同位素變化形式(例如其中併入放射性同位素(例如3 H或14 C)之彼等)可用於藥物及/或受質組織分佈研究中。含氚(即3 H)及碳-14 (即14 C)同位素因其易於製備及可檢測性而尤佳。經18 F標記之化合物尤其適於諸如PET等成像應用。此外,用諸如氘(即2 H)等同位素進行取代因具有更強之代謝穩定性從而可提供某些治療優點,例如活體內半衰期延長或劑量需要減少,且因此可在一些情況下較佳。本發明化合物之同位素變化形式通常可藉由習用程序、例如藉由說明性方法或藉由下文實例及製備中所闡述之製備使用適宜試劑之適當同位素變化形式來製備。The invention also includes all suitable isotopic variations of the compounds of the invention. Isotopic variations of the compounds of the present invention are defined as those in which at least one atom is replaced by an atom having the same atomic number but an atomic mass different from that usually or found in nature. Examples of isotopes that may be incorporated into the compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, sulfur, fluorine and chlorine, such as 2 H, 3 H, 13 C, 14 C, 15 N, 17 O, respectively. 18 O, 35 S, 18 F and 36 Cl. Certain isotopic variations of the present invention, such as those in which radioactive isotopes (eg, 3 H or 14 C) are incorporated, may be used in drug and/or substrate tissue distribution studies. Isotopes containing tritium (i.e. 3 H) and carbon-14 (i.e. 14 C) are particularly preferred because of their ease of preparation and detectability. 18 F-labeled compounds are particularly suitable for imaging applications such as PET. In addition, substitution with isotopes such as deuterium (ie, 2 H) may provide certain therapeutic advantages, such as increased half-life in vivo or reduced dosage requirements, due to greater metabolic stability, and may therefore be preferred in some circumstances. Isotopic variations of the compounds of the present invention may generally be prepared by conventional procedures, for example by the illustrative methods or by preparation as set forth in the Examples and Preparations below, using appropriate isotopic variations of suitable reagents.

本發明之化合物可藉由以下方案中所示一般方法中之一者來合成。該等方法僅出於說明性目的而給出,且不應解釋為具有限制性。Compounds of the invention may be synthesized by one of the general methods shown in the following schemes. These methods are given for illustrative purposes only and should not be construed as limiting.

用於製備本發明之構建單元之一般合成方案:General synthetic scheme for preparing the building blocks of the invention:

方案1 plan 1

於適宜溶劑中在酸性條件下加熱市售苯基肼衍生物(Z = H、F、Cl、Me或OMe;Ra = H、CH3 )與市售4-側氧基六氫吡啶-1-甲酸第三丁基酯(費舍爾吲哚(Fischer-Indole)合成),在純化後得到三環2,3,4,5-四氫-1H -吡啶并[4,3-b ]吲哚構建單元。在使用2-或3-取代之苯基肼衍生物之情形下,藉由超臨界流體層析(SFC)分離區域異構物。進一步利用Boc2 O處理在吲哚環上帶有NH-部分之三環構建單元以選擇性地保護脂肪族二級胺部分且在純化後獲得。Heating commercially available phenylhydrazine derivatives ( Z = H, F, Cl, Me or OMe; R a = H, CH 3 ) and commercially available 4-side oxyhexahydropyridine-1 in a suitable solvent under acidic conditions -Tertiary butyl formate (Fischer-Indole synthesis), which after purification gives tricyclic 2,3,4,5-tetrahydro- 1H -pyrido[4,3- b ] Indole building block. In the case of using 2- or 3-substituted phenylhydrazine derivatives, the regioisomers are separated by supercritical fluid chromatography (SFC). The tricyclic building block bearing the NH-moiety on the indole ring was further treated with Boc2O to selectively protect the aliphatic secondary amine moiety and was obtained after purification.

方案2 為避免因採用2-或3-取代之苯基肼衍生物所致之區域異構物形成,使用具有毗鄰肼部分之額外鹵素原子(Br、Cl)之相應苯基肼衍生物。因此,利用市售4-側氧基六氫吡啶-1-甲酸第三丁基酯於適宜溶劑中在酸性條件下之費舍爾吲哚合成在純化後僅得到具有額外鹵素原子之單一產物。脂肪族二級胺經Boc保護且在純化後獲得產物。然後藉由使用適宜鹼於適當溶劑中利用鈀觸媒進行氫化來去除額外鹵素原子,以在純化後得到期望三環2,3,4,5-四氫-1H -吡啶并[4,3-b ]吲哚構建單元。Scenario 2 To avoid the formation of regioisomers resulting from the use of 2- or 3-substituted phenylhydrazine derivatives, the corresponding phenylhydrazine derivatives with additional halogen atoms (Br, Cl) adjacent to the hydrazine moiety are used. Therefore, Fisher's indole synthesis using commercially available tert-butyl 4-pendantoxyhexahydropyridine-1-carboxylate in a suitable solvent under acidic conditions yields only a single product with an additional halogen atom after purification. The aliphatic secondary amine was Boc protected and the product was obtained after purification. The additional halogen atoms are then removed by hydrogenation with a suitable base in an appropriate solvent using a palladium catalyst to obtain the desired tricyclic 2,3,4,5-tetrahydro-1 H -pyrido[4,3 - b ] indole building blocks.

方案3 然後於適當溶劑中使用適宜鹼利用碘甲烷或甲苯磺醯氯處理吲哚部分之NH-部分,以在純化後得到N-甲基或N-甲苯磺醯基衍生物。於適當溶劑中藉由酸處理去除Boc-保護基團,以在純化後得到期望三環2,3,4,5-四氫-1H -吡啶并[4,3-b ]吲哚構建單元。在無鹼處理之情形下,獲得相應鹽。Option 3 The NH-portion of the indole moiety is then treated with methyl iodide or tosyl chloride using a suitable base in an appropriate solvent to obtain an N-methyl or N-toluenesulfonyl derivative after purification. The Boc-protecting group is removed by acid treatment in an appropriate solvent to obtain the desired tricyclic 2,3,4,5-tetrahydro- 1H -pyrido[4,3- b ]indole building block after purification . In the absence of alkali treatment, the corresponding salt is obtained.

方案4 於適當溶劑中利用甲基肼處理在3位、4位或5位具有F原子且在2位具有額外鹵素原子(Br、Cl)之市售氟吡啶衍生物,以在純化後得到相應N-甲基肼衍生物。於適宜溶劑中在酸性條件下利用市售4-側氧基六氫吡啶-1-甲酸第三丁基酯之費舍爾吲哚合成在純化後得到期望三環9-甲基-6,7,8,9-四氫-5H -吡咯并[2,3-b :4,5-c ']二吡啶衍生物。Option 4 Commercially available fluoropyridine derivatives having an F atom at position 3, 4 or 5 and an additional halogen atom (Br, Cl) at position 2 are treated with methylhydrazine in an appropriate solvent to obtain the corresponding N- Methylhydrazine derivatives. Fisher indole synthesis using commercially available tert-butyl 4-pentanoxyhexahydropyridine-1-carboxylate in a suitable solvent under acidic conditions gave the desired tricyclic 9-methyl-6,7 after purification. ,8,9-tetrahydro- 5H -pyrrolo[2,3- b :4,5- c ']dipyridine derivatives.

方案5 於適宜溶劑中在酸性條件下加熱市售苯基肼衍生物(Z = F)與市售3-側氧基-8-氮雜二環[3.2.1]辛烷-8-甲酸第三丁基酯(費舍爾吲哚合成),在純化後得到三環5,6,7,8,9,10-六氫-7,10-環亞胺基環庚[b ]吲哚衍生物。在2-或3-取代之苯基肼衍生物之情形下,區域異構物必須藉由超臨界流體層析(SFC)分離。然後使脂肪族二級胺部分經Boc保護且在純化後獲得產物。然後使用適宜鹼於適當溶劑中利用甲苯磺醯氯處理吲哚部分之NH-部分,以在純化後得到N-甲苯磺醯基衍生物。於適當溶劑中藉由酸處理去除Boc-保護基團,以在純化後得到期望三環5,6,7,8,9,10-六氫-7,10-環亞胺基環庚[b ]吲哚構建單元。在無鹼處理之情形下,獲得相應鹽。Option 5 The commercially available phenylhydrazine derivative ( Z = F) and the commercially available 3-pentanoxy-8-azabicyclo[3.2.1]octane-8-carboxylic acid tert-butyl are heated in a suitable solvent under acidic conditions. ester (Fisher indole synthesis), and after purification, tricyclic 5,6,7,8,9,10-hexahydro-7,10-cycloiminocycloheptyl [ b ] indole derivatives were obtained. In the case of 2- or 3-substituted phenylhydrazine derivatives, the regioisomers must be separated by supercritical fluid chromatography (SFC). The aliphatic secondary amine moiety is then Boc protected and the product is obtained after purification. The NH-portion of the indole moiety is then treated with toluenesulfonyl chloride using a suitable base in an appropriate solvent to obtain the N-toluenesulfonyl derivative after purification. The Boc-protecting group is removed by acid treatment in an appropriate solvent to obtain the desired tricyclic 5,6,7,8,9,10-hexahydro-7,10-cycloiminocycloheptane after purification [ b ]Indole building block. In the absence of alkali treatment, the corresponding salt is obtained.

方案6 利用N-羥基酞醯亞胺及氯化銅(I)及吡啶於適當溶劑中處理市售苯基酸酯衍生物(Z = F),以在純化後得到含有酞醯亞胺保護基團之相應羥胺衍生物。利用水合肼裂解酞醯亞胺保護基團且獲得呈鹽形式之相應羥胺衍生物。於適宜溶劑中在酸性條件下加熱羥胺衍生物與市售1,4-二氧雜-8-氮雜螺[4.5]癸烷(費舍爾吲哚合成),在純化後得到三環1,2,3,4-四氫苯并呋喃并[3,2-c ]吡啶衍生物。Option 6 Use N-hydroxyphthalimide, copper(I) chloride and pyridine to treat commercially available phenyl groups in appropriate solvents The acid ester derivative (Z = F) is purified to obtain the corresponding hydroxylamine derivative containing a phthalimine protecting group. The phthalimine protecting group is cleaved using hydrazine hydrate and the corresponding hydroxylamine derivative is obtained in salt form. The hydroxylamine derivative and commercially available 1,4-dioxa-8-azaspiro[4.5]decane (Fisher Indole Synthesis) were heated in a suitable solvent under acidic conditions to obtain tricyclic 1, 2,3,4-Tetrahydrobenzofuro[3,2- c ]pyridine derivatives.

方案7利用一級或二級胺於適當溶劑中且利用額外鹼處理含有兩個鹵素(Br、Cl)原子之市售苯并[d ]噻唑(G = Ph)或苯并[d ]噁唑(G = Ph)衍生物。脫離基X經由親核取代由一級或二級胺替代,以在純化後得到相應胺基取代之苯并[d ]噻唑或苯并[d ]噁唑衍生物。在反應性胺較少之情形下,藉由在微波條件下實施親核取代反應獲得期望苯并[d ]噻唑或苯并[d ]噁唑衍生物。利用嗎啉於適當溶劑中且利用額外鹼處理含有兩個鹵素(Br、Cl)原子之相應噻唑并[5,4-b ]吡啶(G = Py)及噻唑并[4,5-b ]吡啶(G = Py)衍生物,以在純化後得到相應嗎啉基-噻唑并[5,4-b ]吡啶(G = Py)及嗎啉基-噻唑并[4,5-b ]吡啶(G = Py)衍生物。利用嗎啉於適當溶劑中且利用額外鹼處理含有兩個鹵素(Br、Cl)原子之相應噻唑并[5,4-d ]嘧啶(G =嘧啶)衍生物,以在純化後得到相應嗎啉基-噻唑并[5,4-d ]嘧啶(G =嘧啶)衍生物。Option 7 Commercially available benzo[ d ]thiazole (G = Ph) or benzo[ d ]oxazole (G = Ph) derivatives. The leaving group In the case of less reactive amines, the desired benzo[ d ]thiazole or benzo[ d ]oxazole derivatives are obtained by performing a nucleophilic substitution reaction under microwave conditions. The corresponding thiazolo[5,4- b ]pyridines (G = Py) and thiazolo[4,5- b ]pyridines containing two halogen (Br, Cl) atoms were treated with morpholine in an appropriate solvent and with an additional base. (G = Py) derivatives to obtain the corresponding morpholino-thiazolo[5,4- b ]pyridine (G = Py) and morpholino-thiazolo[4,5- b ]pyridine (G = Py) derivatives. The corresponding thiazolo[5,4- d ]pyrimidine (G = pyrimidine) derivative containing two halogen (Br, Cl) atoms is treated with morpholine in an appropriate solvent and with an additional base to obtain the corresponding morpholine after purification Gyl-thiazolo[5,4- d ]pyrimidine (G = pyrimidine) derivatives.

方案8 利用嗎啉在微波條件下處理含有兩個鹵素(Br、Cl)原子之市售[1,2,4]三唑并[1,5-a ]吡啶衍生物,以在純化後得到親核置換產物。Option 8 Commercially available [1,2,4]triazolo[1,5- a ]pyridine derivatives containing two halogen (Br, Cl) atoms were treated with morpholine under microwave conditions to obtain nucleophilic substitution after purification product.

方案8a 利用嗎啉、3-氧雜-8-氮雜二環[3.2.1]辛烷或8-氧雜-3-氮雜二環[3.2.1]辛烷在微波條件下處理含有兩個溴原子之市售3,6-二溴嗒嗪及2,5-二溴吡嗪衍生物,以在純化後得到親核置換產物。Option 8a Utilizing morpholine, 3-oxa-8-azabicyclo[3.2.1]octane or 8-oxa-3-azabicyclo[3.2.1]octane under microwave conditions containing two bromines Atomized commercially available 3,6-dibromopyridazine and 2,5-dibromopyrazine derivatives to obtain nucleophilic substitution products after purification.

方案8b 利用嗎啉在微波條件下處理含有兩個鹵素(Br、Cl)原子之市售2,7-二氯喹啉、2-溴-6-氯-1,7-萘啶、2-溴-6-氯-1,8-萘啶及2,6-二氯喹啉衍生物,以在純化後得到親核置換產物。Option 8b Using morpholine to treat commercially available 2,7-dichloroquinoline, 2-bromo-6-chloro-1,7-naphthyridine, and 2-bromo-6-containing two halogen (Br, Cl) atoms under microwave conditions Chloro-1,8-naphthyridine and 2,6-dichloroquinoline derivatives to obtain nucleophilic substitution products after purification.

用於製備本發明化合物之一般合成方案:General synthetic scheme for preparing compounds of the invention:

方案9 將具有B = NCH3 或B = O之三環構建單元與胺基取代之苯并[d ]噻唑或苯并[d ]噁唑衍生物或經取代之吡啶衍生物經由鈀化學利用適宜鈀觸媒(氯-(2-二環己基膦基-2′,6′-二異丙氧基-1,1′-聯苯)[2-(2-胺基乙基)苯基]鈀(II)-甲基-第三丁基醚加成物;Pd(RuPhos) G1)、配體(2-二環己基膦基-2′,6′-二異丙氧基聯苯;RuPhos)及鹼(雙(三甲基矽基)胺基鋰;LiHMDS)於適宜溶劑中(四氫呋喃;THF)偶合,以在純化後得到期望之式(I)化合物。Option 9 The tricyclic building blocks with B= NCH3 or B=O are reacted with amino-substituted benzo[ d ]thiazole or benzo[ d ]oxazole derivatives or substituted pyridine derivatives via palladium chemistry using appropriate palladium contacts. (Chloro-(2-dicyclohexylphosphino-2′,6′-diisopropoxy-1,1′-biphenyl)[2-(2-aminoethyl)phenyl]palladium(II) )-Methyl-tert-butyl ether adduct; Pd(RuPhos) G1), ligand (2-dicyclohexylphosphino-2′,6′-diisopropoxybiphenyl; RuPhos) and base (Lithium bis(trimethylsilyl)amide; LiHMDS) is coupled in a suitable solvent (tetrahydrofuran; THF) to obtain the desired compound of formula (I) after purification.

方案10 將在吲哚/氮雜吲哚部分處含有N-甲苯磺醯基之三環構建單元與胺基取代之苯并[d ]噻唑、苯并[d ]噁唑、噻唑并[5,4-b ]吡啶(G = Py)、噻唑并[4,5-b ]吡啶(G = Py)衍生物、噻唑并[5,4-d ]嘧啶(G =嘧啶)或胺基取代之[1,2,4]三唑并[1,5-a ]吡啶衍生物或胺基取代之吡啶(L = Py)、吡嗪(L =吡嗪)、嗒嗪(L =嗒嗪)、嘧啶(L =嘧啶)衍生物或胺基取代之異喹啉、萘啶、喹唑啉衍生物經由鈀化學利用適宜鈀觸媒(參(二亞苄基丙酮)二鈀(0);Pd2 (dba)3 )、配體(2-二環己基膦基-2′,6′-二異丙氧基聯苯;RuPhos)及鹼(第三丁醇鈉;NaOtBu)於適宜溶劑中(1,4-二噁烷)偶合,以在純化後得到期望之式(I)化合物。或者,將在吲哚/氮雜吲哚部分處含有N-甲苯磺醯基之三環構建單元與胺基取代之苯并[d ]噻唑、苯并[d ]噁唑、噻唑并[5,4-b ]吡啶(G = Py)、噻唑并[4,5-b ]吡啶(G = Py)衍生物、噻唑并[5,4-d ]嘧啶(G =嘧啶)或胺基取代之[1,2,4]三唑并[1,5-a ]吡啶衍生物或胺基取代之吡啶(L = Py)、吡嗪(L =吡嗪)、嗒嗪(L =嗒嗪)、嘧啶(L =嘧啶)衍生物或胺基取代之異喹啉、萘啶、喹唑啉衍生物經由鈀化學利用適宜鈀觸媒(參(二亞苄基丙酮)二鈀(0);Pd2 (dba)3 )、配體(2-二環己基膦基-2′,6′-二異丙氧基聯苯;RuPhos)及弱鹼(碳酸銫;Cs2 CO3 )於適宜溶劑(1,4-二噁烷)中偶合,以在純化後得到經N-甲苯磺醯基保護之化合物。然後使用適宜鹼(碳酸銫;Cs2 CO3 )於適宜溶劑中(2-甲基THF、甲醇)在升高溫度(回流)下去除甲苯磺醯基保護基團,以在純化後得到期望之式(I)化合物。Option 10 The tricyclic building block containing N-toluenesulfonyl group at the indole/azaindole moiety and the amino-substituted benzo[ d ]thiazole, benzo[ d ]oxazole, thiazolo[5,4- b ]pyridine (G = Py), thiazolo[4,5- b ]pyridine (G = Py) derivatives, thiazolo[5,4- d ]pyrimidine (G = pyrimidine) or amino-substituted [1, 2,4]Triazolo[1,5- a ]pyridine derivatives or amino-substituted pyridine (L = Py), pyrazine (L = pyrazine), pyridazine (L = pyridazine), pyrimidine (L =pyrimidine) derivatives or amino-substituted isoquinoline, naphthyridine, quinazoline derivatives via palladium chemistry using a suitable palladium catalyst (see (dibenzylideneacetone) dipalladium (0); Pd 2 (dba) 3 ), ligand (2-dicyclohexylphosphino-2′,6′-diisopropoxybiphenyl; RuPhos) and base (sodium tert-butoxide; NaOtBu) in a suitable solvent (1,4- Dioxane) coupling to obtain the desired compound of formula (I) after purification. Alternatively, benzo[ d ]thiazole, benzo[ d ]oxazole, thiazolo[5, 4- b ]pyridine (G = Py), thiazolo[4,5- b ]pyridine (G = Py) derivatives, thiazolo[5,4- d ]pyrimidine (G = pyrimidine) or amino-substituted [ 1,2,4]Triazolo[1,5- a ]pyridine derivatives or amino-substituted pyridine (L = Py), pyrazine (L =pyrazine), pyridazine (L =pyridazine), pyrimidine (L = pyrimidine) derivatives or amino-substituted isoquinoline, naphthyridine, quinazoline derivatives via palladium chemistry using a suitable palladium catalyst (see (dibenzylideneacetone) dipalladium (0); Pd 2 ( dba) 3 ), ligand (2-dicyclohexylphosphino-2′,6′-diisopropoxybiphenyl; RuPhos) and weak base (cesium carbonate; Cs 2 CO 3 ) in a suitable solvent (1, 4-dioxane) to obtain the compound protected by N-toluenesulfonyl group after purification. The tosyl protecting group is then removed using a suitable base (cesium carbonate; Cs 2 CO 3 ) in a suitable solvent (2-methylTHF, methanol) at elevated temperature (reflux) to obtain the desired product after purification Compounds of formula (I).

方案11 利用適宜鹼(碳酸鉀;K2 CO3 )於適宜溶劑(DMF)中使在吲哚部分處含有N-甲苯磺醯基之三環構建單元與二-鹵化苯并[d ]噻唑及二-鹵化苯并[d ]噁唑偶合,以得到親核置換產物。然後使產物經由鈀化學與適宜鈀觸媒(參(二亞苄基丙酮)二鈀(0);Pd2 (dba)3 )、配體(2-二環己基膦基-2′,6′-二異丙氧基聯苯;RuPhos)及鹼(第三丁醇鈉;NaOtBu)於適宜溶劑中(1,4-二噁烷)中偶合,以在純化後得到期望之式(I)化合物。Plan 11 Using a suitable base (potassium carbonate; K 2 CO 3 ) in a suitable solvent (DMF), a tricyclic building block containing an N-toluenesulfonyl group at the indole moiety is combined with di-halogenated benzo[ d ]thiazole and di- Halogenated benzo[ d ]oxazoles are coupled to give nucleophilic displacement products. The product is then subjected to palladium chemistry with a suitable palladium catalyst (see (dibenzylideneacetone) dipalladium (0); Pd 2 (dba) 3 ), ligand (2-dicyclohexylphosphino-2′,6′ - Diisopropoxybiphenyl; RuPhos) and a base (sodium tert-butoxide; NaOtBu) are coupled in a suitable solvent (1,4-dioxane) to obtain the desired compound of formula (I) after purification .

用於製備本發明之氚標記化合物之一般合成方案:General synthetic scheme for preparing tritium-labeled compounds of the invention:

方案12 將觸媒添加至氚反應容器,之後添加本發明化合物於二氯甲烷中之溶液。將容器附接至氚管線並在-200℃下對氚氣加壓。將溶液在室溫下攪拌8小時,冷卻至-200℃且去除過量氣體。用甲醇沖洗反應燒瓶且使合併之有機相在真空下蒸發。藉由HPLC純化粗製材料,使移動相在真空下蒸發且將產物重新溶解於無水乙醇中。藉由質譜測定比活性。 可使用業內已知之任何適宜分析來量測Tau K18及全長(fl) Tau之解聚。闡述用於量測解聚能力之標準活體外分析。Plan 12 Catalyst is added to the tritium reaction vessel, followed by a solution of the compound of the invention in dichloromethane. Attach the container to the tritium line and pressurize the tritium gas at -200°C. The solution was stirred at room temperature for 8 hours, cooled to -200°C and excess gas removed. The reaction flask was rinsed with methanol and the combined organic phases were evaporated under vacuum. The crude material was purified by HPLC, the mobile phase was evaporated under vacuum and the product was redissolved in absolute ethanol. Specific activity was determined by mass spectrometry. Depolymerization of Tau K18 and full-length (fl) Tau can be measured using any suitable assay known in the art. Describes standard in vitro assays for measuring disaggregation capacity.

實例 所有試劑及溶劑均自商業來源獲得且不經進一步純化即使用。在Bruker AV 300及400 MHz光譜儀上於氘化溶劑中記錄1 H NMR光譜。化學位移(δ)係以百萬分率報告且偶合常數(J值)係以赫茲報告。自旋多重性係由以下符號指示:s (單峰)、d (雙重峰)、t (三重峰)、q (四重峰)、m (多重峰)、bs (寬單峰)。在具有6130 Chemstation之Agilent 1290 Infinity II光譜儀上及具有6130 Chemstation之Agilent 1200 Infinity II光譜儀上獲得質譜。使用Agilent 7890B氣相層析儀及5977B質譜儀來收集GC-MS數據。在PerkinElmer光譜儀上獲得紅外光譜。使用矽膠(Fluka:矽膠60, 0.063-0.2 mm)及如在具體實例中所指示之適宜溶劑來實施層析。利用具有HP-Sil或KP-NH SNAP柱之Biotage Isolera (Biotage)及具體實例中所指示之溶劑梯度進行急速純化。在矽膠板上利用UV檢測實施薄層層析(TLC)。 Examples All reagents and solvents were obtained from commercial sources and used without further purification. 1 H NMR spectra were recorded on Bruker AV 300 and 400 MHz spectrometers in deuterated solvents. Chemical shifts (δ) are reported in parts per million and coupling constants (J values) are reported in Hertz. Spin multiplicity is indicated by the following symbols: s (singlet), d (doublet), t (triplet), q (quartet), m (multiplet), bs (broad singlet). Mass spectra were acquired on an Agilent 1290 Infinity II spectrometer with a 6130 Chemstation and an Agilent 1200 Infinity II spectrometer with a 6130 Chemstation. GC-MS data were collected using an Agilent 7890B gas chromatograph and 5977B mass spectrometer. Infrared spectra were obtained on a PerkinElmer spectrometer. Chromatography was performed using silica gel (Fluka: Silica gel 60, 0.063-0.2 mm) and appropriate solvents as indicated in the specific examples. Flash purification was performed using a Biotage Isolera (Biotage) with an HP-Sil or KP-NH SNAP column and a solvent gradient as indicated in the specific examples. Thin layer chromatography (TLC) was performed on silica plates using UV detection.

製備實例 1 步驟 A 在冰浴溫度下向4-氟苯基肼(1 g, 7.9 mmol)及4-側氧基六氫吡啶-1-甲酸第三丁基酯(1.2 g, 8.3 mmol)於1,4-二噁烷(10 mL)中之溶液添加濃H2 SO4 (1 mL)。然後將反應混合物在110℃下加熱3 h。將該反應混合物冷卻至室溫,將沈澱物過濾出。將固體溶解於水中,經NaOH溶液鹼化並用DCM (二氯甲烷)萃取。將有機相分離且經Na2 SO4 乾燥並去除溶劑,得到呈淡黃色固體之標題化合物(950 mg, 59%)。 MS: 191 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 10.91 (s, 1H), 7.23-7.24 (m, 1H), 7.09-7.09 (m, 1H), 6.80-6.81 (m, 1H), 3.91 (s, 2H), 3.11 (t,J = 5.56 Hz, 2H), 2.75 (d,J = 4.96 Hz, 2H)。步驟 B 向來自上文步驟A之標題化合物(0.95 g, 4.77 mmol)於THF (四氫呋喃)中之溶液添加二碳酸二-第三丁基酯(Boc2 O) (1.5 g),且將混合物攪拌過夜。如藉由TLC所證實,在反應完成後,將溶劑去除且使用Biotage Isolera One純化系統,採用EtOAc/庚烷梯度(10/80 => 80/20),在矽膠管柱上純化粗製反應混合物,得到呈淡黃色黏性液體之標題化合物(1.1 g, 78%)。 MS: 291 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 11.00 (s, 1H), 7.26 (q,J = 4.52 Hz, 1H), 7.18 (t,J = 8.12 Hz, 1H), 6.83-6.83 (m, 1H), 4.49 (s, 2H), 3.69 (t,J = 5.64 Hz, 2H), 2.76 (s, 2H), 1.43 (s, 9H)。步驟 C 向來自上文步驟B之標題化合物(0.41 g, 1.41 mmol)於THF (5 mL)中之溶液添加氫化鈉(0.15 g, 6.25 mmol),之後添加對甲苯磺醯氯(TsCl) (0.29 g, 1.45 mmol)。將反應混合物攪拌10 min。將該混合物溶解於EtOAc (20 ml)中,並用水及鹽水洗滌且經Na2 SO4 乾燥。使用Biotage Isolera One純化系統,採用EtOAc/庚烷梯度(20/80 => 80/20),在矽膠管柱上純化粗產物,得到標題化合物(0.45 g, 72%)。 MS: 445 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 8.03-8.04 (m, 1H), 7.77 (d,J = 8.20 Hz, 2H), 7.36-7.38 (m, 3H), 7.15-7.16 (m, 1H), 4.43 (s, 2H), 3.69 (t,J = 5.64 Hz, 2H), 3.08 (s, 2H), 2.32 (s, 3H), 1.43 (s, 9H)。步驟 D 向來自上文步驟C之標題化合物(0.42 g, 0.915 mmol)於二氯甲烷中之溶液添加於1,4-二噁烷中之2N HCl (5 mL)。將反應混合物攪拌過夜。在反應完成後,使該反應混合物蒸發以去除溶劑並用二乙醚洗滌,得到呈灰白色固體之標題化合物(0.2 g, 58%)。 MS: 345 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 9.61 (s, 1H), 8.01-8.02 (m, 1H), 7.82 (d,J = 8.40 Hz, 2H), 7.45-7.45 (m, 1H), 7.39 (d,J = 8.40 Hz, 2H), 7.20-7.21 (m, 1H), 4.25 (s, 2H), 3.49 (s, 2H), 3.35 (d, J = 3.60 Hz, 2H), 2.34 (s, 3H)。步驟 E 向來自上文步驟D之標題化合物(5.0 g, 13 mmol)於二氯甲烷(50 mL)中之溶液添加三乙胺(5 mL)並攪拌10 min。將反應混合物用二氯甲烷(20 mL)稀釋,用水(2 × 30 mL)及NaCl飽和溶液(30 mL)洗滌。使合併之有機層經硫酸鈉乾燥並在真空下濃縮,得到呈游離鹼之標題化合物(定量產率)。 Preparation Example 1 Step A 4-Fluorophenylhydrazine (1 g, 7.9 mmol) and 4-pentyloxyhexahydropyridine-1-carboxylic acid tert-butyl ester (1.2 g, 8.3 mmol) were added to 1,4-di To a solution in oxane (10 mL) was added concentrated H 2 SO 4 (1 mL). The reaction mixture was then heated at 110 °C for 3 h. The reaction mixture was cooled to room temperature and the precipitate was filtered off. The solid was dissolved in water, basified with NaOH solution and extracted with DCM (dichloromethane). The organic phase was separated and dried over Na2SO4 and the solvent was removed to give the title compound as a pale yellow solid (950 mg, 59%). MS: 191 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 10.91 (s, 1H), 7.23-7.24 (m, 1H), 7.09-7.09 (m, 1H), 6.80-6.81 (m, 1H), 3.91 (s, 2H), 3.11 (t, J = 5.56 Hz, 2H), 2.75 (d, J = 4.96 Hz, 2H). Step B To a solution of the title compound from Step A above (0.95 g, 4.77 mmol) in THF (tetrahydrofuran) was added di-tert-butyl dicarbonate (Boc 2 O) (1.5 g) and the mixture was stirred Stay overnight. After the reaction was complete, as confirmed by TLC, the solvent was removed and the crude reaction mixture was purified on a silica column using a Biotage Isolera One purification system using an EtOAc/heptane gradient (10/80 => 80/20), The title compound (1.1 g, 78%) was obtained as a pale yellow viscous liquid. MS: 291 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 11.00 (s, 1H), 7.26 (q, J = 4.52 Hz, 1H), 7.18 (t, J = 8.12 Hz, 1H), 6.83-6.83 ( m, 1H), 4.49 (s, 2H), 3.69 (t, J = 5.64 Hz, 2H), 2.76 (s, 2H), 1.43 (s, 9H). Step C To a solution of the title compound from Step B above (0.41 g, 1.41 mmol) in THF (5 mL) was added sodium hydride (0.15 g, 6.25 mmol), followed by p-toluenesulfonyl chloride (TsCl) (0.29 g, 1.45 mmol). The reaction mixture was stirred for 10 min. The mixture was dissolved in EtOAc ( 20 ml) and washed with water and brine and dried over Na2SO4 . The crude product was purified on a silica column using a Biotage Isolera One purification system using an EtOAc/heptane gradient (20/80 => 80/20) to obtain the title compound (0.45 g, 72%). MS: 445 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 8.03-8.04 (m, 1H), 7.77 (d, J = 8.20 Hz, 2H), 7.36-7.38 (m, 3H), 7.15-7.16 (m , 1H), 4.43 (s, 2H), 3.69 (t, J = 5.64 Hz, 2H), 3.08 (s, 2H), 2.32 (s, 3H), 1.43 (s, 9H). Step D To a solution of the title compound from Step C above (0.42 g, 0.915 mmol) in dichloromethane was added 2N HCl in 1,4-dioxane (5 mL). The reaction mixture was stirred overnight. After the reaction was complete, the reaction mixture was evaporated to remove the solvent and washed with diethyl ether to give the title compound as an off-white solid (0.2 g, 58%). MS: 345 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 9.61 (s, 1H), 8.01-8.02 (m, 1H), 7.82 (d, J = 8.40 Hz, 2H), 7.45-7.45 (m, 1H ), 7.39 (d, J = 8.40 Hz, 2H), 7.20-7.21 (m, 1H), 4.25 (s, 2H), 3.49 (s, 2H), 3.35 (d, J = 3.60 Hz, 2H), 2.34 (s, 3H). Step E To a solution of the title compound from Step D above (5.0 g, 13 mmol) in dichloromethane (50 mL) was added triethylamine (5 mL) and stirred for 10 min. The reaction mixture was diluted with dichloromethane (20 mL), washed with water (2 × 30 mL) and saturated NaCl solution (30 mL). The combined organic layers were dried over sodium sulfate and concentrated in vacuo to afford the title compound as the free base (quantitative yield).

製備實例 2 步驟 A 向來自製備實例1步驟B之標題化合物(0.41 g, 1.41 mmol)於THF (5 mL)中之溶液添加氫化鈉(0.067 g, 2.82 mmol),將懸浮液在室溫下攪拌20分鐘。將反應混合物再次冷卻至0℃且添加碘甲烷(0.24 g, 1.45 mmol)。將反應混合物攪拌3 h。使反應混合物溶解於EtOAc (15 mL)中,並用水及鹽水洗滌且經Na2 SO4 乾燥。使用Biotage Isolera One純化系統,採用EtOAc/己烷梯度(20/80 => 80/20),在矽膠管柱上純化粗產物,得到標題化合物(0.3 g, 70%)。 MS: 304 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 7.38-7.39 (m, 1H), 7.22 (dd,J = 2.40, 10.00 Hz, 1H), 6.90-6.91 (m, 1H), 4.49 (s, 2H), 3.71 (t,J = 6.00 Hz, 2H), 3.62 (s, 3H), 2.79 (t,J = 5.20 Hz, 2H), 1.40 (s, 9H) 步驟 B 向來自上文步驟A之標題化合物(0.3 mg, 0.986 mmol)於二氯甲烷中之溶液添加於1,4-二噁烷中之2 N HCl (5 mL)。將反應混合物攪拌過夜。使該反應混合物蒸發以去除溶劑並用二乙醚洗滌,得到呈灰白色固體之標題化合物(0.12 g, 60%)。 MS: 205.08 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 9.45 (br, 1H), 7.45-7.46 (m, 1H), 7.30-7.31 (m, 1H), 6.98-7.00 (m, 1H), 4.26 (s, 2H), 3.66 (s, 3H), 3.49 (d,J = 4.80 Hz, 2H), 3.06 (t,J = 6.00 Hz, 2H), 2.50 (s, 9H)。 Preparation Example 2 Step A To a solution of the title compound from Preparative Example 1, Step B (0.41 g, 1.41 mmol) in THF (5 mL) was added sodium hydride (0.067 g, 2.82 mmol) and the suspension was stirred at room temperature for 20 min. The reaction mixture was cooled again to 0°C and methyl iodide (0.24 g, 1.45 mmol) was added. The reaction mixture was stirred for 3 h. The reaction mixture was dissolved in EtOAc (15 mL) and washed with water and brine and dried over Na2SO4 . The crude product was purified on a silica column using a Biotage Isolera One purification system using an EtOAc/hexane gradient (20/80 => 80/20) to obtain the title compound (0.3 g, 70%). MS: 304 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 7.38-7.39 (m, 1H), 7.22 (dd, J = 2.40, 10.00 Hz, 1H), 6.90-6.91 (m, 1H), 4.49 (s , 2H), 3.71 (t, J = 6.00 Hz, 2H), 3.62 (s, 3H), 2.79 (t, J = 5.20 Hz, 2H), 1.40 (s, 9H) . Step B To a solution of the title compound from Step A above (0.3 mg, 0.986 mmol) in dichloromethane was added 2 N HCl in 1,4-dioxane (5 mL). The reaction mixture was stirred overnight. The reaction mixture was evaporated to remove the solvent and washed with diethyl ether to give the title compound as an off-white solid (0.12 g, 60%). MS: 205.08 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 9.45 (br, 1H), 7.45-7.46 (m, 1H), 7.30-7.31 (m, 1H), 6.98-7.00 (m, 1H), 4.26 (s, 2H), 3.66 (s, 3H), 3.49 (d, J = 4.80 Hz, 2H), 3.06 (t, J = 6.00 Hz, 2H), 2.50 (s, 9H).

製備實例 3 步驟 A 在冰浴溫度下向1-(4-氟苯基)-1-甲基肼(2 g, 14.0 mmol)及4-側氧基六氫吡啶-1-甲酸第三丁基酯(2.84 g, 14.0 mmol)於1,4-二噁烷(10 mL)中之溶液添加濃H2 SO4 (1 mL)。然後將反應混合物在110℃下加熱過夜。將該反應混合物冷卻至室溫,將沈澱物過濾出。棄掉濾液。將固體溶解於水(10 mL)中,利用NaOH溶液將pH調整至14,並用二氯甲烷(150 mL)萃取混合物。將有機相用水及鹽水洗滌且經Na2 SO4 乾燥。將溶劑去除,得到呈游離鹼之標題化合物(1.3 g, 46%)。 MS: 205.08 (M+H)+ Preparation Example 3 Step A : 1-(4-fluorophenyl)-1-methylhydrazine (2 g, 14.0 mmol) and 4-side oxyhexahydropyridine-1-carboxylic acid tert-butyl ester (2.84 g, 14.0 mmol) in 1,4-dioxane (10 mL) was added concentrated H 2 SO 4 (1 mL). The reaction mixture was then heated at 110°C overnight. The reaction mixture was cooled to room temperature and the precipitate was filtered off. Discard the filtrate. The solid was dissolved in water (10 mL), the pH was adjusted to 14 using NaOH solution, and the mixture was extracted with dichloromethane (150 mL). The organic phase was washed with water and brine and dried over Na2SO4 . The solvent was removed to give the title compound as the free base (1.3 g, 46%). MS: 205.08 (M+H) + .

製備實例 4 步驟 A 在冰浴溫度下向(4-甲氧基苯基)肼(1 g, 5.6 mmol)及4-側氧基六氫吡啶-1-甲酸第三丁基酯(1.13 g, 5.6 mmol)於1,4-二噁烷(10 mL)中之溶液添加濃H2 SO4 (1 mL)。然後將反應混合物在110℃下加熱3 h。使該反應混合物冷卻至室溫,將沈澱物過濾出。將固體溶解於水中,經NaOH溶液鹼化並用二氯甲烷萃取。將有機相分離且經Na2 SO4 乾燥,過濾並將溶劑去除,得到呈淡黃色黏性液體之標題化合物(0.60 g, 53%)。粗產物原樣用於下一步驟。 MS: 203 (M+H)+步驟 B 向來自上文步驟A之標題化合物(0.60 mg, 2.9 mmol)於THF中之溶液添加二碳酸二-第三丁基酯(0.65 g, 2.9 mmol),且將混合物攪拌過夜。將溶劑去除且使用Biotage Isolera One純化系統,採用EtOAc/己烷梯度(80/20),在矽膠管柱上純化粗產物,得到呈淡黃色固體之標題化合物(0.55 g, 62%)。 MS: 303 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 10.71 (bs, 1H), 7.17 (d,J = 8.40 Hz, 1H), 6.89 (s, 1H), 6.65-6.66 (m, 1H), 4.49 (s, 2H), 3.75 (s, 3H), 3.69 (t,J = 5.60 Hz, 2H), 2.74 (t,J = 5.60 Hz, 2H), 1.44 (s, 9H)。步驟 C 向來自上文步驟B之標題化合物(0.55 g, 1.8 mmol)於THF (5 mL)中之溶液添加氫化鈉(0.087 g, 3.6 mmol),之後添加對甲苯磺醯氯(0.342 g, 1.8 mmol)。將反應混合物攪拌45分鐘。在反應完成後,將該反應混合物溶解於EtOAc (200 mL)中,並用水及鹽水洗滌且經Na2 SO4 乾燥。使用Biotage Isolera One純化系統,採用EtOAc/己烷梯度(20/80),在矽膠管柱上純化粗產物,得到呈灰白色固體之標題化合物(0.45 g ,45%)。 MS: 457 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 7.92 (d,J = 9.20 Hz, 1H), 7.71 (d,J = 8.40 Hz, 2H), 7.35 (d,J = 8.00 Hz, 2H), 7.01 (s, 1H), 6.91-6.92 (m, 1H), 4.42 (s, 2H), 3.77 (s, 3H), 3.68 (t,J = 5.60 Hz, 2H), 3.05 (bs, 2H), 2.31 (s, 3H), 1.43 (s, 9H)。步驟 D 向來自上文步驟C之標題化合物(0.450 g, 0.986 mmol)於二氯甲烷中之溶液添加於1,4-二噁烷中之2 N HCl (5 mL)。將反應混合物攪拌過夜。在反應完成後,使該反應混合物蒸發以去除溶劑並用二乙醚洗滌,得到呈灰白色固體之化合物(0.23 g, 65%)。 MS: 357 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 9.67 (bs, 1H), 7.89 (d,J = 9.20 Hz, 1H), 7.77 (d,J = 8.40 Hz, 2H), 7.36 (d,J = 8.00 Hz, 2H), 7.11 (d,J = 2.40 Hz, 1H), 6.93-6.94 (m, 1H), 4.24 (s, 2H), 3.77 (s, 3H), 3.48-3.49 (m, 2H), 3.34-3.35 (m, 2H), 2.33 (s, 3H)。 Preparation Example 4 Step A : Add (4-methoxyphenyl)hydrazine (1 g, 5.6 mmol) and 4-side oxyhexahydropyridine-1-carboxylic acid tert-butyl ester (1.13 g, 5.6 mmol) at ice bath temperature. To a solution in 1,4-dioxane (10 mL) was added concentrated H2SO4 (1 mL). The reaction mixture was then heated at 110 °C for 3 h. The reaction mixture was allowed to cool to room temperature and the precipitate was filtered off. The solid was dissolved in water, basified with NaOH solution and extracted with dichloromethane. The organic phase was separated and dried over Na2SO4 , filtered and the solvent removed to give the title compound (0.60 g, 53%) as a pale yellow viscous liquid. The crude product was used as such in the next step. MS: 203 (M+H) + . Step B To a solution of the title compound from Step A above (0.60 mg, 2.9 mmol) in THF was added di-tert-butyl dicarbonate (0.65 g, 2.9 mmol) and the mixture was stirred overnight. The solvent was removed and the crude product was purified on a silica column using a Biotage Isolera One purification system using an EtOAc/hexane gradient (80/20) to give the title compound as a pale yellow solid (0.55 g, 62%). MS: 303 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 10.71 (bs, 1H), 7.17 (d, J = 8.40 Hz, 1H), 6.89 (s, 1H), 6.65-6.66 (m, 1H), 4.49 (s, 2H), 3.75 (s, 3H), 3.69 (t, J = 5.60 Hz, 2H), 2.74 (t, J = 5.60 Hz, 2H), 1.44 (s, 9H). Step C To a solution of the title compound from Step B above (0.55 g, 1.8 mmol) in THF (5 mL) was added sodium hydride (0.087 g, 3.6 mmol) followed by p-toluenesulfonyl chloride (0.342 g, 1.8 mmol). The reaction mixture was stirred for 45 minutes. After the reaction was complete, the reaction mixture was dissolved in EtOAc ( 200 mL) and washed with water and brine and dried over Na2SO4 . The crude product was purified on a silica column using a Biotage Isolera One purification system and an EtOAc/hexane gradient (20/80) to obtain the title compound (0.45 g, 45%) as an off-white solid. MS: 457 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 7.92 (d, J = 9.20 Hz, 1H), 7.71 (d, J = 8.40 Hz, 2H), 7.35 (d, J = 8.00 Hz, 2H) , 7.01 (s, 1H), 6.91-6.92 (m, 1H), 4.42 (s, 2H), 3.77 (s, 3H), 3.68 (t, J = 5.60 Hz, 2H), 3.05 (bs, 2H), 2.31 (s, 3H), 1.43 (s, 9H). Step D To a solution of the title compound from Step C above (0.450 g, 0.986 mmol) in dichloromethane was added 2 N HCl in 1,4-dioxane (5 mL). The reaction mixture was stirred overnight. After the reaction was completed, the reaction mixture was evaporated to remove the solvent and washed with diethyl ether to obtain the compound as an off-white solid (0.23 g, 65%). MS: 357 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 9.67 (bs, 1H), 7.89 (d, J = 9.20 Hz, 1H), 7.77 (d, J = 8.40 Hz, 2H), 7.36 (d, J = 8.00 Hz, 2H), 7.11 (d, J = 2.40 Hz, 1H), 6.93-6.94 (m, 1H), 4.24 (s, 2H), 3.77 (s, 3H), 3.48-3.49 (m, 2H ), 3.34-3.35 (m, 2H), 2.33 (s, 3H).

製備實例 5 步驟 A 在0℃下向來自製備實例4步驟B之標題化合物(0.55 g, 1.8 mmol)於THF (5 mL)中之溶液添加氫化鈉(0.087 g, 3.6 mmol)。將懸浮液在室溫下攪拌20分鐘。將反應混合物再次冷卻至0℃且添加碘甲烷(0.255 g, 1.8 mmol)。將反應混合物攪拌45分鐘。在反應完成後,將該反應混合物溶解於EtOAc (20 ml)中,並用水及鹽水洗滌且經Na2 SO4 乾燥,過濾並濃縮。使用Biotage Isolera One純化系統,採用EtOAc/己烷梯度(80/20),在矽膠管柱上純化粗製混合物,得到呈淡棕色固體之標題化合物(0.40 g, 45%)。 MS: 317 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 7.29 (d,J = 8.80 Hz, 2H), 6.93 (s, 1H), 6.72-6.73 (m, 1H), 4.50 (s, 2H), 3.76 (s, 3H), 3.70-3.72 (m, 2H), 3.59 (s, 3H), 2.78-2.79 (m, 2H), 1.44 (s, 9H)。步驟 B 向來自上文步驟A之標題化合物(0.4 g, 1.26 mmol)於二氯甲烷中之溶液添加於1,4-二噁烷中之2 N HCl (5 mL)。將反應混合物攪拌過夜。在反應完成後,使該反應混合物蒸發以去除溶劑並用二乙醚洗滌,得到呈白色固體之標題化合物(0.2 g, 73%)。 MS: 217 (M+H)+ Preparation Example 5 Step A To a solution of the title compound from Preparative Example 4, Step B (0.55 g, 1.8 mmol) in THF (5 mL) was added sodium hydride (0.087 g, 3.6 mmol) at 0°C. The suspension was stirred at room temperature for 20 minutes. The reaction mixture was cooled again to 0°C and iodomethane (0.255 g, 1.8 mmol) was added. The reaction mixture was stirred for 45 minutes. After the reaction was complete, the reaction mixture was dissolved in EtOAc ( 20 ml) and washed with water and brine and dried over Na2SO4 , filtered and concentrated. The crude mixture was purified on a silica column using a Biotage Isolera One purification system using an EtOAc/hexane gradient (80/20) to obtain the title compound (0.40 g, 45%) as a light brown solid. MS: 317 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 7.29 (d, J = 8.80 Hz, 2H), 6.93 (s, 1H), 6.72-6.73 (m, 1H), 4.50 (s, 2H), 3.76 (s, 3H), 3.70-3.72 (m, 2H), 3.59 (s, 3H), 2.78-2.79 (m, 2H), 1.44 (s, 9H). Step B To a solution of the title compound from Step A above (0.4 g, 1.26 mmol) in dichloromethane was added 2 N HCl in 1,4-dioxane (5 mL). The reaction mixture was stirred overnight. After the reaction was complete, the reaction mixture was evaporated to remove the solvent and washed with diethyl ether to give the title compound as a white solid (0.2 g, 73%). MS: 217 (M+H) + .

製備實例 6 步驟 A 於微波爐中將2-氯-5-氟吡啶(10 g, 76.34 mmol)及N- 甲基肼(6 mL)之混合物在180℃下輻照1 h。在反應完成後,使反應混合物冷卻並將反應混合物傾倒於冰冷水中且用二氯甲烷(2 × 50 mL)萃取。將有機相分離且經Na2 SO4 乾燥並去除溶劑,得到呈淡棕色固體之標題化合物(10 g,粗製)。粗產物原樣用於下一步驟。 MS: 143 (M+H)+步驟 B 向來自上文步驟A之標題化合物(10 g, 70.84 mmol)於甲醇(100 mL)中之溶液添加4-側氧基六氫吡啶-1-甲酸第三丁基酯(16.9 g, 85 mmol)並攪拌過夜。將溶劑去除,使用Biotage Isolera One純化系統,採用甲醇/DCM梯度(1/99),在矽膠管柱上純化粗製反應混合物,得到呈棕色黏性油狀物之標題化合物(5 g, 22%)。 MS: 323 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 7.82-7.84 (m, 1H), 7.13-7.14 (m, 1H), 6.72-6.73 (m, 1H), 4.51 (bs, 2H), 3.77 (s, 3H), 3.71-3.73 (m, 2H), 2.76-2.78 (m, 2H), 1.43 (s, 9H)。步驟 C 於微波爐中將來自上文步驟B之標題化合物(5 g, 15.50 mmol)及二乙二醇(5 mL)之混合物在180℃下輻照1 h。在反應完成後,使反應混合物冷卻並將反應混合物傾倒於冰冷水中並用二氯甲烷(2 × 50 mL)萃取。將有機相分離且經Na2 SO4 乾燥並去除溶劑,藉由製備型HPLC (管柱:Phenomenex Gemini C18 (150*4.6)mm, 3.0 μm。移動相A:於Milli-Q水中之10 mM乙酸銨。移動相B:乙腈。流速:1.0 ml\min)純化粗製混合物,得到呈棕色固體之標題化合物(0.9 g, 29%)。 MS: 206 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 7.87-7.89 (m, 1H), 7.46-7.47 (m, 1H), 4.53 (bs, 2H), 3.77 (s, 3H), 3.71-3.73 (m, 2H), 2.76-2.78 (m, 2H)。 Preparation Example 6 Step A : Irradiate a mixture of 2-chloro-5-fluoropyridine (10 g, 76.34 mmol) and N- methylhydrazine (6 mL) in a microwave oven at 180°C for 1 h. After the reaction was completed, the reaction mixture was allowed to cool and poured into ice-cold water and extracted with dichloromethane (2 × 50 mL). The organic phase was separated and dried over Na2SO4 and the solvent was removed to give the title compound (10 g, crude) as a light brown solid. The crude product was used as such in the next step. MS: 143 (M+H) + . Step B To a solution of the title compound from Step A above (10 g, 70.84 mmol) in methanol (100 mL) was added tert-butyl 4-pentanoxyhexahydropyridine-1-carboxylate (16.9 g, 85 mmol) and stir overnight. The solvent was removed, and the crude reaction mixture was purified on a silica column using a Biotage Isolera One purification system using a methanol/DCM gradient (1/99) to obtain the title compound (5 g, 22%) as a brown viscous oil. . MS: 323 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 7.82-7.84 (m, 1H), 7.13-7.14 (m, 1H), 6.72-6.73 (m, 1H), 4.51 (bs, 2H), 3.77 (s, 3H), 3.71-3.73 (m, 2H), 2.76-2.78 (m, 2H), 1.43 (s, 9H). Step C A mixture of the title compound from Step B above (5 g, 15.50 mmol) and diethylene glycol (5 mL) was irradiated in a microwave oven at 180°C for 1 h. After the reaction was completed, the reaction mixture was allowed to cool and poured into ice-cold water and extracted with dichloromethane (2 × 50 mL). The organic phase was separated and dried over Na 2 SO 4 and the solvent was removed by preparative HPLC (column: Phenomenex Gemini C18 (150*4.6) mm, 3.0 μm. Mobile phase A: 10 mM acetic acid in Milli-Q water Ammonium. Mobile phase B: acetonitrile. Flow rate: 1.0 ml\min). The crude mixture was purified to obtain the title compound (0.9 g, 29%) as a brown solid. MS: 206 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 7.87-7.89 (m, 1H), 7.46-7.47 (m, 1H), 4.53 (bs, 2H), 3.77 (s, 3H), 3.71-3.73 (m, 2H), 2.76-2.78 (m, 2H).

製備實例 7 步驟 A 在0℃下向3-(氟苯基)肼(1 g, 6.1 mmol)及4-側氧基六氫吡啶-1-甲酸第三丁基酯(1.2 g, 6.1 mmol)於1,4-二噁烷(10 mL)中之溶液添加濃H2 SO4 (1 mL)。然後將反應混合物升溫至25℃並在110℃下加熱3 h。將該反應混合物冷卻至室溫且將沈澱物過濾出。將固體溶解於水中,經NaOH溶液鹼化並用二氯甲烷萃取。將有機相分離且經Na2 SO4 乾燥並去除溶劑,得到呈淡黃色固體之區域異構物混合物(0.65 g, 56%)。 MS: 191.1 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 10.87 (bs, 1H), 7.26-7.30 (m, 1H), 7.02-7.05 (m, 1H), 6.74-6.79 (m, 1H), 3.83 (bs, 2H), 2.99-3.02 (m, 2H), 2.65-2.66 (m, 2H)。步驟 B 向區域異構物混合物(0.65 g, 3.15 mmol)於THF中之溶液添加二碳酸二-第三丁基酯(0.757 g, 3.47 mmol),且將混合物攪拌12 h。在反應完成後(藉由TLC監測),將溶劑在減壓下濃縮以產生粗產物。藉由矽膠(60-120目)管柱層析使用己烷: EtOAc (70:30)純化該粗產物,得到呈黃色固體之區域異構物7-氟-1,3,4,5-四氫-2H -吡啶并[4,3-b ]吲哚-2-甲酸第三丁基酯及9-氟-1,3,4,5-四氫-2H -吡啶并[4,3-b ]吲哚-2-甲酸第三丁基酯之混合物(0.750 g, 61%),其比率為分別約70:30。 MS: 291.2 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 11.01 (bs, 1H), 7.36-7.39 (m, 1H), 7.06-7.09 (m, 1H), 6.79-6.84 (m, 1H), 4.51 (bs, 2H), 3.68-3.71 (m, 2H), 2.74-2.76 (m, 2H), 1.38 (s, 9H)。步驟 C 藉由SFC手性管柱(Chiracel OJ-H;管柱:X-bridge C8 (50 × 4.6) mm, 3.5 μm,移動相A:於水中之0.1% TFA,移動相B:0.1% TFA乙腈)分離區域異構物混合物(0.750 mg, 70:30),得到具有100%手性純度之呈淡黃色固體之第二溶析標題化合物7-氟-1,3,4,5-四氫-2H -吡啶并[4,3-b ]吲哚-2-甲酸第三丁基酯(0.4 mg, 53%)。第一溶析標題化合物9-氟-1,3,4,5-四氫-2H -吡啶并[4,3-b ]吲哚-2-甲酸第三丁基酯分離為具有100%手性純度之淡黃色固體(0.25 g, 33%)。 第二溶析標題化合物: MS: 291.2 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 11.01 (bs, 1H), 7.36-7.39 (m, 1H), 7.06-7.09 (m, 1H), 6.79-6.84 (m, 1H), 4.51 (bs, 2H), 3.68-3.71 (m, 2H), 2.74-2.77 (m, 2H), 1.44 (s, 9H)。 RT=2.08 min。 第一溶析標題化合物: MS: 291.2 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 11.22 (s, 1H), 7.13 (d,J = 8.08 Hz, 1H), 6.96-6.97 (m, 1H), 6.69-6.71 (m, 1H), 4.63 (s, 2H), 3.69-3.70 (m, 2H), 2.68-2.76 (m, 2H), 1.44 (s, 9H)。 RT=1.74 min。步驟 D 向來自上文步驟C之第二溶析標題化合物(0.4 g, 1.37 mmol)於THF (5 mL)中之溶液添加氫化鈉(0.099 mg, 4.137 mmol),之後添加對甲苯磺醯氯(0.288 g, 1.51 mmol)。將反應混合物攪拌30分鐘。將該混合物溶解於EtOAc (20 ml)中,並用水及鹽水洗滌且經Na2 SO4 乾燥。使用Biotage Isolera One純化系統,採用EtOAc/庚烷梯度(20/80 => 80/20),在矽膠管柱上純化粗產物,得到標題化合物(0.3 g, 49%)。 MS: 445 (M+H)+1 H-NMR (400 MHz,氯仿-d ) δ = 7.92-7.94 (m, 1H), 7.68-7.70 (m, 1H), 7.25-7.29 (m, 4H), 7.00-7.04 (m, 1H), 4.50 (bs, 2H), 3.76 (bs, 2H), 3.12 (bs, 2H), 2.38 (s, 3H), 1.51 (s, 9H)。步驟 E 向來自上文步驟D之標題化合物(0.3 g, 0.676 mmol)於二氯甲烷中之溶液添加於1,4-二噁烷中之2 N HCl (5 mL)。將反應混合物攪拌12 h。在反應完成後,使該反應混合物蒸發以去除溶劑並用二乙醚洗滌,得到呈灰白色固體之標題化合物(0.2 g 78%)。 MS: 345 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 9.50 (bs, 2H), 7.80-7.87 (m, 2H), 7.78 (d,J = 2.00 Hz, 1H), 7.57-7.61 (m, 1H), 7.40 (d,J = 8.16 Hz, 2H), 7.18-7.23 (m, 1H), 4.27 (bs, 2H), 3.56 (bs, 2H), 3.47 (bs, 2H), 2.34 (s, 3H)。 Preparation Example 7 Step A To 3-(fluorophenyl)hydrazine (1 g, 6.1 mmol) and 4-pentyloxyhexahydropyridine-1-carboxylic acid tert-butyl ester (1.2 g, 6.1 mmol) in 1,4- To a solution in dioxane (10 mL) was added concentrated H 2 SO 4 (1 mL). The reaction mixture was then warmed to 25 °C and heated at 110 °C for 3 h. The reaction mixture was cooled to room temperature and the precipitate was filtered off. The solid was dissolved in water, basified with NaOH solution and extracted with dichloromethane. The organic phase was separated and dried over Na2SO4 and the solvent was removed to give a mixture of regioisomers as a pale yellow solid (0.65 g, 56%). MS: 191.1 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 10.87 (bs, 1H), 7.26-7.30 (m, 1H), 7.02-7.05 (m, 1H), 6.74-6.79 (m, 1H), 3.83 (bs, 2H), 2.99-3.02 (m, 2H), 2.65-2.66 (m, 2H). Step B To a solution of the regioisomer mixture (0.65 g, 3.15 mmol) in THF was added di-tert-butyl dicarbonate (0.757 g, 3.47 mmol), and the mixture was stirred for 12 h. After the reaction was complete (monitored by TLC), the solvent was concentrated under reduced pressure to yield crude product. The crude product was purified by silica gel (60-120 mesh) column chromatography using hexane: EtOAc (70:30) to obtain the regioisomer 7-fluoro-1,3,4,5-tetrakis as a yellow solid. Hydro- 2H -pyrido[4,3- b ]indole-2-carboxylic acid tert-butyl ester and 9-fluoro-1,3,4,5-tetrahydro- 2H -pyrido[4,3 - b ] A mixture of tert-butyl indole-2-carboxylate (0.750 g, 61%) in a ratio of approximately 70:30 respectively. MS: 291.2 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 11.01 (bs, 1H), 7.36-7.39 (m, 1H), 7.06-7.09 (m, 1H), 6.79-6.84 (m, 1H), 4.51 (bs, 2H), 3.68-3.71 (m, 2H), 2.74-2.76 (m, 2H), 1.38 (s, 9H). Step C is carried out by SFC chiral column (Chiracel OJ-H; column: X-bridge C8 (50 × 4.6) mm, 3.5 μm, mobile phase A: 0.1% TFA in water, mobile phase B: 0.1% TFA Acetonitrile) separated the regioisomer mixture (0.750 mg, 70:30) to obtain the second eluted title compound 7-fluoro-1,3,4,5-tetrahydro as a pale yellow solid with 100% chiral purity. -2H -pyrido[4,3- b ]indole-2-carboxylic acid tert-butyl ester (0.4 mg, 53%). First elution of the title compound 9-fluoro-1,3,4,5-tetrahydro- 2H -pyrido[4,3- b ]indole-2-carboxylic acid tert-butyl ester was isolated as having 100% chiral Light yellow solid with high purity (0.25 g, 33%). The title compound was eluted second: MS: 291.2 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 11.01 (bs, 1H), 7.36-7.39 (m, 1H), 7.06-7.09 (m, 1H), 6.79-6.84 (m, 1H), 4.51 (bs, 2H), 3.68-3.71 (m, 2H), 2.74-2.77 (m, 2H), 1.44 (s, 9H). RT=2.08 min. First eluate the title compound: MS: 291.2 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 11.22 (s, 1H), 7.13 (d, J = 8.08 Hz, 1H), 6.96-6.97 (m, 1H), 6.69-6.71 (m, 1H ), 4.63 (s, 2H), 3.69-3.70 (m, 2H), 2.68-2.76 (m, 2H), 1.44 (s, 9H). RT=1.74 min. Step D To a second solution of the title compound from Step C above (0.4 g, 1.37 mmol) in THF (5 mL) was added sodium hydride (0.099 mg, 4.137 mmol) followed by p-toluenesulfonyl chloride ( 0.288 g, 1.51 mmol). The reaction mixture was stirred for 30 minutes. The mixture was dissolved in EtOAc ( 20 ml) and washed with water and brine and dried over Na2SO4 . The crude product was purified on a silica column using a Biotage Isolera One purification system using an EtOAc/heptane gradient (20/80 => 80/20) to obtain the title compound (0.3 g, 49%). MS: 445 (M+H) + . 1 H-NMR (400 MHz, chloroform- d ) δ = 7.92-7.94 (m, 1H), 7.68-7.70 (m, 1H), 7.25-7.29 (m, 4H), 7.00-7.04 (m, 1H), 4.50 (bs, 2H), 3.76 (bs, 2H), 3.12 (bs, 2H), 2.38 (s, 3H), 1.51 (s, 9H). Step E To a solution of the title compound from Step D above (0.3 g, 0.676 mmol) in dichloromethane was added 2 N HCl in 1,4-dioxane (5 mL). The reaction mixture was stirred for 12 h. After the reaction was complete, the reaction mixture was evaporated to remove the solvent and washed with diethyl ether to give the title compound as an off-white solid (0.2 g 78%). MS: 345 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 9.50 (bs, 2H), 7.80-7.87 (m, 2H), 7.78 (d, J = 2.00 Hz, 1H), 7.57-7.61 (m, 1H ), 7.40 (d, J = 8.16 Hz, 2H), 7.18-7.23 (m, 1H), 4.27 (bs, 2H), 3.56 (bs, 2H), 3.47 (bs, 2H), 2.34 (s, 3H) .

製備實例 8 步驟 A 在0℃下向(2-氯-3-氟苯基)肼(10 g, 62.5 mmol)及4-側氧基六氫吡啶-1-甲酸第三丁基酯(12 g, 62.5 mmol)於1,4-二噁烷(100 mL)中之溶液添加濃H2 SO4 (10 mL)。然後將反應混合物升溫至25℃並在110℃下加熱3 h。使該反應混合物冷卻至室溫且將沈澱物過濾出。將固體溶解於水中,經NaOH溶液鹼化並用二氯甲烷萃取。將有機相分離且經Na2 SO4 乾燥並去除溶劑,得到呈淡黃色固體之標題化合物(10 g, 72%)。 MS: 225 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 11.23 (bs, 1H), 7.27-7.28 (m, 1H), 6.94-6.96 (m, 1H), 3.82 (s, 2H), 2.98-3.00 (m, 2H), 2.68 (d,J = 4.72 Hz, 2H)。步驟 B 向來自上文步驟A之標題化合物(10 g, 44.5 mmol)於THF (100 mL)中之溶液添加二碳酸二-第三丁基酯(10.5 g, 46.5 mmol),且將混合物攪拌12 h。在反應完成後(藉由TLC監測),將溶劑在減壓下濃縮以產生粗產物。藉由矽膠(60-120目)管柱層析純化該粗產物,得到呈黃色固體之標題化合物(12 g, 85%)。 MS: 325.1 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 11.43 (s, 1H), 7.36-7.38 (m, 1H), 6.97-7.00 (m, 1H), 4.51 (s, 2H), 3.68-3.69 (m, 2H), 2.76-2.78 (m, 2H), 1.43 (s, 9H)。步驟 C 向來自上文步驟B之標題化合物(5 g, 15.3 mmol)於無水甲醇(50 mL)中之溶液添加三乙胺(6.74 mL, 46.18 mmol)及10% Pd/C (0.2 mg, 20% wt)。在10巴壓力下進行氫化達16小時。經由矽藻土墊過濾反應混合物並在真空下濃縮,得到標題化合物(4 g, 90%)。 MS: 291.2 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 11.01 (bs, 1H), 7.36-7.39 (m, 1H), 7.06-7.09 (m, 1H), 6.79-6.84 (m, 1H), 4.51 (bs, 2H), 3.68-3.71 (m, 2H), 2.74-2.77 (m, 2H), 1.44 (s, 9H)。步驟 D 向來自上文步驟C之標題化合物(4 g, 13.7 mmol)於THF (40 mL)中之溶液添加氫化鈉(9.9 g, 41.23 mmol),之後添加對甲苯磺醯氯(2.88 g, 15.1 mmol)。將反應混合物攪拌30 min。將該混合物溶解於EtOAc (200 ml)中,並用水及鹽水洗滌且經Na2 SO4 乾燥。使用Biotage Isolera One純化系統,採用EtOAc/庚烷梯度(20/80 => 80/20),在矽膠管柱上純化粗產物,得到標題化合物(5 g, 82%)。 MS: 445 (M+H)+1 H-NMR (400 MHz,氯仿-d ) δ = 7.92-7.94 (m, 1H), 7.68-7.70 (m, 1H), 7.25-7.29 (m, 4H), 7.00-7.04 (m, 1H), 4.50 (bs, 2H), 3.76 (bs, 2H), 3.12 (bs, 2H), 2.38 (s, 3H), 1.51 (s, 9H)。步驟 E 向來自上文步驟D之標題化合物(3 g, 6.76 mmol)於二氯甲烷(30 mL)中之溶液添加於1,4-二噁烷中之2 N HCl (15 mL)。將反應混合物攪拌12 h。在反應完成後,使該反應混合物蒸發以去除溶劑並用二乙醚洗滌,得到呈灰白色固體之標題化合物(2 g, 78%)。 MS: 345 (M+H)+1 H NMR (400 MHz, DMSO-d 6 ) δ = 9.50 (bs, 2H), 7.80-7.87 (m, 2H), 7.78 (d,J = 2.00 Hz, 1H), 7.57-7.61 (m, 1H), 7.40 (d,J = 8.16 Hz, 2H), 7.18-7.23 (m, 1H), 4.27 (bs, 2H), 3.56 (bs, 2H), 3.47 (bs, 2H), 2.34 (s, 3H)。 Preparation Example 8 Step A : (2-Chloro-3-fluorophenyl)hydrazine (10 g, 62.5 mmol) and 4-pentanoxyhexahydropyridine-1-carboxylic acid tert-butyl ester (12 g, 62.5 mmol) at 0°C ) in 1,4-dioxane (100 mL) was added concentrated H 2 SO 4 (10 mL). The reaction mixture was then warmed to 25 °C and heated at 110 °C for 3 h. The reaction mixture was allowed to cool to room temperature and the precipitate was filtered off. The solid was dissolved in water, basified with NaOH solution and extracted with dichloromethane. The organic phase was separated and dried over Na2SO4 and the solvent was removed to give the title compound as a pale yellow solid (10 g, 72%). MS: 225 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 11.23 (bs, 1H), 7.27-7.28 (m, 1H), 6.94-6.96 (m, 1H), 3.82 (s, 2H), 2.98-3.00 (m, 2H), 2.68 (d, J = 4.72 Hz, 2H). Step B To a solution of the title compound from Step A above (10 g, 44.5 mmol) in THF (100 mL) was added di-tert-butyl dicarbonate (10.5 g, 46.5 mmol) and the mixture was stirred for 12 h. After the reaction was complete (monitored by TLC), the solvent was concentrated under reduced pressure to yield crude product. The crude product was purified by silica gel (60-120 mesh) column chromatography to obtain the title compound (12 g, 85%) as a yellow solid. MS: 325.1 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 11.43 (s, 1H), 7.36-7.38 (m, 1H), 6.97-7.00 (m, 1H), 4.51 (s, 2H), 3.68-3.69 (m, 2H), 2.76-2.78 (m, 2H), 1.43 (s, 9H). Step C To a solution of the title compound from Step B above (5 g, 15.3 mmol) in anhydrous methanol (50 mL) was added triethylamine (6.74 mL, 46.18 mmol) and 10% Pd/C (0.2 mg, 20 %wt). The hydrogenation was carried out at a pressure of 10 bar for 16 hours. The reaction mixture was filtered through a pad of celite and concentrated in vacuo to give the title compound (4 g, 90%). MS: 291.2 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 11.01 (bs, 1H), 7.36-7.39 (m, 1H), 7.06-7.09 (m, 1H), 6.79-6.84 (m, 1H), 4.51 (bs, 2H), 3.68-3.71 (m, 2H), 2.74-2.77 (m, 2H), 1.44 (s, 9H). Step D To a solution of the title compound from Step C above (4 g, 13.7 mmol) in THF (40 mL) was added sodium hydride (9.9 g, 41.23 mmol) followed by p-toluenesulfonyl chloride (2.88 g, 15.1 mmol). The reaction mixture was stirred for 30 min. The mixture was dissolved in EtOAc ( 200 ml) and washed with water and brine and dried over Na2SO4 . The crude product was purified on a silica column using a Biotage Isolera One purification system using an EtOAc/heptane gradient (20/80 => 80/20) to obtain the title compound (5 g, 82%). MS: 445 (M+H) + . 1 H-NMR (400 MHz, chloroform- d ) δ = 7.92-7.94 (m, 1H), 7.68-7.70 (m, 1H), 7.25-7.29 (m, 4H), 7.00-7.04 (m, 1H), 4.50 (bs, 2H), 3.76 (bs, 2H), 3.12 (bs, 2H), 2.38 (s, 3H), 1.51 (s, 9H). Step E To a solution of the title compound from Step D above (3 g, 6.76 mmol) in dichloromethane (30 mL) was added 2 N HCl in 1,4-dioxane (15 mL). The reaction mixture was stirred for 12 h. After the reaction was complete, the reaction mixture was evaporated to remove the solvent and washed with diethyl ether to give the title compound as an off-white solid (2 g, 78%). MS: 345 (M+H) + . 1 H NMR (400 MHz, DMSO- d 6 ) δ = 9.50 (bs, 2H), 7.80-7.87 (m, 2H), 7.78 (d, J = 2.00 Hz, 1H), 7.57-7.61 (m, 1H) , 7.40 (d, J = 8.16 Hz, 2H), 7.18-7.23 (m, 1H), 4.27 (bs, 2H), 3.56 (bs, 2H), 3.47 (bs, 2H), 2.34 (s, 3H).

製備實例 9 步驟 A 向來自製備實例8步驟C之標題化合物(0.4 mg, 1.37 mmol)於THF (5 mL)中之溶液添加氫化鈉(0.099 g, 4.137 mmol),之後添加碘甲烷(0.102 ml, 1.64 mmol)。將反應混合物攪拌2 h。將該混合物溶解於EtOAc (20 ml)中,並用水及鹽水洗滌且經Na2 SO4 乾燥。使用Biotage Isolera One純化系統,採用EtOAc/庚烷梯度(20/80 => 80/20),在矽膠管柱上純化粗產物,得到標題化合物(0.3 mg, 73%)。 MS: 305.37 (M+H)+步驟 B 向來自上文步驟A之標題化合物(0.3 mg, 0.986 mmol)於二氯甲烷中之溶液添加於1,4-二噁烷中之2 N HCl (5 mL)。將反應混合物攪拌過夜。在反應完成後,使該反應混合物蒸發以去除溶劑並用二乙醚洗滌,得到呈灰白色固體之標題化合物(0.18 g 75%)。 MS: 205 (M+H)+ Preparation Example 9 Step A To a solution of the title compound from Preparative Example 8, Step C (0.4 mg, 1.37 mmol) in THF (5 mL) was added sodium hydride (0.099 g, 4.137 mmol) followed by methyl iodide (0.102 ml, 1.64 mmol). . The reaction mixture was stirred for 2 h. The mixture was dissolved in EtOAc ( 20 ml) and washed with water and brine and dried over Na2SO4 . The crude product was purified on a silica column using a Biotage Isolera One purification system using an EtOAc/heptane gradient (20/80 => 80/20) to obtain the title compound (0.3 mg, 73%). MS: 305.37 (M+H) + . Step B To a solution of the title compound from Step A above (0.3 mg, 0.986 mmol) in dichloromethane was added 2 N HCl in 1,4-dioxane (5 mL). The reaction mixture was stirred overnight. After the reaction was complete, the reaction mixture was evaporated to remove the solvent and washed with diethyl ether to give the title compound as an off-white solid (0.18 g 75%). MS: 205 (M+H) + .

製備實例 10 步驟 A 向來自製備實例7步驟C之第一溶析標題化合物(0.2 mg, 0.67 mmol)於THF (5 mL)中之溶液添加氫化鈉(0.048 g, 2.137 mmol),之後添加對甲苯磺醯氯(0.144 g, 0.76 mmol)。將反應混合物攪拌30分鐘。將該混合物溶解於EtOAc (20 ml)中,並用水及鹽水洗滌且經Na2 SO4 乾燥。使用Biotage Isolera One純化系統,採用EtOAc/庚烷梯度(20/80 => 80/20),在矽膠管柱上純化粗產物,得到標題化合物(0.155 g, 50%)。 MS: 445 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 7.80-7.82 (m, 3H), 7.31-7.32 (m, 3H), 7.07-7.09 (m, 1H), 4.56 (s, 2H), 3.68-3.69 (m, 2H), 3.09 (bs, 2H), 2.33 (s, 3H), 1.43 (s, 9H)。步驟 B 向來自上文步驟A之標題化合物(0.15 g, 0.337 mmol)於二氯甲烷中之溶液添加於1,4-二噁烷中之2 N HCl (5 mL)。將反應混合物攪拌12 h。在反應完成後,使該反應混合物蒸發以去除溶劑並用二乙醚洗滌,得到呈灰白色固體之標題化合物(0.1 g, 71%)。 MS: 345 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 9.49 (bs, 2H), 7.85-7.87 (m, 3H), 7.35-7.36 (m, 3H), 7.12-7.14 (m, 1H), 4.39 (s, 2H), 3.48 (bs, 2H), 3.17 (bs, 2H), 2.35 (s, 3H)。 Preparation Example 10 Step A To the first solution of the title compound (0.2 mg, 0.67 mmol) in THF (5 mL) from Preparative Example 7, Step C, was added sodium hydride (0.048 g, 2.137 mmol) followed by p-toluenesulfonyl chloride. (0.144 g, 0.76 mmol). The reaction mixture was stirred for 30 minutes. The mixture was dissolved in EtOAc ( 20 ml) and washed with water and brine and dried over Na2SO4 . The crude product was purified on a silica column using a Biotage Isolera One purification system using an EtOAc/heptane gradient (20/80 => 80/20) to obtain the title compound (0.155 g, 50%). MS: 445 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 7.80-7.82 (m, 3H), 7.31-7.32 (m, 3H), 7.07-7.09 (m, 1H), 4.56 (s, 2H), 3.68 -3.69 (m, 2H), 3.09 (bs, 2H), 2.33 (s, 3H), 1.43 (s, 9H). Step B To a solution of the title compound from Step A above (0.15 g, 0.337 mmol) in dichloromethane was added 2 N HCl in 1,4-dioxane (5 mL). The reaction mixture was stirred for 12 h. After the reaction was complete, the reaction mixture was evaporated to remove the solvent and washed with diethyl ether to give the title compound as an off-white solid (0.1 g, 71%). MS: 345 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 9.49 (bs, 2H), 7.85-7.87 (m, 3H), 7.35-7.36 (m, 3H), 7.12-7.14 (m, 1H), 4.39 (s, 2H), 3.48 (bs, 2H), 3.17 (bs, 2H), 2.35 (s, 3H).

製備實例 11 步驟 A 向2-(4-氟苯基)-4,4,5,5-四甲基-1,3,2-二氧雜硼雜環戊烷(5g, 22.51 mmol)於二氯乙烷(250 mL)中之溶液添加N-羥基酞醯亞胺(7.34 g, 45.03 mmol)、氯化銅(I)(2.22 g, 22.51 mmol)、吡啶(2.75 mL, 33.75 mmol)、分子篩(5 g),且將反應混合物加熱至70℃持續12 h。在反應完成後,經由矽藻土床過濾該反應混合物並用乙酸乙酯洗滌。將乙酸乙酯層濃縮且使用Biotage Isolera One純化系統,採用己烷/EtOAc梯度(90/10 => 80/20),在矽膠上純化粗產物,得到呈白色固體之標題化合物(1.4 g, 24%)。 MS: 258.22 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 7.92-7.93 (m, 4H), 7.33-7.34 (m, 2H), 7.20-7.22 (m, 2H)。步驟 B 向來自上文步驟A之標題化合物(1.4g, 5.44 mmol)於氯仿:甲醇(9:1, 100 mL)中之溶液添加水合肼(0.81 g, 16.32 mmol),且將反應混合物在25℃下攪拌12 h。在反應完成後,經由矽藻土床過濾該反應混合物並用二氯甲烷洗滌。將二氯甲烷層濃縮,且在0℃下將粗產物添加至乙醚(5 mL)及於乙醚(2 mL)中之2 N HCl,然後將反應混合物在25℃下攪拌30 min。在反應完成後,將該反應混合物過濾,在真空下乾燥,得到灰白色固體(0.44 g, 50%)。 MS: 128.12 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 8.03-8.04 (m, 2H), 7.76-7.76 (m, 2H)。步驟 C 在0℃下向來自上文步驟B之標題化合物(0.44 g, 2.7 mmol)及1,4-二氧雜-8-氮雜螺[4.5]癸烷(0.44 g, 3.07 mmol)於1,4-二噁烷(5 mL)中之溶液添加濃H2 SO4 (0.5 mL)。將反應混合物升溫至25℃,且然後在微波條件下在150℃下進一步加熱1 h。使反應混合物冷卻至25℃,且將沈澱物過濾出。將固體溶解於水中,經NaOH溶液鹼化並用二氯甲烷萃取。將有機相分離且經Na2 SO4 乾燥,並將溶劑濃縮,粗產物原樣用於下一步驟(0.270 g, 52%)。 MS: 192.21 (M+H)+ Preparation Example 11 Step A : 2-(4-Fluorophenyl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (5g, 22.51 mmol) in dichloroethane (250 mL), add N-hydroxyphthalimide (7.34 g, 45.03 mmol), copper (I) chloride (2.22 g, 22.51 mmol), pyridine (2.75 mL, 33.75 mmol), and molecular sieve (5 g ), and the reaction mixture was heated to 70 °C for 12 h. After the reaction was complete, the reaction mixture was filtered through a bed of celite and washed with ethyl acetate. The ethyl acetate layer was concentrated and the crude product was purified on silica gel using a Biotage Isolera One purification system using a hexane/EtOAc gradient (90/10 => 80/20) to give the title compound as a white solid (1.4 g, 24 %). MS: 258.22 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 7.92-7.93 (m, 4H), 7.33-7.34 (m, 2H), 7.20-7.22 (m, 2H). Step B To a solution of the title compound from Step A above (1.4 g, 5.44 mmol) in chloroform:methanol (9:1, 100 mL) was added hydrazine hydrate (0.81 g, 16.32 mmol), and the reaction mixture was heated at 25 Stir for 12 h at ℃. After the reaction was complete, the reaction mixture was filtered through a bed of celite and washed with dichloromethane. The dichloromethane layer was concentrated and the crude product was added to diethyl ether (5 mL) and 2 N HCl in diethyl ether (2 mL) at 0 °C, and the reaction mixture was stirred at 25 °C for 30 min. After the reaction was completed, the reaction mixture was filtered and dried under vacuum to obtain an off-white solid (0.44 g, 50%). MS: 128.12 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 8.03-8.04 (m, 2H), 7.76-7.76 (m, 2H). Step C To the title compound from Step B above (0.44 g, 2.7 mmol) and 1,4-dioxa-8-azaspiro[4.5]decane (0.44 g, 3.07 mmol) were added to 1 , to a solution of 4-dioxane (5 mL) was added concentrated H 2 SO 4 (0.5 mL). The reaction mixture was warmed to 25°C and then further heated at 150°C for 1 h under microwave conditions. The reaction mixture was cooled to 25°C and the precipitate was filtered off. The solid was dissolved in water, basified with NaOH solution and extracted with dichloromethane. The organic phase was separated and dried over Na2SO4 , and the solvent was concentrated and the crude product was used unchanged in the next step (0.270 g, 52%). MS: 192.21 (M+H) + .

製備實例 12 步驟 A 在0℃下向4-(氟苯基)肼鹽酸鹽(10 g, 0.061 mol)及3-側氧基-8-氮雜二環[3.2.1]辛烷-8-甲酸第三丁基酯(13.9 g, 0.61 mol)於1,4-二噁烷(100 mL)中之溶液添加濃H2 SO4 (10 mL),且然後將混合物加熱至100℃持續12 h。在反應完成後,將反應混合物冷卻至室溫,並藉由高真空將溶劑去除以得到粗製材料。使用30%氫氧化鈉溶液使粗製材料鹼化,且沈澱出固體。將沈澱固體過濾,用水(100 mL)及二乙醚(100 mL)洗滌,然後將固體在管線真空下乾燥16 h,得到呈淡棕色固體之標題化合物(11 g, 83%)。 MS: 216.10 (M+H)+步驟 B 在0℃下向來自上文步驟A之標題化合物(11 g, 0.051 mol)於THF (50 mL)中之溶液添加三乙胺(11 mL, 0.076 mol)及二碳酸二-第三丁基酯(11.13 g, 0.051 mol),且然後在室溫下攪拌12 h。如藉由TLC所證實,在反應完成後,將溶劑去除且藉由矽膠管柱使用於石油醚中之40%至50%乙酸乙酯純化粗製反應混合物,得到呈淡黃色固體之標題化合物(7.4 g, 46%)。 MS: 316.15 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 10.94 (bs, 1H), 7.23-7.24 (m, 2H), 6.80-6.81 (m, 1H), 5.09-5.11 (m, 1H), 4.46 (bs, 1H), 3.23-3.27 (m, 1H), 2.51-2.57 (m, 1H), 2.22-2.26 (m, 1H), 2.00-2.07 (m, 1H), 1.59-1.60 (m, 1H), 1.41-1.44 (m, 2H), 1.20-1.27 (m, 9H)。步驟 C 在0℃下向來自上文步驟B之標題化合物(3 g, 0.009 mol)於THF (30 mL)中之溶液逐份添加氫化鈉(60%於礦物油中;0.57 g, 0.014 mol)。添加完成後,將反應混合物在室溫下攪拌30分鐘,且然後將反應混合物再次冷卻至0℃。在0℃下向此混合物逐滴添加溶解於THF (20 mL)中之對甲苯磺醯氯(2.16 g, 0.11 mol)。添加完成後,將反應混合物在室溫下攪拌2 h。如藉由TLC所證實,在反應完成後,將反應混合物冷卻至0℃並用冰水淬滅,之後使用乙酸乙酯(100 mL)萃取。將乙酸乙酯層用水(30 mL)及鹽水溶液(30 mL)洗滌。使有機層經Na2 SO4 乾燥,過濾並蒸發以得到粗產物,其藉由矽膠管柱使用於石油醚中之15%至25%乙酸乙酯進行純化,得到呈灰白色固體之標題化合物(2.9 g, 65%)。 MS: 470.16 (M+H)+步驟 D 在0℃下向來自上文步驟C之標題化合物(2.9 g , 0.006 mol)於二氯甲烷(10 mL)中之溶液添加於1,4-二噁烷中之4 N HCl (20 mL)。將反應混合物在環境溫度下攪拌12 h。在反應完成後,使該反應混合物蒸發以去除溶劑並用二乙醚(10 mL)洗滌,得到呈灰白色固體之標題化合物(2.4 g, 98%)。 MS: 370.15 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 8.07-8.08 (m, 1H), 7.78-7.80 (m, 1H), 7.30-7.31 (m, 3H), 7.10-7.11 (m, 1H), 5.13 (d,J = 4.80 Hz, 1H), 4.52 (d,J = 4.80 Hz, 1H), 3.64-3.65 (m, 4H), 3.30-3.30 (m, 1H), 2.22-2.24 (m, 7H), 1.92 (bs, 1H)。 Preparation Example 12 Step A : Add 4-(fluorophenyl)hydrazine hydrochloride (10 g, 0.061 mol) and 3-side oxy-8-azabicyclo[3.2.1]octane-8-carboxylic acid at 0°C. To a solution of tributyl ester (13.9 g, 0.61 mol) in 1,4-dioxane (100 mL) was added concentrated H2SO4 (10 mL), and the mixture was then heated to 100 °C for 12 h . After the reaction was completed, the reaction mixture was cooled to room temperature and the solvent was removed by high vacuum to obtain crude material. The crude material was basified using 30% sodium hydroxide solution and a solid precipitated. The precipitated solid was filtered, washed with water (100 mL) and diethyl ether (100 mL), and the solid was dried under line vacuum for 16 h to obtain the title compound (11 g, 83%) as a light brown solid. MS: 216.10 (M+H) + . Step B To a solution of the title compound from Step A above (11 g, 0.051 mol) in THF (50 mL) was added triethylamine (11 mL, 0.076 mol) and di-tert-butyl dicarbonate at 0°C. ester (11.13 g, 0.051 mol) and then stirred at room temperature for 12 h. After the reaction was complete, as confirmed by TLC, the solvent was removed and the crude reaction mixture was purified via a silica column using 40% to 50% ethyl acetate in petroleum ether to give the title compound (7.4) as a pale yellow solid. g, 46%). MS: 316.15 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 10.94 (bs, 1H), 7.23-7.24 (m, 2H), 6.80-6.81 (m, 1H), 5.09-5.11 (m, 1H), 4.46 (bs, 1H), 3.23-3.27 (m, 1H), 2.51-2.57 (m, 1H), 2.22-2.26 (m, 1H), 2.00-2.07 (m, 1H), 1.59-1.60 (m, 1H) , 1.41-1.44 (m, 2H), 1.20-1.27 (m, 9H). Step C To a solution of the title compound from Step B above (3 g, 0.009 mol) in THF (30 mL) was added sodium hydride (60% in mineral oil; 0.57 g, 0.014 mol) portionwise at 0°C. . After the addition was complete, the reaction mixture was stirred at room temperature for 30 minutes, and then the reaction mixture was cooled to 0°C again. To this mixture was added dropwise p-toluenesulfonyl chloride (2.16 g, 0.11 mol) dissolved in THF (20 mL) at 0°C. After the addition was complete, the reaction mixture was stirred at room temperature for 2 h. After the reaction was complete, as confirmed by TLC, the reaction mixture was cooled to 0°C and quenched with ice water, followed by extraction with ethyl acetate (100 mL). The ethyl acetate layer was washed with water (30 mL) and brine solution (30 mL). The organic layer was dried over Na2SO4 , filtered and evaporated to give the crude product, which was purified by a silica column using 15% to 25% ethyl acetate in petroleum ether to give the title compound (2.9 ) as an off-white solid. g, 65%). MS: 470.16 (M+H) + . Step D To a solution of the title compound from Step C above (2.9 g, 0.006 mol) in dichloromethane (10 mL) was added 4 N HCl in 1,4-dioxane (20 mL) at 0°C. ). The reaction mixture was stirred at ambient temperature for 12 h. After the reaction was complete, the reaction mixture was evaporated to remove the solvent and washed with diethyl ether (10 mL) to afford the title compound as an off-white solid (2.4 g, 98%). MS: 370.15 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 8.07-8.08 (m, 1H), 7.78-7.80 (m, 1H), 7.30-7.31 (m, 3H), 7.10-7.11 (m, 1H) , 5.13 (d, J = 4.80 Hz, 1H), 4.52 (d, J = 4.80 Hz, 1H), 3.64-3.65 (m, 4H), 3.30-3.30 (m, 1H), 2.22-2.24 (m, 7H ), 1.92 (bs, 1H).

製備實例 13 步驟 A 向2,6-二氯苯并[d]噻唑(5 g, 24.5 mmol)於無水二氯甲烷(50 mL)中之溶液添加嗎啉(3.19 g, 36.6 mmol),且將混合物冷卻至0℃。向此冷的反應混合物逐滴添加三乙胺(3.71 g, 36.7 mmol),且將混合物在室溫下攪拌4 h。在反應完成後,利用H2 O (2 × 20 mL)處理反應混合物並用二氯甲烷萃取。將有機層分離,經Na2 SO4 乾燥,過濾並蒸發,得到白色固體,將其與二乙醚一起研磨,得到標題化合物(5 g, 96%)。 MS: 213.4 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 7.80 (d,J = 8.00 Hz, 1H), 7.53(d,J = 2.00 Hz, 1H), 7.13-7.14 (m, 1H), 3.74-3.75 (m, 4H), 3.56-3.57 (m, 4H)。 Preparation Example 13 Step A To a solution of 2,6-dichlorobenzo[d]thiazole (5 g, 24.5 mmol) in dry dichloromethane (50 mL) was added morpholine (3.19 g, 36.6 mmol) and the mixture was cooled to 0℃. To the cold reaction mixture, triethylamine (3.71 g, 36.7 mmol) was added dropwise, and the mixture was stirred at room temperature for 4 h. After the reaction was complete, the reaction mixture was treated with H 2 O (2 × 20 mL) and extracted with dichloromethane. The organic layer was separated, dried over Na2SO4 , filtered and evaporated to give a white solid, which was triturated with diethyl ether to give the title compound (5 g, 96%). MS: 213.4 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 7.80 (d, J = 8.00 Hz, 1H), 7.53(d, J = 2.00 Hz, 1H), 7.13-7.14 (m, 1H), 3.74- 3.75 (m, 4H), 3.56-3.57 (m, 4H).

製備實例 14 步驟 A 向2,5-二氯苯并[d]噻唑(5 g, 24.5 mmol)於無水二氯甲烷(50 mL)中之溶液添加嗎啉(3.19 g, 36.6 mmol),且將混合物冷卻至0℃。向此冷的反應混合物逐滴添加三乙胺(3.71 g, 36.7 mmol),且將混合物在室溫下攪拌4 h。在反應完成後,利用H2 O (2 × 20 mL)處理反應混合物並用二氯甲烷萃取。將有機層分離,經Na2 SO4 乾燥,過濾並蒸發,得到白色固體,將其與二乙醚一起研磨,得到標題化合物(4.5 g, 86%)。 MS: 255.4 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 7.82 (d,J = 8.00 Hz, 1H), 7.50 (d,J = 2.00 Hz, 1H), 7.11-7.11 (m, 1H), 3.72-3.73 (m, 4H), 3.55-3.56 (m, 4H)。 Preparation Example 14 Step A To a solution of 2,5-dichlorobenzo[d]thiazole (5 g, 24.5 mmol) in dry dichloromethane (50 mL) was added morpholine (3.19 g, 36.6 mmol) and the mixture was cooled to 0℃. To the cold reaction mixture, triethylamine (3.71 g, 36.7 mmol) was added dropwise, and the mixture was stirred at room temperature for 4 h. After the reaction was complete, the reaction mixture was treated with H 2 O (2 × 20 mL) and extracted with dichloromethane. The organic layer was separated, dried over Na2SO4 , filtered and evaporated to give a white solid which was triturated with diethyl ether to give the title compound (4.5 g, 86%). MS: 255.4 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 7.82 (d, J = 8.00 Hz, 1H), 7.50 (d, J = 2.00 Hz, 1H), 7.11-7.11 (m, 1H), 3.72- 3.73 (m, 4H), 3.55-3.56 (m, 4H).

製備實例 15 步驟 A 向2,6-二氯苯并[d]噁唑(5 g, 26.8 mmol)於無水二氯甲烷(50 mL)中之溶液添加嗎啉(3.50 g, 40.3 mmol),且將混合物冷卻至0℃。向此冷的反應混合物逐滴添加三乙胺(4.0 g, 39.6 mmol),且將混合物在室溫下攪拌4 h。在反應完成後,利用H2 O (2 × 20 mL)處理反應混合物並用二氯甲烷萃取。將有機層分離,經Na2 SO4 乾燥,過濾並蒸發,得到白色固體,將其與二乙醚一起研磨,得到標題化合物(5 g, 78%)。 MS: 239.2 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 7.59 (d,J = 2.80 Hz, 1H), 7.30 (d,J = 11.20 Hz, 1H), 7.21 (dd,J = 2.80, 11.20 Hz, 1H), 3.71-3.74 (m, 4H), 3.57-3.60 (m, 4H)。 Preparation Example 15 Step A To a solution of 2,6-dichlorobenzo[d]oxazole (5 g, 26.8 mmol) in dry dichloromethane (50 mL) was added morpholine (3.50 g, 40.3 mmol) and the mixture was cooled to 0℃. To the cold reaction mixture, triethylamine (4.0 g, 39.6 mmol) was added dropwise, and the mixture was stirred at room temperature for 4 h. After the reaction was complete, the reaction mixture was treated with H 2 O (2 × 20 mL) and extracted with dichloromethane. The organic layer was separated, dried over Na2SO4 , filtered and evaporated to give a white solid which was triturated with diethyl ether to give the title compound (5 g, 78%). MS: 239.2 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 7.59 (d, J = 2.80 Hz, 1H), 7.30 (d, J = 11.20 Hz, 1H), 7.21 (dd, J = 2.80, 11.20 Hz, 1H), 3.71-3.74 (m, 4H), 3.57-3.60 (m, 4H).

製備實例 16 步驟 A 向2,5-二氯苯并[d]噁唑(5 g, 26.8 mmol)於無水二氯甲烷(50 mL)中之溶液添加嗎啉(3.50 g, 40.3 mmol),且將混合物冷卻至0℃。向此冷的反應混合物逐滴添加三乙胺(4.0 g, 39.6 mmol),且將混合物在室溫下攪拌4 h。在反應完成後,利用H2 O (2 × 20 mL)處理反應混合物並用二氯甲烷萃取。將有機層分離,經Na2 SO4 乾燥,過濾並蒸發,得到白色固體,將其與二乙醚一起研磨,得到標題化合物(5.2 g, 81%)。 MS: 239.2 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 7.44 (d,J = 8.40 Hz, 1H), 7.36 (d,J = 2.40 Hz, 1H), 7.06 (dd,J = 2.00, 8.40 Hz, 1H), 3.71-3.73 (m, 4H), 3.59-3.61 (m, 4H)。 Preparation Example 16 Step A To a solution of 2,5-dichlorobenzo[d]oxazole (5 g, 26.8 mmol) in anhydrous dichloromethane (50 mL) was added morpholine (3.50 g, 40.3 mmol) and the mixture was cooled to 0℃. To the cold reaction mixture, triethylamine (4.0 g, 39.6 mmol) was added dropwise, and the mixture was stirred at room temperature for 4 h. After the reaction was completed, the reaction mixture was treated with H 2 O (2 × 20 mL) and extracted with dichloromethane. The organic layer was separated, dried over Na2SO4 , filtered and evaporated to give a white solid which was triturated with diethyl ether to give the title compound (5.2 g, 81%). MS: 239.2 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 7.44 (d, J = 8.40 Hz, 1H), 7.36 (d, J = 2.40 Hz, 1H), 7.06 (dd, J = 2.00, 8.40 Hz, 1H), 3.71-3.73 (m, 4H), 3.59-3.61 (m, 4H).

製備實例 17 步驟 A 向2,6-二氯苯并[d]噻唑(5 g, 24.5 mmol)於無水二氯甲烷(50 mL)中之溶液添加於THF中之2 M二甲胺(18.37 mL, 36.65 mol),且將混合物冷卻至0℃。向此冷的反應混合物逐滴添加三乙胺(6.8 mL, 49 mmol),且將混合物在室溫下攪拌4 h。在反應完成後,利用H2 O (2 × 20 mL)處理反應混合物並用二氯甲烷萃取。將有機層分離,經Na2 SO4 乾燥,過濾並蒸發,得到白色固體,將其與二乙醚一起研磨,得到標題化合物(4.8 g, 94%)。 MS: 213.4 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 7.88 (d,J = 2.10 Hz, 1H), 7.41 (d,J = 8.70 Hz, 1H), 7.25-7.26 (m, 1H), 3.14 (s, 6H)。 Preparation Example 17 Step A To a solution of 2,6-dichlorobenzo[d]thiazole (5 g, 24.5 mmol) in dry dichloromethane (50 mL) was added 2 M dimethylamine in THF (18.37 mL, 36.65 mol ), and the mixture was cooled to 0°C. To the cold reaction mixture, triethylamine (6.8 mL, 49 mmol) was added dropwise, and the mixture was stirred at room temperature for 4 h. After the reaction was completed, the reaction mixture was treated with H 2 O (2 × 20 mL) and extracted with dichloromethane. The organic layer was separated, dried over Na2SO4 , filtered and evaporated to give a white solid which was triturated with diethyl ether to give the title compound (4.8 g, 94%). MS: 213.4 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 7.88 (d, J = 2.10 Hz, 1H), 7.41 (d, J = 8.70 Hz, 1H), 7.25-7.26 (m, 1H), 3.14 ( s, 6H).

製備實例 18 步驟 A 向2,5-二氯苯并[d]噻唑(5 g, 24.5 mmol)於無水二氯甲烷(50 mL)中之溶液添加於THF中之2 M二甲胺(18.37 mL, 36.65 mol),且將混合物冷卻至0℃。向此冷的反應混合物逐滴添加三乙胺(6.8 mL, 49 mmol),且將混合物在室溫下攪拌4 h。在反應完成後,利用H2 O (2 × 20 mL)處理反應混合物並用二氯甲烷萃取。將有機層分離,經Na2 SO4 乾燥,過濾並蒸發,得到白色固體,將其與二乙醚一起研磨,得到標題化合物(4.5 g, 88%)。 MS: 213.4 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 7.77 (d,J = 11.20 Hz, 1H), 7.46 (d,J = 2.40 Hz, 1H), 7.05-7.05 (m, 1H), 3.14 (s, 6H)。 Preparation Example 18 Step A To a solution of 2,5-dichlorobenzo[d]thiazole (5 g, 24.5 mmol) in dry dichloromethane (50 mL) was added 2 M dimethylamine in THF (18.37 mL, 36.65 mol ), and the mixture was cooled to 0°C. To the cold reaction mixture, triethylamine (6.8 mL, 49 mmol) was added dropwise, and the mixture was stirred at room temperature for 4 h. After the reaction was complete, the reaction mixture was treated with H 2 O (2 × 20 mL) and extracted with dichloromethane. The organic layer was separated, dried over Na2SO4 , filtered and evaporated to give a white solid which was triturated with diethyl ether to give the title compound (4.5 g, 88 %). MS: 213.4 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 7.77 (d, J = 11.20 Hz, 1H), 7.46 (d, J = 2.40 Hz, 1H), 7.05-7.05 (m, 1H), 3.14 ( s, 6H).

製備實例 19 步驟 A 向6-溴-2-氯苯并[d]噻唑(0.45 g, 1.81 mmol)於乙醇(12 mL)中之溶液添加2 M二甲胺溶液(3 mL),且使用Biotage微波爐將混合物在100℃下加熱60分鐘。將反應混合物冷卻至室溫。將溶劑去除,使用乙酸乙酯及庚烷梯度(40/60 => 60/40),在矽膠管柱上純化粗產物,得到呈固體之標題化合物(0.441 g, 95%)。1 H-NMR (400 MHz,氯仿-d ) δ = 7.70 (d,J = 1.9 Hz, 1H), 7.43 - 7.35 (m, 2H), 3.20 (s, 6H)。 Preparation Example 19 Step A. To a solution of 6-bromo-2-chlorobenzo[d]thiazole (0.45 g, 1.81 mmol) in ethanol (12 mL) was added 2 M dimethylamine solution (3 mL) and the mixture was microwaved using a Biotage microwave. Heat at 100°C for 60 minutes. The reaction mixture was cooled to room temperature. The solvent was removed, and the crude product was purified on a silica column using an ethyl acetate and heptane gradient (40/60 => 60/40) to obtain the title compound as a solid (0.441 g, 95%). 1 H-NMR (400 MHz, chloroform- d ) δ = 7.70 (d, J = 1.9 Hz, 1H), 7.43 - 7.35 (m, 2H), 3.20 (s, 6H).

製備實例 20 步驟 A 向2,6-二氯苯并[d]噁唑(5 g, 26.6 mmol)於無水二氯甲烷(50 mL)中之溶液添加於THF中之2 M二甲胺(26.6 mL, 53.2 mmol),且將混合物冷卻至0℃。向此冷的反應混合物逐滴添加三乙胺(5.6 mL, 39.9 mmol),且將混合物在室溫下攪拌4 h。在反應完成後,利用H2 O (2 × 20 mL)處理反應混合物並用二氯甲烷萃取。將有機層分離,經Na2 SO4 乾燥,過濾並蒸發,得到白色固體,將其與二乙醚一起研磨,得到標題化合物(5 g, 96%)。 MS: 197.2 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 7.56 (s, 1H), 7.23-7.24 (m, 1H), 7.16-7.16 (m, 1H), 3.13 (s, 6H)。 Preparation Example 20 Step A To a solution of 2,6-dichlorobenzo[d]oxazole (5 g, 26.6 mmol) in dry dichloromethane (50 mL) was added 2 M dimethylamine in THF (26.6 mL, 53.2 mmol) and the mixture was cooled to 0 °C. To the cold reaction mixture, triethylamine (5.6 mL, 39.9 mmol) was added dropwise, and the mixture was stirred at room temperature for 4 h. After the reaction was completed, the reaction mixture was treated with H 2 O (2 × 20 mL) and extracted with dichloromethane. The organic layer was separated, dried over Na2SO4 , filtered and evaporated to give a white solid which was triturated with diethyl ether to give the title compound (5 g, 96%). MS: 197.2 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 7.56 (s, 1H), 7.23-7.24 (m, 1H), 7.16-7.16 (m, 1H), 3.13 (s, 6H).

製備實例 21 步驟 A 向2,5-二氯苯并[d]噁唑(5 g, 26.6 mmol)於無水二氯甲烷(50 mL)中之溶液添加於THF中之2 M二甲胺(26.6 mL, 53.2 mmol),且將混合物冷卻至0℃。向此冷的反應混合物逐滴添加三乙胺(5.6 mL, 39.9 mmol),且將混合物在室溫下攪拌4 h。在反應完成後,利用H2 O (2 × 20 mL)處理反應混合物並用二氯甲烷萃取。將有機層分離,經Na2 SO4 乾燥,過濾並蒸發,得到白色固體,將其與二乙醚一起研磨,得到標題化合物(4.9 g, 94%)。 MS: 197.2 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 7.40 (d,J = 8.40 Hz, 1H), 7.30 (d,J = 2.00 Hz, 1H), 6.99-6.99 (m, 1H), 3.13 (s, 6H)。 Preparation Example 21 Step A To a solution of 2,5-dichlorobenzo[d]oxazole (5 g, 26.6 mmol) in dry dichloromethane (50 mL) was added 2 M dimethylamine in THF (26.6 mL, 53.2 mmol) and the mixture was cooled to 0 °C. To the cold reaction mixture, triethylamine (5.6 mL, 39.9 mmol) was added dropwise, and the mixture was stirred at room temperature for 4 h. After the reaction was complete, the reaction mixture was treated with H 2 O (2 × 20 mL) and extracted with dichloromethane. The organic layer was separated, dried over Na2SO4 , filtered and evaporated to give a white solid which was triturated with diethyl ether to give the title compound (4.9 g, 94%). MS: 197.2 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 7.40 (d, J = 8.40 Hz, 1H), 7.30 (d, J = 2.00 Hz, 1H), 6.99-6.99 (m, 1H), 3.13 ( s, 6H).

製備實例 22 步驟 A 向6-溴-2-氯苯并[d]噻唑(0.8 g, 3.2 mmol)於乙醇(12 mL)中之溶液添加異丙胺溶液(1 mL),且使用Biotage微波爐將混合物在100℃下加熱45分鐘。將反應混合物冷卻至室溫。去除溶劑,將粗產物自EtOAc及正庚烷混合物結晶,得到呈固體之標題化合物(0.663 g, 75.8%)。1 H-NMR (400 MHz,氯仿-d ) δ = 7.69 (t,J = 1.3 Hz, 1H), 7.38 (d,J = 1.3 Hz, 2H), 7.27 (s, 1H), 5.51 - 5.26 (m, 1H), 3.92 (h,J = 6.5 Hz, 1H), 1.33 (d,J = 6.4 Hz, 6H)。 Preparation Example 22 Step A. To a solution of 6-bromo-2-chlorobenzo[d]thiazole (0.8 g, 3.2 mmol) in ethanol (12 mL) was added isopropylamine solution (1 mL), and the mixture was heated at 100°C using a Biotage microwave oven. Heat for 45 minutes. The reaction mixture was cooled to room temperature. The solvent was removed and the crude product was crystallized from a mixture of EtOAc and n-heptane to give the title compound as a solid (0.663 g, 75.8%). 1 H-NMR (400 MHz, chloroform- d ) δ = 7.69 (t, J = 1.3 Hz, 1H), 7.38 (d, J = 1.3 Hz, 2H), 7.27 (s, 1H), 5.51 - 5.26 (m , 1H), 3.92 (h, J = 6.5 Hz, 1H), 1.33 (d, J = 6.4 Hz, 6H).

製備實例 23 步驟 A 向6-溴-2-氯苯并[d]噻唑(1 g, 4.0 mmol)於乙醇(6 mL)中之溶液添加異丙胺溶液(1.5 mL),且使用Biotage微波爐將混合物在100℃下加熱90分鐘。將反應混合物冷卻至室溫。去除溶劑,將粗產物溶解於二氯甲烷(150 mL)中,且用1 M NaOH溶液、水及鹽水洗滌並經Na2 SO4 乾燥。將溶劑在減壓下去除以得到粗產物,使用EtOAc及庚烷梯度(20/80 => 50/50),在矽膠管柱上純化該粗產物,得到呈固體之標題化合物(0.35 g, 36%)。1 H-NMR (400 MHz,氯仿-d ) δ = 7.72 (s, 1H), 7.41 (s, 1H), 7.29 (d,J = 3.3 Hz, 1H), 5.40 (s, 1H), 3.13 (s, 3H)。 Preparation Example 23 Step A. To a solution of 6-bromo-2-chlorobenzo[d]thiazole (1 g, 4.0 mmol) in ethanol (6 mL) was added isopropylamine solution (1.5 mL), and the mixture was heated at 100°C using a Biotage microwave oven. Heat for 90 minutes. The reaction mixture was cooled to room temperature. The solvent was removed and the crude product was dissolved in dichloromethane (150 mL) and washed with 1 M NaOH solution, water and brine and dried over Na2SO4 . The solvent was removed under reduced pressure to obtain the crude product, which was purified on a silica column using an EtOAc and heptane gradient (20/80 => 50/50) to obtain the title compound as a solid (0.35 g, 36 %). 1 H-NMR (400 MHz, chloroform- d ) δ = 7.72 (s, 1H), 7.41 (s, 1H), 7.29 (d, J = 3.3 Hz, 1H), 5.40 (s, 1H), 3.13 (s , 3H).

製備實例 24 步驟 A 向5-溴-2-氯苯并[d]噻唑(0.9 g, 3.62 mmol)於乙醇(12 mL)中之溶液添加4 M甲胺溶液(1 mL),且使用Biotage微波爐將混合物在100℃下加熱90分鐘。將反應混合物冷卻至室溫。去除溶劑,使用EtOAc及庚烷梯度(20/80 => 50/50),在矽膠管柱上純化粗產物,得到呈固體之標題化合物(0.57 g, 65%)。1 H-NMR (400 MHz,氯仿-d ) δ = 7.68 (t,J = 2.0 Hz, 1H), 7.46 (d,J = 8.3 Hz, 1H), 7.22 (dd,J = 8.4, 1.9 Hz, 1H), 5.90 (s, 1H), 3.13 (s, 3H)。 Preparation Example 24 Step A To a solution of 5-bromo-2-chlorobenzo[d]thiazole (0.9 g, 3.62 mmol) in ethanol (12 mL) was added 4 M methylamine solution (1 mL) and the mixture was incubated using a Biotage microwave. Heat at 100°C for 90 minutes. The reaction mixture was cooled to room temperature. The solvent was removed and the crude product was purified on a silica column using an EtOAc and heptane gradient (20/80 => 50/50) to obtain the title compound as a solid (0.57 g, 65%). 1 H-NMR (400 MHz, chloroform- d ) δ = 7.68 (t, J = 2.0 Hz, 1H), 7.46 (d, J = 8.3 Hz, 1H), 7.22 (dd, J = 8.4, 1.9 Hz, 1H ), 5.90 (s, 1H), 3.13 (s, 3H).

製備實例 25 步驟 A 向5-溴-2-氯苯并[d]噻唑(0.87 g, 3.5 mmol)於乙醇(12 mL)中之溶液添加異丙胺溶液(1.5 mL),且使用Biotage微波爐將混合物在100℃下加熱60分鐘。將反應混合物冷卻至室溫。去除溶劑,將粗產物溶解於二氯甲烷(150 mL)中,且用1 M NaOH溶液、水及鹽水洗滌並經Na2 SO4 乾燥。將溶劑在減壓下去除以得到粗產物,使用EtOAc及庚烷梯度(20/80 => 50/50),在矽膠管柱上純化該粗產物,得到呈固體之標題化合物(0.75 g, 79%)。1 H-NMR (400 MHz,氯仿-d ) δ = 7.67 (d,J = 1.9 Hz, 1H), 7.43 (d,J = 8.5 Hz, 1H), 7.19 (dd,J = 8.3, 1.9 Hz, 1H), 5.47 (s, 1H), 4.04 - 3.79 (m, 1H), 1.34 (d,J = 6.5 Hz, 6H)。 Preparation Example 25 Step A. To a solution of 5-bromo-2-chlorobenzo[d]thiazole (0.87 g, 3.5 mmol) in ethanol (12 mL) was added isopropylamine solution (1.5 mL), and the mixture was heated at 100°C using a Biotage microwave oven. Heat for 60 minutes. The reaction mixture was cooled to room temperature. The solvent was removed and the crude product was dissolved in dichloromethane (150 mL) and washed with 1 M NaOH solution, water and brine and dried over Na2SO4 . The solvent was removed under reduced pressure to obtain the crude product, which was purified on a silica column using an EtOAc and heptane gradient (20/80 => 50/50) to obtain the title compound as a solid (0.75 g, 79 %). 1 H-NMR (400 MHz, chloroform- d ) δ = 7.67 (d, J = 1.9 Hz, 1H), 7.43 (d, J = 8.5 Hz, 1H), 7.19 (dd, J = 8.3, 1.9 Hz, 1H ), 5.47 (s, 1H), 4.04 - 3.79 (m, 1H), 1.34 (d, J = 6.5 Hz, 6H).

製備實例 26 步驟 A 將5-溴-3-碘吡啶-2-胺(5 g, 16.72 mmol)及異硫氰酸苯甲醯基酯(3.29, 20.07 mmol)於丙酮(10 mL)中之溶液在60℃下攪拌12小時,藉由TLC監測反應。將溶劑蒸發且將固體過濾,用正己烷(200 mL)洗滌並乾燥,得到呈灰白色固體之標題化合物(4 g, 52%)。 MS: 461.5 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 12.35 (s, 1H), 11.86 (s, 1H), 8.64-8.65 (m, 2H), 7.99-7.99 (m, 2H), 7.67 (s, 1H), 7.56 (d,J = 9.40 Hz, 2H)。步驟 B 向來自上文步驟A之標題化合物(4 g, 12.1 mmol)於1,4-二噁烷(60 mL)中之溶液添加碳酸鉀(2.5 g, 18.15 mmol)、L-脯胺酸(0.28 g, 2.43 mmol)及碘化銅(I)(0.462 g, 2.43 mmol)。然後,將反應混合物在80℃下攪拌16小時,藉由TLC監測反應。將反應混合物傾倒至1.0 L水及1.0 L NH4 Cl飽和水溶液中。將懸浮液在室溫下攪拌1小時。將固體過濾,用NH4 Cl之飽和水溶液(2 × 300 mL)及水(2 × 300 mL)洗滌並乾燥,得到呈灰白色固體之標題化合物(2.5 g,粗製)。 MS: 334.51 (M+H)+步驟 C 將來自上文步驟B之標題化合物(2 g, 5.98 mmol)於70% H2 SO4 (6, 3.0 vol)中之懸浮液在120℃下加熱2小時。將反應混合物冷卻至室溫,且將該反應混合物緩慢傾倒至100 mL冷水(0℃)中。然後,藉由添加50% NaOH水溶液將該反應混合物調整至鹼性pH。然後,用EtOAc (6 × 150 mL)萃取化合物。使合併之有機層經Na2 SO4 乾燥並過濾,然後將溶液濃縮,得到呈淺黃色固體之標題化合物(0.3 g, 23%)。 MS: 230.4 (M)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 8.27-8.31 (m, 2H), 8.11 (s, 2H)。步驟 D 在0℃下利用注射器經10 min之時期向來自上文步驟C之標題化合物(0.3 mg, 1.3 mmol)於乙腈(5 mL)中之懸浮液添加亞硝酸第三丁基酯( 0.2 ml, 1.95 mmol)。然後,逐份添加氯化銅(II)(0.2 g, 1.56 mmol)。在0℃下30分鐘後,將反應混合物升溫至室溫持續1小時並加熱至65℃,然後攪拌4小時。藉由TLC監測反應之進展。在反應完成後,將溶劑蒸發且用水(20 mL)及5% MeOH/DCM (3 × 20 mL)稀釋產物。將合併之有機物用鹽水(10 mL)洗滌,經Na2 SO4 乾燥,過濾並在減壓下濃縮。藉由矽膠(60-120)管柱層析,利用1%甲醇/二氯甲烷進行溶析來純化粗製化合物,得到呈灰白色固體之標題化合物(0.15 g, 46%)。 MS: 250.9 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 8.91 (d,J = 2.40 Hz, 1H), 8.82 (d,J = 1.60 Hz, 1H)。步驟 E 向來自上文步驟D之標題化合物(0.18 g, 0.72 mmol)於無水二氯甲烷(5 mL)中之溶液添加三乙胺(0.3 mL, 2.16 mmol)及嗎啉(0.074 g, 0.86 mmol),且將混合物在室溫下攪拌6小時。將反應混合物在真空下濃縮。藉由矽膠(60-120)管柱層析,利用石油醚/乙酸乙酯進行溶析來純化粗製化合物,得到呈灰黃色固體之標題化合物(0.18 g, 83%)。 MS: 300.0 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 8.49 (d,J = 2.00 Hz, 1H), 8.38 (d,J = 1.60 Hz, 1H), 3.72-3.74 (m, 4H), 3.61-3.62 (m, 4H)。 Preparation Example 26 Step A : Prepare a solution of 5-bromo-3-iodopyridin-2-amine (5 g, 16.72 mmol) and benzyl isothiocyanate (3.29, 20.07 mmol) in acetone (10 mL) at 60°C. Stir for 12 hours at low temperature and monitor the reaction by TLC. The solvent was evaporated and the solid was filtered, washed with n-hexane (200 mL) and dried to give the title compound as an off-white solid (4 g, 52%). MS: 461.5 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 12.35 (s, 1H), 11.86 (s, 1H), 8.64-8.65 (m, 2H), 7.99-7.99 (m, 2H), 7.67 (s , 1H), 7.56 (d, J = 9.40 Hz, 2H). Step B To a solution of the title compound from Step A above (4 g, 12.1 mmol) in 1,4-dioxane (60 mL) was added potassium carbonate (2.5 g, 18.15 mmol), L-proline ( 0.28 g, 2.43 mmol) and copper(I) iodide (0.462 g, 2.43 mmol). The reaction mixture was then stirred at 80°C for 16 hours and the reaction was monitored by TLC. The reaction mixture was poured into 1.0 L water and 1.0 L saturated aqueous NH 4 Cl solution. The suspension was stirred at room temperature for 1 hour. The solid was filtered, washed with saturated aqueous NH 4 Cl (2 × 300 mL) and water (2 × 300 mL) and dried to give the title compound (2.5 g, crude) as an off-white solid. MS: 334.51 (M+H) + . Step C A suspension of the title compound from Step B above (2 g, 5.98 mmol) in 70 % H2SO4 (6, 3.0 vol) was heated at 120°C for 2 hours. The reaction mixture was cooled to room temperature and slowly poured into 100 mL of cold water (0°C). The reaction mixture was then adjusted to alkaline pH by adding 50% aqueous NaOH solution. Then, the compound was extracted with EtOAc (6 × 150 mL). The combined organic layers were dried over Na2SO4 and filtered, and the solution was concentrated to give the title compound as a pale yellow solid (0.3 g , 23%). MS: 230.4 (M) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 8.27-8.31 (m, 2H), 8.11 (s, 2H). Step D To a suspension of the title compound from Step C above (0.3 mg, 1.3 mmol) in acetonitrile (5 mL) was added tert-butyl nitrite ( 0.2 ml) using a syringe over a period of 10 min at 0°C. , 1.95 mmol). Then, copper(II) chloride (0.2 g, 1.56 mmol) was added portionwise. After 30 minutes at 0°C, the reaction mixture was warmed to room temperature for 1 hour and heated to 65°C, then stirred for 4 hours. The progress of the reaction was monitored by TLC. After the reaction was complete, the solvent was evaporated and the product was diluted with water (20 mL) and 5% MeOH/DCM (3 × 20 mL). The combined organics were washed with brine (10 mL), dried over Na2SO4 , filtered and concentrated under reduced pressure . The crude compound was purified by silica gel (60-120) column chromatography and elution with 1% methanol/dichloromethane to obtain the title compound (0.15 g, 46%) as an off-white solid. MS: 250.9 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 8.91 (d, J = 2.40 Hz, 1H), 8.82 (d, J = 1.60 Hz, 1H). Step E To a solution of the title compound from Step D above (0.18 g, 0.72 mmol) in dry dichloromethane (5 mL) was added triethylamine (0.3 mL, 2.16 mmol) and morpholine (0.074 g, 0.86 mmol) ), and the mixture was stirred at room temperature for 6 hours. The reaction mixture was concentrated in vacuo. The crude compound was purified by silica gel (60-120) column chromatography and elution with petroleum ether/ethyl acetate to obtain the title compound (0.18 g, 83%) as a grayish-yellow solid. MS: 300.0 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 8.49 (d, J = 2.00 Hz, 1H), 8.38 (d, J = 1.60 Hz, 1H), 3.72-3.74 (m, 4H), 3.61- 3.62 (m, 4H).

製備實例 27 步驟 A 將2-溴-6-氯吡啶-3-胺(5 g, 24.1 mmol)及硫氰酸鉀(7 g, 72.3 mmol)於乙醇(50 mL)中之溶液及濃鹽酸(37%, 100 mL)在100℃下攪拌40-45 h。藉由TLC確認反應完成(PE/EA = 7.5/2.5)。將反應混合物冷卻至室溫並濃縮以提供棕色固體,將其在二氯甲烷(150 mL)及1 N NaOH水溶液(50 mL)中分配。將固體過濾並乾燥,得到呈淺黃色固體之標題化合物(3.5 g,79%產率)。 MS: 186.1 (M+H)+步驟 B 在0℃下利用注射器經10 min之時期向來自上文步驟A之標題化合物(1.5 g, 8.08 mmol)於乙腈(25 mL)中之懸浮液添加亞硝酸第三丁基酯( 1.4 ml, 12.12 mmol)。然後,逐份添加溴化銅(II)(2.16 g, 9.69 mmol)。在0℃下30分鐘後,使反應混合物升溫至室溫持續2分鐘。藉由TLC監測反應之進展。在反應完成後,將溶劑蒸發,且用水(20 mL)及5% MeOH/DCM (3 × 20 mL)稀釋混合物。將合併之有機物用鹽水(10 mL)洗滌,經Na2 SO4 乾燥,過濾並在減壓下濃縮。藉由矽膠(60-120)管柱層析且利用1%甲醇/二氯甲烷進行溶析來純化粗製化合物,得到呈淡黃色固體之標題化合物(0.65 g, 32%)。 MS: 248.5 (M+H)+步驟 C 向來自上文步驟B之標題化合物(0.65 g, 2.61 mmol)於無水二氯甲烷(5 mL)中之溶液添加三乙胺(1.1 mL, 7.83 mmol)及嗎啉(0.34 g, 3.91 mmol),且將混合物在室溫下攪拌6小時。將反應混合物在真空下濃縮。藉由矽膠(60-120)管柱層析,利用石油醚/乙酸乙酯進行溶析來純化粗製化合物,得到呈灰黃色固體之標題化合物(0.6 g, 90%)。 MS: 256.0 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 7.83 (d,J = 8.40 Hz, 1H), 7.41 (d,J = 8.44 Hz, 1H), 3.72-3.74 (m, 2H), 3.59-3.60 (m, 2H)。 Preparation Example 27 Step A : A solution of 2-bromo-6-chloropyridin-3-amine (5 g, 24.1 mmol) and potassium thiocyanate (7 g, 72.3 mmol) in ethanol (50 mL) and concentrated hydrochloric acid (37%, 100 mL) and stir at 100°C for 40-45 h. The reaction was confirmed to be complete by TLC (PE/EA = 7.5/2.5). The reaction mixture was cooled to room temperature and concentrated to provide a brown solid, which was partitioned between dichloromethane (150 mL) and 1 N aqueous NaOH (50 mL). The solid was filtered and dried to give the title compound as a pale yellow solid (3.5 g, 79% yield). MS: 186.1 (M+H) + . Step B To a suspension of the title compound from Step A above (1.5 g, 8.08 mmol) in acetonitrile (25 mL) was added tert-butyl nitrite ( 1.4 ml) using a syringe over a period of 10 min at 0°C. , 12.12 mmol). Then, copper (II) bromide (2.16 g, 9.69 mmol) was added portionwise. After 30 minutes at 0°C, the reaction mixture was allowed to warm to room temperature for 2 minutes. The progress of the reaction was monitored by TLC. After the reaction was complete, the solvent was evaporated and the mixture was diluted with water (20 mL) and 5% MeOH/DCM (3 × 20 mL). The combined organics were washed with brine (10 mL), dried over Na2SO4 , filtered and concentrated under reduced pressure . The crude compound was purified by silica gel (60-120) column chromatography and elution with 1% methanol/dichloromethane to obtain the title compound (0.65 g, 32%) as a light yellow solid. MS: 248.5 (M+H) + . Step C To a solution of the title compound from Step B above (0.65 g, 2.61 mmol) in dry dichloromethane (5 mL) was added triethylamine (1.1 mL, 7.83 mmol) and morpholine (0.34 g, 3.91 mmol) ), and the mixture was stirred at room temperature for 6 hours. The reaction mixture was concentrated in vacuo. The crude compound was purified by silica gel (60-120) column chromatography and elution with petroleum ether/ethyl acetate to obtain the title compound (0.6 g, 90%) as a grayish-yellow solid. MS: 256.0 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 7.83 (d, J = 8.40 Hz, 1H), 7.41 (d, J = 8.44 Hz, 1H), 3.72-3.74 (m, 2H), 3.59- 3.60 (m, 2H).

製備實例 28 步驟 A 使2,7-二溴-[1,2,4]三唑并[1,5-a]吡啶(0.4 g, 1.44 mmol)及嗎啉(4 mL)之混合物在N2 氣氛下回流4小時。將反應混合物濃縮至乾燥。藉由矽膠(60-120)管柱層析,利用石油醚/乙酸乙酯10%-100%作為溶析液進行溶析來純化粗製化合物,得到呈白色固體之標題化合物(0.27 g, 66%)。 MS: 285.0 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 8.64 (d,J = 7.08 Hz, 1H), 7.83 (s, 1H), 7.13-7.14 (m, 1H), 3.69-3.71 (m, 2H), 3.45-3.46 (m, 2H)。 Preparation Example 28 Step A : Reflux a mixture of 2,7-dibromo-[1,2,4]triazolo[1,5-a]pyridine (0.4 g, 1.44 mmol) and morpholine (4 mL) under N2 atmosphere 4 hours. The reaction mixture was concentrated to dryness. The crude compound was purified by silica gel (60-120) column chromatography, using petroleum ether/ethyl acetate 10%-100% as the eluent, to obtain the title compound as a white solid (0.27 g, 66% ). MS: 285.0 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 8.64 (d, J = 7.08 Hz, 1H), 7.83 (s, 1H), 7.13-7.14 (m, 1H), 3.69-3.71 (m, 2H ), 3.45-3.46 (m, 2H).

製備實例 29 將3,6-二溴嗒嗪(0.2 g, 0.841 mmol)溶解於乙腈(2.5 mL)中。然後添加嗎啉(0.110 mL, 1.261 mmol)及三乙胺(0.176 mL, 1.261 mmol),且將懸浮液於微波中在160℃下輻照1 h及20分鐘。用二氯甲烷及水稀釋反應混合物。分離有機層,且將水層用二氯甲烷萃取兩次。使合併之有機層經Na2 SO4 乾燥,過濾並在減壓下濃縮。使用Biotage Isolera One純化系統,採用正庚烷/乙酸乙酯梯度(100/0 -> 0/100),在矽膠管柱上純化粗產物,得到呈白色固體之標題化合物(0.168 g, 82%)。 MS: 245.2 (M+H)+1 H-NMR (400 MHz,氯仿-d ) δ = 7.35 (d,J = 9.5 Hz, 1H), 6.79 (d,J = 9.5 Hz, 1H), 3.88 - 3.79 (m, 4H), 3.66 - 3.55 (m, 4H)。 Preparation Example 29 Dissolve 3,6-dibromopyridazine (0.2 g, 0.841 mmol) in acetonitrile (2.5 mL). Morpholine (0.110 mL, 1.261 mmol) and triethylamine (0.176 mL, 1.261 mmol) were then added, and the suspension was irradiated in the microwave at 160 °C for 1 h and 20 min. Dilute the reaction mixture with dichloromethane and water. The organic layer was separated, and the aqueous layer was extracted twice with dichloromethane. The combined organic layers were dried over Na2SO4 , filtered and concentrated under reduced pressure. The crude product was purified on a silica column using a Biotage Isolera One purification system using n-heptane/ethyl acetate gradient (100/0 -> 0/100) to obtain the title compound (0.168 g, 82%) as a white solid. . MS: 245.2 (M+H) + . 1 H-NMR (400 MHz, chloroform- d ) δ = 7.35 (d, J = 9.5 Hz, 1H), 6.79 (d, J = 9.5 Hz, 1H), 3.88 - 3.79 (m, 4H), 3.66 - 3.55 (m, 4H).

製備實例 30 在氮下在0℃下向氫化鈉(0.0404 g, 1.682 mmol)於無水THF (3 mL)中之懸浮液添加嗎啉(0.146 mL, 1.682 mmol)。將反應混合物在0℃下攪拌15分鐘,然後添加溶解於無水THF (3 mL)中之2,5-二溴吡嗪(0.400 g, 1.682 mmol)。將反應混合物在回流條件下攪拌過夜。用水使其淬滅且將產物用EtOAc萃取三次。使合併之有機層經Na2 SO4 乾燥,過濾並在減壓下濃縮。使用Biotage Isolera One純化系統,採用正庚烷/乙酸乙酯梯度(100/0 -> 0/100),在矽膠管柱上純化粗產物,得到呈白色固體之標題化合物(0.245 g, 60%)。1 H-NMR (400 MHz,氯仿-d ) δ = 8.15 (d,J = 1.5 Hz, 1H), 7.86 (d,J = 1.5 Hz, 1H), 3.89 - 3.76 (m, 4H), 3.60 - 3.45 (m, 4H)。 Preparation Example 30 To a suspension of sodium hydride (0.0404 g, 1.682 mmol) in dry THF (3 mL) was added morpholine (0.146 mL, 1.682 mmol) under nitrogen at 0°C. The reaction mixture was stirred at 0 °C for 15 min, then 2,5-dibromopyrazine (0.400 g, 1.682 mmol) dissolved in anhydrous THF (3 mL) was added. The reaction mixture was stirred under reflux overnight. It was quenched with water and the product was extracted three times with EtOAc. The combined organic layers were dried over Na2SO4 , filtered and concentrated under reduced pressure. The crude product was purified on a silica column using a Biotage Isolera One purification system using n-heptane/ethyl acetate gradient (100/0 -> 0/100) to obtain the title compound (0.245 g, 60%) as a white solid. . 1 H-NMR (400 MHz, chloroform- d ) δ = 8.15 (d, J = 1.5 Hz, 1H), 7.86 (d, J = 1.5 Hz, 1H), 3.89 - 3.76 (m, 4H), 3.60 - 3.45 (m, 4H).

製備實例 31 步驟 A 在120℃下向2-氯-5-硝基-4-氰硫基嘧啶(10 g, 46.2 mmol)於乙酸(264 mL, 4617 mmol)中之熱溶液添加鐵(15.47 g, 277 mmol)。將混合物在相同溫度下攪拌1小時。使該混合物冷卻至室溫且藉由過濾去除不溶性材料並用乙酸洗滌。將濾液在減壓下濃縮並溶解於THF (200 mL)及EtOAc (400 mL)中,且用飽和NH4 Cl水溶液(100 mL)、飽和NaHCO3 溶液(100 mL)洗滌,經Na2 SO4 乾燥且在減壓下去除溶劑,得到標題化合物(6.82 g, 79%)。1 H-NMR (400 MHz, DMSO-d 6 ) δ = 9.42 (s, 1H)。步驟 B 在0℃下經30分鐘向來自上文步驟A之標題化合物(8.2 g, 43 mmol)於乙腈(20 mL)中之懸浮液添加亞硝酸第三丁基酯(6.8 g, 65.9 mmol)。然後,分多次添加溴化銅(II)(11.78 g, 52.8 mmol)。在0℃下30分鐘後,使反應混合物升溫至室溫並攪拌12小時。添加水(50 mL)及EtOAc (100 mL)且分離各相。將水層用EtOAc萃取兩次且將有機層合併,經Na2 SO4 乾燥且將溶劑在減壓下蒸發。使用Biotage Isolera One純化系統,採用EtOAc/正庚烷(20/80)在HP-Sil SNAP柱上純化殘餘物,得到標題化合物(4.94 g, 45%)。1 H-NMR (400 MHz, DMSO-d 6 ) δ = 8.54 (s, 1H), 8.30 (s, 2H)。步驟 C 將來自上文步驟B之標題化合物(0.157 g, 0.627 mmol)溶解於嗎啉(3 mL, 0.627 mmol)中,且在室溫下攪拌2 h。用二氯甲烷及水稀釋反應混合物。分離有機層,且將水層用二氯甲烷萃取兩次。使合併之有機層經Na2 SO4 乾燥,過濾並在減壓下濃縮。使用Biotage Isolera One純化系統,採用正庚烷/乙酸乙酯梯度(100/0 -> 0/100),在矽膠管柱上純化粗產物,得到呈白色固體之標題化合物(0.048 g, 30%)。 MS: 258.6 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 8.71 (s, 1H), 3.74 (dd,J = 6.0, 3.6 Hz, 4H), 3.70 - 3.58 (m, 4H)。 Preparation Example 31 Step A To a hot solution of 2-chloro-5-nitro-4-cyanothiopyrimidine (10 g, 46.2 mmol) in acetic acid (264 mL, 4617 mmol) at 120 °C was added iron (15.47 g, 277 mmol). The mixture was stirred at the same temperature for 1 hour. The mixture was allowed to cool to room temperature and insoluble material was removed by filtration and washed with acetic acid. The filtrate was concentrated under reduced pressure and dissolved in THF (200 mL) and EtOAc (400 mL), and washed with saturated aqueous NH 4 Cl solution (100 mL), saturated NaHCO 3 solution (100 mL), and washed with Na 2 SO 4 Drying and removal of solvent under reduced pressure gave the title compound (6.82 g, 79%). 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 9.42 (s, 1H). Step B To a suspension of the title compound from step A above (8.2 g, 43 mmol) in acetonitrile (20 mL) was added tert-butyl nitrite (6.8 g, 65.9 mmol) at 0°C over 30 min. Then, copper (II) bromide (11.78 g, 52.8 mmol) was added in several portions. After 30 minutes at 0°C, the reaction mixture was allowed to warm to room temperature and stirred for 12 hours. Water (50 mL) and EtOAc (100 mL) were added and the phases separated. The aqueous layer was extracted twice with EtOAc and the organic layers were combined , dried over Na2SO4 and the solvent was evaporated under reduced pressure. The residue was purified on a HP-Sil SNAP column using EtOAc/n-heptane (20/80) using a Biotage Isolera One purification system to give the title compound (4.94 g, 45%). 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 8.54 (s, 1H), 8.30 (s, 2H). Step C The title compound from step B above (0.157 g, 0.627 mmol) was dissolved in morpholine (3 mL, 0.627 mmol) and stirred at room temperature for 2 h. Dilute the reaction mixture with dichloromethane and water. The organic layer was separated, and the aqueous layer was extracted twice with dichloromethane. The combined organic layers were dried over Na2SO4 , filtered and concentrated under reduced pressure. The crude product was purified on a silica column using a Biotage Isolera One purification system using n-heptane/ethyl acetate gradient (100/0 -> 0/100) to obtain the title compound (0.048 g, 30%) as a white solid. . MS: 258.6 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 8.71 (s, 1H), 3.74 (dd, J = 6.0, 3.6 Hz, 4H), 3.70 - 3.58 (m, 4H).

製備實例 32 在氮下在0℃下向氫化鈉(0.077 g, 3.21 mmol)於無水THF (5 mL)中之懸浮液添加(1R ,5S )-3-氧雜-8-氮雜二環[3.2.1]辛烷鹽酸鹽(0.457 g, 3.06 mmol)。將反應混合物攪拌15分鐘,然後添加溶解於無水THF (5 mL)中之2,5-二溴吡嗪(0.800 g, 3.36 mmol)。將反應混合物在回流條件下攪拌過夜。用水使反應混合物淬滅且將產物用乙酸乙酯萃取三次。使合併之有機層經Na2 SO4 乾燥,過濾並在減壓下濃縮。使用Biotage Isolera One純化系統,採用正庚烷/乙酸乙酯梯度(100/0 -> 0/100),在矽膠管柱上純化粗產物,得到呈白色固體之標題化合物(0.038 mg, 5%)。 MS: 271.8 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 8.24 (d,J = 1.4 Hz, 1H), 8.11 (d,J = 1.4 Hz, 1H), 4.49 (s, 2H), 3.62 (d,J = 10.9 Hz, 2H), 3.52 (d,J = 11.0 Hz, 2H), 2.02 - 1.95 (m, 2H), 1.91 (m, 2H)。 Preparation Example 32 To a suspension of sodium hydride (0.077 g, 3.21 mmol) in dry THF (5 mL) was added (1 R ,5 S )-3-oxa-8-azabicyclo [3.2 .1] Octane hydrochloride (0.457 g, 3.06 mmol). The reaction mixture was stirred for 15 minutes, then 2,5-dibromopyrazine (0.800 g, 3.36 mmol) dissolved in anhydrous THF (5 mL) was added. The reaction mixture was stirred under reflux overnight. The reaction mixture was quenched with water and the product was extracted three times with ethyl acetate. The combined organic layers were dried over Na2SO4 , filtered and concentrated under reduced pressure. The crude product was purified on a silica column using a Biotage Isolera One purification system using n-heptane/ethyl acetate gradient (100/0 -> 0/100) to obtain the title compound (0.038 mg, 5%) as a white solid. . MS: 271.8 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 8.24 (d, J = 1.4 Hz, 1H), 8.11 (d, J = 1.4 Hz, 1H), 4.49 (s, 2H), 3.62 (d, J = 10.9 Hz, 2H), 3.52 (d, J = 11.0 Hz, 2H), 2.02 - 1.95 (m, 2H), 1.91 (m, 2H).

製備實例 33 步驟 A 將乙酸鈀(II)(0.00651 g, 0.029 mmol)及2-二環己基膦基-2′,4′,6′-三異丙基聯苯(XPhos) (0.0415 g, 0.087 mmol)置於反應小瓶中,且添加經脫氣之1,4-二噁烷(2 mL)。將所得溶液短暫脫氣。將懸浮液在100℃下(在預加熱之加熱塊上)加熱少於1分鐘,直至溶液之色彩自橙色變為深粉色為止。然後,將小瓶自加熱塊移除,且添加來自製備實例1之標題化合物(0.110 g, 0.290 mmol)及來自製備實例29之標題化合物(0.078 g, 0.319 mmol)及碳酸銫(0.331 g, 1.015 mmol)。將反應小瓶填充氬,之後將其關閉。將反應混合物在100℃下加熱12 h。用乙酸乙酯及水稀釋該反應混合物。分離有機層,且將水層用乙酸乙酯再萃取兩次。使合併之有機層經Na2 SO4 乾燥,過濾並將溶劑在減壓下蒸發。使用Biotage Isolera One純化系統,採用正庚烷/乙酸乙酯梯度(90/10 -> 0/100),在矽膠管柱上純化粗產物,得到呈黃色固體之標題化合物(0.072 g, 49%)。 MS: 508.2 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 8.03 (dd,J = 9.1, 4.5 Hz, 1H), 7.70 (d,J = 8.4 Hz, 2H), 7.42 (d,J = 9.9 Hz, 1H), 7.38 (dd,J = 8.9, 2.6 Hz, 1H), 7.32 (dd,J = 9.0, 6.1 Hz, 2H), 7.17 (td,J = 9.2, 2.6 Hz, 1H), 6.93 (s, 1H), 4.55 (s, 2H), 3.88 (t,J = 5.6 Hz, 2H), 3.78 - 3.65 (m, 4H), 3.41 - 3.34 (m, 4H), 3.21 - 3.14 (m, 2H), 2.30 (s, 3H)。 Preparation Example 33 Step A: Place palladium(II) acetate (0.00651 g, 0.029 mmol) and 2-dicyclohexylphosphino-2′,4′,6′-triisopropylbiphenyl (XPhos) (0.0415 g, 0.087 mmol). To the reaction vial, add degassed 1,4-dioxane (2 mL). The resulting solution was briefly degassed. Heat the suspension at 100°C (on a preheated heating block) for less than 1 minute until the color of the solution changes from orange to dark pink. The vial was then removed from the heating block and the title compound from Preparative Example 1 (0.110 g, 0.290 mmol) and the title compound from Preparative Example 29 (0.078 g, 0.319 mmol) and cesium carbonate (0.331 g, 1.015 mmol) were added ). The reaction vial was filled with argon before being closed. The reaction mixture was heated at 100 °C for 12 h. The reaction mixture was diluted with ethyl acetate and water. The organic layer was separated, and the aqueous layer was extracted twice more with ethyl acetate. The combined organic layers were dried over Na2SO4 , filtered and the solvent evaporated under reduced pressure . The crude product was purified on a silica column using a Biotage Isolera One purification system using n-heptane/ethyl acetate gradient (90/10 -> 0/100) to obtain the title compound (0.072 g, 49%) as a yellow solid. . MS: 508.2 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 8.03 (dd, J = 9.1, 4.5 Hz, 1H), 7.70 (d, J = 8.4 Hz, 2H), 7.42 (d, J = 9.9 Hz, 1H), 7.38 (dd, J = 8.9, 2.6 Hz, 1H), 7.32 (dd, J = 9.0, 6.1 Hz, 2H), 7.17 (td, J = 9.2, 2.6 Hz, 1H), 6.93 (s, 1H ), 4.55 (s, 2H), 3.88 (t, J = 5.6 Hz, 2H), 3.78 - 3.65 (m, 4H), 3.41 - 3.34 (m, 4H), 3.21 - 3.14 (m, 2H), 2.30 ( s, 3H).

製備實例 34 41f 除使用下表中所指示之三環胺基衍生物及溴/氯衍生物以外,遵循如製備實例33中所闡述之鈀偶合程序,製備以下化合物: 1 Preparation Examples 34 to 41f Following the palladium coupling procedure as set forth in Preparative Example 33, except using the tricyclic amine derivatives and the bromo/chlorine derivatives indicated in the table below, the following compounds were prepared: Table 1

製備實例 42 步驟 A 將乙酸鈀(II)(0.0045 g, 0.020 mmol)及4,5-雙(二苯基膦基)-9,9-二甲基𠮿(XANTPHOS) (0.035 g, 0.061 mmol)置於反應小瓶中,且添加經脫氣之1,4-二噁烷(4 mL)。將所得溶液短暫脫氣。將懸浮液在100℃下(在預加熱之加熱塊上)加熱少於1分鐘,直至溶液之色彩自橙色變為深粉色為止。然後,將小瓶自加熱塊移除,且添加來自製備實例1步驟E之標題化合物(70 mg, 0.203 mmol)及來自製備實例26之標題化合物(0.067 g, 0.224 mmol)及碳酸銫(0.232 g, 0.771 mmol)。將反應小瓶填充氬,之後將其關閉。將反應混合物在100℃下加熱18 h。用乙酸乙酯及水稀釋該反應混合物。分離有機層,且將水層用乙酸乙酯再萃取兩次。使合併之有機層經Na2 SO4 乾燥,過濾並將溶劑在減壓下蒸發。在矽膠管柱上使用Biotage Isolera One純化系統,採用二氯甲烷/甲醇(100/00 -> 95/05)純化粗產物,得到呈黃色固體之標題化合物(0.052 g, 45%)。1 H NMR (400 MHz,氯仿-d ) δ = 8.24 (d,J = 2.7 Hz, 1H), 8.16 - 8.03 (m, 1H), 7.64 - 7.59 (m, 2H), 7.55 (d,J = 2.7 Hz, 1H), 7.18 (d,J = 8.2 Hz, 2H), 7.02 (dd,J = 8.6, 1.9 Hz, 2H), 4.23 (d,J = 2.0 Hz, 2H), 3.84 - 3.78 (m, 4H), 3.69 - 3.63 (m, 4H), 3.59 (t,J = 5.6 Hz, 2H), 3.48 (s, 1H), 3.25 (dq,J = 5.7, 3.5, 2.8 Hz, 2H), 2.33 (s, 3H)。 Preparation Example 42 Step A Palladium(II) acetate (0.0045 g, 0.020 mmol) and 4,5-bis(diphenylphosphino)-9,9-dimethyl 𠮿 (XANTPHOS) (0.035 g, 0.061 mmol) was placed in the reaction vial and degassed 1,4-dioxane (4 mL) was added. The resulting solution was briefly degassed. Heat the suspension at 100°C (on a preheated heating block) for less than 1 minute until the color of the solution changes from orange to dark pink. The vial was then removed from the heating block and the title compound from Preparative Example 1, Step E (70 mg, 0.203 mmol) and the title compound from Preparative Example 26 (0.067 g, 0.224 mmol) and cesium carbonate (0.232 g, 0.771 mmol). The reaction vial was filled with argon before being closed. The reaction mixture was heated at 100 °C for 18 h. The reaction mixture was diluted with ethyl acetate and water. The organic layer was separated, and the aqueous layer was extracted twice more with ethyl acetate. The combined organic layers were dried over Na2SO4 , filtered and the solvent evaporated under reduced pressure . The crude product was purified on a silica column using a Biotage Isolera One purification system and methylene chloride/methanol (100/00 -> 95/05) to obtain the title compound (0.052 g, 45%) as a yellow solid. 1 H NMR (400 MHz, chloroform- d ) δ = 8.24 (d, J = 2.7 Hz, 1H), 8.16 - 8.03 (m, 1H), 7.64 - 7.59 (m, 2H), 7.55 (d, J = 2.7 Hz, 1H), 7.18 (d, J = 8.2 Hz, 2H), 7.02 (dd, J = 8.6, 1.9 Hz, 2H), 4.23 (d, J = 2.0 Hz, 2H), 3.84 - 3.78 (m, 4H ), 3.69 - 3.63 (m, 4H), 3.59 (t, J = 5.6 Hz, 2H), 3.48 (s, 1H), 3.25 (dq, J = 5.7, 3.5, 2.8 Hz, 2H), 2.33 (s, 3H).

製備實例 43 步驟 A 將乙酸鈀(II)(0.0045 g, 0.020 mmol)及4,5-雙(二苯基膦基)-9,9-二甲基𠮿(XANTPHOS) (0.035 g, 0.061 mmol)置於反應小瓶中,且添加經脫氣之1,4-二噁烷(4 mL)。將所得溶液短暫脫氣。將懸浮液在100℃下(在預加熱之加熱塊上)加熱少於1分鐘,直至溶液之色彩自橙色變為深粉色為止。然後,將小瓶自加熱塊移除,且添加來自製備實例1步驟E之標題化合物(70 mg, 0.203 mmol)及來自製備實例27之標題化合物 (0.067 g, 0.224 mmol)及碳酸銫(0.232 g, 0.771 mmol)。將反應小瓶填充氬,之後將其關閉。將反應混合物在100℃下加熱18 h。用乙酸乙酯及水稀釋該反應混合物。分離有機層,且將水層用乙酸乙酯再萃取兩次。使合併之有機層經Na2 SO4 乾燥,過濾並將溶劑在減壓下蒸發。使用Biotage Isolera One純化系統,採用二氯甲烷/甲醇(100/00 -> 95/05),在矽膠管柱上純化粗產物,得到呈黃色固體之標題化合物(0.048 g, 42%)。1 H NMR (400 MHz,氯仿-d ) δ = 8.10 (dd,J = 9.0, 4.4 Hz, 1H), 7.65 (d,J = 8.9 Hz, 1H), 7.63 - 7.57 (m, 2H), 7.15 (d,J = 8.2 Hz, 2H), 7.09 - 6.97 (m, 2H), 6.75 (d,J = 8.9 Hz, 1H), 4.51 (d,J = 2.0 Hz, 2H), 3.94 (t,J = 5.6 Hz, 2H), 3.82 (q,J = 5.2 Hz, 4H), 3.57 (dd,J = 5.8, 4.0 Hz, 4H), 3.25 (td,J = 5.5, 2.7 Hz, 2H), 2.31 (s, 3H)。 Preparation Example 43 Step A Palladium(II) acetate (0.0045 g, 0.020 mmol) and 4,5-bis(diphenylphosphino)-9,9-dimethyl 𠮿 (XANTPHOS) (0.035 g, 0.061 mmol) was placed in the reaction vial and degassed 1,4-dioxane (4 mL) was added. The resulting solution was briefly degassed. Heat the suspension at 100°C (on a preheated heating block) for less than 1 minute until the color of the solution changes from orange to dark pink. The vial was then removed from the heating block and the title compound from Preparative Example 1, Step E (70 mg, 0.203 mmol) and the title compound from Preparative Example 27 (0.067 g, 0.224 mmol) and cesium carbonate (0.232 g, 0.771 mmol). The reaction vial was filled with argon before being closed. The reaction mixture was heated at 100 °C for 18 h. The reaction mixture was diluted with ethyl acetate and water. The organic layer was separated, and the aqueous layer was extracted twice more with ethyl acetate. The combined organic layers were dried over Na2SO4 , filtered and the solvent evaporated under reduced pressure . The crude product was purified on a silica column using a Biotage Isolera One purification system using methylene chloride/methanol (100/00 -> 95/05) to obtain the title compound (0.048 g, 42%) as a yellow solid. 1 H NMR (400 MHz, chloroform- d ) δ = 8.10 (dd, J = 9.0, 4.4 Hz, 1H), 7.65 (d, J = 8.9 Hz, 1H), 7.63 - 7.57 (m, 2H), 7.15 ( d, J = 8.2 Hz, 2H), 7.09 - 6.97 (m, 2H), 6.75 (d, J = 8.9 Hz, 1H), 4.51 (d, J = 2.0 Hz, 2H), 3.94 (t, J = 5.6 Hz, 2H), 3.82 (q, J = 5.2 Hz, 4H), 3.57 (dd, J = 5.8, 4.0 Hz, 4H), 3.25 (td, J = 5.5, 2.7 Hz, 2H), 2.31 (s, 3H ).

製備實例 44 步驟 A 將3,6-二溴嗒嗪(0.2 g, 0.841 mmol)溶解於乙腈(2.5 mL)中。然後添加8-氧雜-3-氮雜二環[3.2.1]辛烷(0.105 g, 0.925 mmol)及三乙胺(0.176 mL, 1.261 mmol),且將懸浮液於微波中輻照至160℃持續1 h及20分鐘。用二氯甲烷及水稀釋反應混合物。分離有機層,且將水層用二氯甲烷萃取兩次。使合併之有機層經Na2 SO4 乾燥,過濾並在減壓下濃縮。使用Biotage Isolera One純化系統,採用正庚烷/乙酸乙酯梯度(100/0 -> 0/100),在矽膠管柱上純化粗產物,得到呈米色固體之標題化合物(0.152 g, 67%)。 MS: 271.5 (M+H)+1 H-NMR (400 MHz,氯仿-d ) δ = 7.32 (d,J = 9.5 Hz, 1H), 6.70 (d,J = 9.5 Hz, 1H), 4.60 - 4.43 (m, 2H), 3.83 (d,J = 12.8 Hz, 2H), 3.23 (dd,J = 12.4, 2.7 Hz, 2H), 2.08 - 1.94 (m, 2H), 1.90 - 1.76 (m, 2H)。 Preparation Example 44 Step A Dissolve 3,6-dibromopyridazine (0.2 g, 0.841 mmol) in acetonitrile (2.5 mL). Then 8-oxa-3-azabicyclo[3.2.1]octane (0.105 g, 0.925 mmol) and triethylamine (0.176 mL, 1.261 mmol) were added, and the suspension was irradiated in the microwave to 160 ℃ for 1 h and 20 minutes. Dilute the reaction mixture with dichloromethane and water. The organic layer was separated, and the aqueous layer was extracted twice with dichloromethane. The combined organic layers were dried over Na2SO4 , filtered and concentrated under reduced pressure. The crude product was purified on a silica column using a Biotage Isolera One purification system using n-heptane/ethyl acetate gradient (100/0 -> 0/100) to obtain the title compound (0.152 g, 67%) as a beige solid. . MS: 271.5 (M+H) + . 1 H-NMR (400 MHz, chloroform- d ) δ = 7.32 (d, J = 9.5 Hz, 1H), 6.70 (d, J = 9.5 Hz, 1H), 4.60 - 4.43 (m, 2H), 3.83 (d , J = 12.8 Hz, 2H), 3.23 (dd, J = 12.4, 2.7 Hz, 2H), 2.08 - 1.94 (m, 2H), 1.90 - 1.76 (m, 2H).

製備實例 45 步驟 A 向2,6-二氯苯并[d ]噁唑之溶液(1.2 g, 6.3 mmol)添加於無水乙醇(8 mL)中之33 wt.%甲胺溶液,且使用Biotage微波爐將混合物在100℃下加熱30分鐘。使反應混合物冷卻至室溫,且溶解於二氯甲烷(150 mL)中。將有機相用水、1 N NaOH溶液及鹽水洗滌,且然後經Na2 SO4 乾燥。將溶劑去除,且獲得標題化合物(1.08 g, 94%)。1 H NMR (400 MHz,氯仿-d ) δ 7.30 - 7.24 (m, 2H), 7.16 (dd,J = 8.4, 1.9 Hz, 1H), 5.35 (s, 1H), 3.14 (s, 3H)。步驟 B 在0℃下向來自上文步驟A之標題化合物(39 mg, 1.643 mmol)於無水THF (1.5 mL)中之攪拌懸浮液緩慢添加6-氯-N-甲基苯并[d]噁唑-2-胺(100 mg, 0.548 mmol)於無水THF (2 mL)中之溶液,且在相同溫度下攪拌30 min。然後在0℃下逐滴添加4-甲苯-1-磺醯氯(107 mg, 0.561 mmol)於無水THF (1.5 mL)中之溶液,且將反應混合物在0℃下攪拌2 h。利用冰水(4 mL)將反應混合物在0℃下淬滅。然後將產物用乙酸乙酯萃取三次。將合併之有機萃取物用水、鹽水洗滌,經Na2 SO4 乾燥,過濾並在減壓下蒸發,得到呈米色固體之標題化合物(154 mg, 83%)。 MS: 337.14 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 7.93 - 7.89 (m, 2H), 7.88 (d,J = 2.0 Hz, 1H), 7.58 (d,J = 8.5 Hz, 1H), 7.49 - 7.44 (m, 2H), 7.36 (dd,J = 8.5, 2.0 Hz, 1H), 3.49 (s, 3H), 2.39 (s, 3H)。 Preparation Example 45 Step A To a solution of 2,6-dichlorobenzo[ d ]oxazole (1.2 g, 6.3 mmol) was added a 33 wt.% methylamine solution in absolute ethanol (8 mL), and the mixture was heated in a Biotage microwave oven. Heat at 100°C for 30 minutes. The reaction mixture was cooled to room temperature and dissolved in dichloromethane (150 mL). The organic phase was washed with water, 1 N NaOH solution and brine, and then dried over Na2SO4 . The solvent was removed and the title compound was obtained (1.08 g, 94%). 1 H NMR (400 MHz, chloroform- d ) δ 7.30 - 7.24 (m, 2H), 7.16 (dd, J = 8.4, 1.9 Hz, 1H), 5.35 (s, 1H), 3.14 (s, 3H). Step B To a stirred suspension of the title compound from Step A above (39 mg, 1.643 mmol) in anhydrous THF (1.5 mL) was slowly added 6-chloro-N-methylbenzo[d]oxazole at 0°C. A solution of azole-2-amine (100 mg, 0.548 mmol) in anhydrous THF (2 mL) was stirred at the same temperature for 30 min. Then a solution of 4-toluene-1-sulfonyl chloride (107 mg, 0.561 mmol) in anhydrous THF (1.5 mL) was added dropwise at 0 °C, and the reaction mixture was stirred at 0 °C for 2 h. The reaction mixture was quenched with ice water (4 mL) at 0 °C. The product was then extracted three times with ethyl acetate. The combined organic extracts were washed with water, brine, dried over Na2SO4 , filtered and evaporated under reduced pressure to give the title compound as a beige solid (154 mg, 83%). MS: 337.14 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 7.93 - 7.89 (m, 2H), 7.88 (d, J = 2.0 Hz, 1H), 7.58 (d, J = 8.5 Hz, 1H), 7.49 - 7.44 (m, 2H), 7.36 (dd, J = 8.5, 2.0 Hz, 1H), 3.49 (s, 3H), 2.39 (s, 3H).

製備實例 46 步驟 A 在0℃下向NaH (0.47 g, 19.55 mmol)於THF (20 mL)中之懸浮液逐滴添加市售1,3,4,5-四氫-2H-吡啶并[4,3-b]吲哚-2-甲酸第三丁基酯(2.0 g, 7.35 mmol) (溶解於THF中)。然後將混合物在室溫下攪拌1 h。之後,在0℃下添加甲苯磺醯氯(1.5 g, 7.82 mmol) (溶解於THF中),且然後將混合物在室溫下攪拌2 h。如藉由TLC所檢查在反應完成後,利用冰水將反應混合物淬滅,之後使用乙酸乙酯進行萃取。將有機層濃縮,得到標題化合物(1.8 g, 65%)。產物原樣用於下一步驟。 MS: 427.1 (M+H)+步驟 B 向來自上文步驟A之標題化合物(1.8 g, 4.22 mmol)於DCM (20 mL)中之溶液添加於二噁烷中之4 M HCl (5 mL)。將反應混合物攪拌過夜。在反應完成後,使該反應混合物蒸發以去除溶劑並用二乙醚洗滌殘餘物,得到呈灰白色固體之標題化合物(1.0 g, 66%)。 MS: 361.3 (M+H)+ Preparation Example 46 Step A To a suspension of NaH (0.47 g, 19.55 mmol) in THF (20 mL) was added dropwise commercially available 1,3,4,5-tetrahydro-2H-pyrido[4,3- b] Indole-2-carboxylic acid tert-butyl ester (2.0 g, 7.35 mmol) (dissolved in THF). The mixture was then stirred at room temperature for 1 h. Afterwards, tosyl chloride (1.5 g, 7.82 mmol) (dissolved in THF) was added at 0 °C, and the mixture was then stirred at room temperature for 2 h. After the reaction was complete as checked by TLC, the reaction mixture was quenched with ice water, followed by extraction with ethyl acetate. The organic layer was concentrated to give the title compound (1.8 g, 65%). The product was used as received in the next step. MS: 427.1 (M+H) + . Step B To a solution of the title compound from Step A above (1.8 g, 4.22 mmol) in DCM (20 mL) was added 4 M HCl in dioxane (5 mL). The reaction mixture was stirred overnight. After the reaction was completed, the reaction mixture was evaporated to remove the solvent and the residue was washed with diethyl ether to give the title compound as an off-white solid (1.0 g, 66%). MS: 361.3 (M+H) + .

製備實例 47 步驟 A 在0℃下向市售(2-氟苯基)肼鹽酸鹽(2 g, 12.34 mmol)及4-側氧基六氫吡啶-1-甲酸第三丁基酯(2.45 g, 12.34 mmol)於二噁烷(20 mL)中之溶液添加濃H2 SO4 (2 mL)。然後將反應混合物在100℃下加熱4 h。如藉由TLC所證實,在反應完成後,將反應混合物冷卻至25℃且然後濃縮。藉由10% NaOH溶液使粗製混合物鹼化,且將沈澱物過濾出。將固體用水洗滌並在真空下乾燥,得到標題化合物(1.7 g, 75%)。 MS: 191.0 (M+H)+步驟 B 在室溫下向來自上文步驟A之標題化合物(1.7g,粗製)於THF (20 mL)中之攪拌溶液添加TEA (3.76 mL, 26.82 mmol)及二碳酸二-第三丁基酯(2.34 mL, 10.73 mmol)。將混合物攪拌12 h。如藉由TLC所證實,在反應完成後,將溶劑去除且使用Biotage Isolera One純化系統,採用EtOAc/庚烷梯度(10/80 => 80/20),在矽膠管柱上純化粗製反應混合物,得到呈淡黃色固體之標題化合物(700 mg, 27%)。 MS: 291.1 (M+H)+ Preparation Example 47 Step A : Commercially available (2-fluorophenyl)hydrazine hydrochloride (2 g, 12.34 mmol) and 4-side oxyhexahydropyridine-1-carboxylic acid tert-butyl ester (2.45 g, 12.34 mmol) were prepared at 0°C. mmol) in dioxane (20 mL) was added concentrated H 2 SO 4 (2 mL). The reaction mixture was then heated at 100 °C for 4 h. After the reaction was complete as confirmed by TLC, the reaction mixture was cooled to 25°C and then concentrated. The crude mixture was basified by 10% NaOH solution and the precipitate was filtered off. The solid was washed with water and dried under vacuum to give the title compound (1.7 g, 75%). MS: 191.0 (M+H) + . Step B To a stirred solution of the title compound from Step A above (1.7 g, crude) in THF (20 mL) was added TEA (3.76 mL, 26.82 mmol) and di-tert-butyl dicarbonate at room temperature. (2.34 mL, 10.73 mmol). The mixture was stirred for 12 h. After the reaction was complete, as confirmed by TLC, the solvent was removed and the crude reaction mixture was purified on a silica column using a Biotage Isolera One purification system using an EtOAc/heptane gradient (10/80 => 80/20), The title compound was obtained as a pale yellow solid (700 mg, 27%). MS: 291.1 (M+H) + .

1H-NMR(400MHz,DMSO-d 6 )δ=11.38(bs,1H),7.22(d,J=7.60Hz,1H),6.96-6.85(m,2H),4.53(s,2H),3.70-3.71(m,2H),2.78(bs,2H),1.44(s,9H)。 1 H-NMR (400MHz, DMSO- d 6 ) δ = 11.38 (bs, 1H), 7.22 (d, J = 7.60Hz, 1H), 6.96-6.85 (m, 2H), 4.53 (s, 2H), 3.70 -3.71(m,2H),2.78(bs,2H),1.44(s,9H).

步驟CStep C

在0℃下向NaH(144mg,3.61mmol)於THF(15mL)中之懸浮液逐滴添加來自上文步驟B之標題化合物(700mg,2.41mmol)(溶解於THF中)。然後將混合物在室溫下攪拌1h。之後,在0℃下添加甲苯磺醯氯(549mg,2.89mmol)(溶解於THF中),且然後將混合物在室溫下攪拌2h。如藉由TLC所證實,在反應完成後,利用冰水將反應混合物淬滅,之後使用乙酸乙酯進行萃取。將有機層濃縮,且使用Biotage Isolera One純化系統,採用EtOAc/己烷梯度(10/80=>80/20),在矽膠管柱上純化粗製反應混合物,得到標題化合物(900mg,84%)。 To a suspension of NaH (144 mg, 3.61 mmol) in THF (15 mL) was added the title compound from step B above (700 mg, 2.41 mmol) (dissolved in THF) dropwise at 0°C. The mixture was then stirred at room temperature for 1 h. Afterwards, tosyl chloride (549 mg, 2.89 mmol) (dissolved in THF) was added at 0 °C, and the mixture was then stirred at room temperature for 2 h. After the reaction was complete, as confirmed by TLC, the reaction mixture was quenched with ice water and then extracted with ethyl acetate. The organic layer was concentrated, and the crude reaction mixture was purified on a silica column using a Biotage Isolera One purification system using an EtOAc/hexane gradient (10/80=>80/20) to give the title compound (900 mg, 84%).

MS:345.1(M-Boc)。 MS: 345.1(M-Boc).

1H-NMR(400MHz,DMSO-d 6 )δ=7.79(d,J=8.04Hz,2H),7.41(d,J=8.24Hz,2H),7.34(d,J=7.68Hz,2H),7.22-7.23(m,1H),7.07-7.09(m,1H),4.50(s,2H),3.70-3.72(m,2H),3.17(bs,2H),2.36(s,3H),1.40(s,9H)。 1 H-NMR(400MHz, DMSO- d 6 )δ=7.79(d, J =8.04Hz,2H),7.41(d, J =8.24Hz,2H),7.34(d, J =7.68Hz,2H), 7.22-7.23(m,1H),7.07-7.09(m,1H),4.50(s,2H),3.70-3.72(m,2H),3.17(bs,2H),2.36(s,3H),1.40( s,9H).

步驟DStep D

向來自上文步驟C之標題化合物(900mg,2.02mmol)於DCM(10mL)中之溶液添加於二噁烷中之4N HCl(5mL)。將反應混合物在室溫下攪拌2h。在反應完成後,使該反應混合物蒸發以去除溶劑並用二乙醚洗滌殘餘物,得到呈淡棕色固體之標題化合物(450mg,65%)。 To a solution of the title compound from Step C above (900 mg, 2.02 mmol) in DCM (10 mL) was added 4N HCl in dioxane (5 mL). The reaction mixture was stirred at room temperature for 2 h. After the reaction was completed, the reaction mixture was evaporated to remove the solvent and the residue was washed with diethyl ether to give the title compound as a light brown solid (450 mg, 65%).

MS:345.1(M+H)+MS: 345.1(M+H) + .

製備實例 48 步驟 A 在0℃下向NaH (300 mg, 12.39 mmol)於THF (15 mL)中之懸浮液逐滴添加來自製備實例47步驟B之標題化合物(1.2 g, 4.13 mmol) (溶解於THF中)。然後將反應混合物在室溫下攪拌1 h。之後,在0℃下添加碘甲烷(0.5 mL, 8.26 mmol),且然後將混合物在室溫下攪拌2 h。完成之後,利用冰水將反應混合物淬滅,之後使用乙酸乙酯進行萃取。將有機層濃縮,且使用Biotage Isolera One純化系統,採用EtOAc/己烷梯度(10/80 => 80/20),在矽膠管柱上純化粗製反應混合物,得到標題化合物(900 mg, 72%)。 MS: 305.3 (M+H)+1 H-NMR (400 MHz, DMSO-d6 ) δ = 7.23 (d,J = 9.60 Hz, 1H), 6.88-6.91 (m, 2H), 4.51 (s, 2H), 3.71-3.73 (m, 5H), 2.73-2.77 (m, 2H), 1.50 (s, 9H)。步驟 B 向來自上文步驟A之標題化合物(900 mg, 2.96 mmol)於DCM (10 mL)中之溶液添加於二噁烷中之4 M HCl (5 mL)。將反應混合物在室溫下攪拌2 h。在反應完成後,使該反應混合物蒸發以去除溶劑並用二乙醚洗滌殘餘物,得到呈淡棕色固體之標題化合物(500 mg, 83%)。 MS: 205.1 (M+H)+ Preparation Example 48 Step A To a suspension of NaH (300 mg, 12.39 mmol) in THF (15 mL) was added the title compound from Preparative Example 47, Step B (1.2 g, 4.13 mmol) (dissolved in THF) dropwise at 0°C. . The reaction mixture was then stirred at room temperature for 1 h. Afterwards, methyl iodide (0.5 mL, 8.26 mmol) was added at 0 °C, and the mixture was then stirred at room temperature for 2 h. After completion, the reaction mixture was quenched with ice water and extracted with ethyl acetate. The organic layer was concentrated, and the crude reaction mixture was purified on a silica column using a Biotage Isolera One purification system using an EtOAc/hexane gradient (10/80 => 80/20) to give the title compound (900 mg, 72%) . MS: 305.3 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 7.23 (d, J = 9.60 Hz, 1H), 6.88-6.91 (m, 2H), 4.51 (s, 2H), 3.71-3.73 (m, 5H ), 2.73-2.77 (m, 2H), 1.50 (s, 9H). Step B To a solution of the title compound from Step A above (900 mg, 2.96 mmol) in DCM (10 mL) was added 4 M HCl in dioxane (5 mL). The reaction mixture was stirred at room temperature for 2 h. After the reaction was completed, the reaction mixture was evaporated to remove the solvent and the residue was washed with diethyl ether to give the title compound as a light brown solid (500 mg, 83%). MS: 205.1 (M+H) + .

製備實例 49 步驟 A 向市售2-溴-6-氯-1,8-萘啶(0.2 g, 0.821 mmol)於無水乙腈(5 mL)中之溶液添加碳酸鉀(0.335 mg, 2.46 mmol)及嗎啉(0.11 g, 1.23 mmol)。將反應混合物加熱至100℃持續3小時。將反應混合物在真空下濃縮,且使用Biotage Isolera One純化系統,採用EtOAc/己烷梯度(10/80 => 80/20),在矽膠管柱上純化粗製反應混合物,得到呈淡黃色固體之標題化合物(170 mg, 83%)。 MS: 250.1 (M+H)+1 H-NMR (400 MHz, DMSO-d6 ) δ = 8.71 (s, 1H), 8.07-8.08 (m, 1H), 7.82-7.95 (m, 1H), 7.55-7.56 (m, 1H), 3.72 (s, 8H)。 Preparation Example 49 Step A : To a solution of commercially available 2-bromo-6-chloro-1,8-naphthyridine (0.2 g, 0.821 mmol) in anhydrous acetonitrile (5 mL), potassium carbonate (0.335 mg, 2.46 mmol) and morpholine ( 0.11 g, 1.23 mmol). The reaction mixture was heated to 100°C for 3 hours. The reaction mixture was concentrated under vacuum, and the crude reaction mixture was purified on a silica column using a Biotage Isolera One purification system using an EtOAc/hexane gradient (10/80 => 80/20) to obtain the title product as a pale yellow solid. compound (170 mg, 83%). MS: 250.1 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 8.71 (s, 1H), 8.07-8.08 (m, 1H), 7.82-7.95 (m, 1H), 7.55-7.56 (m, 1H), 3.72 (s, 8H).

製備實例 50 步驟 A 向市售2-溴-6-氯-1,7-萘啶(0.5 g, 2.05 mmol)於無水乙腈(5 mL)中之溶液添加碳酸鉀(0.837 mg, 6.16 mmol)及嗎啉(0.27 g, 3.08 mmol)。將反應混合物加熱至100℃持續3小時。將反應混合物在真空下濃縮,且使用Biotage Isolera One純化系統,採用EtOAc/己烷梯度(10/80 => 80/20),在矽膠管柱上純化粗製反應混合物,得到呈淡黃色固體之標題化合物(270 mg, 53%)。 MS: 250.1 (M+H)+1 H-NMR (400 MHz, DMSO-d6 ) δ = 9.01 (s, 1H), 7.91-7.93 (m, 2H), 6.92-6.93 (m, 1H), 3.71 (s, 8H)。 Preparation Example 50 Step A : To a solution of commercially available 2-bromo-6-chloro-1,7-naphthyridine (0.5 g, 2.05 mmol) in anhydrous acetonitrile (5 mL), potassium carbonate (0.837 mg, 6.16 mmol) and morpholine ( 0.27 g, 3.08 mmol). The reaction mixture was heated to 100°C for 3 hours. The reaction mixture was concentrated under vacuum, and the crude reaction mixture was purified on a silica column using a Biotage Isolera One purification system using an EtOAc/hexane gradient (10/80 => 80/20) to obtain the title product as a pale yellow solid. compound (270 mg, 53%). MS: 250.1 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 9.01 (s, 1H), 7.91-7.93 (m, 2H), 6.92-6.93 (m, 1H), 3.71 (s, 8H).

製備實例 51 步驟 A 向市售2,7-二氯喹啉(0.5 g, 2.52 mmol)於無水DMF (5 mL)中之溶液添加碳酸鉀(1 g, 7.52 mmol)及嗎啉(0.32 g, 3.78 mmol)。將反應混合物加熱至100℃持續3小時。將反應混合物在真空下濃縮,得到呈淡黃色固體之標題化合物(500 mg, 80%)。 MS: 249.1 (M+H)+ Preparation Example 51 Step A. To a solution of commercially available 2,7-dichloroquinoline (0.5 g, 2.52 mmol) in anhydrous DMF (5 mL), potassium carbonate (1 g, 7.52 mmol) and morpholine (0.32 g, 3.78 mmol) were added. The reaction mixture was heated to 100°C for 3 hours. The reaction mixture was concentrated in vacuo to give the title compound as a pale yellow solid (500 mg, 80%). MS: 249.1 (M+H) + .

製備實例 52 步驟 A 向市售2,6-二氯喹啉(0.5 g, 2.52 mmol)於無水DMF (5 mL)中之溶液添加碳酸鉀(1 g, 7.52 mmol)及嗎啉(0.32 g, 3.78 mmol)。將反應混合物加熱至100℃持續3小時。將反應混合物在真空下濃縮,且使用Biotage Isolera One純化系統,採用EtOAc/己烷梯度(10/80 => 80/20),在矽膠管柱上純化粗製反應混合物,得到呈淡黃色固體之標題化合物(500 mg, 80%)。 MS: 249.1 (M+H)+ 1 H-NMR (400 MHz, DMSO-d6 ) δ = 8.06 (d,J = 12.40 Hz, 1H), 7.84 (d,J = 2.40 Hz, 1H), 7.51-7.52 (m, 2H), 7.31 (d,J = 12.40 Hz, 1H), 3.66-3.67 (m, 8H)。 Preparation Example 52 Step A. To a solution of commercially available 2,6-dichloroquinoline (0.5 g, 2.52 mmol) in anhydrous DMF (5 mL) was added potassium carbonate (1 g, 7.52 mmol) and morpholine (0.32 g, 3.78 mmol). The reaction mixture was heated to 100°C for 3 hours. The reaction mixture was concentrated under vacuum, and the crude reaction mixture was purified on a silica column using a Biotage Isolera One purification system using an EtOAc/hexane gradient (10/80 => 80/20) to obtain the title product as a pale yellow solid. compound (500 mg, 80%). MS: 249.1 (M+H) + 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 8.06 (d, J = 12.40 Hz, 1H), 7.84 (d, J = 2.40 Hz, 1H), 7.51- 7.52 (m, 2H), 7.31 (d, J = 12.40 Hz, 1H), 3.66-3.67 (m, 8H).

製備實例 53 步驟 A 將市售4,6-二氯吡啶-3-胺(8.0 g, 49.07 mmol)及異硫氰酸苯甲醯基酯(7.3 mL, 53.98 mmol)於丙酮(120 mL)中之溶液在60℃下攪拌3小時。藉由TLC監測反應。將溶劑蒸發且將固體過濾,用正己烷(100 mL)洗滌並乾燥,得到呈灰白色固體之期望產物(14.0 g, 87%)。 MS: 328.0 (M+H)+1 H-NMR (400 MHz, DMSO-d6 ) δ =12.39 (s, 1H), 12.02 (s, 1H), 8.74 (s, 1H), 7.98-7.99 (m, 3H), 7.67-7.68 (m, 1H), 7.56 (t,J = 7.60 Hz, 2H)。步驟 B 在0℃向來自上文步驟A之標題化合物(14.0 g, 42.94 mmol)於N-甲基-2-吡咯啶酮(NMP) (70 mL)中之溶液添加甲醇鈉(NaOMe) (4.6 g, 85.88 mmol)。然後將混合物加熱至120℃且繼續攪拌4小時。藉由TLC監測反應。將反應混合物傾倒至冷水(300 mL)中且獲得白色沈澱物。將固體過濾,用水(300 mL)及正己烷(200 mL)洗。將化合物在真空下乾燥6 h,得到呈白色固體之期望產物(14.0 g,粗製)。產物原樣用於下一步驟。 MS: 290.0 (M+H)+步驟 C 將來自上文步驟B之標題化合物(14.0 g, 48.4 mmol)於70% H2 SO4 (50.0 mL)中之懸浮液在110℃加熱4小時。將反應混合物冷卻至室溫,且將反應混合物緩慢傾倒至200 mL冷水(0℃)中。然後,藉由添加50% NaOH水溶液將反應混合物調整至鹼性pH。然後,用EtOAc (6 × 100 mL)萃取化合物。使合併之有機層經Na2 SO4 乾燥並過濾,然後將溶劑濃縮,得到呈淺黃色固體之期望產物(6 g, 67%)。 MS: 186.1 (M+H)+ 1 H-NMR (400 MHz, DMSO-d6 ) δ = 8.30 (s, 1H), 7.86 (s, 1H)。步驟 D 在0℃利用注射器經10 min之時期向來自上文步驟C之標題化合物(5.0 g, 27.02 mmol)於乙腈(120 mL)中之懸浮液添加亞硝酸第三丁基酯( 4.8 mL, 40.54 mmol)。然後,逐份添加溴化銅(II)(9.0 g, 40.54 mmol)。在0℃下30分鐘後,將反應混合物升溫至室溫2.5小時,藉由TLC監測反應之進展。在反應完成後,將溶劑蒸發且用水(200 mL)及5% MeOH/ DCM (3 × 200 mL)稀釋。將合併之有機物用鹽水(50 mL)洗,經Na2 SO4 乾燥,過濾並在減壓下濃縮,得到呈白色固體之標題化合物(6.5 g)。產物原樣用於下一步驟。 MS: 250.9 (M+H)+ 步驟 E 向來自上文步驟D之標題化合物(6.5 g, 26.09 mmol)於無水DCM (100 mL)中之溶液添加三乙胺(11.2 mL, 81.5 mmol)及嗎啉(2.8 mL, 28.13 mmol)。將反應混合物在室溫攪拌4小時。將反應混合物在真空下濃縮。在矽膠管柱上使用Biotage Isolera One純化系統,採用EtOAc/己烷梯度(10/80 => 80/20)純化粗製反應混合物,得到呈淡黃色固體之標題化合物(4.7g, 71%)。 MS: 256.1 (M+H)+1 H-NMR (400 MHz, DMSO-d6 ) δ = 8.48 (s, 1H), 8.05 (s, 1H), 3.73-3.74 (m, 4H), 3.60-3.61 (m, 4H)。 Preparation Example 53 Step A : Add a solution of commercially available 4,6-dichloropyridin-3-amine (8.0 g, 49.07 mmol) and benzyl isothiocyanate (7.3 mL, 53.98 mmol) in acetone (120 mL). Stir at 60°C for 3 hours. The reaction was monitored by TLC. The solvent was evaporated and the solid was filtered, washed with n-hexane (100 mL) and dried to give the desired product as an off-white solid (14.0 g, 87%). MS: 328.0 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ =12.39 (s, 1H), 12.02 (s, 1H), 8.74 (s, 1H), 7.98-7.99 (m, 3H), 7.67-7.68 (m , 1H), 7.56 (t, J = 7.60 Hz, 2H). Step B To a solution of the title compound from Step A above (14.0 g, 42.94 mmol) in N-methyl-2-pyrrolidone (NMP) (70 mL) was added sodium methoxide (NaOMe) (4.6 g, 85.88 mmol). The mixture was then heated to 120°C and stirring continued for 4 hours. The reaction was monitored by TLC. The reaction mixture was poured into cold water (300 mL) and a white precipitate was obtained. The solid was filtered and washed with water (300 mL) and n-hexane (200 mL). The compound was dried under vacuum for 6 h to obtain the desired product (14.0 g, crude) as a white solid. The product was used as received in the next step. MS: 290.0 (M+H) + . Step C A suspension of the title compound from Step B above (14.0 g, 48.4 mmol) in 70% H2SO4 (50.0 mL) was heated at 110°C for 4 hours. The reaction mixture was cooled to room temperature, and the reaction mixture was slowly poured into 200 mL of cold water (0°C). The reaction mixture was then adjusted to alkaline pH by adding 50% aqueous NaOH solution. Then, the compound was extracted with EtOAc (6 × 100 mL). The combined organic layers were dried over Na2SO4 and filtered, and the solvent was concentrated to afford the desired product as a pale yellow solid (6 g, 67%). MS: 186.1 (M+H) + 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 8.30 (s, 1H), 7.86 (s, 1H). Step D To a suspension of the title compound from Step C above (5.0 g, 27.02 mmol) in acetonitrile (120 mL) was added tert-butyl nitrite ( 4.8 mL, 4.8 mL, via syringe over 10 min at 0°C). 40.54 mmol). Then, copper (II) bromide (9.0 g, 40.54 mmol) was added portionwise. After 30 minutes at 0°C, the reaction mixture was warmed to room temperature for 2.5 hours and the progress of the reaction was monitored by TLC. After the reaction was complete, the solvent was evaporated and diluted with water (200 mL) and 5% MeOH/DCM (3 × 200 mL). The combined organics were washed with brine (50 mL), dried over Na2SO4 , filtered and concentrated under reduced pressure to give the title compound as a white solid ( 6.5 g). The product was used as received in the next step. MS: 250.9 (M+H) + Step E To a solution of the title compound from Step D above (6.5 g, 26.09 mmol) in anhydrous DCM (100 mL) was added triethylamine (11.2 mL, 81.5 mmol) and methyl Phenoline (2.8 mL, 28.13 mmol). The reaction mixture was stirred at room temperature for 4 hours. The reaction mixture was concentrated in vacuo. The crude reaction mixture was purified on a silica column using a Biotage Isolera One purification system and an EtOAc/hexane gradient (10/80 => 80/20) to obtain the title compound (4.7 g, 71%) as a pale yellow solid. MS: 256.1 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 8.48 (s, 1H), 8.05 (s, 1H), 3.73-3.74 (m, 4H), 3.60-3.61 (m, 4H).

製備實例 54 向2,5-二氯-1,3-苯并噁唑(0.25 g, 0.0013 mol)於乙腈(15 mL)中之攪拌溶液添加K2 CO3 (0.538 g, 0.0039 mol)及5-氧雜-8-氮雜螺[3.5]壬烷(0.178 g, 0.0014 mol)。之後,將反應混合物加熱至70℃持續12 h。根據TLC在反應完成後,向反應混合物添加DCM及水(50 mL)。將有機層分離,經硫酸鈉乾燥,過濾且然後濃縮,得到呈白色固體之標題化合物(0.30 g, 80.21%)。 MS: 279.1 (M+H)+1 H-NMR (400 MHz, DMSO-d6 ) δ =7.44 (d,J = 8.40 Hz, 1H), 7.36-7.37 (m, 1H), 7.06 (dd,J = 2.00, 8.40 Hz, 1H), 3.64-3.65 (m, 2H), 3.60 (s, 2H), 3.54-3.55 (m, 2H), 1.96-1.97 (m, 4H), 1.71-1.74 (m, 2H)。 Preparation Example 54 To a stirred solution of 2,5-dichloro-1,3-benzoxazole (0.25 g, 0.0013 mol) in acetonitrile (15 mL) was added K 2 CO 3 (0.538 g, 0.0039 mol) and 5-oxa -8-Azaspiro[3.5]nonane (0.178 g, 0.0014 mol). Afterwards, the reaction mixture was heated to 70 °C for 12 h. After the reaction was complete according to TLC, DCM and water (50 mL) were added to the reaction mixture. The organic layer was separated, dried over sodium sulfate, filtered and then concentrated to give the title compound as a white solid (0.30 g, 80.21%). MS: 279.1 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ =7.44 (d, J = 8.40 Hz, 1H), 7.36-7.37 (m, 1H), 7.06 (dd, J = 2.00, 8.40 Hz, 1H), 3.64-3.65 (m, 2H), 3.60 (s, 2H), 3.54-3.55 (m, 2H), 1.96-1.97 (m, 4H), 1.71-1.74 (m, 2H).

製備實例 55 向2,5-二氯-1,3-苯并噁唑(0.60 g, 0.0032 mol)於乙腈(15 mL)中之攪拌溶液添加K2 CO3 (1.32 g, 0.0096 mol)及2-甲氧基乙烷-1-胺(0.266 g, 0.0035 mol)。之後,將反應混合物加熱至70℃持續12 h。根據TLC在反應完成後,向反應混合物添加DCM及水(50 mL)。將有機層分離,經硫酸鈉乾燥,過濾且然後濃縮,得到呈棕色固體之標題化合物(0.41 g, 56.24%)。 MS: 226.9 (M+H)+1 H-NMR (400 MHz, DMSO-d6 ) δ = 8.26 (s, 1H), 7.35 (d,J = 11.20 Hz, 1H), 7.28 (d,J = 2.80 Hz, 1H), 6.99 (dd,J = 2.80, 11.20 Hz, 1H), 3.48 (d,J = 7.20 Hz, 4H), 3.27 (s, 3H)。 Preparation Example 55 To a stirred solution of 2,5-dichloro-1,3-benzoxazole (0.60 g, 0.0032 mol) in acetonitrile (15 mL) was added K 2 CO 3 (1.32 g, 0.0096 mol) and 2-methoxy Ethane-1-amine (0.266 g, 0.0035 mol). Afterwards, the reaction mixture was heated to 70 °C for 12 h. After the reaction was complete according to TLC, DCM and water (50 mL) were added to the reaction mixture. The organic layer was separated, dried over sodium sulfate, filtered and then concentrated to give the title compound as a brown solid (0.41 g, 56.24%). MS: 226.9 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 8.26 (s, 1H), 7.35 (d, J = 11.20 Hz, 1H), 7.28 (d, J = 2.80 Hz, 1H), 6.99 (dd, J = 2.80, 11.20 Hz, 1H), 3.48 (d, J = 7.20 Hz, 4H), 3.27 (s, 3H).

製備實例 56 向2,5-二氯-1,3-苯并噁唑(0.719 g, 0.0038 mol)於DCM (15 mL)中之攪拌溶液添加TEA (2.67 ml, 0.0191 mol)及(2R)-2-甲基嗎啉(0.5 g, 0.00494 mol),且在25℃下攪拌12 h。根據TLC在反應完成後,向反應混合物添加DCM及水(50 mL)。將有機層分離,經硫酸鈉乾燥,過濾且然後濃縮,得到呈白色固體之標題化合物(0.6 g, 45.4%)。 MS: 253.2 (M+H)+1 H-NMR (400 MHz, DMSO-d6 ) δ = 7.43 (d,J = 11.20 Hz, 1H), 7.35 (d,J = 2.40 Hz, 1H), 7.05 (dd,J = 2.80, 11.20 Hz, 1H), 3.89-3.90 (m, 3H), 3.56-3.59 (m, 2H), 3.17-3.18 (m, 1H), 2.86-2.89 (m, 1H), 1.15 (d,J = 8.40 Hz, 3H)。 Preparation Example 56 To a stirred solution of 2,5-dichloro-1,3-benzoxazole (0.719 g, 0.0038 mol) in DCM (15 mL) was added TEA (2.67 ml, 0.0191 mol) and (2R)-2-methyl morpholine (0.5 g, 0.00494 mol) and stirred at 25°C for 12 h. After the reaction was complete according to TLC, DCM and water (50 mL) were added to the reaction mixture. The organic layer was separated, dried over sodium sulfate, filtered and then concentrated to give the title compound as a white solid (0.6 g, 45.4%). MS: 253.2 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 7.43 (d, J = 11.20 Hz, 1H), 7.35 (d, J = 2.40 Hz, 1H), 7.05 (dd, J = 2.80, 11.20 Hz, 1H), 3.89-3.90 (m, 3H), 3.56-3.59 (m, 2H), 3.17-3.18 (m, 1H), 2.86-2.89 (m, 1H), 1.15 (d, J = 8.40 Hz, 3H) .

製備實例 57 向2,5-二氯-1,3-苯并噁唑(0.360 g, 0.0019 mol)於DCM (15 mL)中之攪拌溶液添加TEA (1.33 ml, 0.0095 mol)及(2S)-2-甲基嗎啉(0.25 g, 0.00247 mol),且在25℃下攪拌12 h。根據TLC在反應完成後,向反應混合物添加DCM及水(50 mL)。將有機層分離,經硫酸鈉乾燥,過濾且然後濃縮。使用Biotage Isolera One純化系統,採用於石油醚中之20%-25%乙酸乙酯,在矽膠管柱上純化粗製反應混合物,得到呈白色固體之標題化合物(0.3 g, 45.5%)。 MS: 253.1 (M+H)+1 H-NMR (400 MHz, DMSO-d6 ) δ = 7.43 (d,J = 8.48 Hz, 1H), 7.34-7.35 (m, 1H), 7.05 (dd,J = 2.08, 8.46 Hz, 1H), 3.90-3.90 (m, 3H), 3.61-3.62 (m, 2H), 3.24-3.25 (m, 1H), 2.87-2.89 (m, 1H), 1.15 (d,J = 6.20 Hz, 3H)。 Preparation Example 57 To a stirred solution of 2,5-dichloro-1,3-benzoxazole (0.360 g, 0.0019 mol) in DCM (15 mL) was added TEA (1.33 ml, 0.0095 mol) and (2S)-2-methyl morpholine (0.25 g, 0.00247 mol) and stirred at 25°C for 12 h. After the reaction was complete according to TLC, DCM and water (50 mL) were added to the reaction mixture. The organic layer was separated, dried over sodium sulfate, filtered and then concentrated. The crude reaction mixture was purified on a silica column using a Biotage Isolera One purification system using 20%-25% ethyl acetate in petroleum ether to obtain the title compound as a white solid (0.3 g, 45.5%). MS: 253.1 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 7.43 (d, J = 8.48 Hz, 1H), 7.34-7.35 (m, 1H), 7.05 (dd, J = 2.08, 8.46 Hz, 1H), 3.90-3.90 (m, 3H), 3.61-3.62 (m, 2H), 3.24-3.25 (m, 1H), 2.87-2.89 (m, 1H), 1.15 (d, J = 6.20 Hz, 3H).

製備實例 58 在0℃下向2,5-二氯-1,3-苯并噁唑(1.20 g, 6.38 mmol)於無水DCM (50 mL)中之溶液添加(3R)-3-甲基嗎啉(0.775 g, 7.65 mmol)及Et3 N (1.94 g, 19.10 mmol),且在25℃下攪拌4 h。在反應完成後(藉由TLC監測),用水(20 mL)稀釋反應混合物並用DCM (20 mL × 2)萃取。使合併之有機萃取物經Na2 SO4 乾燥,過濾並在減壓下蒸發。 使用Biotage Isolera One純化系統,採用於石油醚中之10%-20%乙酸乙酯,在矽膠管柱上純化粗製反應混合物,得到標題化合物(1.0 g, 59.9%)。 MS: 252.9 (M+H)+1 H-NMR (400 MHz, DMSO-d6 ) δ = 7.43 (d,J = 11.20 Hz, 1H), 7.35 (d,J = 2.80 Hz, 1H), 7.04 (dd,J = 2.80, 11.40 Hz, 1H), 4.17-4.19 (m, 1H), 3.69-3.76 (m, 3H), 3.42-3.43 (m, 5H), 1.30 (d,J = 9.20 Hz, 3H)。 Preparation Example 58 To a solution of 2,5-dichloro-1,3-benzoxazole (1.20 g, 6.38 mmol) in anhydrous DCM (50 mL) at 0 °C was added (3R)-3-methylmorpholine (0.775 g, 7.65 mmol) and Et 3 N (1.94 g, 19.10 mmol), and stirred at 25°C for 4 h. After the reaction was complete (monitored by TLC), the reaction mixture was diluted with water (20 mL) and extracted with DCM (20 mL × 2). The combined organic extracts were dried over Na2SO4 , filtered and evaporated under reduced pressure . The crude reaction mixture was purified on a silica column using a Biotage Isolera One purification system using 10%-20% ethyl acetate in petroleum ether to give the title compound (1.0 g, 59.9%). MS: 252.9 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 7.43 (d, J = 11.20 Hz, 1H), 7.35 (d, J = 2.80 Hz, 1H), 7.04 (dd, J = 2.80, 11.40 Hz, 1H), 4.17-4.19 (m, 1H), 3.69-3.76 (m, 3H), 3.42-3.43 (m, 5H), 1.30 (d, J = 9.20 Hz, 3H).

製備實例 59 在0℃下向2,5-二氯-1,3-苯并噁唑(1.20 g, 6.38 mmol)於無水DCM (50 mL)中之溶液添加(3S)-3-甲基嗎啉(0.775 g, 7.65 mmol)及Et3 N (1.94 g, 19.10 mmol),且在25℃下攪拌4 h。在反應完成後(藉由TLC監測),用H2 O (20 mL)稀釋反應混合物並用DCM (20 mL × 2)萃取。使合併之有機萃取物經Na2 SO4 乾燥,過濾並在減壓下蒸發。 使用Biotage Isolera One純化系統,採用於石油醚中之10%-20%乙酸乙酯,在矽膠管柱上純化粗製反應混合物,得到標題化合物(0.8 g,49.6%)。 MS: 252.9 (M+H)+1 H-NMR (400 MHz, DMSO-d6 ) δ = 7.43 (d,J = 11.20 Hz, 1H), 7.35 (d,J = 2.80 Hz, 1H), 7.04 (dd,J = 2.80, 11.20 Hz, 1H), 4.17-4.19 (m, 1H), 3.73-3.75 (m, 4H), 3.46-3.47 (m, 2H), 1.30 (d,J = 9.20 Hz, 3H), 。 Preparation Example 59 To a solution of 2,5-dichloro-1,3-benzoxazole (1.20 g, 6.38 mmol) in anhydrous DCM (50 mL) at 0 °C was added (3S)-3-methylmorpholine (0.775 g, 7.65 mmol) and Et 3 N (1.94 g, 19.10 mmol), and stirred at 25°C for 4 h. After the reaction was complete (monitored by TLC), the reaction mixture was diluted with H2O (20 mL) and extracted with DCM (20 mL × 2). The combined organic extracts were dried over Na2SO4 , filtered and evaporated under reduced pressure . The crude reaction mixture was purified on a silica column using a Biotage Isolera One purification system using 10%-20% ethyl acetate in petroleum ether to give the title compound (0.8 g, 49.6%). MS: 252.9 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 7.43 (d, J = 11.20 Hz, 1H), 7.35 (d, J = 2.80 Hz, 1H), 7.04 (dd, J = 2.80, 11.20 Hz, 1H), 4.17-4.19 (m, 1H), 3.73-3.75 (m, 4H), 3.46-3.47 (m, 2H), 1.30 (d, J = 9.20 Hz, 3H), .

製備實例 60 向2,5-二氯-1,3-苯并噁唑(0.5 g, 0.00265 mol)於乙腈(10 mL)中之攪拌溶液添加K2 CO3 (1.1 g, 0.00797 mol)及2-甲氧基-N-甲基乙烷-1-胺(0.284 g, 0.00319 mol)。之後,將反應混合物加熱至70℃持續12 h。根據TLC在反應完成後,向反應混合物添加DCM及水(50 mL)。將有機層分離,經硫酸鈉乾燥,過濾且然後濃縮,得到呈棕色液體之標題化合物(0.61 g, 95%)。 MS: 241.1 (M+H)+1 H-NMR (400 MHz, DMSO-d6 ) δ = 7.40 (d,J = 11.20 Hz, 1H), 7.30 (d,J = 2.80 Hz, 1H), 7.00 (dd,J = 2.80, 11.20 Hz, 1H), 3.68 (t,J = 6.40 Hz, 2H), 3.58 (t,J = 7.20 Hz, 2H), 3.27 (s, 3H), 3.16 (s, 3H)。 Preparation Example 60 To a stirred solution of 2,5-dichloro-1,3-benzoxazole (0.5 g, 0.00265 mol) in acetonitrile (10 mL) was added K 2 CO 3 (1.1 g, 0.00797 mol) and 2-methoxy 1-N-Methylethane-1-amine (0.284 g, 0.00319 mol). Afterwards, the reaction mixture was heated to 70 °C for 12 h. After the reaction was complete according to TLC, DCM and water (50 mL) were added to the reaction mixture. The organic layer was separated, dried over sodium sulfate, filtered and then concentrated to give the title compound as a brown liquid (0.61 g, 95%). MS: 241.1 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 7.40 (d, J = 11.20 Hz, 1H), 7.30 (d, J = 2.80 Hz, 1H), 7.00 (dd, J = 2.80, 11.20 Hz, 1H), 3.68 (t, J = 6.40 Hz, 2H), 3.58 (t, J = 7.20 Hz, 2H), 3.27 (s, 3H), 3.16 (s, 3H).

製備實例 61 步驟 A 向2-胺基-3-氯苯酚(4 g, 0.0280 mol)於乙醇(80 mL)中之攪拌溶液添加O-乙基二硫代碳酸酯鉀鹽(4.49 g, 0.0280 mol),然後在N2 下加熱至85℃持續12 h。根據LCMS在反應完成後,將反應混合物濃縮,藉由使用乙酸(pH = 5)使粗產物酸化且將固體過濾,用水洗滌,在真空下乾燥6 h,得到呈淡棕色固體之4-氯苯并[d]噁唑-2-硫醇(4.6 g, 89%)。 MS: 186.0 (M+H)+1 H-NMR (400 MHz, DMSO-d6 ) δ = 14.44 (bs, 1H), 7.46 (d,J = 10.80 Hz, 1H), 7.35 (d,J = 11.20 Hz, 1H), 7.23 (t,J = 10.80 Hz, 1H)。步驟 B 在0℃下向來自上文步驟A之標題化合物(4.6 g, 0.0244 mol)於DCM (90 mL)中之溶液添加草醯氯(3.20 mL, 0.0374 mol),之後添加DMF (1 mL),然後在25℃下攪拌1 h。然後在0℃下添加TEA (10 mL, 0.0734 mol)及嗎啉(2.5 mL, 0.0293 mol)且在氮下在25℃下攪拌2 h。在藉由TLC跟蹤反應完成後,添加水(40 mL)並用DCM (2×50 mL)萃取。將有機層濃縮且使用Biotage Isolera One純化系統,採用於石油醚中之10%-15%乙酸乙酯,在矽膠管柱上純化粗製物,得到呈淡黃色固體之標題化合物(3.4 g, 58%)。 MS: 239.1 (M+H)+1 H-NMR (400 MHz, DMSO-d6 ) δ = 7.40 (d,J = 10.80 Hz, 1H), 7.23 (d,J = 10.40 Hz, 1H), 7.03 (t,J = 10.80 Hz, 1H), 3.71-3.73 (m, 4H), 3.60-3.62 (m, 4H)。 Preparation Example 61 Step A. To a stirred solution of 2-amino-3-chlorophenol (4 g, 0.0280 mol) in ethanol (80 mL) was added O-ethyldithiocarbonate potassium salt (4.49 g, 0.0280 mol), then Heat to 85 °C under N for 12 h. After the reaction was completed according to LCMS, the reaction mixture was concentrated, the crude product was acidified by using acetic acid (pH = 5) and the solid was filtered, washed with water, and dried under vacuum for 6 h to obtain 4-chlorobenzene as a light brown solid. and[d]oxazole-2-thiol (4.6 g, 89%). MS: 186.0 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 14.44 (bs, 1H), 7.46 (d, J = 10.80 Hz, 1H), 7.35 (d, J = 11.20 Hz, 1H), 7.23 (t, J = 10.80 Hz, 1H). Step B To a solution of the title compound from Step A above (4.6 g, 0.0244 mol) in DCM (90 mL) was added oxalate chloride (3.20 mL, 0.0374 mol) at 0°C, followed by DMF (1 mL) , and then stirred at 25°C for 1 h. Then TEA (10 mL, 0.0734 mol) and morpholine (2.5 mL, 0.0293 mol) were added at 0 °C and stirred at 25 °C for 2 h under nitrogen. After the reaction was followed to completion by TLC, water (40 mL) was added and extracted with DCM (2×50 mL). The organic layer was concentrated and the crude material was purified on a silica column using a Biotage Isolera One purification system using 10%-15% ethyl acetate in petroleum ether to obtain the title compound as a pale yellow solid (3.4 g, 58% ). MS: 239.1 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 7.40 (d, J = 10.80 Hz, 1H), 7.23 (d, J = 10.40 Hz, 1H), 7.03 (t, J = 10.80 Hz, 1H) , 3.71-3.73 (m, 4H), 3.60-3.62 (m, 4H).

製備實例 62 步驟 A 在0℃下向NaH (0.47 g,19.8 mmol)於THF (10 mL)中之懸浮液逐滴添加1,3,4,5-四氫-2H-吡啶并[4,3-b]吲哚-2-甲酸第三丁基酯(1.8 g, 6.61 mmol) (溶解於THF中),且然後在室溫下攪拌1 h。之後,在0℃下添加碘甲烷(0.7 mL, 11.76 mmol),且然後在室溫下攪拌2 h。在反應完成後(藉由TLC監測),利用冰水將反應混合物淬滅,之後使用乙酸乙酯進行萃取。將有機層濃縮以得到粗製反應混合物5-甲基-1,3,4,5-四氫-2H-吡啶并[4,3-b]吲哚-2-甲酸第三丁基酯(1.5 g,粗製)。產物原樣用於下一步驟。 MS: 287.2 (M+H)+步驟 B 向來自上文步驟A之標題化合物(1.5 g, 5.24 mmol)於DCM (20 mL)中之溶液添加於二噁烷中之4.0 M HCl (5 mL)。將反應混合物攪拌過夜。在反應完成後,使該反應混合物蒸發以去除溶劑並用二乙醚洗滌,得到呈棕色固體之標題化合物(800 mg,粗製)。 MS: 187.1 (M+H)+ Preparation Example 62 Step A To a suspension of NaH (0.47 g, 19.8 mmol) in THF (10 mL) was added dropwise 1,3,4,5-tetrahydro-2H-pyrido[4,3-b] at 0°C. Indole-2-carboxylic acid tert-butyl ester (1.8 g, 6.61 mmol) (dissolved in THF) and then stirred at room temperature for 1 h. Afterwards, methyl iodide (0.7 mL, 11.76 mmol) was added at 0 °C, and then stirred at room temperature for 2 h. After the reaction was completed (monitored by TLC), the reaction mixture was quenched with ice water and extracted with ethyl acetate. The organic layer was concentrated to give a crude reaction mixture 5-methyl-1,3,4,5-tetrahydro-2H-pyrido[4,3-b]indole-2-carboxylic acid tert-butyl ester (1.5 g , crude). The product was used as received in the next step. MS: 287.2 (M+H) + . Step B To a solution of the title compound from Step A above (1.5 g, 5.24 mmol) in DCM (20 mL) was added 4.0 M HCl in dioxane (5 mL). The reaction mixture was stirred overnight. After the reaction was completed, the reaction mixture was evaporated to remove the solvent and washed with diethyl ether to give the title compound (800 mg, crude) as a brown solid. MS: 187.1 (M+H) + .

製備實例 63 步驟 A 向(3-甲氧基苯基)肼鹽酸鹽(10 g, 57.4 mmol)及4-側氧基六氫吡啶-1-甲酸第三丁基酯(11.3 g, 57.4 mmol)於乙醇(100 mL)中之溶液添加酒石酸。添加二甲基脲(30:70, 10 g),且在70℃下加熱16 h。將反應混合物冷卻至25℃並在真空下濃縮。將殘餘物溶解於水中且利用NaOH溶液(30%)鹼化至pH = 14並用DCM萃取。將有機相分離且經Na2 SO4 乾燥,過濾並將溶劑在減壓下蒸發,得到呈淡黃色膠狀物之7-甲氧基-2,3,4,5-四氫-1H-吡啶并[4,3-b]吲哚(2.5 g, 21%)。粗產物原樣用於下一步驟。 MS: 203.0 (M+H)+步驟 B 在25℃下向來自上文步驟A之標題化合物(600 mg, 2.9 mmol)於THF (6 mL)中之溶液添加三乙胺(0.8 mL, 5.8 mmol)及Boc酸酐(650 mg, 3 mmol),且攪拌12 h。在反應完成後(藉由TLC監測),將反應混合物在減壓下濃縮且藉由急速管柱層析使用己烷: EtOAc (80:20)純化粗產物,得到呈淡黃色固體之7-甲氧基-1,3,4,5-四氫-2H-吡啶并[4,3-b]吲哚-2-甲酸第三丁基酯(610 mg, 68%)。 MS: 303.2 (M+H)+1 H-NMR (400 MHz, DMSO-d6 ) δ = 10.69 (bs, 1H), 7.25 (d,J = 11.60 Hz, 1H), 6.80 (d,J = 2.80 Hz, 1H), 6.61 (dd,J = 2.80, 11.60 Hz, 1H), 4.48 (s, 2H), 3.81 (s, 3H), 3.67-3.68 (m, 2H), 2.73 (bs, 2H), 1.44 (s, 9H)。 MS: 187.1 (M+H)+步驟 C 在0℃下向來自上文步驟B之標題化合物(550 mg, 1.8 mmol)於THF (5 mL)中之溶液添加NaH (60%礦物油,0.14 g, 3.6 mmol),且攪拌30 min。然後添加對甲苯磺醯氯(342 mg, 1.8 mmol)並攪拌45 min。在反應完成後(藉由TLC監測),利用水將反應混合物淬滅並用EtOAc (20 mL × 3)萃取。將合併之有機萃取物用水、鹽水洗滌,且經Na2 SO4 乾燥。過濾且使溶劑在減壓下蒸發,產生呈灰白色固體之7-甲氧基-5-甲苯磺醯基-1,3,4,5-四氫-2H-吡啶并[4,3-b]吲哚-2-甲酸第三丁基酯(550 mg, 66%)。產物原樣用於下一步驟。 MS: 357.0 (M+H)+步驟 D 在0℃下向來自上文步驟C之標題化合物(550 mg, 1.204 mmol)於無水DCM (5 mL)中之溶液緩慢添加於二噁烷中之HCl (g) (2 M, 5 mL),且在25℃下攪拌12 h。在反應完成後(藉由TLC監測),使反應混合物在減壓下蒸發以產生粗產物。用二乙醚洗滌粗產物,得到呈灰白色固體之7-甲氧基-5-甲苯磺醯基-2,3,4,5-四氫-1H-吡啶并[4,3-b]吲哚(350 mg, 81%)。 MS: 357.2 (M+H)+1 H-NMR (400 MHz, DMSO-d6 ) δ = 7.73 (d,J = 8.40 Hz, 1H), 7.56 (s, 1H), 7.37 (d,J = 8.00 Hz, 2H), 7.27 (d,J = 8.40 Hz, 1H), 6.85-6.86 (m, 1H), 3.82 (s, 3H), 3.72 (s, 2H), 2.90-2.97 (m, 4H), 2.32 (s, 3H)。 Preparation Example 63 Step A: Add (3-methoxyphenyl)hydrazine hydrochloride (10 g, 57.4 mmol) and 4-pentyloxyhexahydropyridine-1-carboxylic acid tert-butyl ester (11.3 g, 57.4 mmol) in ethanol (100 mL) was added tartaric acid. Dimethylurea (30:70, 10 g) was added and heated at 70 °C for 16 h. The reaction mixture was cooled to 25°C and concentrated in vacuo. The residue was dissolved in water and basified with NaOH solution (30%) to pH = 14 and extracted with DCM. The organic phase was separated and dried over Na2SO4 , filtered and the solvent was evaporated under reduced pressure to obtain 7-methoxy-2,3,4,5-tetrahydro - 1H-pyridine as a pale yellow gum. and [4,3-b]indole (2.5 g, 21%). The crude product was used as such in the next step. MS: 203.0 (M+H) + . Step B To a solution of the title compound from Step A above (600 mg, 2.9 mmol) in THF (6 mL) was added triethylamine (0.8 mL, 5.8 mmol) and Boc anhydride (650 mg, 3 mmol) and stirred for 12 h. After the reaction was completed (monitored by TLC), the reaction mixture was concentrated under reduced pressure and the crude product was purified by flash column chromatography using Hexane:EtOAc (80:20) to obtain 7-methane as a pale yellow solid. Oxy-1,3,4,5-tetrahydro-2H-pyrido[4,3-b]indole-2-carboxylic acid tert-butyl ester (610 mg, 68%). MS: 303.2 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 10.69 (bs, 1H), 7.25 (d, J = 11.60 Hz, 1H), 6.80 (d, J = 2.80 Hz, 1H), 6.61 (dd, J = 2.80, 11.60 Hz, 1H), 4.48 (s, 2H), 3.81 (s, 3H), 3.67-3.68 (m, 2H), 2.73 (bs, 2H), 1.44 (s, 9H). MS: 187.1 (M+H) + . Step C To a solution of the title compound from Step B above (550 mg, 1.8 mmol) in THF (5 mL) was added NaH (60% mineral oil, 0.14 g, 3.6 mmol) at 0 °C and stirred for 30 min. . Then p-toluenesulfonyl chloride (342 mg, 1.8 mmol) was added and stirred for 45 min. After the reaction was complete (monitored by TLC), the reaction mixture was quenched with water and extracted with EtOAc (20 mL × 3). The combined organic extracts were washed with water, brine, and dried over Na2SO4 . Filtration and evaporation of the solvent under reduced pressure yielded 7-methoxy-5-toluenesulfonyl-1,3,4,5-tetrahydro-2H-pyrido[4,3-b] as an off-white solid. Indole-2-carboxylic acid tert-butyl ester (550 mg, 66%). The product was used as received in the next step. MS: 357.0 (M+H) + . Step D To a solution of the title compound from Step C above (550 mg, 1.204 mmol) in anhydrous DCM (5 mL) was slowly added HCl (g) in dioxane (2 M, 5 mL) at 0°C. ) and stir at 25°C for 12 h. After the reaction was complete (monitored by TLC), the reaction mixture was evaporated under reduced pressure to yield crude product. The crude product was washed with diethyl ether to obtain 7-methoxy-5-toluenesulfonyl-2,3,4,5-tetrahydro-1H-pyrido[4,3-b]indole ( 350 mg, 81%). MS: 357.2 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 7.73 (d, J = 8.40 Hz, 1H), 7.56 (s, 1H), 7.37 (d, J = 8.00 Hz, 2H), 7.27 (d, J = 8.40 Hz, 1H), 6.85-6.86 (m, 1H), 3.82 (s, 3H), 3.72 (s, 2H), 2.90-2.97 (m, 4H), 2.32 (s, 3H).

製備實例 64 步驟 A 在0℃下向8-甲基-2,3,4,5-四氫-1H-吡啶并[4,3-b]吲哚(1 g, 5.37 mmol)於DCM (20 mL)中之攪拌溶液添加TEA (2.25 mL, 16.1 mmol)及BOC酸酐(1.85 mL, 8.05 mol),然後在25℃下攪拌12 h。根據TLC在反應完成後,向反應混合物添加水,之後使用DCM進行萃取。將有機層分離並用鹽水溶液洗滌,濃縮並用己烷洗滌,得到呈灰白色固體之標題化合物(1.1 g, 71%)。 MS: 287.1 (M+H)+1 H-NMR (400 MHz, DMSO-d6 ) δ = 10.74 (s, 1H), 7.16 (d,J = 8.44 Hz, 1H), 6.85 (d,J = 8.28 Hz, 1H), 4.49 (s, 2H), 3.69 (t,J = 5.64 Hz, 2H), 2.74 (t,J = 5.28 Hz, 2H), 2.35 (s, 3H), 1.47 (s, 9H)。步驟 B 在0℃下向氫化鈉(0.125 g, 3.13 mmol)於THF (10 mL)中之懸浮液逐滴添加來自上文步驟A之標題化合物(0.6 g, 2.08 mmol) (溶解於20 mL THF中),然後在室溫下攪拌30 min。之後,在0℃下逐滴添加甲苯磺醯氯(1.19 g, 6.25 mmol) (溶解於20 mL THF中),且然後在室溫下攪拌3 h。根據TLC在反應完成後,利用冰水將反應混合物淬滅,過濾所形成之固體且用水洗滌並乾燥,得到呈白色固體之標題化合物(0.550 g, 57%)。 MS: 341.1 (M+H)+ -Boc。1 H-NMR (400 MHz, DMSO-d6 ) δ = 7.91 (d,J = 8.52 Hz, 1H), 7.73 (d,J = 8.20 Hz, 2H), 7.34 (d,J = 8.40 Hz, 2H), 7.26 (s, 1H), 7.15 (d, J = 8.56 Hz, 1H), 4.41 (s, 2H), 3.68 (t,J = 5.60 Hz, 2H), 3.06 (s, 2H), 2.29-2.30 (m, 6H), 1.43 (s, 9H),步驟 C 在0℃下向來自上文步驟B之標題化合物(0.55 g, 1.20 mmol)於DCM中之溶液添加於二噁烷中之4 M HCl (4 mL)。將反應混合物攪拌2 h。在反應完成後,使該反應混合物蒸發以去除溶劑並利用二乙醚過濾,得到呈白色固體之標題化合物(0.40 g, 85.9%)。 Preparation Example 64 Step A 8-Methyl-2,3,4,5-tetrahydro-1H-pyrido[4,3-b]indole (1 g, 5.37 mmol) in DCM (20 mL) at 0 °C TEA (2.25 mL, 16.1 mmol) and BOC anhydride (1.85 mL, 8.05 mol) were added to the stirred solution, and then stirred at 25°C for 12 h. After the reaction was completed according to TLC, water was added to the reaction mixture, followed by extraction with DCM. The organic layer was separated and washed with brine solution, concentrated and washed with hexane to give the title compound as an off-white solid (1.1 g, 71%). MS: 287.1 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 10.74 (s, 1H), 7.16 (d, J = 8.44 Hz, 1H), 6.85 (d, J = 8.28 Hz, 1H), 4.49 (s, 2H), 3.69 (t, J = 5.64 Hz, 2H), 2.74 (t, J = 5.28 Hz, 2H), 2.35 (s, 3H), 1.47 (s, 9H). Step B To a suspension of sodium hydride (0.125 g, 3.13 mmol) in THF (10 mL) was added the title compound from Step A above (0.6 g, 2.08 mmol) (dissolved in 20 mL THF) dropwise at 0°C. medium), then stir at room temperature for 30 min. Afterwards, tosyl chloride (1.19 g, 6.25 mmol) (dissolved in 20 mL THF) was added dropwise at 0 °C, and then stirred at room temperature for 3 h. After the reaction was complete according to TLC, the reaction mixture was quenched with ice water and the solid formed was filtered and washed with water and dried to give the title compound as a white solid (0.550 g, 57%). MS: 341.1 (M+H) + -Boc. 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 7.91 (d, J = 8.52 Hz, 1H), 7.73 (d, J = 8.20 Hz, 2H), 7.34 (d, J = 8.40 Hz, 2H) , 7.26 (s, 1H), 7.15 (d, J = 8.56 Hz, 1H), 4.41 (s, 2H), 3.68 (t, J = 5.60 Hz, 2H), 3.06 (s, 2H), 2.29-2.30 ( m, 6H), 1.43 (s, 9H), Step C To a solution of the title compound from Step B above (0.55 g, 1.20 mmol) in DCM was added 4 M HCl in dioxane ( 4 mL). The reaction mixture was stirred for 2 h. After the reaction was complete, the reaction mixture was evaporated to remove the solvent and filtered with diethyl ether to give the title compound as a white solid (0.40 g, 85.9%).

製備實例 65 步驟 A 在0℃下向8-氯-2,3,4,5-四氫-1H-吡啶并[4,3-b]吲哚(1 g, 4.84 mmol)於DCM (20 mL)中之攪拌溶液添加TEA (2.02 mL, 14.5 mmol)及BOC酸酐(1.67 mL, 7.26 mmol),然後在25℃下攪拌12 h。根據TLC在反應完成後,向反應混合物添加水,之後使用DCM進行萃取。將有機層分離並用鹽水溶液洗滌,濃縮得到呈白色固體之8-氯-1,3,4,5-四氫吡啶并[4,3-b]吲哚-2-甲酸第三丁基酯(1 g, 66.5%)。 MS: 305.1 (M+H)+1 H-NMR (400 MHz, DMSO-d6 ) δ = 11.12 (bs, 1H), 7.46 (d,J = 1.68 Hz, 1H), 7.30 (d,J = 8.56 Hz, 1H), 7.03 (dd,J = 1.96, 8.52 Hz, 1H), 4.51 (s, 2H), 3.70 (t, J = 5.64 Hz, 2H), 2.77 (t,J = 5.32 Hz, 2H), 1.47 (s, 9H)。步驟 B 在0℃下向冷卻至0℃之氫化鈉(0.096 g, 2.42 mmol)於THF (10 mL)中之懸浮液逐滴添加來自上文步驟A之標題化合物(0.5 g, 1.61 mol) (溶解於20 mL THF中),然後在室溫下攪拌30 min。之後,在0℃下逐滴添加甲苯磺醯氯(0.921 g, 48.3 mol) (溶解於20 mL THF中),且然後在室溫下攪拌3 h。根據TLC在反應完成後,利用冰水將反應混合物淬滅,之後使用乙酸乙酯進行萃取(100 mL)。將有機層分離,經硫酸鈉乾燥,過濾且然後濃縮。藉由矽膠管柱層析使用石油醚:乙酸乙酯(75:25)純化產物,得到呈白色固體之8-氯-5-(對甲苯基磺醯基)-3,4-二氫-1H-吡啶并[4,3-b]吲哚-2-甲酸第三丁基酯(0.450 g, 60%)。 MS: 361.1 (M+H)+ -Boc。1 H-NMR (400 MHz, DMSO-d6 ) δ = 8.05 (d,J = 8.88 Hz, 1H), 7.78 (d,J = 8.16 Hz, 2H), 7.64 (d,J = 1.96 Hz, 1H), 7.35-7.36 (m, 3H), 4.44 (s, 2H), 3.68 (t,J = 5.60 Hz, 2H), 3.08 (bs, 2H), 2.32 (s, 3H), 1.43 (s, 9H)。步驟 C 在0℃下向來自上文步驟B之標題化合物(0.45 g, 0.97 mol)於DCM中之溶液添加於二噁烷中之4 M HCl (3 mL)。將反應混合物攪拌2 h。在反應完成後,使該反應混合物蒸發以去除溶劑並利用二乙醚濃縮,得到呈白色固體之標題化合物 (0.32 g, 91%)。產物原樣用於下一步驟。 MS: 361.1 (M+H)+ Preparation Example 65 Step A. 8-Chloro-2,3,4,5-tetrahydro-1H-pyrido[4,3-b]indole (1 g, 4.84 mmol) in DCM (20 mL) at 0 °C. TEA (2.02 mL, 14.5 mmol) and BOC anhydride (1.67 mL, 7.26 mmol) were added to the stirred solution, and then stirred at 25°C for 12 h. After the reaction was completed according to TLC, water was added to the reaction mixture, followed by extraction with DCM. The organic layer was separated, washed with brine solution, and concentrated to obtain 8-chloro-1,3,4,5-tetrahydropyrido[4,3-b]indole-2-carboxylic acid tert-butyl ester ( 1 g, 66.5%). MS: 305.1 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 11.12 (bs, 1H), 7.46 (d, J = 1.68 Hz, 1H), 7.30 (d, J = 8.56 Hz, 1H), 7.03 (dd, J = 1.96, 8.52 Hz, 1H), 4.51 (s, 2H), 3.70 (t, J = 5.64 Hz, 2H), 2.77 (t, J = 5.32 Hz, 2H), 1.47 (s, 9H). Step B To a suspension of sodium hydride (0.096 g, 2.42 mmol) in THF (10 mL) cooled to 0°C was added dropwise the title compound from Step A above (0.5 g, 1.61 mol) at 0°C ( Dissolve in 20 mL THF) and stir at room temperature for 30 min. Afterwards, tosyl chloride (0.921 g, 48.3 mol) (dissolved in 20 mL THF) was added dropwise at 0 °C, and then stirred at room temperature for 3 h. After the reaction was completed according to TLC, the reaction mixture was quenched with ice water and then extracted with ethyl acetate (100 mL). The organic layer was separated, dried over sodium sulfate, filtered and then concentrated. The product was purified by silica gel column chromatography using petroleum ether:ethyl acetate (75:25) to obtain 8-chloro-5-(p-tolylsulfonyl)-3,4-dihydro-1H as a white solid. -Pyrido[4,3-b]indole-2-carboxylic acid tert-butyl ester (0.450 g, 60%). MS: 361.1 (M+H) + -Boc. 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 8.05 (d, J = 8.88 Hz, 1H), 7.78 (d, J = 8.16 Hz, 2H), 7.64 (d, J = 1.96 Hz, 1H) , 7.35-7.36 (m, 3H), 4.44 (s, 2H), 3.68 (t, J = 5.60 Hz, 2H), 3.08 (bs, 2H), 2.32 (s, 3H), 1.43 (s, 9H). Step C To a solution of the title compound from Step B above (0.45 g, 0.97 mol) in DCM was added 4 M HCl in dioxane (3 mL) at 0°C. The reaction mixture was stirred for 2 h. After the reaction was complete, the reaction mixture was evaporated to remove the solvent and concentrated with diethyl ether to afford the title compound as a white solid (0.32 g, 91%). The product was used as received in the next step. MS: 361.1 (M+H) + .

製備實例 66 步驟 A 在0℃下向5-溴-2-氯苯并[d]噁唑(1 g, 4.30 mmol)於無水DCM (10 mL)中之溶液添加嗎啉(0.56 g, 6.42 mmol)及Et3 N (1.7 mL, 12.9 mmol),且在25℃下攪拌4 h。在反應完成後(藉由TLC監測),用H2 O (10 mL)稀釋反應混合物並用DCM (10 mL × 2)萃取。將合併之有機萃取物經Na2 SO4 乾燥,過濾並在減壓下蒸發以產生粗產物。使粗產物與二乙醚(100 mL)一起研磨,過濾,用二乙醚(5 mL)洗滌並乾燥,得到呈灰白色固體之標題化合物(0.85 g, 71%)。1 H-NMR (400 MHz, DMSO-d6 ) δ = 7.48 (d,J = 2.40 Hz, 1H), 7.34-7.38 (m, 1H), 7.16-7.17 (m, 1H), 3.70-3.72 (m, 4H), 3.58-3.59 (m, 4H)。 Preparation Example 66 Step A To a solution of 5-bromo-2-chlorobenzo[d]oxazole (1 g, 4.30 mmol) in anhydrous DCM (10 mL) at 0°C was added morpholine (0.56 g, 6.42 mmol) and Et 3 N (1.7 mL, 12.9 mmol) and stirred at 25°C for 4 h. After the reaction was complete (monitored by TLC), the reaction mixture was diluted with H2O (10 mL) and extracted with DCM (10 mL × 2). The combined organic extracts were dried over Na2SO4 , filtered and evaporated under reduced pressure to give crude product. The crude product was triturated with diethyl ether (100 mL), filtered, washed with diethyl ether (5 mL) and dried to give the title compound as an off-white solid (0.85 g, 71%). 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 7.48 (d, J = 2.40 Hz, 1H), 7.34-7.38 (m, 1H), 7.16-7.17 (m, 1H), 3.70-3.72 (m , 4H), 3.58-3.59 (m, 4H).

製備實例 67 步驟 A 向4,6-二氯吡啶-3-胺(2.5 g, 15.3 mmol)於THF (50 mL)中之攪拌溶液逐滴添加於THF中之三光氣(4.55 g, 15.3 mol),之後添加TEA (4.28 mL, 30.7 mol)且加熱至回流2 h。將反應混合物在真空下濃縮。將殘餘物溶解於乙腈(50 mL)及甲苯(50 mL)中,且添加嗎啉(1.34 g, 15.3 mmol)並加熱至110℃持續12 h。之後,檢查TLC,將粗製物濃縮並藉由矽膠管柱層析使用石油醚:乙酸乙酯(20:80)純化,得到呈白色固體之N-(4,6-二氯-3-吡啶基)嗎啉-4-甲醯胺(3.0 g, 70.1%)。 MS: 276.0 (M+H)+1 H-NMR (400 MHz, DMSO-d6 ) δ = 8.55 (s, 1H), 8.38-8.40 (m, 1H), 7.78-7.79 (m, 1H), 3.57-3.58 (m, 4H), 3.40-3.42 (m, 4H)。步驟 B 向來自上文步驟A之標題化合物(3.0 g, 10.8 mmol)於1,4-二噁烷(5 mL)中之溶液添加Cs2 CO3 (10.5 g, 32.4 mol)、1,10-菲咯啉(0.972 g, 5.40 mol)及碘化銅(1.03 g, 5.40 mol),然後加熱至120℃持續12 h。經由矽藻土過濾反應混合物並用DCM/MeOH洗滌,濃縮且使用Biotage Isolera One純化系統,採用EtOAc/己烷梯度(40/60),在矽膠管柱上純化粗製物,得到呈灰白色固體之標題化合物(0.150 g,粗製)。粗產物原樣用於下一步驟。 MS: 240.1 (M+H)+ Preparation Example 67 Step A : To a stirred solution of 4,6-dichloropyridin-3-amine (2.5 g, 15.3 mmol) in THF (50 mL) was added triphosgene (4.55 g, 15.3 mol) in THF dropwise, followed by TEA (4.28 mL, 30.7 mol) and heated to reflux for 2 h. The reaction mixture was concentrated in vacuo. The residue was dissolved in acetonitrile (50 mL) and toluene (50 mL), and morpholine (1.34 g, 15.3 mmol) was added and heated to 110 °C for 12 h. Afterwards, TLC was checked, the crude material was concentrated and purified by silica column chromatography using petroleum ether:ethyl acetate (20:80) to obtain N-(4,6-dichloro-3-pyridyl) as a white solid ) Morpholine-4-methamide (3.0 g, 70.1%). MS: 276.0 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 8.55 (s, 1H), 8.38-8.40 (m, 1H), 7.78-7.79 (m, 1H), 3.57-3.58 (m, 4H), 3.40 -3.42 (m, 4H). Step B To a solution of the title compound from Step A above (3.0 g, 10.8 mmol) in 1,4-dioxane (5 mL) was added Cs 2 CO 3 (10.5 g, 32.4 mol), 1,10- phenanthroline (0.972 g, 5.40 mol) and copper iodide (1.03 g, 5.40 mol), and then heated to 120°C for 12 h. The reaction mixture was filtered through celite and washed with DCM/MeOH, concentrated and the crude material was purified on a silica column using a Biotage Isolera One purification system with an EtOAc/hexane gradient (40/60) to give the title compound as an off-white solid (0.150 g, crude). The crude product was used as such in the next step. MS: 240.1 (M+H) + .

製備實例 68 步驟 A 在0℃下向8-乙氧基-2,3,4,5-四氫-1H-吡啶并[4,3-b]吲哚(1 g, 4.62 mmol)於DCM (20 mL)中之攪拌溶液添加TEA (1.97 mL, 13.87 mmol)及BOC酸酐(1.5 mL, 6.93 mol),然後在25℃下攪拌12 h。根據TLC在反應完成後,向反應混合物添加水,之後使用DCM進行萃取。將有機層分離並用鹽水溶液洗滌,濃縮並用己烷洗滌,獲得呈棕色固體之8-乙氧基-1,3,4,5-四氫-2H-吡啶并[4,3-b]吲哚-2-甲酸第三丁基酯(0.8 g, 54%)。 MS: 317.2 (M+H)+1 H-NMR (400 MHz, DMSO-d6 ) δ = 10.69 (bs, 1H), 7.16 (d,J = 8.68 Hz, 1H), 6.87 (s, 1H), 6.66 (t,J = 6.76 Hz, 1H), 4.48 (s, 1H), 3.97-3.99 (m, 2H), 3.69 (t,J = 5.48 Hz, 2H), 2.74 (bs, 2H), 1.44 (s, 9H), 1.23 (t,J = 6.84 Hz, 3H)。步驟 B 在0℃下向氫化鈉(0.136 g, 2.84 mmol)於THF (5 mL)中之懸浮液逐滴添加來自上文步驟A之標題化合物(0.3 g, 0.95 mmol) (溶解於10 mL THF中),然後在室溫下攪拌30 min。之後,在0℃下逐滴添加甲苯磺醯氯(0.27 g, 1.42 mmol) (溶解於10 mL THF中),且然後在室溫下攪拌3 h。根據TLC在反應完成後,利用冰水將反應混合物淬滅,過濾所形成之固體且用水洗滌並乾燥,得到呈白色固體之8-乙氧基-5-甲苯磺醯基-1,3,4,5-四氫-2H-吡啶并[4,3-b]吲哚-2-甲酸第三丁基酯(0.32 g, 72%)。產物原樣用於下一步驟。 MS: 371.2 (M+H)+ -Boc。步驟 C 在0℃下向來自上文步驟B之標題化合物(0.32 g, 0.68 mmol)於DCM中之溶液添加於二噁烷中之4 M HCl (4 mL)。將反應混合物攪拌2 h。在反應完成後,使該反應混合物蒸發以去除溶劑並利用二乙醚過濾,得到呈棕色固體之標題化合物(0.13 g, 56% )。產物原樣用於下一步驟。 MS: 371.2 (M+H)+ Preparation Example 68 Step A 8-Ethoxy-2,3,4,5-tetrahydro-1H-pyrido[4,3-b]indole (1 g, 4.62 mmol) in DCM (20 mL) at 0 °C TEA (1.97 mL, 13.87 mmol) and BOC anhydride (1.5 mL, 6.93 mol) were added to the stirring solution, and then stirred at 25°C for 12 h. After the reaction was completed according to TLC, water was added to the reaction mixture, followed by extraction with DCM. The organic layer was separated and washed with brine solution, concentrated and washed with hexane to obtain 8-ethoxy-1,3,4,5-tetrahydro-2H-pyrido[4,3-b]indole as a brown solid -Tert-Butyl 2-carboxylate (0.8 g, 54%). MS: 317.2 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 10.69 (bs, 1H), 7.16 (d, J = 8.68 Hz, 1H), 6.87 (s, 1H), 6.66 (t, J = 6.76 Hz, 1H), 4.48 (s, 1H), 3.97-3.99 (m, 2H), 3.69 (t, J = 5.48 Hz, 2H), 2.74 (bs, 2H), 1.44 (s, 9H), 1.23 (t, J = 6.84 Hz, 3H). Step B To a suspension of sodium hydride (0.136 g, 2.84 mmol) in THF (5 mL) was added the title compound from Step A above (0.3 g, 0.95 mmol) (dissolved in 10 mL THF) dropwise at 0°C. medium), then stir at room temperature for 30 min. Afterwards, tosyl chloride (0.27 g, 1.42 mmol) (dissolved in 10 mL THF) was added dropwise at 0 °C, and then stirred at room temperature for 3 h. After the reaction is completed according to TLC, the reaction mixture is quenched with ice water, the solid formed is filtered, washed with water and dried to obtain 8-ethoxy-5-toluenesulfonyl-1,3,4 as a white solid , tert-butyl 5-tetrahydro-2H-pyrido[4,3-b]indole-2-carboxylate (0.32 g, 72%). The product was used as received in the next step. MS: 371.2 (M+H) + -Boc. Step C To a solution of the title compound from Step B above (0.32 g, 0.68 mmol) in DCM was added 4 M HCl in dioxane (4 mL) at 0°C. The reaction mixture was stirred for 2 h. After the reaction was completed, the reaction mixture was evaporated to remove the solvent and filtered with diethyl ether to give the title compound as a brown solid (0.13 g, 56%). The product was used as received in the next step. MS: 371.2 (M+H) + .

製備實例 69 步驟 A 向4-側氧基六氫吡啶-1-甲酸第三丁基酯(10.00 g, 0.0502 mol)於乙醇(100 mL)中之攪拌溶液添加羥胺鹽酸鹽(6.98 g, 0.100 mol)及CH3 COONa (8.23 g, 0.100 mol),然後在氮氣氛下加熱至90℃持續12 h。根據LCMS在反應完成後,將反應混合物濃縮,且向粗製材料添加水(100 mL),之後藉由使用二氯甲烷(250 mL)進行萃取。將有機層濃縮且藉由矽膠管柱(Biotage)使用於石油醚中之18%-30%乙酸乙酯純化粗產物,獲得呈白色固體之4-(羥基亞胺基)六氫吡啶-1-甲酸第三丁基酯(5 g, 46.4%)。 MS: 159.1 (M+H)+ -第三丁基1 H-NMR (400 MHz, DMSO-d6 ) δ = 10.45 (s, 1H), 3.33-3.36 (m, 4H), 2.42-2.44 (m, 2H), 2.20-2.22 (m, 2H), 1.41 (s, 9H)。步驟 B 在0℃下向氫化鈉(0.268 g, 7.00 mmol)於DMF (3 mL)中之懸浮液逐滴添加來自上文步驟A之標題化合物(0.500 g, 2.33 mmol) (溶解於5 mL DMF中),然後在室溫下攪拌60 min。之後,在0℃下逐滴添加2-氟吡啶(0.340 g, 3.50 mmol) (溶解於2 mL DMF中),且然後在室溫下攪拌3 h。根據TLC在反應完成後,利用冰水將反應混合物淬滅,之後使用乙酸乙酯(30 mL)進行萃取。將有機層分離,經硫酸鈉乾燥,過濾且然後濃縮,獲得呈淡棕色固體之4-側氧基-3-(2-側氧基-1,2-二氫吡啶-3-基)六氫吡啶-1-甲酸第三丁基酯(300 mg,粗製)。粗產物原樣用於下一步驟。 MS: 293.2 (M+H)+步驟 C 將來自上文步驟B之標題化合物(0.5 g, 1.71 Mmol)於濃H2 SO4 (2.0 mL)中之懸浮液在氮氣氛下在室溫下攪拌16 h。之後檢查LCMS,其僅指示起始材料,然後在氮氣氛下將反應混合物加熱至60℃持續16 h。之後檢查LCMS,其指示80%之產物質量。將反應混合物冷卻至室溫,向此添加於水中之10%乙腈(20.0 mL),且藉由使用固體K2 CO3 使反應混合物鹼化,然後將固體過濾。將濾液濃縮,得到呈棕色黏性油狀物之標題化合物(250 mg,粗製)。產物原樣用於下一步驟。 MS: 175.1 (M+H)+步驟 D 在0℃下向來自上文步驟C之標題化合物(0.6 g, 3.15 mmol)於四氫呋喃(10.0 mL)中之攪拌溶液添加TEA (1.32 mL, 9.46 m0mol)及BOC酸酐(0.869 mL, 3.78 mmol),然後在氮氣氛下在25℃下攪拌12 h。根據TLC在反應完成後,將反應混合物濃縮且藉由矽膠管柱(Biotage)使用於石油醚中之15%-20%乙酸乙酯純化粗產物,得到呈灰白色固體之-7,8-二氫呋喃并[2,3-b:4,5-c']二吡啶-6(5H)-甲酸第三丁基酯(500 mg, 53%)。 MS: 275.2 (M+H)+1 H-NMR (400 MHz, DMSO-d6 ) δ = 8.21-8.22 (m, 1H), 8.05-8.06 (m, 1H), 7.30-7.31 (m, 1H), 4.51 (s, 2H), 3.76 (t,J = 5.60 Hz, 2H), 2.86 (t,J = 5.60 Hz, 2H), 1.44 (s, 9H)。步驟 E 在0℃下向來自上文步驟D之標題化合物(0.5 g, 1.67 mmol)於二氯甲烷(5 mL)中之攪拌溶液添加於二噁烷中之4.0 M HCl (2 mL),然後在0℃-20℃下攪拌2 h。根據TLC及LCMS在反應完成後,將反應混合物濃縮,得到呈灰白色固體之標題化合物(350 mg,定量)。 MS: 175.1 (M+H)+ Preparation Example 69 Step A : To a stirred solution of tert-butyl 4-pentanoxyhexahydropyridine-1-carboxylate (10.00 g, 0.0502 mol) in ethanol (100 mL) was added hydroxylamine hydrochloride (6.98 g, 0.100 mol) and CH 3 COONa (8.23 g, 0.100 mol) and then heated to 90 °C under nitrogen atmosphere for 12 h. After the reaction was completed according to LCMS, the reaction mixture was concentrated and water (100 mL) was added to the crude material, followed by extraction with dichloromethane (250 mL). The organic layer was concentrated and the crude product was purified through a silica gel column (Biotage) using 18%-30% ethyl acetate in petroleum ether to obtain 4-(hydroxyimino)hexahydropyridine-1- as a white solid. tert-Butyl formate (5 g, 46.4%). MS: 159.1 (M+H) + -tert-butyl 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 10.45 (s, 1H), 3.33-3.36 (m, 4H), 2.42-2.44 (m , 2H), 2.20-2.22 (m, 2H), 1.41 (s, 9H). Step B To a suspension of sodium hydride (0.268 g, 7.00 mmol) in DMF (3 mL) was added the title compound from Step A above (0.500 g, 2.33 mmol) (dissolved in 5 mL DMF) dropwise at 0°C. medium), then stir at room temperature for 60 min. Afterwards, 2-fluoropyridine (0.340 g, 3.50 mmol) (dissolved in 2 mL DMF) was added dropwise at 0 °C, and then stirred at room temperature for 3 h. After the reaction was completed according to TLC, the reaction mixture was quenched with ice water, and then extracted with ethyl acetate (30 mL). The organic layer was separated, dried over sodium sulfate, filtered and then concentrated to obtain 4-Pendantoxy-3-(2-Pendantoxy-1,2-dihydropyridin-3-yl)hexahydrogen as a light brown solid tert-Butylpyridine-1-carboxylate (300 mg, crude). The crude product was used as such in the next step. MS: 293.2 (M+H) + . Step C A suspension of the title compound from Step B above (0.5 g, 1.71 Mmol) in concentrated H2SO4 (2.0 mL) was stirred at room temperature under a nitrogen atmosphere for 16 h. The LCMS was checked afterwards, which indicated only starting material, and the reaction mixture was heated to 60 °C under nitrogen atmosphere for 16 h. The LCMS was then checked and indicated 80% product quality. The reaction mixture was cooled to room temperature, 10% acetonitrile in water (20.0 mL) was added, and the reaction mixture was basified by using solid K2CO3 , and the solid was filtered. The filtrate was concentrated to give the title compound (250 mg, crude) as a brown viscous oil. The product was used as received in the next step. MS: 175.1 (M+H) + . Step D To a stirred solution of the title compound from Step C above (0.6 g, 3.15 mmol) in tetrahydrofuran (10.0 mL) was added TEA (1.32 mL, 9.46 mol) and BOC anhydride (0.869 mL, 3.78 mmol) at 0°C. ) and then stirred at 25°C for 12 h under nitrogen atmosphere. After the reaction was completed according to TLC, the reaction mixture was concentrated and the crude product was purified through a silica gel column (Biotage) using 15%-20% ethyl acetate in petroleum ether to obtain -7,8-dihydrogen as an off-white solid. Furo[2,3-b:4,5-c']bipyridine-6(5H)-carboxylic acid tert-butyl ester (500 mg, 53%). MS: 275.2 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 8.21-8.22 (m, 1H), 8.05-8.06 (m, 1H), 7.30-7.31 (m, 1H), 4.51 (s, 2H), 3.76 (t, J = 5.60 Hz, 2H), 2.86 (t, J = 5.60 Hz, 2H), 1.44 (s, 9H). Step E To a stirred solution of the title compound from Step D above (0.5 g, 1.67 mmol) in dichloromethane (5 mL) was added 4.0 M HCl in dioxane (2 mL) at 0°C and then Stir at 0°C-20°C for 2 h. After the reaction was completed according to TLC and LCMS, the reaction mixture was concentrated to obtain the title compound (350 mg, quantitative) as an off-white solid. MS: 175.1 (M+H) + .

製備實例 70 步驟 A 向2,5-二氯-1,3-苯并噁唑(0.25 g, 0.0013 mol)於乙腈(15 mL)中之攪拌溶液添加K2 CO3 (0.55 g, 0.0040 mol)及(2S,6R)-2,6-二甲基嗎啉(0.17 g, 0.0014 mol)。之後將反應混合物加熱至70℃持續12 h。 根據TLC在反應完成後,向反應混合物添加DCM及水(50 mL)。將有機層分離,經硫酸鈉乾燥,過濾且然後濃縮至呈白色固體之標題化合物(0.2 g, 56%)。 MS: 267.0 (M+H)+1 H-NMR (400 MHz, DMSO-d6 ) δ = 7.43 (d,J = 8.44 Hz, 1H), 7.34 (d,J = 1.96 Hz, 1H), 7.04-7.04 (m, 1H), 3.99 (d,J = 13.16 Hz, 2H), 3.66-3.67 (m, 2H), 2.82 (t,J = 10.84 Hz, 2H), 1.00 (d,J = 6.28 Hz, 6H)。 Preparation Example 70 Step A To a stirred solution of 2,5-dichloro-1,3-benzoxazole (0.25 g, 0.0013 mol) in acetonitrile (15 mL) was added K 2 CO 3 (0.55 g, 0.0040 mol) and (2S ,6R)-2,6-dimethylmorpholine (0.17 g, 0.0014 mol). The reaction mixture was then heated to 70 °C for 12 h. After the reaction was complete according to TLC, DCM and water (50 mL) were added to the reaction mixture. The organic layer was separated, dried over sodium sulfate, filtered and then concentrated to the title compound as a white solid (0.2 g, 56%). MS: 267.0 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 7.43 (d, J = 8.44 Hz, 1H), 7.34 (d, J = 1.96 Hz, 1H), 7.04-7.04 (m, 1H), 3.99 ( d, J = 13.16 Hz, 2H), 3.66-3.67 (m, 2H), 2.82 (t, J = 10.84 Hz, 2H), 1.00 (d, J = 6.28 Hz, 6H).

製備實例 71 步驟 A 向2,5-二氯-1,3-苯并噁唑(0.6 g, 0.0032 mol)於乙腈(15 mL)中之攪拌溶液添加K2 CO3 (1.32 g, 0.0096 mol)及第三丁基胺(0.255 g, 0.0035 mol)。之後將反應混合物加熱至70℃持續12 h。 根據TLC在反應完成後,向反應混合物添加DCM及水(50 mL)。將有機層分離,經硫酸鈉乾燥,過濾且然後濃縮,得到呈棕色固體之標題化合物(0.4 g, 55%)。 MS: 225.0 (M+H)+1 H-NMR (400 MHz, DMSO-d6 ) δ = 7.97 (s, 1H), 7.31-7.32 (m, 2H), 6.96-6.97 (m, 1H), 1.40 (s, 9H)。 Preparation Example 71 Step A. To a stirred solution of 2,5-dichloro-1,3-benzoxazole (0.6 g, 0.0032 mol) in acetonitrile (15 mL) was added K 2 CO 3 (1.32 g, 0.0096 mol) and a third Butylamine (0.255 g, 0.0035 mol). The reaction mixture was then heated to 70 °C for 12 h. After the reaction was complete according to TLC, DCM and water (50 mL) were added to the reaction mixture. The organic layer was separated, dried over sodium sulfate, filtered and then concentrated to give the title compound as a brown solid (0.4 g, 55%). MS: 225.0 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 7.97 (s, 1H), 7.31-7.32 (m, 2H), 6.96-6.97 (m, 1H), 1.40 (s, 9H).

製備實例 72 步驟 A 向6-溴-2-氯喹唑啉(0.3 g, 1.234 mmol)於乙腈(10 mL)中之攪拌溶液添加K2 CO3 (0.34 g, 2.467 mmol)及嗎啉(0.17 g, 1.85 mmol)。之後將反應混合物加熱至70℃持續12 h。 根據TLC在反應完成後,向反應混合物添加DCM及水(50 mL)。將有機層分離,經硫酸鈉乾燥,過濾且然後濃縮,得到呈淡黃色固體之標題化合物(0.25 g, 69%)。產物原樣用於下一步驟。 MS: 294.0 (M+H)+ Preparation Example 72 Step A. To a stirred solution of 6-bromo-2-chloroquinazoline (0.3 g, 1.234 mmol) in acetonitrile (10 mL) was added K 2 CO 3 (0.34 g, 2.467 mmol) and morpholine (0.17 g, 1.85 mmol). ). The reaction mixture was then heated to 70 °C for 12 h. After the reaction was complete according to TLC, DCM and water (50 mL) were added to the reaction mixture. The organic layer was separated, dried over sodium sulfate, filtered and then concentrated to give the title compound as a pale yellow solid (0.25 g, 69%). The product was used as received in the next step. MS: 294.0 (M+H) + .

製備實例 73 步驟 A 向2,6-二氯-1,5-萘啶(100 mg, 0.502 mmol)於二噁烷(3 mL)中之攪拌溶液添加三乙胺(0.210 ml, 1.507 mmol)及嗎啉(0.052 ml, 0.603 mmol)。然後在110℃下攪拌反應混合物。4 h後,反應未完成,因此添加三乙胺(0.210 ml, 1.507 mmol),且將反應混合物進一步在110℃下攪拌過夜。將反應混合物濃縮至乾燥且然後溶解於二氯甲烷中,並用飽和NH4 Cl水溶液洗滌。用二氯甲烷將水層萃取兩次。使合併之有機層經Na2 SO4 乾燥,過濾並在減壓下蒸發,得到呈米色固體之標題產物(93 mg, 74%)。 MS: 250.02 (M+H)+1 H-NMR (400 MHz, DMSO-d6 ) δ = 8.05 (d,J = 9.4 Hz, 1H), 7.98 (d,J = 8.8 Hz, 1H), 7.59 (d,J = 8.7 Hz, 1H), 7.53 (d,J = 9.5 Hz, 1H), 3.71 (hept,J = 3.3, 2.6 Hz, 8H)。 Preparation Example 73 Step A. To a stirred solution of 2,6-dichloro-1,5-naphthyridine (100 mg, 0.502 mmol) in dioxane (3 mL) was added triethylamine (0.210 ml, 1.507 mmol) and morpholine ( 0.052 ml, 0.603 mmol). The reaction mixture was then stirred at 110°C. After 4 h, the reaction was not complete, so triethylamine (0.210 ml, 1.507 mmol) was added and the reaction mixture was further stirred at 110 °C overnight. The reaction mixture was concentrated to dryness and then dissolved in dichloromethane and washed with saturated aqueous NH4Cl solution. The aqueous layer was extracted twice with dichloromethane. The combined organic layers were dried over Na2SO4 , filtered and evaporated under reduced pressure to give the title product as a beige solid (93 mg, 74%). MS: 250.02 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 8.05 (d, J = 9.4 Hz, 1H), 7.98 (d, J = 8.8 Hz, 1H), 7.59 (d, J = 8.7 Hz, 1H) , 7.53 (d, J = 9.5 Hz, 1H), 3.71 (hept, J = 3.3, 2.6 Hz, 8H).

製備實例 74 步驟 A 將2-溴-5-氯吡啶(200 mg, 1.039 mmol)溶解於乙腈(2.5 mL)中,向此添加(1S,4S)-2-氧雜-5-氮雜二環[2.2.1]庚烷鹽酸鹽(211 mg, 1.559 mmol)及三乙胺(0.362 mL, 2.60 mmol),且將懸浮液於微波中輻照至160℃持續1h20。然後將樣品在水(20 mL)與二氯甲烷(20 mL)之間進行萃取。將水層用二氯甲烷洗滌兩次。使合併之有機層經Na2 SO4 乾燥,過濾並在減壓下濃縮。使用Biotage Isolera One純化系統利用二氯甲烷/甲醇梯度(100/0 -> 90/10)在矽膠管柱上純化粗製物,得到呈米色固體之產物(57 mg, 26%)。 MS: 211.03 (M+H)+1 H-NMR (400 MHz, DMSO-d6 ) δ = 8.06 (d,J = 2.6 Hz, 1H), 7.56 (dd,J = 9.0, 2.7 Hz, 1H), 6.57 (d, J = 9.0 Hz, 1H), 4.80 (d,J = 2.3 Hz, 1H), 4.65 (d,J = 2.4 Hz, 1H), 3.76 (dd,J = 7.3, 1.5 Hz, 1H), 3.61 (d,J = 7.3 Hz, 1H), 3.43 (dd,J = 10.1, 1.5 Hz, 1H), 3.20 (d,J = 10.3 Hz, 1H), 1.90 (dd,J = 9.7, 2.3 Hz, 1H), 1.87 - 1.81 (m, 1H)。 Preparation Example 74 Step A Dissolve 2-bromo-5-chloropyridine (200 mg, 1.039 mmol) in acetonitrile (2.5 mL), and add (1S,4S)-2-oxa-5-azabicyclo [2.2. 1] Heptane hydrochloride (211 mg, 1.559 mmol) and triethylamine (0.362 mL, 2.60 mmol), and the suspension was irradiated in microwave to 160°C for 1h20. The sample was then extracted between water (20 mL) and dichloromethane (20 mL). The aqueous layer was washed twice with dichloromethane. The combined organic layers were dried over Na2SO4 , filtered and concentrated under reduced pressure. The crude product was purified on a silica column using a Biotage Isolera One purification system using a methylene chloride/methanol gradient (100/0 -> 90/10) to obtain the product as a beige solid (57 mg, 26%). MS: 211.03 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 8.06 (d, J = 2.6 Hz, 1H), 7.56 (dd, J = 9.0, 2.7 Hz, 1H), 6.57 (d, J = 9.0 Hz, 1H), 4.80 (d, J = 2.3 Hz, 1H), 4.65 (d, J = 2.4 Hz, 1H), 3.76 (dd, J = 7.3, 1.5 Hz, 1H), 3.61 (d, J = 7.3 Hz, 1H), 3.43 (dd, J = 10.1, 1.5 Hz, 1H), 3.20 (d, J = 10.3 Hz, 1H), 1.90 (dd, J = 9.7, 2.3 Hz, 1H), 1.87 - 1.81 (m, 1H ).

製備實例 75 步驟 A 將2-溴-5-氯吡啶(200 mg, 1.039 mmol)溶解於乙腈(2.5 mL)中,向此添加(1R,4R)-2-氧雜-5-氮雜二環[2.2.1]庚烷鹽酸鹽(211 mg, 1.559 mmol)及三乙胺(0.362 mL, 2.60 mmol),且將懸浮液於微波中輻照至160℃持續1h20。然後將樣品在水(20 mL)與二氯甲烷(20 mL)之間進行萃取。將水層用二氯甲烷洗滌兩次。使合併之有機層經Na2 SO4 乾燥,過濾並在減壓下濃縮。使用Biotage Isolera One純化系統利用二氯甲烷/甲醇梯度(100/0 -> 90/10)在矽膠管柱上純化粗製物,得到呈米色固體之產物(51 mg, 23%)。 MS: 211.04 (M+H)+1 H-NMR (400 MHz, DMSO-d6 ) δ = 8.06 (dd,J = 2.7, 0.7 Hz, 1H), 7.56 (dd,J = 9.0, 2.7 Hz, 1H), 6.63 - 6.50 (m, 1H), 4.80 (s, 1H), 4.64 (s, 1H), 3.75 (dd,J = 7.3, 1.5 Hz, 1H), 3.61 (d,J = 7.3, 0.9 Hz, 1H), 3.43 (dd,J = 10.1, 1.6 Hz, 1H), 3.20 (d,J = 10.1, 1.1 Hz, 1H), 1.94 - 1.78 (m, 2H)。 Preparation Example 75 Step A Dissolve 2-bromo-5-chloropyridine (200 mg, 1.039 mmol) in acetonitrile (2.5 mL), and add (1R,4R)-2-oxa-5-azabicyclo [2.2. 1] Heptane hydrochloride (211 mg, 1.559 mmol) and triethylamine (0.362 mL, 2.60 mmol), and the suspension was irradiated in microwave to 160°C for 1h20. The sample was then extracted between water (20 mL) and dichloromethane (20 mL). The aqueous layer was washed twice with dichloromethane. The combined organic layers were dried over Na2SO4 , filtered and concentrated under reduced pressure. The crude product was purified on a silica column using a Biotage Isolera One purification system using a methylene chloride/methanol gradient (100/0 -> 90/10) to obtain the product as a beige solid (51 mg, 23%). MS: 211.04 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 8.06 (dd, J = 2.7, 0.7 Hz, 1H), 7.56 (dd, J = 9.0, 2.7 Hz, 1H), 6.63 - 6.50 (m, 1H ), 4.80 (s, 1H), 4.64 (s, 1H), 3.75 (dd, J = 7.3, 1.5 Hz, 1H), 3.61 (d, J = 7.3, 0.9 Hz, 1H), 3.43 (dd, J = 10.1, 1.6 Hz, 1H), 3.20 (d, J = 10.1, 1.1 Hz, 1H), 1.94 - 1.78 (m, 2H).

實例 1 步驟 A 向經脫氣之四氫呋喃(5 mL)添加氯-(2-二環己基膦基-2′,6′-二異丙氧基-1,1′-聯苯)[2-(2-胺基乙基)苯基]鈀(II)-甲基-第三丁基醚加成物(PdRuPhos G1) (0.017 g, 0.024 mmol)、2-二環己基膦基-2′,6′-二異丙氧基聯苯(RuPho) (0.011 g, 0.024 mmol)、來自製備實例2 之標題化合物(0.05 g, 0.024 mmol)及市售4-(6-溴苯并[d]噻唑-2-基)嗎啉(0.073 g, 0.029 mmol)。然後,添加雙(三甲基矽基)胺基鋰(LiHMDS)於四氫呋喃(1 mL, 1 mmol)中之1 M溶液。將所得反應混合物在回流下加熱2小時。將反應混合物冷卻至室溫,溶解於二氯甲烷(100 mL)中。將有機相用水及鹽水洗滌且經Na2 SO4 乾燥。將溶劑在減壓下去除。使用Biotage Isolera One純化系統,採用乙酸乙酯/正庚烷梯度(80/20 => 100/0),在矽膠管柱上純化粗產物,得到標題化合物(0.070 g, 69%)。1 H-NMR (400 MHz,氯仿-d ) δ = 7.51 (d,J = 8.8 Hz, 1H), 7.31 (d,J = 2.4 Hz, 1H), 7.27 (s, 1H), 7.20 (dd,J = 8.8, 4.2 Hz, 1H), 7.14 (td,J = 8.6, 2.4 Hz, 2H), 6.94 (td,J = 9.1, 2.5 Hz, 1H), 4.40 (s, 2H), 3.88 - 3.81 (m, 4H), 3.70 (t,J = 5.7 Hz, 2H), 3.64 (s, 3H), 3.59 (t,J = 4.9 Hz, 4H), 2.92 (t,J = 5.7 Hz, 2H)。 Example 1 Step A To degassed tetrahydrofuran (5 mL) was added chloro-(2-dicyclohexylphosphino-2′,6′-diisopropoxy-1,1′-biphenyl)[2-(2- Aminoethyl)phenyl]palladium(II)-methyl-tert-butyl ether adduct (PdRuPhos G1) (0.017 g, 0.024 mmol), 2-dicyclohexylphosphino-2′,6′- Diisopropoxybiphenyl (RuPho) (0.011 g, 0.024 mmol), the title compound from Preparative Example 2 (0.05 g, 0.024 mmol) and commercially available 4-(6-bromobenzo[d]thiazole-2- base) morpholine (0.073 g, 0.029 mmol). Then, a 1 M solution of lithium bis(trimethylsilyl)amide (LiHMDS) in tetrahydrofuran (1 mL, 1 mmol) was added. The resulting reaction mixture was heated at reflux for 2 hours. The reaction mixture was cooled to room temperature and dissolved in dichloromethane (100 mL). The organic phase was washed with water and brine and dried over Na2SO4 . The solvent was removed under reduced pressure. The crude product was purified on a silica column using a Biotage Isolera One purification system using an ethyl acetate/n-heptane gradient (80/20 => 100/0) to obtain the title compound (0.070 g, 69%). 1 H-NMR (400 MHz, chloroform- d ) δ = 7.51 (d, J = 8.8 Hz, 1H), 7.31 (d, J = 2.4 Hz, 1H), 7.27 (s, 1H), 7.20 (dd, J = 8.8, 4.2 Hz, 1H), 7.14 (td, J = 8.6, 2.4 Hz, 2H), 6.94 (td, J = 9.1, 2.5 Hz, 1H), 4.40 (s, 2H), 3.88 - 3.81 (m, 4H), 3.70 (t, J = 5.7 Hz, 2H), 3.64 (s, 3H), 3.59 (t, J = 4.9 Hz, 4H), 2.92 (t, J = 5.7 Hz, 2H).

實例 2 步驟 A 向製備實例1之標題化合物(0.150 g, 1 eq)於無水1,4-二噁烷(5 mL)中之攪拌溶液添加市售4-(6-溴苯并[d]噻唑-2-基)嗎啉(1 eq)、第三丁醇鈉(3 eq),且將混合物在N2 氣氛下脫氣10分鐘。向此反應混合物添加Pd2 (dba)3 (0.05 eq)及Ru-Phos (0.1 eq),且將混合物加熱至100℃直至反應完成為止。在反應完成後,經由矽藻土床過濾該反應混合物,並用EtOAc洗滌。將濾液濃縮且藉由管柱層析或製備型HPLC純化粗產物,得到如表2中所指示之標題化合物6 Example 2 Step A To a stirred solution of the title compound of Preparative Example 1 (0.150 g, 1 eq) in anhydrous 1,4-dioxane (5 mL) was added commercially available 4-(6-bromobenzo[d]thiazole-2 -ethyl)morpholine (1 eq), sodium tert-butoxide (3 eq), and the mixture was degassed under N2 atmosphere for 10 min. To this reaction mixture were added Pd 2 (dba) 3 (0.05 eq) and Ru-Phos (0.1 eq), and the mixture was heated to 100°C until the reaction was complete. After the reaction was complete, the reaction mixture was filtered through a bed of celite and washed with EtOAc. The filtrate was concentrated and the crude product was purified by column chromatography or preparative HPLC to afford the title compound 6 as indicated in Table 2.

實例 3 96e 除使用下表中所指示之三環胺基衍生物及溴/氯衍生物以外,遵循如實例12 中所闡述之鈀偶合程序,製備以下化合物。實例7172 係分別遵循如製備實例4243 中所闡述之程序,之後實例97 中所闡述之去保護程序來製備。 2 Examples 3 to 96e The following compounds were prepared following the palladium coupling procedure as set forth in Examples 1 and 2 , except using the tricyclic amine derivatives and bromo/chlorine derivatives indicated in the table below. Examples 71 and 72 were prepared following the procedures set forth in the preparation of Examples 42 and 43 , respectively, followed by the deprotection procedure set forth in Example 97 . Table 2

實例 97 將來自製備實例33之標題化合物(0.072 g, 0.142 mmol)及碳酸銫(0.107 g, 0.328 mmol)置入至微波管中,之後置入MeOH (1.5 mL)及THF (3 mL)。將反應混合物於微波中輻照至110℃持續30分鐘,且然後使其冷卻至室溫。將溶劑在減壓下去除,且使用Biotage Isolera One純化系統,利用正庚烷/乙酸乙酯梯度(80/20-> 0/100)在矽膠管柱上純化殘餘物,得到呈黃色固體之標題化合物(0.027 g, 53%)。 MS: 354.2 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 10.96 (s, 1H), 7.39 (d,J = 9.9 Hz, 1H), 7.28 (s, 1H), 7.26 (t,J = 4.4 Hz, 1H), 7.20 (dd,J = 10.0, 2.6 Hz, 1H), 6.85 (td,J = 9.2, 2.6 Hz, 1H), 4.62 (s, 2H), 3.91 (t,J = 5.7 Hz, 2H), 3.79 - 3.67 (m, 4H), 3.37 - 3.32 (m, 4H), 2.88 (t,J = 5.6 Hz, 2H)。 Example 97 The title compound from Preparative Example 33 (0.072 g, 0.142 mmol) and cesium carbonate (0.107 g, 0.328 mmol) were placed in a microwave tube, followed by MeOH (1.5 mL) and THF (3 mL). The reaction mixture was irradiated in the microwave to 110°C for 30 minutes and then allowed to cool to room temperature. The solvent was removed under reduced pressure, and the residue was purified on a silica column using a Biotage Isolera One purification system using n-heptane/ethyl acetate gradient (80/20->0/100) to obtain the title product as a yellow solid. compound (0.027 g, 53%). MS: 354.2 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 10.96 (s, 1H), 7.39 (d, J = 9.9 Hz, 1H), 7.28 (s, 1H), 7.26 (t, J = 4.4 Hz, 1H), 7.20 (dd, J = 10.0, 2.6 Hz, 1H), 6.85 (td, J = 9.2, 2.6 Hz, 1H), 4.62 (s, 2H), 3.91 (t, J = 5.7 Hz, 2H), 3.79 - 3.67 (m, 4H), 3.37 - 3.32 (m, 4H), 2.88 (t, J = 5.6 Hz, 2H).

實例 98 105f 除使用下表中所指示之甲苯磺醯基保護之前體以外,遵循如實例97中所闡述之甲苯磺酸鹽裂解程序,製備以下化合物: 3 Examples 98 to 105f Following the tosylate cleavage procedure as set forth in Example 97, except using the tosylate protected precursor indicated in the table below, the following compounds were prepared: Table 3

實例 106 步驟 A 向來自製備實例7之標題化合物(500 mg, 1.31 mmol)於DMF (5 mL)中之溶液添加2,5-二氯苯并噁唑(327 mg, 1.74 mmol)及碳酸鉀(602 mg, 4.36 mmol),且加熱至100℃持續8小時。在反應完成後,將反應混合物傾倒至冰冷卻之水中;將沈澱出之固體過濾並乾燥,得到標題化合物。產物不經進一步純化即原樣用於下一步驟(0.500 g, 77%)。 MS: 496.1 (M+H)+步驟 B 於50 mL雙頸圓底燒瓶中添加無水1,4-二噁烷(5 mL)並脫氣20 min。向此添加乙酸鈀(67 mg, 0.1008 mmol)及2-二環己基膦基-2′,4′,6′-三異丙基聯苯(144 mg, 0.302 mmol)。所得溶液呈橙色且再繼續脫氣10 min。將懸浮液在預先加熱之油浴上在100℃下加熱數秒(20- 30秒)。色彩變為深綠色。將油浴移除並脫氣5-10 min。向此添加來自上文步驟A之標題化合物(500 mg, 1.008 mmol)、嗎啉(88 mg, 1.008 mmol)及碳酸銫(0.98 g, 3.024 mmol),且將反應混合物加熱至100℃持續3小時。3 h後,LC-MS顯示產物為主要峰。經由矽藻土墊過濾反應混合物並在真空下濃縮,得到標題化合物。產物不經進一步純化即原樣用於下一步驟(0.450 g, 82%)。 MS: 547.3 (M+H)+步驟 C 向來自上文步驟B之標題化合物(450 mg, 0.82 mmol)於1,4-二噁烷:甲醇(1:1;5 mL)中之溶液添加第三丁醇鈉(230 mg, 2.4 mmol),且加熱至70℃持續16小時。將反應混合物在真空下濃縮。使用Biotage Isolera One純化系統,採用石油醚/ EtOAc梯度(0 - 100%),在矽膠管柱上實施純化,得到呈固體之標題化合物(0.270 g, 84%)。 MS: 393.2 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 11.10 (s, 1H), 7.47-7.48 (m, 1H), 7.28 (d,J = 8.72 Hz, 1H), 7.08-7.09 (m, 1H), 6.92 (d,J = 2.20 Hz, 1H), 6.83-6.84 (m, 1H), 6.62-6.63 (m, 1H), 4.81 (s, 2H), 3.98-4.00 (m, 2H), 3.73-3.74 (m, 4H), 3.04-3.05 (m, 4H), 2.94-2.95 (m, 2H)。 Example 106 Step A To a solution of the title compound from Preparative Example 7 (500 mg, 1.31 mmol) in DMF (5 mL) was added 2,5-dichlorobenzoxazole (327 mg, 1.74 mmol) and potassium carbonate (602 mg , 4.36 mmol) and heated to 100°C for 8 hours. After the reaction was completed, the reaction mixture was poured into ice-cooled water; the precipitated solid was filtered and dried to obtain the title compound. The product was used as received in the next step without further purification (0.500 g, 77%). MS: 496.1 (M+H) + . Step B : Add anhydrous 1,4-dioxane (5 mL) to a 50 mL double-neck round-bottom flask and degas for 20 min. To this were added palladium acetate (67 mg, 0.1008 mmol) and 2-dicyclohexylphosphino-2′,4′,6′-triisopropylbiphenyl (144 mg, 0.302 mmol). The resulting solution was orange in color and degassed for an additional 10 min. Heat the suspension at 100°C for a few seconds (20-30 seconds) on a preheated oil bath. The color changes to dark green. Remove the oil bath and degas for 5-10 minutes. To this were added the title compound from step A above (500 mg, 1.008 mmol), morpholine (88 mg, 1.008 mmol) and cesium carbonate (0.98 g, 3.024 mmol) and the reaction mixture was heated to 100°C for 3 hours . After 3 h, LC-MS showed that the product was the main peak. The reaction mixture was filtered through a pad of celite and concentrated in vacuo to give the title compound. The product was used as received in the next step without further purification (0.450 g, 82%). MS: 547.3 (M+H) + . Step C To a solution of the title compound from Step B above (450 mg, 0.82 mmol) in 1,4-dioxane:methanol (1:1; 5 mL) was added sodium tert-butoxide (230 mg, 2.4 mmol) and heated to 70°C for 16 hours. The reaction mixture was concentrated in vacuo. Purification was performed on a silica column using a Biotage Isolera One purification system using a petroleum ether/EtOAc gradient (0 - 100%) to give the title compound as a solid (0.270 g, 84%). MS: 393.2 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 11.10 (s, 1H), 7.47-7.48 (m, 1H), 7.28 (d, J = 8.72 Hz, 1H), 7.08-7.09 (m, 1H ), 6.92 (d, J = 2.20 Hz, 1H), 6.83-6.84 (m, 1H), 6.62-6.63 (m, 1H), 4.81 (s, 2H), 3.98-4.00 (m, 2H), 3.73- 3.74 (m, 4H), 3.04-3.05 (m, 4H), 2.94-2.95 (m, 2H).

實例 107 向來自製備實例7之標題化合物(500 mg, 1.318 mmol)於DMF (5 mL)中之溶液添加2,5-二氯苯并噻唑(268 mg, 1.32 mmol)及碳酸鉀(545 mg, 3.95 mmol),且加熱至100℃持續8小時。在反應完成後,將反應混合物傾倒至冰冷卻之水中,將沈澱出之固體過濾並乾燥,得到標題化合物(500 mg, 74%)。產物不經進一步純化即原樣用於下一步驟。 MS: 513.1 (M+H)+步驟 B 向來自上文步驟A之標題化合物(200 mg, 0.39 mmol)於無水二噁烷(5 mL)中之攪拌溶液添加嗎啉(51 mg, 0.58 mmol)、第三丁醇鈉(112 mg, 1.17 mmol),且在N2 氣氛下脫氣10 min。向此反應混合物添加參(二亞苄基丙酮)二鈀(0) (17.9 mg, 0.019 mmol)及2-二環己基膦基-2′,6′-二異丙氧基聯苯(18.2 mg, 0.039 mmol),且加熱至100℃直至反應完成為止。在反應完成後(藉由LCMS監測),經由矽藻土過濾反應混合物並用EtOAc洗滌。將濾液在減壓下濃縮以產生粗產物。使用Biotage Isolera One純化系統,採用石油醚/ EtOAc梯度(0 - 100%),在矽膠管柱上實施純化,得到呈固體之標題化合物(50 mg, 33%)。 MS: 409.1 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 11.11 (s, 1H), 7.60 (d,J = 8.68 Hz, 1H), 7.48-7.49 (m, 1H), 7.07-7.08 (m, 2H), 6.86-6.88 (m, 2H), 4.77 (s, 2H), 3.97-3.98 (m, 2H), 3.74-3.76 (m, 4H), 3.11-3.12 (m, 4H), 2.95-2.96 (m, 2H)。 Example 107 To a solution of the title compound from Preparative Example 7 (500 mg, 1.318 mmol) in DMF (5 mL) was added 2,5-dichlorobenzothiazole (268 mg, 1.32 mmol) and potassium carbonate (545 mg, 3.95 mmol) ), and heated to 100°C for 8 hours. After the reaction was completed, the reaction mixture was poured into ice-cooled water, and the precipitated solid was filtered and dried to obtain the title compound (500 mg, 74%). The product was used as received in the next step without further purification. MS: 513.1 (M+H) + . Step B To a stirred solution of the title compound from Step A above (200 mg, 0.39 mmol) in anhydrous dioxane (5 mL) was added morpholine (51 mg, 0.58 mmol), sodium tert-butoxide (112 mg , 1.17 mmol) and degassed under N atmosphere for 10 min. To this reaction mixture were added ginseng(dibenzylideneacetone)dipalladium(0) (17.9 mg, 0.019 mmol) and 2-dicyclohexylphosphino-2′,6′-diisopropoxybiphenyl (18.2 mg , 0.039 mmol) and heated to 100°C until the reaction was completed. After the reaction was complete (monitored by LCMS), the reaction mixture was filtered through celite and washed with EtOAc. The filtrate was concentrated under reduced pressure to give crude product. Purification was performed on a silica column using a Biotage Isolera One purification system using a petroleum ether/EtOAc gradient (0 - 100%) to give the title compound as a solid (50 mg, 33%). MS: 409.1 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 11.11 (s, 1H), 7.60 (d, J = 8.68 Hz, 1H), 7.48-7.49 (m, 1H), 7.07-7.08 (m, 2H ), 6.86-6.88 (m, 2H), 4.77 (s, 2H), 3.97-3.98 (m, 2H), 3.74-3.76 (m, 4H), 3.11-3.12 (m, 4H), 2.95-2.96 (m , 2H).

實例 108 步驟 A 向來自製備實例7之標題化合物(1 g, 2.63 mmol)於DMF (10 mL)中之溶液添加2,6-二氯苯并噻唑(0.537 g, 2.63 mmol)及碳酸鉀(1.09 g, 7.89 mmol),且加熱至100℃持續8小時。在反應完成後,將反應混合物傾倒至冰冷卻之水中,將沈澱出之固體過濾並乾燥,得到標題化合物(1.0 g, 74%)。產物不經進一步純化即原樣用於下一步驟。 MS: 513.2 (M+H)+步驟 B 向來自上文步驟A之標題化合物(150 mg, 0.293 mmol)於無水二噁烷(5 mL)中之攪拌溶液添加嗎啉(39 mg, 0.44 mmol)、第三丁醇鈉(85 mg, 0.87 mmol),且在N2 氣氛下脫氣10 min。向此反應混合物添加參(二亞苄基丙酮)二鈀(0) (13.4 mg, 0.0015 mmol)及2-二環己基膦基-2′,6′-二異丙氧基聯苯(13.65 mg, 0.029 mmol),且加熱至100℃直至反應完成為止。在反應完成後(藉由LCMS監測),經由矽藻土過濾反應混合物並用EtOAc洗滌。將濾液在減壓下濃縮以產生粗產物。使用Biotage Isolera One純化系統,採用石油醚/ EtOAc梯度(0 - 100%),在矽膠管柱上實施純化,得到呈固體之標題化合物(60 mg, 50%)。 MS: 409.1 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 11.10 (s, 1H), 7.48-7.50 (m, 1H), 7.36-7.38 (m, 2H), 7.08-7.09 (m, 1H), 6.96-6.97 (m, 1H), 6.85-6.86 (m, 1H), 4.74 (s, 2H), 3.94-3.95 (m, 2H), 3.74-3.75 (m, 4H), 3.06-3.07 (m, 4H), 2.94-2.95 (m, 2H)。 Example 108 Step A To a solution of the title compound from Preparative Example 7 (1 g, 2.63 mmol) in DMF (10 mL) was added 2,6-dichlorobenzothiazole (0.537 g, 2.63 mmol) and potassium carbonate (1.09 g, 7.89 mmol) and heated to 100°C for 8 hours. After the reaction was completed, the reaction mixture was poured into ice-cooled water, and the precipitated solid was filtered and dried to obtain the title compound (1.0 g, 74%). The product was used as received in the next step without further purification. MS: 513.2 (M+H) + . Step B To a stirred solution of the title compound from Step A above (150 mg, 0.293 mmol) in anhydrous dioxane (5 mL) was added morpholine (39 mg, 0.44 mmol), sodium tert-butoxide (85 mg , 0.87 mmol) and degassed under N2 atmosphere for 10 min. To this reaction mixture were added ginseng(dibenzylideneacetone)dipalladium(0) (13.4 mg, 0.0015 mmol) and 2-dicyclohexylphosphino-2′,6′-diisopropoxybiphenyl (13.65 mg , 0.029 mmol) and heated to 100°C until the reaction was completed. After the reaction was complete (monitored by LCMS), the reaction mixture was filtered through celite and washed with EtOAc. The filtrate was concentrated under reduced pressure to give crude product. Purification was performed on a silica column using a Biotage Isolera One purification system using a petroleum ether/EtOAc gradient (0 - 100%) to give the title compound as a solid (60 mg, 50%). MS: 409.1 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 11.10 (s, 1H), 7.48-7.50 (m, 1H), 7.36-7.38 (m, 2H), 7.08-7.09 (m, 1H), 6.96 -6.97 (m, 1H), 6.85-6.86 (m, 1H), 4.74 (s, 2H), 3.94-3.95 (m, 2H), 3.74-3.75 (m, 4H), 3.06-3.07 (m, 4H) , 2.94-2.95 (m, 2H).

實例 109 步驟 A 向來自製備實例7之標題化合物(250 mg, 0.725 mmol)於DMF (5 mL)中之溶液添加2,6-二氯苯并噁唑(166 mg, 0.88 mmol)及碳酸鉀(326 mg, 2.36 mmol),且加熱至100℃持續8小時。在反應完成後,將反應混合物傾倒至冰冷卻之水中,將沈澱出之固體過濾並乾燥,得到標題化合物(250 mg, 70%)。產物不經進一步純化即原樣用於下一步驟。 MS: 496.1 (M+H)+步驟 B 向來自上文步驟A之標題化合物(250 mg, 0.505 mmol)於無水二噁烷(5 mL)中之攪拌溶液添加嗎啉(53 mg, 0.60 mmol)、第三丁醇鈉(145 mg, 1.52 mmol),且在N2 氣氛下脫氣10 min。向此反應混合物添加參(二亞苄基丙酮)二鈀(0) (23.1 mg, 0.0252 mmol)及2-二環己基膦基-2′,6′-二異丙氧基聯苯(23.53 mg, 0.051 mmol),且加熱至100℃直至反應完成為止。在反應完成後(藉由LCMS監測),經由矽藻土過濾反應混合物並用EtOAc洗滌。將濾液在減壓下濃縮以產生粗產物。使用Biotage Isolera One純化系統,採用石油醚/ EtOAc梯度(0 - 100%),在矽膠管柱上實施純化,得到呈固體之標題化合物(50 mg, 25%)。 MS: 393.2 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ = 11.09 (s, 1H), 7.45-7.46 (m, 1H), 7.17 (d,J = 8.56 Hz, 1H), 7.08-7.09 (m, 2H), 6.87-6.79 (m, 2H), 4.77 (s, 2H), 3.95-3.96 (m, 2H), 3.72-3.74 (m, 4H), 3.03-3.04 (m, 4H), 2.91-2.93 (m, 2H)。 Example 109 Step A To a solution of the title compound from Preparative Example 7 (250 mg, 0.725 mmol) in DMF (5 mL) was added 2,6-dichlorobenzoxazole (166 mg, 0.88 mmol) and potassium carbonate (326 mg , 2.36 mmol) and heated to 100°C for 8 hours. After the reaction was completed, the reaction mixture was poured into ice-cooled water, and the precipitated solid was filtered and dried to obtain the title compound (250 mg, 70%). The product was used as received in the next step without further purification. MS: 496.1 (M+H) + . Step B To a stirred solution of the title compound from Step A above (250 mg, 0.505 mmol) in anhydrous dioxane (5 mL) was added morpholine (53 mg, 0.60 mmol), sodium tert-butoxide (145 mg , 1.52 mmol) and degassed under N atmosphere for 10 min. To this reaction mixture were added ginseng(dibenzylideneacetone)dipalladium(0) (23.1 mg, 0.0252 mmol) and 2-dicyclohexylphosphino-2′,6′-diisopropoxybiphenyl (23.53 mg , 0.051 mmol) and heated to 100°C until the reaction was completed. After the reaction was complete (monitored by LCMS), the reaction mixture was filtered through celite and washed with EtOAc. The filtrate was concentrated under reduced pressure to give crude product. Purification was performed on a silica column using a Biotage Isolera One purification system using a petroleum ether/EtOAc gradient (0 - 100%) to give the title compound as a solid (50 mg, 25%). MS: 393.2 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ = 11.09 (s, 1H), 7.45-7.46 (m, 1H), 7.17 (d, J = 8.56 Hz, 1H), 7.08-7.09 (m, 2H ), 6.87-6.79 (m, 2H), 4.77 (s, 2H), 3.95-3.96 (m, 2H), 3.72-3.74 (m, 4H), 3.03-3.04 (m, 4H), 2.91-2.93 (m , 2H).

實例 110 步驟 A 向來自製備實例1 之標題化合物(0.15 g, 0.43 mmol)於無水1,4-二噁烷(5 mL)中之攪拌溶液添加來自製備實例50 之標題化合物(0.17 g, 0.43 mmol)及Cs2 CO3 (0.420 g, 1.29 mmol)。將反應混合物在N2 氣氛下脫氣10 min。然後添加Pd(OAc)2 (0.009 g, 0.043 mmol)及2-二環己基膦基-2′,4′,6′-三異丙基聯苯‎(0.062 g; 0.129 mmol),且將反應混合物加熱至100℃直至反應完成為止。在反應完成後(藉由LCMS監測),經由矽藻土過濾反應混合物並用乙酸乙酯洗滌。將濾液在減壓下濃縮以產生粗產物。藉由急速管柱層析或製備型HPLC純化粗製材料,得到經甲苯磺醯基保護之化合物。向甲苯磺醯基化合物(1.0 eq)於二噁烷:MeOH (1:1, 10 vol)中之溶液添加NaOtBu (3 eq),且加熱至70℃持續6小時。將反應混合物在真空下濃縮,且使粗產物進行管柱純化,得到期望產物。藉由急速管柱層析或製備型HPLC純化粗製材料,得到標題化合物(0.042 g, 24%)。1 H-NMR (400 MHz, DMSO-d 6 ): δ 11.00 (bs, 1H), 8.70 (s, 1H), 7.96 (d,J = 9.60 Hz, 1H), 7.45-7.46 (m, 1H), 7.39 (d,J = 9.20 Hz, 1H), 7.07-7.08 (m, 2H), 6.82-6.83 (m, 1H), 4.64 (s, 2H), 4.02-4.04 (m, 2H), 3.73 (d,J = 4.80 Hz, 4H), 3.59 (d,J = 4.40 Hz, 4H), 2.90 (bs, 2H)。 MS: 404.2 (M+H)+ Example 110 Step A To a stirred solution of the title compound from Preparative Example 1 (0.15 g, 0.43 mmol) in anhydrous 1,4-dioxane (5 mL) was added the title compound from Preparative Example 50 (0.17 g, 0.43 mmol) and Cs 2 CO 3 (0.420 g, 1.29 mmol). The reaction mixture was degassed under N2 atmosphere for 10 min. Then Pd(OAc) 2 (0.009 g, 0.043 mmol) and 2-dicyclohexylphosphino-2′,4′,6′-triisopropylbiphenyl‎ (0.062 g; 0.129 mmol) were added, and the reaction was The mixture was heated to 100°C until the reaction was complete. After the reaction was complete (monitored by LCMS), the reaction mixture was filtered through celite and washed with ethyl acetate. The filtrate was concentrated under reduced pressure to give crude product. The crude material is purified by flash column chromatography or preparative HPLC to obtain the tosyl-protected compound. To a solution of tosyl compound (1.0 eq) in dioxane:MeOH (1:1, 10 vol) was added NaOtBu (3 eq) and heated to 70°C for 6 h. The reaction mixture was concentrated in vacuo and the crude product was column purified to give the desired product. The crude material was purified by flash column chromatography or preparative HPLC to afford the title compound (0.042 g, 24%). 1 H-NMR (400 MHz, DMSO- d 6 ): δ 11.00 (bs, 1H), 8.70 (s, 1H), 7.96 (d, J = 9.60 Hz, 1H), 7.45-7.46 (m, 1H), 7.39 (d, J = 9.20 Hz, 1H), 7.07-7.08 (m, 2H), 6.82-6.83 (m, 1H), 4.64 (s, 2H), 4.02-4.04 (m, 2H), 3.73 (d, J = 4.80 Hz, 4H), 3.59 (d, J = 4.40 Hz, 4H), 2.90 (bs, 2H). MS: 404.2 (M+H) + .

實例 111 112 在甲苯磺醯基去保護之後,遵循如實例110 中所闡述之鈀偶合程序,製備以下化合物。 4 實例 113 步驟 A 將乙酸鈀(II)(0.013 g, 0.058 mmol)及4,5-雙(二苯基膦基)-9,9-二甲基𠮿(XANTPHOS) (0.101 g, 0.174 mmol)置於反應小瓶中並脫氣。添加1,4-二噁烷(6 mL)。將所得溶液短暫脫氣。將懸浮液在100℃下(在預加熱之加熱塊上)加熱少於1分鐘,直至溶液之色彩自橙色變為深粉色為止。然後,將小瓶自加熱塊移除,且添加來自製備實例52之標題化合物(148 mg, 0.581 mmol)、來自製備實例1之標題化合物(0.200 g, 0.581 mmol)及碳酸銫(0.662 g, 2.033 mmol)。將反應小瓶填充氬,之後將其關閉。將反應混合物在100℃下加熱18 h。用乙酸乙酯及水稀釋該反應混合物。分離有機層,且將水層用乙酸乙酯再萃取兩次。使合併之有機層經Na2 SO4 乾燥,過濾並將溶劑在減壓下蒸發。使用Biotage Isolera One純化系統,採用正庚烷/乙酸乙酯(100/00 --> 50/50)梯度,在矽膠管柱上純化粗產物。將所獲得之粗製反應混合物(0.261 g, 0.469 mmol)及碳酸銫(0.214 g, 0.658 mmol)置入至微波管中,之後置入MeOH (4 mL)及THF (8 mL)。將反應混合物於微波中輻照至110℃持續30分鐘,且然後使其冷卻至室溫。將溶劑在減壓下去除,且使用Biotage Isolera One純化系統,利用二氯甲烷/甲醇梯度(100/00 --> 95/05)在矽膠管柱上純化殘餘物,得到呈黃色固體之標題化合物(0.020 g, 9%)。1 H NMR (400 MHz, DMSO-d6 ) δ 10.97 (s, 1H), 7.95 (d, 1H), 7.54 (d, 2H), 7.33 - 7.10 (m, 4H), 6.86 (td, 1H), 4.41 (s, 2H), 3.72 (dt, 6H), 3.55 (t, 4H), 2.95 (d, 2H)。 MS: 403.19 (M+H)+ Examples 111 and 112 Following deprotection of the toluenesulfonyl group, the following compounds were prepared following the palladium coupling procedure as set forth in Example 110 . Table 4 Example 113 Step A : Palladium(II) acetate (0.013 g, 0.058 mmol) and 4,5-bis(diphenylphosphino)-9,9-dimethylmethacrylate (XANTPHOS) (0.101 g, 0.174 mmol) was placed in the reaction vial and degassed. Add 1,4-dioxane (6 mL). The resulting solution was briefly degassed. Heat the suspension at 100°C (on a preheated heating block) for less than 1 minute until the color of the solution changes from orange to dark pink. The vial was then removed from the heating block and the title compound from Preparative Example 52 (148 mg, 0.581 mmol), the title compound from Preparative Example 1 (0.200 g, 0.581 mmol) and cesium carbonate (0.662 g, 2.033 mmol) were added ). The reaction vial was filled with argon before being closed. The reaction mixture was heated at 100 °C for 18 h. The reaction mixture was diluted with ethyl acetate and water. The organic layer was separated, and the aqueous layer was extracted twice more with ethyl acetate. The combined organic layers were dried over Na2SO4 , filtered and the solvent evaporated under reduced pressure . The crude product was purified on a silica column using a Biotage Isolera One purification system using n-heptane/ethyl acetate (100/00 --> 50/50) gradient. The obtained crude reaction mixture (0.261 g, 0.469 mmol) and cesium carbonate (0.214 g, 0.658 mmol) were placed into a microwave tube, followed by MeOH (4 mL) and THF (8 mL). The reaction mixture was irradiated in the microwave to 110°C for 30 minutes and then allowed to cool to room temperature. The solvent was removed under reduced pressure, and the residue was purified on a silica column using a Biotage Isolera One purification system using a methylene chloride/methanol gradient (100/00 --> 95/05) to obtain the title compound as a yellow solid. (0.020 g, 9%). 1 H NMR (400 MHz, DMSO-d 6 ) δ 10.97 (s, 1H), 7.95 (d, 1H), 7.54 (d, 2H), 7.33 - 7.10 (m, 4H), 6.86 (td, 1H), 4.41 (s, 2H), 3.72 (dt, 6H), 3.55 (t, 4H), 2.95 (d, 2H). MS: 403.19 (M+H) + .

實例 114 步驟 A 將乙酸鈀(II)(0.013 g, 0.058 mmol)及4,5-雙(二苯基膦基)-9,9-二甲基𠮿(XANTPHOS) (0.101 g, 0.174 mmol)置於反應小瓶中並脫氣。添加1,4-二噁烷(6 mL)。將所得溶液短暫脫氣。將懸浮液在100℃下(在預加熱之加熱塊上)加熱少於1分鐘,直至溶液之色彩自橙色變為深粉色為止。然後,將小瓶自加熱塊移除,且添加來自製備實例51之標題化合物(144 mg, 0.581 mmol)、來自製備實例1之標題化合物(0.200 g, 0.581 mmol)及碳酸銫(0.662 g, 2.033 mmol)。將反應小瓶填充氬,之後將其關閉。將反應混合物在100℃下加熱18 h。用乙酸乙酯及水稀釋該反應混合物。分離有機層,且將水層用乙酸乙酯再萃取兩次。使合併之有機層經Na2 SO4 乾燥,過濾並將溶劑在減壓下蒸發。使用Biotage Isolera One純化系統,採用正庚烷/乙酸乙酯梯度(100/00 --> 50/50),在矽膠管柱上純化粗產物。將所獲得之粗製反應混合物(0.122 g, 0.219 mmol)及碳酸銫(0.214 g, 0.658 mmol)置入至微波管中,之後置入MeOH (4 mL)及THF (8 mL)。將反應混合物於微波中輻照至110℃持續30分鐘,且然後使其冷卻至室溫。將溶劑在減壓下去除,且使用Biotage Isolera One純化系統,利用二氯甲烷/甲醇梯度(100/00 --> 95/05)在矽膠管柱上純化殘餘物,得到呈黃色固體之標題化合物(0.081 g, 35%)。1 H NMR (400 MHz, DMSO-d6 ) δ 10.97 (s, 1H), 7.88 (d, 1H), 7.56 (d, 1H), 7.40 - 7.13 (m, 3H), 7.02 (d, 1H), 6.97 - 6.79 (m, 2H), 4.49 (s, 2H), 3.80 (t, 2H), 3.72 (t, 4H), 3.60 (t, 4H), 2.93 (d, 2H)。 MS: 403.20 (M+H)+ Example 114 Step A : Palladium(II) acetate (0.013 g, 0.058 mmol) and 4,5-bis(diphenylphosphino)-9,9-dimethylmethacrylate (XANTPHOS) (0.101 g, 0.174 mmol) was placed in the reaction vial and degassed. Add 1,4-dioxane (6 mL). The resulting solution was briefly degassed. Heat the suspension at 100°C (on a preheated heating block) for less than 1 minute until the color of the solution changes from orange to dark pink. The vial was then removed from the heating block and the title compound from Preparative Example 51 (144 mg, 0.581 mmol), the title compound from Preparative Example 1 (0.200 g, 0.581 mmol) and cesium carbonate (0.662 g, 2.033 mmol) were added ). The reaction vial was filled with argon before being closed. The reaction mixture was heated at 100 °C for 18 h. The reaction mixture was diluted with ethyl acetate and water. The organic layer was separated, and the aqueous layer was extracted twice more with ethyl acetate. The combined organic layers were dried over Na2SO4 , filtered and the solvent evaporated under reduced pressure . The crude product was purified on a silica column using a Biotage Isolera One purification system using n-heptane/ethyl acetate gradient (100/00 --> 50/50). The obtained crude reaction mixture (0.122 g, 0.219 mmol) and cesium carbonate (0.214 g, 0.658 mmol) were placed into a microwave tube, followed by MeOH (4 mL) and THF (8 mL). The reaction mixture was irradiated in the microwave to 110°C for 30 minutes and then allowed to cool to room temperature. The solvent was removed under reduced pressure, and the residue was purified on a silica column using a Biotage Isolera One purification system using a methylene chloride/methanol gradient (100/00 --> 95/05) to obtain the title compound as a yellow solid. (0.081 g, 35%). 1 H NMR (400 MHz, DMSO-d 6 ) δ 10.97 (s, 1H), 7.88 (d, 1H), 7.56 (d, 1H), 7.40 - 7.13 (m, 3H), 7.02 (d, 1H), 6.97 - 6.79 (m, 2H), 4.49 (s, 2H), 3.80 (t, 2H), 3.72 (t, 4H), 3.60 (t, 4H), 2.93 (d, 2H). MS: 403.20 (M+H) + .

實例 115 步驟 A 將Pd(OAc)2 (16 mg,0.072 mmol)及2-二環己基膦基-2′,4′,6′-三異丙基聯苯‎(XPhos;103 mg, 0.2177 mmol)添加至反應小瓶,且添加經脫氣之二噁烷(5 ml)。將小瓶填充氬氣且密封。將懸浮液在100℃下加熱1分鐘,然後添加8-氟-5-甲苯磺醯基-2,3,4,5-四氫-1H-吡啶并[4,3-b]吲哚(製備實例1 ) (250 mg, 0.725 mmol)、4-(4-溴苯基)嗎啉(210 mg, 0.87 mmol)及Cs2 CO3 (707 mg, 2.177 mmol),且將溶液在100℃下加熱18小時。用乙酸乙酯(30 mL)及水(30 mL)稀釋反應混合物。分離有機相,且將水相用乙酸乙酯再萃取兩次。使合併之有機相經Na2 SO4 乾燥,過濾並將溶劑在減壓下蒸發。藉由採用DCM/MeOH梯度(100/0 -> 95/05),在HP-Sil管柱(Biotage)上純化粗製物,得到呈黃色固體之4-(4-(8-氟-5-甲苯磺醯基-1,3,4,5-四氫-2H-吡啶并[4,3-b]吲哚-2-基)苯基)嗎啉(235 mg, 64%)。 向4-(4-(8-氟-5-甲苯磺醯基-1,3,4,5-四氫-2H-吡啶并[4,3-b]吲哚-2-基)苯基)嗎啉(0.235 mg, 0.465 mmol)於二噁烷:MeOH (1:1, 5 vol)中之溶液添加NaOtBu (133 mg,1.39 mmol),且加熱至70℃持續6小時。將反應混合物在真空下濃縮,且藉由採用DCM/MeOH梯度(100/0 -> 95/05),在HP-Sil管柱(Biotage)上管柱純化粗產物,得到呈灰白色固體之標題化合物(57 mg, 35%) MS: 352.0 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ 10.93 (bs, 1H), 7.21-7.24 (m, 2H), 6.99-7.01 (m, 2H), 6.85-6.86 (m, 3H), 4.24 (s, 2H), 3.73 (bs, 4H), 3.52-3.53 (m, 2H), 2.98 (bs, 4H), 2.86 (bs, 2H)。 Example 115 Step A Add Pd(OAc) 2 (16 mg, 0.072 mmol) and 2-dicyclohexylphosphino-2′,4′,6′-triisopropylbiphenyl‎ (XPhos; 103 mg, 0.2177 mmol) to the reaction vial and add degassed dioxane (5 ml). The vial was filled with argon and sealed. The suspension was heated at 100 °C for 1 min and 8-fluoro-5-toluenesulfonyl-2,3,4,5-tetrahydro-1H-pyrido[4,3-b]indole (Preparation Example 1 ) (250 mg, 0.725 mmol), 4-(4-bromophenyl)morpholine (210 mg, 0.87 mmol) and Cs 2 CO 3 (707 mg, 2.177 mmol), and the solution was heated at 100°C 18 hours. Dilute the reaction mixture with ethyl acetate (30 mL) and water (30 mL). The organic phase was separated and the aqueous phase was extracted twice more with ethyl acetate. The combined organic phases were dried over Na2SO4 , filtered and the solvent evaporated under reduced pressure . The crude material was purified on a HP-Sil column (Biotage) using a DCM/MeOH gradient (100/0 -> 95/05) to obtain 4-(4-(8-fluoro-5-toluene) as a yellow solid Sulfonyl-1,3,4,5-tetrahydro-2H-pyrido[4,3-b]indol-2-yl)phenyl)morpholine (235 mg, 64%). To 4-(4-(8-fluoro-5-toluenesulfonyl-1,3,4,5-tetrahydro-2H-pyrido[4,3-b]indol-2-yl)phenyl) A solution of morpholine (0.235 mg, 0.465 mmol) in dioxane:MeOH (1:1, 5 vol) was added NaOtBu (133 mg, 1.39 mmol) and heated to 70°C for 6 h. The reaction mixture was concentrated in vacuo and the crude product was column purified on a HP-Sil column (Biotage) using a DCM/MeOH gradient (100/0 -> 95/05) to give the title compound as an off-white solid (57 mg, 35%) MS: 352.0 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ 10.93 (bs, 1H), 7.21-7.24 (m, 2H), 6.99-7.01 (m, 2H), 6.85-6.86 (m, 3H), 4.24 ( s, 2H), 3.73 (bs, 4H), 3.52-3.53 (m, 2H), 2.98 (bs, 4H), 2.86 (bs, 2H).

實例 116 向7-氟-5-甲苯磺醯基-2,3,4,5-四氫-1H-吡啶并[4,3-b]吲哚(製備實例7 ) (250 mg, 0.7267 mmol)於無水二噁烷(5 mL)中之攪拌溶液添加5-氯-N-(2-甲氧基乙基)-N-甲基苯并[d]噁唑-2-胺(製備實例60 ) (190 mg, 0.7994 mmol)、第三丁醇鈉(202 mg, 2.180 mmol),且在N2 氣氛下脫氣10 min。向此反應混合物添加Ruphos G4 Pd (60 mg, 0.0726 mmol),且加熱至100℃直至反應完成為止。在反應完成後,經由矽藻土床過濾該反應混合物,用EtOAc洗滌。將濾液濃縮,且藉由採用DCM/MeOH梯度(100/0 -> 95/05)在HP-Sil管柱(Biotage)上藉由管柱層析純化粗製物,得到標題化合物(37 mg, 13%) MS: 395.0 (M+H)+ 。 1H-NMR (400 MHz, DMSO-d 6 ) δ 10.94 (bs, 1H), 7.43-7.44 (m, 1H), 7.23 (d,J = 8.80 Hz, 1H), 7.06-7.06 (m, 1H), 6.99 (d,J = 2.40 Hz, 1H), 6.80-6.81 (m, 1H), 6.71-6.72 (m, 1H), 4.32 (s, 2H), 3.64-3.65 (m, 2H), 3.56-3.58 (m, 4H), 3.27 (s, 3H), 3.13 (s, 3H), 2.86 (t,J = 5.20 Hz, 2H)。 Example 116 To 7-fluoro-5-toluenesulfonyl-2,3,4,5-tetrahydro-1H-pyrido[4,3-b]indole (Preparation Example 7 ) (250 mg, 0.7267 mmol) was dissolved in anhydrous To a stirred solution in dioxane (5 mL) was added 5-chloro-N-(2-methoxyethyl)-N-methylbenzo[d]oxazole-2-amine (Preparation Example 60 ) (190 mg, 0.7994 mmol), sodium tert-butoxide (202 mg, 2.180 mmol), and degassed under N2 atmosphere for 10 min. To this reaction mixture was added Ruphos G4 Pd (60 mg, 0.0726 mmol) and heated to 100°C until the reaction was complete. After the reaction was complete, the reaction mixture was filtered through a bed of celite, washing with EtOAc. The filtrate was concentrated and the crude material was purified by column chromatography on a HP-Sil column (Biotage) using a DCM/MeOH gradient (100/0 -> 95/05) to give the title compound (37 mg, 13 %) MS: 395.0 (M+H) + . 1H-NMR (400 MHz, DMSO- d 6 ) δ 10.94 (bs, 1H), 7.43-7.44 (m, 1H), 7.23 (d, J = 8.80 Hz, 1H), 7.06-7.06 (m, 1H), 6.99 (d, J = 2.40 Hz, 1H), 6.80-6.81 (m, 1H), 6.71-6.72 (m, 1H), 4.32 (s, 2H), 3.64-3.65 (m, 2H), 3.56-3.58 ( m, 4H), 3.27 (s, 3H), 3.13 (s, 3H), 2.86 (t, J = 5.20 Hz, 2H).

實例 117 145 遵循實例115 及實例116 中所報導之程序,製備以下化合物。 5 Examples 117 to 145 Following the procedures reported in Example 115 and Example 116 , the following compounds were prepared. Table 5

實例 146 步驟 A 在0℃下向氫化鈉(0.0293 g, 0.764 mmol)於DMF (3 mL)中之懸浮液逐滴添加5-(7-氟-1,3,4,5-四氫吡啶并[4,3-b]吲哚-2-基)-2-嗎啉基-1,3-苯并噁唑(實例45 ) (0.100 g, 0.255 mmol) (溶解於3 mL DMF中),然後在室溫下攪拌60 min。之後,在0℃下逐滴添加碘乙烷(0.0596 g, 0.382 mmol) (溶解於2 mL DMF中),且然後在室溫下攪拌3 h。根據TLC在反應完成後,利用冰水將反應混合物淬滅,之後使用乙酸乙酯(20 mL)進行萃取。將有機層分離,經硫酸鈉乾燥,過濾且然後濃縮以得到粗製物,且然後藉由矽膠管柱(Biotage)使用於石油醚中之40%-50%乙酸乙酯純化為呈灰白色固體之5-(5-乙基-7-氟-3,4-二氫-1H-吡啶并[4,3-b]吲哚-2-基)-2-嗎啉基-1,3-苯并噁唑。(15 mg, 14%) MS: 421.2 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ 7.47-7.48 (m, 1H), 7.26-7.28 (m, 2H), 7.06 (d,J = 2.40 Hz, 1H), 6.84-6.85 (m, 1H), 6.78-6.79 (m, 1H), 4.33 (s, 2H), 4.10 (q,J = 7.20 Hz, 2H), 3.55-3.57 (m, 10H), 2.90 (t,J = 4.80 Hz, 2H), 1.22 (t,J = 7.20 Hz, 3H)。 Example 146 Step A To a suspension of sodium hydride (0.0293 g, 0.764 mmol) in DMF (3 mL) was added dropwise 5-(7-fluoro-1,3,4,5-tetrahydropyrido[4 ,3-b]indol-2-yl)-2-morpholinyl-1,3-benzoxazole (Example 45 ) (0.100 g, 0.255 mmol) (dissolved in 3 mL DMF), then incubated in the chamber Stir at room temperature for 60 minutes. Afterwards, ethyl iodide (0.0596 g, 0.382 mmol) (dissolved in 2 mL DMF) was added dropwise at 0 °C, and then stirred at room temperature for 3 h. After the reaction was completed according to TLC, the reaction mixture was quenched with ice water, and then extracted with ethyl acetate (20 mL). The organic layer was separated, dried over sodium sulfate, filtered and then concentrated to give crude material, which was then purified by silica column (Biotage) using 40%-50% ethyl acetate in petroleum ether to 5 as an off-white solid. -(5-ethyl-7-fluoro-3,4-dihydro-1H-pyrido[4,3-b]indol-2-yl)-2-morpholinyl-1,3-benzox Azole. (15 mg, 14%) MS: 421.2 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ 7.47-7.48 (m, 1H), 7.26-7.28 (m, 2H), 7.06 (d, J = 2.40 Hz, 1H), 6.84-6.85 (m, 1H), 6.78-6.79 (m, 1H), 4.33 (s, 2H), 4.10 (q, J = 7.20 Hz, 2H), 3.55-3.57 (m, 10H), 2.90 (t, J = 4.80 Hz, 2H ), 1.22 (t, J = 7.20 Hz, 3H).

實例 147 153 除使用下表中所指示之三環胺基衍生物及溴/氯衍生物以外,遵循如實例1 2115 中所闡述之鈀偶合程序,製備以下化合物。 5a Examples 147 to 153 The following compounds were prepared following the palladium coupling procedure as set forth in Examples 1 , 2 , and 115 , except using the tricyclic amine derivatives and bromo/chlorine derivatives indicated in the table below. Table 5a

本發明化合物之 HCl 實例 89 HCl 步驟 A 向冷卻至0℃之4-(6-(7-氟-1,3,4,5-四氫-2H-吡啶并[4,3-b]吲哚-2-基)噻唑并[4,5-c]吡啶-2-基)嗎啉(實例89 ) (0.1 g, 0.244 mmol)於無水DCM (5 mL)中之溶液添加4 M HCl於二噁烷(0.2 mL)並攪拌1小時。將反應混合物在真空下濃縮且與二乙醚一起研磨,得到呈棕色固體之標題化合物(0.09 g, 83%)。 MS: 410.2 (M+H)+1 H-NMR (400 MHz, DMSO-d 6 ) δ 11.14 (bs, 1H), 8.28 (s, 1H), 7.98 (s, 1H), 7.38-7.40 (m, 1H), 7.11-7.12 (m, 1H), 6.85-6.86 (m, 1H), 4.75 (s, 2H), 4.02 (t,J = 5.64 Hz, 2H), 3.73-3.75 (m, 8H), 2.97 (s, 2H)。 Example of HCl salt of the compound of the present invention 89 HCl Step A : 4-(6-(7-fluoro-1,3,4,5-tetrahydro-2H-pyrido[4,3-b]indol-2-yl)thiazolo[ A solution of 4,5-c]pyridin-2-yl)morpholine (Example 89 ) (0.1 g, 0.244 mmol) in anhydrous DCM (5 mL) was added 4 M HCl in dioxane (0.2 mL) and stirred for 1 hours. The reaction mixture was concentrated in vacuo and triturated with diethyl ether to give the title compound as a brown solid (0.09 g, 83%). MS: 410.2 (M+H) + . 1 H-NMR (400 MHz, DMSO- d 6 ) δ 11.14 (bs, 1H), 8.28 (s, 1H), 7.98 (s, 1H), 7.38-7.40 (m, 1H), 7.11-7.12 (m, 1H), 6.85-6.86 (m, 1H), 4.75 (s, 2H), 4.02 (t, J = 5.64 Hz, 2H), 3.73-3.75 (m, 8H), 2.97 (s, 2H).

實例 遵循如實例89 HCl 中所闡述之鹽酸鹽程序,製備以下化合物。 6 EXAMPLE Following the hydrochloride salt procedure as set forth in Example 89 HCl , the following compounds were prepared. Table 6

本發明化合物之放射性配體合成 實例 3 H-12 將觸媒添加至氚反應容器,之後添加實例12 起始材料(1.0 mg, 0.003 mmol)於二氯甲烷中之溶液。將容器附接至氚管線並在-200℃下對氚氣加壓。將溶液攪拌8小時,冷卻至-200℃且去除過量氣體。用4 × 1 mL甲醇沖洗反應燒瓶,將其各自轉移至100 mL回收燒瓶。使合併之有機相在真空下蒸發(粗產率:124 mCi)。藉由HPLC純化粗製材料,使移動相在真空下蒸發且將產物重新溶解於無水乙醇中(產率:17 mCi,純度> 97%)。藉由質譜測定比活性為80.6 Ci/mmol。 Example 3 H-12 for the synthesis of radioactive ligands of the compounds of the present invention The catalyst was added to the tritium reaction vessel, followed by a solution of Example 12 starting material (1.0 mg, 0.003 mmol) in dichloromethane. Attach the container to the tritium line and pressurize the tritium gas at -200°C. The solution was stirred for 8 hours, cooled to -200°C and excess gas removed. Rinse the reaction flask with 4 × 1 mL of methanol and transfer each to a 100 mL recovery flask. The combined organic phases were evaporated in vacuo (crude yield: 124 mCi). The crude material was purified by HPLC, the mobile phase was evaporated under vacuum and the product was redissolved in absolute ethanol (yield: 17 mCi, purity >97%). The specific activity was determined by mass spectrometry to be 80.6 Ci/mmol.

實例 3 H-45 將觸媒添加至氚反應容器,之後添加實例45 起始材料(1.0 mg, 0.003 mmol)於二氯甲烷中之溶液。將容器附接至氚管線並在-200℃下對氚氣加壓。將溶液攪拌8小時,冷卻至-200℃且去除過量氣體。用4 × 1 mL甲醇沖洗反應燒瓶,將其各自轉移至100 mL回收燒瓶。使合併之有機相在真空下蒸發(粗產率:124 mCi)。藉由HPLC純化粗製材料,使移動相在真空下蒸發且將產物重新溶解於無水乙醇中(產率:17 mCi,純度> 99%)。藉由質譜測定比活性為96.8 Ci/mmol。 Example 3 H-45 The catalyst was added to the tritium reaction vessel, followed by a solution of Example 45 starting material (1.0 mg, 0.003 mmol) in dichloromethane. Attach the container to the tritium line and pressurize the tritium gas at -200°C. The solution was stirred for 8 hours, cooled to -200°C and excess gas removed. Rinse the reaction flask with 4 × 1 mL of methanol and transfer each to a 100 mL recovery flask. The combined organic phases were evaporated in vacuo (crude yield: 124 mCi). The crude material was purified by HPLC, the mobile phase was evaporated under vacuum and the product was redissolved in absolute ethanol (yield: 17 mCi, purity >99%). The specific activity was determined by mass spectrometry to be 96.8 Ci/mmol.

生物分析描述 藉由硫代黃素 T (ThT) 之全長 Tau (flTau) 解聚分析 將人類Tau之最長同種型(2N4R;441個胺基酸)在細菌中表現並純化。對於藉由ThT之Tau解聚分析,使35 µM於PBS中之重組全長(fl)Tau在37℃下在50 µM肝素(Sigma-Aldrich)及10 mM DTT (Sigma-Aldrich)存在下在750 RPM振盪下聚集24小時。將化合物溶解於無水二甲亞碸(DMSO, Sigma-Aldrich)中以達到10 mM之濃度。將flTau聚集體及化合物之連續稀釋液一起混合於PBS (體積50 µL)中至2 μM flTau聚集體及160 µM至0.04 µM化合物之終濃度。將混合物在室溫(RT)下培育30分鐘,然後將40 µL之此混合物轉移至黑色384孔板分析(Perkin-Elmer)中,且與10 µL之100 µM ThT (於PBS中之250 mM甘胺酸中(二者均來自Sigma-Aldrich))混合。在Tecan讀數儀(激發:440 nm;發射:485 nm)上以一式一份或一式兩份量測螢光(相對螢光單位;RFU)。然後計算flTau解聚百分比,且使用GraphPad Prism第5版(GraphPad軟體)假設一個結合位點擬合模型來測定半最大有效濃度(EC50 )。藉由 ThT Tau K18 解聚分析 將涵蓋人類Tau441之最長同種型(2N4R)之胺基酸244至372之Tau K18片段在細菌中表現並純化,或自SignalChem購買。對於藉由ThT之K18解聚分析,使35 µM於PBS中之重組K18在37℃下在50 µM肝素(Sigma-Aldrich)及10 mM DTT (Sigma-Aldrich)存在下在750 RPM振盪下聚集24小時。將化合物溶解於無水二甲亞碸 (DMSO, Sigma-Aldrich)中以達到10 mM之濃度。將K18聚集體及化合物之連續稀釋液一起混合於PBS (體積50 µL)中至2 μM K18聚集體及160 µM至0.04 µM化合物之終濃度。將混合物在室溫(RT)下培育30分鐘,然後將40 µL之此混合物轉移至黑色384孔板分析(Perkin-Elmer)中,且與10 µL之100 µM ThT (於PBS中之250 mM甘胺酸中(二者均來自Sigma-Aldrich))混合。在Tecan讀數儀(激發:440 nm;發射:485 nm)上以一式一份或一式兩份量測螢光(相對螢光單位;RFU)。然後計算K18解聚百分比,且使用GraphPad Prism第5版(GraphPad軟體)假設一個結合位點擬合模型來測定半最大有效濃度(EC50 )。 Bioassay Description Full-length Tau (flTau) depolymerization assay by thioflavin T (ThT) The longest isoform of human Tau (2N4R; 441 amino acids) was expressed in bacteria and purified. For analysis of Tau depolymerization by ThT, 35 µM recombinant full-length (fl) Tau in PBS was incubated at 37°C in the presence of 50 µM heparin (Sigma-Aldrich) and 10 mM DTT (Sigma-Aldrich) at 750 RPM. Aggregate under shaking for 24 hours. Compounds were dissolved in anhydrous dimethylsulfoxide (DMSO, Sigma-Aldrich) to a concentration of 10 mM. Serial dilutions of flTau aggregates and compounds were mixed together in PBS (50 µL volume) to a final concentration of 2 µM flTau aggregates and 160 µM to 0.04 µM compound. The mixture was incubated at room temperature (RT) for 30 minutes, then 40 µL of this mixture was transferred to a black 384-well assay plate (Perkin-Elmer) and mixed with 10 µL of 100 µM ThT (250 mM Glycerol in PBS). Amino acids (both from Sigma-Aldrich) were mixed. Fluorescence (relative fluorescence units; RFU) was measured in duplicate or in duplicate on a Tecan reader (excitation: 440 nm; emission: 485 nm). The percent flTau depolymerization was then calculated and the half-maximum effective concentration ( EC50 ) was determined assuming a binding site fitting model using GraphPad Prism version 5 (GraphPad Software). Tau K18 Depolymerization Analysis by ThT Tau K18 fragments covering amino acids 244 to 372 of the longest isoform of human Tau441 (2N4R) were expressed in bacteria and purified, or purchased from SignalChem . For K18 depolymerization assay by ThT, 35 µM recombinant K18 in PBS was aggregated at 37°C in the presence of 50 µM heparin (Sigma-Aldrich) and 10 mM DTT (Sigma-Aldrich) with shaking at 750 RPM 24 hours. Compounds were dissolved in anhydrous dimethylsulfoxide (DMSO, Sigma-Aldrich) to a concentration of 10 mM. Serial dilutions of K18 aggregates and compounds were mixed together in PBS (volume 50 µL) to a final concentration of 2 μM K18 aggregates and 160 µM to 0.04 µM compound. The mixture was incubated at room temperature (RT) for 30 minutes, then 40 µL of this mixture was transferred to a black 384-well assay plate (Perkin-Elmer) and mixed with 10 µL of 100 µM ThT (250 mM Glycerol in PBS). Amino acids (both from Sigma-Aldrich) were mixed. Fluorescence (relative fluorescence units; RFU) was measured in duplicate or in duplicate on a Tecan reader (excitation: 440 nm; emission: 485 nm). The percent K18 depolymerization was then calculated and the half-maximum effective concentration ( EC50 ) was determined assuming a binding site fitting model using GraphPad Prism version 5 (GraphPad Software).

量測以下實例化合物: 7 圖例:+++ EC50 < 10 uM;++ EC50 10<x<25 uM;+ EC50 25<x<50 uM。The following example compounds were measured: Table 7 Legend: +++ EC 50 < 10 uM; ++ EC 50 10<x<25 uM; + EC 50 25<x<50 uM.

細胞內 Tau 聚集之降低 於完全培養基[補充有2.5 µg/ml G418 (Sigma-Aldrich)選擇抗生素之DMEM-F12 4.5 g/L Glutamax (Invitrogen)、15% FBS (Biochrom)、1% Peni/Strep (Invitrogen)]中培養過表現攜帶P301L突變之人類Tau之全長形式之人類神經胚細胞瘤細胞系。在實驗前一天,將5×105 個細胞/孔平鋪於6孔板中之3 mL完全培養基中。第二天,使細胞與DMSO或5 µM之本發明化合物一起在37℃下再培育24 h。培育後,使細胞胰蛋白酶化,重新懸浮於100 µl均質緩衝液[25 mM Tris-HCl pH 7.4、150 mM NaCl、1 mM EDTA、1 mM含有磷酸酶抑制劑(30 mM NaF、0.2 mM Na3 VO4 、1 nM岡田酸(Okadaic acid)、1 mM PMSF、5 mM Na4 P2 O7 )之EGTA及蛋白酶抑制劑混合液(CompleteTM , Roche)]中,且然後利用三次快速冷凍及解凍循環進行物理溶解。然後在AlphaLISA分析中直接測試樣品。 Intracellular Tau aggregation is reduced compared to complete medium [DMEM-F12 supplemented with 2.5 µg/ml G418 (Sigma-Aldrich) selection antibiotic 4.5 g/L Glutamax (Invitrogen), 15% FBS (Biochrom), 1% Peni/Strep ( A human neuroblastoma cell line expressing the full-length form of human Tau carrying the P301L mutation was cultured in Invitrogen. The day before the experiment, 5 × 10 5 cells/well were plated in 3 mL of complete culture medium in a 6-well plate. The next day, cells were incubated with DMSO or 5 µM of a compound of the invention for an additional 24 h at 37°C. After incubation, cells were trypsinized and resuspended in 100 µl of homogenization buffer [25 mM Tris-HCl pH 7.4, 150 mM NaCl, 1 mM EDTA, 1 mM containing phosphatase inhibitors (30 mM NaF, 0.2 mM Na VO 4 , 1 nM Okadaic acid, 1 mM PMSF, 5 mM Na 4 P 2 O 7 ) in EGTA and protease inhibitor mixture (Complete TM , Roche)], and then rapidly frozen and thawed three times Cycle for physical dissolution. The samples are then tested directly in the AlphaLISA assay.

藉由AlphaLisa使用以下抗體對來量化磷酸化、聚集及總Tau: ·   HT7-接受者珠粒+生物素(BT)-Tau13-供體珠粒:   總Tau ·   HT7-接受者珠粒+生物素(BT)-HT7-供體珠粒:     聚集人類TauPhosphorylation, aggregation and total Tau were quantified by AlphaLisa using the following antibody pairs: · HT7-Receiver Beads + Biotin (BT)-Tau13-Donor Beads: Total Tau · HT7-recipient beads + biotin (BT)-HT7-donor beads: Aggregation of human Tau

使用EZ-Link® NHS-PEO固相生物素化套組(Thermo Scientific)使Tau13 (Abcam)生物素化,同時HT7-生物素係來自商業來源(Thermo Scientific)。Tau13 (Abcam) was biotinylated using the EZ-Link® NHS-PEO Solid Phase Biotinylation Kit (Thermo Scientific), while the HT7-biotin system was from a commercial source (Thermo Scientific).

對於每一抗體對,接受者珠粒及生物素化抗體之濃度係最佳化的。所有樣品均首先於PBS中之稀釋系列中進行測試以鑑別每一樣品及分析之線性範圍及最佳稀釋度。對於最終方案,將以下試劑添加於384孔白色OptiPlate (PerkinElmer)中: ·   5 µL測試稀釋樣品 ·   20 µL以下終濃度之混合物生物素-mAb接受者珠粒: ·   1.25 nM HT7-BT與10 µg/ml HT7-Acc珠粒之組合 ·   5 nM Tau13-BT與2.5 µg/ml HT7-Acc珠粒之組合For each antibody pair, the concentrations of acceptor beads and biotinylated antibodies were optimized. All samples were first tested in a dilution series in PBS to identify the linear range and optimal dilution for each sample and analysis. For the final protocol, the following reagents were added to a 384-well white OptiPlate (PerkinElmer): · 5 µL test dilution sample · Mixed biotin-mAb receptor beads at a final concentration below 20 µL: · Combination of 1.25 nM HT7-BT and 10 µg/ml HT7-Acc beads · Combination of 5 nM Tau13-BT and 2.5 µg/ml HT7-Acc beads

將此混合物在室溫下培育1 h之後,在黑暗中添加25 µL之25 µg/mL鏈黴抗生物素蛋白(Streptavidin)供體珠粒(Perkin Elmer)。在30 min培育後,使用EnSpire Alpha儀器及EnSpire工作站第3.00版來分析板。將聚集Tau之數據正規化為總Tau,且然後表示為DMSO處理細胞之百分比。After the mixture was incubated for 1 h at room temperature, 25 µL of 25 µg/mL Streptavidin donor beads (Perkin Elmer) was added in the dark. After 30 min incubation, plates were analyzed using the EnSpire Alpha instrument and EnSpire Workstation version 3.00. Data for aggregated Tau were normalized to total Tau and then expressed as percentage of DMSO-treated cells.

量測以下實例化合物: 8 圖例:+++%>50;++% 50<x<25;+% 25<x<10。The following example compounds were measured: Table 8 Legend: +++%>50; ++% 50<x<25; +% 25<x<10.

本發明化合物之活體內效能 測試實例 12 及實例 45 之活體內研究設計 雙轉基因rTg4510小鼠在tet誘導型CaMKII啟動子控制下表現攜帶P301L (Tau4R0N-P301L)突變之全長人類Tau (Ramsden等人,J.Neurosci., 2005 • 25(46):10637-10647)。使用僅表現四環素控制之反式激活蛋白(tTA)之單一轉基因小鼠作為基因型對照。該研究包含4個治療組(對於tTa組n= 15隻雌性且n = 11隻雌性小鼠/組用於治療),其具有以下組分配(參見表9)。自5月齡開始,藉由胃管灌食每日一次或每日兩次投與化合物或媒劑對照達1個月。所有小鼠均在飼料中接受200 ppm去氧羥四環素達3週,加上在前兩天在飲用水中接受於4%蔗糖中1.5 mg/mL之負荷劑量。投藥時之去氧羥四環素投與係在第2週開始且持續整個投藥期。 In vivo efficacy test of the compounds of the present invention . In vivo study design of Example 12 and Example 45. Double transgenic rTg4510 mice express full-length human Tau carrying the P301L (Tau4RON-P301L) mutation under the control of the tet-inducible CaMKII promoter (Ramsden et al., J. Neurosci., 2005 • 25(46):10637-10647). A single transgenic mouse expressing only the tetracycline-controlled transactivator (tTA) was used as a genotype control. The study contained 4 treatment groups (n=15 females for tTa group and n=11 female mice/group for treatment) with the following group allocation (see Table 9). Beginning at 5 months of age, compound or vehicle control was administered via gastric tube feeding once daily or twice daily for 1 month. All mice received 200 ppm deoxytetracycline in the feed for 3 weeks, plus a loading dose of 1.5 mg/mL in 4% sucrose in the drinking water for the first two days. Dosing of deoxytetracycline began at week 2 and continued throughout the dosing period.

對所有動物均實施腦脊液(CSF)分析,而對10隻動物/組實施錯誤摺疊之Tau (MC1)、總小神經膠質細胞(Iba1)及吞噬小神經膠質細胞(CD68)之組織學。 9 測試實例12及實例45之活體內研究設計 (a)             媒劑:0.5% w/v羥丙基甲基纖維素4000 cps;0.5% w/v Tween 80Cerebrospinal fluid (CSF) analysis was performed on all animals, and histology of misfolded Tau (MC1), total microglia (Iba1), and phagocytic microglia (CD68) was performed on 10 animals/group. Table 9 : In vivo study design for Test Example 12 and Example 45 (a) Vehicle: 0.5% w/v Hydroxypropyl Methylcellulose 4000 cps; 0.5% w/v Tween 80

腦脊液 (CSF) 收集及分析 藉由標準可注射麻醉使小鼠深度麻醉,且經心臟灌注冷的PBS。利用手術剪去除來自頸部之皮毛及皮膚(小鼠未斷頭)。經由鈍性分離以最少出血小心地去除圍繞顱底及腦幹之組織。一旦腦膜暴露在顱底之枕骨大孔區域中,則使27ga蝶針保持垂直於顱骨且橫向插入至腦脊髓液(CSF)空間中。將樣品快速浸入液氮中且在-80℃下儲存直至藉由AlphaLISA實施生物化學分析。使用以下抗體對量化CSF Tau中之總人類Tau:HT7-接受者珠粒+生物素(BT)-Tau13-供體珠粒。使用EZ-Link® NHS-PEO固相生物素化套組(Thermo Scientific)使Tau13 (Abcam)抗體生物素化。對於最終方案,將以下試劑添加於384孔白色OptiPlate (PerkinElmer)中: ·   5 µL測試稀釋樣品 ·   20 µL混合物生物素-mAb 接受者珠粒,終濃度為:0.6 nM Tau13-BT與2.5 µg/ml HT7-Acc珠粒之組合 Cerebrospinal fluid (CSF) collection and analysis Mice were deeply anesthetized by standard injectable anesthesia and perfused transcardially with cold PBS. Use surgical scissors to remove the fur and skin from the neck (the mice are not decapitated). The tissue surrounding the skull base and brainstem is carefully removed via blunt dissection with minimal bleeding. Once the meninges are exposed in the foramen magnum region of the skull base, a 27ga sphenoidal needle is held perpendicular to the skull and inserted laterally into the cerebrospinal fluid (CSF) space. Samples were quickly immersed in liquid nitrogen and stored at -80°C until biochemical analysis by AlphaLISA. Total human Tau in CSF Tau was quantified using the following antibody pair: HT7-recipient beads + biotin (BT)-Tau13-donor beads. Tau13 (Abcam) antibody was biotinylated using the EZ-Link® NHS-PEO Solid Phase Biotinylation Kit (Thermo Scientific). For the final protocol, add the following reagents to a 384-well white OptiPlate (PerkinElmer): 5 µL of test dilution sample 20 µL of mixed biotin-mAb receptor beads, final concentration: 0.6 nM Tau13-BT with 2.5 µg/ ml HT7-Acc Beads Combination

將此混合物在室溫下培育1 h之後,在黑暗中添加25 µL之25 µg/mL鏈黴抗生物素蛋白供體珠粒(Perkin Elmer)。在30分鐘培育後,使用EnSpire Alpha儀器及EnSpire工作站第3.00版來分析板。藉由單因子ANOVA、之後進行事後比較來分析數據,且所示統計學係指與媒劑治療之rTg4510小鼠相比之差異。After the mixture was incubated for 1 h at room temperature, 25 µL of 25 µg/mL streptavidin donor beads (Perkin Elmer) was added in the dark. After a 30 minute incubation, the plates were analyzed using the EnSpire Alpha instrument and EnSpire Workstation version 3.00. Data were analyzed by one-way ANOVA followed by post hoc comparisons, and statistics shown refer to differences compared to vehicle-treated rTg4510 mice.

如圖1中所示,實例12及實例45二者均降低CSF中之Tau含量。As shown in Figure 1, both Example 12 and Example 45 reduced Tau content in CSF.

經實例 12 及實例 45 治療之 rTg4510 小鼠之組織學評估 在灌注後,然後將腦移除且半矢狀地進行半切。將右半球於PBS中之4%多聚甲醛中在室溫下固定3小時,藉由浸入4℃下之15%蔗糖中冷藏保存3天,且準備用於冷凍切片。然後將經固定之半球在低溫模具中在OCT介質上於乾冰中冷凍,且用Leica CM3050冷凍切片機進行矢狀冷凍切片(10 mm厚度)。自大約12個內外側位準收集來自10隻小鼠/組之切片。使用來自七個矢狀水準/隻動物之系統隨機切片組來量化Iba1及CD68免疫反應性,而使用來自內外側位準4之1個切片/動物來評價Tau錯誤摺疊(MC1單株抗體)。將冷凍切片自-20℃移除且在室溫下風乾25分鐘。利用pap筆液體阻斷劑將腦組織圈起,且在室溫下於PBS中洗滌一次5分鐘及一次10分鐘。在室溫下利用10%正常山羊血清(NGS)及於PBS中之0.25% Triton X-100實施封阻以及可滲透化處理2小時。然後將切片用紙吸乾且與於含有5% NGS及0.25% Triton X-100之PBS中稀釋1:1000之小鼠單株MC1抗體一起在4℃下於濕度箱中培育過夜。將切片在室溫下於PBS中洗滌三次10分鐘,且與於PBS中稀釋1:1000之經Cy3標記之山羊抗小鼠IgG (H+L)二級抗體(Jackson)一起在室溫下避光培育30分鐘。於PBS中洗滌三次10分鐘之後,使切片與0.1%蘇丹黑(Sudan Black)(Sigma)於70%乙醇中之溶液一起在室溫下培育30秒以減少組織之自發螢光。將切片於PBS中洗滌三次10分鐘,使用含有DAPI之ProLong Gold抗褪色試劑(Molecular Probes)封固並蓋上蓋玻片。使用數位載玻片掃描儀(Panoramic 250 Flash, 3D Histech Ltd.)使切片成像,且使用影像可視化軟體Visiopharm來量化。 Histological evaluation of rTg4510 mice treated with Example 12 and Example 45 After perfusion, the brains were then removed and hemisectioned hemisagittally. The right hemisphere was fixed in 4% paraformaldehyde in PBS for 3 hours at room temperature, refrigerated by immersion in 15% sucrose at 4°C for 3 days, and prepared for cryosectioning. The fixed hemispheres were then frozen in dry ice on OCT media in a cryo-mold and sagittal cryosections (10 mm thickness) were performed using a Leica CM3050 cryomicrotome. Sections from 10 mice/group were collected from approximately 12 medial and lateral planes. Systematic randomized sets of sections from seven sagittal levels/animal were used to quantify Iba1 and CD68 immunoreactivity, while 1 section/animal from medial and lateral levels 4 was used to evaluate Tau misfolding (MC1 monoclonal antibody). Frozen sections were removed from -20°C and air-dried at room temperature for 25 minutes. Brain tissue was circled using Pap Pen Liquid Blocker and washed once for 5 minutes and once for 10 minutes in PBS at room temperature. Block and permeabilize using 10% normal goat serum (NGS) and 0.25% Triton X-100 in PBS for 2 hours at room temperature. The sections were then blotted dry with paper and incubated overnight in a humidity chamber at 4°C with mouse monoclonal MC1 antibody diluted 1:1000 in PBS containing 5% NGS and 0.25% Triton X-100. Sections were washed three times for 10 min in PBS at room temperature and incubated with Cy3-labeled goat anti-mouse IgG (H+L) secondary antibody (Jackson) diluted 1:1000 in PBS at room temperature. Incubate in light for 30 minutes. After washing three times in PBS for 10 minutes, the sections were incubated with a solution of 0.1% Sudan Black (Sigma) in 70% ethanol for 30 seconds at room temperature to reduce tissue autofluorescence. The sections were washed three times in PBS for 10 minutes, mounted using ProLong Gold anti-fade reagent (Molecular Probes) containing DAPI, and covered with coverslips. Sections were imaged using a digital slide scanner (Panoramic 250 Flash, 3D Histech Ltd.) and quantified using the image visualization software Visiopharm.

在上皮質層中之額葉皮質中分析MC1染色。繪製額葉皮質周圍對應於20×放大率之視野之一個所關注區域(ROI)。在Visiopharm中使用MC1染色之預定臨限值分析此ROI,其中像素強度臨限值高於30 (8位元圖片)且排除小於20 μm2 之所有檢測到之物體。藉由單因子ANOVA、之後事後測試來分析數據。MC1 staining was analyzed in the frontal cortex among the upper cortical layers. A region of interest (ROI) corresponding to a field of view at 20× magnification was drawn around the frontal cortex. This ROI was analyzed in Visiopharm using predetermined thresholds for MC1 staining, where the pixel intensity threshold was above 30 (8-bit image) and all detected objects smaller than 20 μm were excluded. Data were analyzed by one-way ANOVA followed by post hoc tests.

如圖2中所示,實例12及實例45二者均降低MC1陽性面積。該等數據指示,在rTg4510中利用實例12及實例45二者治療均降低此攻擊性Tau轉基因模型中錯誤摺疊之Tau之含量。As shown in Figure 2, both Example 12 and Example 45 reduced MC1 positive area. These data indicate that treatment with both Example 12 and Example 45 in rTg4510 reduced the amount of misfolded Tau in this aggressive Tau transgenic model.

為評價所測試化合物對Tau之上述效應是否亦對神經發炎標記物具有效應,對大鼠抗小鼠CD68純系FA-11 (BD Biosciences)、山羊多株抗Iba1抗體(Abcam)之切片進行標記,並用DAPI複染。使用高度交叉吸收之螢光標記之二級抗體(Thermo Fisher)使抗體結合可視化。將抗體稀釋於抗體稀釋液(Dako)中,在初步培育之前利用M.O.M.血清(Vector)阻斷非特異性內源性IgG結合。使用LED (Colibri2)照明及靈敏之Orca Flash 4.0單色照相機,在由ZEN軟體驅動之Axio Scan Z1載玻片掃描儀上以20×放大率(平場複消色差物鏡)使封固切片整體成像。藉由單因子ANOVA、之後進行事後比較來分析數據,且所示統計學係指與媒劑治療之rTg4510小鼠相比之差異。In order to evaluate whether the above-mentioned effects of the tested compounds on Tau also have effects on neuroinflammation markers, sections were labeled with rat anti-mouse CD68 pure line FA-11 (BD Biosciences) and goat multi-strain anti-Iba1 antibodies (Abcam). And counterstained with DAPI. Antibody binding was visualized using highly cross-absorbing fluorescently labeled secondary antibodies (Thermo Fisher). Antibodies were diluted in Antibody Diluent (Dako) and non-specific endogenous IgG binding was blocked using M.O.M. serum (Vector) before initial incubation. Mounted sections were imaged in their entirety at 20× magnification (plan apochromatic objective) on an Axio Scan Z1 slide scanner driven by ZEN software using LED (Colibri2) illumination and a sensitive Orca Flash 4.0 monochrome camera. Data were analyzed by one-way ANOVA followed by post hoc comparisons, and statistics shown refer to differences compared to vehicle-treated rTg4510 mice.

如圖3A中所示,實例12及實例45二者均降低小神經膠質細胞增生,如Iba1免疫反應性面積所量測。此外,實例12及45二者均減少對CD68呈陽性之高度活化之吞噬小神經膠質細胞(圖3B)。總之,該等數據指示,在rTg4510中利用實例12及實例45二者治療均降低此攻擊性Tau轉基因模型中神經發炎之程度。As shown in Figure 3A, both Example 12 and Example 45 reduced microgliosis as measured by Iba1 immunoreactive area. Furthermore, both Examples 12 and 45 reduced highly activated phagocytic microglia that were positive for CD68 (Figure 3B). Taken together, these data indicate that treatment with both Example 12 and Example 45 in rTg4510 reduced the extent of neuroinflammation in this aggressive Tau transgenic model.

減少來自人類 AD PSP 腦切片之神經纖維纏結 (NFT) 中之 Tau 錯誤摺疊 自商業經銷商(Tissue Solutions, UK)獲得來自一名AD病例及兩名PSP病例之新鮮冷凍組織切片。將組織切片與100 µM之實例45或DMSO一起於濕度箱中在室溫下培育大約60小時。在於PBS中將培育切片洗滌三次之後,在4℃下利用多聚甲醛(PFA; Sigma)固定10 min,且然後於PBS中再洗滌三次。然後在室溫下於封阻緩衝液(PBS、10%純淨山羊血清(NGS)、0.25% Triton X)中將切片可滲透化處理1小時。封阻之後,將切片與Tau構形抗體(MC1 單株抗體)一起於封阻緩衝液(5% NGS, 0.25% Triton X)中在4℃下培育過夜。第二天,於PBS中將切片洗滌三次5 min,且與Cy3-偶聯之AffiniPure山羊抗小鼠抗體(Jackson laboratories)一起在室溫下培育1h。於PBS中洗滌三次5 min將過量抗體洗出。為減少自發螢光,將切片與溶解於70%乙醇中之0.1%蘇丹黑(Sigma)一起在室溫下培育8 min。最後,將切片於PBS中洗滌5次5 min,且使用含有DAPI之ProLong Gold抗褪色試劑(Invitrogen)在蓋玻片下封固。然後將切片在室溫下乾燥24小時,之後使用數位載玻片掃描儀(Pannoramic P250 Flash III, 3D Histech Ltd.)成像,且使用Visiopharm軟體量化NFT中之Tau錯誤摺疊。如圖4中所示,實例45使NFT中之Tau錯誤摺疊降低大約30%,其對AD及PSP人類腦樣品具有類似功效。Reduction of Tau misfolding in neurofibrillary tangles (NFTs) from human AD and PSP brain sections Fresh frozen tissue sections from one AD case and two PSP cases were obtained from a commercial distributor (Tissue Solutions, UK). Incubate tissue sections with 100 µM of Example 45 or DMSO in a humidity chamber at room temperature for approximately 60 hours. After the cultured sections were washed three times in PBS, they were fixed with paraformaldehyde (PFA; Sigma) for 10 min at 4°C and then washed three more times in PBS. Sections were then permeabilized in blocking buffer (PBS, 10% pure goat serum (NGS), 0.25% Triton X) for 1 hour at room temperature. After blocking, sections were incubated with Tau conformational antibody (MC1 monoclonal antibody) in blocking buffer (5% NGS, 0.25% Triton X) overnight at 4°C. The next day, sections were washed three times for 5 min in PBS and incubated with Cy3-conjugated AffiniPure goat anti-mouse antibody (Jackson laboratories) for 1 h at room temperature. Wash three times in PBS for 5 minutes to wash out excess antibody. To reduce autofluorescence, sections were incubated with 0.1% Sudan black (Sigma) dissolved in 70% ethanol for 8 min at room temperature. Finally, the sections were washed 5 times in PBS for 5 min and mounted under coverslips using ProLong Gold anti-fade reagent (Invitrogen) containing DAPI. The sections were then dried at room temperature for 24 hours before being imaged using a digital slide scanner (Pannoramic P250 Flash III, 3D Histech Ltd.) and using Visiopharm software to quantify Tau misfolding in NFTs. As shown in Figure 4, Example 45 reduced Tau misfolding in NFTs by approximately 30%, with similar efficacy on AD and PSP human brain samples.

在人類 AD 腦切片中利用 3 H- 實例 45 進行離體高解析度放射自顯影 自商業經銷商(Tissue Solutions, UK)獲得來自一名AD病例之新鮮冷凍組織切片。對於放射自顯影,將腦切片在4℃下利用4% PFA (Sigma) 固定15分鐘。將切片與單獨之20 nM [3 H]-實例45或連同5 μM非放射性實例45一起在室溫培育1小時。然後如下洗滌切片:首先,於冰冷緩衝液中洗滌1分鐘,然後於冰冷70%乙醇中洗滌兩次1分鐘、於冰冷緩衝液中洗滌1分鐘且最後於冰冷蒸餾水中簡短沖洗。隨後使切片在氣流下乾燥1小時,且然後於不透光之載玻片儲存盒中在4℃下暴露於Ilford核乳膠類型K5 (Agar Scientific)達5天。根據製造商說明書,一直在用安全燈照明之暗室中暴露於乳膠,且將乳膠碎片在等體積之40℃預先加熱水中熔融。5天後,根據製造商說明書使切片顯影。使用ProLong Gold抗褪色試劑(Invitrogen)使切片封固且使用數位載玻片掃描儀(Pannoramic P250 Flash III, 3D Histech Ltd.)成像。Ex vivo high-resolution autoradiography in human AD brain sections using 3 H- Example 45. Fresh frozen tissue sections from an AD case were obtained from a commercial distributor (Tissue Solutions, UK). For autoradiography, brain sections were fixed with 4% PFA (Sigma) for 15 min at 4°C. Sections were incubated with 20 nM [ 3H ]-Example 45 alone or together with 5 μM non-radioactive Example 45 for 1 hour at room temperature. The sections were then washed as follows: first in ice-cold buffer for 1 minute, then twice in ice-cold 70% ethanol for 1 minute, in ice-cold buffer for 1 minute and finally with a brief rinse in ice-cold distilled water. Sections were then allowed to dry under airflow for 1 hour and then exposed to Ilford core latex type K5 (Agar Scientific) for 5 days at 4°C in a light-tight slide storage box. Exposure to latex was carried out in a dark room illuminated by a safety light according to the manufacturer's instructions, and latex fragments were melted in an equal volume of 40°C preheated water. After 5 days, sections were developed according to the manufacturer's instructions. Sections were mounted using ProLong Gold anti-fade reagent (Invitrogen) and imaged using a digital slide scanner (Pannoramic P250 Flash III, 3D Histech Ltd.).

如圖5中所示,實例45顯示人類AD腦樣品中Tau NFT上之特異性靶標接合。As shown in Figure 5, Example 45 shows specific target engagement on Tau NFTs in human AD brain samples.

1 在經實例12及實例45治療之rTg4510小鼠中量化之CSF中之Tau。 2 經實例12及實例45治療之rTg4510小鼠中之Tau錯誤摺疊量化。 3 經實例12及實例45治療之rTg4510小鼠中之Iba1 (A)及CD68 (B)量化。 4 實例45降低人類AD及PSP腦切片中NFT中之Tau錯誤摺疊。 5 使用3 H-實例45 (A)及3 H-實例45及冷實例45 (B)對人類AD腦切片之高解析度放射自顯影。 Figure 1 : Tau in CSF quantified in rTg4510 mice treated with Example 12 and Example 45. Figure 2 : Quantification of Tau misfolding in rTg4510 mice treated with Example 12 and Example 45. Figure 3 : Quantification of Ibal (A) and CD68 (B) in rTg4510 mice treated with Example 12 and Example 45. Figure 4 Example 45 reduces Tau misfolding in NFTs in human AD and PSP brain slices. Figure 5 High-resolution autoradiography of human AD brain sections using 3H -Example 45 (A) and 3H -Example 45 and Cold Example 45 (B).

Claims (29)

一種式(I)化合物及其所有立體異構物、外消旋混合物、互變異構物、醫藥上可接受之鹽、水合物及多形體,
Figure 108107135-A0305-02-0211-1
其中 A係選自由以下組成之群:
Figure 108107135-A0305-02-0211-2
Figure 108107135-A0305-02-0211-3
Figure 108107135-A0305-02-0211-4
Figure 108107135-A0305-02-0211-5
Figure 108107135-A0305-02-0211-6
Figure 108107135-A0305-02-0211-7
Figure 108107135-A0305-02-0211-8
Figure 108107135-A0305-02-0211-9
Figure 108107135-A0305-02-0211-10
,其中
Figure 108107135-A0305-02-0211-11
Figure 108107135-A0305-02-0211-12
Figure 108107135-A0305-02-0211-13
Figure 108107135-A0305-02-0211-14
Figure 108107135-A0305-02-0211-15
Figure 108107135-A0305-02-0211-16
Figure 108107135-A0305-02-0211-17
Figure 108107135-A0305-02-0211-18
Figure 108107135-A0305-02-0211-19
可在任一可用位置連接至Q,其中
Figure 108107135-A0305-02-0211-20
Figure 108107135-A0305-02-0211-21
Figure 108107135-A0305-02-0211-22
Figure 108107135-A0305-02-0211-23
經一或多個取代基R j 取代,其中當A為
Figure 108107135-A0305-02-0212-24
時,R j 不為-CN、鹵素或- CF3;其中當A為
Figure 108107135-A0305-02-0212-25
Figure 108107135-A0305-02-0212-26
時,R j 不為鹵素,且其中
Figure 108107135-A0305-02-0212-27
Figure 108107135-A0305-02-0212-28
Figure 108107135-A0305-02-0212-29
Figure 108107135-A0305-02-0212-30
可視情況經一或多個取代基R取代;B係選自由O及NRa組成之群;EV獨立地選自由以下組成之群:N、NR5、O及S;G係選自由以下組成之群:苯環及吡啶環;J係選自由O及N-R1組成之群;Q係選自由N及C-R1組成之群;Y係選自由CZ及N組成之群,條件係當Y係N且Y 1 Y 2 Y 3 係CZ時,B係N-烷基或O;Y 1 係選自由CZ及N組成之群;Y 2 係選自由CZ及N組成之群;Y 3 係選自由CZ及N組成之群;Z獨立地選自由以下組成之群:H、鹵素、O-烷基、烷基及CN; R獨立地選自由
Figure 108107135-A0305-02-0212-33
及-NR3R4組成之群;R a 係選自由H及烷基組成之群;R b R c R d R e R f R g 獨立地選自由H及烷基組成之群,或R b R c R d R e R f R g 中之任兩者可連結形成3員至8員環; R j 獨立地選自由以下組成之群:-鹵素、-O-烷基、-CF3、-CN、- NR3R4
Figure 108107135-A0305-02-0213-34
Figure 108107135-A0305-02-0213-35
,其中含C1-2碳原子之橋可存在於a碳原子與c或d碳原子之間或其中含C1-2碳原子之橋可存在於b碳原子與c或d碳原子之間;R 1 係選自由H及烷基組成之群;R 2 獨立地選自由烷基、F及=O組成之群,其中該烷基可視情況經鹵素、-OH或-O-烷基取代,且其中若兩個R 2 係孿位的,則其可連結形成3員至6員環;R 3 R 4 獨立地選自由H及烷基組成之群,其中該烷基可視情況經鹵素、-OH或-O-烷基取代;R 5 係選自由H及烷基組成之群;n係0、1、2、3或4,rs獨立地係0、1、2或3;及tu獨立地係1、2或3。
A compound of formula (I) and all stereoisomers, racemic mixtures, tautomers, pharmaceutically acceptable salts, hydrates and polymorphs thereof,
Figure 108107135-A0305-02-0211-1
Among them, series A is selected from the group consisting of:
Figure 108107135-A0305-02-0211-2
,
Figure 108107135-A0305-02-0211-3
,
Figure 108107135-A0305-02-0211-4
,
Figure 108107135-A0305-02-0211-5
,
Figure 108107135-A0305-02-0211-6
,
Figure 108107135-A0305-02-0211-7
,
Figure 108107135-A0305-02-0211-8
,
Figure 108107135-A0305-02-0211-9
and
Figure 108107135-A0305-02-0211-10
,in
Figure 108107135-A0305-02-0211-11
,
Figure 108107135-A0305-02-0211-12
,
Figure 108107135-A0305-02-0211-13
,
Figure 108107135-A0305-02-0211-14
,
Figure 108107135-A0305-02-0211-15
,
Figure 108107135-A0305-02-0211-16
,
Figure 108107135-A0305-02-0211-17
,
Figure 108107135-A0305-02-0211-18
and
Figure 108107135-A0305-02-0211-19
Can be connected to Q at any available location, where
Figure 108107135-A0305-02-0211-20
,
Figure 108107135-A0305-02-0211-21
,
Figure 108107135-A0305-02-0211-22
and
Figure 108107135-A0305-02-0211-23
Substituted with one or more substituents R j , where when A is
Figure 108107135-A0305-02-0212-24
When , R j is not -CN, halogen or - CF 3 ; where A is
Figure 108107135-A0305-02-0212-25
or
Figure 108107135-A0305-02-0212-26
When , R j is not halogen, and where
Figure 108107135-A0305-02-0212-27
,
Figure 108107135-A0305-02-0212-28
,
Figure 108107135-A0305-02-0212-29
and
Figure 108107135-A0305-02-0212-30
Optionally substituted by one or more substituents R ; B is selected from the group consisting of O and NR a ; E and V are independently selected from the group consisting of: N, NR 5 , O and S; G is selected from the following The group consisting of: benzene ring and pyridine ring; J is selected from the group consisting of O and NR 1 ; Q is selected from the group consisting of N and CR 1 ; Y is selected from the group consisting of CZ and N, provided that Y is When N and Y 1 , Y 2 and Y 3 are CZ, B is N-alkyl or O; Y 1 is selected from the group consisting of CZ and N; Y 2 is selected from the group consisting of CZ and N; Y 3 is selected from the group consisting of CZ and N Selected from the group consisting of CZ and N; Z is independently selected from the group consisting of: H, halogen, O-alkyl, alkyl, and CN; R is independently selected from
Figure 108107135-A0305-02-0212-33
and -NR 3 R 4 ; R a is selected from the group consisting of H and alkyl groups; R b , R c , R d , R e , R f and R g are independently selected from the group consisting of H and alkyl groups group, or any two of R b , R c , R d , Re , R f and R g can be connected to form a 3- to 8-membered ring; R j is independently selected from the group consisting of : - halogen, - O-alkyl, -CF 3 , -CN, - NR 3 R 4 ,
Figure 108107135-A0305-02-0213-34
and
Figure 108107135-A0305-02-0213-35
, where a bridge containing C 1-2 carbon atoms can exist between a carbon atom and c or d carbon atom or where a bridge containing C 1-2 carbon atoms can exist between b carbon atom and c or d carbon atom ; R 1 is selected from the group consisting of H and alkyl; R 2 is independently selected from the group consisting of alkyl, F and =O, wherein the alkyl is optionally substituted by halogen, -OH or -O-alkyl, And if two R 2 are twin-positioned, they can be connected to form a 3- to 6-membered ring; R 3 and R 4 are independently selected from the group consisting of H and an alkyl group, wherein the alkyl group may be optionally treated with halogen, -OH or -O-alkyl substitution; R 5 is selected from the group consisting of H and alkyl; n is 0, 1, 2, 3 or 4, r and s are independently 0, 1, 2 or 3; and t and u are independently 1, 2 or 3.
如請求項1之化合物及其所有立體異構物、外消旋混合物、互變異構物、醫藥上可接受之鹽、水合物及多形體,其中該化合物係式(Ia)化合物:
Figure 108107135-A0305-02-0214-36
其中ABR b R c R d R e R f R g Y及Z係如請求項1中所定義。
For example, the compound of claim 1 and all stereoisomers, racemic mixtures, tautomers, pharmaceutically acceptable salts, hydrates and polymorphs thereof, wherein the compound is a compound of formula (Ia):
Figure 108107135-A0305-02-0214-36
Among them , A , B , Rb , Rc , Rd , Re , Rf , Rg , Y and Z are as defined in claim 1 .
如請求項1之化合物及其所有立體異構物、外消旋混合物、互變異構物、醫藥上可接受之鹽、水合物及多形體,其係式(Ib)化合物:
Figure 108107135-A0305-02-0214-37
其中ABR b R c R d R e R f R g Z係如請求項1中所定義。
For example, the compound of claim 1 and all stereoisomers, racemic mixtures, tautomers, pharmaceutically acceptable salts, hydrates and polymorphs thereof are compounds of formula (Ib):
Figure 108107135-A0305-02-0214-37
Among them, A , B , Rb , Rc, Rd, Re, Rf , Rg and Z are as defined in claim 1 .
如請求項1至3中任一項之化合物及其所有立體異構物、外消旋混合物、互變異構物、醫藥上可接受之鹽、水合物及多形體,其中A
Figure 108107135-A0305-02-0214-38
,其中
Figure 108107135-A0305-02-0214-39
可在任一可用位置連接至Q或N,且其中
Figure 108107135-A0305-02-0215-40
可視情況經一或多個取代基R取代。
For example, the compound of any one of claims 1 to 3 and all stereoisomers, racemic mixtures, tautomers, pharmaceutically acceptable salts, hydrates and polymorphs thereof, wherein A is
Figure 108107135-A0305-02-0214-38
,in
Figure 108107135-A0305-02-0214-39
Can be connected to Q or N at any available location, and where
Figure 108107135-A0305-02-0215-40
Optionally substituted with one or more substituents R.
如請求項1至3中任一項之化合物及其所有立體異構物、外消旋混合物、互變異構物、醫藥上可接受之鹽、水合物及多形體,其係式(Ic)化合物:
Figure 108107135-A0305-02-0215-41
其中ERVZ係如請求項1中所定義。
For example, the compound of any one of claims 1 to 3 and all stereoisomers, racemic mixtures, tautomers, pharmaceutically acceptable salts, hydrates and polymorphs thereof are compounds of formula (Ic) :
Figure 108107135-A0305-02-0215-41
Where E , R , V and Z are as defined in claim 1.
如請求項1之化合物及其所有立體異構物、外消旋混合物、互變異構物、醫藥上可接受之鹽、水合物及多形體,其中該化合物係選自由以下組成之群:
Figure 108107135-A0305-02-0216-42
Figure 108107135-A0305-02-0217-43
Figure 108107135-A0305-02-0218-44
Figure 108107135-A0305-02-0219-45
Figure 108107135-A0305-02-0220-46
Figure 108107135-A0305-02-0221-47
For example, the compound of claim 1 and all stereoisomers, racemic mixtures, tautomers, pharmaceutically acceptable salts, hydrates and polymorphs thereof, wherein the compound is selected from the group consisting of:
Figure 108107135-A0305-02-0216-42
Figure 108107135-A0305-02-0217-43
Figure 108107135-A0305-02-0218-44
Figure 108107135-A0305-02-0219-45
Figure 108107135-A0305-02-0220-46
Figure 108107135-A0305-02-0221-47
一種化合物及其所有立體異構物、外消旋混合物、互變異構物、醫藥上可接受之鹽、水合物及多形體,其係選自由以下組成之群:
Figure 108107135-A0305-02-0221-48
Figure 108107135-A0305-02-0222-49
A compound and all its stereoisomers, racemic mixtures, tautomers, pharmaceutically acceptable salts, hydrates and polymorphs, selected from the group consisting of:
Figure 108107135-A0305-02-0221-48
Figure 108107135-A0305-02-0222-49
一種醫藥組合物,其包含如請求項1至7中任一項之化合物及其所有立體異構物、外消旋混合物、互變異構物、醫藥上可接受之鹽、水合物及多形體及視情況醫藥上可接受之載劑或賦形劑。 A pharmaceutical composition comprising a compound as claimed in any one of claims 1 to 7 and all stereoisomers, racemic mixtures, tautomers, pharmaceutically acceptable salts, hydrates and polymorphs thereof, and Pharmaceutically acceptable carriers or excipients, as appropriate. 如請求項1至3、6及7中任一項之化合物及其所有立體異構物、外消旋混合物、互變異構物、醫藥上可接受之鹽、水合物及多形體,其用作藥劑。 For example, the compounds of any one of claims 1 to 3, 6 and 7 and all stereoisomers, racemic mixtures, tautomers, pharmaceutically acceptable salts, hydrates and polymorphs thereof are used as Potion. 一種式(I)化合物及其所有立體異構物、外消旋混合物、互變異構物、醫藥上可接受之鹽、水合物及多形體之用途,
Figure 108107135-A0305-02-0222-50
其係用於製造用於治療、改善或預防與Tau蛋白聚集體有關的病症或 異常、用於製造用於降低Tau聚集、用於防止Tau聚集體之形成及/或用於抑制Tau聚集、用於細胞內干擾Tau聚集體、用於降低活體內Tau錯誤摺疊及過度磷酸化、或用於減少神經發炎標記物之藥劑,其中 A係選自由以下組成之群:
Figure 108107135-A0305-02-0223-52
Figure 108107135-A0305-02-0223-53
Figure 108107135-A0305-02-0223-54
Figure 108107135-A0305-02-0223-58
Figure 108107135-A0305-02-0223-55
,其中
Figure 108107135-A0305-02-0223-56
Figure 108107135-A0305-02-0223-57
Figure 108107135-A0305-02-0223-59
Figure 108107135-A0305-02-0223-60
Figure 108107135-A0305-02-0223-61
Figure 108107135-A0305-02-0223-62
Figure 108107135-A0305-02-0223-63
Figure 108107135-A0305-02-0223-64
Figure 108107135-A0305-02-0223-65
可在任一可用位置連接至Q,其中
Figure 108107135-A0305-02-0223-66
Figure 108107135-A0305-02-0223-67
Figure 108107135-A0305-02-0223-68
Figure 108107135-A0305-02-0223-69
經一或多個取代基R j 取代,其中當A為
Figure 108107135-A0305-02-0223-70
時,R j 不為-CN、鹵素或- CF3,且其中
Figure 108107135-A0305-02-0223-71
Figure 108107135-A0305-02-0223-72
Figure 108107135-A0305-02-0223-73
Figure 108107135-A0305-02-0223-74
可視情況經一或多個取代基R取代;B係選自由O及NRa組成之群;EV獨立地選自由以下組成之群:N、NR5、O及S;G係選自由以下組成之群:苯環、嘧啶環及吡啶環; J係選自由O及N-R1組成之群;Q係選自由N及C-R1組成之群;Y係選自由CZ及N組成之群;Y 1 係選自由CZ及N組成之群;Y 2 係選自由CZ及N組成之群;Y 3 係選自由CZ及N組成之群;Z獨立地選自由以下組成之群:H、鹵素、O-烷基、烷基及CN;R獨立地選自由
Figure 108107135-A0305-02-0224-75
及-NR3R4組成之群;R a 係選自由H及烷基組成之群;R b R c R d R e R f R g 獨立地選自由H及烷基組成之群,或R b R c R d R e R f R g 中之任兩者可連結形成3員至8員環;R j 獨立地選自由以下組成之群:-鹵素、-O-烷基、-CF3、-CN、-NR3R4
Figure 108107135-A0305-02-0224-76
Figure 108107135-A0305-02-0224-77
,其中含C1-2碳原子之橋可存在於a碳原子與c或d碳原子之間或其中含C1-2碳原子之橋可存在於b碳原子與c或d碳原子之間;R 1 係選自由H及烷基組成之群;R 2 獨立地選自由烷基、F及=O組成之群,其中該烷基可視情況經鹵素、-OH或-O-烷基取代,且其中若兩個R 2 係孿位的,則其可連結形成3員至6員環;R 3 R 4 獨立地選自由H及烷基組成之群,其中該烷基可視情況經鹵素、-OH或-O-烷基取代; R 5 係選自由H及烷基組成之群;n係0、1、2、3或4;rs獨立地係0、1、2或3;及tu獨立地係1、2或3。
The use of a compound of formula (I) and all stereoisomers, racemic mixtures, tautomers, pharmaceutically acceptable salts, hydrates and polymorphs thereof,
Figure 108107135-A0305-02-0222-50
It is used for manufacturing, for treating, ameliorating or preventing diseases or abnormalities related to Tau protein aggregates, for manufacturing, for reducing Tau aggregation, for preventing the formation of Tau aggregates and/or for inhibiting Tau aggregation, for Agents that interfere with Tau aggregates in cells, are used to reduce Tau misfolding and hyperphosphorylation in vivo, or are used to reduce neuroinflammatory markers, wherein A is selected from the group consisting of:
Figure 108107135-A0305-02-0223-52
,
Figure 108107135-A0305-02-0223-53
,
Figure 108107135-A0305-02-0223-54
,
Figure 108107135-A0305-02-0223-58
and
Figure 108107135-A0305-02-0223-55
,in
Figure 108107135-A0305-02-0223-56
,
Figure 108107135-A0305-02-0223-57
,
Figure 108107135-A0305-02-0223-59
,
Figure 108107135-A0305-02-0223-60
,
Figure 108107135-A0305-02-0223-61
,
Figure 108107135-A0305-02-0223-62
,
Figure 108107135-A0305-02-0223-63
,
Figure 108107135-A0305-02-0223-64
and
Figure 108107135-A0305-02-0223-65
Can be connected to Q at any available location, where
Figure 108107135-A0305-02-0223-66
,
Figure 108107135-A0305-02-0223-67
,
Figure 108107135-A0305-02-0223-68
and
Figure 108107135-A0305-02-0223-69
Substituted with one or more substituents R j , where when A is
Figure 108107135-A0305-02-0223-70
, R j is not -CN, halogen or -CF 3 , and where
Figure 108107135-A0305-02-0223-71
,
Figure 108107135-A0305-02-0223-72
,
Figure 108107135-A0305-02-0223-73
and
Figure 108107135-A0305-02-0223-74
Optionally substituted by one or more substituents R ; B is selected from the group consisting of O and NR a ; E and V are independently selected from the group consisting of: N, NR 5 , O and S; G is selected from the following The group consisting of: benzene ring, pyrimidine ring and pyridine ring; J is selected from the group consisting of O and NR 1 ; Q is selected from the group consisting of N and CR 1 ; Y is selected from the group consisting of CZ and N; Y 1 Y 2 is selected from the group consisting of CZ and N; Y 3 is selected from the group consisting of CZ and N; Z is independently selected from the group consisting of: H, halogen, O- Alkyl, alkyl and CN; R is independently selected from
Figure 108107135-A0305-02-0224-75
and -NR 3 R 4 ; R a is selected from the group consisting of H and alkyl groups; R b , R c , R d , R e , R f and R g are independently selected from the group consisting of H and alkyl groups group, or any two of R b , R c , R d , Re , R f and R g can be connected to form a 3- to 8-membered ring; R j is independently selected from the group consisting of: - halogen, - O-alkyl, -CF 3 , -CN, -NR 3 R 4 ,
Figure 108107135-A0305-02-0224-76
and
Figure 108107135-A0305-02-0224-77
, where a bridge containing C 1-2 carbon atoms can exist between a carbon atom and c or d carbon atom or where a bridge containing C 1-2 carbon atoms can exist between b carbon atom and c or d carbon atom ; R 1 is selected from the group consisting of H and alkyl; R 2 is independently selected from the group consisting of alkyl, F and =O, wherein the alkyl is optionally substituted by halogen, -OH or -O-alkyl, And if two R 2 are twin-positioned, they can be connected to form a 3- to 6-membered ring; R 3 and R 4 are independently selected from the group consisting of H and an alkyl group, wherein the alkyl group may be optionally treated with halogen, -OH or -O-alkyl substitution; R 5 is selected from the group consisting of H and alkyl; n is 0, 1, 2, 3 or 4; r and s are independently 0, 1, 2 or 3; and t and u are independently 1, 2 or 3.
如請求項10之用途,其中該式(I)化合物係如請求項1至7中任一項中所定義。 The use of claim 10, wherein the compound of formula (I) is as defined in any one of claims 1 to 7. 如請求項10或11之用途,其中該病症係選自阿茲海默氏病(Alzheimer’s disease,AD)、家族性AD、原發性年齡相關Tau蛋白病變(PART)、庫賈氏病(Creutzfeldt-Jacob disease)、拳擊手型失智症、唐氏症候群(Down’s Syndrome)、傑茨曼-斯脫司勒-史茵克病(Gerstmann-Sträussler-Scheinker disease,GSS)、包涵體肌炎、普里昂蛋白(prion protein)腦類澱粉血管病變、創傷性腦損傷(TBI)、肌肉萎縮性脊髓側索硬化症(ALS)、關島帕金森氏症-失智複合症(Parkinsonism-dementia complex of Guam)、非關島運動神經元病伴神經纖維纏結(non-Guamanian motor neuron disease with neurofibrillary tangles)、嗜銀顆粒病、皮質基底核退化症(CBD)、瀰漫性神經纖維纏結伴鈣化、與染色體17相關之額顳葉失智症伴帕金森氏症(FTDP-17)、哈勒沃登-施帕茨病(Hallervorden-Spatz disease)、多系統萎縮(MSA)、C型尼曼匹克症(Niemann-Pick disease type C)、蒼白球-腦橋-黑質退化、匹克氏病(Pick’s disease,PiD)、進行性皮質下神經膠瘤病、進行性核上性麻痺(PSP)、亞急性硬化性泛腦炎、纏結主導型失智症、腦炎後帕金森氏症、 肌強直性營養不良、亞急性硬化性泛腦病變、LRRK2突變、慢性創傷性腦病變(CTE)、家族性英國型失智症(familial British dementia)、家族性丹麥型失智症(familial Danish dementia)、其他額顳葉退化、瓜德羅普帕金森氏症(Guadeloupean Parkinsonism)、神經退化伴腦內鐵累積、SLC9A6相關性智力遲鈍、白質tau蛋白病變伴球狀膠質細胞包涵體、癲癇、路易氏體失智症(Lewy body dementia,LBD)、輕度認知損害(MCI)、多發性硬化、帕金森氏病(Parkinson's disease)、HIV相關性失智症、成人發作型糖尿病、老年性心臟類澱粉變性、青光眼、缺血性中風、AD之精神病及亨庭頓氏病(Huntington's disease)。 The use of claim 10 or 11, wherein the disease is selected from Alzheimer's disease (AD), familial AD, primary age-related tauopathy (PART), Creutzfeldt- Jacob disease), boxer dementia, Down's Syndrome, Gerstmann-Sträussler-Scheinker disease (GSS), inclusion body myositis, Prion Protein (prion protein) cerebral amyloid angiopathy, traumatic brain injury (TBI), amyotrophic lateral sclerosis (ALS), Parkinsonism-dementia complex of Guam, Non-Guamanian motor neuron disease with neurofibrillary tangles, argyrophilic granulopathy, corticobasal degeneration (CBD), diffuse neurofibrillary tangles with calcification, chromosome 17-related Frontotemporal dementia with Parkinson's disease (FTDP-17), Hallervorden-Spatz disease, multiple system atrophy (MSA), Niemann-Pick disease type C disease type C), pallidum-pontine-substantia nigra degeneration, Pick's disease (PiD), progressive subcortical gliomatosis, progressive supranuclear palsy (PSP), subacute sclerosing panencephalitis , tangle-dominant dementia, post-encephalitic Parkinson's disease, Myotonic dystrophy, subacute sclerosing panencephalopathy, LRRK2 mutation, chronic traumatic encephalopathy (CTE), familial British dementia, familial Danish dementia ), other frontotemporal degeneration, Guadeloupe Parkinsonism, neurodegeneration with iron accumulation in the brain, SLC9A6-related mental retardation, white matter tauopathy with glomerular inclusions, epilepsy, Lewy Lewy body dementia (LBD), mild cognitive impairment (MCI), multiple sclerosis, Parkinson's disease, HIV-related dementia, adult-onset diabetes, senile heart disease Amylase, glaucoma, ischemic stroke, AD psychosis and Huntington's disease. 如請求項10或11之用途,其中該病症係選自阿茲海默氏病(AD)、皮質基底核退化症(CBD)、匹克氏病(PiD)及進行性核上性麻痺(PSP)。 The use of claim 10 or 11, wherein the disease is selected from Alzheimer's disease (AD), corticobasal degeneration (CBD), Pick's disease (PiD) and progressive supranuclear palsy (PSP) . 如請求項1至3、6及7中任一項之化合物及其所有立體異構物、外消旋混合物、互變異構物、醫藥上可接受之鹽、水合物及多形體,其用於降低Tau聚集。 For example, the compounds of any one of claims 1 to 3, 6 and 7 and all stereoisomers, racemic mixtures, tautomers, pharmaceutically acceptable salts, hydrates and polymorphs thereof are used for Reduce Tau aggregation. 如請求項1至3、6及7中任一項之化合物及其所有立體異構物、外消旋混合物、互變異構物、醫藥上可接受之鹽、水合物及多形體,其用於防止Tau聚集體之形成及/或用於抑制Tau聚集。 For example, the compounds of any one of claims 1 to 3, 6 and 7 and all stereoisomers, racemic mixtures, tautomers, pharmaceutically acceptable salts, hydrates and polymorphs thereof are used for To prevent the formation of Tau aggregates and/or to inhibit Tau aggregation. 如請求項1至3、6及7中任一項之化合物及其所有立體異構物、外消旋混合物、互變異構物、醫藥上可接受之鹽、水合物及多形體,其用於細 胞內干擾Tau聚集體。 For example, the compounds of any one of claims 1 to 3, 6 and 7 and all stereoisomers, racemic mixtures, tautomers, pharmaceutically acceptable salts, hydrates and polymorphs thereof are used for thin Intracellular interference with Tau aggregates. 如請求項1至3、6及7中任一項之化合物及其所有立體異構物、外消旋混合物、互變異構物、醫藥上可接受之鹽、水合物及多形體,其用於降低活體內Tau錯誤摺疊及過度磷酸化。 For example, the compounds of any one of claims 1 to 3, 6 and 7 and all stereoisomers, racemic mixtures, tautomers, pharmaceutically acceptable salts, hydrates and polymorphs thereof are used for Reduce Tau misfolding and hyperphosphorylation in vivo. 如請求項1至3、6及7中任一項之化合物及其所有立體異構物、外消旋混合物、互變異構物、醫藥上可接受之鹽、水合物及多形體,其用於減少神經發炎標記物。 For example, the compounds of any one of claims 1 to 3, 6 and 7 and all stereoisomers, racemic mixtures, tautomers, pharmaceutically acceptable salts, hydrates and polymorphs thereof are used for Reduce markers of neuroinflammation. 一種如請求項1至7中任一項之化合物及其所有立體異構物、外消旋混合物、互變異構物、醫藥上可接受之鹽、水合物及多形體或如請求項8之醫藥組合物之用途,其用於製造用於降低Tau聚集之藥劑。 A compound as claimed in any one of claims 1 to 7 and all stereoisomers, racemic mixtures, tautomers, pharmaceutically acceptable salts, hydrates and polymorphs thereof or a medicament as claimed in claim 8 Use of the composition for the manufacture of a medicament for reducing Tau aggregation. 一種如請求項1至7中任一項之化合物及其所有立體異構物、外消旋混合物、互變異構物、醫藥上可接受之鹽、水合物及多形體或如請求項8之醫藥組合物之用途,其用於製造用於防止Tau聚集體之形成及/或用於抑制Tau聚集之藥劑。 A compound as claimed in any one of claims 1 to 7 and all stereoisomers, racemic mixtures, tautomers, pharmaceutically acceptable salts, hydrates and polymorphs thereof or a medicament as claimed in claim 8 Use of the composition for manufacturing agents for preventing the formation of Tau aggregates and/or for inhibiting Tau aggregation. 一種如請求項1至7中任一項之化合物及其所有立體異構物、外消旋混合物、互變異構物、醫藥上可接受之鹽、水合物及多形體或如請求項8之醫藥組合物之用途,其用於製造用於細胞內干擾Tau聚集體之藥劑。 A compound as claimed in any one of claims 1 to 7 and all stereoisomers, racemic mixtures, tautomers, pharmaceutically acceptable salts, hydrates and polymorphs thereof or a medicament as claimed in claim 8 Use of compositions for the manufacture of agents for interfering with Tau aggregates within cells. 一種如請求項1至7中任一項之化合物及其所有立體異構物、外消旋混合物、互變異構物、醫藥上可接受之鹽、水合物及多形體或如請求項8之醫藥組合物之用途,其用於製造用於降低活體內Tau錯誤摺疊及過度磷酸化之藥劑。 A compound as claimed in any one of claims 1 to 7 and all stereoisomers, racemic mixtures, tautomers, pharmaceutically acceptable salts, hydrates and polymorphs thereof or a medicament as claimed in claim 8 Use of the composition for manufacturing agents for reducing Tau misfolding and hyperphosphorylation in vivo. 一種如請求項1至7中任一項之化合物及其所有立體異構物、外消旋混合物、互變異構物、醫藥上可接受之鹽、水合物及多形體或如請求項8之醫藥組合物之用途,其用於製造用於減少神經發炎標記物之藥劑。 A compound as claimed in any one of claims 1 to 7 and all stereoisomers, racemic mixtures, tautomers, pharmaceutically acceptable salts, hydrates and polymorphs thereof or a medicament as claimed in claim 8 Use of a composition for the manufacture of a medicament for reducing markers of neuroinflammation. 一種混合物,其包含如請求項1至7中任一項之化合物及其所有立體異構物、外消旋混合物、互變異構物、醫藥上可接受之鹽、水合物及多形體及至少一種其他生物活性化合物,選自與如請求項1至7中任一項之化合物不同之治療劑,其係選自由以下組成之群:抗氧化壓力之化合物、抗細胞凋亡化合物、金屬螯合劑、DNA修復抑制劑、哌侖西平(pirenzepine)或其代謝物、3-胺基-1-丙烷磺酸(3APS)、1,3-丙烷二磺酸鹽(1,3PDS)、α-分泌酶活化劑、β-及γ-分泌酶抑制劑、肝醣合酶激酶3抑制劑、O連接之N-乙醯葡糖胺水解酶(O-glcnacase,OGA)抑制劑、神經傳遞質、β褶板碎裂劑(β-sheet breakers)、類澱粉β清除/消除(depleting)細胞組分引誘劑、N端截短之類澱粉β(包括焦麩胺酸化之類澱粉β 3-42)之抑制劑、抗發炎分子、膽鹼酯酶抑制劑(ChEI)、塔克寧(tacrine)、利凡斯的明(rivastigmine)、多奈派齊(donepezil)、加蘭他敏(galantamine)、M1激動劑、類澱粉或Tau改質藥物及營養補充劑、抗體、功能等效抗體或其功能部分、或疫苗, 其中該混合物進一步包含醫藥上可接受之載劑、稀釋劑及賦形劑中之至少一者。 A mixture comprising a compound as claimed in any one of claims 1 to 7 and all stereoisomers, racemic mixtures, tautomers, pharmaceutically acceptable salts, hydrates and polymorphs thereof and at least one Other biologically active compounds selected from therapeutic agents different from the compounds of any one of claims 1 to 7, which are selected from the group consisting of: compounds that resist oxidative stress, anti-apoptotic compounds, metal chelators, DNA repair inhibitor, pirenzepine or its metabolites, 3-amino-1-propanesulfonic acid (3APS), 1,3-propanedisulfonate (1,3PDS), α-secretase activation Agents, β- and γ-secretase inhibitors, glycogen synthase kinase 3 inhibitors, O-linked N-acetylglucosamine hydrolase (O-glcnacase, OGA) inhibitors, neurotransmitters, β-pleated plates β-sheet breakers, amyloid β-like scavenging/depleting cellular component attractants, N-terminally truncated starch-like β-like (including pyroglutamate-like β-amyloid 3-42) inhibitors , anti-inflammatory molecules, cholinesterase inhibitors (ChEI), tacrine, rivastigmine, donepezil, galantamine, M1 agonist , starch-like or Tau-modified drugs and nutritional supplements, antibodies, functionally equivalent antibodies or functional parts thereof, or vaccines, The mixture further includes at least one of a pharmaceutically acceptable carrier, diluent and excipient. 如請求項24之混合物,其中該化合物及/或該其他生物活性化合物係以治療有效量存在。 The mixture of claim 24, wherein the compound and/or the other biologically active compound is present in a therapeutically effective amount. 如請求項24或25之混合物,其中該其他生物活性化合物係膽鹼酯酶抑制劑(ChEI)。 The mixture of claim 24 or 25, wherein the other biologically active compound is a cholinesterase inhibitor (ChEI). 如請求項24或25之混合物,其中該其他生物活性化合物係選自由以下組成之群:塔克寧、利凡斯的明、多奈派齊、加蘭他敏、菸鹼酸及美金剛(memantine)。 Such as the mixture of claim 24 or 25, wherein the other biologically active compound is selected from the group consisting of: tacnin, rivastigmine, donepezil, galantamine, nicotinic acid and memantine ( memantine). 如請求項24或25之混合物,其中該其他生物活性化合物係單株抗體或功能等效抗體或其功能部分。 Such as the mixture of claim 24 or 25, wherein the other biologically active compound is a monoclonal antibody or a functionally equivalent antibody or a functional part thereof. 一種如請求項1至6或7中任一項之化合物及其所有立體異構物、外消旋混合物、互變異構物、醫藥上可接受之鹽、水合物及多形體之用途,其用作用於表徵具有Tau病理學之組織及用於在此組織上測試靶向Tau病理學之化合物之分析參考或活體外篩選工具。The use of a compound as claimed in any one of claims 1 to 6 or 7 and all stereoisomers, racemic mixtures, tautomers, pharmaceutically acceptable salts, hydrates and polymorphs thereof, Analytical reference or in vitro screening tools for characterizing tissues with Tau pathology and for testing compounds targeting Tau pathology on such tissues.
TW108107135A 2018-06-04 2019-03-04 Novel compounds for the treatment, alleviation or prevention of disorders associated with tau aggregates TWI812677B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP18175852 2018-06-04
EP18175852.5 2018-06-04

Publications (2)

Publication Number Publication Date
TW202017923A TW202017923A (en) 2020-05-16
TWI812677B true TWI812677B (en) 2023-08-21

Family

ID=62530124

Family Applications (1)

Application Number Title Priority Date Filing Date
TW108107135A TWI812677B (en) 2018-06-04 2019-03-04 Novel compounds for the treatment, alleviation or prevention of disorders associated with tau aggregates

Country Status (3)

Country Link
AR (1) AR117560A1 (en)
EA (1) EA202091395A1 (en)
TW (1) TWI812677B (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008036021A1 (en) * 2006-09-20 2008-03-27 Astrazeneca Ab Tetrahydro-lh-pyrido [3,4 -b] indole derivatives as cbl receptor ligands
WO2010080253A1 (en) * 2008-12-18 2010-07-15 Merck Patent Gmbh Tricyclic azaindoles
WO2011128455A1 (en) * 2010-04-16 2011-10-20 Ac Immune S.A. Novel compounds for the treatment of diseases associated with amyloid or amyloid-like proteins
TWI498328B (en) * 2008-01-25 2015-09-01 Medivation Technologies Inc New 2,3,4,5-tetrahydro-1h-pyrido(4,3-b)indole compounds and methods of use thereof

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008036021A1 (en) * 2006-09-20 2008-03-27 Astrazeneca Ab Tetrahydro-lh-pyrido [3,4 -b] indole derivatives as cbl receptor ligands
TWI498328B (en) * 2008-01-25 2015-09-01 Medivation Technologies Inc New 2,3,4,5-tetrahydro-1h-pyrido(4,3-b)indole compounds and methods of use thereof
WO2010080253A1 (en) * 2008-12-18 2010-07-15 Merck Patent Gmbh Tricyclic azaindoles
WO2011128455A1 (en) * 2010-04-16 2011-10-20 Ac Immune S.A. Novel compounds for the treatment of diseases associated with amyloid or amyloid-like proteins

Also Published As

Publication number Publication date
EA202091395A1 (en) 2020-10-01
TW202017923A (en) 2020-05-16
AR117560A1 (en) 2021-08-18

Similar Documents

Publication Publication Date Title
JP7225251B2 (en) 1,3,4,5-Tetrahydro-2H-pyrido(4,3-B) Indole Derivatives for the Treatment, Alleviation or Prevention of Tau Aggregate-Related Disorders Such as Alzheimer's Disease
TWI743484B (en) Novel compounds for the treatment, alleviation or prevention of disorders associated with tau aggregates
JP7232931B2 (en) Novel compounds for the treatment, alleviation or prevention of disorders associated with tau aggregates
US11814378B2 (en) Compounds for the treatment, alleviation or prevention of disorders associated with Tau aggregates
TWI812677B (en) Novel compounds for the treatment, alleviation or prevention of disorders associated with tau aggregates
EA043389B1 (en) 1,3,4,5-TETRAHYDRO-2H-PYRIDO[4,3-b]INDOLE DERIVATIVES FOR THE TREATMENT, RELIEF, OR PREVENTION OF TAU AGGREGATE ASSOCIATED DISORDERS, SUCH AS ALZHEIMER'S DISEASE
WO2022078971A1 (en) Novel compounds