TW201725202A - Novel substituted indazoles, processes for preparation thereof, pharmaceutical preparations comprising them and use thereof for production of medicaments - Google Patents

Novel substituted indazoles, processes for preparation thereof, pharmaceutical preparations comprising them and use thereof for production of medicaments Download PDF

Info

Publication number
TW201725202A
TW201725202A TW105142812A TW105142812A TW201725202A TW 201725202 A TW201725202 A TW 201725202A TW 105142812 A TW105142812 A TW 105142812A TW 105142812 A TW105142812 A TW 105142812A TW 201725202 A TW201725202 A TW 201725202A
Authority
TW
Taiwan
Prior art keywords
hydroxy
trifluoromethyl
carboxamide
pyridine
group
Prior art date
Application number
TW105142812A
Other languages
Chinese (zh)
Inventor
厄瑞屈 波斯
賀爾格 席比尼奇
尼可 史密特
茱蒂斯 剛瑟
霍格爾 史堤柏爾
沃夫 波默
馬汀 蘭奇
蘭哈德 努伯梅爾
史文 林
克里西汀 史德曼
Original Assignee
拜耳製藥公司
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 拜耳製藥公司 filed Critical 拜耳製藥公司
Publication of TW201725202A publication Critical patent/TW201725202A/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/04Ortho-condensed systems

Abstract

The present application relates to novel substituted indazoles, to processes for preparation thereof, to the use thereof alone or in combinations for treatment and/or prophylaxis of diseases, and to the use thereof for production of medicaments for treatment and/or prophylaxis of diseases, especially for treatment and/or prophylaxis of endometriosis and endometriosis-associated pain and other endometriosis-associated symptoms such as dysmenorrhoea, dyspareunia, dysuria and dyschezia, of lymphoma, rheumatoid arthritis, spondyloarthritis (especially psoriatic spondyloarthritis and Bekhterev's disease), lupus erythematosus, multiple sclerosis, macular degeneration, COPD, gout, fatty liver disorders, insulin resistance, neoplastic disorders and psoriasis.

Description

新穎經取代吲唑、其製法、包含其之醫藥製劑及其製備藥劑之用途Novel substituted carbazole, its preparation method, pharmaceutical preparation containing the same, and use thereof

本發明係關於新穎經取代吲唑、關於其製法、關於用於製備該等新穎化合物之中間體、關於該等新穎經取代吲唑用於治療及/或預防疾病之用途,且關於其用於製備用以治療及/或預防疾病、尤其增殖病症(例如自體免疫病症)、代謝及發炎病症(例如類風濕性關節炎)、脊椎關節炎(尤其牛皮癬性脊椎關節炎及別赫捷列夫氏病(Bekhterev's disease))、慢性阻塞性肺病(縮寫:COPD)、多發性硬化、全身性紅斑性狼瘡、痛風、代謝症候群、脂肪肝肝炎、胰島素抗性、子宮內膜異位症及發炎誘導之疼痛或慢性疼痛及淋巴瘤之藥劑的用途。 本發明係關於通式(I)之新穎經取代吲唑,其抑制介白素-1受體相關之激酶4 (IRAK4)。The present invention relates to novel substituted carbazoles, to processes for their preparation, to intermediates useful in the preparation of such novel compounds, to the use of such novel substituted carbazoles for the treatment and/or prophylaxis of diseases, and to their use Prepared to treat and/or prevent diseases, especially proliferative disorders (eg autoimmune disorders), metabolic and inflammatory conditions (eg rheumatoid arthritis), spondylarthritis (especially psoriatic spondylarthritis and Behetylev) Bekhterev's disease, chronic obstructive pulmonary disease (abbreviation: COPD), multiple sclerosis, systemic lupus erythematosus, gout, metabolic syndrome, fatty liver hepatitis, insulin resistance, endometriosis and inflammation induction The use of a medicament for pain or chronic pain and lymphoma. The present invention relates to a novel substituted carbazole of the general formula (I) which inhibits the interleukin-1 receptor-associated kinase 4 (IRAK4).

人類IRAK4 (介白素-1受體相關之激酶4)在免疫系統的活化作用扮演關鍵角色。因此,此激酶係用於開發發炎抑制性物質之重要治療靶分子。IRAK4由多種細胞表現且介導類鐸受體(TLR) (TLR3除外)及介白素(IL)-1β家族受體(由IL-1R (受體)、IL-18R、IL-33R及IL-36R組成)之信號轉導(Janeway及Medzhitov, Annu. Rev. Immunol., 2002;Dinarello, Annu. Rev. Immunol., 2009;Flannery及Bowie, Biochemical Pharmacology, 2010)。 IRAK4剔除小鼠及來自缺乏IRAK4之患者之人類細胞皆不會對TLR (TLR3除外)及IL-1β家族之刺激有反應(Suzuki、Suzuki等人,Nature, 2002;Davidson、Currie等人,The Journal of Immunology, 2006;Ku、von Bernuth等人,JEM, 2007;Kim、Staschke等人,JEM, 2007)。 TLR配體或IL-1β家族配體結合至各別受體導致MyD88 [髓樣分化原發反應基因(88)]募集並結合至受體。因此,MyD88與IRAK4發生相互作用,導致形成活性複合物,從而與激酶IRAK1或IRAK2發生相互作用並活化該等激酶(Kollewe、Mackensen等人,Journal of Biological Chemistry, 2004;Precious等人,J. Biol. Chem., 2009)。因此,NF (核因子)-κB信號傳導路徑及MAPK (促分裂原活化之蛋白激酶)信號路徑得到活化(Wang、Deng等人,Nature, 2001)。NF-κB信號路徑及MAPK信號路徑之活化會引起與不同免疫過程相關之過程。舉例而言,各種發炎信號分子及酶(例如細胞介素、趨化介素及COX-2 (環加氧酶-2))之表現有所增加,且發炎相關之基因(例如COX-2、IL-6 (介白素-6)、IL-8)之mRNA穩定性有所增加(Holtmann、Enninga等人,Journal of Biological Chemistry, 2001;Datta、Novotny等人,The Journal of Immunology, 2004)。此外,該等過程可能與特定細胞類型(例如單核球、巨噬細胞、樹突細胞、T細胞及B細胞)之增殖及分化相關(Wan、Chi等人,Nat Immunol, 2006;McGettrick及J. O'Neill, British Journal of Haematology, 2007)。 藉由野生型(WT)小鼠與具有IRAK4之激酶去活化形式(IRAK4 KDKI)之經遺傳修飾動物的直接比較已經證明IRAK4在各種發炎病症之病理學中的重要作用。在多發性硬化、動脈粥樣硬化、心肌梗塞及阿茲海默氏病(Alzheimer's disease)之動物模型中,IRAK4 KDKI動物具有改良之臨床表現(Rekhter、Staschke等人,Biochemical and Biophysical Research Communication, 2008;Maekawa、Mizue等人,Circulation, 2009;Staschke、Dong等人,The Journal of Immunology, 2009;Kim、Febbraio等人,The Journal of Immunology, 2011;Cameron、Tse等人,The Journal of Neuroscience, 2012)。此外,咸發現,動物模型中IRAK4之缺失會由於改良之抗病毒反應以及同時減少之全身發炎而保護抵抗病毒誘導之心肌炎(Valaperti、Nishii等人,Circulation, 2013)。亦已顯示IRAK4之表現與格特-小柳-原田三氏症候群(Vogt-Koyanagi-Harada syndrome)程度相關聯(Sun、Yang等人,PLoS ONE, 2014)。另外, IRAK4對漿細胞樣樹突細胞之免疫複合物介導之IFNα (干擾素-α)產生的高相關性業已證明,該IFNα產生係全身性紅斑性狼瘡(SLE)之發病機制中之關鍵過程(Chiang等人,The Journal of Immunology, 2010)。此外,信號傳導路徑與肥胖症相關(Ahmad, R., P. Shihab等人,Diabetology &Metabolic Syndrome, 2015)。 除IRAK4在先天免疫性中之必不可少的作用以外,亦暗示IRAK4會影響稱為Th17 T細胞(適應性免疫性之組份)之分化。在不存在IRAK4激酶活性下,與WT小鼠相比,會生成較少的產生IL-17之T細胞(Th17 T細胞)。IRAK4之抑制使得能夠預防及/或治療動脈粥樣硬化、1型糖尿病、類風濕性關節炎、脊椎關節炎(尤其牛皮癬性脊椎關節炎及別赫捷列夫氏病)、紅斑性狼瘡、牛皮癬、白斑病、巨細胞動脈炎、慢性發炎腸病症及病毒病症(例如HIV (人類免疫缺失病毒)、肝炎病毒) (Staschke等人,The Journal of Immunology, 2009;Marquez等人,Ann Rheum Dis, 2014;Zambrano-Zaragoza等人,International Journal of Inflammation, 2014;Wang等人,Experimental and Therapeutic Medicine, 2015;Ciccia等人,Rheumatology, 2015)。 由於IRAK4在TLR (TLR3除外)及IL-1受體家族之MyD88介導之信號級聯中具有重要作用,故IRAK4之抑制可用於預防及/或治療由所提及受體介導之病症。TLR以及IL-1受體家族之組份參與以下疾病之發病機制:風濕性關節炎、牛皮癬關節炎、重症肌無力、血管炎(例如貝歇氏症(Behçet’s disease)、具有多血管炎之肉芽腫病及巨細胞動脈炎)、胰臟炎、全身性紅斑性狼瘡、皮肌炎及多發性肌炎、代謝症候群,包括(例如)胰島素抗性、高血壓、異常脂蛋白血症及肥胖症、糖尿病(1型及2型)、糖尿病腎病變、骨關節炎、薛格連氏症候群(Sjögren syndrome)及敗血症(Yang、Tuzun等人,J Immunol, 2005;Candia、Marquez等人,The Journal of Rheumatology, 2007;Scanzello、Plaas等人Curr Opin Rheumatol, 2008;Deng、Ma-Krupa等人,Circ Res, 2009;Roger、Froidevaux等人,PNAS, 2009;Devaraj、Tobias等人,Arterioscler Thromb Vasc Biol, 2011;Kim、Cho等人,Clin Rheumatol, 2010;Carrasco等人,Clinical and Experimental Rheumatology, 2011;Gambuzza、Licata等人,Journal of Neuroimmunology, 2011;Fresno, Archives Of Physiology And Biochemistry, 2011;Volin及Koch,J Interferon Cytokine Res, 2011;Akash、Shen等人,Journal of Pharmaceutical Sciences, 2012;Goh及Midwood,Rheumatology, 2012;Dasu、Ramirez等人,Clinical Science, 2012;Ouziel、Gustot等人,Am J Patho, 2012;Ramirez及Dasu,Curr Diabetes Rev, 2012,Okiyama等人,Arthritis Rheum, 2012;Chen等人,Arthritis Research & Therapy, 2013;Holle、Windmoller等人,Rheumatology (Oxford), 2013;Li、Wang等人,Pharmacology & Therapeutics, 2013;Sedimbi、Hagglof等人,Cell Mol Life Sci, 2013;Caso、Costa等人,Mediators of Inflammation, 2014;Cordiglieri、Marolda等人,J Autoimmun, 2014;Jialal、Major等人,J Diabetes Complications, 2014;Kaplan、Yazgan等人,Scand J Gastroenterol, 2014;Talabot-Aye等人,Cytokine, 2014;Zong、Dorph等人,Ann Rheum Di, 2014;Ballak、Stienstra等人,Cytokine, 2015;Timper、Seelig等人,J Diabetes Complications, 2015)。皮膚病(例如牛皮癬、異位性皮膚炎、金德勒氏症候群(Kindler's syndrome)、大皰性類天皰瘡、過敏性接觸性皮膚炎、斑禿、反常性痤瘡及尋常性痤瘡)與IRAK4介導之TLR信號傳導路徑或IL-1R家族相關(Schmidt、Mittnacht等人,J Dermatol Sci, 1996;Hoffmann, J Investig Dermatol Symp Proc, 1999;Gilliet, Conrad等人,Archives of Dermatology, 2004;Niebuhr、Langnickel等人,Allergy, 2008;Miller, Adv Dermatol, 2008;Terhorst、Kalali等人,Am J Clin Dermatol, 2010;Viguier、Guigue等人,Annals of Internal Medicine, 2010;Cevikbas, Steinhoff, J Invest Dermatol, 2012;Minkis、Aksentijevich等人,Archives of Dermatology, 2012;Dispenza、Wolpert等人,J Invest Dermatol, 2012;Minkis、Aksentijevich等人,Archives of Dermatology, 2012;Gresnigt及van de Veerdonk,Seminars in Immunology, 2013;Selway、Kurczab等人,BMC Dermatology, 2013;Sedimbi、Hagglof等人,Cell Mol Life Sci, 2013;Wollina、Koch等人Indian Dermatol Online, 2013;Foster、Baliwag等人,The Journal of Immunology, 2014)。 肺部病症(例如肺纖維化、阻塞性肺病(COPD)、急性呼吸窘迫症候群(ARDS)、急性肺損傷(ALI)、間質性肺病(ILD)、類肉瘤病及肺高血壓)亦顯示與各種TLR介導之信號路徑相關。肺部病症之發病機制係為以傳染方式介導或以非傳染方式介導之過程(Ramirez Cruz、Maldonado Bernal等人,Rev Alerg Mex, 2004;Jeyaseelan、Chu等人,Infection and Immunity, 2005;Seki、Tasaka等人,Inflammation Research, 2010;Xiang、Fan等人,Mediators of Inflammation, 2010;Margaritopoulos、Antoniou等人,Fibrogenesis & Tissue Repair, 2010;Hilberath、Carlo等人,The FASEB Journal, 2011;Nadigel、Prefontaine等人,Respiratory Research, 2011;Kovach及Standiford, International Immunopharmacology, 2011;Bauer、Shapiro等人,Mol Med, 2012;Deng、Yang等人,PLoS One, 2013;Freeman、Martinez等人,Respiratory Research, 2013;Dubaniewicz, A., Human Immunology, 2013)。TLR亦及IL-1R家族成員亦參與其他發炎病症之發病機制,例如過敏、貝歇氏症、痛風、紅斑性狼瘡、成人發作性斯蒂爾氏病(adult-onset Still's disease)、心包炎及慢性發炎腸病症(例如潰瘍性結腸炎及克隆氏病(Crohn's disease))、移植排斥及移植物抗宿主反應,且因此此處IRAK4之抑制係適宜預防及/或治療方法(Liu-Bryan、Scott等人,Arthritis& Rheumatism, 2005;Piggott、Eisenbarth等人,J Clin Inves, 2005;Christensen、Shupe等人,Immunity, 2006;Cario, Inflammatory Bowel Diseases, 2010;Nickerson, Christensen等人,The Journal of Immunology, 2010;Rakoff-Nahoum、Hao等人,Immunity, 2006;Heimesaat、Fischer等人,PLoS ONE, 2007;Heimesaat、Nogai等人,Gut, 2010;Kobori、Yagi等人,J Gastroenterol, 2010;Schmidt、Raghavan等人,Nat Immunol, 2010;Shi、Mucsi等人,Immunological Reviews, 2010;Leventhal及Schroppel, Kidney Int, 2012;Chen、Lin等人,Arthritis Res Ther, 2013;Hao、Liu等人,Curr Opin Gastroenterol, 2013;Kreisel及Goldstein, Transplant International, 2013;Li、Wang等人,Pharmacology & Therapeutics, 2013;Walsh、Carthy等人,Cytokine & Growth Factor Reviews, 2013;Zhu、Jiang等人,Autoimmunity, 2013;Yap及Lai, Nephrology, 2013;Vennegaard、Dyring-Andersen等人,Contact Dermatitis, 2014;D'Elia、Brucato等人,Clin Exp Rheumatol, 2015;Jain、Thongprayoon等人,Am J Cardiol., 2015;Li、Zhang等人,Oncol Rep., 2015)。 由TLR及IL-1R家族介導之婦科病症(例如子宮肌腺症、痛經、性交疼痛及子宮內膜異位症、尤其子宮內膜異位症相關性疼痛及其他子宮內膜異位症相關性症狀(例如痛經、性交疼痛、排尿困難及排糞困難)可受到IRAK4抑制劑之預防性及/或治療性使用之正面影響(Akoum、Lawson等人,Human Reproduction, 2007;Allhorn、Boing等人,Reproductive Biology and Endocrinology, 2008;Lawson、Bourcier等人,Journal of Reproductive Immunology, 2008;Sikora、Mielczarek-Palacz等人,American Journal of Reproductive Immunology, 2012;Khan、Kitajima等人,Journal of Obstetrics and Gynaecology Research, 2013;Santulli、Borghese等人,Human Reproduction, 2013)。IRAK4抑制劑之預防性及/或治療性使用亦可對動脈粥樣硬化具有積極影響(Seneviratne、Sivagurunathan等人,Clinica Chimica Acta, 2012;Falck-Hansen、Kassiteridi等人,International Journal of Molecular Sciences, 2013;Sedimbi、Hagglof等人,Cell Mol Life Sci, 2013)。 除已提及之病症以外,已在眼部病症(例如視網膜缺血、角膜炎、過敏性結膜炎、乾燥性角結膜炎、黃斑部病變及眼色素層炎)之發病機制中闡述IRAK4介導之TLR過程(Kaarniranta及Salminen, J Mol Med (Berl), 2009;Sun及Pearlman, Investigative Ophthalmology & Visual Science, 2009;Redfern及McDermott, Experimental Eye Research, 2010;Kezic、Taylor等人,J Leukoc Biol, 2011;Chang、McCluskey等人,Clinical & Experimental Ophthalmology, 2012;Guo、Gao等人,Immunol Cell Biol, 2012;Lee、Hattori等人,Investigative Ophthalmology & Visual Science, 2012;Qi、Zhao等人,Investigative Ophthalmology & Visual Science, 2014)。 IRAK4之抑制亦係纖維變性病症(例如肝纖維化、心肌炎、原發性膽汁性肝硬化、囊性纖維化)的適宜治療方法(Zhao, Zhao等人,Scand J Gastroenterol, 2011;Benias、Gopal等人,Clin Res Hepatol Gastroenterol, 2012;Yang, L.及E. Seki, Front Physiol, 2012;Liu、Hu等人,Biochim Biophys Acta., 2015)。 由於IRAK4在由TLR及IL-1R家族介導之病症中具有關鍵位置,故可利用IRAK4抑制劑以預防性及/或治療性方式治療慢性肝病症,例如脂肪肝肝炎及尤其非酒精性脂肪肝疾病(NAFLD)及/或非酒精性脂肪性肝炎(NASH)、酒精性脂肪性肝炎(ASH) (Nozaki, Saibara等人,Alcohol Clin Exp Res, 2004;Csak, T., A. Velayudham等人,Am J Physiol Gastrointest Liver Physiol, 2011;Miura, Kodama等人,Gastroenterology, 2010;Kamari, Shaish等人,J Hepatol, 2011;Ye、Li等人,Gut, 2012;Roh, Seki, J Gastroenterol Hepatol, 2013;Ceccarelli, S., V. Nobili等人,World J Gastroenterol, 2014;Miura, Ohnishi, World J Gastroenterol, 2014;Stojsavljevic, Palcic等人,World J Gastroenterol, 2014)。 由於IRAK4在TLR介導之過程中之重要作用,故IRAK4之抑制亦使得能夠治療及/或預防心血管及神經病症,例如心肌再灌注損害、心肌梗塞、高血壓(Oyama、Blais等人,Circulation, 2004;Timmers、Sluijter等人,Circulation Research, 2008;Fang及Hu,Med Sci Monit, 2011;Bijani, International Reviews of Immunology, 2012;Bomfim、Dos Santos等人,Clin Sci (Lond), 2012;Christia及Frangogiannis,European Journal of Clinical Investigation, 2013;Thompson及Webb,Clin Sci (Lond), 2013;Hernanz、Martínez-Revelles等人,British Journal of Pharmacology, 2015;Frangogiannis, Curr Opin Cardiol, 2015;Bomfim、Echem等人,Life Sciences, 2015),亦及阿茲海默氏病、中風、顱腦創傷、肌肉萎縮性脊髓側索硬化症(ALS)及帕金森病(Parkinson's) (Brough、Tyrrell等人,Trends in Pharmacological Sciences, 2011;Carty及Bowie,Biochemical Pharmacology, 2011;Denes、Kitazawa、Cheng等人,The Journal of Immunology, 2011;Lim、Kou等人,The American Journal of Pathology, 2011;Béraud及Maguire-Zeiss,Parkinsonism& Related Disorders, 2012;Denes、Wilkinson等人,Disease Models & Mechanisms, 2013;Noelker、Morel等人,Sci. Rep., 2013;Wang、Wang等人,Stroke, 2013;Xiang、Chao等人,Rev Neurosci, 2015;Lee、Lee等人,J Neuroinflammation, 2015)。 由於在搔癢症及疼痛(包括急性、慢性、發炎性及神經病性疼痛)之情形下涉及經由IRAK4之TLR介導之信號及IL-1受體家族介導之信號,故可透過IRAK4之抑制推論所提及之適應症中存在治療效應。疼痛之實例包括感覺過敏、觸摸痛、經前疼痛、子宮內膜異位症相關性疼痛、手術後疼痛、間質性膀胱炎、CRPS (複雜性區域疼痛症候群)、三叉神經痛、前列腺炎、由脊髓損傷造成之疼痛、發炎誘導之疼痛、腰部疼痛、癌性疼痛、化學療法相關性疼痛、HIV治療誘導之神經病變、燒傷誘導之疼痛及慢性疼痛(Wolf、Livshits等人,Brain, Behavior, and Immunity, 2008;Kim、Lee等人,Toll-like Receptors:Roles in Infection and Neuropathology, 2009;del Rey、Apkarian等人,Annals of the New York Academy of Sciences, 2012;Guerrero、Cunha等人,European Journal of Pharmacology, 2012;Kwok、Hutchinson等人,PLoS ONE, 2012;Nicotra、Loram等人,Experimental Neurology, 2012;Chopra及Cooper,J Neuroimmune Pharmacol, 2013;David、Ratnayake等人,Neurobiology of Disease, 2013;Han、Zhao等人,Neuroscience, 2013;Liu及Ji,Pflugers Arch., 2013;Stokes、Cheung等人,Journal of Neuroinflammation, 2013;Zhao、Zhang等人,Neuroscience, 2013;Liu、Zhang等人,Cell Research, 2014;Park、Stokes等人,Cancer Chemother Pharmacol, 2014;Van der Watt、Wilkinson等人,BMC Infect Dis, 2014;Won, K. A.、M. J. Kim等人,J Pain, 2014;Min、Ahmad等人,Photochem Photobiol., 2015;Schrepf、Bradley等人,Brain Behav Immun, 2015;Wong, L.、J. D. Done等人,Prostate, 2015)。 此亦適用於一些腫瘤病症。特定淋巴瘤(例如ABC-DLBCL (經活化B細胞擴散大細胞B細胞淋巴瘤)、外套細胞淋巴瘤及瓦爾登斯特倫氏病(Waldenström's disease))亦及慢性淋巴白血病、黑色素瘤、胰臟腫瘤及肝細胞癌之特徵在於MyD88突變或MyD88活性變化,其可藉由IRAK4抑制劑來治療(Ngo、Young等人,Nature, 2011;Puente、Pinyol等人,Nature, 2011;Ochi、Nguyen等人,J Exp Med, 2012;Srivastava、Geng等人,Cancer Research, 2012;Treon、Xu等人,New England Journal of Medicine, 2012;Choi、Kim等人,Human Pathology, 2013;(Liang、Chen等人,Clinical Cancer Research, 2013)。另外,MyD88在ras依賴性腫瘤中起重要作用,且因此IRAK4抑制劑亦適於該腫瘤之治療(Kfoury, A.、K. L. Corf等人,Journal of the National Cancer Institute, 2013)。亦可透過IRAK4之抑制而推論在乳癌、卵巢癌、結腸直腸癌、頭頸癌、肺癌、前列腺癌中具有治療效應,乃因所提及之適應症與信號傳導路徑相關(Szczepanski、Czystowska等人,Cancer Res, 2009;Zhang、He等人,Mol Biol Rep, 2009;Wang、Qian等人,Br J Cancer Kim, 2010;Jo等人,World J Surg Oncol, 2012;Zhao、Zhang等人;Front Immunol, 2014;Chen、Zhao等人,Int J Clin Exp Pathol, 2015)。 發炎病症(例如CAPS (隱熱蛋白相關性週期症候群),包括FCAS (家族性寒冷性自體發炎症候群)、MWS (穆-韋二氏症候群(Muckle-Wells syndrome))、NOMID (新生兒發作型多系統發炎性疾病)及CONCA (慢性嬰兒、神經、皮膚及關節)症候群)、FMF (家族性地中海熱)、HIDS (高IgD症候群)、TRAPS (腫瘤壞死因子受體1相關之週期性症候群)、幼年型特發性關節炎、成人發作性斯蒂爾氏病、阿達替德-貝歇氏症(Adamantiades-Behçet's disease)、類風濕性關節炎、骨關節炎、乾燥性角結膜炎、PAPA症候群(化膿性關節炎、壞疽性膿皮病及痤瘡)、施尼茨勒氏症候群(Schnitzler's syndrome)及薛格連氏症候群可藉由阻斷IL-1信號路徑來治療;因此,在本文中,IRAK4抑制劑亦適於治療所提及之疾病(Narayanan、Corrales等人,Cornea, 2008;Brenner、Ruzicka等人,British Journal of Dermatology, 2009;Henderson及Goldbach-Mansky,Clinical Immunology, 2010;Dinarello, European Journal of Immunology, 2011;Gul、Tugal-Tutkun等人,Ann Rheum Dis, 2012;Pettersson, Annals of MedicinePetterson, 2012;Ruperto、Brunner等人,New England Journal of Medicine, 2012;Nordström、Knight等人,The Journal of Rheumatology, 2012;Vijmasi、Chen等人,Mol Vis, 2013;Yamada、Arakaki等人,Opinion on Therapeutic Targets, 2013;de Koning, Clin Transl Allergy, 2014)。IL-33R之配體IL-33特定地參與急性腎衰竭之發病機制,且因此抑制IRAK4用於預防及/或治療係適宜治療方法(Akcay、Nguyen等人,Journal of the American Society of Nephrology, 2011)。IL-1受體家族之組份與心肌梗塞、不同肺部病症(例如氣喘、COPD、特發性間質性肺炎、過敏性鼻炎、肺纖維化及急性呼吸窘迫症候群(ARDS))相關,且因此預計藉助IRAK4之抑制在所提及之適應症中具有預防及/或治療作用(Kang、Homer等人,The Journal of Immunology, 2007;Imaoka、Hoshino等人,European Respiratory Journal, 2008;Couillin、Vasseur等人,The Journal of Immunology, 2009;Abbate、Kontos等人,The American Journal of Cardiology, 2010;Lloyd, Current Opinion in Immunology, 2010;Pauwels、Bracke等人,European Respiratory Journal, 2011;Haenuki、Matsushita等人,Journal of Allergy及Clinical Immunology, 2012;Yin、Li等人,Clinical & Experimental Immunology, 2012;Abbate、Van Tassell等人,The American Journal of Cardiology, 2013;Alexander-Brett等人,The Journal of Clinical Investigation, 2013;Bunting、Shadie等人,BioMed Research International, 2013;Byers、Alexander-Brett等人,The Journal of Clinical Investigation, 2013;Kawayama、Okamoto等人,J Interferon Cytokine Res, 2013;Martínez-González、Roca等人,American Journal of Respiratory Cell amd Molecular Biology, 2013;Nakanishi、Yamaguchi等人,PLoS ONE, 2013;Qiu、Li等人,Immunology, 2013;Li、Guabiraba等人,Journal of Allergy and Clinical Immunology, 2014;Saluja、Ketelaar等人,Molecular Immunology, 2014;Lugrin、Parapanov等人,The Journal of Immunology, 2015)。 先前技術揭示多種IRAK4抑制劑(例如參見Annual Reports in Medicinal Chemistry (2014), 49, 117 – 133)。 US8293923及US20130274241揭示具有3-取代之吲唑結構之IRAK4抑制劑。未闡述2-取代之吲唑。 WO2013106254及WO2011153588揭示2,3-二取代之吲唑衍生物。 WO2007091107闡述2-取代之吲唑衍生物用於治療杜興氏肌肉營養不良症(Duchenne muscular dystrophy)。僅明確闡述在2位具有苯基之吲唑衍生物。 WO2015091426闡述在2位經甲醯胺側鏈取代之吲唑,例如實例WO2015091426-64。未公開之申請案EP 14195032.9 (申請日期2014年11月26日)闡述在6位經烷基取代之2-取代之吲唑,其中烷基經羥基取代。未闡述在6位具有氯原子或烷氧基之2-取代之吲唑。 WO2015104662揭示具有以下通式之2-取代之吲唑:其中R2 係烷基或環烷基。明確闡述在2位具有甲基、2-甲氧基乙基及環戊基之2-取代之吲唑(實例1、4及76)。實例117亦闡述在1位具有羥基乙基取代基之吲唑衍生物。然而,未闡述在1位或2位具有3-羥基-3-甲基丁基取代基之吲唑衍生物。 在2位具有羥基取代之烷基之吲唑通常由通式涵蓋,但其在WO2015104662中未明確揭示。 除在吲唑上1及2位處之上文所述取代型式以外,WO2015104662闡述在6位具有取代之吲唑,其中R1 定義為如下:烷基、氰基、-NRa Rb 或選自環烷基、芳基或雜環基之視情況經取代之基團,其中取代基獨立地係烷基、烷氧基、鹵素、羥基、羥基烷基、胺基、胺基烷基、硝基、氰基、鹵代烷基、鹵代烷氧基、-OCOCH2 -O-烷基、-OP(O)(O-烷基)2 或-CH2 -OP(O)(O-烷基)2 。在WO2015104662中針對R1 闡述之6位處取代基之實例係環丙基、環己基、氰基、3-氟苯基及飽和雜環取代基。 因此,未闡述在6位具有烷氧基或氯原子之吲唑衍生物。 本發明解決之問題係提供充當介白素-1受體相關性激酶-4 (IRAK4)之抑制劑之新穎化合物。Human IRAK4 (interleukin-1 receptor-associated kinase 4) plays a key role in the activation of the immune system. Therefore, this kinase is an important therapeutic target molecule for the development of inflammatory inhibitory substances. IRAK4 is expressed by a variety of cells and mediates steroid receptors (TLR) (except TLR3) and interleukin (IL)-1β family receptors (by IL-1R (receptor), IL-18R, IL-33R and IL) Signal transduction of -36R composition) (Janeway and Medzhitov, Annu. Rev. Immunol., 2002; Dinarello, Annu. Rev. Immunol., 2009; Flannery and Bowie, Biochemical Pharmacology, 2010). IRAK4 knockout mice and human cells from patients lacking IRAK4 do not respond to TLR (except TLR3) and IL-1β family stimuli (Suzuki, Suzuki et al, Nature, 2002; Davidson, Currie et al, The Journal Of Immunology, 2006; Ku, von Bernuth et al, JEM, 2007; Kim, Staschke et al, JEM, 2007). Binding of TLR ligands or IL-1β family ligands to individual receptors results in the recruitment and binding of MyD88 [myeloid differentiation primary response gene (88)] to the receptor. Thus, MyD88 interacts with IRAK4, resulting in the formation of an active complex that interacts with the kinase IRAK1 or IRAK2 and activates the kinases (Kollewe, Mackensen et al, Journal of Biological Chemistry, 2004; Precious et al, J. Biol Chem., 2009). Thus, the NF (nuclear factor)-kappaB signaling pathway and the MAPK (mitogen-activated protein kinase) signaling pathway are activated (Wang, Deng et al, Nature, 2001). Activation of the NF-κB signaling pathway and the MAPK signaling pathway can result in processes associated with different immune processes. For example, various inflammatory signaling molecules and enzymes (such as interleukins, chemokines, and COX-2 (cyclooxygenase-2)) have increased expression, and inflammation-related genes (such as COX-2, The mRNA stability of IL-6 (interleukin-6), IL-8) is increased (Holtmann, Enninga et al, Journal of Biological Chemistry, 2001; Datta, Novotny et al, The Journal of Immunology, 2004). In addition, these processes may be associated with proliferation and differentiation of specific cell types (eg, mononuclear spheres, macrophages, dendritic cells, T cells, and B cells) (Wan, Chi et al, Nat Immunol, 2006; McGettrick and J). O'Neill, British Journal of Haematology, 2007). A direct comparison of wild-type (WT) mice with genetically modified animals with a kinase-deactivated form of IRAK4 (IRAK4 KDKI) has demonstrated an important role for IRAK4 in the pathology of various inflammatory conditions. In animal models of multiple sclerosis, atherosclerosis, myocardial infarction, and Alzheimer's disease, IRAK4 KDKI animals have improved clinical manifestations (Rekhter, Staschke et al, Biochemical and Biophysical Research Communication, 2008) Maekawa, Mizue et al., Circulation, 2009; Staschke, Dong et al, The Journal of Immunology, 2009; Kim, Febbraio et al, The Journal of Immunology, 2011; Cameron, Tse et al, The Journal of Neuroscience, 2012) . In addition, it was found that the absence of IRAK4 in animal models protects against viral-induced myocarditis due to improved antiviral responses and reduced systemic inflammation (Valaperti, Nishii et al., Circulation, 2013). It has also been shown that the performance of IRAK4 is associated with the degree of Vogt-Koyanagi-Harada syndrome (Sun, Yang et al., PLoS ONE, 2014). In addition, the high correlation of IRAK4 to immune complex-mediated IFNα (interferon-α) production by plasmacytoid dendritic cells has demonstrated that this IFNα production is key in the pathogenesis of systemic lupus erythematosus (SLE). Process (Chiang et al., The Journal of Immunology, 2010). In addition, signaling pathways are associated with obesity (Ahmad, R., P. Shihab et al., Diabetology & Metabolic Syndrome, 2015). In addition to the essential role of IRAK4 in innate immunity, it is also suggested that IRAK4 affects the differentiation of cells known as Th17 T cells (adaptive immune components). In the absence of IRAK4 kinase activity, less IL-17 producing T cells (Th17 T cells) were produced compared to WT mice. Inhibition of IRAK4 enables prevention and/or treatment of atherosclerosis, type 1 diabetes, rheumatoid arthritis, spondylarthritis (especially psoriatic spondyloarthritis and Behdejelev's disease), lupus erythematosus, psoriasis , leukoplakia, giant cell arteritis, chronic inflammatory bowel disease and viral disorders (eg HIV (human immunodeficiency virus), hepatitis virus) (Staschke et al, The Journal of Immunology, 2009; Marquez et al, Ann Rheum Dis, 2014) ;Zambrano-Zaragoza et al, International Journal of Inflammation, 2014; Wang et al, Experimental and Therapeutic Medicine, 2015; Ciccia et al, Rheumatology, 2015). Since IRAK4 plays an important role in the TLR (except TLR3) and the MyD88-mediated signaling cascade of the IL-1 receptor family, inhibition of IRAK4 can be used to prevent and/or treat conditions mediated by the mentioned receptors. The components of the TLR and IL-1 receptor families are involved in the pathogenesis of rheumatoid arthritis, psoriatic arthritis, myasthenia gravis, vasculitis (eg Behçet's disease, granulation with polyangiitis) Swollen disease and giant cell arteritis), pancreatitis, systemic lupus erythematosus, dermatomyositis and polymyositis, metabolic syndrome, including, for example, insulin resistance, hypertension, dyslipoproteinemia, and obesity , diabetes (type 1 and type 2), diabetic nephropathy, osteoarthritis, Sjögren syndrome, and sepsis (Yang, Tuzun et al, J Immunol, 2005; Candia, Marquez et al, The Journal of Rheumatology, 2007; Scanzello, Plaas et al. Curr Opin Rheumatol, 2008; Deng, Ma-Krupa et al, Circ Res, 2009; Roger, Froidevaux et al, PNAS, 2009; Devaraj, Tobias et al, Arterioscler Thromb Vasc Biol, 2011; Kim Cho et al., Clin Rheumatol, 2010; Carrasco et al., Clinical and Experimental Rheumatology, 2011; Gambuzza, Licata et al., Journal of Neuroimmunology, 2011; Fresno, Archives Of Physiology And Biochemistry, 2011; Volin and Koch, J Interferon Cytokine Res, 2011; Akash, Shen et al, Journal of Pharmaceutical Sciences, 2012; Goh and Midwood, Rheumatology, 2012; Dasu, Ramirez et al, Clinical Science, 2012; Ouziel , Gustot et al, Am J Patho, 2012; Ramirez and Dasu, Curr Diabetes Rev, 2012, Okiyama et al, Arthritis Rheum, 2012; Chen et al, Arthritis Research & Therapy, 2013; Holle, Windmoller et al, Rheumatology (Oxford ), 2013; Li, Wang et al, Pharmacology & Therapeutics, 2013; Sedimbi, Hagglof et al, Cell Mol Life Sci, 2013; Caso, Costa et al, Mediators of Inflammation, 2014; Cordiglieri, Marolda et al, J Autoimmun, 2014; Jiaal, Major et al, J Diabetes Complications, 2014; Kaplan, Yazgan et al, Scand J Gastroenterol, 2014; Talabot-Aye et al, Cytokine, 2014; Zong, Dorph et al, Ann Rheum Di, 2014; Ballak, Stienstra et al., Cytokine, 2015; Timper, Seelig et al., J Diabetes Complications, 2015). Skin diseases (eg psoriasis, atopic dermatitis, Kindler's syndrome, bullous pemphigoid, allergic contact dermatitis, alopecia areata, abnormal acne and acne vulgaris) and IRAK4 Guided TLR signaling pathway or IL-1R family correlation (Schmidt, Mittnacht et al, J Dermatol Sci, 1996; Hoffmann, J Investig Dermatol Symp Proc, 1999; Gilliet, Conrad et al, Archives of Dermatology, 2004; Niebuhr, Langnickel Et al, Allergy, 2008; Miller, Adv Dermatol, 2008; Terhorst, Kalali et al, Am J Clin Dermatol, 2010; Viguier, Guigue et al, Annals of Internal Medicine, 2010; Cevikbas, Steinhoff, J Invest Dermatol, 2012; Minkis, Aksentijevich et al., Archives of Dermatology, 2012; Dispenza, Wolpert et al, J Invest Dermatol, 2012; Minkis, Aksentijevich et al, Archives of Dermatology, 2012; Gresnigt and van de Veerdonk, Seminars in Immunology, 2013; Selway, Kurczab et al, BMC Dermatology, 2013; Sedimbi, Hagglof et al, Cell Mol Life Sci, 2013; Wollina, Koch et al I Ndian Dermatol Online, 2013; Foster, Baliwag et al., The Journal of Immunology, 2014). Pulmonary disorders such as pulmonary fibrosis, obstructive pulmonary disease (COPD), acute respiratory distress syndrome (ARDS), acute lung injury (ALI), interstitial lung disease (ILD), sarcoma-like disease, and pulmonary hypertension are also shown Various TLR-mediated signal pathway correlations. The pathogenesis of pulmonary disorders is a process mediated by infection or mediated by non-infectious methods (Ramirez Cruz, Maldonado Bernal et al, Rev Alerg Mex, 2004; Jeyaseelan, Chu et al, Infection and Immunity, 2005; Seki , Tasaka et al, Inflammation Research, 2010; Xiang, Fan et al, Mediators of Inflammation, 2010; Margaritopoulos, Antoniou et al, Fibrogenesis & Tissue Repair, 2010; Hilberath, Carlo et al, The FASEB Journal, 2011; Nadiger, Prefontaine Et al, Respiratory Research, 2011; Kovach and Standiford, International Immunopharmacology, 2011; Bauer, Shapiro et al, Mol Med, 2012; Deng, Yang et al, PLoS One, 2013; Freeman, Martinez et al, Respiratory Research, 2013; Dubaniewicz, A., Human Immunology, 2013). TLR and members of the IL-1R family are also involved in the pathogenesis of other inflammatory conditions such as allergies, Beth's disease, gout, lupus erythematosus, adult-onset Still's disease, pericarditis and Chronic inflammatory bowel disorders (eg, ulcerative colitis and Crohn's disease), transplant rejection, and graft versus host response, and thus inhibition of IRAK4 is a suitable preventive and/or therapeutic method (Liu-Bryan, Scott) Et al, Arthritis & Rheumatism, 2005; Piggott, Eisenbarth et al, J Clin Inves, 2005; Christensen, Shupe et al, Immunity, 2006; Cario, Inflammatory Bowel Diseases, 2010; Nickerson, Christensen et al, The Journal of Immunology, 2010 ; Rakoff-Nahoum, Hao et al, Immunity, 2006; Heimesaat, Fischer et al, PLoS ONE, 2007; Heimesaat, Nogai et al, Gut, 2010; Kobori, Yagi et al, J Gastroenterol, 2010; Schmidt, Raghavan, etc. , Nat Immunol, 2010; Shi, Mucsi et al, Immunological Reviews, 2010; Leventhal and Schroppel, Kidney Int, 2012; Chen, Lin et al, Arthritis Res Ther, 2013; Hao, Liu et al, Curr Opin Gastroenterol, 2013; Kreisel and Goldstein, Transplant International, 2013; Li, Wang et al, Pharmacology & Therapeutics, 2013; Walsh, Carthy et al, Cytokine & Growth Factor Reviews, 2013; Zhu , Jiang et al, Autoimmunity, 2013; Yap and Lai, Nephrology, 2013; Vennegaard, Dyring-Andersen, et al, Contact Dermatitis, 2014; D'Elia, Brucato et al, Clin Exp Rheumatol, 2015; Jain, Thongprayoon, etc. Am J Cardiol., 2015; Li, Zhang et al., Oncol Rep., 2015). Gynecological disorders mediated by the TLR and IL-1R families (eg, uterine adenomyosis, dysmenorrhea, painful intercourse and endometriosis, especially endometriosis-related pain and other endometriosis) Sexual symptoms (such as dysmenorrhea, painful intercourse, dysuria, and difficulty in defecation) can be positively affected by the prophylactic and/or therapeutic use of IRAK4 inhibitors (Akoum, Lawson et al., Human Reproduction, 2007; Allhorn, Boing et al. Reproductive Biology and Endocrinology, 2008; Lawson, Bourcier et al, Journal of Reproductive Immunology, 2008; Sikora, Mielczarek-Palacz et al, American Journal of Reproductive Immunology, 2012; Khan, Kitajima et al, Journal of Obstetrics and Gynaecology Research, 2013; Santulli, Borghese et al, Human Reproduction, 2013). Prophylactic and/or therapeutic use of IRAK4 inhibitors can also have a positive effect on atherosclerosis (Seneviratne, Sivagurunathan et al, Clinica Chimica Acta, 2012; Falck - Hansen, Kassiteridi et al., International Journal of Molecular Sciences, 2013; Sedimbi Hagglof et al, Cell Mol Life Sci, 2013). In addition to the conditions already mentioned, already in ocular conditions (eg retinal ischemia, keratitis, allergic conjunctivitis, keratoconjunctivitis sicca, macular degeneration and uveal layer) The IRAK4-mediated TLR process is described in the pathogenesis of inflammation (Kaarniranta and Salminen, J Mol Med (Berl), 2009; Sun and Pearlman, Investigative Ophthalmology & Visual Science, 2009; Redfern and McDermott, Experimental Eye Research, 2010; Kezic , Taylor et al, J Leukoc Biol, 2011; Chang, McCluskey et al, Clinical & Experimental Ophthalmology, 2012; Guo, Gao et al, Immunol Cell Biol, 2012; Lee, Hattori et al, Investigative Ophthalmology & Visual Science, 2012; Qi, Zhao et al., Investigative Ophthalmology & Visual Science, 2014). Inhibition of IRAK4 is also a suitable treatment for fibrotic disorders (eg liver fibrosis, myocarditis, primary biliary cirrhosis, cystic fibrosis) (Zhao, Zhao et al, Scand J Gastroenterol, 2011; Benias, Gopal, etc.) People, Clin Res Hepatol Gastroenterol, 2012; Yang, L. and E. Seki, Front Physiol, 2012; Liu, Hu et al, Biochim Biophys Acta., 2015). Since IRAK4 has a critical role in the conditions mediated by the TLR and IL-1R families, it is possible to treat chronic liver conditions, such as fatty liver hepatitis and especially nonalcoholic fatty liver, in a prophylactic and/or therapeutic manner using IRAK4 inhibitors. Disease (NAFLD) and/or nonalcoholic steatohepatitis (NASH), alcoholic steatohepatitis (ASH) (Nozaki, Saibara et al, Alcohol Clin Exp Res, 2004; Csak, T., A. Velayudham et al, Am J Physiol Gastrointest Liver Physiol, 2011; Miura, Kodama et al, Gastroenterology, 2010; Kamari, Shaish et al, J Hepatol, 2011; Ye, Li et al, Gut, 2012; Roh, Seki, J Gastroenterol Hepatol, 2013; Ceccarelli, S., V. Nobili et al, World J Gastroenterol, 2014; Miura, Ohnishi, World J Gastroenterol, 2014; Stojsavljevic, Palcic et al, World J Gastroenterol, 2014). Due to the important role of IRAK4 in TLR-mediated processes, inhibition of IRAK4 also enables the treatment and/or prevention of cardiovascular and neurological disorders such as myocardial reperfusion injury, myocardial infarction, hypertension (Oyama, Blais et al., Circulation) , 2004; Timmers, Sluijter et al, Circulation Research, 2008; Fang and Hu, Med Sci Monit, 2011; Bijani, International Reviews of Immunology, 2012; Bomfim, Dos Santos et al, Clin Sci (Lond), 2012; Frangogiannis, European Journal of Clinical Investigation, 2013; Thompson and Webb, Clin Sci (Lond), 2013; Hernanz, Martínez-Revelles et al, British Journal of Pharmacology, 2015; Frangogiannis, Curr Opin Cardiol, 2015; Bomfim, Echem et al. , Life Sciences, 2015), and Alzheimer's disease, stroke, craniocerebral trauma, amyotrophic lateral sclerosis (ALS), and Parkinson's disease (Brough, Tyrrell et al., Trends in Pharmacological Sciences, 2011; Carty and Bowie, Biochemical Pharmacology, 2011; Denes, Kitazawa, Cheng et al, The Journal of Immunol Ogy, 2011; Lim, Kou et al, The American Journal of Pathology, 2011; Béraud and Maguire-Zeiss, Parkinsonism & Related Disorders, 2012; Denes, Wilkinson et al, Disease Models & Mechanisms, 2013; Noelker, Morel et al, Sci Rep., 2013; Wang, Wang et al., Stroke, 2013; Xiang, Chao et al., Rev Neurosci, 2015; Lee, Lee et al., J Neuroinflammation, 2015). Inhibition of IRAK4 is implicated in the context of pruritus and pain (including acute, chronic, inflammatory, and neuropathic pain) involving TLR-mediated signaling via IRAK4 and IL-1 receptor-mediated signaling. There is a therapeutic effect in the indications mentioned. Examples of pain include hyperesthesia, touch pain, premenstrual pain, endometriosis-related pain, post-operative pain, interstitial cystitis, CRPS (complex regional pain syndrome), trigeminal neuralgia, prostatitis, Pain caused by spinal cord injury, inflammation-induced pain, lumbar pain, cancer pain, chemotherapy-related pain, HIV-induced neuropathy, burn-induced pain, and chronic pain (Wolf, Livshits et al, Brain, Behavior, And Immunity, 2008; Kim, Lee et al, Toll-like Receptors: Roles in Infection and Neuropathology, 2009; del Rey, Apkarian et al, Annals of the New York Academy of Sciences, 2012; Guerrero, Cunha et al, European Journal Of Pharmacology, 2012; Kwok, Hutchinson et al, PLoS ONE, 2012; Nicotra, Loram et al, Experimental Neurology, 2012; Chopra and Cooper, J Neuroimmune Pharmacol, 2013; David, Ratnayake et al, Neurobiology of Disease, 2013; Han , Zhao et al., Neuroscience, 2013; Liu and Ji, Pflugers Arch., 2013; Stokes, Cheung et al., Jou Rnal of Neuroinflammation, 2013; Zhao, Zhang et al, Neuroscience, 2013; Liu, Zhang et al, Cell Research, 2014; Park, Stokes et al, Cancer Chemother Pharmacol, 2014; Van der Watt, Wilkinson et al, BMC Infect Dis , 2014; Won, KA, MJ Kim, et al, J Pain, 2014; Min, Ahmad, etc., Photochem Photobiol., 2015; Schrepf, Bradley et al, Brain Behav Immun, 2015; Wong, L., JD Done, etc. , Prostate, 2015). This also applies to some tumor conditions. Specific lymphomas (eg ABC-DLBCL (activated B-cell-diffuse large cell B-cell lymphoma), mantle cell lymphoma, and Waldenström's disease) as well as chronic lymphocytic leukemia, melanoma, pancreas Tumor and hepatocellular carcinoma are characterized by changes in MyD88 mutation or MyD88 activity, which can be treated by IRAK4 inhibitors (Ngo, Young et al, Nature, 2011; Puente, Pinyol et al, Nature, 2011; Ochi, Nguyen et al. , J Exp Med, 2012; Srivastava, Geng et al, Cancer Research, 2012; Treon, Xu et al, New England Journal of Medicine, 2012; Choi, Kim et al, Human Pathology, 2013; (Liang, Chen et al, Clinical Cancer Research, 2013). In addition, MyD88 plays an important role in ras-dependent tumors, and thus IRAK4 inhibitors are also suitable for the treatment of this tumor (Kfoury, A., KL Corf et al, Journal of the National Cancer Institute, 2013). It can also be inferred through the inhibition of IRAK4 in breast cancer, ovarian cancer, colorectal cancer, head and neck cancer, lung cancer, prostate cancer, because of the indications and signal transmission mentioned. Path correlation (Szczepanski, Czystowska et al, Cancer Res, 2009; Zhang, He et al, Mol Biol Rep, 2009; Wang, Qian et al, Br J Cancer Kim, 2010; Jo et al, World J Surg Oncol, 2012; Zhao, Zhang et al; Front Immunol, 2014; Chen, Zhao et al, Int J Clin Exp Pathol, 2015) Inflammatory disorders (eg, CAPS (Hidden Heat Protein Associated Cycle Syndrome), including FCAS (familial cold autologous) Inflammation group), MWS (Muckle-Wells syndrome), NOMID (neonatal onset multisystem inflammatory disease) and CONCA (chronic infant, nerve, skin and joint syndrome), FMF (family) Sexual Mediterranean fever), HIDS (high IgD syndrome), TRAPS (tumor syndrome associated with tumor necrosis factor receptor 1), juvenile idiopathic arthritis, adult onset Stil's disease, adalter-bescher Adamantiades-Behçet's disease, rheumatoid arthritis, osteoarthritis, keratoconjunctivitis sicca, PAPA syndrome (suppurative arthritis, gangrenous pyoderma and acne), Schnitzler's syndrome And Xue Gelian The population can be treated by blocking the IL-1 signaling pathway; therefore, IRAK4 inhibitors are also suitable herein for the treatment of the mentioned diseases (Narayanan, Corrales et al, Cornea, 2008; Brenner, Ruzicka et al, British Journal of Dermatology, 2009; Henderson and Goldbach-Mansky, Clinical Immunology, 2010; Dinarello, European Journal of Immunology, 2011; Gul, Tugal-Tutkun et al, Ann Rheum Dis, 2012; Pettersson, Annals of MedicinePetterson, 2012; , Brunner et al, New England Journal of Medicine, 2012; Nordström, Knight et al, The Journal of Rheumatology, 2012; Vijmasi, Chen et al, Mol Vis, 2013; Yamada, Arakaki et al, Opinion on Therapeutic Targets, 2013; De Koning, Clin Transl Allergy, 2014). The ligand IL-33 of IL-33R is specifically involved in the pathogenesis of acute renal failure, and thus inhibits IRAK4 for prevention and/or treatment of a suitable therapeutic method (Akcay, Nguyen et al., Journal of the American Society of Nephrology, 2011) ). The components of the IL-1 receptor family are associated with myocardial infarction, different pulmonary conditions (eg, asthma, COPD, idiopathic interstitial pneumonia, allergic rhinitis, pulmonary fibrosis, and acute respiratory distress syndrome (ARDS)), and It is therefore expected that inhibition by IRAK4 has a prophylactic and/or therapeutic effect in the mentioned indications (Kang, Homer et al, The Journal of Immunology, 2007; Imaoka, Hoshino et al, European Respiratory Journal, 2008; Couillin, Vasseur Et al, The Journal of Immunology, 2009; Abbate, Kontos et al, The American Journal of Cardiology, 2010; Lloyd, Current Opinion in Immunology, 2010; Pauwels, Bracket et al, European Respiratory Journal, 2011; Haenuki, Matsushita et al. , Journal of Allergy and Clinical Immunology, 2012; Yin, Li et al, Clinical & Experimental Immunology, 2012; Abbate, Van Tassell et al, The American Journal of Cardiology, 2013; Alexander-Brett et al, The Journal of Clinical Investigation, 2013; Bunting, Shadie et al, BioMed Research International, 2013; Byers, Alexander-Brett People, The Journal of Clinical Investigation, 2013; Kawayama, Okamoto et al, J Interferon Cytokine Res, 2013; Martínez-González, Roca et al, American Journal of Respiratory Cell amd Molecular Biology, 2013; Nakanishi, Yamaguchi et al, PLoS ONE , 2013; Qiu, Li et al, Immunology, 2013; Li, Guabiraba et al, Journal of Allergy and Clinical Immunology, 2014; Saluja, Ketelaar et al, Molecular Immunology, 2014; Lugrin, Parapanov et al, The Journal of Immunology, 2015). The prior art discloses a variety of IRAK4 inhibitors (see, for example, Annual Reports in Medicinal Chemistry (2014), 49, 117-133). US8293923 and US20130274241 disclose IRAK4 inhibitors having a 3-substituted carbazole structure. 2-substituted carbazoles are not illustrated. WO2013106254 and WO2011153588 disclose 2,3-disubstituted oxazole derivatives. WO2007091107 describes 2-substituted carbazole derivatives for the treatment of Duchenne muscular dystrophy. Only the carbazole derivatives having a phenyl group at the 2-position are specifically described. WO2015091426 describes carbazoles substituted at the 2-position vialamine, for example from WO2015091426-64. The unpublished application EP 14195032.9 (filed on November 26, 2014) describes a 2-substituted oxazole substituted with an alkyl group at the 6-position, wherein the alkyl group is substituted by a hydroxy group. A 2-substituted carbazole having a chlorine atom or an alkoxy group at the 6-position is not illustrated. WO2015104662 discloses 2-substituted carbazoles having the general formula: Wherein R 2 is an alkyl group or a cycloalkyl group. The 2-substituted oxazoles having a methyl group, a 2-methoxyethyl group and a cyclopentyl group at the 2-position are exemplified (Examples 1, 4 and 76). Example 117 also illustrates a carbazole derivative having a hydroxyethyl substituent at the 1-position. However, a carbazole derivative having a 3-hydroxy-3-methylbutyl substituent at the 1- or 2-position is not illustrated. The oxazole having a hydroxy-substituted alkyl group at the 2-position is generally encompassed by the general formula, but it is not explicitly disclosed in WO2015104662. In addition to the above-described substitution patterns at the 1 and 2 positions on the carbazole, WO2015104662 describes substituted carbazoles at the 6 position, wherein R 1 is defined as follows: alkyl, cyano, -NR a R b or selected An optionally substituted group derived from a cycloalkyl, aryl or heterocyclic group, wherein the substituent is independently alkyl, alkoxy, halogen, hydroxy, hydroxyalkyl, amine, aminoalkyl, nitrate Base, cyano, haloalkyl, haloalkoxy, -OCOCH 2 -O-alkyl, -OP(O)(O-alkyl) 2 or -CH 2 -OP(O)(O-alkyl) 2 . Examples of substituents at the 6 position described for R 1 in WO2015104662 are cyclopropyl, cyclohexyl, cyano, 3-fluorophenyl and saturated heterocyclic substituents. Therefore, a carbazole derivative having an alkoxy group or a chlorine atom at the 6-position is not explained. The problem addressed by the present invention is to provide novel compounds that act as inhibitors of interleukin-1 receptor-associated kinase-4 (IRAK4).

本發明提供通式(I)化合物其中 R1 代表氯或O-R4 ; R2 代表C1 -C8 -烷基, 其可視情況彼此獨立地經選自由以下組成之群之相同或不同取代基單取代或多取代:1至5個氟原子、1至3個羥基、氧雜環丁基、四氫呋喃基、吡喃基、側氧基及C1 -C6 -烷氧基, 其中C1 -C6 -烷氧基 可經氟或 經羥基或 經C(=O)OH、C(=O)Me、C(=O)Et、C(=O)NH2 單取代至三取代; R3 代表選自以下之基團: , 其中R5 代表氫、C3 -C6 -環烷基或C1 -C6 -烷基,其中C1 -C6 -烷基可視情況經環丙基、氰基及羥基單取代至三取代,其中每一取代基可僅出現一次,或經氟原子單取代至五取代; R6 代表氫、氟或甲基, 或 R3 代表選自以下之基團:其中R7 代表氫、C3 -C6 -環烷基、氰基、NH2 、NH(C1 -C6 -烷基)、N(C1 -C6 -烷基)2 、吡咯啶-1-基、六氫吡啶-1-基、嗎啉-4-基、4-甲基六氫吡嗪-1-基 或代表C1 -C6 -烷基,其中C1 -C6 -烷基可視情況經環丙基、氰基及羥基單取代至三取代,其中每一取代基可僅出現一次,或經氟原子單取代至五取代; R8 、R9 、R10 代表氫、甲基或氟, 或 R3 代表其中R11 代表 C3 -C6 -環烷基或C1 -C6 -烷基,其中C1 -C6 -烷基可視情況經環丙基、氰基及羥基單取代至三取代,其中每一取代基可僅出現一次,或經氟原子單取代至五取代; 且R12 代表氫、氟或C1 -C6 -烷基; 或 R3 代表 6,7-二氫-4H-吡唑并[5,1-c][1,4]噁嗪-2-基、5-甲基-4,5,6,7-四氫吡唑并[1,5-a]吡嗪-2-基或4,5,6,7-四氫吡唑并[1,5-a]吡嗪-2-基 或代表 吡唑并[1,5-a]嘧啶-3-基、吡咯并[2,1-f][1,2,4]三嗪-7-基、吡咯并[1,2-b]嗒嗪-7-基、噻吩并[2,3-b]吡嗪-7-基、5-胺基吡唑并[1,5-a]嘧啶-3-基、2-胺基吡咯并[2,1-f][1,2,4]三嗪-7-基、2-胺基吡咯并[1,2-b]嗒嗪-7-基、2-胺基噻吩并[2,3-b]吡嗪-7-基; R4 代表飽和4員至7員雜環基或C3 -C7 -環烷基,其中該飽和雜環基及該C3 -C7 -環烷基可視情況經來自由以下組成之群之相同或不同取代基單取代或多取代:C1 -C6 -烷基、三氟甲基、2,2,2-三氟乙基、環丙基、C(=O)OH、C1 -C6 -烷氧基、三氟甲氧基、2,2,2-三氟乙氧基、氟、氯、氰基、羥基、NH2 、NHRa 、N(Ra )Rb , 或代表 C1 -C6 -烷基,其中C1 -C6 -烷基可視情況經來自由以下組成之群之相同或不同取代基單取代或多取代:氟、氯、氰基、羥基、C(=O)OC1 -C6 -烷基、C(=O)OH、C(=O)NH2 、C(=O)N(H)Ra 、C(=O)N(Ra )Rb 、SC1 -C6 -烷基、S(=O)2 -C1 -C6 -烷基、S(=O)2 NH2 、C1 -C6 -烷氧基、三氟甲氧基、2,2,2-三氟乙氧基、NH2 、NHRa 、N(Ra )Rb 、C3 -C7 -環烷基、四唑及4員至7員飽和雜環基, 其中C3 -C7 -環烷基及4員至7員飽和雜環基可視情況經來自由以下組成之群之相同或不同取代基單取代至四取代:氟、C1 -C4 -烷基、羥基、C(=O)OH、三氟甲基、2,2,2-三氟乙基、甲氧基、乙氧基、三氟甲氧基、環丙基、環丙基甲基、甲基磺醯基、NH2 、NHRa 、N(Ra )Rb ; Ra 代表C1 -C6 -烷基或C3 -C7 -環烷基, 其中C1 -C6 -烷基及C3 -C7 -環烷基可視情況經來自由以下組成之群之相同或不同取代基單取代或多取代:氟、羥基、氰基、C1 -C4 -烷基、C1 -C4 -烷氧基及C3 -C7 -環烷基; Rb 代表C1 -C6 -烷基或C3 -C7 -環烷基; 鍵處之*表示分子中之附接點, 及其非鏡像異構物、鏡像異構物、代謝物、鹽、溶劑合物或鹽之溶劑合物。 新穎IRAK4抑制劑尤其適於治療及預防特徵在於過反應免疫系統之增殖、代謝及發炎病症。此處尤其應提及發炎皮膚病症、心血管病症、肺病症、眼部病症、神經病症、疼痛病症及癌症。 另外,新穎IRAK4抑制劑適於治療及預防 •  自體免疫及發炎病症,尤其類風濕性關節炎、多發性硬化、全身性紅斑性狼瘡、脊椎關節炎及痛風, •  代謝失調,尤其肝臟病症(例如脂肪肝),及 •  婦科病症,尤其子宮內膜異位症及子宮內膜異位症相關性疼痛及其他子宮內膜異位症相關性症狀,例如痛經、性交疼痛、排尿困難及排糞困難。The present invention provides a compound of the formula (I) Wherein R 1 represents chloro or OR 4 ; R 2 represents C 1 -C 8 -alkyl, which may optionally be mono- or polysubstituted, respectively, via the same or different substituents selected from the group consisting of: 1 to 5 a fluorine atom, 1 to 3 hydroxyl groups, oxetanyl group, tetrahydrofuranyl group, pyranyl group, pendant oxy group, and C 1 -C 6 -alkoxy group, wherein the C 1 -C 6 -alkoxy group may be fluorine or Monosubstituted to trisubstituted by hydroxy or by C(=O)OH, C(=O)Me, C(=O)Et, C(=O)NH 2 ; R 3 represents a group selected from the group consisting of: Wherein R 5 represents hydrogen, C 3 -C 6 -cycloalkyl or C 1 -C 6 -alkyl, wherein the C 1 -C 6 -alkyl group may optionally be monosubstituted by cyclopropyl, cyano and hydroxy to three Substituting wherein each substituent may occur only once, or may be mono-substituted to penta-substituted with a fluorine atom; R 6 represents hydrogen, fluorine or methyl, or R 3 represents a group selected from the group consisting of: Wherein R 7 represents hydrogen, C 3 -C 6 -cycloalkyl, cyano, NH 2 , NH(C 1 -C 6 -alkyl), N(C 1 -C 6 -alkyl) 2 , pyrrolidine- 1-yl, hexahydropyridin-1-yl, morpholin-4-yl, 4-methylhexahydropyrazin-1-yl or a C 1 -C 6 -alkyl group, wherein C 1 -C 6 -alkane The group may be mono-substituted to trisubstituted by cyclopropyl, cyano and hydroxy, wherein each substituent may occur only once, or may be mono-substituted to penta-substituted through a fluorine atom; R 8 , R 9 , R 10 represent hydrogen, A Base or fluorine, or R 3 represents Wherein R 11 represents a C 3 -C 6 -cycloalkyl group or a C 1 -C 6 -alkyl group, wherein the C 1 -C 6 -alkyl group may be monosubstituted to trisubstituted by a cyclopropyl group, a cyano group and a hydroxy group, wherein Each substituent may occur only once, or may be mono-substituted to penta-substituted with a fluorine atom; and R 12 represents hydrogen, fluorine or C 1 -C 6 -alkyl; or R 3 represents 6,7-dihydro-4H-pyridyl Zoxa[5,1-c][1,4]oxazin-2-yl, 5-methyl-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrazine-2 -yl or 4,5,6,7-tetrahydropyrazolo[1,5-a]pyrazin-2-yl or represents pyrazolo[1,5-a]pyrimidin-3-yl, pyrrole [ 2,1-f][1,2,4]triazin-7-yl, pyrrolo[1,2-b]pyridazin-7-yl, thieno[2,3-b]pyrazine-7- 5-Aminopyrazolo[1,5-a]pyrimidin-3-yl, 2-aminopyrrolo[2,1-f][1,2,4]triazin-7-yl, 2 -Aminopyrrolo[1,2-b]pyridazin-7-yl, 2-aminothieno[2,3-b]pyrazin-7-yl; R 4 represents a saturated 4- to 7-membered heterocyclic ring Or a C 3 -C 7 -cycloalkyl group, wherein the saturated heterocyclic group and the C 3 -C 7 -cycloalkyl group are optionally mono- or polysubstituted by the same or different substituents from the group consisting of: C 1 -C 6 -alkyl, trifluoromethyl, 2,2,2-trifluoroethyl, cyclopropyl , C(=O)OH, C 1 -C 6 -alkoxy, trifluoromethoxy, 2,2,2-trifluoroethoxy, fluoro, chloro, cyano, hydroxy, NH 2 , NHR a And N(R a )R b , or represents C 1 -C 6 -alkyl, wherein the C 1 -C 6 -alkyl group may be mono- or polysubstituted by the same or different substituents from the group consisting of: Fluorine, chlorine, cyano, hydroxy, C(=O)OC 1 -C 6 -alkyl, C(=O)OH, C(=O)NH 2 , C(=O)N(H)R a , C(=O)N(R a )R b , SC 1 -C 6 -alkyl, S(=O) 2 -C 1 -C 6 -alkyl, S(=O) 2 NH 2 , C 1 - C 6 -alkoxy, trifluoromethoxy, 2,2,2-trifluoroethoxy, NH 2 , NHR a , N(R a )R b , C 3 -C 7 -cycloalkyl, tetra An azole and a 4- to 7-membered saturated heterocyclic group, wherein the C 3 -C 7 -cycloalkyl group and the 4 to 7 membered saturated heterocyclic group are optionally monosubstituted by the same or different substituents from the group consisting of Tetrasubstituted: fluorine, C 1 -C 4 -alkyl, hydroxy, C(=O)OH, trifluoromethyl, 2,2,2-trifluoroethyl, methoxy, ethoxy, trifluoromethyl Oxyl, cyclopropyl, cyclopropylmethyl, methylsulfonyl, NH 2 , NHR a , N(R a )R b ; R a represents C 1 -C 6 -alkyl or C 3 -C 7 a cycloalkyl group, wherein the C 1 -C 6 -alkyl group and the C 3 -C 7 -cycloalkyl group are optionally mono- or polysubstituted by the same or different substituents from the group consisting of fluorine, hydroxyl, cyanide a C 1 -C 4 -alkyl group, a C 1 -C 4 -alkoxy group and a C 3 -C 7 -cycloalkyl group; R b represents a C 1 -C 6 -alkyl group or a C 3 -C 7 - ring Alkyl; * at the bond indicates an attachment point in the molecule, and a solvate thereof, a non-image isomer, a mirror image isomer, a metabolite, a salt, a solvate or a salt. Novel IRAK4 inhibitors are particularly suitable for the treatment and prevention of proliferative, metabolic and inflammatory conditions characterized by an overreactive immune system. In particular, inflammatory skin disorders, cardiovascular disorders, pulmonary disorders, ocular disorders, neurological disorders, pain disorders and cancers should be mentioned herein. In addition, novel IRAK4 inhibitors are suitable for the treatment and prevention of autoimmune and inflammatory conditions, especially rheumatoid arthritis, multiple sclerosis, systemic lupus erythematosus, spondyloarthritis and gout, • metabolic disorders, especially liver disorders ( For example, fatty liver), and • gynecological conditions, especially endometriosis and endometriosis-related pain and other endometriosis-related symptoms, such as dysmenorrhea, painful intercourse, dysuria, and feces difficult.

在下文所闡述之本發明之合成中間體及工作實例之情形下,以相應鹼或酸之鹽形式指定之任一化合物通常係具有未知確切化學計量組成之鹽,如藉由各別製備及/或純化製程所獲得。除非更詳細的指定,否則因此在該等鹽之情形下至名稱及結構式(例如「鹽酸鹽」、「三氟乙酸鹽」、「鈉鹽」或「x HCl」、「x CF3 COOH」、「x Na+ 」)之添加不能以化學計量方式理解,而僅具有關於其中所呈現之鹽形成組份之闡述性特徵。 若合成中間體或工作實例或其鹽係藉由所述製備及/或純化製程以具有未知化學計量組成之溶劑合物形式(例如水合物) (若其具有所定義類型)獲得,則此相應地適用。 若由式(I)涵蓋且在下文提及之化合物不為鹽、溶劑合物及鹽之溶劑合物,則本發明化合物係式(I)化合物及其鹽、溶劑合物及鹽之溶劑合物,由式(I)涵蓋且具有下文所提及之各式之化合物及其鹽、溶劑合物及鹽之溶劑合物,及由式(I)涵蓋且在下文作為工作實例引用之化合物及其鹽、溶劑合物及鹽之溶劑合物。 在本發明之上下文中,較佳鹽係本發明化合物之生理上可接受之鹽。然而,本發明亦涵蓋其本身並不適於醫藥應用但可用於(例如)分離或純化本發明化合物之鹽。 本發明化合物之生理上可接受之鹽包括礦物酸、羧酸及磺酸之酸加成鹽,例如以下酸之鹽:鹽酸、氫溴酸、硫酸、磷酸、甲磺酸、乙磺酸、甲苯磺酸、苯磺酸、萘二磺酸、乙酸、三氟乙酸、丙酸、乳酸、酒石酸、蘋果酸、檸檬酸、富馬酸、馬來酸及苯甲酸。 本發明化合物之生理上可接受之鹽亦包括習用鹼之鹽,以舉例方式且優選的鹼金屬鹽(例如鈉鹽及鉀鹽)、鹼土金屬鹽(例如鈣鹽及鎂鹽);及衍生自氨或具有1至16個碳原子之有機胺之銨鹽,該等有機胺以舉例方式且優選的係乙胺、二乙胺、三乙胺、乙基二異丙基胺、單乙醇胺、二乙醇胺、三乙醇胺、二環己胺、二甲基胺基乙醇、普魯卡因(procaine)、二苄基胺、N-甲基嗎啉、精胺酸、離胺酸、乙二胺及N-甲基六氫吡啶。 溶劑合物在本發明上下文中闡述為藉由與溶劑分子配位形成呈固體或液體狀態之複合物之彼等形式之本發明化合物。水合物係與水配位之溶劑合物之具體形式。 本發明化合物可端視其結構以不同立體異構形式存在,即,呈構形異構物之形式或者若適當作為構象異構物(鏡像異構物及/或非鏡像異構物,包括彼等在構型異構物情形中之彼等)。因此,本發明涵蓋鏡像異構物及非鏡像異構物及其各別混合物。立體異構均質成份可以習知方式自鏡像異構物及/或非鏡像異構物之該等混合物分離;較佳使用層析製程、尤其於非對掌性或對掌性相上之HPLC層析用於此目的。 若本發明化合物可以互變異構物形式存在,則本發明涵蓋所有互變異構物形式。 本發明亦涵蓋本發明化合物之所有適宜同位素變體。此處,本發明化合物之同位素變體應理解為意指其中本發明化合物內之至少一個原子已替換為具有相同原子序數但原子量不同於自然界中通常或主要存在原子量之另一原子的化合物。可納入本發明化合物中之同位素之實例係氫、碳、氮、氧、磷、硫、氟、氯、溴及碘之彼等同位素,例如2 H (氘)、3 H (氚)、13 C、14 C、15 N、17 O、18 O、32 P、33 P、33 S、34 S、35 S、36 S、18 F、36 Cl、82 Br、123 I、124 I、129 I及131 I。本發明化合物之特定同位素變體、例如特定而言其中已納入一或多種放射性同位素之彼等可有益的(例如)用於檢查作用機制或活性化合物在體內之分佈;由於相對比較容易之可製備性及可檢測性,特定而言經3 H或14 C同位素標記之化合物適用於此目的。另外,同位素(例如氘)之納入可產生特定治療益處,此乃因化合物之代謝穩定性提高,例如在體內之半衰期延長或所需要之活性劑量降低;因此,本發明化合物之該等改質形式在一些情形下亦可構成本發明之較佳實施例。本發明化合物之同位素變體可藉由熟悉此項技術者已知之製程(例如藉由下文進一步闡述之方法及在工作實例中闡述之程序)藉由使用各別試劑及/或起始化合物之相應同位素改質形式來製備。 本發明進一步提供本發明化合物之所有可能結晶及多形形式,其中多形體在所有濃度範圍中皆可作為單一多形體或複數種多形體之混合物存在。 本發明另外亦涵蓋本發明化合物之前藥。術語「前藥」在此上下文中係指本身可具有生物活性或無活性但在其於體內之滯留時間期間反應(例如代謝或水解)以產生本發明化合物之化合物。 在本發明之上下文中,除非另外指明,否則取代基具有以下含義:烷基 在本發明之上下文中係具有指定的特定碳原子數之直鏈或具支鏈烷基。實例包括甲基、乙基、正丙基、異丙基、正丁基、異丁基、1-甲基丙基、2-甲基丙基、第三丁基、正戊基、1-乙基丙基、1-甲基丁基、2-甲基丁基、3-甲基丁基、2,2-二甲基丙基、正己基、1-甲基戊基、2-甲基戊基、3-甲基戊基、4-甲基戊基、1-乙基丁基及2-乙基丁基。較佳者係甲基、乙基、正丙基、正丁基、2-甲基丁基、3-甲基丁基及2,2-二甲基丙基。 在本發明之上下文中,環烷基 係在每一情形下具有所指定碳原子數之單環飽和烷基。較佳實例包括環丙基、環丁基、環戊基及環己基。 在本發明之上下文中,雜環烷基、雜環基或雜環 係總共具有3至10個環原子且含有一或兩個來自由N、O、S、SO及/或SO2 組成之群之環雜原子之飽和雜環。可提及(例如)以下:氮雜環丁基、氧雜環丁基、吡咯啶基、吡唑啶基、四氫呋喃基、六氫吡啶基、六氫吡嗪基、四氫吡喃基、嗎啉基、硫嗎啉基、二氧橋硫嗎啉基、二氫吲哚基及二氫異吲哚基。較佳者係:氮雜環丁基、氧雜環丁基、吡咯啶基、六氫吡啶基、六氫吡嗪基及嗎啉基。烷氧基 在本發明之上下文中代表具有所指定特定碳原子數之直鏈或具支鏈烷氧基。1至6個碳原子較佳。實例包括甲氧基、乙氧基、正丙氧基、異丙氧基、1-甲基丙氧基、正丁氧基、異丁氧基、第三丁氧基、正戊氧基、異戊氧基、1-乙基丙氧基、1-甲基丁氧基、2-甲基丁氧基、3-甲基丁氧基及正己氧基。尤佳者係具有1至4個碳原子之直鏈或具支鏈烷氧基。可提及為較佳之實例係甲氧基、乙氧基、正丙氧基、1-甲基丙氧基、正丁氧基及異丁氧基。 在本發明之上下文中,鹵素 係氟、氯及溴。較佳者係氟。羥基 在本發明之上下文中係OH。 鍵處之符號*表示分子中之鍵結位點。 除非另外指定,否則當本發明化合物中之基團經取代時,該等基團可經單取代或多取代。在本發明之上下文中,出現一次以上之所有基團皆彼此獨立地定義。較佳者係經一個、兩個或三個相同或不同的取代基取代。 R1 之較佳實施例係氯。 R1 之另一較佳實施例係O-R4 。 R2 之較佳實施例係C2 -C6 -烷基,其經羥基取代。尤佳地,R2 代表C2 -C5 -烷基,其經羥基取代。極尤佳者係其中R2 代表3-羥基丙基、3-羥基丁基、2-羥基乙基或3-羥基-3-甲基丁基之化合物。尤佳者係羥基-3-甲基丁基。 R2 之另一較佳實施例係C2 -C6 -烷基,其經兩個羥基取代。尤佳地,R2 代表2,3-二羥基丙基、2,3-二羥基-3-甲基丁基或2,3-二羥基丁基。極尤佳地,R2 代表2,3-二羥基丙基。 R2 之另一較佳實施例係C1 -C3 -烷基,其經氧雜環丁基或四氫呋喃基取代。尤佳者係其中R2 代表氧雜環丁-3-基甲基、氧雜環丁-2-基甲基、四氫呋喃-3-基甲基或四氫呋喃-2-基甲基之化合物。極尤佳者係其中R2 代表氧雜環丁-3-基甲基或四氫呋喃-3-基甲基之化合物。 R2 之另一較佳實施例係C2 -C6 -烷基,其經C1 -C3 -烷氧基取代。尤佳地,R2 代表C2 -C6 -烷基,其經甲氧基取代。極尤佳者係其中R2 代表3-甲氧基-3-甲基丁基、3-甲氧基丙基或2-甲氧基乙基之化合物。 R2 之另一較佳實施例係C2 -C6 -烷基,其經2-羥基乙氧基取代。尤佳者係其中R2 代表2-(2-羥基乙氧基)乙基或3-(2-羥基乙氧基)丙基之化合物。極尤佳者係2-(2-羥基乙氧基)乙基。 R2 之另一較佳實施例係C2 -C6 -烷基,其經OCH2 (C=O)OH或OCH2 (C=O)OCH3 取代。尤佳者係其中R2 代表CH2 CH2 OCH2 C(=O)OH或CH2 CH2 OCH2 C(=O)OEt之化合物。 R2 之另一較佳實施例係3-側氧基丁基。 亦較佳者係其中R2 係4,4,4-三氟丁基、3,3,3-三氟丙基、2,2,2-三氟乙基或3,3-二氟丁基之化合物。尤佳地,R2 代表4,4,4-三氟丁基或3,3,3-三氟丙基。極佳者係4,4,4-三氟丁基。 在較佳實施例中,R3 代表2-(C1 -C6 -烷基)-1,3-噻唑-4-基,其中C1 -C6 -烷基取代基可視情況經環丙基單取代、經氟原子單取代至五取代且視情況經羥基單取代。尤佳地,R3 代表2-(三氟甲基)-1,3-噻唑-4-基、2-甲基-1,3-噻唑-4-基、2-乙基-1,3-噻唑-4-基、2-丙基-1,3-噻唑-4-基、2-異丙基-1,3-噻唑-4-基或2-第三丁基-1,3-噻唑-4-基。 在另一較佳實施例中,R3 代表2-環丙基-1,3-噻唑-4-基。 在另一較佳實施例中,R3 代表4-(C1 -C6 -烷基)-1,3-噻唑-2-基,其中C1 -C6 -烷基取代基可視情況經環丙基單取代、經氟原子單取代至五取代且視情況經羥基單取代。尤佳地,R3 代表4-(三氟甲基)-1,3-噻唑-2-基、4-甲基-1,3-噻唑-2-基、4-乙基-1,3-噻唑-2-基、4-異丙基-1,3-噻唑-2-基或4-第三丁基-1,3-噻唑-2-基。極尤佳地,R3 代表4-(三氟甲基)-1,3-噻唑-2-基。 在另一較佳實施例中,R3 代表4-環丙基-1,3-噻唑-2-基。 在另一較佳實施例中,R3 代表2-(C1 -C6 -烷基)-1,3-噁唑-4-基,其中C1 -C6 -烷基取代基可視情況經環丙基單取代、經氟原子單取代至五取代且視情況經羥基單取代。尤佳地,R3 代表2-甲基-1,3-噁唑-4-基、2-(三氟甲基)-1,3-噁唑-4-基、2-乙基-1,3-噁唑-4-基、2-(1,1-二氟乙基)-1,3-噁唑-4-基、2-(2,2,2-三氟乙基)-1,3-噁唑-4-基、2-異丙基-1,3-噁唑-4-基、2-第三丁基-1,3-噁唑-4-基或2-(環丙基甲基)-1,3-噁唑-4-基。極尤佳地,R3 代表2-(三氟甲基)-1,3-噁唑-4-基。在另一較佳實施例中,R3 代表2-環丙基-1,3-噁唑-4-基。 在另一較佳實施例中,R3 代表2-(C1 -C6 -烷基)-1,3-噁唑-5-基,其中C1 -C6 -烷基取代基可視情況經環丙基單取代、經氟原子單取代至五取代且視情況經羥基單取代。尤佳地,R3 代表2-甲基-1,3-噁唑-5-基、2-乙基-1,3-噁唑-5-基、2-異丙基-1,3-噁唑-5-基。極尤佳地,R3 代表2-甲基-1,3-噁唑-5-基。 在另一較佳實施例中,R3 代表1-(C1 -C6 -烷基)-1H-吡唑-3-基,其中C1 -C6 -烷基取代基可視情況經環丙基單取代、經氟原子單取代至五取代且視情況經羥基單取代。尤佳地,R3 代表1-(二氟甲基)-1H-吡唑-3-基、1-甲基-1H-吡唑-3-基、1-乙基-1H-吡唑-3-基、1-(2,2,2-三氟乙基)-1H-吡唑-3-基、1-異丙基-1H-吡唑-3-基、1-丙基-1H-吡唑-3-基、1-第三丁基-1H-吡唑-3-基、1-異丁基-1H-吡唑-3-基。極尤佳地,R3 代表1-(二氟甲基)-1H-吡唑-3-基、1-乙基-1H-吡唑-3-基、1-異丙基-1H-吡唑-3-基。 在另一較佳實施例中,R3 代表6-(C1 -C6 -烷基)吡啶-2-基,其中C1 -C6 -烷基取代基可視情況經環丙基單取代、經氟原子單取代至五取代且視情況經羥基單取代。尤佳地,R3 代表6-甲基吡啶-2-基、6-(二氟甲基)吡啶-2-基、6-(三氟甲基)吡啶-2-基、6-乙基吡啶-2-基、6-(1,1-二氟乙基)吡啶-2-基、6-(五氟乙基)吡啶-2-基、6-(2,2,2-三氟乙基)吡啶-2-基、6-丙基吡啶-2-基、6-異丙基吡啶-2-基、6-(2-羥基丙-2-基)吡啶-2-基、6-第三丁基吡啶-2-基。極尤佳地,R3 代表6-(1,1-二氟乙基)吡啶-2-基、6-(2-羥基丙-2-基)吡啶-2-基、6-(二氟甲基)吡啶-2-基、6-(五氟乙基)吡啶-2-基或6-(三氟甲基)吡啶-2-基。尤佳地,R3 代表6-(1,1-二氟乙基)吡啶-2-基或6-(三氟甲基)吡啶-2-基。 在另一較佳實施例中,R3 代表6-胺基吡啶-2-基、6-((C1 -C4 -烷基)胺基)吡啶-2-基、6-(二-(C1 -C4 -烷基)胺基)吡啶-2-基、6-(吡咯啶-1-基)吡啶-2-基、6-(六氫吡啶-1-基)吡啶-2-基、6-(嗎啉-4-基)吡啶-2-基、6-(4-甲基六氫吡嗪-1-基)吡啶-2-基。尤佳地,R3 代表6-胺基吡啶-2-基、6-(甲基胺基)吡啶-2-基、6-(乙基胺基)吡啶-2-基或6-(二甲基胺基)吡啶-2-基。極尤佳地,R3 代表6-胺基吡啶-2-基。 在另一較佳實施例中,R3 代表6-環丙基吡啶-2-基。 在另一較佳實施例中,R3 代表6,7-二氫-4H-吡唑并[5,1-c][1,4]噁嗪-2-基、5-甲基-4,5,6,7-四氫吡唑并[1,5-a]吡嗪-2-基或4,5,6,7-四氫吡唑并[1,5-a]吡嗪-2-基、吡唑并[1,5-a]嘧啶-3-基、吡咯并[2,1-f][1,2,4]三嗪-7-基、吡咯并[1,2-b]嗒嗪-7-基、噻吩并[2,3-b]吡嗪-7-基、5-胺基吡唑并[1,5-a]嘧啶-3-基、2-胺基吡咯并[2,1-f][1,2,4]三嗪-7-基、2-胺基吡咯并[1,2-b]嗒嗪-7-基、2-胺基噻吩并[2,3-b]吡嗪-7-基。尤佳地,R3 代表6,7-二氫-4H-吡唑并[5,1-c][1,4]噁嗪-2-基、吡唑并[1,5-a]嘧啶-3-基。 R4 之較佳實施例係環丙基、六氫吡啶-4-基、1-甲基六氫吡啶-4-基、吡咯啶-3-基、1-甲基吡咯啶-3-基、氮雜環丁-3-基、1-甲基氮雜環丁-3-基、氧雜環丁-3-基、四氫呋喃-3-基或四氫-2H-吡喃-4-基。尤佳地,R4 代表氧雜環丁-3-基、四氫呋喃-3-基或四氫-2H-吡喃-4-基。極尤佳地,R4 代表氧雜環丁-3-基或四氫呋喃-3-基。 R4 之另一較佳實施例係C1 -C6 -烷基。尤佳地,R4 代表甲基、乙基或異丙基。極尤佳地,R4 代表甲基或乙基。 R4 之另一較佳實施例係C2 -C6 -烷基,其經羥基取代。尤佳地,R4 代表C2 -C4 -烷基,其經羥基取代。極尤佳地,R4 代表2-羥基乙基、2-羥基丙基或2-羥基-2-甲基丙基。尤佳地,R4 代表2-羥基乙基。 R4 之另一較佳實施例係C2 -C6 -烷基,其經選自以下之基團取代:C(=O)OMe、C(=O)OEt、C(=O)OH、C(=O)NH2 、SMe、SEt、S(=O)2 Me、S(=O)2 Et、甲氧基、乙氧基、N(CH3 )2 、環丙基、環丁基、環戊基、氧雜環丁-3-基、四氫呋喃-3-基及四氫-2H-吡喃-4-基。在尤佳實施例中,R4 代表環丙基甲基、氧雜環丁-3-基甲基、2-(甲基磺醯基)乙基、3-(甲基磺醯基)丙基、2-(甲基硫基)乙基、CH2 C(=O)OEt、CH(CH3 )C(=O)OH或CH2 C(=O)OH、CH2 CH2 C(=O)OH。極尤佳地,R4 代表環丙基甲基或氧雜環丁-3-基甲基。 在另一較佳實施例之背景下,R4 代表三氟甲基、2,2,2-三氟乙基或2,2-二氟乙基。尤佳地,R4 代表三氟甲基。 較佳地,R6 代表氫或氟。尤佳地,R6 代表氫。 較佳地,R8 代表氫或氟。尤佳地,R8 代表氫。 較佳地,R9 代表氫或氟。尤佳地,R9 代表氫。 較佳地,R10 代表氫或氟。尤佳地,R10 代表氫。 較佳地,R12 代表氫或氟。尤佳地,R12 代表氫。 較佳地,Ra 代表甲基、乙基或環丙基。 較佳地,Rb 代表甲基、乙基或環丙基。 本發明之一個實施例提供 R2 代表C1 -C8 -烷基, 其可視情況彼此獨立地經選自由以下組成之群之相同或不同取代基單取代或多取代:1至5個氟原子、1至3個羥基、氧雜環丁基、四氫呋喃基、吡喃基及C1 -C6 -烷氧基, 其中C1 -C6 -烷氧基 可經氟或 經羥基或 經C(=O)OH、C(=O)Me、C(=O)Et、C(=O)NH2 單取代至三取代; 且取代基R1 及R3 皆可具有上文所提及之含義。 較佳者係式(I)化合物,其中 R1 代表氯或O-R4 , R2 代表C1 -C6 -烷基, 其可視情況彼此獨立地經選自由以下組成之群之相同或不同取代基單取代或多取代:1至3個氟原子、1至2個羥基、氧雜環丁基、四氫呋喃基及C1 -C3 -烷氧基, 其中C1 -C3 -烷氧基 可經羥基或 經C(=O)OH、C(=O)Me、C(=O)Et取代; 或R2 代表3-側氧基丁基; R3 代表選自以下之基團: , 其中R5 代表氫、環丙基或C1 -C6 -烷基,其中C1 -C6 -烷基可視情況經環丙基、氰基及羥基單取代至三取代,其中每一取代基可僅出現一次,或經氟原子單取代至五取代; 且R6 代表氫; 或 R3 代表基團其中R7 代表氫、環丙基、氰基、NH2 、NH(C1-C6 -烷基)、N(C1 -C6 -烷基)2 、吡咯啶-1-基、六氫吡啶-1-基、嗎啉-4-基、4-甲基六氫吡嗪-1-基 或代表C1 -C6 -烷基,其中C1 -C6 -烷基可視情況經環丙基、氰基及羥基單取代至三取代,其中每一取代基可僅出現一次,或經氟原子單取代至五取代; 且R8 、R9 、R10 代表氫 或 R3 代表基團其中R11 代表 C3 -C6 -環烷基或C1 -C6 -烷基,其中C1 -C6 -烷基可視情況經環丙基、氰基及羥基單取代至三取代,其中每一取代基可僅出現一次,或經氟原子單取代至五取代; 且R12 代表氫 或 R3 代表 6,7-二氫-4H-吡唑并[5,1-c][1,4]噁嗪-2-基、5-甲基-4,5,6,7-四氫吡唑并[1,5-a]吡嗪-2-基或4,5,6,7-四氫吡唑并[1,5-a]吡嗪-2-基, 或代表 吡唑并[1,5-a]嘧啶-3-基、吡咯并[2,1-f][1,2,4]三嗪-7-基、吡咯并[1,2-b]嗒嗪-7-基、噻吩并[2,3-b]吡嗪-7-基、5-胺基吡唑并[1,5-a]嘧啶-3-基、2-胺基吡咯并[2,1-f][1,2,4]三嗪-7-基、2-胺基吡咯并[1,2-b]嗒嗪-7-基、2-胺基噻吩并[2,3-b]吡嗪-7-基; R4 代表環丙基、六氫吡啶-4-基、1-甲基六氫吡啶-4-基、吡咯啶-3-基、1-甲基吡咯啶-3-基、氮雜環丁-3-基、1-甲基氮雜環丁-3-基、氧雜環丁-3-基、四氫呋喃-3-基或四氫-2H-吡喃-4-基,其中氧雜環丁-3-基、四氫呋喃-3-基、四氫-2H-吡喃-4-基可視情況經甲基單取代或二取代, 或代表 C1 -C6 -烷基,其中C1 -C6 -烷基可視情況經以下取代:羥基、C(=O)OMe、C(=O)OEt、C(=O)OH、C(=O)NH2 、SMe、SEt、S(=O)2 Me、S(=O)2 Et、甲氧基、乙氧基、N(CH3 )2 、環丙基、環丁基、環戊基、氧雜環丁-3-基、四氫呋喃-3-基、四氫-2H-吡喃-4-基, 或為 三氟甲基、2,2,2-三氟乙基或2,2-二氟乙基; 及其非鏡像異構物、鏡像異構物、代謝物、鹽、溶劑合物或鹽之溶劑合物。 另外較佳者係式(I)化合物,其中 R1 代表氯或O-R4 ; R2 代表C2 -C5 -烷基,其可 經1至2個羥基或 經氧雜環丁-3-基或經四氫呋喃-3-基或 經基團OCH2 C(=O)OH、OCH2 C(=O)OMe、OCH2 CH2 OH或 經C1 -C3 -烷氧基取代, 其中C1 -C3 -烷氧基 可經羥基或 經C(=O)OH、C(=O)Me、C(=O)Et取代; 或R2 代表3,3,3-三氟丙基、4,4,4-三氟丁基或3-側氧基丁基; R3 代表1-(C1 -C6 -烷基)-1H-吡唑-3-基,其中C1 -C6 -烷基取代基可視情況經環丙基單取代、經氟原子單取代至五取代且視情況經羥基單取代, 或 代表2-環丙基-1,3-噻唑-4-基或2-(C1 -C6 -烷基)-1,3-噻唑-4-基,其中C1 -C6 -烷基取代基可視情況經環丙基單取代、經氟原子單取代至五取代且視情況經羥基單取代, 或 代表4-(C1 -C6 -烷基)-1,3-噻唑-2-基,其中C1 -C6 -烷基取代基可視情況經環丙基單取代、經氟原子單取代至五取代且視情況經羥基單取代, 或 代表2-環丙基-1,3-噁唑-4-基或2-(C1 -C6 -烷基)-1,3-噁唑-4-基,其中C1 -C6 -烷基取代基可視情況經環丙基單取代、經氟原子單取代至五取代且視情況經羥基單取代, 或 代表2-(C1 -C6 -烷基)-1,3-噁唑-5-基,其中C1 -C6 -烷基取代基可視情況經環丙基單取代、經氟原子單取代至五取代且視情況經羥基單取代, 或 代表6-(C1 -C6 -烷基)吡啶-2-基,其中C1 -C6 -烷基取代基可視情況 經環丙基單取代、經氟原子單取代至五取代且視情況經羥基單取代, 或代表6-胺基吡啶-2-基、6-((C1 -C4 -烷基)胺基)吡啶-2-基、6-(二-(C1 -C4 -烷基)胺基)吡啶-2-基、6-(吡咯啶-1-基)吡啶-2-基、6-(六氫吡啶-1-基)吡啶-2-基、6-(嗎啉-4-基)吡啶-2-基、6-(4-甲基六氫吡嗪-1-基)吡啶-2-基, 或 代表6,7-二氫-4H-吡唑并[5,1-c][1,4]噁嗪-2-基、5-甲基-4,5,6,7-四氫吡唑并[1,5-a]吡嗪-2-基或4,5,6,7-四氫吡唑并[1,5-a]吡嗪-2-基, 或代表 吡唑并[1,5-a]嘧啶-3-基、吡咯并[2,1-f][1,2,4]三嗪-7-基、吡咯并[1,2-b]嗒嗪-7-基、噻吩并[2,3-b]吡嗪-7-基、5-胺基吡唑并[1,5-a]嘧啶-3-基、2-胺基吡咯并[2,1-f][1,2,4]三嗪-7-基、2-胺基吡咯并[1,2-b]嗒嗪-7-基、2-胺基噻吩并[2,3-b]吡嗪-7-基; R4 代表C1 -C4 -烷基或 代表氧雜環丁-3-基、四氫呋喃-3-基或四氫-2H-吡喃-4-基 或代表 2-羥基乙基、3-羥基丙基、2-羥基丙基、2-羥基-2-甲基丙基 或代表 CH2 C(=O)OMe、CH2 C(=O)OEt、CH2 C(=O)OH、CH2 CH2 SMe、2-(甲基磺醯基)乙基、3-(甲基磺醯基)丙基、2-甲氧基乙基、2-乙氧基乙基、CH2 CH2 N(CH3 )2 、環丙基甲基、環丁基甲基、環戊基甲基、氧雜環丁-3-基甲基、四氫呋喃-3-基甲基、四氫-2H-吡喃-4-基甲基 或代表 三氟甲基、2,2,2-三氟乙基或2,2-二氟乙基; R5 代表氫或氟; 及其非鏡像異構物、鏡像異構物、代謝物、鹽、溶劑合物或鹽之溶劑合物。 亦尤佳者係通式(I)之化合物,其中 R1 代表氯或O-R4 ; R2 代表3-羥基丙基、3-羥基丁基、2-羥基乙基、3-羥基-3-甲基丁基、4-羥基丁基、4-羥基戊基、2,3-二羥基丙基、2,3-二羥基-2-甲基丙基、2-羥基丙基, 氧雜環丁-3-基甲基、氧雜環丁-3-基乙基, 四氫呋喃-3-基甲基、四氫呋喃-3-基乙基, 3-側氧基丁基, 3-甲氧基-3-甲基丁基、3-甲氧基丙基、2-甲氧基乙基, 4,4,4-三氟丁基、3,3,3-三氟丙基, 2-(2-羥基乙氧基)乙基, CH2 CH2 OCH2 C(=O)OH、CH2 CH2 OCH2 C(=O)OEt; R3 代表1-(二氟甲基)-1H-吡唑-3-基、1-甲基-1H-吡唑-3-基、1-乙基-1H-吡唑-3-基、1-(2,2,2-三氟乙基)-1H-吡唑-3-基、1-異丙基-1H-吡唑-3-基、1-丙基-1H-吡唑-3-基、1-第三丁基-1H-吡唑-3-基、1-異丁基-1H-吡唑-3-基, 2-(三氟甲基)-1,3-噻唑-4-基、2-甲基-1,3-噻唑-4-基、2-乙基-1,3-噻唑-4-基、2-丙基-1,3-噻唑-4-基、2-異丙基-1,3-噻唑-4-基、2-第三丁基-1,3-噻唑-4-基、2-環丙基-1,3-噻唑-4-基, 4-(三氟甲基)-1,3-噻唑-2-基、4-甲基-1,3-噻唑-2-基、4-乙基-1,3-噻唑-2-基、4-異丙基-1,3-噻唑-2-基、4-第三丁基-1,3-噻唑-2-基、4-環丙基-1,3-噻唑-2-基, 2-甲基-1,3-噁唑-4-基、2-(三氟甲基)-1,3-噁唑-4-基、2-乙基-1,3-噁唑-4-基、2-(1,1-二氟乙基)-1,3-噁唑-4-基、2-(2,2,2-三氟乙基)-1,3-噁唑-4-基、2-異丙基-1,3-噁唑-4-基、2-第三丁基-1,3-噁唑-4-基、2-環丙基-1,3-噁唑-4-基、2-(環丙基甲基)-1,3-噁唑-4-基, 2-甲基-1,3-噁唑-5-基、2-乙基-1,3-噁唑-5-基、2-異丙基-1,3-噁唑-5-基, 6-甲基吡啶-2-基、6-(二氟甲基)吡啶-2-基、6-(三氟甲基)吡啶-2-基、6-乙基吡啶-2-基、6-(1,1-二氟乙基)吡啶-2-基、6-(五氟乙基)吡啶-2-基、6-(2,2,2-三氟乙基)吡啶-2-基、6-丙基吡啶-2-基、6-異丙基吡啶-2-基、6-(2-羥基丙-2-基)吡啶-2-基、6-第三丁基吡啶-2-基、6-環丙基吡啶-2-基、6-胺基吡啶-2-基、6-(甲基胺基)吡啶-2-基、6-(乙基胺基)吡啶-2-基、6-(二甲基胺基)吡啶-2-基, 6,7-二氫-4H-吡唑并[5,1-c][1,4]噁嗪-2-基、5-甲基-4,5,6,7-四氫吡唑并[1,5-a]吡嗪-2-基、4,5,6,7-四氫吡唑并[1,5-a]吡嗪-2-基, 吡唑并[1,5-a]嘧啶-3-基、吡咯并[2,1-f][1,2,4]三嗪-7-基、吡咯并[1,2-b]嗒嗪-7-基、噻吩并[2,3-b]吡嗪-7-基; R4 代表甲基、乙基、異丙基、丙基、環丙基甲基、氧雜環丁-3-基、氧雜環丁-3-基甲基、四氫呋喃-3-基、2-羥基乙基、三氟甲基、2,2,2-三氟乙基、2,2-二氟乙基、2-(甲基磺醯基)乙基、3-(甲基磺醯基)丙基、2-(甲基硫基)乙基、CH2 C(=O)OEt、CH2 C(=O)OH; R5 代表氫; 及其非鏡像異構物、鏡像異構物、代謝物、鹽、溶劑合物或鹽之溶劑合物。 極尤佳者係如下化合物:其中 R1 代表氯或O-R4 ; R2 代表3-羥基丙基、3-羥基丁基、2-羥基乙基、3-羥基-3-甲基丁基、2,3-二羥基丙基, 氧雜環丁-3-基甲基、四氫呋喃-3-基甲基、3-側氧基丁基, 3-甲氧基-3-甲基丁基、3-甲氧基丙基、2-甲氧基乙基, 4,4,4-三氟丁基, 2-(2-羥基乙氧基)乙基, CH2 CH2 OCH2 C(=O)OH、CH2 CH2 OCH2 C(=O)OEt; R3 代表1-(二氟甲基)-1H-吡唑-3-基、1-乙基-1H-吡唑-3-基、1-異丙基-1H-吡唑-3-基、2-(三氟甲基)-1,3-噁唑-4-基、2-(三氟甲基)-1,3-噻唑-4-基、2-環丙基-1,3-噁唑-4-基、2-甲基-1,3-噁唑-5-基、2-甲基-1,3-噻唑-4-基、4-(三氟甲基)-1,3-噻唑-2-基、6-(1,1-二氟乙基)吡啶-2-基、6-(2-羥基丙-2-基)吡啶-2-基、6-(二氟甲基)吡啶-2-基、6-(五氟乙基)吡啶-2-基、6-(三氟甲基)吡啶-2-基、6,7-二氫-4H-吡唑并[5,1-c][1,4]噁嗪-2-基、6-胺基吡啶-2-基、吡唑并[1,5-a]嘧啶-3-基; R4 代表甲基、乙基、異丙基、環丙基甲基、氧雜環丁-3-基、氧雜環丁-3-基甲基、四氫呋喃-3-基、2-羥基乙基、三氟甲基、3-(甲基磺醯基)丙基、2-(甲基硫基)乙基、CH2 C(=O)OEt、CH2 C(=O)OH; 及其非鏡像異構物、鏡像異構物、代謝物、鹽、溶劑合物或鹽之溶劑合物。 亦極尤佳者係如下化合物:其中 R1 代表O-R4 ; R2 代表3-羥基-3-甲基丁基; R3 代表1-(二氟甲基)-1H-吡唑-3-基、1-乙基-1H-吡唑-3-基、2-(三氟甲基)-1,3-噁唑-4-基、2-(三氟甲基)-1,3-噻唑-4-基、2-環丙基-1,3-噁唑-4-基、2-甲基-1,3-噁唑-5-基、2-甲基-1,3-噻唑-4-基、4-(三氟甲基)-1,3-噻唑-2-基、6-(1,1-二氟乙基)吡啶-2-基、6-(2-羥基丙-2-基)吡啶-2-基、6-(二氟甲基)吡啶-2-基、6-(三氟甲基)吡啶-2-基、6-胺基吡啶-2-基、吡唑并[1,5-a]嘧啶-3-基; R4 代表甲基、乙基、異丙基、環丙基甲基、氧雜環丁-3-基、氧雜環丁-3-基甲基, 2-羥基乙基; 及其非鏡像異構物、鏡像異構物、代謝物、鹽、溶劑合物或鹽之溶劑合物。 特定而言,本發明提供以下化合物: 在上文所提及之化合物中,尤佳者係以下化合物: 1-(二氟甲基)-N-[2-(3-羥基-3-甲基丁基)-6-甲氧基-2H-吲唑-5-基]-1H-吡唑-3-甲醯胺 N-[6-乙氧基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]-2-(三氟甲基)-1,3-噻唑-4-甲醯胺 1-(二氟甲基)-N-[6-乙氧基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]-1H-吡唑-3-甲醯胺 N-[6-乙氧基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]-2-甲基-1,3-噻唑-4-甲醯胺 2-環丙基-N-[6-乙氧基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]-1,3-噁唑-4-甲醯胺 6-胺基-N-[6-乙氧基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]吡啶-2-甲醯胺 N-[6-乙氧基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 N-[2-(3-羥基-3-甲基丁基)-6-(氧雜環丁-3-基氧基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 N-[2-(3-羥基-3-甲基丁基)-6-(氧雜環丁-3-基氧基)-2H-吲唑-5-基]-2-(三氟甲基)-1,3-噻唑-4-甲醯胺 1-(二氟甲基)-N-[2-(3-羥基-3-甲基丁基)-6-(氧雜環丁-3-基甲氧基)-2H-吲唑-5-基]-1H-吡唑-3-甲醯胺 6-(二氟甲基)-N-[2-(3-羥基-3-甲基丁基)-6-(氧雜環丁-3-基甲氧基)-2H-吲唑-5-基]吡啶-2-甲醯胺 N-2-(3-羥基-3-甲基丁基)-6-[(3S)-四氫呋喃-3-基氧基]-2H-吲唑-5-基-2-甲基-1,3-噁唑-5-甲醯胺 N-2-(3-羥基-3-甲基丁基)-6-[(3S)-四氫呋喃-3-基氧基]-2H-吲唑-5-基-6-(三氟甲基)吡啶-2-甲醯胺 6-胺基-N-2-(3-羥基-3-甲基丁基)-6-[(3S)-四氫呋喃-3-基氧基]-2H-吲唑-5-基吡啶-2-甲醯胺 6-(二氟甲基)-N-[6-乙氧基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]吡啶-2-甲醯胺 N-[2-(3-羥基-3-甲基丁基)-6-(氧雜環丁-3-基甲氧基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 N-[2-(3-羥基-3-甲基丁基)-6-甲氧基-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 N-[6-(2-羥基乙氧基)-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 6-(1,1-二氟乙基)-N-[6-乙氧基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]吡啶-2-甲醯胺 N-[6-乙氧基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]-1-乙基-1H-吡唑-3-甲醯胺。 本發明化合物充當IRAK4激酶之抑制劑且具有驚奇有用之藥理學活性譜。 因此,除上文所提及之標的物以外,本發明亦提供本發明化合物之用途,其用於治療及/或預防人及動物之疾病。 尤佳者係利用本發明之IRAK4抑制劑來治療及/或預防婦科病症、發炎皮膚病症、心血管病症、肺部病症、眼部病症、自體免疫病症、疼痛病症、代謝失調、痛風、肝臟病症、代謝症候群、胰島素抗性及癌症。 本發明化合物適於預防及/或治療各種病症及疾病相關狀態,尤其由TLR (TLR3除外)及/或IL-1受體家族介導之病症及/或病理學由IRAK4直接介導之病症。IRAK4相關之病症包括多發性硬化、動脈粥樣硬化、心肌梗塞、阿茲海默氏病、病毒誘導之心肌炎、痛風、格特-小柳-原田三氏症候群、紅斑性狼瘡、牛皮癬、脊椎關節炎及關節炎。 本發明化合物亦可用於預防及/或治療由MyD88及TLR (TLR3除外)介導之病症。此包括多發性硬化、類風濕性關節炎、脊椎關節炎(尤其牛皮癬性脊椎關節炎及別赫捷列夫氏病)、代謝症候群(包括胰島素抗性、糖尿病)、骨關節炎、薛格連氏症候群、巨細胞動脈炎、敗血症、多發性肌炎及皮肌炎、皮膚病症(例如牛皮癬、異位性皮膚炎、斑禿、反常性痤瘡及尋常性痤瘡)、肺部病症(例如肺纖維化、慢性阻塞性肺病(COPD)、急性呼吸窘迫症候群(ARDS)、急性肺損傷(ALI)、間質性肺病(ILD)、類肉瘤病及肺高血壓)。 由於本發明化合物之作用機制,故其適於預防及/或治療TLR介導之病症貝歇氏症、痛風、子宮內膜異位症及子宮內膜異位症相關性疼痛及其他子宮內膜異位症相關性症狀(例如痛經、性交疼痛、排尿困難及排糞困難)。另外,本發明化合物適於預防及/或治療移植排斥、紅斑性狼瘡、成人發作性斯蒂爾氏病及慢性發炎腸病症(例如潰瘍性結腸炎及克隆氏病)。 除已列示之病症以外,使用本發明化合物亦適於治療及/或預防以下病症:眼部病症,例如角膜炎、過敏性結膜炎、乾燥性角結膜炎、黃斑部病變及眼色素層炎;心血管病症,例如動脈粥樣硬化、心肌再灌注損害、心肌梗塞、高血壓及神經病症(例如阿茲海默氏病、中風及帕金森病)。 本發明化合物之作用機制亦使得能夠預防及/或治療由TLR及IL-1受體家族介導之肝臟病症,尤其NAFLD、NASH、ASH、肝纖維化及肝硬化。 本發明化合物亦可預防及/或治療搔癢症及疼痛、尤其急性、慢性、發炎性及神經病性疼痛。 由於本發明化合物之作用機制,故其適於預防及/或治療腫瘤病症,例如淋巴瘤、慢性淋巴白血病、黑色素瘤及肝細胞癌、乳癌及Ras依賴性腫瘤。 此外,本發明化合物適於治療及/或預防經由IL-1受體家族介導之病症。該等病症包括CAPS (隱熱蛋白相關週期症候群) (包括FCAS (家族性寒冷性自體發炎症候群)、MWS (穆-韋二氏症候群)、NOMID (新生兒發作型多系統發炎性疾病)及CONCA (慢性嬰兒、神經、皮膚及關節)症候群)、FMF (家族性地中海熱)、HIDS (高IgD症候群)、TRAPS (腫瘤壞死因子受體1相關之週期症候群)、幼年型特發性關節炎、成人發作性斯蒂爾氏病、阿達替德-貝歇氏症、類風濕性關節炎、牛皮癬、關節炎、別赫捷列夫氏病、骨關節炎、乾燥性角結膜炎及薛格連氏症候群、多發性硬化、紅斑性狼瘡、斑禿、1型糖尿病、2型糖尿病及心肌梗塞後遺症。肺部病症(例如氣喘、COPD、特發性間質性肺炎及ARDS)、婦科病症(例如子宮內膜異位症及子宮內膜異位症相關性疼痛及其他子宮內膜異位症相關性症狀(例如痛經、性交疼痛、排尿困難及排糞困難))、慢性-發炎腸病症(例如克隆氏病及潰瘍性結腸炎)與IL-1受體家族之失調相關且適於治療及/或預防性使用本發明化合物。 本發明化合物亦可用於治療及/或預防IL1受體家族介導之神經病症(例如中風、阿茲海默氏病、顱腦創傷)及皮膚病學病症(例如牛皮癬、異位性皮膚炎、反常性痤瘡、斑禿及過敏性接觸性皮膚炎)。 另外,本發明化合物適於治療及/或預防疼痛病症、尤其急性、慢性、發炎性及神經病性疼痛。此較佳包括感覺過敏、觸摸痛、關節炎疼痛(例如骨關節炎、類風濕性關節炎及脊椎關節炎)、經前疼痛、子宮內膜異位症相關性疼痛、手術後疼痛、間質性膀胱炎疼痛、CRPS (複雜性區域疼痛症候群)、三叉神經痛、前列腺炎疼痛、由脊髓損傷造成之疼痛、發炎誘導之疼痛、腰部疼痛、癌性疼痛、化學療法相關性疼痛、HIV治療誘導之神經病變、燒傷誘導之疼痛及慢性疼痛。 本發明亦進一步提供使用有效量之本發明化合物中之至少一者治療及/或預防病症、尤其上文所提及病症之方法。 在本發明之上下文中,術語「治療(treatment或treating)」包括抑制、延遲、遏製、減輕、減弱、限制、減少、抑制、逐退或治癒疾病、病況、病症、損傷或健康問題,或該等狀態及/或該等狀態之症狀之發生、病程或進展。術語「療法」在本文中應理解為與術語「治療」同義。 術語「預防(prevention、prophylaxis及preclusion)」在本發明之上下文中同義使用,且係指避免或減少感染、經歷、罹患或患有疾病、病況、病症、損傷或健康問題或該等狀態及/或該等狀態之症狀之發生或推進之風險。 可部分地或完全地治療或預防疾病、病況、病症、損傷或健康問題。 本發明化合物可單獨使用,或若需要可與其他活性化合物組合使用。本發明進一步提供包含本發明化合物中之至少一者及一或多種其他活性化合物之藥劑,特定而言其用於治療及/或預防上文所提及之病症。適於組合之活性化合物之較佳實例包括: 通常可提及以下活性化合物:例如抗細菌物質(例如青黴素(penicillins)、萬古黴素(vancomycin)、環丙沙星(ciprofloxacin))、抗病毒物質(例如阿昔洛韋(aciclovir)、奧司他韋(oseltamivir))及抗黴菌物質(例如那夫替非(naftifin)、製黴素(nystatin))及γ免疫球蛋白)、免疫調節及免疫抑制化合物(例如環孢素(cyclosporin)、Methotrexat®、TNF拮抗劑(例如Humira®、依那西普(Etanercept)、英利昔單抗(Infliximab))、IL-1抑制劑(例如阿那白滯素(Anakinra)、卡那單抗(Canakinumab)、利羅西普(Rilonacept))、磷酸二酯酶抑制劑(例如阿普斯特(Apremilast))、Jak/STAT抑制劑(例如托法替尼(Tofacitinib)、巴瑞替尼(Baricitinib)、GLPG0634)、來氟米特(leflunomid)、環磷醯胺、利妥昔單抗(rituximab)、貝利木單抗(belimumab)、他克莫司(tacrolimus)、雷帕黴素(rapamycin)、嗎替麥考酚酯(mycophenolate mofetil)、干擾素、皮質類固醇(例如普賴松(prednisone)、普賴蘇濃(prednisolone)、甲基普賴蘇濃、氫化可體松(hydrocortisone)、倍他米松(betamethasone))、環磷醯胺、硫唑嘌呤及磺胺塞拉金(sulfasalazine));對乙醯胺基酚、非類固醇抗發炎物質(NSAIDS) (阿斯匹林(aspirin)、布洛芬(ibuprofen)、萘普生(naproxen)、依託度酸(etodolac)、塞來昔布(celecoxib)、秋水仙鹼(colchicine))。 對於腫瘤療法應提及以下各項:免疫療法(例如阿地介白素(aldesleukin)、阿倫單抗(alemtuzumab)、巴利昔單抗(basiliximab)、卡妥索單抗(catumaxomab)、西莫白介素(celmoleukin)、地尼白介素2 (denileukin-diftitox)、依庫珠單抗(eculizumab)、依決洛單抗(edrecolomab)、吉妥珠單抗(gemtuzumab)、替伊莫單抗(ibritumomab-tiuxetan)、咪喹莫特(imiquimod)、干擾素-α、干擾素-β、干擾素-γ、伊匹單抗(ipilimumab)、來那度胺(lenalidomid)、來格司亭(lenograstim)、米伐木肽(mifamurtid)、奧法木單抗(ofatumumab)、奧普瑞白介素(oprelvekin)、溶鏈菌製劑(picibanil)、普樂沙福(plerixafor)、多醣-K、沙格司亭(sargramostim)、西普魯塞-T(sipuleucel-T)、他索那敏(tasonermin)、替西白介素(teceleukin)、托珠單抗(tocilizumab))、抗增殖物質例如(但不限於)安吖啶(amsacrine)、阿格拉濱(arglabin)、三氧化砷、天冬醯胺酶、博來黴素(bleomycin)、白消安(busulfan)、放線菌素D(dactinomycin)、多西他賽(docetaxel)、泛艾黴素(epirubicin)、培洛黴素(peplomycin)、曲妥珠單抗(trastuzumab)、利妥昔單抗、奧妥珠單抗(obinutuzumab)、奧法木單抗(ofatumumab)、托西莫單抗(tositumomab)、芳香酶抑制劑(例如依西美坦(exemestane)、法曲唑(fadrozole)、福美坦(formestane)、來曲唑(letrozole)、阿那曲唑(anastrozole)、伏氯唑(vorozole))、抗雌激素(例如氯地孕酮(chlormadinone)、氟維司群(fulvestrant)、美雄烷(mepitiostane)、他莫昔芬(tamoxifen)、雷洛昔芬(raloxifen)、托瑞米芬(toremifen))、雌激素(例如雌二醇、磷酸聚雌二醇)、促孕素(例如甲羥助孕酮(medroxyprogesteron)、甲地孕酮(megestrol))、拓樸異構酶I抑制劑(例如伊立替康(irinotecan)、托泊替康(topotecan))、拓樸異構酶II抑制劑(例如胺柔比星(amrubicin)、道諾黴素(daunorubicin)、依利醋銨(elliptinium acetate)、依託泊苷(etoposide)、伊達比星(idarubicin)、米托蒽醌(mitoxantrone)、替尼泊苷(teniposide))、微管活性物質(例如卡巴他賽(cabazitaxel)、埃雷布林(eribulin)、太平洋紫杉醇(paclitaxel)、長春鹼(vinblastine)、長春新鹼(vincristine)、長春地辛(vindesine)、長春瑞濱(vinorelbine))、端粒酶抑制劑(例如伊美司他(imetelstat))、烷基化物質及組織蛋白去乙醯酶抑制劑(例如苯達莫司汀(bendamustine)、卡莫司汀(carmustine)、甲川氯(chlormethine)、達卡巴嗪(dacarbazine)、雌氮芥(estramustine)、異環磷醯胺(ifosfamid)、洛莫司汀(lomustine)、二溴甘露醇(mitobronitol)、二溴衛矛醇(mitolactol)、尼莫司汀(nimustine)、潑尼莫司汀(prednimustine)、丙卡巴肼(procarbazine)、雷莫司汀(ranimustine)、鏈佐黴素(streptozotocine)、替莫唑胺(temozolomide)、噻替派(thiotepa)、曲奧舒凡(treosulfan)、曲洛磷胺(trofosfamid)、伏立諾他(vorinostat)、羅米地辛(romidepsin)、帕比司他(panobinostat));影響細胞分化過程之物質,例如阿巴瑞克(abarelix)、胺魯米特(aminoglutethimide)、貝沙羅汀(bexarotene)、MMP抑制劑(肽模擬物(peptide mimetica)、非肽模擬物及四環素(tetracycline),例如馬立馬司他(Marimastat)、BAY 12-9566、BMS-275291、氯膦酸、普啉司他(prinomastat)、去氧羥四環素(doxycycline))、mTOR抑制劑(例如西羅莫司(sirolimus)、依維莫司(everolimus)、替西羅莫司(temsirolimus)、佐他莫司(zotarolimus))、抗代謝物(例如氯法拉濱(clofarabine)、去氧尿苷、胺甲喋呤(methotrexate)、5-氟尿嘧啶、克拉屈濱(cladribine)、阿糖胞苷(cytarabine)、氟達拉濱(fludarabine)、巰嘌呤、培美曲塞(pemetrexed)、雷替曲塞(raltitrexed)、替加氟(tegafur)、硫鳥嘌呤)、鉑化合物(例如卡鉑(carboplatin)、順鉑(cisplatin)、順氯胺鉑(cisplatinum)、依他鉑(eptaplatin)、洛鉑(lobaplatin)、米利鉑(miriplatin)、奈達鉑(nedaplatin)、奧沙利鉑(oxaliplatin));抗血管生成化合物(例如貝伐珠單抗(bevacizumab))、抗雄激素化合物(例如貝伐珠單抗、恩雜魯胺(enzalutamide)、氟他胺(flutamide)、尼魯米特(nilutamide)、比卡魯胺(bicalutamide)、環丙孕酮(cyproterone)、乙酸環丙孕酮)、蛋白酶體抑制劑(例如硼替佐米(bortezomib)、卡非佐米(carfilzomib)、沃洛佐米(oprozomib)、ONYX0914)、促性腺素釋放素激動劑及拮抗劑(例如阿巴瑞克(abarelix)、布舍瑞林(buserelin)、地洛瑞林(deslorelin)、加尼瑞克(ganirelix)、戈舍瑞林(goserelin)、組胺瑞林(histrelin)、曲普瑞林(triptorelin)、地加瑞克(degarelix)、亮丙瑞林(leuprorelin))、甲硫胺酸胺基肽酶抑制劑(例如本阿米德(bengamide)衍生物、TNP-470、PPI-2458)、類肝素酶抑制劑(例如SST0001、PI-88);遺傳修飾之Ras蛋白質之抑制劑(例如法尼基轉移酶抑制劑,例如羅那法尼(lonafarnib)、替吡法尼(tipifarnib))、HSP90抑制劑(例如格爾德黴素(geldamycin)衍生物,例如17-丙烯胺基格爾德黴素、17-去甲氧基格爾德黴素(17AAG)、17-DMAG、鹽酸瑞他黴素(retaspimycin hydrochloride)、IPI-493、AUY922、BIIB028、STA-9090、KW-2478)、紡錘體驅動蛋白抑制劑(例如SB715992、SB743921、噴他脒/氯丙嗪(pentamidine/chlorpromazine))、MEK (促分裂原活化之蛋白激酶激酶)抑制劑(例如曲美替尼(trametinib)、BAY 86-9766 (雷法替尼(refametinib))、AZD6244)、激酶抑制劑(例如:索拉菲尼(sorafenib)、瑞格菲尼(regorafenib)、拉帕替尼(lapatinib)、舒癌特(sutent)、達沙替尼(dasatinib)、西妥昔單抗(cetuximab)、BMS-908662、GSK2118436、AMG 706、厄洛替尼(erlotinib)、吉非替尼(gefitinib)、伊馬替尼(imatinib)、尼羅替尼(nilotinib)、帕唑帕尼(pazopanib)、羅尼西布(roniciclib)、舒尼替尼(sunitinib)、凡德他尼(vandetanib)、威羅菲尼(vemurafenib))、刺蝟信號抑制劑(例如環杷明(cyclopamine)、維莫德吉(vismodegib))、BTK (布魯頓氏酪胺酸激酶(Bruton's tyrosine kinase))抑制劑(例如依魯替尼(ibrutinib))、JAK/pan-JAK (傑納斯激酶(Janus kinase))抑制劑(例如SB-1578、巴瑞替尼(baricitinib)、托法替尼(tofacitinib)、帕利替尼(pacritinib)、莫羅替尼(momelotinib)、魯索替尼(ruxolitinib)、VX-509、AZD-1480、TG-101348)、PI3K抑制劑(例如BAY 1082439、BAY 80-6946 (庫盤裡斯(copanlisib))、ATU-027、SF-1126、DS-7423、GSK-2126458、布帕裡斯(buparlisib)、PF-4691502、BYL-719、XL-147、XL-765、艾代拉裡斯(idelalisib))、SYK (脾酪胺酸激酶)抑制劑(例如弗馬替尼(fostamatinib)、伊西來(Excellair)、PRT-062607)、p53基因療法、雙膦酸鹽(例如依替膦酸鹽(etidronate)、氯膦酸鹽、替魯膦酸鹽(tiludronate)、帕米膦酸鹽(pamidronate)、阿侖膦酸(alendronic acid)、伊班膦酸鹽(ibandronate)、利塞膦酸鹽(risedronate)、唑來膦酸鹽(zoledronate))。對於組合而言,亦應提及以下活性化合物,例如(但不限於):利妥昔單抗、環磷醯胺、多柔比星、多柔比星與雌酮之組合、長春新鹼、氮芥苯丁酸(chlorambucil)、氟達拉濱、地塞米松(dexamethasone)、克拉屈濱、普賴松、131I-chTNT、阿比特龍(abirateron)、阿柔比星(aclarubicin)、阿曲諾英(alitretinoin)、比生群(bisantrene)、亞葉酸鈣、左亞葉酸鈣、卡培他濱(capecitabin)、卡莫氟(carmofur)、氯膦酸、羅米司亭(romiplostim)、克立他酶(crisantaspase)、阿法達依泊汀(darbepoetin alfa)、地西他濱(decitabin)、地諾單抗(denosumab)、二溴螺氯銨(dibrospidium chloride)、伊屈潑帕(eltrombopag)、內皮抑素、環硫雄醇(epitiostanol)、阿法依伯汀(epoetin alfa)、非格司亭(filgrastim)、福莫司汀(fotemustin)、硝酸鎵、吉西他濱(gemcitabin)、格魯妥昔(glutoxim)、組織胺二鹽酸鹽、羥基脲、英丙舒凡(improsulfan)、伊沙匹隆(ixabepilon)、蘭瑞肽(lanreotid)、香菇多醣、左旋咪唑(levamisole)、利舒脲(lisurid)、氯尼達明(lonidamin)、馬索羅酚(masoprocol)、甲基睪固酮、甲氧沙林(methoxsalen)、胺基酮戊酸甲酯(methyl aminolevulinate)、米替福新(miltefosin)、米托胍腙(mitoguazon)、絲裂黴素、米托坦(mitotan)、奈拉濱(nelarabin)、尼妥珠單抗、硝胺丙吖啶(nitracrin)、奧美拉唑(omeprazol)、帕利夫明(palifermin)、帕尼單抗(panitumumab)、培門冬酶、PEG依泊亭β (甲氧基-PEG依泊亭β)、聚乙二醇非格司亭、聚乙二醇干擾素α-2b、噴他佐辛(pentazocin)、噴司他汀(pentostatin)、培磷醯胺(perfosfamid)、吡柔比星(pirarubicin)、普卡黴素(plicamycin)、聚胺葡糖(poliglusam)、卟吩姆鈉(porfimer-sodium)、普拉曲沙(pralatrexate)、喹高利特(quinagolid)、雷佐生(razoxan)、西左非蘭(sizofiran)、索布佐生(sobuzoxan)、甘胺雙唑鈉(sodium glycididazole)、他米巴羅汀(tamibaroten)、替加氟及吉美嘧啶(gimeracil)及奧替拉西(oteracil)之組合、睪固酮、替曲膦(tetrofosmin)、沙利竇邁(thalidomide)、胸腺法新(thymalfasin)、曲貝替定(trabectedin)、維甲酸、曲洛司坦(trilostan)、色胺酸、烏苯美司(ubenimex)、伐普肽(vapreotid)、釔-90玻璃微珠粒、淨司他汀(zinostatin)、淨司他汀斯酯(zinostatin stimalamer)。 亦適於腫瘤療法的係諸如化學療法(例如阿紮胞苷(azacitidine)、貝洛替康(belotecan)、依諾他濱(enocitabine)、美法侖(melphalan)、戊柔比星(valrubicin)、長春氟寧(vinflunin)、佐柔比星(zorubicin))、放射療法(例如I-125晶粒、鈀-103晶粒、氯化鐳-223)或光療法(例如替莫泊芬(temoporfin)、他拉泊芬(talaporfin))等非藥物療法之組合,該組合伴有利用本發明之IRAK4抑制劑之藥物治療,或其在非藥物腫瘤療法(例如化學療法、放射療法或光療法)結束後由利用本發明之IRAK4抑制劑之藥物治療來補充。 除上文所提及之彼等以外,本發明之IRAK4抑制劑亦可與以下活性化合物組合: 用於阿茲海默氏療法之活性化合物,例如乙醯基膽鹼酯酶抑制劑(例如多奈派齊(donepezil)、利凡斯的明(rivastigmine)、加藍他敏(galantamin)、塔克寧(tacrine))、NMDA (N-甲基-D-天冬胺酸鹽)受體拮抗劑(例如美金剛(memantine));用於治療帕金森病之L-DOPA/卡比多巴(carbidopa) (L-3,4-二羥基苯丙胺酸)、COMT (兒茶酚-O-甲基轉移酶)抑制劑(例如恩他卡朋(entacapon))、多巴胺激動劑(例如羅頻羅(ropinrol)、普拉克索(pramipexol)、溴隱亭(bromocriptine))、MAO-B (單胺基氧化酶-B)抑制劑(例如司來吉蘭(selegiline))、抗膽鹼劑(例如三己芬迪(trihexyphenidyl))及NMDA拮抗劑(例如金剛烷胺(amantadin));用於治療多發性硬化之β-干擾素(IFN-β) (例如IFN β-1b、IFN β-1a Avonex®及Betaferon®)、乙酸格拉替雷(glatiramer acetate)、免疫球蛋白、那他珠單抗(natalizumab)、芬戈莫德(fingolimod)及免疫抑制劑(例如米托蒽醌、硫唑嘌呤及環磷醯胺);用於治療肺部病症之物質,例如β-2-擬交感神經藥(例如沙丁胺醇(salbutamol))、抗膽鹼劑(例如吡咯醣)、甲基黃嘌呤(例如茶鹼)、白三烯受體拮抗劑(例如孟魯斯特(montelukast))、PDE-4 (4型磷酸二酯酶)抑制劑(例如羅氟司特(roflumilast))、胺甲喋呤、IgE抗體、硫唑嘌呤及環磷醯胺、含皮質醇製劑;用於治療骨關節炎之物質,例如非類固醇抗發炎物質(NSAID)。除所提及之兩種療法以外,對於類風濕病症(例如類風濕性關節炎、脊椎關節炎及幼年型特發性關節炎)應提及胺甲喋呤及用於B細胞及T細胞療法之生物劑(例如利妥昔單抗、阿巴西普(abatacept))。神經營養物質,例如乙醯基膽鹼酯酶抑制劑(例如多奈派齊)、MAO (單胺基氧化酶)抑制劑(例如司來吉蘭)、干擾素及抗驚厥藥(例如加巴噴丁(gabapentin));用於治療心血管病症之活性化合物,例如β阻斷劑(例如美托洛爾(metoprolol))、ACE抑制劑(例如貝那普利(benazepril))、血管收縮肽受體阻斷劑(例如氯沙坦(losartan)、纈沙坦(valsartan))、利尿劑(例如氫氯噻嗪)、鈣通道阻斷劑(例如硝苯地平(nifedipine))、他汀類藥物(statins) (例如斯伐他汀(simvastatin)、氟伐他汀(fluvastatin));抗糖尿病藥物,例如二甲雙胍(metformin)、格列奈(glinide)(例如那格列奈(nateglinide))、DPP-4 (二肽基肽酶-4)抑制劑(例如利拉利汀(linagliptin)、沙格列汀(saxagliptin)、西他列汀(sitagliptin)、維格列汀(vildagliptin))、SGLT2 (鈉/葡萄糖共運輸蛋白2)抑制劑/格列淨(gliflozin) (例如達格列淨(dapagliflozin)、恩格列淨(empagliflozin))、腸促胰島素模擬物(激素葡萄糖依賴性促胰島素性肽(GIP)及升糖素樣肽1 (GLP-1)類似物/激動劑) (例如艾塞那肽(exenatide)、利拉魯肽(liraglutide)、利西拉肽(lixisenatide))、α-葡萄糖苷酶抑制劑(例如阿卡波糖(acarbose)、米格列醇(miglitol)、伏格列波糖(voglibiose))及磺脲(例如格列本脲(glibenclamide)、甲苯磺丁脲(tolbutamide))、胰島素增敏劑(例如吡格列酮(pioglitazone))及胰島素療法(例如NPH胰島素、賴脯胰島素(insulin lispro)),用於治療低血糖用於治療糖尿病及代謝症候群之物質。脂質降低藥,例如貝特類藥(fibrates)(例如苯紮貝特(bezafibrate)、依託貝特(etofibrate)、非諾貝特(fenofibrate)、吉非羅齊(gemfibrozil))、菸鹼酸衍生物(例如菸鹼酸/拉羅皮蘭(laropiprant))、依折麥布(ezetimib)、他汀類藥物(例如斯伐他汀、氟伐他汀)、陰離子交換劑(例如考來烯胺(colestyramine)、考來替泊(colestipol)、考來維侖(colesevelam))。用於治療慢性發炎腸病症之活性化合物,例如美沙拉嗪(mesalazine)、磺胺塞拉金(sulfasalazine)、硫唑嘌呤、6-巰嘌呤或胺甲喋呤、益生菌(目他弗(Mutaflor)、VSL#3®、乳酸桿菌GG (Lactobacillus GG)、胚芽乳桿菌(Lactobacillus plantarum)、嗜酸乳酸桿菌(L. acidophilus)、乳酪桿菌(L. casei)、嬰兒雙歧桿菌35624 (Bifidobacterium infantis 35624)、屎腸球菌SF68 (Enterococcus fecium SF68)、長雙歧桿菌(Bifidobacterium longum)、尼氏大腸桿菌1917 (Escherichia coli Nissle 1917))、抗生素(例如環丙沙星及甲硝唑(metronidazole))、抗腹瀉藥(例如洛哌丁胺)或輕瀉藥(比沙可啶(bisacodyl))。用於治療紅斑性狼瘡之免疫抑制劑,例如糖皮質激素及非類固醇抗發炎物質(NSAID)、可體松、氯喹(chloroquin)、環孢素、硫唑嘌呤、貝利木單抗、利妥昔單抗、環磷醯胺。用於器官移植之藥物,例如(但不限於)鈣調神經磷酸酶抑制劑(例如他克莫司及環孢素)、細胞分裂抑制劑(例如硫唑嘌呤、嗎替麥考酚酯、黴酚酸(mycophenolic acid)、依維莫司或西羅莫司)、雷帕黴素、巴利昔單抗、達克珠單抗(daclizumab)、抗CD3抗體、抗T-淋巴球免疫球蛋白/抗淋巴球免疫球蛋白。用於皮膚病學病症之維生素D3類似物,例如卡泊三醇(calcipotriol)、他卡西醇(tacalcitol)或骨化三醇、柳酸、尿素、環孢素、胺甲喋呤、依法利珠單抗(efalizumab)。 對於上文所提及之病症之治療及/或預防,亦應提及包含本發明化合物中之至少一者及一或多種其他活性化合物(特定而言EP4抑制劑(前列腺素E2受體4抑制劑)、P2X3抑制劑(P2X嘌呤受體3)、PTGES抑制劑(前列腺素E合酶抑制劑)或AKR1C3抑制劑(醛酮還原酶家族1成員C3抑制劑))之藥劑。 本發明化合物可以全身及/或局部方式起作用。出於此目的,其可以適宜方式投與,例如藉由經口、非經腸、經肺、經鼻、經舌下、經舌、經頰、經直腸、經真皮、經皮、或結膜途徑,經由耳部或作為植入物或支架。 本發明化合物可以適於該等投與途徑之投與形式投與。 適用於經口投與之投與形式係彼等以下形式:根據先前技術工作並快速及/或以改良方式釋放根據本發明化合物且含有呈結晶及/或非晶形及/或溶解形式之本發明化合物,例如錠劑(未包衣或包衣錠劑,例如具有控制本發明化合物之釋放之抗胃液或延遲溶解或不溶解包衣)、在口腔中快速崩解之錠劑或膜/扁圓劑、膜/凍乾物、膠囊(例如硬或軟明膠膠囊)、糖包衣之錠劑、顆粒、丸劑、粉末、乳液、懸浮液、氣溶膠或溶液。 非經腸投與可避免再吸收步驟(例如藉由靜脈內、動脈內、心內、脊柱內或腰髓內途徑)或包括再吸收(例如藉由肌內、皮下、皮內、經皮或腹膜內途徑)來實現。適於非經腸投與之投與形式包括呈溶液、懸浮液、乳液、凍乾劑或無菌粉劑之形式的注射及輸注用製劑。 其他投與途徑之適宜實例係吸入藥劑(包括粉劑吸入器、噴霧器)、滴鼻劑、溶液或噴霧劑;用於經舌、舌下或經頰投與之錠劑、膜/扁圓劑或膠囊、栓劑、耳或眼製劑、陰道膠囊、水性懸浮液(洗劑、振盪混合物)、親脂性懸浮液、軟膏、乳霜、經皮治療系統(例如貼片)、牛奶、膏糊、泡沫、撒粉、移植物或支架。 較佳者係經口或非經腸投與,尤其經口投與。 可將本發明化合物轉化成所提及之投與形式。此可以本身已知之方式藉由與惰性、無毒性、醫藥上適宜之助劑混合實現。該等賦形劑包括載劑(例如微晶纖維素、乳糖、甘露醇)、溶劑(例如液體聚乙二醇)、乳化劑及分散劑或潤濕劑(例如十二烷基硫酸鈉、聚氧山梨醇酐油酸酯)、黏合劑(例如聚乙烯吡咯啶酮)、合成及天然聚合物(例如白蛋白)、穩定劑(例如氧化劑,例如抗壞血酸)、著色劑(例如無機顏料,例如氧化鐵)及矯味劑及/或矯臭劑。 本發明進一步提供包含至少一種本發明化合物通常連同一或多種惰性、無毒、醫藥上適宜之賦形劑之藥劑以及其用於上文所提及目的之用途。 一般而言,在非經腸投與之情形下有利於投與約0.001 mg/kg體重至1 mg/kg體重、較佳約0.01 mg/kg體重至0.5 mg/kg體重之量以達成有效結果。在經口投與之情形下,劑量為約0.01 mg/kg體重至100 mg/kg體重,較佳約0.01 mg/kg體重至20 mg/kg體重,且最佳0.1 mg/kg體重至10 mg/kg體重。 然而,在一些情形下偏離所述量可能係必需的,具體而言隨體重、投與途徑、個體對活性化合物之反應、製劑性質及進行投與之時間或間隔而變。因此,在一些情形下,以小於上文所提及之最低量進行管控可能即已足夠,而在其他情形下則必須超出所提及上限。在投與較大量之情形下,可適當的將該等分成若干個別劑量並全天投與。 下列工作實例闡釋本發明。本發明並非限於該等實例。 除非另外陳述,否則以下測試及實例中之百分比係重量百分比;份數係重量份數。液/液溶液之溶劑比率、稀釋比率及濃度數據在每一情形下均基於體積。 於以下合成方案1至6中闡釋本發明之式(I)物質之製備。 如合成方案1中所顯示,6-氯-5-硝基-1H-吲唑(CAS 101420-98-8)可以烷基化反應或光延反應(Mitsunobu reaction)中吲轉化為中間體1。適於烷基化反應之反應物係具有視情況經取代之烷基氯化物、烷基溴化物、烷基碘化物或4-甲苯磺酸烷基酯之彼等。所使用之烷基鹵化物或4-甲苯磺酸烷基酯可自市面購得或可以與自文獻已知之途徑類似之方式來製備(以4-甲苯磺酸烷基酯之製備而言,一個實例係將適當醇與4-甲苯磺醯基氯化物在三乙胺或吡啶存在下反應;例如參見Bioorganic and Medicinal Chemistry, 2006, 14, 12 4277 - 4294)。視情況地,在使用烷基氯化物或烷基溴化物之情形下,亦可添加諸如碘化鉀或碘化鈉等鹼金屬碘化物。所用的鹼可為(例如)碳酸鉀、碳酸銫或氫化鈉。在反應性烷基鹵化物之情形下,在一些情形下亦可使用N-環己基-N-甲基環己胺。有用溶劑包括(例如) 1-甲基吡咯啶-2-酮、DMF、DMSO或THF。視情況地,所使用之烷基鹵化物或4-甲苯磺酸烷基酯係具有已視情況預先經保護基團保護之官能基(亦參見P. G. M. Wuts、T. W. Greene,Greene’s Protective Groups in Organic Synthesis , 第四版, ISBN: 9780471697541)。若使用(例如)具有一或多個羥基之烷基鹵化物或4-甲苯磺酸烷基酯,則該等羥基可視情況經熟習此項技術者熟悉之第三丁基(二甲基)矽基或類似的含矽保護基團保護。或者,該羥基亦可經四氫-2H-吡喃(THP)基團或經乙醯基或苯甲醯基保護。然後可在合成中間體1或中間體2之後、或者在合成(I)-a之後將所使用之保護基團脫離。若使用(例如)第三丁基(二甲基矽基)基團作為保護基團,則可例如在諸如THF等溶劑中使用四丁基氟化銨使其脫離。可使用(例如) 4-甲苯磺酸(視情況呈一水合物形式)使THP保護基團脫離。可藉由利用氫氧化鈉水溶液處理使乙醯基或苯甲醯基脫離。 所採用之烷基鹵化物或4-甲苯磺酸烷基酯可視情況含有可藉由熟習此項技術者已知之反應轉化為替代官能基之官能基。所提及之實例係羧酸烷基酯之氧化或還原反應或水解反應(例如比較Science of Synthesis , Georg Thieme Verlag)。 或者,在光延反應(例如參見K. C. K. Swamy等人Chem. Rev. 2009, 109, 2551 – 2651)中利用視情況經取代之烷基醇將6-氯-5-硝基-1H-吲唑轉化為中間體1。可利用各種膦(例如三苯基膦、三丁基膦或1,2-二苯基膦基乙烷)與偶氮二甲酸二異丙基酯(CAS 2446-83-5)或在文獻中提及之其他二氮烯衍生物之組合(K. C. K. Swamy等人Chem. Rev. 2009, 109, 2551 - 2651)。較佳者係使用三苯基膦及偶氮二甲酸二異丙基酯。若烷基醇具有官能基,則可使用已知之保護基團策略(如在上文所提及與烷基鹵化物反應之情形下) (其他指示可參見P. G. M. Wuts、T. W. Greene,Greene’s Protective Groups in Organic Synthesis, 第四版, ISBN: 9780471697541)且可在合成中間體1之後或者在合成中間體2之後或在合成本發明之通式(I)化合物之後實現氧化或還原步驟(如在上文所提及與烷基鹵化物反應之情形下)。 可藉由還原硝基將中間體1轉化為中間體2。出於此目的可使用於水及乙醇中之鐵及氯化銨(亦例如參見Journal of the Chemical Society, 1955, 2412 -2419)。為自中間體2製備本發明之式(I)化合物之子集(I)-a,可採用醯胺合成。可採用自文獻(Amino Acids, Peptides and Proteins in Organic Chemistry, 第3卷- Building Blocks, Catalysis and Coupling Chemistry, Andrew B. Hughes, Wiley, Chapter 12 - Peptide-Coupling Reagents, 407-442;Chem. Soc. Rev., 2009, 38, 606)已知之各種偶合劑。舉例而言,可使用1-(3-二甲基胺基丙基)-3-乙基碳二亞胺鹽酸鹽與1-羥基-1H-苯并三唑水合物(HOBt, WO2012107475;Bioorg. Med. Chem. Lett., 2008 , 18, 2093)、四氟硼酸(1H-苯并三唑-1-基氧基)(二甲基胺基)-N,N-二甲基甲銨鹽(TBTU, CAS 125700-67-6)、六氟磷酸(二甲基胺基)-N,N-二甲基(3H-[1,2,3]三唑并[4,5-b]吡啶-3-基氧基)甲銨鹽(HATU, CAS 148893-10-1)、丙烷膦酸酐(呈於乙酸乙酯或DMF中之溶液形式,CAS68957-94-8)或二-1H-咪唑-1-基甲酮(CDI)之組合作為偶合劑,且在每一情形下可向反應混合物中添加諸如三乙胺或N-乙基-N-異丙基丙-2-胺等鹼。較佳者係使用於DMF中之HATU及N-乙基-N-異丙基丙-2-胺。合成方案1 本發明之式(I)化合物之子集(I)-a (其中R1 = Cl)之製備. R2 及R3 係如針對通式(I)所定義。 本發明之式(I)化合物之另一子集(I)-b可如合成方案2中所顯示獲得。此處,使6-氯-1H-吲唑-5-胺(CAS 221681-75-0)與羧酸反應。於合成方案1中闡述之醯胺合成之方法適於此目的。較佳者係使用於THF中1-羥基-1H-苯并三唑水合物、1-(3-二甲基胺基丙基)-3-乙基碳二亞胺鹽酸鹽及三乙胺。此得到中間體3,然後以與合成方案1類似之方式使該中間體與烷基鹵化物反應,得到(I)-b。合成方案2 本發明之式(I)化合物之子集(I)-b(其中R1 = Cl)之製備. R2 及R3 係如針對通式(I)所定義。 本發明之式(I)化合物之另一子集(I)-c可如合成方案3中所顯示獲得。此處,將5-硝基-1H-吲唑-6-醇(CAS 1082041-56-2)轉化為中間體4。已知一些中間體4之製備(例如比較WO2013174744、WO201574986、Bioorganic and Medicinal Chemistry, 2004, 12, 2115 – 2137)。對於中間體4自5-硝基-1H-吲唑-6-醇之製備較佳的係光延反應(比較合成方案1)。尤佳者係使用於THF中之三苯基膦及偶氮二甲酸二異丙基酯。 利用中間體4開始,藉由還原反應獲得中間體5 (對於使用於水及乙醇中之鐵及氯化銨比較合成方案1;然而,亦可使用於氫氣氛中之碳載鈀(WO2015/74986)或於甲醇中之氯化錫(II) (比較WO2013174744))。如合成方案1中所闡述之醯胺合成產生中間體6。與合成方案1類似,烷基化反應得到(I)-c。合成方案3 本發明之式(I)化合物之子集(I)-c (其中R1 = OR4 )之製備.R2 、R3 及R4 係如針對通式(I)所定義。 本發明之式(I)化合物之另一子集(I)-d可如合成方案4中所顯示獲得。如合成方案1中所闡述之中間體4係在烷基化反應中轉化為中間體7。硝基之還原(比較合成方案4)產生中間體8,然後藉由醯胺合成(比較合成方案1)將該中間體轉化為(I)-d。合成方案4 本發明之式(I)化合物之子集(I)-d (其中R1 = OR4 )之製備. R2 、R3 及R4 係如針對通式(I)所定義。 如合成方案5中所闡述,中間體8可藉由替代途徑自中間體5來製備。首先利用第三丁氧基羰基保護中間體5 (比較WO2015091426),且此後如合成方案1中所闡述進行烷基化並在二氯甲烷中在三氟乙酸存在下去除保護基團。合成方案5 中間體8之替代製備. R2 及R4 係如針對通式(I)所定義。 亦可根據合成方案6自中間體11、12及13開始藉由利用烷基鹵化物或4-甲苯磺酸烷基酯之烷基化反應引入取代基R4 ,此容許獲得中間體6 (本發明之式(I)化合物之子集(I)-e)及中間體10。此處較佳者係使用於DMF中利用烷基溴化物及碳酸鉀之烷基化反應。所使用之烷基鹵化物可視情況具有在烷基化反應之後可藉由熟習此項技術者已知之轉變轉化為其他官能基之官能基。可提及之實例係使用硼氫化鈉將羧酸烷基酯基團還原為醇基團或在水及THF中使用氫氧化鋰將羧酸烷基酯水解為羧酸基團。合成方案6 本發明之式(I)化合物之子集(I)-e (其中R1 = OR4 )之製備及中間體6及10之製備。R2 、R3 及R4 係如針對通式(I)所定義。實例化合物之合成 縮寫及說明 術語飽和鹽水意指飽和氯化鈉水溶液。 中間體及實例之化學名稱係使用ACD / LABS (Batch 12.01.版)軟體生成。方法 在一些情形下,藉由LC-MS分析本發明化合物及其前體及/或中間體。 方法A1:UPLC(MeCN-HCOOH): 儀器:Waters Acquity UPLC-MS SQD 3001;管柱:Acquity UPLC BEH C18 1.7 50×2.1 mm;流動相A:水 + 0.1體積%之甲酸(99%),流動相B:乙腈;梯度:0-1.6 min 1-99% B,1.6-2.0 min 99% B;流速0.8 ml/min;溫度:60℃;注入:2 µl;DAD掃描:210-400 nm;MS ESI+,ESI-,掃描範圍160-1000 m/z;ELSD。 方法A2:UPLC (MeCN-NH3 ): 儀器:Waters Acquity UPLC-MS SQD 3001;管柱:Acquity UPLC BEH C18 1.7 50×2.1 mm;流動相A:水 + 0.2體積%之甲酸(32%),流動相B:乙腈;梯度:0-1.6 min 1-99% B,1.6-2.0 min 99% B;流速0.8 ml/min;溫度:60℃;注入:2 µl;DAD掃描:210-400 nm;MS ESI+,ESI-,掃描範圍160-1000 m/z;ELSD。 方法A3:(LC-MS) 儀器:Agilent 1290 Infinity LC;管柱:Acquity UPLC BEH C18 1.7 50×2.1mm;移動相A:水 + 0.05體積%甲酸,移動相B:乙腈 + 0.05體積%甲酸;梯度:0-1.7 min 2-90% B,1.7-2.0 min 90% B;流速1.2 ml/min;溫度:60℃;注入:2 µl;DAD掃描:190-390 nm;MS:Agilent TOF 6230。 方法A4:(LC-MS) 儀器:Waters Acquity;管柱:Kinetex (Phenomenex), 50×2 mm;移動相A:水 + 0.05體積%甲酸,移動相B:乙腈 + 0.05體積%甲酸;梯度:0-1.9 min 1-99% B,1.9-2.1 min 99% B;流速1.5 ml/min;溫度:60℃;注入:0.5 µl;DAD掃描:200-400 nm。 在一些情形下,本發明化合物及其前體及/或中間體係藉由以下實例性製備型HPLC方法來純化: 方法P1:系統:Waters自純化系統:幫浦2545, 試樣管控器2767, CFO, DAD 2996, ELSD 2424, SQD;管柱:XBridge C18 5µm 100×30 mm;移動相:A:水 + 0.1體積%甲酸,移動相B:乙腈;梯度:0-8 min 10-100% B, 8-10 min 100% B;流速:50 ml/min;溫度:室溫;溶液:最大250 mg / 最大2.5 ml DMSO或DMF;注入:1 × 2.5 ml;檢測:DAD掃描範圍210-400 nm;MS ESI+, ESI-,掃描範圍160-1000 m/z。 方法P2:系統:Waters自純化系統:幫浦254, 試樣管控器2767, CFO, DAD 2996, ELSD 2424, SQD 3100;管柱:XBridge C18 5µm 100×30 mm;移動相:A:水 + 0.2體積%氨(32%),移動相B:甲醇;梯度:0-8 min 30-70% B;流速:50 ml/min;溫度:室溫;檢測:DAD掃描範圍210-400 nm;MS ESI+, ESI-,掃描範圍160-1000 m/z;ELSD。 方法P3:系統:Labomatic, 幫浦:HD-5000, 流份收集器:LABOCOL Vario-4000,UV檢測器:Knauer UVD 2.1S;管柱:XBridge C18 5µm 100×30 mm;移動相A:水 + 0.2體積%氨(25%),移動相B:乙腈;梯度:0-1 min 15% B, 1-6.3 min 15-55% B, 6.3-6.4min 55-100% B, 6.4-7.4min 100% B;流速:60 ml/min;溫度:室溫;溶液:最大250 mg / 2 ml DMSO;注入:2 × 2 ml;檢測:UV 218 nm;軟體:SCPA PrepCon5。 方法P4:系統:Labomatic, 幫浦:HD-5000,流份收集器:LABOCOL Vario-4000,UV檢測器:Knauer UVD 2.1S;管柱:Chromatorex RP C18 10µm 125×30 mm, 移動相:A:水 + 0.1體積%甲酸,移動相B:乙腈;梯度:0 - 15 min 65 – 100% B;流速:60 ml/min;溫度:室溫;溶液:最大250 mg / 2 ml DMSO;注入:2 × 2 ml;檢測:UV 254 nm;軟體:SCPA PrepCon5。 方法P5:系統:Sepiatec:Prep SFC100;管柱:Chiralpak IA 5µm 250×20 mm;移動相A:二氧化碳,移動相B:乙醇;梯度:等度20% B;流速:80 ml/min;溫度:40℃;溶液:最大250 mg / 2 ml DMSO;注入:5 × 0.4 ml檢測:UV 254 nm。 方法P6:系統:Agilent:Prep 1200, 2×Prep Pump, DLA, MWD, Gilson:液體處置器215;管柱:Chiralcel OJ-H 5µm 250×20 mm;移動相A:己烷,移動相B:乙醇;梯度:等度30% B;流速:25 ml/min;溫度:25℃;溶液:187 mg / 8 ml乙醇/甲醇;注入:8 × 1.0 ml檢測:UV 280 nm。 方法P7:系統:Labomatic,幫浦:HD-5000,流份收集器:LABOCOL Vario-4000,UV檢測器:Knauer UVD 2.1S;管柱:XBridge C18 5µm 100×30 mm;移動相A:水 + 0.1體積%甲酸,移動相B:乙腈;梯度:0-3 min:65%B等度,3-13 min: 65-100% B;流速:60 ml/min;溫度:室溫;溶液:最大250 mg / 2 ml DMSO;注入:2 × 2 ml;檢測:UV 254 nm。 方法P8:系統:Agilent:Prep 1200, 2×Prep Pump, DLA, MWD, Gilson:液體處置器215;管柱:Chiralpak IF 5µm 250×20 mm;移動相A:乙醇,移動相B:甲醇;梯度:等度50% B;流速:25 ml/min;溫度:25℃;溶液:600 mg / 7 ml N,N-二甲基甲醯胺;注入:10 × 0.7 ml檢測:UV 254 nm。 對於一些本發明化合物及其前體及/或中間體之製備,在矽膠上使用來自Biotage之Isolera® 裝置執行管柱層析純化(「急速層析」)。此係使用來自Biotage之筒柱(例如不同大小之「SNAP Cartridge, KP_SIL」筒柱及來自Interchim之不同大小之「Interchim Puriflash Silica HP 15UM驟沸塔」筒柱)進行。中間體 1-1 6- -2-(3- 甲氧基 -3- 甲基丁基 )-5- 硝基 -2H- 吲唑 首先在5 ml THF中裝填500 mg (2.53 mmol) 6-氯-5-硝基-1H-吲唑(CAS編號:101420-98-8)、390 µl (3.0 mmol) 3-甲氧基-3-甲基丁-1-醇及996 mg (3.80 mmol)三苯基膦,緩慢逐滴添加540 µl (2.8 mmol)偶氮二甲酸二異丙基酯並將混合物在25℃下攪拌21.5小時。用水稀釋反應混合物並用乙酸乙酯萃取三次。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。藉由急速層析(Interchim 15µm筒柱, KP-Sil, 移動相:己烷/乙酸乙酯)純化殘餘物。此得到792 mg (粗製產物)標題化合物。 LC-MS (方法A1):Rt = 1.22 min (UV檢測器:TIC Smooth),質量測定值297.001 H-NMR (500MHz, DMSO-d6, 所選信號):δ = 2.10 - 2.16 (m, 1H), 3.12 (s, 1H), 4.50 - 4.55 (m, 1H), 8.00 (d, 1H), 8.67 (s, 1H), 8.82 (d, 1H)。中間體 2-1 6- -2-(3- 甲氧基 -3- 甲基丁基 )-2H- 吲唑 -5- 將791 mg 6-氯-2-(3-甲氧基-3-甲基丁基)-5-硝基-2H-吲唑(粗製產物)溶解於8 ml乙醇中,並添加2 ml水。然後添加1.48 g (26.6 mmol)鐵粉末及71.1 mg (1.33 mmol)氯化銨,並將混合物在90℃下攪拌1.5小時。冷卻反應混合物並藉助矽藻土(Celite)過濾。用乙醇將濾液洗滌三次並濃縮。用乙酸乙酯將水相萃取三次。用飽和氯化鈉溶液洗滌合併之有機相,過濾(疏水過濾器)並濃縮。此得到715 mg標題化合物之粗製產物。 LC-MS (方法A1):Rt = 0.92 min (UV檢測器:TIC Smooth),質量測定值267.00。中間體 3-1 N-(6- -1H- 吲唑 -5- )-6-( 二氟甲基 ) 吡啶 -2- 甲醯胺 將2.00 g (11.9 mmol) 6-氯-1H-吲唑-5-胺(CAS編號:221681-75-0)、2.48 g (14.3 mmol) 6-(二氟甲基)吡啶-2-甲酸、1.83 g (11.9 mmol) 1-羥基-1H-苯并三唑水合物、4.58 g (23.9 mmol) 1-(3-二甲基胺基丙基)-3-乙基碳二亞胺鹽酸鹽及5.0 ml (36 mmol)三乙胺於40 ml THF中之混合物在25℃下攪拌20.5小時。用水稀釋反應混合物並濃縮。利用抽吸濾出固體並用水洗滌三次並用乙醚洗滌三次。然後添加醚,並攪拌混合物。利用抽吸濾出固體,用乙醚洗滌三次並在減壓下乾燥。此得到3.45 g標題化合物之粗製產物。 LC-MS (方法A1):Rt = 1.09 min (UV檢測器:TIC Smooth),質量測定值322.00。中間體 4-1 5- 硝基 -6-( 氧雜環丁 -3- 基氧基 )-1H- 吲唑 首先在50 ml THF中裝填4.00 g (22.3 mmol) 5-硝基-1H-吲唑-6-醇(CAS編號:1082041-56-2)、1.74 g (23.4 mmol)氧雜環丁-3-醇及8.79 g (33.5 mmol)三苯基膦,緩慢逐滴添加6.5 ml (33 mmol)偶氮二甲酸二異丙基酯並將混合物在25℃下攪拌17.5小時。用水稀釋反應混合物並用乙酸乙酯萃取三次。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。藉由急速層析(Biotage SNAP筒柱(100 g;KP-Sil), 移動相:己烷/乙酸乙酯)純化殘餘物。此得到4.58 g標題化合物之粗製產物。 LC-MS (方法A1):Rt = 0.78 min (UV檢測器:TIC Smooth),質量測定值235.001 H NMR (500MHz, DMSO-d6): δ = 4.59 - 4.64 (m, 2H), 4.95 - 5.02 (m, 2H), 5.50 - 5.56 (m, 1H), 6.84 (s, 1H), 8.21 (s, 1H), 8.49 (s, 1H), 13.33 (s, 1H)。中間體 4-2 5- 硝基 -6-( 氧雜環丁 -3- 基甲氧基 )-1H- 吲唑 首先在25 ml THF中裝填2.00 g (11.2 mmol) 5-硝基-1H-吲唑-6-醇(CAS編號:1082041-56-2)、1.03 g (11.7 mmol)氧雜環丁-3-基甲醇及4.39 g (16.7 mmol)三苯基膦,緩慢逐滴添加3.2 ml (17 mmol)偶氮二甲酸二異丙基酯並將混合物在25℃下攪拌18.5小時。用水稀釋反應混合物並用乙酸乙酯萃取三次。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。藉由急速層析(Biotage SNAP筒柱(100 g;KP-Sil), 移動相:己烷/乙酸乙酯)純化殘餘物。此得到1.03 g標題化合物之粗製產物。 LC-MS (方法A1):Rt = 0.79 min (UV檢測器:TIC Smooth),質量測定值249.001 H NMR (500MHz, DMSO-d 6 ): δ = 3.41 - 3.48 (m, 1H), 4.40 (d, 2H), 4.48 (t, 2H), 4.70 (dd, 2H), 7.28 (s, 1H), 8.20 (s, 1H), 8.43 (s, 1H), 13.38 (s, 1H)。中間體 4-3 5- 硝基 -6-[(3S)- 四氫呋喃 -3- 基氧基 ]-1H- 吲唑 首先在25 ml THF中裝填2.00 g (11.2 mmol) 5-硝基-1H-吲唑-6-醇(CAS編號:1082041-56-2)、940 µl (12 mmol) (3S)-四氫呋喃-3-醇及4.39 g (16.7 mmol)三苯基膦,緩慢逐滴添加3.2 ml (17 mmol)偶氮二甲酸二異丙基酯並將混合物在25℃下攪拌67小時。濃縮反應混合物並藉由急速層析(Biotage SNAP筒柱(100 g;KP-Sil), 移動相:己烷/乙酸乙酯)純化。此得到3.08 g標題化合物。 LC-MS (方法A1):Rt = 0.82 min (UV檢測器:TIC Smooth),質量測定值249.00。中間體 6-1 N-(6- 異丙氧基 -1H- 吲唑 -5- )-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 首先在400 ml THF中裝填7.02 g (36.73 mmol) 6-(三氟甲基)吡啶-2-甲酸以及13.48 g (41.98 mmol) O-(苯并三唑-1-基)-N,N,N',N'-四甲基脲鎓四氟硼酸鹽及7.31 ml (41.98 mmol) N,N-二異丙基乙胺。將混合物在25℃下攪拌30分鐘,且然後添加6.69 g (34.98 mmol) 6-異丙氧基-1H-吲唑-5-胺(CAS編號:1498329-80-8)。將反應混合物在25℃下攪拌16小時。濃縮反應混合物並將殘餘物分配於水與乙酸乙酯之間。用乙酸乙酯將混合物萃取兩次,並用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。藉由急速層析(Biotage SNAP筒柱(340 g;KP-Sil), 移動相:己烷/乙酸乙酯)純化殘餘物。濃縮合併之產物流份,將其懸浮於水中並劇烈攪拌20分鐘。利用抽吸濾出產物並用水及乙醚洗滌。此得到8.56 g標題化合物。 LC-MS (方法A1):Rt = 1.23 min (UV檢測器:TIC Smooth),質量測定值364.001 H NMR (400 MHz, DMSO-d6): δ = 1.41 (d, 6 H), 4.85 (spt, 1 H), 7.18 (s, 1 H), 8.01 (s, 1 H), 8.21 (dd, 1 H), 8.40 (t, 1 H), 8.47 (d, 1 H), 8.81 (s, 1 H), 10.67 (s, 1 H), 12.85 (s, 1 H)。中間體 6-2 6-( 二氟甲基 )-N-(6- 乙氧基 -1H- 吲唑 -5- ) 吡啶 -2- 甲醯胺 將512 mg (2.89 mmol) 6-乙氧基-1H-吲唑-5-胺、600 mg (3.47 mmol) 6-(二氟甲基)吡啶-2-甲酸、442 mg (2.89 mmol) 1-羥基-1H-苯并三唑水合物、1.11 g (5.78 mmol) 1-(3-二甲基胺基丙基)-3-乙基碳二亞胺鹽酸鹽及1.2 ml (8.7 mmol)三乙胺於10 ml THF中之混合物在25℃下攪拌20.5小時。用水稀釋反應混合物並用乙酸乙酯萃取三次。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。將乙醚添加至殘餘物中,攪拌混合物並利用抽吸濾出所形成之固體,用乙醚洗滌三次並乾燥。此得到499 mg標題化合物。 LC-MS (方法A1):Rt = 1.10 min (UV檢測器:TIC Smooth),質量測定值332.001 H-NMR (400MHz, DMSO-d6): δ = 1.51 (t, 3H), 4.23 (q, 2H), 6.96 (t, 1H), 7.10 (s, 1H), 7.94 - 8.02 (m, 2H), 8.26 - 8.36 (m, 2H), 8.76 (s, 1H), 10.71 (s, 1H), 12.86 (s, 1H)。中間體 6-3 N-[6-( 苄基氧基 )-1H- 吲唑 -5- ]-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 首先在150 ml THF中裝填5.40 g (22.57 mmol) 6-(三氟甲基)吡啶-2-甲酸以及10.87 g (33.85 mmol) O-(苯并三唑-1-基)-N,N,N',N'-四甲基脲鎓四氟硼酸鹽及5.90 ml (33.85 mmol) N,N-二異丙基乙胺。將混合物在25℃下攪拌30分鐘,且然後添加5.40 g (22.57 mmol) 6-(苄基氧基)-1H-吲唑-5-胺(CAS編號:1499162-36-5)。將反應混合物在25℃下攪拌16小時。濃縮反應混合物並將殘餘物分配於水與乙酸乙酯之間。使用乙酸乙酯萃取混合物,且使用飽和氯化鈉溶液洗滌合併之有機相,經硫酸鈉乾燥,過濾並濃縮。將乙醚添加至粗製產物中,將混合物劇烈攪拌10分鐘並利用抽吸濾出所形成之沈澱物。此得到6.72 g標題化合物。 LC-MS (方法A1):Rt = 1.28 min (UV檢測器:TIC Smooth),質量測定值412.001 H NMR (400 MHz, DMSO-d6): δ = 5.34 (s, 2 H), 7.31 (s, 1 H), 7.41 (d, 3 H), 7.58 (dd, 2 H), 8.04 (s, 1 H), 8.18 (d, 1 H), 8.40 (d, 1 H), 8.45 - 8.53 (m, 1 H), 8.85 (s, 1 H), 10.41 (s, 1 H), 12.97 (s, 1 H)。中間體 6-4 N-[6-( 環丙基甲氧基 )-1H- 吲唑 -5- ]-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 將1.00 g N-(6-羥基-1H-吲唑-5-基)-6-(三氟甲基)吡啶-2-甲醯胺溶解於20 ml DMF中,並在攪拌下添加1.72 g (12.41 mmol)碳酸鉀。將懸浮液在25℃下攪拌30分鐘,且然後添加431 µl (4.65 mmol) (溴甲基)環丙烷。將反應混合物在25℃下攪拌16小時。然後用水稀釋反應混合物,並利用抽吸濾出所形成之固體,用乙醚洗滌並在減壓下乾燥。此得到585 mg標題化合物。 LC-MS (方法A1):Rt = 1.25 min (UV檢測器:TIC Smooth),質量測定值376.001 H NMR (300 MHz, DMSO-d6): δ = 0.40 - 0.51 (m, 2 H), 0.59 - 0.71 (m, 2 H), 1.27 - 1.41 (m, 1 H), 4.05 (d, 2 H), 7.08 (s, 1 H), 8.01 (s, 1 H), 8.16 - 8.25 (m, 1 H), 8.41 (t, 1 H), 8.48 (d, 1 H), 8.82 (s, 1 H), 10.65 (s, 1 H), 12.90 (s, 1 H)。中間體 6-5 N-[6-( 三氟甲氧基 )-1H- 吲唑 -5- ]-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 將600 mg 6-(三氟甲氧基)-1H-吲唑-5-胺(CAS編號:1499162-39-8, 作為粗製產物使用)及673 mg (3.52 mmol) 6-(三氟甲基)吡啶-2-甲酸溶解於15 ml THF中,並在25℃下添加360 mg (2.35 mmol) 1-羥基-1H-苯并三唑水合物、900 mg (4.70 mmol) 1-(3-二甲基胺基丙基)-3-乙基碳二亞胺鹽酸鹽及980 µl (7.0 mmol)三乙胺。將反應混合物在RT下攪拌24小時且然後用水稀釋,並濾出沈澱之固體。用水及乙醚洗滌固體並在減壓下乾燥。此得到406 mg標題化合物。 LC-MS (方法A1):Rt = 1.26 min (UV檢測器:TIC Smooth),質量測定值390.00。中間體 6-6 2- 環丙基 -N-(6- 甲氧基 -1H- 吲唑 -5- )-1,3- 噁唑 -4- 甲醯胺 將586 mg (3.59 mmol) 6-甲氧基-1H-吲唑-5-胺(CAS編號:749223-61-8)及500 mg (3.27 mmol) 2-環丙基-1,3-噁唑-4-甲酸(CAS編號:1060816-04-7)溶解於10 ml THF中,並在25℃下添加500 mg (3.3 mmol) 1-羥基-1H-苯并三唑水合物、1.25 g (6.5 mmol) 1-(3-二甲基胺基丙基)-3-乙基碳二亞胺鹽酸鹽及1.37 ml (9.8 mmol)三乙胺。添加水,部分地去除溶劑並濾出固體,用水及乙醚洗滌並乾燥。此得到758 mg標題化合物。 LC-MS (方法A1):Rt = 1.05 min (UV檢測器:TIC Smooth),質量測定值298.00。中間體 6-7 N-(6- 甲氧基 -1H- 吲唑 -5- )-6-( 吡咯啶 -1- ) 吡啶 -2- 甲醯胺 將692 mg HATU添加至248 mg 6-甲氧基-1H-吲唑-5-胺、350 mg 6-(吡咯啶-1-基)吡啶-2-甲酸及0.79 ml N,N-二異丙基乙胺於5.3 ml THF中之混合物中,並將混合物在室溫下攪拌過夜。添加水並利用抽吸濾出固體並在減壓下乾燥。此得到503 mg標題化合物之粗製產物。1 H-NMR (400MHz, DMSO-d6 ): δ [ppm]= 1.98 - 2.07 (m, 4H), 3.46 – 3.58 (m, 4H), 4.00 (s, 3H), 6.74 (d, 1H), 7.10 (s, 1H), 7.33 (d, 1H), 7.72 (dd, 1H), 7.98 (s, 1H), 8.74 (s, 1H), 10.85 (s, 1H), 12.88 (s, 1H)。中間體 7-1 4-(6- 乙氧基 -5- 硝基 -2H- 吲唑 -2- )-2- 甲基丁 -2- 首先在100 ml DMF中裝填10 g (48 mmol) 6-乙氧基-5-硝基-1H-吲唑(參見WO2015091426)及20 g碳酸鉀,並將混合物攪拌15分鐘。然後添加8.8 ml (72.3 mmol) 4-溴-2-甲基丁-2-醇,並將混合物在70℃下攪拌過夜。添加另一2.9 ml (24 mmol) 4-溴-2-甲基丁-2-醇及6.6 g碳酸鉀,並在70℃下繼續攪拌另一夜。利用抽吸濾出混合物並將濾液吸收於甲苯中並用水洗滌三次。用飽和氯化鈉溶液洗滌混合物並經硫酸鎂乾燥且然後濃縮。急速層析(Biotage, 己烷/乙酸乙酯梯度)得到4.87 g標題化合物。 LC-MS (方法A1):Rt = 0.99 min (UV檢測器:TIC Smooth),質量測定值293.00。 1H-NMR (400MHz, DMSO-d6): δ [ppm]= 1.15 (s, 6 H) 1.35 (t, 3H) 1.99 - 2.06 (m, 2 H) 4.14 – 4.21 (q, 2 H) 4.45 - 4.53 (m, 2 H) 4.54 (s, 1H) 7.23 (s, 1H) 8.37 ( s, 1 H) 8.60 (s, 1 H)中間體 7-2 2- 甲基 -4-[5- 硝基 -6-( 氧雜環丁 -3- 基氧基 )-2H- 吲唑 -2- ] -2- 將4.58 g 5-硝基-6-(氧雜環丁-3-基氧基)-1H-吲唑(粗製產物)溶解於50 ml DMF中,並在攪拌下添加6.06 g (43.8 mmol)碳酸鉀及7.16 g (作為粗製產物使用) 3-羥基-3-甲基丁基-4-甲苯磺酸酯(M. Shimizu等人,Bioorganic and Medicinal Chemistry 2006 ,14, 4277 – 4294)。將反應混合物於80℃下攪拌18小時,用水稀釋並用乙酸乙酯萃取三次。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。藉由急速層析(Biotage SNAP筒柱, KP-Sil, 移動相:己烷/乙酸乙酯)純化殘餘物。此得到627 mg標題化合物。 LC-MS (方法A1):Rt = 0.87 min (UV檢測器:TIC Smooth),質量測定值321.001 H NMR (500MHz, DMSO-d6): δ = 1.14 (s, 6H), 1.99 - 2.04 (m, 2H), 4.47 - 4.51 (m, 2H), 4.53 (s, 1H), 4.57 (dd, 2H), 4.97 - 5.02 (m, 2H), 5.42 - 5.49 (m, 1H), 6.90 (s, 1H), 8.47 (s, 1H), 8.63 (d, 1H)。中間體 7-3 2- 甲基 -4-[5- 硝基 -6-( 氧雜環丁 -3- 基甲氧基 )-2H- 吲唑 -2- ] -2- 將1.03 g 5-硝基-6-(氧雜環丁-3-基甲氧基)-1H-吲唑(粗製產物)溶解於7.5 ml DMF中,並在攪拌下添加777 mg (4.65 mmol) 4-溴-2-甲基丁-2-醇及1.29 g (9.30 mmol)碳酸鉀。將反應混合物在100℃下攪拌16.5小時。用水稀釋反應混合物並用乙酸乙酯萃取三次。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。將反應混合物分配於乙酸乙酯與水之間。用飽和氯化鈉溶液將有機相洗滌兩次,藉助疏水過濾器過濾並濃縮。藉由製備型HPLC純化殘餘物。此得到287 mg標題化合物。 LC-MS (方法A1):Rt = 0.84 min;MS (ESIpos):m/z = 335 [M+H]+ 1 H NMR (500MHz, DMSO-d6): δ = 1.15 (s, 6H), 2.00 - 2.06 (m, 2H), 3.40 - 3.47 (m, 1H), 4.35 (d, 2H), 4.46 (t, 2H), 4.48 - 4.52 (m, 2H), 4.70 (dd, 2H), 7.30 (s, 1H), 8.39 (s, 1H), 8.61 (d, 1H)。中間體 7-4 2- 甲基 -4-{5- 硝基 -6-[(3S)- 四氫呋喃 -3- 基氧基 ]-2H- 吲唑 -2- } -2- 將3.08 g 5-硝基-6-[(3S)-四氫呋喃-3-基氧基]-1H-吲唑(粗製產物)溶解於50 ml DMF中,並在攪拌下添加3.84 g (27.8 mmol)碳酸鉀及4.55 g (作為粗製產物使用) 3-羥基-3-甲基丁基-4-甲苯磺酸酯。將反應混合物在80℃下攪拌22小時。用水稀釋反應混合物並用乙酸乙酯萃取三次。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。藉由急速層析(Biotage SNAP筒柱(100 g;KP-Sil), 移動相:己烷/乙酸乙酯)純化殘餘物。此得到618 mg標題化合物之粗製產物。 LC-MS (方法A1):Rt = 0.91 min (UV檢測器:TIC Smooth),質量測定值335.00。中間體 8-1 [5- 胺基 -6-( 環丙基甲氧基 )-2H- 吲唑 -2- ] 乙酸乙酯 將5.52 ml (71.6 mmol)三氟乙酸添加至2.79 g (7.16 mmol)於50 ml二氯甲烷中之{5-[(第三丁氧基羰基)胺基]-6-(環丙基甲氧基)-2H-吲唑-2-基}乙酸乙酯中,並將混合物在25℃下攪拌6小時。將混合物倒入飽和碳酸氫鈉溶液中並攪拌10分鐘,分離各相並用二氯甲烷將水相萃取兩次。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。此得到2.03 g標題化合物。 LC-MS (方法A1):Rt = 0.75 min (UV檢測器:TIC Smooth),質量測定值289.00 1H NMR (400 MHz, DMSO-d6): δ = 0.32 - 0.43 (m, 2 H), 0.53 - 0.64 (m, 2 H), 1.20 (t, 3 H), 1.29 (s, 1 H), 3.86 (d, 2 H), 4.14 (q, 2 H), 4.65 (br. s., 2 H), 5.17 (s, 2 H), 6.65 (s, 1 H), 6.76 (s, 1 H), 7.84 (d, 1 H)。中間體 8-2 4-[5- 胺基 -6-( 苄基氧基 )-2H- 吲唑 -2- ]-2- 甲基丁 -2- 將2.06 ml (26.79 mmol)三氟乙酸添加至1.14 g (2.68 mmol)於20 ml二氯甲烷中之[6-(苄基氧基)-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]胺基甲酸第三丁基酯中,並將混合物在25℃下攪拌3小時。將混合物倒入飽和碳酸氫鈉溶液中。利用抽吸濾出所形成之固體,用水洗滌並乾燥。此得到840 mg標題化合物。 LC-MS (方法A1):Rt = 0.82 min (UV檢測器:TIC Smooth),質量測定值325.00 1H NMR (400 MHz, DMSO-d6): δ = 1.12 (s, 6 H), 1.90 - 2.02 (m, 2 H), 4.21 - 4.38 (m, 2 H), 4.46 (s, 1 H), 4.76 (br. s., 2 H), 5.17 (s, 2 H), 6.67 (s, 1 H), 6.91 (s, 1 H), 7.28 - 7.36 (m, 1 H), 7.36 - 7.45 (m, 2 H), 7.48 - 7.55 (m, 2 H), 7.85 (s, 1 H)。中間體 8-3 4-(5- 胺基 -6- 甲氧基 -2H- 吲唑 -2- )-2- 甲基丁 -2- 將610 mg [2-(3-羥基-3-甲基丁基)-6-甲氧基-2H-吲唑-5-基]胺基甲酸第三丁基酯於10 ml二氯甲烷及1.3 ml (16 mmol)三氟乙酸中之混合物在25℃下攪拌19.5小時。將混合物倒入飽和碳酸氫鈉溶液中並攪拌10分鐘,分離各相用二氯甲烷並將水相萃取兩次。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。此得到318 mg標題化合物之粗製產物。 LC-MS (方法A2):Rt = 0.72 min (UV檢測器:TIC Smooth),質量測定值249.00。中間體 8-4 6- 甲氧基 -2-[3-( 四氫 -2H- 吡喃 -2- 基氧基 ) 丙基 ]-2H- 吲唑 -5- 首先在5 ml二氯甲烷中裝填789 mg (0.95 mmol) {6-甲氧基-2-[3-(四氫-2H-吡喃-2-基氧基)丙基]-2H-吲唑-5-基}胺基甲酸第三丁基酯。添加735 µl (9.53 mmol)三氟乙酸,並將混合物在25℃下攪拌22小時。添加另一735 µl (9.53 mmol)三氟乙酸,並將混合物在25℃下攪拌68小時。用水萃取反應混合物並用二氯甲烷洗滌三次。用飽和碳酸氫鈉溶液及飽和氯化鈉溶液洗滌合併之有機相,過濾(疏水過濾器)並濃縮。再一次,將飽和碳酸氫鈉溶液(pH約等於9)添加至水溶液中,並用乙酸乙酯將混合物萃取三次。用飽和氯化鈉溶液洗滌合併之有機相,過濾(疏水過濾器)並濃縮。將氯化鈉及乙酸乙酯添加至水相中並將混合物攪拌30分鐘。分離各相,並用乙酸乙酯將水相萃取兩次以上。過濾(疏水過濾器)合併之有機相並濃縮。此得到199 mg標題化合物。 LC-MS (方法A2):Rt = 0.55 min (UV檢測器:TIC Smooth),質量測定值221.00。中間體 8-5 4-(5- 胺基 -6- 乙氧基 -2H- 吲唑 -2- )-2- 甲基丁 -2- 將4.87 g (16.6 mmol) 4-(6-乙氧基-5-硝基-2H-吲唑-2-基)-2-甲基丁-2-醇溶解於164 ml THF及70 ml甲醇中,並使用1.46 g (0.1 mmol)活性碳載鈀,在氫氣氛中在25℃下氫化3小時。藉助矽藻土過濾反應混合物,用乙酸乙酯洗滌過濾器,並濃縮濾液。此得到4.37 g標題化合物。 1H-NMR (400MHz, DMSO-d6): δ = 1.13 (s, 6 H), 1.40 (t, 3H), 1.92 – 1.99 (m, 2 H), 4.00 – 4.07 (q, 2 H), 4.26 - 4.34 (m, 2 H), 4.49 (s, 1H), 4.57 (s, 2H), 6.62 (s, 1H), 6.80 ( s, 1 H), 7.83 (s, 1 H)。中間體 8-6 4-[5- 胺基 -6-( 氧雜環丁 -3- 基氧基 )-2H- 吲唑 -2- ]-2- 甲基丁 -2- 將616 mg 2-甲基-4-[5-硝基-6-(氧雜環丁-3-基氧基)-2H-吲唑-2-基]丁-2-醇溶解於12 ml乙醇中,並添加3 ml水。然後添加963 mg (17.3 mmol)鐵粉末及46.1 mg (863 µmol)氯化銨,並將混合物在90℃下攪拌17小時。藉助矽藻土過濾反應混合物並濃縮並將殘餘物吸收於THF中,再次過濾並濃縮。此得到574 mg標題化合物之粗製產物。 LC-MS (方法A2):Rt = 0.68 min (UV檢測器:TIC Smooth),質量測定值291.00。中間體 8-7 4-[5- 胺基 -6-( 氧雜環丁 -3- 基甲氧基 )-2H- 吲唑 -2- ]-2- 甲基丁 -2- 將281 mg 2-甲基-4-[5-硝基-6-(氧雜環丁-3-基甲氧基)-2H-吲唑-2-基]丁-2-醇溶解於7.5 ml乙醇中,並添加2.5 ml水。然後添加426 mg (7.62 mmol)鐵粉末及20.4 mg (381 µmol)氯化銨,並將混合物在90℃下攪拌21小時。冷卻反應混合物並過濾。濃縮濾液並將殘餘物吸收於THF中,再次過濾並濃縮。此得到227 mg標題化合物之粗製產物。 LC-MS (方法A2):Rt = 0.70 min (UV檢測器:TIC Smooth),質量測定值303.00。中間體 8-8 4-{5- 胺基 -6-[(3S)- 四氫呋喃 -3- 基氧基 ]-2H- 吲唑 -2- }-2- 甲基丁 -2- 將610 mg 2-甲基-4-{5-硝基-6-[(3S)-四氫呋喃-3-基氧基]-2H-吲唑-2-基}丁-2-醇(粗製產物)溶解於10 ml乙醇中,並添加3 ml水。然後添加843 mg (15.1 mmol)鐵粉末及40 mg氯化銨,並將混合物在90℃下攪拌5小時。冷卻反應混合物並藉助矽藻土過濾。濃縮濾液並將殘餘物吸收於THF中,再次過濾並濃縮。將殘餘物吸收於2 ml DMF中並藉由製備型HPLC純化。此得到348 mg標題化合物之粗製產物。 LC-MS (方法A4):Rt = 0.72 min;MS (ESIpos):m/z = 305 [M+H]+中間體 9-1 (6- 甲氧基 -1H- 吲唑 -5- ) 胺基甲酸第三丁基酯 合成闡述於WO2015091426中。中間體 10-1 [2-(3- 羥基 -3- 甲基丁基 )-6- 甲氧基 -2H- 吲唑 -5- ] 胺基甲酸第三丁基酯 將2.00 g (7.60 mmol) (6-甲氧基-1H-吲唑-5-基)胺基甲酸第三丁基酯溶解於20 ml 1-甲基-2-吡咯啶酮中,並在攪拌下添加1.90 g (11.4 mmol) 4-溴-2-甲基丁-2-醇、3.15 g (22.8 mmol)碳酸鉀及1.89 g (11.4 mmol)碘化鉀。將懸浮液在80℃下攪拌21小時。添加另一525 mg (3.8 mmol)碳酸鉀及630 mg (3.8 mmol)碘化鉀,並將混合物在80℃下再攪拌6小時。用水稀釋反應混合物並用乙酸乙酯萃取三次。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。藉由急速層析(Biotage Interchim 15µm筒柱(80 g;KP-Sil), 移動相:己烷/乙酸乙酯)純化殘餘物。濃縮合併之產物流份並乾燥。此得到610 mg標題化合物。 LC-MS (方法A1):Rt = 1.16 min (UV檢測器:TIC Smooth),質量測定值349.001 H-NMR (500MHz, DMSO-d 6 ): δ = 1.13 (s, 6H), 1.46 (s), 1.96 - 2.03 (m, 2H), 3.85 (s, 3H), 4.36 - 4.41 (m, 2H), 4.50 (s, 1H), 6.97 (s, 1H), 7.77 (s, 1H), 7.89 (br. s., 1H), 8.17 (d, 1H)。中間體 10-2 {6- 甲氧基 -2-[3-( 四氫 -2H- 吡喃 -2- 基氧基 ) 丙基 ]-2H- 吲唑 -5- } 胺基甲酸第三丁基酯 將1.00 g (6-甲氧基-1H-吲唑-5-基)胺基甲酸第三丁基酯溶解於15 ml DMF中,並在攪拌下添加967 µl (5.7 mmol) 2-(3-溴丙氧基)四氫-2H-吡喃、1.58 g (11.4 mmol)碳酸鉀及757 mg (4.6 mmol)碘化鉀。將反應混合物在100℃下攪拌20小時。添加另一484 µl (2.9 mmol) 2-(3-溴丙氧基)四氫-2H-吡喃,並將混合物在100℃下攪拌24小時。然後用水稀釋反應混合物並用乙酸乙酯萃取。用飽和氯化鈉溶液洗滌合併之有機相,分離各相並藉助疏水過濾器過濾。將殘餘物吸收於二氯甲烷中且在濃縮期間吸附於Isolute上。藉由急速層析(Biotage SNAP筒柱(100 g;KP-Sil), 移動相:己烷/乙酸乙酯)純化殘餘物。濃縮合併之產物流份並乾燥。此得到790 mg標題化合物。 LC-MS (方法A1):Rt = 1.33 min (UV檢測器:TIC Smooth),質量測定值405.00。中間體 10-3 [6-( 苄基氧基 )-2-(3- 羥基 -3- 甲基丁基 )-2H- 吲唑 -5- ] 胺基甲酸第三丁基酯 將3.50 (10.3 mmol) [6-(苄基氧基)-1H-吲唑-5-基]胺基甲酸第三丁基酯(CAS編號:1799835-17-8)溶解於30 ml DMF中,並在攪拌下添加4.28 g (30.94 mmol)碳酸鉀及1.71 g (10.31 mmol)碘化鉀。將懸浮液在25℃下攪拌30分鐘,且然後添加2.58 g (15.47 mmol) 4-溴-2-甲基丁-2-醇。將反應混合物在100℃下攪拌16小時。添加另一1.72 g (10.31 mmol) 4-溴-2-甲基丁-2-醇及1.42 g (10.31 mmol)碳酸鉀,並將混合物在100℃下攪拌另一24小時。然後用水稀釋反應混合物並用乙酸乙酯萃取兩次。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。藉由急速層析(Biotage SNAP筒柱(340 g;KP-Sil), 移動相:己烷/乙酸乙酯)純化殘餘物。此得到1.14 g標題化合物。 LC-MS (方法A1):Rt = 1.35 min (UV檢測器:TIC Smooth),質量測定值425.00 1H NMR (400 MHz, DMSO-d6): δ = 1.13 (s, 6 H), 1.45 (s, 9 H), 1.92 - 2.02 (m, 2 H), 4.34 - 4.42 (m, 2 H), 4.47 (s, 1 H), 5.20 (s, 2 H), 7.03 (s, 1 H), 7.29 - 7.37 (m, 1 H), 7.37 - 7.44 (m, 2 H), 7.53 (d, 2 H), 7.81 - 7.87 (m, 2 H), 8.18 (s, 1 H)。中間體 10-4 {5-[( 第三丁氧基羰基 ) 胺基 ]-6-( 環丙基甲氧基 )-2H- 吲唑 -2- } 乙酸乙酯 將3.00 g (8.95 mmol){5-[(第三丁氧基羰基)胺基]-6-羥基-2H-吲唑-2-基}乙酸乙酯溶解於45 ml DMF中,並在攪拌下添加3.71 g (26.84 mmol)碳酸鉀。將懸浮液在25℃下攪拌10分鐘,且然後添加995 µl (10.73 mmol) (溴甲基)環丙烷。將反應混合物在25℃下攪拌16小時且在80℃下攪拌4小時。將反應混合物分配於水與乙酸乙酯之間,並分離各相。使用乙酸乙酯萃取水相。使用飽和氯化鈉溶液洗滌合併之有機相並濃縮。藉由急速層析(Biotage SNAP筒柱(340 g;KP-Sil), 移動相:己烷/乙酸乙酯)純化殘餘物。此得到2.79 g標題化合物。 LC-MS (方法A1):Rt = 1.37 min (UV檢測器:TIC Smooth),質量測定值389.00 1H NMR (400 MHz, 氯仿-d): δ = 0.36 - 0.44 (m, 2 H), 0.66 - 0.74 (m, 2 H), 1.28 (t, 3 H), 1.32 - 1.42 (m, 1 H), 1.57 (s, 9 H), 3.91 (d, 2 H), 4.25 (q, 2 H), 5.10 (s, 2 H), 6.92 (s, 1 H), 7.30 (s, 1 H), 7.81 (s, 1 H), 8.28 (br. s., 1 H)。中間體 11-1 N-(6- 羥基 -1H- 吲唑 -5- )-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 將7.20 g (17.5 mmol) N-[6-(苄基氧基)-1H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺溶解於400 ml甲醇中,並將燒瓶抽真空且然後用氮沖洗(此操作重複兩次以上)。添加9.29 g (8.73 mmol)碳載鈀並燒瓶將抽真空並用氫沖洗。將反應混合物在標準氫壓力下在25℃下氫化3小時。然後將燒瓶抽真空並用氮填充。添加5.50 g甲酸銨並將混合物在25℃下攪拌1小時。藉助矽藻土過濾反應混合物並濃縮濾液。此得到499 mg標題化合物。 LC-MS (方法A1):Rt = 0.95 min (UV檢測器:TIC Smooth),質量測定值322.00 1H NMR (400 MHz, DMSO-d6): δ = 6.98 (s, 1 H), 7.95 (s, 1 H), 8.17 - 8.23 (m, 1 H), 8.40 (t, 1 H), 8.47 (d, 1 H), 8.72 (s, 1 H), 10.46 (s, 1 H), 10.76 (br. s., 1 H), 12.63 (s, 1 H)。中間體 12-1 N-[6- 羥基 -2-(3- 羥基 -3- 甲基丁基 )-2H- 吲唑 -5- ]-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 步驟 A將840 mg (2.58 mmol) 4-[5-胺基-6-(苄基氧基)-2H-吲唑-2-基]-2-甲基丁-2-醇及542 mg (2.84 mmol) 6-(三氟甲基)吡啶-2-甲酸溶解於50 ml THF中,並添加395 mg (2.58 mmol) 1-羥基-1H-苯并三唑水合物、990 mg (5.16 mmol) 1-(3-二甲基胺基丙基)-3-乙基碳二亞胺鹽酸鹽及1.1 ml三乙胺並將混合物在25℃下攪拌2小時。在濃縮溶液後,將水添加至所形成之沈澱物中並利用抽吸濾出沈澱物,用水及乙醚洗滌並在減壓下乾燥。此得到1.02 g N-[6-(苄基氧基)-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺。 LC-MS (方法A1):Rt = 1.37 min (UV檢測器:TIC Smooth),質量測定值498.00 1H NMR (400 MHz, DMSO-d6): δ = 1.15 (s, 6H), 1.99 - 2.06 (m, 2H), 4.40 - 4.46 (m, 2H), 4.51 (s, 1H), 5.30 (s, 2H), 7.32 (s, 1H), 7.37 - 7.45 (m, 3H), 7.54 - 7.60 (m, 2H), 8.18 (dd, 1H), 8.33 (s, 1H), 8.38 (t, 1H), 8.46 (d, 1H), 8.77 (s, 1H), 10.46 (s, 1H)。步驟 B 將940 mg (1.89 mmol) N-[6-(苄基氧基)-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺溶解於94 ml甲醇中,並將燒瓶抽真空且然後用氮沖洗(此操作重複兩次以上)。添加201 mg (0.20 mmol)碳載鈀並將燒瓶抽真空並用氫沖洗。將反應混合物在標準氫壓力下在25℃下氫化3小時。藉助矽藻土過濾反應混合物並用甲醇洗滌,並濃縮濾液。此得到731 mg N-[6-羥基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺。 LC-MS (方法A1):Rt = 1.03 min (UV檢測器:TIC Smooth),質量測定值408.00 1H NMR (400 MHz, DMSO-d6): δ = 1.15 (s, 6 H), 1.93 - 2.04 (m, 2 H), 4.28 - 4.44 (m, 2 H), 4.50 (s, 1 H), 6.93 (s, 1 H), 8.21 (d, 1 H), 8.26 (s, 1 H), 8.40 (t, 1 H), 8.47 (d, 1 H), 8.66 (s, 1 H), 10.53 (s, 1 H), 10.61 (s, 1 H)。中間體 13-1 {5-[( 第三丁氧基羰基 ) 胺基 ]-6- 羥基 -2H- 吲唑 -2- } 乙酸乙酯 將4.50 g (10.6 mmol) {6-(苄基氧基)-5-[(第三丁氧基羰基)胺基]-2H-吲唑-2-基}乙酸乙酯(CAS編號: 1799835-24-7)溶解於225 ml乙醇中,並將燒瓶抽真空且然後用氮沖洗(此程序重複兩次以上)。添加1.13 g (1.06 mmol)碳載鈀並將燒瓶抽真空並用氫沖洗。將反應混合物在標準氫壓力下在25℃下氫化4小時。藉助矽藻土過濾反應混合物並濃縮濾液。此得到3.44 g標題化合物。 LC-MS (方法A1):Rt = 1.05 min (UV檢測器:TIC Smooth),質量測定值335.00 1H NMR (300 MHz, DMSO-d6): δ = 1.21 (t, 3 H), 1.47 (s, 9 H), 4.15 (q, 2 H), 5.24 (s, 2 H), 6.82 (s, 1 H), 7.64 (s, 1 H), 7.93 (s, 1 H), 8.10 - 8.16 (m, 1 H), 10.26 (s, 1 H)。 在ExpNo UBOT8758-1下實例 1 1-( 二氟甲基 )-N-[2-(3- 羥基 -3- 甲基丁基 )-6- 甲氧基 -2H- 吲唑 -5- ]-1H- 吡唑 -3- 甲醯胺 將158 mg (66%純, 418 µmol) 4-(5-胺基-6-甲氧基-2H-吲唑-2-基)-2-甲基丁-2-醇(CAS編號:1799835-12-3), 81.4 mg (502 µmol) 1-(二氟甲基)-1H-吡唑-3-甲酸、191 mg (502 µmol) HATU及87 µl (500 µmol) N,N-二異丙基乙胺溶解於2 ml THF中。將混合物於25℃下攪拌23小時。用水稀釋反應混合物並用乙酸乙酯萃取兩次。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。藉由製備型HPLC純化殘餘物。將合併之產物流份凍乾。此得到95.2 mg標題化合物。 LC-MS (方法A2):Rt = 1.01 min (UV檢測器:TIC Smooth),質量測定值393.001 H-NMR (400MHz, DMSO-d6): δ = 1.15 (s, 6H), 1.96 - 2.07 (m, 2H), 3.34 (s, 3 H), 4.38 - 4.47 (m, 2H), 4.53 (s, 1H), 7.04 (d, 1H), 7.12 (s, 1H), 7.98 (t, 1H), 8.30 (s, 1H), 8.46 (d, 1H), 8.50 (s, 1H), 9.39 (s, 1H)。實例 2 N-[2-(3- 羥基 -3- 甲基丁基 )-6- 甲氧基 -2H- 吲唑 -5- ]-2-( 三氟甲基 )-1,3- 噁唑 -4- 甲醯胺 將158 mg (66%純, 418 µmol) 4-(5-胺基-6-甲氧基-2H-吲唑-2-基)-2-甲基丁-2-醇、90.9 mg (502 µmol) 2-(三氟甲基)-1,3-噁唑-4-甲酸、191 mg (502 µmol) HATU及87 µl (500 µmol) N,N-二異丙基乙胺溶解於2 ml THF中。將混合物於25℃下攪拌23小時。用水稀釋反應混合物並用乙酸乙酯萃取兩次。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。藉由製備型HPLC純化殘餘物。將合併之產物流份凍乾。此得到133 mg標題化合物。 LC-MS (方法A3):Rt = 1.12 min (UV檢測器:TIC Smooth),質量測定值412.001 H-NMR (400MHz, DMSO-d6): δ = 1.15 (s, 6H), 1.99 - 2.05 (m, 2H), 3.95 (s, 3H), 4.39 - 4.47 (m, 2H), 4.52 (s, 1H), 7.13 (s, 1H), 8.32 (s, 1H), 8.44 (s, 1H), 9.23 (s, 1H), 9.41 (s, 1H)。實例 3 6-( 二氟甲基 )-N-[2-(3- 羥基丙基 )-6- 甲氧基 -2H- 吲唑 -5- ] 吡啶 -2- 甲醯胺 將100 mg (0.45 mmol) 3-(5-胺基-6-甲氧基-2H-吲唑-2-基)丙-1-醇及94 mg (0.54 mmol) 6-(二氟甲基)吡啶-2-甲酸溶解於3.0 ml DMF中,添加69 mg (0.45 mmol) 1-羥基-1H-苯并三唑水合物、173 mg (0.90 mmol) 1-(3-二甲基胺基丙基)-3-乙基碳二亞胺鹽酸鹽及189 µl (1.36 mmol)三乙胺並將混合物在25℃下攪拌27小時。添加水,並用乙酸乙酯將混合物萃取三次。藉助疏水過濾器過濾合併之有機相並濃縮。藉由製備型HPLC (管柱:YMC-Triart 5µm 100×30 mm)純化殘餘物。冷凍乾燥含有產物之流份。此得到18 mg標題化合物。 LC-MS (方法A1):Rt = 1.02 min (UV檢測器:TIC Smooth),質量測定值376.001 H NMR (500 MHz, DMSO-d6): δ = 2.04 (五重峰, 2 H), 3.37 - 3.43 (m, 2 H), 4.00 (s, 3 H), 4.40 (t, 2 H), 4.62 (t, 1 H), 7.15 (t, 1 H), 7.14 (s, 1 H), 7.95 - 8.02 (m, 1 H), 8.26 - 8.36 (m, 3 H), 8.69 (s, 1 H), 10.55 (s, 1 H)。實例 4 N-[6- 乙氧基 -2-(3- 羥基 -3- 甲基丁基 )-2H- 吲唑 -5- ]-4-( 三氟甲基 )-1,3- 噻唑 -2- 甲醯胺 將於2 ml DMF中之80 mg (0.3 mmol) 4-(5-胺基-6-乙氧基-2H-吲唑-2-基)-2-甲基丁-2-醇、90 mg (0.45 mmol) 4-(三氟甲基)-1,3-噻唑-2-甲酸、63 µl ( 0.45 mmol)三乙胺及174 mg (0.45 mmol) HATU在RT下攪拌過夜。藉由製備型HPLC純化得到95 mg(70 %)目標化合物。 LC-MS (方法A1):Rt = 1.29 min (UV檢測器:TIC Smooth),質量測定值442.00 1H-NMR (400MHz, DMSO-d6): δ = 1.15 (s, 6 H) 1.45 (t, 3H) 1.98 – 2.06 (m, 2 H) 4.16 – 4.25 (q, 2 H) 4.38 - 4.48 (m, 2 H) 4.52 (s, 1H) 7.14 (s, 1H) 8.33 ( s, 1 H) 8.46 (s, 1 H) 8.90 (s, 1H) 9.92 (s, 1H)。實例 5 N-[6- 乙氧基 -2-(3- 羥基 -3- 甲基丁基 )-2H- 吲唑 -5- ]-2-( 三氟甲基 )-1,3- 噻唑 -4- 甲醯胺 將於2 ml DMF中之80 mg (0.3 mmol) 4-(5-胺基-6-乙氧基-2H-吲唑-2-基)-2-甲基丁-2-醇、90 mg (0.45 mmol) 2-(三氟甲基)-1,3-噻唑-4-甲酸、63 µl ( 0.45 mmol)三乙胺及174 mg (0.45 mmol) HATU在RT下攪拌過夜。藉由製備型HPLC純化得到90 mg(67%)目標化合物。 LC-MS (方法A1):Rt = 1.27 min (UV檢測器:TIC Smooth),質量測定值442.00 1H-NMR (400MHz, DMSO-d6): δ = 1.15 (s, 6 H) 1.47 (t, 3H) 1.98 – 2.05 (m, 2 H) 4.15 – 4.24 (q, 2 H) 4.38 - 4.46 (m, 2 H) 4.53 (s, 1H) 7.11 (s, 1H) 8.31 ( s, 1 H) 8.57 (s, 1 H) 8.87 (s, 1H) 9.98 (s, 1H)。實例 6 N-[6- 乙氧基 -2-(3- 羥基 -3- 甲基丁基 )-2H- 吲唑 -5- ]-2- 甲基 -1,3- 噁唑 -5- 甲醯胺 將於2 ml DMF中之80 mg (0.3 mmol) 4-(5-胺基-6-乙氧基-2H-吲唑-2-基)-2-甲基丁-2-醇、58 mg (0.45 mmol) 2-甲基-1,3-噁唑-5-甲酸、63 µl ( 0.45 mmol)三乙胺及174 mg (0.45 mmol) HATU在RT下攪拌過夜。藉由製備型HPLC純化得到80 mg (70 %)目標化合物。 LC-MS (方法A1):Rt = 0.91 min (UV檢測器:TIC Smooth),質量測定值372.00 1H-NMR (400MHz, DMSO-d6): δ = 1.14 (s, 6H), 1.43 (t, 3H), 1.98 – 2.05 (m, 2H), 2.54 (s, 3H), 4.11 – 4.29 (q, 2H), 4.38 - 4.45 (m, 2H), 4.52 (s, 1H), 7.07 (s, 1H), 7.82 ( s, 1H), 8.17 (s, 1H), 8.29 (s, 1H), 9.21 (s, 1H)。實例 7 1-( 二氟甲基 )-N-[6- 乙氧基 -2-(3- 羥基 -3- 甲基丁基 )-2H- 吲唑 -5- ]-1H- 吡唑 -3- 甲醯胺 將於2 ml DMF中之80 mg (0.3 mmol) 4-(5-胺基-6-乙氧基-2H-吲唑-2-基)-2-甲基丁-2-醇、74 mg (0.45 mmol) 1-(二氟甲基)-1H-吡唑-3-甲酸、63 µl ( 0.45 mmol)三乙胺及174 mg (0.45 mmol) HATU在RT下攪拌過夜。藉由製備型HPLC純化得到20 mg(16 %)目標化合物。 LC-MS (方法A1):Rt = 1.05 min (UV檢測器:TIC Smooth),質量測定值407.00 1H-NMR (400MHz, DMSO-d6): δ = 1.15 (s, 6 H) 1.46 (t, 3H), 1.97 – 2.05 (m, 2 H), 4.16 – 4.25 (q, 2H), 4.37 - 4.45 (m, 2H), 4.52 (s, 1H), 7.03 (m, 1H), 7.10 ( s, 1H), 7.96 (t, 1H), 8.29 (s, 1 H), 8.48 (m, 1H), 8.52 (s, 1H), 9.53 (s, 1H)。實例 8 N-[6- 乙氧基 -2-(3- 羥基 -3- 甲基丁基 )-2H- 吲唑 -5- ]-2- 甲基 -1,3- 噻唑 -5- 甲醯胺 將於2 ml DMF中之80 mg (0.3 mmol) 4-(5-胺基-6-乙氧基-2H-吲唑-2-基)-2-甲基丁-2-醇、65 mg (0.45 mmol) 2-甲基-1,3-噻唑-5-甲酸、63 µl ( 0.45 mmol)三乙胺及174 mg (0.45 mmol) HATU在RT下攪拌過夜。藉由製備型HPLC純化得到70 mg(59 %)目標化合物。 LC-MS (方法A1):Rt = 1.12 min (UV檢測器:TIC Smooth),質量測定值388.00 1H-NMR (400MHz, DMSO-d6): δ = 1.14 (s, 6 H), 1.49 (t, 3 H), 1.96 – 2.06 (m, 2 H), 2.75 (s, 3 H), 4.16 – 4.25 (q, 2 H), 4.36 - 4.45 (m, 2 H), 4.52 (s, 1 H), 7.09 (s, 1 H), 8.29 (m, 2 H), 8.61 (s, 1 H), 10.03 (s, 1 H)。實例 9 2- 環丙基 -N-[6- 乙氧基 -2-(3- 羥基 -3- 甲基丁基 )-2H- 吲唑 -5- ]-1,3- 噁唑 -4- 甲醯胺 將於2 ml DMF中之80 mg (0.3 mmol) 4-(5-胺基-6-乙氧基-2H-吲唑-2-基)-2-甲基丁-2-醇、70 mg (0.45 mmol) 2-環丙基-1,3-噁唑-4-甲酸、63 µl ( 0.45 mmol)三乙胺及174 mg (0.45 mmol) HATU在RT下攪拌過夜。藉由製備型HPLC純化得到65 mg(53%)目標化合物。 LC-MS (方法A1):Rt = 1.15 min (UV檢測器:TIC Smooth),質量測定值398.00 1H-NMR (400MHz, DMSO-d6): δ = 1.00 – 1.06 (m, 2 H), 1.11 – 1.18 (m, 8 H), 1.48 (t, 3 H), 1.96 – 2.04 (m, 2 H), 2.17 – 2.26 (m, 1 H), 4.15 – 4.23 (q, 2 H), 4.36 - 4.44 (m, 2 H), 4.52 (s, 1 H), 7.09 (s, 1 H), 8.27 (s, 1 H), 8.54 (s, 1 H), 8.64 (s, 1 H), 9.55 (s, 1 H)。實例 10 6- 胺基 -N-[6- 乙氧基 -2-(3- 羥基 -3- 甲基丁基 )-2H- 吲唑 -5- ] 吡啶 -2- 甲醯胺 將於4 ml DMF中之160 mg (0.6 mmol) 4-(5-胺基-6-乙氧基-2H-吲唑-2-基)-2-甲基丁-2-醇、126 mg (0.91 mmol) 6-胺基吡啶-4-甲酸、127 µl ( 0.91 mmol)三乙胺及347 mg (0.91 mmol) HATU在RT下攪拌過夜。藉由製備型HPLC純化得到100 mg(43%)目標化合物。 LC-MS (方法A1):Rt = 0.94 min (UV檢測器:TIC Smooth),質量測定值383.00 1H-NMR (400MHz, DMSO-d6): δ = 1.15 (s, 6 H), 1.53 (t, 3 H), 1.96 – 2.06 (m, 2 H), 4.16 – 4.27 (q, 2 H), 4.35 - 4.45 (m, 2 H), 4.51 (s br, 1 H), 6.22 (s br, 2 H), 6.72 (d, 1 H), 7.08 (s, 1 H), 7.31 (d, 1 H), 7.62 (t, 1 H), 8.26 (s, 1 H), 8.66 (s, 1 H), 10.58 (s, 1 H)。實例 11 N-[6- 乙氧基 -2-(3- 羥基 -3- 甲基丁基 )-2H- 吲唑 -5- ]-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 將於1.5 ml DMF中之80 mg (0.3 mmol) 4-(5-胺基-6-乙氧基-2H-吲唑-2-基)-2-甲基丁-2-醇、87 mg (0.45 mmol) 6-(三氟甲基)吡啶-2-甲酸、63 µl ( 0.45 mmol)三乙胺及174 mg (0.45 mmol) HATU在RT下攪拌過夜。藉由製備型HPLC純化得到100 mg(75%)目標化合物。 LC-MS (方法A1):Rt = 1.25 min (UV檢測器:TIC Smooth),質量測定值436.00 1H-NMR (400MHz, DMSO-d6): δ = 1.15 (s, 6 H), 1.51 (t, 3 H) 1.98 – 2.06 (m, 2 H) 4.16 – 4.25 (q, 2 H), 4.38 - 4.46 (m, 2 H), 4.53 (s, 1 H), 7.12 (s, 1 H), 8.22 (d, 1 H) 8.32 (s, 1 H) 8.37 – 8.49 (m, 2 H), 8.70 (s, 1 H), 10.74 (s, 1 H)。實例 12 N-[2-(3- 羥基 -3- 甲基丁基 )-6-( 氧雜環丁 -3- 基氧基 )-2H- 吲唑 -5- ]-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 將150 mg (515 µmol) 4-[5-胺基-6-(氧雜環丁-3-基氧基)-2H-吲唑-2-基]-2-甲基丁-2-醇、118 mg (618 µmol) 6-(三氟甲基)吡啶-2-甲酸、198 mg (618 µmol) 2-(1H-苯并三唑-1-基)-1,1,3,3-四甲基脲鎓四氟硼酸鹽及110 µl (620 µmol)N ,N -二異丙基乙胺溶解於3 ml THF中。將混合物於25℃下攪拌16小時。用水稀釋反應混合物並用乙酸乙酯萃取兩次。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。藉由製備型HPLC純化殘餘物。將合併之產物流份凍乾。此得到117 mg標題化合物。 LC-MS (方法A1):Rt = 0.90 min (UV檢測器:TIC Smooth),質量測定值464.001 H-NMR (400MHz, DMSO-d6): δ = 1.15 (s, 6H), 1.97- 2.06 (m, 2H), 4.38 - 4.46 (m, 2H), 4.52 (s, 1H), 4.66 (dd, 2H), 5.10 (t, 2H), 5.50 (五重峰, 1H), 6.82 (s, 1H), 8.23 (dd, 1H), 8.34 (s, 1H), 8.39 - 8.45 (m, 1H), 8.46 - 8.50 (m, 1H), 8.73 (s, 1H), 10.70 (s, 1H)。實例 13 N-[2-(3- 羥基 -3- 甲基丁基 )-6-( 氧雜環丁 -3- 基氧基 )-2H- 吲唑 -5- ]-2-( 三氟甲基 )-1,3- 噻唑 -4- 甲醯胺 將80.0 mg (275 µmol) 4-[5-胺基-6-(氧雜環丁-3-基氧基)-2H-吲唑-2-基]-2-甲基丁-2-醇、65.0 mg (329 µmol) 2-(三氟甲基)-1,3-噻唑-4-甲酸、125 mg (329 µmol) HATU及57 µl (330 µmol)N ,N -二異丙基乙胺溶解於2 ml THF中。將混合物於25℃下攪拌16.5小時。用水稀釋反應混合物並用乙酸乙酯萃取兩次。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。藉由製備型純化殘餘物。將合併之產物流份凍乾。此得到64.1 mg標題化合物。 LC-MS (方法A3):Rt = 0.90 min (UV檢測器:TIC Smooth),質量測定值470.001 H-NMR (500MHz, DMSO-d6): δ = 1.14 (s, 6H), 1.97 - 2.04 (m, 2H), 4.37 - 4.46 (m, 2H), 4.51 (s, 1H), 4.61 (dd, 2H), 5.07 (t, 2H), 5.44 - 5.51 (m, 1H), 6.80 (s, 1H), 8.33 (s, 1H), 8.56 (s, 1H), 8.88 (s, 1H), 9.96 (s, 1H)。實例 14 N-[2-(3- 羥基 -3- 甲基丁基 )-6-( 氧雜環丁 -3- 基氧基 )-2H- 吲唑 -5- ]-4-( 三氟甲基 )-1,3- 噻唑 -2- 甲醯胺 將80.0 mg (275 µmol) 4-[5-胺基-6-(氧雜環丁-3-基氧基)-2H-吲唑-2-基]-2-甲基丁-2-醇、65.0 mg (329 µmol) 4-(三氟甲基)-1,3-噻唑-2-甲酸、125 mg (329 µmol) HATU及57 µl (330 µmol)N ,N -二異丙基乙胺溶解於2 ml THF中。將混合物於25℃下攪拌16.5小時。用水稀釋反應混合物並用乙酸乙酯萃取兩次。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。藉由製備型HPLC純化殘餘物。將合併之產物流份凍乾。此得到61 mg標題化合物。 LC-MS (方法A3):Rt = 0.95 min (UV檢測器:TIC Smooth),質量測定值470.001 H-NMR (500MHz, DMSO-d6): δ = 1.14 (s, 6H), 1.97 - 2.04 (m, 2H), 4.38 - 4.45 (m, 2H), 4.51 (s, 1H), 4.60 (dd, 2H), 5.06 (t, 2H), 5.46 (五重峰, 1H), 6.81 (s, 1H), 8.35 (s, 1H), 8.41 (s, 1H), 8.89 (s, 1H), 9.96 (s, 1H)。實例 15 1-( 二氟甲基 )-N-[2-(3- 羥基 -3- 甲基丁基 )-6-( 氧雜環丁 -3- 基甲氧基 )-2H- 吲唑 -5- ]-1H- 吡唑 -3- 甲醯胺 將113 mg (63%純, 233 µmol) 4-[5-胺基-6-(氧雜環丁-3-基甲氧基)-2H-吲唑-2-基]-2-甲基丁-2-醇、45.3 mg (280 µmol) 1-(二氟甲基)-1H-吡唑-3-甲酸、89.8 mg (280 µmol) 2-(1H-苯并三唑-1-基)-1,1,3,3-四甲基脲鎓四氟硼酸鹽及49 µl (280 µmol)N ,N -二異丙基乙胺溶解於2 ml THF中。將混合物在25℃下攪拌67.5小時。用水稀釋反應混合物並用乙酸乙酯萃取三次。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。藉由製備型HPLC純化殘餘物。將合併之產物流份凍乾。此得到66.5 mg標題化合物。 LC-MS (方法A3):Rt = 0.95 min (UV檢測器:TIC Smooth),質量測定值449.001 H NMR (400MHz, DMSO-d6): δ = 1.15 (s, 6H), 1.97 - 2.07 (m, 2H), 3.46 - 3.66 (m, 1H), 4.33 - 4.47 (m, 4H), 4.53 (s, 1H), 4.58 (t, 2H), 4.78 (dd, 2H), 7.02 (d, 1H), 7.17 (s, 1H), 7.90 (t, 1H), 8.31 (s, 1H), 8.45 (d, 1H), 8.50 (s, 1H), 9.58 (s, 1H)。實例 16 6-( 二氟甲基 )-N-[2-(3- 羥基 -3- 甲基丁基 )-6-( 氧雜環丁 -3- 基甲氧基 )-2H- 吲唑 -5- ] 吡啶 -2- 甲醯胺 將113 mg (63%純, 233 µmol) 4-[5-胺基-6-(氧雜環丁-3-基甲氧基)-2H-吲唑-2-基]-2-甲基丁-2-醇、48.4 mg (280 µmol) 6-(二氟甲基)-吡啶-2-甲酸、89.8 mg (280 µmol) 2-(1H-苯并三唑-1-基)-1,1,3,3-四甲基脲鎓四氟硼酸鹽及49 µl (280 µmol)N ,N -二異丙基乙胺溶解於2 ml THF中。將混合物在25℃下攪拌67.5小時。用水稀釋反應混合物並用乙酸乙酯萃取三次。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。藉由製備型HPLC純化殘餘物。將合併之產物流份凍乾。此得到66.0 mg標題化合物。 LC-MS (方法A3):Rt = 1.07 min (UV檢測器:TIC Smooth),質量測定值460.001 H NMR (400MHz, DMSO-d6): δ = 1.15 (s, 6H), 1.98 - 2.07 (m, 2H), 3.53 - 3.66 (m, 1H), 4.37 (d, 2H), 4.39 - 4.48 (m, 2H), 4.53 (s, 1H), 4.65 (t, 2H), 4.82 (dd, 2H), 7.12 (t, 1H), 7.19 (s, 1H), 8.00 (dd, 1H), 8.26 - 8.40 (m, 3H), 8.76 (s, 1H), 10.65 (s, 1H)。實例 17 N-{2-(3- 羥基 -3- 甲基丁基 )-6-[(3S)- 四氫呋喃 -3- 基氧基 ]-2H- 吲唑 -5- }-2- 甲基 -1,3- 噁唑 -5- 甲醯胺 將116 mg (379 µmol) 4-{5-胺基-6-[(3S)-四氫呋喃-3-基氧基]-2H-吲唑-2-基}-2-甲基丁-2-醇、57.8 mg (455 µmol) 2-甲基-1,3-噁唑-5-甲酸、173 mg (455 µmol) HATU及79 µl (460 µmol)N ,N -二異丙基乙胺溶解於2 ml中。將混合物於25℃下攪拌16.5小時。用水稀釋反應混合物並用乙酸乙酯萃取兩次。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。將殘餘物吸收於2 ml DMF中並藉由製備型HPLC純化。將合併之產物流份凍乾。此得到96.9 mg標題化合物。 LC-MS (方法A3):Rt = 0.93 min (UV檢測器:TIC Smooth),質量測定值414.001 H-NMR (400MHz, DMSO-d6): δ = 1.15 (s, 6H), 1.98 - 2.05 (m, 2H), 2.06 - 2.17 (m, 1H), 2.29 - 2.41 (m, 1H), 3.34 (s, 3 H), 3.79 - 3.92 (m, 2H), 3.95 - 4.04 (m, 2H), 4.38 - 4.46 (m, 2H), 4.52 (s, 1H), 5.23 (dd, 1H), 7.12 (s, 1H), 8.30 (s, 1H), 8.56 (s, 1H), 8.68 (s, 1H), 9.47 (s, 1H)。實例 18 N-{2-(3- 羥基 -3- 甲基丁基 )-6-[(3S)- 四氫呋喃 -3- 基氧基 ]-2H- 吲唑 -5- }-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 將116 mg (379 µmol) 4-{5-胺基-6-[(3S)-四氫呋喃-3-基氧基]-2H-吲唑-2-基}-2-甲基丁-2-醇、87.0 mg (455 µmol) 6-(三氟甲基)吡啶-2-甲酸、173 mg (455 µmol) HATU及79 µl (460 µmol)N ,N -二異丙基乙胺溶解於2 ml中。將混合物在25℃下攪拌16.5小時。用水稀釋反應混合物並用乙酸乙酯萃取三次。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。將殘餘物吸收於2 ml DMF中並藉由製備型HPLC純化。將合併之產物流份凍乾。此得到105 mg標題化合物。 LC-MS (方法A3):Rt = 1.14 min (UV檢測器:TIC Smooth),質量測定值478.001 H-NMR (400MHz, DMSO-d6): δ = 1.16 (s, 6H), 1.97 - 2.06 (m, 2H), 2.10 - 2.21 (m, 1H), 2.31 - 2.43 (m, 1H), 3.83 - 3.98 (m, 3H), 4.06 (dd, 1H), 4.39 - 4.46 (m, 2H), 4.54 (s, 1H), 7.12 (s, 1H), 8.22 (dd, 1H), 8.33 (s, 1H), 8.38 - 8.49 (m, 2H), 8.73 (s, 1H), 10.63 (s, 1H)。實例 19 6- 胺基 -N-{2-(3- 羥基 -3- 甲基丁基 )-6-[(3S)- 四氫呋喃 -3- 基氧基 ]-2H- 吲唑 -5- } 吡啶 -2- 甲醯胺 將116 mg (379 µmol) 4-{5-胺基-6-[(3S)-四氫呋喃-3-基氧基]-2H-吲唑-2-基}-2-甲基丁-2-醇、62.9 mg (455 µmol) 6-胺基吡啶-2-甲酸、173 mg (455 µmol) HATU及79 µl (460 µmol)N ,N -二異丙基乙胺溶解於2 ml中。將混合物在25℃下攪拌16.5小時。用水稀釋反應混合物並用乙酸乙酯萃取三次。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。藉由製備型HPLC純化殘餘物。將合併之產物流份凍乾。此得到18.9 mg標題化合物。 LC-MS (方法A3):Rt = 0.89 min (UV檢測器:TIC Smooth),質量測定值425.001 H-NMR (400MHz, DMSO-d6): δ = 1.15 (s, 6H), 1.97 - 2.05 (m, 2H), 2.18 - 2.40 (m, 2H), 3.83 (td, 1H), 3.96 - 4.07 (m, 3H), 4.36 - 4.47 (m, 2H), 4.53 (s, 1H), 5.25 (br d, 1H), 6.17 (s, 2H), 6.73 (dd, 1H), 7.12 (s, 1H), 7.31 (dd, 1H), 7.63 (dd, 1H), 8.29 (s, 1H), 8.66 (s, 1H), 10.61 (s, 1H)。實例 20 N-[6- -2-(3- 甲氧基 -3- 甲基丁基 )-2H- 吲唑 -5- ]-6-(2- 羥基丙 -2- ) 吡啶 -2- 甲醯胺 將150 mg (20%純, 112 µmol) 6-氯-2-(3-甲氧基-3-甲基丁基)-2H-吲唑-5-胺、36.9 mg (168 µmol) 6-(2-羥基丙-2-基)吡啶-2-甲酸鉀(參見WO2015091426)、46.9 mg (123 µmol) HATU及21 µl (120 µmol)N ,N -二異丙基乙胺溶解於2 ml DMF中。將混合物於25℃下攪拌17.5小時。用水稀釋反應混合物並用乙酸乙酯萃取兩次。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。藉由製備型HPLC純化殘餘物。將合併之產物流份凍乾。此得到33.5 mg標題化合物。 LC-MS (方法A3):Rt = 1.22 min (UV檢測器:TIC Smooth),質量測定值430.001 H-NMR (400MHz, DMSO-d6): δ = 1.17 (s, 6H), 1.57 (s, 6H), 2.09 - 2.17 (m, 2H), 3.14 (s, 3H), 4.41 - 4.50 (m, 2H), 5.49 (s, 1H), 7.92 - 7.98 (m, 2H), 8.01 - 8.12 (m, 2H), 8.53 (d, 1H), 8.71 (s, 1H), 10.88 (s, 1H)。實例 21 N-[6- -2-(3- 甲氧基 -3- 甲基丁基 )-2H- 吲唑 -5- ]-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 將150 mg (20%純, 112 µmol) 6-氯-2-(3-甲氧基-3-甲基丁基)-2H-吲唑-5-胺、32.1 mg (168 µmol) 6-(三氟甲基)吡啶-2-甲酸、46.9 mg (123 µmol) HATU及21 µl (120 µmol)N ,N -二異丙基乙胺溶解於2 ml DMF中。將混合物於25℃下攪拌17.5小時。用水稀釋反應混合物並用乙酸乙酯萃取兩次。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。藉由製備型HPLC純化殘餘物。將合併之產物流份凍乾。此得到48 mg標題化合物。 LC-MS (方法A3):Rt = 1.43 min (UV檢測器:TIC Smooth),質量測定值440.00 1H-NMR (400MHz, DMSO-d6): δ = 1.17 (s, 6H), 2.10 - 2.17 (m, 2H), 3.14 (s, 3H), 4.43 - 4.50 (m, 2H), 7.93 (s, 1H), 8.24 (dd, 1H), 8.39 - 8.45 (m, 1H), 8.45 - 8.50 (m, 1H), 8.54 (s, 1H), 8.63 (s, 1H), 10.52 (s, 1H)。實例 22 N-[6- 異丙氧基 -2-(2- 甲氧基乙基 )-2H- 吲唑 -5- ]-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 將150 mg (0.41 mmol) N-(6-異丙氧基-1H-吲唑-5-基)-6-(三氟甲基)吡啶-2-甲醯胺、171 mg (1.24 mmol)碳酸鉀及34 mg (0.21 mmol)碘化鉀懸浮於2.0 ml DMF中,並將混合物在25℃下攪拌30分鐘。隨後,添加58 µl (0.62 mmol) 2-溴乙基甲醚,並將混合物在25℃下攪拌16小時並在80℃下攪拌48小時。將反應混合物分配於乙酸乙酯與水之間。分離各相。用飽和氯化鈉溶液將有機相洗滌兩次,藉助疏水過濾器過濾並濃縮。藉由製備型HPLC純化殘餘物。凍乾產物流份。此得到41 mg標題化合物。 LC-MS (方法A1):Rt = 1.31 min (UV檢測器:TIC Smooth),質量測定值422.00 1H NMR (400 MHz, DMSO-d6): δ = 1.41 (d, 6 H), 3.31 (s, 3 H), 3.81 (t, 2 H), 4.50 (t, 2 H), 4.84 (dt, 1 H), 7.19 (s, 1 H), 8.21 (dd, 1 H), 8.28 (s, 1 H), 8.37 - 8.43 (m, 1 H), 8.43 - 8.48 (m, 1 H), 8.72 (s, 1 H), 10.73 (s, 1 H)。實例 23 N-[6- 異丙氧基 -2-(3- 甲氧基丙基 )-2H- 吲唑 -5- ]-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 將150 mg (0.41 mmol) N-(6-異丙氧基-1H-吲唑-5-基)-6-(三氟甲基)吡啶-2-甲醯胺、170 mg (1.24 mmol)碳酸鉀及34 mg (0.21 mmol)碘化鉀懸浮於2.0 ml DMF中,並將混合物在25℃下攪拌30分鐘。隨後,添加69 µl (0.62 mmol) 1-溴-3-甲氧基丙烷,並將混合物在25℃下攪拌16小時並在80℃下攪拌24小時。將反應混合物分配於乙酸乙酯與水之間。分離各相。用飽和氯化鈉溶液將有機相洗滌兩次,藉助疏水過濾器過濾並濃縮。藉由製備型HPLC純化殘餘物。凍乾產物流份。此得到45 mg標題化合物。 LC-MS (方法A1):Rt = 1.35 min (UV檢測器:TIC Smooth),質量測定值436.00 1H NMR (400 MHz, DMSO-d6): δ = 1.41 (d, 6 H), 2.12 (五重峰, 2 H), 3.24 (s, 3 H), 3.32 - 3.35 (m, 2 H), 4.39 (t, 2 H), 4.84 (五重峰, 1 H), 7.19 (s, 1 H), 8.21 (dd, 1 H), 8.28 (s, 1 H), 8.40 (t, 1 H), 8.46 (d, 1 H), 8.72 (s, 1 H), 10.73 (s, 1 H)。實例 24 6-( 二氟甲基 )-N-[6- 乙氧基 -2-(3- 羥基 -3- 甲基丁基 )-2H- 吲唑 -5- ] 吡啶 -2- 甲醯胺 將150 mg (89%純, 402 µmol) 6-(二氟甲基)-N-(6-乙氧基-1H-吲唑-5-基)吡啶-2-甲醯胺溶解於4 ml DMF中,並在攪拌下添加101 mg (603 µmol) 4-溴-2-甲基丁-2-醇、167 mg (1.21 mmol)碳酸鉀及100 mg (603 µmol)碘化鉀。將懸浮液在120℃下攪拌6小時。用水稀釋反應混合物並用乙酸乙酯萃取三次。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。藉由製備型HPLC純化殘餘物。將合併之產物流份凍乾。此得到36.6 mg標題化合物。 LC-MS (方法A1):Rt = 1.19 min (UV檢測器:TIC Smooth),質量測定值418.001 H-NMR(400MHz, DMSO-d 6 ): δ = 1.15 (s, 6H), 1.52 (t, 3H), 1.98 - 2.06 (m, 2H), 4.21 (q, 2H), 4.38 - 4.46 (m, 2H), 4.50 (s, 1H), 6.93 - 7.27 (m, 2H), 7.98 (dd, 1H), 8.27 - 8.36 (m, 3H), 8.69 (s, 1H), 10.78 (s, 1H)。實例 25 N-[6-( 環丙基甲氧基 )-2-(3- 甲氧基丙基 )-2H- 吲唑 -5- ]-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 將200 mg (0.53 mmol) N-[6-(環丙基甲氧基)-1H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺、220 mg (1.59 mmol)碳酸鉀及一刮勺碘化鉀懸浮於3.0 ml DMF中,並將混合物在25℃下攪拌30分鐘。隨後,添加89 µl (0.80 mmol) 1-溴-3-甲氧基丙烷,並將混合物在25℃下攪拌72小時。將反應混合物分配於乙酸乙酯與水之間。分離各相。用飽和氯化鈉溶液將有機相洗滌兩次,藉助疏水過濾器過濾並濃縮。藉由製備型HPLC純化殘餘物。凍乾產物流份。此得到25 mg標題化合物。 LC-MS (方法A1):Rt = 1.37 min (UV檢測器:TIC Smooth),質量測定值448.001 H-NMR (400 MHz, DMSO-d6): δ = 0.41 - 0.47 (m, 2 H), 0.60 - 0.69 (m, 2 H), 1.30 - 1.40 (m, 1 H), 2.12 (五重峰, 2 H), 3.24 (s, 3 H), 3.30 (t, 2 H), 4.02 (d, 2 H), 4.38 (t, 2 H), 7.09 (s, 1 H), 8.21 (dd, 1 H), 8.28 (s, 1 H), 8.41 (t, 1 H), 8.47 (d, 1 H), 8.74 (s, 1 H), 10.69 (s, 1 H)。實例 26 N-[6-( 環丙基甲氧基 )-2-(2- 甲氧基乙基 )-2H- 吲唑 -5- ]-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 將280 mg (0.74 mmol) N-[6-(環丙基甲氧基)-1H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺、308 mg (2.23 mmol)碳酸鉀及148 mg (0.89 mmol)碘化鉀懸浮於5.0 ml DMF中,並將混合物在25℃下攪拌30分鐘。隨後,添加105 µl (1.12 mmol) 2-溴甲基甲醚,並將混合物在25℃下攪拌72小時。將反應混合物分配於乙酸乙酯與水之間。分離各相。用飽和氯化鈉溶液將有機相洗滌兩次,藉助疏水過濾器過濾並濃縮。藉由製備型HPLC純化殘餘物。凍乾產物流份。此得到50 mg標題化合物。 LC-MS (方法A1):Rt = 1.33 min (UV檢測器:TIC Smooth),質量測定值434.00 1H NMR (400 MHz, DMSO-d6): δ = 0.39 - 0.47 (m, 2 H), 0.63 - 0.69 (m, 2 H), 1.29 - 1.40 (m, 1 H), 3.23 (s, 3 H), 3.80 (t, 2 H), 4.02 (d, 2 H), 4.49 (t, 2 H), 7.08 (s, 1 H), 8.21 (dd, 1 H), 8.26 - 8.29 (m, 1 H), 8.38 - 8.44 (m, 1 H), 8.45 - 8.50 (m, 1 H), 8.74 (s, 1 H), 10.70 (s, 1 H)。實例 27 N-[6-( 環丙基甲氧基 )-2-( 氧雜環丁 -3- 基甲基 )-2H- 吲唑 -5- ]-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 將200 mg (0.53 mmol) N-[6-(環丙基甲氧基)-1H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺、220 mg (1.59 mmol)碳酸鉀及一刮勺碘化鉀懸浮於3.0 ml DMF中,並將混合物在25℃下攪拌30分鐘。隨後,添加120 mg (0.80 mmol) 3-溴甲基氧雜環丁烷,並將混合物在25℃下攪拌72小時。將反應混合物分配於乙酸乙酯與水之間。分離各相。用飽和氯化鈉溶液將有機相洗滌兩次,藉助疏水過濾器過濾並濃縮。藉由製備型HPLC根據方法P4來純化殘餘物。凍乾產物流份。藉由製備型HPLC使用方法P2 (管柱:YMC Triart C18 5µm 100×30 mm, 梯度:0–0.5 min 25 ml/min至70 ml/min 52% B;0.5-5.5 min 52-61% B;流速:70 ml/min)再純化凍乾物。凍乾產物流份。此得到14 mg標題化合物。 LC-MS (方法A1):Rt =1.28 min (UV檢測器:TIC Smooth),質量測定值446.00 1H NMR (400 MHz, DMSO-d6): δ = 0.38 - 0.47 (m, 2 H), 0.59 - 0.70 (m, 2 H), 1.29 - 1.40 (m, 1 H), 3.44 - 3.58 (m, 1 H), 4.02 (d, 2 H), 4.47 (t, 2 H), 4.62 - 4.70 (m, 4 H), 7.08 (s, 1 H), 8.18 - 8.24 (m, 1 H), 8.33 (s, 1 H), 8.37 - 8.45 (m, 1 H), 8.45 - 8.50 (m, 1 H), 8.73 (s, 1 H), 10.69 (s, 1 H)。實例 28 N-[2-(3- 羥基 -3- 甲基丁基 )-6-( 三氟甲氧基 )-2H- 吲唑 -5- ]-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 將406 mg (94%純, 978 µmol) N-[6-(三氟甲氧基)-1H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺溶解於5 ml DMF中,並在攪拌下添加245 mg (1.47 mmol) 4-溴-2-甲基丁-2-醇、405 mg (2.93 mmol)碳酸鉀及244 mg (1.47 mmol)碘化鉀。將懸浮液在120℃下攪拌19小時。用水稀釋反應混合物並用乙酸乙酯萃取三次。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。藉由製備型HPLC純化殘餘物。將合併之產物流份凍乾。此得到19 mg標題化合物。 LC-MS (方法A1):Rt = 1.33 min (UV檢測器:TIC Smooth),質量測定值476.001 H-NMR (400MHz, DMSO-d6): δ = 1.16 (s, 6H), 2.01 - 2.10 (m, 2H), 4.48 - 4.58 (m, 3H), 7.77 (s, 1H), 8.23 (dd, 1H), 8.39 - 8.43 (m, 1H), 8.46 (t, 1H), 8.56 (s, 1H), 8.69 (s, 1H), 10.38 (s, 1H)。實例 29 2- 環丙基 -N-[6- 甲氧基 -2-(3- 甲氧基丙基 )-2H- 吲唑 -5- ]-1,3- 噁唑 -4- 甲醯胺 將378 mg (0.63 mmol) 2-環丙基-N-(6-甲氧基-1H-吲唑-5-基)-1,3-噁唑-4-甲醯胺溶解於4 ml DMF中,並在攪拌下添加108 µl (0.95 mmol) 1-溴-3-甲氧基丙烷、263 mg (1.90 mmol)碳酸鉀及158 mg (0.95 mmol)碘化鉀。將反應混合物在100℃下攪拌23小時。隨後,用水稀釋混合物並用乙酸乙酯萃取。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。藉由製備型HPLC純化殘餘物。冷凍乾燥含有產物之流份。此得到36 mg標題化合物。 LC-MS (方法A4):Rt = 1.12 min (UV檢測器:TIC Smooth),質量測定值370.001 H-NMR (400MHz, DMSO-d6): δ = 1.01 - 1.07 (m, 2H), 1.09 - 1.16 (m, 2H), 2.06 - 2.16 (m, 2H), 2.18 - 2.27 (m, 1H), 3.23 (s, 3H), 3.27 - 3.30 (m, 2H), 3.97 (s, 3H), 4.38 (t, 2H), 7.11 (s, 1H), 8.24 (s, 1H), 8.54 (s, 1H), 8.60 (s, 1H), 9.34 (s, 1H)。實例 30 N-[6- -2-(3- 羥基 -3- 甲基丁基 )-2H- 吲唑 -5- ]-6-( 二氟甲基 ) 吡啶 -2- 甲醯胺 將3.15 g (51%純, 4.98 mmol) N-(6-氯-1H-吲唑-5-基)-6-(二氟甲基)吡啶-2-甲醯胺溶解於20 ml DMF中,並在攪拌下添加1.25 g (7.47 mmol) 4-溴-2-甲基丁-2-醇、2.06 g (14.9 mmol)碳酸鉀及1.24 g (7.47 mmol)碘化鉀。將懸浮液在120℃下攪拌16.5小時。添加另一1.03 mg (7.5 mmol)碳酸鉀及620 mg (3.8 mmol)碘化鉀,並將混合物在120℃下再攪拌24小時。用水稀釋反應混合物並用乙酸乙酯萃取三次。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。藉由急速層析(Biotage Interchim 15 µm筒柱(80 g;KP-Sil), 移動相:己烷/乙酸乙酯)純化殘餘物。濃縮合併之產物流份。利用抽吸濾出固體,用乙醚洗滌三次並乾燥。濃縮濾液並藉由製備型純化殘餘物。將合併之產物流份凍乾。此得到總共314 mg標題化合物。 LC-MS (方法A1):Rt = 1.17 min (UV檢測器:TIC Smooth),質量測定值408.001 H-NMR (400MHz, DMSO-d6): δ = 1.16 (s, 6H), 1.99 - 2.13 (m, 2H), 4.45 - 4.58 (m, 3H), 7.14 (t, 1H), 7.92 (s, 1H), 8.03 (dd, 1H), 8.29 - 8.40 (m, 2H), 8.52 (s, 1H), 8.63 (s, 1H), 10.59 (s, 1H)。實例 31 N-[2-(2- 羥基乙基 )-6- 異丙氧基 -2H- 吲唑 -5- ]-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 步驟A:將200 mg (0.55 mmol) N-(6-異丙氧基-1H-吲唑-5-基)-6-(三氟甲基)吡啶-2-甲醯胺溶解於2 ml DMF中,並在攪拌下添加228 mg (1.65 mmol)碳酸鉀及46 mg (0.27 mmol)碘化鉀。將懸浮液在25℃下攪拌30分鐘,且然後添加177 µl (0.82 mmol) (2-溴乙氧基)(第三丁基)二甲基矽烷。將反應混合物在25℃下攪拌16小時並在80℃下攪拌4小時。然後用水稀釋反應混合物並用乙酸乙酯萃取兩次。藉助疏水過濾器過濾合併之有機相並濃縮。藉由急速層析(Biotage SNAP筒柱(50 g;KP-Sil), 移動相:己烷/乙酸乙酯)純化殘餘物。此得到56 mg N-[2-(2-{[第三丁基(二甲基)矽基]氧基}乙基)-6-異丙氧基-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺。 LC-MS (方法A1):Rt = 1.71 min (UV檢測器:TIC Smooth),質量測定值522.00 1H NMR (400 MHz, DMSO-d6): δ = -0.13 - -0.10 (m, 6 H), 0.76 - 0.79 (m, 9 H), 1.41 (d, 6 H), 4.03 (t, 2 H), 4.42 (t, 2 H), 4.84 (dt, 1 H), 7.19 (s, 1 H), 8.21 (dd, 1 H), 8.25 (s, 1 H), 8.40 (t, 1 H), 8.46 (d, 1 H), 8.72 (s, 1 H), 10.73 (s, 1 H)。 步驟B:將53 mg (0.10 mmol) N-[2-(2-{[第三丁基(二甲基)矽基]氧基}乙基)-6-異丙氧基-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺溶解於0.5 ml THF中並與304 µl (0.30 mmol)四丁基氟化銨於THF中之1 M溶液混合。將反應混合物在25℃下攪拌2小時並將其添加至10 ml水中,並利用抽吸濾出所形成之沈澱物,用水及乙醚洗滌並在減壓下乾燥。此得到29 mg N-[2-(2-羥基乙基)-6-異丙氧基-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺。 LC-MS (方法A1):Rt = 1.19 min (UV檢測器:TIC Smooth),質量測定值408.00 1H NMR (400 MHz, DMSO-d6): δ = 1.41 (d, 6 H), 3.85 (q, 2 H), 4.38 (t, 2 H), 4.84 (dt, 1 H), 4.94 (t, 1 H), 7.18 (s, 1 H), 8.21 (dd, 1 H), 8.27 (s, 1 H), 8.40 (t, 1 H), 8.46 (d, 1 H), 8.73 (s, 1 H), 10.73 (s, 1 H)。實例 32 N-[2-(3- 羥基丙基 )-6- 異丙氧基 -2H- 吲唑 -5- ]-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 步驟A:將200 mg (0.55 mmol) N-(6-異丙氧基-1H-吲唑-5-基)-6-(三氟甲基)吡啶-2-甲醯胺溶解於2 ml DMF中,並在攪拌下添加228 mg (1.65 mmol)碳酸鉀及46 mg (0.27 mmol)碘化鉀。將懸浮液在25℃下攪拌30分鐘,且然後添加191 µl (0.82 mmol) (3-溴丙氧基)(第三丁基)二甲基矽烷。將反應混合物在25℃下攪拌16小時並在80℃下攪拌48小時。然後用水稀釋反應混合物並用乙酸乙酯萃取兩次。藉助疏水過濾器過濾合併之有機相並濃縮。藉由急速層析(Biotage SNAP筒柱(50 g;KP-Sil), 移動相:己烷/乙酸乙酯)純化殘餘物。藉由製備型HPLC純化所獲得之粗製產物。此得到59 mg N-[2-(3-{[第三丁基(二甲基)矽基]氧基}丙基)-6-異丙氧基-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺。 LC-MS (方法A1):Rt = 1.75 min (UV檢測器:TIC Smooth),質量測定值536.00 1H NMR (400 MHz, DMSO-d6): δ = 0.03 (s, 6 H), 0.87 (s, 9 H), 1.41 (d, 6 H), 2.09 (五重峰, 2 H), 3.60 (t, 2 H), 4.40 (t, 2 H), 4.84 (五重峰, 1 H), 7.18 (s, 1 H), 8.21 (dd, 1 H), 8.26 (s, 1 H), 8.40 (t, 1 H), 8.46 (d, 1 H), 8.72 (s, 1 H), 10.73 (s, 1 H)。 步驟B:將59 mg (0.11 mmol) N-[2-(3-{[第三丁基(二甲基)矽基]氧基}丙基)-6-異丙氧基-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺溶解於0.5 ml THF中並與330 µl (0.33 mmol)四丁基氟化銨於THF中之1 M溶液混合。將反應混合物在25℃下攪拌90分鐘並將其添加至10 ml水中,並利用抽吸濾出所形成之沈澱物,用水及乙醚洗滌並在減壓下乾燥。此得到15 mg N-[2-(3-羥基丙基)-6-異丙氧基-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺。 1H NMR (400 MHz, DMSO-d6): δ = 1.41 (d, 6 H), 1.99 - 2.09 (m, 2 H), 3.41 (q, 2 H), 4.40 (t, 2 H), 4.62 (t, 1 H), 4.79 - 4.88 (m, 1 H), 7.18 (s, 1 H), 8.21 (dd, 1 H), 8.28 (s, 1 H), 8.40 (t, 1 H), 8.46 (d, 1 H), 8.72 (s, 1 H), 10.73 (s, 1 H)。實例 33 N-2-(3- 羥基 -3- 甲基丁基 )-6-[3-( 甲基磺醯基 ) 丙氧基 ]-2H- 吲唑 -5- -6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 將50 mg (0.12 mmol) N-[6-羥基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺溶解於735 µl DMF中,並在攪拌下添加33 mg (0.24 mmol)碳酸鉀及29 mg (0.15 mmol) 1-溴-3-(甲基磺醯基)丙烷。將反應混合物在微波中在100℃下攪拌60分鐘。添加水,並用乙酸乙酯萃取混合物。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。將殘餘物溶解於2 ml乙腈中並藉由製備型HPLC純化。凍乾產物流份。此得到46 mg標題化合物。 LC-MS (方法A1):Rt = 1.04 min (UV檢測器:TIC Smooth),質量測定值528.00 1H NMR (300 MHz, DMSO-d6): δ = -1.15 (s, 6 H), 1.95 - 2.07 (m, 2 H), 2.23 - 2.37 (m, 2 H), 3.01 (s, 3 H), 3.34 - 3.40 (m, 2 H), 4.30 (t, 2 H), 4.37 - 4.46 (m, 2 H), 4.51 (s, 1 H), 7.13 (s, 1 H), 8.22 (d, 1 H), 8.33 (s, 1 H), 8.41 (t, 1 H), 8.47 (d, 1 H), 8.71 (s, 1 H), 10.55 (s, 1 H)。實例 34 N-{2-(3- 羥基 -3- 甲基丁基 )-6-[2-( 甲基硫基 ) 乙氧基 ]-2H- 吲唑 -5- }-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 將50 mg (0.12 mmol) N-[6-羥基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺溶解於1 ml DMF中,並在攪拌下添加33 mg (0.24 mmol)碳酸鉀及23 mg (0.15 mmol) 2-溴乙基甲基硫化物。將反應混合物在微波中在100℃下攪拌60分鐘。添加水,並用乙酸乙酯萃取混合物。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。藉由製備型HPLC純化殘餘物。凍乾產物流份。此得到32 mg標題化合物。 LC-MS (方法A1):Rt = 1.26 min (UV檢測器:TIC Smooth),質量測定值482.00 1H NMR (400 MHz, DMSO-d6): δ = 1.15 (s, 6 H), 1.97 - 2.06 (m, 2 H), 2.15 (s, 3 H), 2.98 (t, 2 H), 4.35 (t, 2 H), 4.39 - 4.46 (m, 2 H), 4.51 (s, 1 H), 7.19 (s, 1 H), 8.22 (dd, 1 H), 8.32 (s, 1 H), 8.40 (t, 1 H), 8.47 (d, 1 H), 8.71 (s, 1 H), 10.60 (s, 1 H)。實例 35 {[2-(3- 羥基 -3- 甲基丁基 )-5-({[6-( 三氟甲基 ) 吡啶 -2- ] 羰基 } 胺基 )-2H- 吲唑 -6- ] 氧基 } 乙酸乙酯 將170 mg (0.42 mmol) N-[6-羥基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺溶解於2.5 ml DMF中,並在攪拌下添加115 mg (0.83 mmol)碳酸鉀及56 µl (0.50 mmol)溴乙酸乙酯。將反應混合物在微波中在100℃下攪拌60分鐘。添加水,並用乙酸乙酯萃取混合物。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。藉由製備型HPLC純化殘餘物。凍乾產物流份。此得到143 mg標題化合物。 LC-MS (方法A1):Rt = 1.19 min (UV檢測器:TIC Smooth),質量測定值494.00 1H NMR (400 MHz, DMSO-d6): δ = 1.15 (s, 6 H), 1.21 (t, 3 H), 1.95 - 2.05 (m, 2 H), 4.20 (q, 2 H), 4.39 - 4.46 (m, 2 H), 4.50 (s, 1 H), 5.00 (s, 2 H), 7.18 (s, 1 H), 8.21 (dd, 1 H), 8.34 (s, 1 H), 8.40 (t, 1 H), 8.48 (d, 1 H), 8.72 (s, 1 H), 10.54 (s, 1 H)。實例 36 N-[2-(3- 羥基 -3- 甲基丁基 )-6-( 氧雜環丁 -3- 基甲氧基 )-2H- 吲唑 -5- ]-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 將75 mg (0.18 mmol) N-[6-羥基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺溶解於1.1 ml DMF中,並在攪拌下添加50 mg (0.37 mmol)碳酸鉀及42 mg (0.28 mmol) 3-(溴甲基)氧雜環丁烷。將反應混合物在微波中在100℃下攪拌60分鐘。添加水,並用乙酸乙酯萃取混合物。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。藉由製備型HPLC純化殘餘物。凍乾產物流份。此得到50 mg標題化合物。 LC-MS (方法A1):Rt = 1.12 min (UV檢測器:TIC Smooth),質量測定值478.00 1H NMR (400 MHz, DMSO-d6): δ = 1.15 (s, 6 H), 1.98 - 2.06 (m, 2 H), 3.43 - 3.54 (m, 1 H), 4.33 - 4.56 (m), 4.79 (dd, 2 H), 7.20 (s, 1 H), 8.21 (dd, 1 H), 8.32 (s, 1 H), 8.36 - 8.44 (m, 1 H), 8.44 - 8.48 (m, 1 H), 8.72 (s, 1 H), 10.44 (s, 1 H)。實例 37 N-[6-( 環丙基甲氧基 )-2-(3- 羥基 -3- 甲基丁基 )-2H- 吲唑 -5- ]-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 將68 mg (0.17 mmol) N-[6-羥基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺溶解於1 ml DMF中,並在攪拌下添加46 mg (0.33 mmol)碳酸鉀及19 µl (0.20 mmol) (溴甲基)環丙烷。將懸浮液在微波中在100℃下攪拌60分鐘,且然後添加431 µl (4.65 mmol) (溴甲基)環丙烷。將反應混合物在25℃下攪拌16小時。將反應混合物分配於乙酸乙酯與水之間。分離各相。用飽和氯化鈉溶液將有機相洗滌兩次,藉助疏水過濾器過濾並濃縮。藉由製備型HPLC純化殘餘物。凍乾產物流份。此得到45 mg標題化合物。 LC-MS (方法A1):Rt = 1.33 min (UV檢測器:TIC Smooth),質量測定值462.00 1H NMR (400 MHz, DMSO-d6): δ = 0.39 - 0.47 (m, 2 H), 0.62 - 0.69 (m, 2 H), 1.15 (s, 6 H), 1.29 - 1.39 (m, 1 H), 1.98 - 2.04 (m, 2 H), 4.02 (d, 2 H), 4.38 - 4.45 (m, 2 H), 4.49 (s, 1 H), 7.07 (s, 1 H), 8.21 (dd, 1 H), 8.30 (s, 1 H), 8.40 (t, 1 H), 8.47 (d, 1 H), 8.73 (s, 1 H), 10.69 (s, 1 H)。實例 38 N-[6-( 環丙基甲氧基 )-2-(3- 羥基丙基 )-2H- 吲唑 -5- ]-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 步驟A:將於4.2 ml DMF中之280 mg (0.74 mmol) N-[6-(環丙基甲氧基)-1H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺、308 mg (2.23 mmol)碳酸鉀及一刮勺碘化鉀在25℃下攪拌30分鐘。隨後,添加253 µl (1.49 mmol) 2-(3-溴丙氧基)四氫-2H-吡喃,並將混合物在25℃下攪拌72小時。將反應混合物分配於乙酸乙酯與水之間。分離各相。用飽和氯化鈉溶液將有機相洗滌兩次,藉助疏水過濾器過濾並濃縮。藉由製備型HPLC純化殘餘物。此得到91 mg N-{6-(環丙基甲氧基)-2-[3-(四氫-2H-吡喃-2-基氧基)丙基]-2H-吲唑-5-基}-6-(三氟甲基)吡啶-2-甲醯胺。 LC-MS (方法A1):Rt = 1.47 min (UV檢測器:TIC Smooth),質量測定值518.00 1H NMR (400 MHz, DMSO-d6, 所選信號):δ = 0.40 - 0.48 (m, 2 H), 0.61 - 0.69 (m, 2 H), 1.30 - 1.39 (m, 1 H), 1.39 - 1.54 (m, 4 H), 1.56 - 1.67 (m, 1 H), 1.67 - 1.79 (m, 1 H), 2.15 (五重峰, 2 H), 3.36 - 3.44 (m, 1 H), 3.63 (dt, 1 H), 3.73 (ddd, 1 H), 3.98 - 4.07 (m, 2 H), 4.42 (t, 2 H), 4.53 (t, 1 H), 7.08 (s, 1 H), 8.17 - 8.24 (m, 1 H), 8.29 (s, 1 H), 8.41 (t, 1 H), 8.47 (d, 1 H), 8.74 (s, 1 H), 10.69 (s, 1 H)。 步驟B:將88 mg (0.17 mmol) N-{6-(環丙基甲氧基)-2-[3-(四氫-2H-吡喃-2-基氧基)丙基]-2H-吲唑-5-基}-6-(三氟甲基)吡啶-2-甲醯胺溶解於1.1 ml二氯甲烷中,並添加96 mg (0.51 mmol)對甲苯磺酸。將反應混合物在25℃下攪拌19小時。添加飽和碳酸氫鈉溶液並用乙酸乙酯萃取混合物。使合併之有機相通過疏水過濾器過濾並濃縮。此得到46 mg N-[6-(環丙基甲氧基)-2-(3-羥基丙基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺。 LC-MS (方法A1):Rt = 1.22 min (UV檢測器:TIC Smooth),質量測定值434.00 1H NMR (400 MHz, DMSO-d6): δ = 0.39 - 0.47 (m, 2 H), 0.62 - 0.70 (m, 2 H), 1.29 - 1.41 (m, 1 H), 2.03 (五重峰, 2 H), 3.36 - 3.42 (m, 2 H), 4.02 (d, 2 H), 4.39 (t, 2 H), 4.63 (t, 1 H), 7.08 (s, 1 H), 8.22 (dd, 1 H), 8.28 (s, 1 H), 8.41 (t, 1 H), 8.48 (d, 1 H), 8.74 (s, 1 H), 10.70 (s, 1 H)。實例 39 N-[6- 甲氧基 -2-(3- 側氧基丁基 )-2H- 吲唑 -5- ]-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 步驟A:將2.50 g (7.4 mmol) N-(6-甲氧基-1H-吲唑-5-基)-6-(三氟甲基)吡啶-2-甲醯胺(CAS編號:1799836-45-5)溶解於15 ml DMF中,並在攪拌下添加1.63 ml (11.9 mmol) 2-(2-溴乙基)-2-甲基-1,3-二氧戊環、3.08 g (22.3 mmol)碳酸鉀及1.85 g (11.2 mmol)碘化鉀。將反應混合物在100℃下攪拌21小時。隨後,用水稀釋混合物並用乙酸乙酯萃取。用飽和氯化鈉溶液洗滌合併之有機相,分離各相,藉助疏水過濾器過濾並濃縮。將乙酸乙酯添加至殘餘物中,並濾出所形成之固體。濃縮濾液並藉由急速層析(Biotage SNAP筒柱(100 g;KP-Sil), 移動相:己烷/乙酸乙酯)純化。此得到860 mg N-{6-甲氧基-2-[2-(2-甲基-1,3-二氧戊環-2-基)乙基]-2H-吲唑-5-基}-6-(三氟甲基)吡啶-2-甲醯胺。 LC-MS (方法A1):Rt = 1.20 min (UV檢測器:TIC Smooth),質量測定值450.00。 步驟B:將855 mg (1.37 mmol) N-{6-甲氧基-2-[2-(2-甲基-1,3-二氧戊環-2-基)乙基]-2H-吲唑-5-基}-6-(三氟甲基)吡啶-2-甲醯胺溶解於20 ml二氯甲烷中,添加520 mg (2.73 mmol)對甲苯磺酸一水合物並將混合物在25℃下攪拌67小時。添加另一260 mg (1.36 mmol)對甲苯磺酸一水合物,並將混合物在25℃下攪拌24小時。濾出所形成之固體並用二氯甲烷洗滌三次。添加飽和碳酸氫鈉溶液,並短暫攪拌反應混合物。分離各相,並用二氯甲烷將水相萃取兩次。用飽和氯化鈉溶液洗滌合併之有機相,過濾(疏水過濾器)並濃縮。將乙酸乙酯添加至殘餘物中,並短暫攪拌混合物。利用抽吸濾出固體,並用乙酸乙酯洗滌三次並乾燥。此得到392 mg N-[6-甲氧基-2-(3-側氧基丁基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺。 LC-MS (方法A1):Rt = 1.13 min (UV檢測器:TIC Smooth),質量測定值406.001 H NMR (400 MHz, DMSO-d6): δ = 2.14 (s, 3 H), 3.15 (t, 2 H), 3.98 (s, 3 H), 4.54 (t, 2 H), 7.13 (s, 1 H), 8.21 (dd, 1 H), 8.28 (s, 1 H), 8.40 (t, 1 H), 8.46 (d, 1 H), 8.67 (s, 1 H), 10.49 (s, 1 H)。實例 40 N-[2-(2- 羥基乙基 )-6- 甲氧基 -2H- 吲唑 -5- ]-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 步驟A:將7.00 g (20.8 mmol) N-(6-甲氧基-1H-吲唑-5-基)-6-(三氟甲基)吡啶-2-甲醯胺、6.60 ml (41.6 mmol)溴乙酸苄基酯及8.83 ml (41.6 mmol) N,N-二環己基甲胺於100 ml THF中之混合物加熱至70℃並保持5小時。添加另一3.30 ml (20.8 mmol)溴乙酸苄基酯及4.42 ml (20.8 mmol) N,N-二環己基甲胺,並將混合物在65℃下攪拌20小時。濾出沈澱固體,用乙酸乙酯洗滌兩次,用水洗滌三次並用乙醚洗滌三次並乾燥。濃縮反應混合物之濾液,添加乙酸乙酯並將混合物攪拌15分鐘。利用抽吸過濾出固體,並用乙酸乙酯洗滌兩次並乾燥。合併該兩批固體。此得到7.08 g固體狀[6-甲氧基-5-({[6-(三氟甲基)吡啶-2-基]羰基}胺基)-2H-吲唑-2-基]乙酸苄基酯。 LC-MS (方法A1):Rt = 1.37 min (UV檢測器:TIC Smooth),質量測定值484.001 H NMR (400 MHz, DMSO-d6): δ = 3.99 (s, 3 H), 5.21 (s, 2 H), 5.41 (s, 2 H), 7.14 (s, 1 H), 7.31 - 7.42 (m, 5 H), 8.22 (dd, 1 H), 8.35 (s, 1 H), 8.40 (t, 1 H), 8.47 (d, 1 H), 8.72 (s, 1 H), 10.51 (s, 1 H)。 步驟B:將7.08 g (14.6 mmol) [6-甲氧基-5-({[6-(三氟甲基)吡啶-2-基]羰基}胺基)-2H-吲唑-2-基]乙酸苄基酯溶解於110 ml THF/乙醇(10:1)中,且每次少許添加總共553 mg (14.6 mmol)硼氫化鈉。將混合物在25℃下攪拌48小時。將水添加至反應混合物中並用乙酸乙酯將水相萃取一次。利用抽吸濾出固體並用乙酸乙酯洗滌兩次。分離濾液之相並用乙酸乙酯將水相萃取兩次以上。使用飽和氯化鈉溶液洗滌合併之有機相並濃縮。合併此粗製產物及已濾出之固體,添加60 ml乙醚並將混合物攪拌20分鐘。利用抽吸濾出固體,用乙醚洗滌三次並乾燥。此得到4.56 g N-[2-(2-羥基乙基)-6-甲氧基-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺。 LC-MS (方法A1):Rt = 1.07 min (UV檢測器:TIC Smooth),質量測定值380.001 H NMR (400 MHz, DMSO-d6): δ = 3.85 (q, 2 H), 3.98 (s, 3 H), 4.38 (t, 2 H), 4.99 (t, 1 H), 7.14 (s, 1 H), 8.19 - 8.23 (m, 1 H), 8.28 (s, 1 H), 8.40 (t, 1 H), 8.46 (d, 1 H), 8.69 (s, 1 H), 10.50 (s, 1 H)。實例 41 在ExpNo UBOT8717-1, BAY 1752003下N-[2-(3- 羥基丙基 )-6- 甲氧基 -2H- 吲唑 -5- ]-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 步驟A:將200 mg (0.60 mmol) N-(6-甲氧基-1H-吲唑-5-基)-6-(三氟甲基)吡啶-2-甲醯胺(CAS編號:1799836-45-5)溶解於4 ml DMF中,並在攪拌下添加151 µl (0.89 mmol) 2-(3-溴丙氧基)四氫-2H-吡喃、247 mg (1.78 mmol)碳酸鉀及118 mg (0.71 mmol)碘化鉀。將反應混合物在100℃下攪拌19小時。用水稀釋反應混合物並用乙酸乙酯萃取三次。藉助疏水過濾器過濾合併之有機相並濃縮。將殘餘物溶解於2 ml二甲亞碸並中藉由製備型HPLC純化。此得到50 mg N-{6-甲氧基-2-[3-(四氫-2H-吡喃-2-基氧基)丙基]-2H-吲唑-5-基}-6-(三氟甲基)吡啶-2-甲醯胺。 LC-MS (方法A1):Rt = 1.46 min (UV檢測器:TIC Smooth),質量測定值478.00。 步驟B:將50 mg (0.10 mmol) N-{6-甲氧基-2-[3-(四氫-2H-吡喃-2-基氧基)丙基]-2H-吲唑-5-基}-6-(三氟甲基)吡啶-2-甲醯胺溶解於4 ml二氯甲烷中,添加10 mg (0.05 mmol)對甲苯磺酸一水合物並將混合物在25℃下攪拌72小時。添加另一50 mg (0.25 mmol)對甲苯磺酸一水合物,並將混合物在25℃下攪拌72小時。將反應混合物與飽和碳酸氫鈉溶液混合並用乙酸乙酯萃取三次。過濾(疏水過濾器)合併之有機相且濃縮。將殘餘物溶解於2.5 ml二甲亞碸中並藉由製備型HPLC純化。冷凍乾燥含有產物之流份。此得到30 mg N-[2-(3-羥基丙基)-6-甲氧基-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺。 LC-MS (方法A1):Rt = 1.19 min (UV檢測器:TIC Smooth),質量測定值394.001 H-NMR (400MHz, DMSO-d6 ): δ = 2.04 (五重峰, 2H), 3.41 (q, 2H), 3.99 (s, 3H), 4.41 (t, 2H), 4.62 (t, 1H), 7.14 (s, 1H), 8.20 (dd, 1H), 8.28 (s, 1H), 8.37 - 8.43 (m, 1H), 8.44 - 8.49 (m, 1H), 8.69 (s, 1H), 10.50 (s, 1H)。實例 42 N-{2-[(2S)-2,3- 二羥基丙基 ]-6- 甲氧基 -2H- 吲唑 -5- }-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 步驟A:將200 mg (595 µmol) N-(6-甲氧基-1H-吲唑-5-基)-6-(三氟甲基)吡啶-2-甲醯胺溶解於5 ml DMF中,並在攪拌下添加120 µl (890 µmol) (4R)-4-(氯甲基)-2,2-二甲基-1,3-二氧戊環、247 mg (0.89 mmol)碳酸鉀及148 mg (892 µmol)碘化鉀。將懸浮液在120℃下攪拌21小時。添加另一124 mg (4.85 mmol)碳酸鉀及74 mg (446 µmol)碘化鉀,並將混合物在120℃下再攪拌23小時。用水稀釋反應混合物並用乙酸乙酯萃取三次。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。將殘餘物吸收於3 ml二甲亞碸中並藉由製備型HPLC純化。此得到81 mg N-(2-{[(4S)-2,2-二甲基-1,3-二氧戊環-4-基]甲基}-6-甲氧基-2H-吲唑-5-基)-6-(三氟甲基)吡啶-2-甲醯胺。 LC-MS (方法A1):Rt = 1.27 min (UV檢測器:TIC Smooth),質量測定值450.00。 步驟B:首先在4 ml二氯甲烷中裝填81.3 mg (180 µmol) N-(2-{[(4S)-2,2-二甲基-1,3-二氧戊環-4-基]甲基}-6-甲氧基-2H-吲唑-5-基)-6-(三氟甲基)吡啶-2-甲醯胺。添加51.5 mg (271 µmol)對甲苯磺酸一水合物,並將混合物在25℃下攪拌19小時。將飽和碳酸氫鈉溶液添加至反應混合物中,並利用抽吸濾出所形成之固體並用水洗滌三次並用乙醚洗滌三次。在減壓下乾燥固體。此得到66.3 mg N-{2-[(2S)-2,3-二羥基丙基]-6-甲氧基-2H-吲唑-5-基}-6-(三氟甲基)吡啶-2-甲醯胺。 LC-MS (方法A1):Rt = 1.02 min (UV檢測器:TIC Smooth),質量測定值410.001 H-NMR (400MHz, DMSO-d6): δ = 3.34 - 3.43 (m, 2H), 3.89 - 4.01 (m, 4H), 4.21 (dd, 1H), 4.45 (dd, 1H), 7.13 (s, 1H), 8.20 (dd, 1H), 8.24 (s, 1H), 8.36 - 8.43 (m, 1H), 8.44 - 8.48 (m, 1H), 8.68 (s, 1H), 10.49 (s, 1H)。實例 43 6-( 二氟甲基 )-N-[2-(2- 羥基乙基 )-6- 甲氧基 -2H- 吲唑 -5- ] 吡啶 -2- 甲醯胺 步驟A:將400 mg (1.29 mmol) (5-胺基-6-甲氧基-2H-吲唑-2-基)乙酸苄基酯(CAS編號:1799835-89-4)及245 mg (1.41 mmol) 6-(二氟甲基)吡啶-2-甲酸溶解於10 ml THF中,並在25℃下添加197 mg (1.29 mmol) 1-羥基-1H-苯并三唑水合物、493 mg (2.57 mmol) 1-(3-二甲基胺基丙基)-3-乙基碳二亞胺鹽酸鹽及537 µl (3.85 mmol)三乙胺。在水性後處理、過濾及濃縮後,將殘餘物吸收於乙醚及少量水中並容許攪拌30分鐘。利用抽吸濾出固體,用乙醚洗滌並乾燥。此得到401 mg [5-({[6-(二氟甲基)吡啶-2-基]羰基}胺基)-6-甲氧基-2H-吲唑-2-基]乙酸苄基酯。 LC-MS (方法A1):Rt = 1.29 min (UV檢測器:TIC Smooth),質量測定值466.00。 步驟B:將128 mg (0.27 mmol) [5-({[6-(二氟甲基)吡啶-2-基]羰基}胺基)-6-甲氧基-2H-吲唑-2-基]乙酸苄基酯溶解於4.2 ml THF/甲醇(10:1)中,並添加10 mg (0.27 mmol)硼氫化鈉。將混合物在25℃下攪拌21小時。將水添加至反應混合物中並利用抽吸濾出所得固體並用水洗滌三次並用乙醚洗滌三次。將粗製產物溶解於2.5 ml二甲亞碸中並藉由製備型HPLC純化。冷凍乾燥含有產物之流份。此得到37 mg 6-(二氟甲基)-N-[2-(2-羥基乙基)-6-甲氧基-2H-吲唑-5-基]吡啶-2-甲醯胺。 LC-MS (方法A1):Rt = 0.98 min (UV檢測器:TIC Smooth),質量測定值362.001 H NMR (400 MHz, DMSO-d6): δ = 3.85 (q, 2 H), 4.00 (s, 3 H), 4.38 (t, 2 H), 4.95 (t, 1 H), 7.15 (t, 1 H), 7.15 (d, 1 H), 7.94 - 8.02 (m, 1 H), 8.25 - 8.37 (m, 3 H), 8.69 (s, 1 H), 10.55 (s, 1 H)。實例 44 N-[6- -2-(2- 羥基乙 )-2H- 吲唑 -5- ]-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 步驟A:將2.50 g (7.4 mmol) N-(6-氯-1H-吲唑-5-基)-6-(三氟甲基)吡啶-2-甲醯胺、75 ml THF、2.33 ml (14.7 mmol)溴乙酸苄基酯及3.11 ml (14.7 mmol) N,N-二環己基甲胺之混合物在65℃下攪拌27小時。添加2.33 ml (14.7 mmol)溴乙酸苄基酯及3.11 ml (14.7 mmol) N,N-二環己基甲胺,並將混合物在65℃下攪拌67小時。濾出沈澱固體並用乙酸乙酯將濾液萃取兩次。用1M鹽酸水溶液、飽和碳酸氫鈉溶液及飽和氯化鈉溶液洗滌合併之有機相,過濾並濃縮。藉由急速層析(Biotage SNAP筒柱(100 g;KP-Sil), 移動相:己烷/乙酸乙酯)純化殘餘物。然後藉由製備型HPLC純化粗製產物。此得到1.08 g [6-氯-5-({[6-(三氟甲基)吡啶-2-基]羰基}胺基)-2H-吲唑-2-基]乙酸苄基酯。 LC-MS (方法A1):Rt = 1.46 min (UV檢測器:TIC Smooth),質量測定值488.001 H NMR (400 MHz, DMSO-d6): δ = 5.22 (s, 2 H), 5.53 (s, 2 H), 7.33 - 7.41 (m, 5 H), 7.95 (s, 1 H), 8.24 (dd, 1 H), 8.42 (d, 1 H), 8.45 - 8.49 (m, 1 H), 8.53 - 8.55 (m, 1 H), 8.66 (s, 1 H), 10.53 (s, 1 H)。 步驟B:將185 mg (0.38 mmol) [6-氯-5-({[6-(三氟甲基)吡啶-2-基]羰基}胺基)-2H-吲唑-2-基]乙酸苄基酯溶解於5 ml THF中,並添加14 mg (0.38 mmol)硼氫化鈉。將混合物在25℃下攪拌21.5小時。然後使用吸量管添加500 µl甲醇,並將混合物在25℃下攪拌5小時。將水添加至反應混合物中。利用抽吸濾出固體,用水洗滌兩次並用乙醚洗滌三次並在減壓下乾燥。此得到144 mg N-[6-氯-2-(2-羥基乙基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺。 LC-MS (方法A1):Rt = 1.16 min (UV檢測器:TIC Smooth),質量測定值384.001 H-NMR (400MHz, DMSO-d6): δ = 3.88 (q, 2H), 4.47 (t, 2H), 4.99 (t, 1H), 7.92 (s, 1H), 8.23 (dd, 1H), 8.38 - 8.44 (m, 1H), 8.45 - 8.50 (m, 2H), 8.65 (s, 1H), 10.52 (s, 1H)。實例 45 N-{6- -2-[(2R)-2,3- 二羥基丙基 ]-2H- 吲唑 -5- }-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 步驟A:將300 mg (881 µmol) N-(6-氯-1H-吲唑-5-基)-6-(三氟甲基)吡啶-2-甲醯胺溶解於4 ml DMF中,並在攪拌下添加180 µl (1.3 mmol) (4S)-4-(氯甲基)-2,2-二甲基-1,3-二氧戊環、365 mg (2.64 mmol)碳酸鉀及219 mg (1.32 mmol)碘化鉀。將懸浮液在120℃下攪拌17小時。用水稀釋反應混合物並用乙酸乙酯萃取三次。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。藉由製備型HPLC純化殘餘物。此得到67.3 mg N-(6-氯-2-{[(4R)-2,2-二甲基-1,3-二氧戊環-4-基]甲基}-2H-吲唑-5-基)-6-(三氟甲基)吡啶-2-甲醯胺。 LC-MS (方法A1):Rt = 1.37 min (UV檢測器:TIC Smooth),質量測定值454.00。 步驟B:將67.3 mg (148 µmol) N-(6-氯-2-{[(4R)-2,2-二甲基-1,3-二氧戊環-4-基]甲基}-2H-吲唑-5-基)-6-(三氟甲基)吡啶-2-甲醯胺溶解於3 ml二氯甲烷中,添加42.2 mg (222 µmol)對甲苯磺酸一水合物並將混合物在25℃下攪拌24小時。添加飽和碳酸氫鈉溶液,並短暫攪拌反應混合物。利用抽吸濾出固體,用水洗滌三次並用乙醚洗滌三次並在乾燥櫥中乾燥過夜。此得到57 mg N-{6-氯-2-[(2R)-2,3-二羥基丙基]-2H-吲唑-5-基}-6-(三氟甲基)吡啶-2-甲醯胺。 LC-MS (方法A1):Rt = 1.07 min (UV檢測器:TIC Smooth),質量測定值414.001 H-NMR (300MHz, DMSO-d6): δ = 3.35 - 3.47 (m), 3.89 – 4.02 (m, 1H), 4.28 (dd, 1H), 4.54 (dd, 1H), 4.85 (br s, 1H), 5.10 (br s, 1H), 7.92 (s, 1H), 8.23 (dd, 1H), 8.36 - 8.51 (m, 3H), 8.62 (s, 1H), 10.51 (s, 1H)。實例 46 N-[2-(3- 羥基 -3- 甲基丁基 )-6- 甲氧基 -2H- 吲唑 -5- ]-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 將150 mg (446 µmol) N-(6-甲氧基-1H-吲唑-5-基)-6-(三氟甲基)吡啶-2-甲醯胺溶解於4 ml DMF中,並在攪拌下添加112 mg (669 µmol) 4-溴-2-甲基丁-2-醇、185 mg (1.34 mmol)碳酸鉀及111 mg (669 µmol)碘化鉀。將懸浮液在120℃下攪拌5.5小時。用水稀釋反應混合物並用乙酸乙酯萃取三次。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。將殘餘物吸收於2 ml二甲亞碸中並藉由製備型HPLC純化。將合併之產物流份凍乾。此得到41.4 mg標題化合物。 LC-MS (方法A1):Rt = 1.19 min (UV檢測器:TIC Smooth),質量測定值422.001 H-NMR (400MHz, DMSO-d6): δ = 1.16 (s, 6H), 1.97 - 2.08 (m, 2H), 3.99 (s, 3H), 4.39 - 4.48 (m, 2H), 4.52 (s, 1H), 7.15 (s, 1H), 8.22 (dd, 1H), 8.33 (s, 1H), 8.37 - 8.51 (m, 2H), 8.69 (s, 1H), 10.50 (s, 1H)。實例 47 N-[2-(3- 羥基丁基 )-6- 甲氧基 -2H- 吲唑 -5- ]-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 ( 鏡像異構物 1) 實例 48 N-[2-(3- 羥基丁基 )-6- 甲氧基 -2H- 吲唑 -5- ]-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 ( 鏡像異構物 2) 將192 mg (0.47 mmol) N-[6-甲氧基-2-(3-側氧基丁基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺溶解於5.5 ml THF/甲醇(10:1)中,並添加18 mg (0.47 mmol)硼氫化鈉。將混合物在25℃下攪拌20小時。將水添加至反應混合物中並用乙酸乙酯將水相萃取三次。用飽和氯化鈉溶液洗滌合併之有機相,過濾並濃縮。將殘餘物溶解於8 ml乙醇及甲醇之混合物中並純化並藉由製備型手性HPLC使用方法P6將其分離成鏡像異構物。冷凍乾燥含有產物之流份。此得到72 mg標題化合物(鏡像異構物1)及75 mg鏡像異構物2 (實例48)。 LC-MS (方法A1):Rt = 1.16 min (UV檢測器:TIC Smooth),質量測定值408.001 H NMR (300 MHz, DMSO-d6): δ = 1.02 - 1.11 (m, 3 H), 1.80 - 2.07 (m, 2 H), 3.48 - 3.63 (m, 1 H), 3.98 (s, 3 H), 4.41 (t, 2 H), 4.68 (d, 1 H), 7.15 (s, 1 H), 8.22 (dd, 1 H), 8.30 (s, 1 H), 8.37 - 8.44 (m, 1 H), 8.44 - 8.50 (m, 1 H), 8.68 (s, 1 H), 10.50 (s, 1 H)。實例 49 N-[6- 甲氧基 -2-(3- 甲氧基丙基 )-2H- 吲唑 -5- ]-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 將300 mg (0.60 mmol) N-(6-甲氧基-1H-吲唑-5-基)-6-(三氟甲基)吡啶-2-甲醯胺溶解於4 ml DMF中,並在攪拌下添加121 µl (1.07 mmol) 1-溴-3-甲氧基丙烷、370 mg (2.68 mmol)碳酸鉀及178 mg (1.07 mmol)碘化鉀。將反應混合物於100℃下攪拌17小時,用水稀釋並用乙酸乙酯萃取。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。藉由製備型HPLC純化殘餘物。冷凍乾燥含有產物之流份。此得到81 mg標題化合物。 LC-MS (方法A1):Rt = 1.28 min (UV檢測器:TIC Smooth),質量測定值408.001 H NMR (300 MHz, DMSO-d6): δ = 2.05 (五重峰, 2 H), 3.22 (s, 3 H), 3.27 (t, 2 H), 4.04 (s, 3 H), 4.42 (t, 2 H), 7.31 (s, 1 H), 8.01 (s, 1 H), 8.17 - 8.25 (m, 1 H), 8.40 (t, 1 H), 8.47 (d, 1 H), 8.72 (s, 1 H), 10.43 (s, 1 H)。實例 50 N-[6-(2- 羥基乙氧基 )-2-(3- 羥基 -3- 甲基丁基 )-2H- 吲唑 -5- ]-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 將40 mg (0.08 mmol){[2-(3-羥基-3-甲基丁基)-5-({[6-(三氟甲基)吡啶-2-基]羰基}胺基)-2H-吲唑-6-基]氧基}乙酸乙酯溶解於1.1 ml THF/甲醇(10:1)中,並添加6 mg (0.16 mmol)硼氫化鈉。將反應混合物在25℃下攪拌2小時。添加水並用乙酸乙酯萃取反應混合物。藉助疏水過濾器過濾合併之有機相並濃縮。此得到28 mg標題化合物。 LC-MS (方法A1):Rt = 1.02 min (UV檢測器:TIC Smooth),質量測定值452.00 1H NMR (400 MHz, DMSO-d6): δ = 1.15 (s, 6 H), 1.96 - 2.04 (m, 2 H), 3.88 (q, 2 H), 4.19 (t, 2 H), 4.38 - 4.45 (m, 2 H), 4.51 (s, 1 H), 4.87 (t, 1 H), 7.14 (s, 1 H), 8.21 (dd, 1 H), 8.31 (s, 1 H), 8.40 (t, 1 H), 8.46 (d, 1 H), 8.70 (s, 1 H), 10.65 (s, 1 H)。實例 51 N-[6-( 環丙基甲氧基 )-2-(2- 羥基乙基 )-2H- 吲唑 -5- ]-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 將3.00 g (6.49 mmol) [6-(環丙基甲氧基)-5-({[6-(三氟甲基)吡啶-2-基]羰基}胺基)-2H-吲唑-2-基]乙酸乙酯溶解於33 ml THF/甲醇(10:1)中,並添加245 mg (6.49 mmol)硼氫化鈉。將反應混合物在25℃下攪拌3小時。添加另一245 mg (6.49 mmol)硼氫化鈉,並將混合物在25℃下攪拌另一2小時。添加水並用乙酸乙酯萃取反應混合物。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。將粗製產物與乙醚一起攪拌並利用抽吸濾出固體。此得到1.91 g標題化合物。 LC-MS (方法A1):Rt = 1.20 min (UV檢測器:TIC Smooth),質量測定值420.00 1H NMR (400 MHz, DMSO-d6): δ = 0.40 - 0.47 (m, 2 H), 0.61 - 0.71 (m, 2 H), 1.35 (br. s., 1 H), 3.85 (q, 2 H), 3.99 - 4.05 (m, 2 H), 4.37 (t, 2 H), 4.94 (t, 1 H), 7.08 (s, 1 H), 8.21 (dd, 1 H), 8.26 (s, 1 H), 8.40 (t, 1 H), 8.47 (d, 1 H), 8.74 (s, 1 H), 10.69 (s, 1 H)。實例 52 N-[6- 甲氧基 -2-( 氧雜環丁 -3- 基甲基 )-2H- 吲唑 -5- ]-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 將1.00 g (2.97 mmol) N-(6-甲氧基-1H-吲唑-5-基)-6-(三氟甲基)吡啶-2-甲醯胺溶解於10 ml DMF中,並在攪拌下添加539 mg (3.57 mmol) 3-(溴甲基)氧雜環丁烷、1.23 g (8.92 mmol)碳酸鉀及592 mg (3.57 mmol)碘化鉀。將反應混合物於100℃下攪拌22.5小時。然後,用水稀釋混合物並用乙酸乙酯萃取兩次。藉助疏水過濾器過濾合併之有機相並濃縮。在乙醚中攪拌殘餘物,再次過濾,用乙醚洗滌並在減壓下乾燥。此得到158 mg標題化合物。 LC-MS (方法A1):Rt = 1.19 min (UV檢測器:TIC Smooth),質量測定值406.001 H-NMR (500MHz, DMSO-d6): δ = 3.48 - 3.58 (m, 1H), 3.98 (s, 3H), 4.48 (t, 2H), 4.64 - 4.70 (m, 4H), 7.15 (s, 1H), 8.21 (dd, 1H), 8.33 (d, 1H), 8.38 - 8.43 (m, 1H), 8.45 - 8.48 (m, 1H), 8.69 (s, 1H), 10.50 (s, 1H)。實例 53 N-[6- 甲氧基 -2-(4,4,4- 三氟丁基 )-2H- 吲唑 -5- ]-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 將120 mg (357 µmol) N-(6-甲氧基-1H-吲唑-5-基)-6-(三氟甲基)吡啶-2-甲醯胺溶解於2 ml DMF中,並在攪拌下添加68 µl (540 µmol) 3-(溴甲基)氧雜環丁烷及148 mg (1.07 mmol)碳酸鉀。將反應混合物在50℃下攪拌4.5小時並在80℃下攪拌16小時。然後,用水稀釋混合物並用乙酸乙酯萃取兩次。藉助疏水過濾器過濾合併之有機相並濃縮。藉由製備型HPLC純化殘餘物。將合併之產物流份凍乾。此得到33 mg標題化合物。 LC-MS (方法A3):Rt = 1.36 min (UV檢測器:TIC Smooth),質量測定值446.001 H-NMR (400MHz, DMSO-d6): δ = 2.08 - 2.38 (m, 4H), 3.99 (s, 3H), 4.45 (t, 2H), 7.19 (s, 1H), 8.22 (dd, 1H), 8.34 (s, 1H), 8.38 - 8.44 (m, 1H), 8.45 - 8.49 (m, 1H), 8.70 (s, 1H), 10.51 (s, 1H)。實例 54 N-[6- -2-(3- 羥基 -3- 甲基丁基 )-2H- 吲唑 -5- ]-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 將1.10 g (3.23 mmol) N-(6-氯-1H-吲唑-5-基)-6-(三氟甲基)吡啶-2-甲醯胺溶解於10 ml DMF中,並在攪拌下添加809 mg (4.84 mmol) 4-溴-2-甲基丁-2-醇、1.34 g (9.69 mmol)碳酸鉀及804 mg (4.84 mmol)碘化鉀。將懸浮液在120℃下攪拌23小時。添加另一670 mg (4.85 mmol)碳酸鉀及402 mg (2.42 mmol)碘化鉀,並將混合物在120℃下再攪拌24小時。用水稀釋反應混合物並用乙酸乙酯萃取三次。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。藉由急速層析(移動相:己烷/乙酸乙酯, Biotage Interchim 15µm筒柱(40 g;KP-Sil))純化殘餘物。濃縮合併之產物流份,添加乙醚並將混合物攪拌15分鐘。利用抽吸濾出固體,用乙醚洗滌三次並在乾燥櫥中乾燥過夜。此得到342 mg標題化合物。 LC-MS (方法A1):Rt = 1.17 min (UV檢測器:TIC Smooth),質量測定值408.001 H-NMR (400MHz, DMSO-d6): δ = 1.16 (s, 6H), 1.98 - 2.13 (m, 2H), 4.46 - 4.58 (m, 3H), 7.14 (t, 1H), 7.92 (s, 1H), 8.03 (dd, 1H), 8.28 - 8.39 (m, 2H), 8.52 (s, 1H), 8.63 (s, 1H), 10.59 (s, 1H)。實例 55 N-[6- 甲氧基 -2-(2- 甲氧基乙基 )-2H- 吲唑 -5- ]-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 將200 mg (0.60 mmol) N-(6-甲氧基-1H-吲唑-5-基)-6-(三氟甲基)吡啶-2-甲醯胺溶解於4 ml DMF中,並在攪拌下添加84 µl (0.89 mmol) 2-溴乙基甲醚、247 mg (1.78 mmol)碳酸鉀及118 mg (0.71 mmol)碘化鉀。將反應混合物於100℃下攪拌19小時,用水稀釋並用乙酸乙酯萃取。藉助疏水過濾器過濾合併之有機相並濃縮。藉由製備型HPLC純化殘餘物。冷凍乾燥含有產物之流份。此得到52 mg標題化合物。 LC-MS (方法A1):Rt = 1.28 min (UV檢測器:TIC Smooth),質量測定值394.001 H-NMR (500MHz, DMSO-d6): δ = 3.24 (s, 3H), 3.81 (t, 2H), 3.99 (s, 3H), 4.51 (t, 2H), 7.15 (s, 1H), 8.21 (dd, 1H), 8.28 (s, 1H), 8.38 - 8.43 (m, 1H), 8.45 - 8.48 (m, 1H), 8.69 (s, 1H), 10.50 (s, 1H)。實例 56 N-{2-[2-(2- 羥基乙氧基 ) 乙基 ]-6- 甲氧基 -2H- 吲唑 -5- }-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 將150 mg (446 µmol) N-(6-甲氧基-1H-吲唑-5-基)-6-(三氟甲基)吡啶-2-甲醯胺溶解於2 ml DMF中,並在攪拌下添加71 µl (670 µmol) 2-(2-氯乙氧基)乙醇、185 mg (1.34 mmol)碳酸鉀及111 mg (669 µmol)碘化鉀。將懸浮液在100℃下攪拌26小時。用水稀釋反應混合物並用乙酸乙酯萃取三次。用飽和氯化鈉溶液洗滌合併之有機相,藉助疏水過濾器過濾並濃縮。藉由急速層析(Interchim 15 µm筒柱(4 g;KP-Sil), 移動相:己烷/乙酸乙酯)純化殘餘物。此得到38.9 mg標題化合物。 LC-MS (方法A1):Rt = 1.08 min (UV檢測器:TIC Smooth),質量測定值424.001 H-NMR (400MHz, DMSO-d6): δ = 3.40 - 3.50 (m, 4H), 3.90 (t, 2H), 3.99 (s, 3H), 4.51 (t, 2H), 4.62 (t, 1H), 7.16 (s, 1H), 8.22 (d, 1H), 8.32 (s, 1H), 8.38 - 8.45 (m, 1H), 8.45 - 8.50 (m, 1H), 8.70 (s, 1H), 10.51 (s, 1H)。實例 57 {2-[6- 甲氧基 -5-({[6-( 三氟甲基 ) 吡啶 -2- ] 羰基 } 胺基 )-2H- 吲唑 -2- ] 乙氧基 } 乙酸乙酯 將300 mg (0.89 mmol) N-(6-甲氧基-1H-吲唑-5-基)-6-甲基吡啶-2-甲醯胺溶解於5 ml DMF中,並在攪拌下添加377 ml (1.78 mmol) (2-溴乙氧基)乙酸乙酯及493 mg (3.57 mmol)碳酸鉀。將反應混合物在25℃下攪拌2小時並在65℃攪拌21小時。用水稀釋反應混合物並用乙酸乙酯萃取三次。用飽和氯化鈉溶液洗滌合併之有機相,分離各相,藉助疏水過濾器過濾並濃縮。藉由製備型HPLC純化殘餘物。冷凍乾燥含有產物之流份。此得到96 mg標題化合物。 LC-MS (方法A1):Rt = 1.26 min (UV檢測器:TIC Smooth),質量測定值466.001 H-NMR (500MHz, DMSO-d6): δ = 1.16 (t, 3H), 3.93 (t, 2H), 4.04 (s, 3H), 4.05 - 4.11 (m, 4H), 4.57 (t, 2H), 7.40 (s, 1H), 8.02 (d, 1H), 8.21 (dd, 1H), 8.38 - 8.43 (m, 1H), 8.45 - 8.49 (m, 1H), 8.72 (s, 1H), 10.43 (s, 1H)。實例 58 {[2-(3- 羥基 -3- 甲基丁基 )-5-({[6-( 三氟甲基 ) 吡啶 -2- ] 羰基 } 胺基 )-2H- 吲唑 -6- ] 氧基 } 乙酸 將50 mg (0.10 mmol){[2-(3-羥基-3-甲基丁基)-5-({[6-(三氟甲基)吡啶-2-基]羰基}胺基)-2H-吲唑-6-基]氧基}乙酸乙酯溶解於1.5 ml THF中,並添加12 mg (0.51 mmol)氫氧化鋰一水合物及150 µl水。將反應混合物於25℃下攪拌60分鐘。用1 M鹽酸將混合物酸化至pH = 3並利用抽吸濾出所得沈澱物並用水及乙醚洗滌。此得到35 mg標題化合物。 LC-MS (方法A1):Rt = 1.00 min (UV檢測器:TIC Smooth),質量測定值466.00 1H NMR (300 MHz, DMSO-d6): δ = 1.15 (s, 6 H), 1.96 - 2.06 (m, 2 H), 4.38 - 4.47 (m, 2 H), 4.51 (s, 1 H), 4.89 (s, 2 H), 7.13 (s, 1 H), 8.14 - 8.24 (m, 1 H), 8.34 (s, 1 H), 8.36 - 8.45 (m, 1 H), 8.45 - 8.50 (m, 1 H), 8.73 (s, 1 H), 10.57 (s, 1 H)。實例 59 {2-[6- 甲氧基 -5-({[6-( 三氟甲基 ) 吡啶 -2- ] 羰基 } 胺基 )-2H- 吲唑 -2- ] 乙氧基 } 乙酸 將82 mg (0.18 mmol){2-[6-甲氧基-5-({[6-(三氟甲基)吡啶-2-基]羰基}胺基)-2H-吲唑-2-基]乙氧基}乙酸乙酯溶解於2 ml THF中並添加300 µl乙醇、73 mg (1.76 mmol)氫氧化鋰一水合物於300 µl水中之溶液,並將混合物在50℃下攪拌17小時。用水稀釋混合物並使用10%濃度之檸檬酸溶液酸化至pH 3。用乙酸乙酯將水相萃取三次並藉助疏水過濾器過濾合併之有機相並濃縮。將乙醚添加至殘餘物中,並將混合物攪拌10分鐘,再次過濾,用乙醚洗滌並在減壓下乾燥。藉由製備型HPLC純化殘餘物。冷凍乾燥含有產物之流份。此得到49 mg標題化合物。 LC-MS (方法A1):Rt = 1.09 min (UV檢測器:TIC Smooth),質量測定值438.001 H NMR (300 MHz, DMSO-d6): δ = 3.94 - 4.02 (m, 7 H), 4.53 (t, 2 H), 7.15 (s, 1 H), 8.21 (dd, 1 H), 8.33 (s, 1 H), 8.36 - 8.44 (m, 1 H), 8.44 - 8.49 (m, 1 H), 8.69 (s, 1 H), 10.50 (s, 1 H)。實例 60 N-[6- 乙氧基 -2-(3- 羥基 -3- 甲基丁基 )-2H- 吲唑 -5- ]-6-( 五氟乙基 ) 吡啶 -2- 甲醯胺 將100 mg (0.38 mmol) 4-(5-胺基-6-乙氧基-2H-吲唑-2-基)-2-甲基丁-2-醇、137 mg (0.57 mmol) 6-(五氟乙基)吡啶-2-甲酸(CAS 1283717-85-0)、80 µl (0.57 mmol)三乙胺及217 mg (0.57 mmol) HATU於2 ml DMF中之混合物在RT下攪拌過夜。藉由製備型HPLC純化混合物,得到150 mg標題化合物。 LC-MS (方法A2):Rt = 1.32 min (UV檢測器:TIC Smooth),質量測定值486.171 H-NMR (400MHz, DMSO-d6 ): δ = 1.14 (s, 6H), 1.47 (t, 3H), 1.97 – 2.05 (m, 2H), 4.15 – 4.24 (q, 2H), 4.37 - 4.48 (m, 2H), 4.52 (s, 1H), 7.11 (s, 1H), 8.23 - 8.27 (m, 1H), 8.31 (s, 1H), 8.39 – 8.51 (m, 2H), 8.73 (s, 1H), 10.60 (s, 1H)。實例 61 6-(1,1- 二氟乙基 )-N-[6- 乙氧基 -2-(3- 羥基 -3- 甲基丁基 )-2H- 吲唑 -5- ] 吡啶 -2- 甲醯胺 將100 mg (0.38 mmol) 4-(5-胺基-6-乙氧基-2H-吲唑-2-基)-2-甲基丁-2-醇、106 mg (0.57 mmol) 6-(1,1-二氟乙基)吡啶-2-甲酸、80 µl (0.57 mmol)三乙胺及217 mg (0.57 mmol) HATU於2 ml DMF中之混合物在RT下攪拌過夜。藉由製備型HPLC純化混合物,得到75 mg標題化合物。 LC-MS (方法A2):Rt = 1.23 min (UV檢測器:TIC Smooth),質量測定值432.20。1 H-NMR (400MHz, DMSO-d6 ): δ = 1.15 (s, 6 H), 1.51 (t, 3H), 1.96 – 2.05 (m, 2H), 2.15 (t, 3H), 4.15 – 4.26 (m, 2 H), 4.37 - 4.45 (m, 2H), 4.52 (s, 1H), 7.11 (s, 1H), 7.97 - 8.04 (m, 1H), 8.24 – 8.37 (m, 3H), 8.72 (s, 1H), 10.70 (s, 1H)。實例 62 N-[6- 乙氧基 -2-(3- 羥基 -3- 甲基丁基 )-2H- 吲唑 -5- ]-6,7- 二氫 -4H- 吡唑并 [5,1-c][1,4] 噁嗪 -2- 甲醯胺 將100 mg 4-(5-胺基-6-乙氧基-2H-吲唑-2-基)-2-甲基丁-2-醇、67 mg 6,7-二氫-4H-吡唑并[5,1-c][1,4]噁嗪-2-甲酸、0.19 ml N-乙基-N-異丙基丙烷-2-胺及165 mg HATU於2 ml THF中之混合物在RT下攪拌18小時。用水稀釋反應混合物並用乙酸乙酯萃取三次,並濃縮合併之有機相。藉由製備型HPLC純化混合物得到124 mg標題化合物。 LC-MS (方法A1):Rt = 0.93 min (UV檢測器:TIC Smooth),質量測定值413.21。1 H-NMR (400MHz, DMSO-d6 ): δ = 1.14 (s, 6H), 1.46 (t, 3H), 1.96 - 2.04 (m, 2H), 4.07 - 4.14 (m, 2H), 4.16 - 4.28 (m, 4H), 4.36 - 4.44 (m, 2H), 4.51 (br s, 1H), 4.84 (s, 2H), 6.59 (s, 1H), 7.08 (s, 1H), 8.25 (s, 1H), 8.55 (s, 1H), 9.46 (s, 1H)。實例 63 N-[6- 乙氧基 -2-(3- 羥基 -3- 甲基丁基 )-2H- 吲唑 -5- ]-1- 乙基 -1H- 吡唑 -3- 甲醯胺 將100 mg 4-(5-胺基-6-乙氧基-2H-吲唑-2-基)-2-甲基丁-2-醇、56 mg 1-乙基-1H-吡唑-3-甲酸、0.19 ml N-乙基-N-異丙基丙烷-2-胺及165 mg HATU於2 ml THF中之混合物在RT下攪拌18小時。用水稀釋反應混合物並用乙酸乙酯萃取三次,並濃縮合併之有機相。藉由製備型HPLC純化混合物得到114 mg標題化合物。 LC-MS (方法A1):Rt = 1.02 min (UV檢測器:TIC Smooth),質量測定值385.21。1 H-NMR (400MHz, DMSO-d6 ): δ = 1.14 (s, 6H), 1.42 - 1.51 (m, 6H), 1.97 - 2.03 (m, 2H), 4.15 - 4.28 (m, 4H), 4.36 - 4.43 (m, 2H), 4.51 (s, 1H), 6.75 (d, 1H), 7.07 (s, 1H), 7.93 (d, 1H), 8.25 (s, 1H), 8.54 (s, 1H), 9.57 (s, 1H)。實例 64 N-[6- 乙氧基 -2-(3- 羥基 -3- 甲基丁基 )-2H- 吲唑 -5- ]-1- 異丙基 -1H- 吡唑 -3- 甲醯胺 將100 mg 4-(5-胺基-6-乙氧基-2H-吲唑-2-基)-2-甲基丁-2-醇、61 mg 1-異丙基-1H-吡唑-3-甲酸、0.19 ml N-乙基-N-異丙基丙烷-2-胺及165 mg HATU於2 ml THF中之混合物在RT下攪拌18小時。用水稀釋反應混合物並用乙酸乙酯萃取三次,並濃縮合併之有機相。藉由製備型HPLC純化混合物得到127 mg標題化合物。 LC-MS (方法A1):Rt = 1.10 min (UV檢測器:TIC Smooth),質量測定值399.23。1 H-NMR (400MHz, DMSO-d6 ): δ [ppm]= 1.14 (s, 6H), 1.44 - 1.53 (m, 9H), 1.97 - 2.03 (m, 2H), 4.18 (q, 2H), 4.36 - 4.44 (m, 2H), 4.51 (s, 1H), 4.62 (spt, 1H), 6.74 (d, 1H), 7.07 (s, 1H), 7.96 (d, 1H), 8.25 (s, 1H), 8.54 (s, 1H), 9.65 (s, 1H)。實例 65 N-[6- 乙氧基 -2-(3- 羥基 -3- 甲基丁基 )-2H- 吲唑 -5- ] 吡唑并 [1,5-a] 嘧啶 -3- 甲醯胺 將100 mg 4-(5-胺基-6-乙氧基-2H-吲唑-2-基)-2-甲基丁-2-醇、65 mg 1-吡唑并[1,5-a]嘧啶-3-甲酸、0.19 ml N-乙基-N-異丙基丙烷-2-胺及165 mg HATU於2 ml THF中之混合物在RT下攪拌18小時。用水稀釋反應混合物並用乙酸乙酯萃取三次,並濃縮合併之有機相。藉由製備型HPLC純化混合物得到57 mg標題化合物。 LC-MS (方法A1):Rt = 0.89 min (UV檢測器:TIC Smooth),質量測定值408.191 H-NMR (400MHz, DMSO-d6): δ = 1.15 (s, 6H), 1.60 (t, 3H), 1.97 - 2.05 (m, 2H), 4.20 (q, 2H), 4.35 - 4.45 (m, 2H), 4.51 (br s, 1H), 7.05 (s, 1H), 7.34 (dd, 1H), 8.25 (s, 1H), 8.71 (s, 2H), 8.86 (dd, 1H), 9.38 (dd, 1H), 10.64 (s, 1H)。實例 66 N-[6- 甲氧基 -2-( 四氫呋喃 -3- 基甲基 )-2H- 吲唑 -5- ]-6-( 三氟甲基 ) 吡啶 -2- 甲醯胺 將400 mg N-(6-甲氧基-1H-吲唑-5-基)-6-(三氟甲基)吡啶-2-甲醯胺(CAS編號:1799836-45-5, 作為粗製產物使用)、310 mg 3-(溴甲基)四氫呋喃及519 mg碳酸鉀於5.0 ml DMF中之混合物在100℃下攪拌5小時並在80℃下攪拌21小時。然後添加水,並用乙酸乙酯將混合物萃取三次。用飽和鹽水洗滌合併之有機相,藉助相分離器過濾並濃縮。藉由製備型HPLC純化得到120 mg標題化合物。1 H-NMR (400MHz, DMSO-d6 ): δ = 1.59 - 1.72 (m, 1H), 1.88 - 2.00 (m, 1H), 2.79 - 2.92 (m, 1H), 3.49 - 3.57 (m, 1H), 3.60 - 3.73 (m, 2H), 3.79 (td, 1H), 3.99 (s, 3H), 4.35 (d, 2H), 7.17 (s, 1H), 8.22 (d, 1H), 8.34 - 8.49 (m, 3H), 8.69 (s, 1H), 10.51 (s, 1H)。實例 67 N-[2-(3- 羥基 -3- 甲基丁基 )-6- 甲氧基 -2H- 吲唑 -5- ]-6-( 吡咯啶 -1- ) 吡啶 -2- 甲醯胺 將277 mg碳酸鉀及125 mg碘化鉀添加至169 mg N-(6-甲氧基-1H-吲唑-5-基)-6-(吡咯啶-1-基)吡啶-2-甲醯胺及126 mg 4-溴-2-甲基丁-2-醇於2.7 ml DMF中之混合物中,並將混合物在100℃下加熱22小時。添加42 mg 4-溴-2-甲基丁-2-醇及327 mg碳酸銫並將混合物在100℃下攪拌6小時。利用抽吸濾出混合物,用DMF洗滌殘餘物,濃縮濾液並藉由製備型HPLC純化殘餘物。此得到49 mg標題化合物。1 H-NMR (500MHz, DMSO-d6): DMSO-d6): δ = 1.15 (s, 6H), 1.98 - 2.06 (m, 6H), 3.51 (br s, 4H), 3.98 (s, 3H), 4.37 - 4.45 (m, 2H), 4.52 (s, 1H), 6.73 (d, 1H), 7.09 (s, 1H), 7.33 (d, 1H), 7.67 - 7.75 (m, 1H), 8.26 (s, 1H), 8.65 (s, 1H), 10.92 (s, 1H)。生理效能之評價 IRAK4 激酶分析 在下文所闡述之Irak4 TR-FRET分析(TR-FRET = 時間解析螢光共振能量轉移)中量測本發明物質之IRAK4-抑制活性。 使用在桿狀病毒感染之昆蟲細胞(Hi5, BTI-TN-5B1-4, 購自Invitrogen之細胞系, 目錄號B855-02)中表現且經由親和層析純化之來自N末端GST (麩胱甘肽S-轉移酶)及人類Irak4之重組體融合蛋白作為酶。激酶反應所使用之受質為生物素化肽生物素-Ahx-KKARFSRFAGSSPSQASFAEPG (呈醯胺形式之C末端),其可購自(例如)Biosyntan GmbH公司(Berlin-Buch)。 為進行該分析,自測試物質於DMSO中之2 mM溶液製備在20 µM至0.073 nM範圍內之11個不同濃度。將50 nl各別溶液吸量至黑色低體積384孔微量滴定板(Greiner Bio-One, Frickenhausen, Germany)中,添加2 µl Irak4於分析緩衝液[50 mM HEPES pH 7.5, 5 mM MgCl2, 1.0 mM二硫蘇糖醇, 30 µM經活化正釩酸鈉, 0.1% (w/v)牛γ-免疫球蛋白(BGG) 0.04% (v/v) nonidet-P40 (Sigma)]中之溶液,並將混合物培育15分鐘,以容許物質在激酶反應前預結合至酶。然後藉由添加3 µl腺苷三磷酸(ATP, 1.67 mM = 於5 µl分析體積中之最終濃度:1 mM)及肽受質(0.83 µM =於5 µl分析體積中之最終濃度:0.5 µM)於分析緩衝液中之溶液來開始激酶反應,並將所得混合物在22℃下培育45分鐘之反應時間。根據酶之各別活性調節Irak4之濃度且加以設置使得該分析在線性範圍內實施。典型濃度為約0.2 nM之數量級。藉由添加5 µl TR-FRET檢測試劑[0.1 µM鏈黴抗生物素蛋白(streptavidin)-XL665 (Cisbio Bioassays;France, 目錄號610SAXLG)]及1.5 nM抗磷絲胺酸抗體[Merck Millipore, 「STK Antibody」, 目錄號35-002]及0.6 nM LANCE EU-W1024標記之抗小鼠IgG抗體(Perkin-Elmer, 產品號AD0077;另一選擇為,可使用來自Cisbio Bioassays之鋱穴狀化合物標記之抗小鼠-IgG抗體)於EDTA水溶液(100 mM EDTA,於25 mM HEPES (pH 7.5)中之0.4 % [w/v]牛血清白蛋白[BSA])中之溶液來終止反應。 將所得混合物在22℃下培育1小時以容許形成生物素化磷酸化受質及檢測試劑之複合物。然後藉由量測銪螯合物標記之抗小鼠-IgG抗體至鏈黴抗生物素蛋白-XL665之共振能量轉移來評估磷酸化受質之量。為此,在TR-FRET量測儀器(例如Rubystar (BMG Labtechnologies, Offenburg, Germany)或Viewlux (Perkin-Elmer))中在350 nm下激發後量測在620 nm及665 nm下之螢光發射。將665 nm及622 nm下之發射之比率視為磷酸化受質之量之量度。將數據正規化(無測試物質之酶反應= 0%抑制;所有其他分析組份但沒有酶= 100%抑制)。通常,在相同微量滴定板上在20 µM至0.073 nM範圍內之11個不同濃度(20 µM、5.7 µM、1.6 µM、0.47 µM、0.13 µM、38 nM、11 nM、3.1 nM、0.89 nM、0.25 nM及0.073 nM)下測試測試物質。在分析前藉由系列稀釋製備稀釋系列(2 mM至7.3 nM,在100% DMSO中)。藉由4參數擬合計算IC50 值。 1 在IRAK4激酶分析中實例化合物之IC50 THP-1 細胞中之 TNF-α 分泌 借助於此測試,可測試物質在THP-1細胞(人類單核球性急性白血病細胞系)中抑制TNF-α (腫瘤壞死因子α)分泌之能力。TNF-α係參與發炎過程之細胞介素。在此測試中,TNF-α分泌係藉由利用與細菌脂多醣(LPS)培育觸發。 THP-1細胞保持在連續懸浮細胞培養物[沒有L-Glutamax (GE Healthcare, 目錄號E15-039)且補充有胎牛血清(FCS) 10% (Invitrogen, 目錄號10082-147)、1% L-麩醯胺酸(Sigma, 目錄號G7513)、1%青黴素/鏈黴素(Streptomycin) (PAA, 目錄號P11-010)及50 µM 2-巰基乙醇(Gibco, 目錄號31350-010)之RPMI 1460培養基]中且不應超過1×106 個細胞/ml之細胞濃度。該分析係在細胞培養基(補充有L-麩醯胺酸、青黴素、鏈黴素及2-巰基乙醇之RPMI 1460培養基)中實施。 將THP-1細胞以2.5×105 個細胞/孔之細胞密度播種於96孔板中。以100% DMSO之恆定體積使本發明化合物經受系列稀釋,且其在該分析中係使用10 µM至3 nM範圍內之8個不同濃度,使得最終DMSO濃度為0.4% DMSO。在實際刺激前,然後將細胞與該等一起預培育30分鐘。為誘導細胞介素分泌,利用1 µg/ml LPS (Sigma, 大腸桿菌0127:B8, 目錄號L4516)將細胞刺激6小時。作為中性對照,用1 µg/ml LPS及0.04% DMSO處理細胞,且作為抑制劑對照,僅用0.04% DMSO處理細胞。使用CellTiter-Glo發光分析(Promega, 目錄號G7571 (G755/G756A))根據製造商之說明書測定細胞存活率。使用人類促炎性9-Plex組織培養套組(MSD, 目錄號K15007B)根據製造商之說明書測定在細胞培養上清液中分泌之TNF-α之量。 物質之活性表示為在中性與抑制劑對照之間之比率(以百分比表示)。使用4參數擬合計算IC50 值。 2 實例化合物關於THP-1細胞中TNF-α之分泌且對細胞存活率無影響之IC50 醫藥組合物之工作實例 可將本發明化合物轉化為如下醫藥製劑:錠劑 組成 100 mg實例11之化合物或實例12之化合物、50 mg乳糖(一水合物)、50 mg玉蜀黍澱粉(天然)、10 mg聚乙烯基吡咯啶酮(PVP 25) (來自BASF, Ludwigshafen, Germany)及2 mg硬脂酸鎂。 錠劑重量212 mg。直徑8 mm,曲率半徑12 mm。生產: 利用5% PVP水溶液(w/w)將本發明化合物、乳糖及澱粉之混合物造粒。將該等顆粒乾燥且然後將其與硬脂酸鎂混合5分鐘。使用習用壓錠機壓縮此混合物(對於錠劑之格式參見上文)。壓製所用引導值為15 kN之壓製力。用於經口投與之懸浮液 組成 1000 mg實例11之化合物或實例12之化合物、1000 mg乙醇(96%)、400 mg Rhodigel® (黃原膠,來自FMC, Pennsylvania, USA)及99 g水。 10 ml口服懸浮液對應於100 mg本發明化合物之單一劑量。生產: 將Rhodigel懸浮於乙醇中;將本發明化合物添加至懸浮液中。在攪拌之同時添加水。將混合物攪拌約6小時直至Rhodigel完成膨脹為止。用於經口投與之溶液: 組成 500 mg實例11之化合物或實例12之化合物、2.5 g聚山梨醇酯及97 g聚乙二醇400。20 g口服溶液對應於100 mg本發明化合物之單一劑量。生產 在攪拌下,將本發明化合物懸浮於聚乙二醇及聚山梨醇酯之混合物中。繼續該攪拌操作直至本發明化合物完全溶解為止。In the case of the synthetic intermediates and working examples of the invention set forth below, any compound specified in the form of the corresponding base or acid salt will generally be a salt having an unknown exact stoichiometric composition, such as by separate preparation and / Or obtained by a purification process. Unless specified in more detail, in the case of such salts, the name and structural formula (eg "hydrochloride", "trifluoroacetate", "sodium salt" or "x HCl", "x CF"3 COOH", "x Na+ The addition of ") is not to be understood stoichiometrically, but only to the illustrative features of the salt-forming components present therein. If the synthetic intermediate or working example or a salt thereof is obtained by the preparation and/or purification process in the form of a solvate having an unknown stoichiometric composition (for example, a hydrate) if it has a defined type, this corresponds Suitable for use. If the compound encompassed by the formula (I) and mentioned below is not a solvate of a salt, a solvate or a salt, the compound of the invention is a solvent of a compound of the formula (I) and a salt, a solvate thereof and a salt thereof. And a solvate of a salt, a solvate and a salt thereof, which are encompassed by the formula (I) and which are exemplified by the formula (I) and which are cited below as working examples, and a salt, a solvate thereof and a solvate of a salt. Preferred salts in the context of the present invention are physiologically acceptable salts of the compounds of the invention. However, the invention also encompasses salts which are not themselves suitable for pharmaceutical use but which are useful, for example, for isolating or purifying the compounds of the invention. Physiologically acceptable salts of the compounds of the present invention include acid addition salts of mineral acids, carboxylic acids and sulfonic acids, such as the following acid salts: hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, toluene. Sulfonic acid, benzenesulfonic acid, naphthalene disulfonic acid, acetic acid, trifluoroacetic acid, propionic acid, lactic acid, tartaric acid, malic acid, citric acid, fumaric acid, maleic acid and benzoic acid. Physiologically acceptable salts of the compounds of the invention also include the salts of conventional bases, exemplified and preferred alkali metal salts (e.g., sodium and potassium), alkaline earth metal salts (e.g., calcium and magnesium); Ammonia or an ammonium salt of an organic amine having from 1 to 16 carbon atoms, such as, by way of example and preferred, ethylamine, diethylamine, triethylamine, ethyldiisopropylamine, monoethanolamine, two Ethanolamine, triethanolamine, dicyclohexylamine, dimethylaminoethanol, procaine, dibenzylamine, N-methylmorpholine, arginine, lysine, ethylenediamine and N -methylhexahydropyridine. Solvates are described in the context of the present invention as compounds of the invention in such forms as complexes formed in solid or liquid state by coordination with solvent molecules. The specific form of the solvate in which the hydrate system is coordinated to water. The compounds of the present invention may exist in different stereoisomeric forms depending on their structure, i.e., in the form of a conformational isomer or, if appropriate, as a conformational isomer (mirroromer and/or non-image isomer, including And so on in the case of configuration isomers). Accordingly, the present invention encompasses mirror image isomers and non-image isomers and individual mixtures thereof. The stereoisomerically homogeneous component can be isolated in a conventional manner from such mixtures of mirror image isomers and/or non-image isomers; preferably a chromatographic process, especially a non-pivoting or palm phase phase HPLC layer Analysis for this purpose. If a compound of the invention is present in tautomeric form, the invention encompasses all tautomeric forms. The invention also encompasses all suitable isotopic variations of the compounds of the invention. Herein, an isotopic variation of a compound of the invention is understood to mean a compound in which at least one atom in the compound of the invention has been replaced by another atom having the same atomic number but an atomic weight different from the atomic amount normally or predominantly present in nature. Examples of isotopes which may be incorporated into the compounds of the invention are isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine, bromine and iodine, for example2 H (氘),3 H (氚),13 C,14 C,15 N,17 O,18 O,32 P,33 P,33 S,34 S,35 S,36 S,18 F,36 Cl,82 Br,123 I,124 I,129 I and131 I. Particular isotopic variations of the compounds of the invention, such as, in particular, those in which one or more radioisotopes have been incorporated, may be beneficial, for example, for examining the mechanism of action or distribution of the active compound in vivo; Sexuality and detectability, in particular3 H or14 Compounds labeled with C isotope are suitable for this purpose. In addition, the incorporation of isotopes (e.g., guanidine) may result in a particular therapeutic benefit due to increased metabolic stability of the compound, e.g., increased half-life in vivo or reduced required dosage of the agent; therefore, such modified forms of the compounds of the invention In some cases, preferred embodiments of the invention may also be constructed. Isotopic variants of the compounds of the invention may be prepared by methods known to those skilled in the art (e.g., by the methods further set forth below and the procedures set forth in the working examples) by using the respective reagents and/or corresponding compounds Isotope modified forms are prepared. The invention further provides all possible crystalline and polymorphic forms of the compounds of the invention, wherein the polymorphs are present as a single polymorph or a mixture of plural polymorphs in all concentration ranges. The invention also encompasses prodrugs of the compounds of the invention. The term "prodrug" in this context refers to a compound that is biologically active or inactive but that reacts (eg, metabolizes or hydrolyzes) during its residence time in the body to produce a compound of the invention. In the context of the present invention, unless otherwise indicated, a substituent has the following meaning:alkyl In the context of the present invention is a straight or branched alkyl group having the specified number of specific carbon atoms. Examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, 1-methylpropyl, 2-methylpropyl, tert-butyl, n-pentyl, 1-B Propyl, 1-methylbutyl, 2-methylbutyl, 3-methylbutyl, 2,2-dimethylpropyl, n-hexyl, 1-methylpentyl, 2-methylpentyl Base, 3-methylpentyl, 4-methylpentyl, 1-ethylbutyl and 2-ethylbutyl. Preferred are methyl, ethyl, n-propyl, n-butyl, 2-methylbutyl, 3-methylbutyl and 2,2-dimethylpropyl. In the context of the present invention,Cycloalkyl A monocyclic saturated alkyl group having the specified number of carbon atoms in each case. Preferred examples include cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl. In the context of the present invention,Heterocycloalkyl, heterocyclic or heterocyclic A total of 3 to 10 ring atoms and one or two from N, O, S, SO, and/or SO2 A saturated heterocyclic ring of a hetero atom of a ring of constituent groups. Mention may be made, for example, of the following: azetidinyl, oxetanyl, pyrrolidinyl, pyrazolyl, tetrahydrofuranyl, hexahydropyridyl, hexahydropyrazinyl, tetrahydropyranyl, A phenyl group, a thiomorpholinyl group, a dioxo bridge thiomorpholinyl group, a dihydroindenyl group, and a dihydroisoindenyl group. Preferred are azetidinyl, oxetanyl, pyrrolidinyl, hexahydropyridyl, hexahydropyrazinyl and morpholinyl.Alkoxy In the context of the present invention is meant a linear or branched alkoxy group having the specified number of specific carbon atoms. From 1 to 6 carbon atoms are preferred. Examples include methoxy, ethoxy, n-propoxy, isopropoxy, 1-methylpropoxy, n-butoxy, isobutoxy, tert-butoxy, n-pentyloxy, iso Pentyloxy, 1-ethylpropoxy, 1-methylbutoxy, 2-methylbutoxy, 3-methylbutoxy and n-hexyloxy. More preferably, it is a linear or branched alkoxy group having 1 to 4 carbon atoms. Mention may be made, by way of example, of methoxy, ethoxy, n-propoxy, 1-methylpropoxy, n-butoxy and isobutoxy. In the context of the present invention,halogen It is fluorine, chlorine and bromine. Preferred is fluorine.Hydroxyl In the context of the present invention is OH. The symbol * at the bond indicates the bonding site in the molecule. Unless otherwise specified, when a group in a compound of the invention is substituted, the groups may be mono- or polysubstituted. In the context of the present invention, all of the groups that occur more than once are defined independently of each other. Preferred are substituted by one, two or three identical or different substituents. R1 The preferred embodiment is chlorine. R1 Another preferred embodiment is an O-R4 . R2 Preferred embodiment is C2 -C6 An alkyl group which is substituted by a hydroxyl group. Especially, R2 Representative C2 -C5 An alkyl group which is substituted by a hydroxyl group. Extremely good among them R2 A compound representing 3-hydroxypropyl, 3-hydroxybutyl, 2-hydroxyethyl or 3-hydroxy-3-methylbutyl. More preferred is hydroxy-3-methylbutyl. R2 Another preferred embodiment is C2 -C6 An alkyl group substituted with two hydroxyl groups. Especially, R2 Represents 2,3-dihydroxypropyl, 2,3-dihydroxy-3-methylbutyl or 2,3-dihydroxybutyl. Extremely better, R2 Represents 2,3-dihydroxypropyl. R2 Another preferred embodiment is C1 -C3 An alkyl group which is substituted by an oxetanyl group or a tetrahydrofuranyl group. Especially good among them R2 A compound representing oxetan-3-ylmethyl, oxetan-2-ylmethyl, tetrahydrofuran-3-ylmethyl or tetrahydrofuran-2-ylmethyl. Extremely good among them R2 A compound representing oxetan-3-ylmethyl or tetrahydrofuran-3-ylmethyl. R2 Another preferred embodiment is C2 -C6 -alkyl, which is C1 -C3 - alkoxy substituted. Especially, R2 Representative C2 -C6 An alkyl group which is substituted with a methoxy group. Extremely good among them R2 A compound representing 3-methoxy-3-methylbutyl, 3-methoxypropyl or 2-methoxyethyl. R2 Another preferred embodiment is C2 -C6 - an alkyl group substituted with a 2-hydroxyethoxy group. Especially good among them R2 A compound representing 2-(2-hydroxyethoxy)ethyl or 3-(2-hydroxyethoxy)propyl. Particularly preferred is 2-(2-hydroxyethoxy)ethyl. R2 Another preferred embodiment is C2 -C6 -alkyl, which is OCH2 (C=O)OH or OCH2 (C=O)OCH3 Replace. Especially good among them R2 Representative CH2 CH2 OCH2 C(=O)OH or CH2 CH2 OCH2 Compound of C(=O)OEt. R2 Another preferred embodiment is a 3-oxo butyl group. Also preferred is where R2 A compound of 4,4,4-trifluorobutyl, 3,3,3-trifluoropropyl, 2,2,2-trifluoroethyl or 3,3-difluorobutyl. Especially, R2 Represents 4,4,4-trifluorobutyl or 3,3,3-trifluoropropyl. Excellent is 4,4,4-trifluorobutyl. In a preferred embodiment, R3 Representative 2-(C1 -C6 -alkyl)-1,3-thiazol-4-yl, wherein C1 -C6 The alkyl substituent may optionally be monosubstituted with a cyclopropyl group, monosubstituted to pentasubstituted with a fluorine atom and optionally monosubstituted with a hydroxy group. Especially, R3 Represents 2-(trifluoromethyl)-1,3-thiazol-4-yl, 2-methyl-1,3-thiazol-4-yl, 2-ethyl-1,3-thiazol-4-yl, 2-propyl-1,3-thiazol-4-yl, 2-isopropyl-1,3-thiazol-4-yl or 2-tert-butyl-1,3-thiazol-4-yl. In another preferred embodiment, R3 Represents 2-cyclopropyl-1,3-thiazol-4-yl. In another preferred embodiment, R3 Representative 4-(C1 -C6 -alkyl)-1,3-thiazol-2-yl, wherein C1 -C6 The alkyl substituent may optionally be monosubstituted with a cyclopropyl group, monosubstituted to pentasubstituted with a fluorine atom and optionally monosubstituted with a hydroxy group. Especially, R3 Represents 4-(trifluoromethyl)-1,3-thiazol-2-yl, 4-methyl-1,3-thiazol-2-yl, 4-ethyl-1,3-thiazol-2-yl, 4-isopropyl-1,3-thiazol-2-yl or 4-tert-butyl-1,3-thiazol-2-yl. Extremely better, R3 Represents 4-(trifluoromethyl)-1,3-thiazol-2-yl. In another preferred embodiment, R3 Represents 4-cyclopropyl-1,3-thiazol-2-yl. In another preferred embodiment, R3 Representative 2-(C1 -C6 -alkyl)-1,3-oxazol-4-yl, of which C1 -C6 The alkyl substituent may optionally be monosubstituted with a cyclopropyl group, monosubstituted to pentasubstituted with a fluorine atom and optionally monosubstituted with a hydroxy group. Especially, R3 Represents 2-methyl-1,3-oxazol-4-yl, 2-(trifluoromethyl)-1,3-oxazol-4-yl, 2-ethyl-1,3-oxazole-4 -yl, 2-(1,1-difluoroethyl)-1,3-oxazol-4-yl, 2-(2,2,2-trifluoroethyl)-1,3-oxazole-4 -yl, 2-isopropyl-1,3-oxazol-4-yl, 2-tert-butyl-1,3-oxazol-4-yl or 2-(cyclopropylmethyl)-1, 3-oxazol-4-yl. Extremely better, R3 Represents 2-(trifluoromethyl)-1,3-oxazol-4-yl. In another preferred embodiment, R3 Represents 2-cyclopropyl-1,3-oxazol-4-yl. In another preferred embodiment, R3 Representative 2-(C1 -C6 -alkyl)-1,3-oxazol-5-yl, of which C1 -C6 The alkyl substituent may optionally be monosubstituted with a cyclopropyl group, monosubstituted to pentasubstituted with a fluorine atom and optionally monosubstituted with a hydroxy group. Especially, R3 Represents 2-methyl-1,3-oxazol-5-yl, 2-ethyl-1,3-oxazol-5-yl, 2-isopropyl-1,3-oxazol-5-yl. Extremely better, R3 Represents 2-methyl-1,3-oxazol-5-yl. In another preferred embodiment, R3 Representative 1-(C1 -C6 -alkyl)-1H-pyrazol-3-yl, wherein C1 -C6 The alkyl substituent may optionally be monosubstituted with a cyclopropyl group, monosubstituted to pentasubstituted with a fluorine atom and optionally monosubstituted with a hydroxy group. Especially, R3 Represents 1-(difluoromethyl)-1H-pyrazol-3-yl, 1-methyl-1H-pyrazol-3-yl, 1-ethyl-1H-pyrazol-3-yl, 1-( 2,2,2-trifluoroethyl)-1H-pyrazol-3-yl, 1-isopropyl-1H-pyrazol-3-yl, 1-propyl-1H-pyrazol-3-yl, 1-Terbutyl-1H-pyrazol-3-yl, 1-isobutyl-1H-pyrazol-3-yl. Extremely better, R3 Represents 1-(difluoromethyl)-1H-pyrazol-3-yl, 1-ethyl-1H-pyrazol-3-yl, 1-isopropyl-1H-pyrazol-3-yl. In another preferred embodiment, R3 Representative 6-(C1 -C6 -alkyl)pyridin-2-yl, wherein C1 -C6 The alkyl substituent may optionally be monosubstituted with a cyclopropyl group, monosubstituted to pentasubstituted with a fluorine atom and optionally monosubstituted with a hydroxy group. Especially, R3 Represents 6-methylpyridin-2-yl, 6-(difluoromethyl)pyridin-2-yl, 6-(trifluoromethyl)pyridin-2-yl, 6-ethylpyridin-2-yl, 6 -(1,1-difluoroethyl)pyridin-2-yl, 6-(pentafluoroethyl)pyridin-2-yl, 6-(2,2,2-trifluoroethyl)pyridin-2-yl , 6-propylpyridin-2-yl, 6-isopropylpyridin-2-yl, 6-(2-hydroxypropan-2-yl)pyridin-2-yl, 6-t-butylpyridine-2- base. Extremely better, R3 Represents 6-(1,1-difluoroethyl)pyridin-2-yl, 6-(2-hydroxypropan-2-yl)pyridin-2-yl, 6-(difluoromethyl)pyridin-2-yl , 6-(pentafluoroethyl)pyridin-2-yl or 6-(trifluoromethyl)pyridin-2-yl. Especially, R3 Represents 6-(1,1-difluoroethyl)pyridin-2-yl or 6-(trifluoromethyl)pyridin-2-yl. In another preferred embodiment, R3 Represents 6-aminopyridin-2-yl, 6-((C1 -C4 -alkyl)amino)pyridin-2-yl, 6-(di-(C1 -C4 -alkyl)amino)pyridin-2-yl, 6-(pyrrolidin-1-yl)pyridin-2-yl, 6-(hexahydropyridin-1-yl)pyridin-2-yl, 6-( Polin-4-yl)pyridin-2-yl, 6-(4-methylhexahydropyrazin-1-yl)pyridin-2-yl. Especially, R3 Represents 6-aminopyridin-2-yl, 6-(methylamino)pyridin-2-yl, 6-(ethylamino)pyridin-2-yl or 6-(dimethylamino)pyridine- 2-based. Extremely better, R3 Represents 6-aminopyridin-2-yl. In another preferred embodiment, R3 Represents 6-cyclopropylpyridin-2-yl. In another preferred embodiment, R3 Represents 6,7-dihydro-4H-pyrazolo[5,1-c][1,4]oxazin-2-yl, 5-methyl-4,5,6,7-tetrahydropyrazole [1,5-a]pyrazin-2-yl or 4,5,6,7-tetrahydropyrazolo[1,5-a]pyrazin-2-yl, pyrazolo[1,5-a Pyrimidin-3-yl, pyrrolo[2,1-f][1,2,4]triazin-7-yl, pyrrolo[1,2-b]pyridazin-7-yl, thieno[2] ,3-b]pyrazin-7-yl, 5-aminopyrazolo[1,5-a]pyrimidin-3-yl, 2-aminopyrrolo[2,1-f][1,2, 4] Triazin-7-yl, 2-aminopyrrolo[1,2-b]pyridazin-7-yl, 2-aminothieno[2,3-b]pyrazin-7-yl. Especially, R3 Represents 6,7-dihydro-4H-pyrazolo[5,1-c][1,4]oxazin-2-yl, pyrazolo[1,5-a]pyrimidin-3-yl. R4 Preferred examples are cyclopropyl, hexahydropyridin-4-yl, 1-methylhexahydropyridin-4-yl, pyrrolidin-3-yl, 1-methylpyrrolidin-3-yl, aza Cyclobut-3-yl, 1-methylazetidin-3-yl, oxetan-3-yl, tetrahydrofuran-3-yl or tetrahydro-2H-pyran-4-yl. Especially, R4 Represents oxetan-3-yl, tetrahydrofuran-3-yl or tetrahydro-2H-pyran-4-yl. Extremely better, R4 Represents oxetan-3-yl or tetrahydrofuran-3-yl. R4 Another preferred embodiment is C1 -C6 -alkyl. Especially, R4 Represents methyl, ethyl or isopropyl. Extremely better, R4 Represents methyl or ethyl. R4 Another preferred embodiment is C2 -C6 An alkyl group which is substituted by a hydroxyl group. Especially, R4 Representative C2 -C4 An alkyl group which is substituted by a hydroxyl group. Extremely better, R4 Represents 2-hydroxyethyl, 2-hydroxypropyl or 2-hydroxy-2-methylpropyl. Especially, R4 Represents 2-hydroxyethyl. R4 Another preferred embodiment is C2 -C6 An alkyl group substituted with a group selected from the group consisting of C(=O)OMe, C(=O)OEt, C(=O)OH, C(=O)NH2 , SMe, SEt, S(=O)2 Me, S(=O)2 Et, methoxy, ethoxy, N (CH)3 )2 , cyclopropyl, cyclobutyl, cyclopentyl, oxetan-3-yl, tetrahydrofuran-3-yl and tetrahydro-2H-pyran-4-yl. In a particularly preferred embodiment, R4 Represents cyclopropylmethyl, oxetan-3-ylmethyl, 2-(methylsulfonyl)ethyl, 3-(methylsulfonyl)propyl, 2-(methylthio) Ethyl, CH2 C(=O)OEt, CH(CH3 )C(=O)OH or CH2 C(=O)OH, CH2 CH2 C(=O)OH. Extremely better, R4 Represents cyclopropylmethyl or oxetan-3-ylmethyl. In the context of another preferred embodiment, R4 Represents trifluoromethyl, 2,2,2-trifluoroethyl or 2,2-difluoroethyl. Especially, R4 Represents trifluoromethyl. Preferably, R6 Represents hydrogen or fluorine. Especially, R6 Represents hydrogen. Preferably, R8 Represents hydrogen or fluorine. Especially, R8 Represents hydrogen. Preferably, R9 Represents hydrogen or fluorine. Especially, R9 Represents hydrogen. Preferably, R10 Represents hydrogen or fluorine. Especially, R10 Represents hydrogen. Preferably, R12 Represents hydrogen or fluorine. Especially, R12 Represents hydrogen. Preferably, Ra Represents methyl, ethyl or cyclopropyl. Preferably, Rb Represents methyl, ethyl or cyclopropyl. One embodiment of the present invention provides R2 Representative C1 -C8 An alkyl group, which may optionally be mono- or polysubstituted, independently of one another, via the same or different substituents selected from the group consisting of 1 to 5 fluorine atoms, 1 to 3 hydroxyl groups, oxetanyl groups, tetrahydrofuranyl groups. , pyranyl and C1 -C6 - alkoxy, where C1 -C6 - alkoxy groups may be fluorine or via hydroxyl groups or via C(=O)OH, C(=O)Me, C(=O)Et, C(=O)NH2 Monosubstituted to trisubstituted; and substituted R1 And R3 All have the meanings mentioned above. Preferred are compounds of formula (I) wherein R1 Represents chlorine or O-R4 , R2 Representative C1 -C6 An alkyl group, which may optionally be mono- or polysubstituted, independently of one another, via the same or different substituents selected from the group consisting of 1 to 3 fluorine atoms, 1 to 2 hydroxyl groups, oxetanyl groups, tetrahydrofuranyl groups. And C1 -C3 - alkoxy, where C1 -C3 - alkoxy group may be substituted by hydroxyl group or by C(=O)OH, C(=O)Me, C(=O)Et; or R2 Representative 3-sided oxybutyl; R3 Represents a group selected from the following: , where R5 Represents hydrogen, cyclopropyl or C1 -C6 -alkyl, where C1 -C6 - an alkyl group may optionally be mono-substituted to trisubstituted with a cyclopropyl group, a cyano group and a hydroxy group, wherein each substituent may occur only once, or may be mono-substituted to penta-substituted via a fluorine atom;6 Represents hydrogen; or R3 Representative groupWhere R7 Represents hydrogen, cyclopropyl, cyano, NH2 , NH (C1-C6 -alkyl), N(C1 -C6 -alkyl)2 , pyrrolidin-1-yl, hexahydropyridin-1-yl, morpholin-4-yl, 4-methylhexahydropyrazin-1-yl or C1 -C6 -alkyl, where C1 -C6 - an alkyl group may optionally be mono-substituted to trisubstituted with a cyclopropyl group, a cyano group and a hydroxy group, wherein each substituent may occur only once, or may be mono-substituted to penta-substituted via a fluorine atom;8 , R9 , R10 Represents hydrogen or R3 Representative groupWhere R11 Representative C3 -C6 -cycloalkyl or C1 -C6 -alkyl, where C1 -C6 - an alkyl group may optionally be mono-substituted to trisubstituted with a cyclopropyl group, a cyano group and a hydroxy group, wherein each substituent may occur only once, or may be mono-substituted to penta-substituted via a fluorine atom;12 Represents hydrogen or R3 Represents 6,7-dihydro-4H-pyrazolo[5,1-c][1,4]oxazin-2-yl, 5-methyl-4,5,6,7-tetrahydropyrazole [1,5-a]pyrazin-2-yl or 4,5,6,7-tetrahydropyrazolo[1,5-a]pyrazin-2-yl, or represents pyrazolo[1,5 -a]pyrimidin-3-yl, pyrrolo[2,1-f][1,2,4]triazin-7-yl, pyrrolo[1,2-b]pyridazin-7-yl, thieno [2,3-b]pyrazin-7-yl, 5-aminopyrazolo[1,5-a]pyrimidin-3-yl, 2-aminopyrrolo[2,1-f][1, 2,4]triazin-7-yl, 2-aminopyrrolo[1,2-b]pyridazin-7-yl, 2-aminothieno[2,3-b]pyrazin-7-yl ; R4 Represents cyclopropyl, hexahydropyridin-4-yl, 1-methylhexahydropyridin-4-yl, pyrrolidin-3-yl, 1-methylpyrrolidin-3-yl, azetidin-3- , 1-methylazetidin-3-yl, oxetan-3-yl, tetrahydrofuran-3-yl or tetrahydro-2H-pyran-4-yl, wherein oxetane-3- Mono-, tetrahydrofuran-3-yl, tetrahydro-2H-pyran-4-yl may optionally be mono- or disubstituted with methyl, or represent C1 -C6 -alkyl, where C1 -C6 - The alkyl group may be substituted by the following: hydroxy, C(=O)OMe, C(=O)OEt, C(=O)OH, C(=O)NH2 , SMe, SEt, S(=O)2 Me, S(=O)2 Et, methoxy, ethoxy, N (CH)3 )2 , cyclopropyl, cyclobutyl, cyclopentyl, oxetan-3-yl, tetrahydrofuran-3-yl, tetrahydro-2H-pyran-4-yl, or trifluoromethyl, 2,2 , 2-trifluoroethyl or 2,2-difluoroethyl; and pharmaceutically acceptable solvates thereof, non-imagewise isomers, mirror image isomers, metabolites, salts, solvates or salts. Further preferred are compounds of formula (I) wherein R1 Represents chlorine or O-R4 ; R2 Representative C2 -C5 - an alkyl group which may be 1 to 2 hydroxyl groups or via oxetan-3-yl or tetrahydrofuran-3-yl or via group OCH2 C(=O)OH, OCH2 C(=O)OMe, OCH2 CH2 OH or via C1 -C3 - alkoxy substitution, where C1 -C3 - alkoxy group may be substituted by hydroxyl group or by C(=O)OH, C(=O)Me, C(=O)Et; or R2 Represents 3,3,3-trifluoropropyl, 4,4,4-trifluorobutyl or 3-sided oxybutyl; R3 Representative 1-(C1 -C6 -alkyl)-1H-pyrazol-3-yl, wherein C1 -C6 The alkyl substituent may optionally be mono-substituted with a cyclopropyl group, monosubstituted to penta-substituted with a fluorine atom and optionally substituted by a hydroxy group, or represented by 2-cyclopropyl-1,3-thiazol-4-yl or 2- (C1 -C6 -alkyl)-1,3-thiazol-4-yl, wherein C1 -C6 - an alkyl substituent may optionally be monosubstituted by cyclopropyl, monosubstituted to pentasubstituted by fluorine atom and optionally substituted by hydroxy, or as 4-(C)1 -C6 -alkyl)-1,3-thiazol-2-yl, wherein C1 -C6 The alkyl substituent may optionally be monosubstituted by cyclopropyl, monosubstituted to pentasubstituted with fluorine atom and optionally substituted by hydroxy group, or represented by 2-cyclopropyl-1,3-oxazol-4-yl or -(C1 -C6 -alkyl)-1,3-oxazol-4-yl, of which C1 -C6 - an alkyl substituent may optionally be monosubstituted by cyclopropyl, monosubstituted to pentasubstituted with a fluorine atom and optionally substituted by hydroxy, or as 2-(C)1 -C6 -alkyl)-1,3-oxazol-5-yl, of which C1 -C6 - an alkyl substituent may optionally be monosubstituted by cyclopropyl, monosubstituted to pentasubstituted by fluorine atom and optionally substituted by hydroxy, or 6-(C1 -C6 -alkyl)pyridin-2-yl, wherein C1 -C6 - Alkyl substituents may optionally be monosubstituted by cyclopropyl, monosubstituted to pentasubstituted by fluorine atom and optionally substituted by hydroxy group, or represented by 6-aminopyridin-2-yl, 6-(C1 -C4 -alkyl)amino)pyridin-2-yl, 6-(di-(C1 -C4 -alkyl)amino)pyridin-2-yl, 6-(pyrrolidin-1-yl)pyridin-2-yl, 6-(hexahydropyridin-1-yl)pyridin-2-yl, 6-( Phenyl-4-yl)pyridin-2-yl, 6-(4-methylhexahydropyrazin-1-yl)pyridin-2-yl, or 6,7-dihydro-4H-pyrazolo[5 , 1-c][1,4]oxazin-2-yl, 5-methyl-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrazin-2-yl or 4 , 5,6,7-tetrahydropyrazolo[1,5-a]pyrazin-2-yl, or represents pyrazolo[1,5-a]pyrimidin-3-yl, pyrrolo[2,1 -f][1,2,4]triazin-7-yl, pyrrolo[1,2-b]pyridazin-7-yl, thieno[2,3-b]pyrazin-7-yl, 5 -aminopyrazolo[1,5-a]pyrimidin-3-yl, 2-aminopyrrolo[2,1-f][1,2,4]triazin-7-yl, 2-amino group Pyrrolo[1,2-b]pyridazin-7-yl, 2-aminothieno[2,3-b]pyrazin-7-yl; R4 Representative C1 -C4 -alkyl or represents oxetan-3-yl, tetrahydrofuran-3-yl or tetrahydro-2H-pyran-4-yl or represents 2-hydroxyethyl, 3-hydroxypropyl, 2-hydroxypropyl , 2-hydroxy-2-methylpropyl or represents CH2 C(=O)OMe, CH2 C(=O)OEt, CH2 C(=O)OH, CH2 CH2 SMe, 2-(methylsulfonyl)ethyl, 3-(methylsulfonyl)propyl, 2-methoxyethyl, 2-ethoxyethyl, CH2 CH2 N (CH3 )2 , cyclopropylmethyl, cyclobutylmethyl, cyclopentylmethyl, oxetan-3-ylmethyl, tetrahydrofuran-3-ylmethyl, tetrahydro-2H-pyran-4-ylmethyl or Represents trifluoromethyl, 2,2,2-trifluoroethyl or 2,2-difluoroethyl; R5 Represents hydrogen or fluorine; and pharmaceutically acceptable solvates thereof, non-image isomers, mirror image isomers, metabolites, salts, solvates or salts. Also preferred are compounds of formula (I) wherein R1 Represents chlorine or O-R4 ; R2 Representative 3-hydroxypropyl, 3-hydroxybutyl, 2-hydroxyethyl, 3-hydroxy-3-methylbutyl, 4-hydroxybutyl, 4-hydroxypentyl, 2,3-dihydroxypropyl , 2,3-dihydroxy-2-methylpropyl, 2-hydroxypropyl, oxetan-3-ylmethyl, oxetan-3-ylethyl, tetrahydrofuran-3-ylmethyl , tetrahydrofuran-3-ylethyl, 3-oxobutyl, 3-methoxy-3-methylbutyl, 3-methoxypropyl, 2-methoxyethyl, 4,4, 4-trifluorobutyl, 3,3,3-trifluoropropyl, 2-(2-hydroxyethoxy)ethyl, CH2 CH2 OCH2 C(=O)OH, CH2 CH2 OCH2 C(=O)OEt; R3 Represents 1-(difluoromethyl)-1H-pyrazol-3-yl, 1-methyl-1H-pyrazol-3-yl, 1-ethyl-1H-pyrazol-3-yl, 1-( 2,2,2-trifluoroethyl)-1H-pyrazol-3-yl, 1-isopropyl-1H-pyrazol-3-yl, 1-propyl-1H-pyrazol-3-yl, 1-t-butyl-1H-pyrazol-3-yl, 1-isobutyl-1H-pyrazol-3-yl, 2-(trifluoromethyl)-1,3-thiazol-4-yl, 2-methyl-1,3-thiazol-4-yl, 2-ethyl-1,3-thiazol-4-yl, 2-propyl-1,3-thiazol-4-yl, 2-isopropyl -1,3-thiazol-4-yl, 2-tert-butyl-1,3-thiazol-4-yl, 2-cyclopropyl-1,3-thiazol-4-yl, 4-(trifluoromethyl) -1,3-1,3-thiazol-2-yl, 4-methyl-1,3-thiazol-2-yl, 4-ethyl-1,3-thiazol-2-yl, 4-isopropyl-1 , 3-thiazol-2-yl, 4-tert-butyl-1,3-thiazol-2-yl, 4-cyclopropyl-1,3-thiazol-2-yl, 2-methyl-1,3 -oxazol-4-yl, 2-(trifluoromethyl)-1,3-oxazol-4-yl, 2-ethyl-1,3-oxazol-4-yl, 2-(1,1 -difluoroethyl)-1,3-oxazol-4-yl, 2-(2,2,2-trifluoroethyl)-1,3-oxazol-4-yl, 2-isopropyl- 1,3-oxazol-4-yl, 2-tert-butyl-1,3-oxazol-4-yl, 2-cyclopropyl-1,3-oxazol-4-yl, 2-(cyclo Propylmethyl)-1,3-oxazol-4-yl, 2-methyl-1,3-oxazol-5-yl 2-ethyl-1,3-oxazol-5-yl, 2-isopropyl-1,3-oxazol-5-yl, 6-methylpyridin-2-yl, 6-(difluoromethyl Pyridin-2-yl, 6-(trifluoromethyl)pyridin-2-yl, 6-ethylpyridin-2-yl, 6-(1,1-difluoroethyl)pyridin-2-yl, 6 -(pentafluoroethyl)pyridin-2-yl, 6-(2,2,2-trifluoroethyl)pyridin-2-yl, 6-propylpyridin-2-yl, 6-isopropylpyridine- 2-yl, 6-(2-hydroxypropan-2-yl)pyridin-2-yl, 6-t-butylpyridin-2-yl, 6-cyclopropylpyridin-2-yl, 6-aminopyridine -2-yl, 6-(methylamino)pyridin-2-yl, 6-(ethylamino)pyridin-2-yl, 6-(dimethylamino)pyridin-2-yl, 6, 7-Dihydro-4H-pyrazolo[5,1-c][1,4]oxazin-2-yl, 5-methyl-4,5,6,7-tetrahydropyrazolo[1, 5-a]pyrazin-2-yl, 4,5,6,7-tetrahydropyrazolo[1,5-a]pyrazin-2-yl, pyrazolo[1,5-a]pyrimidine- 3-yl, pyrrolo[2,1-f][1,2,4]triazin-7-yl, pyrrolo[1,2-b]pyridazin-7-yl, thieno[2,3- b] pyrazin-7-yl; R4 Represents methyl, ethyl, isopropyl, propyl, cyclopropylmethyl, oxetan-3-yl, oxetan-3-ylmethyl, tetrahydrofuran-3-yl, 2-hydroxyethyl , trifluoromethyl, 2,2,2-trifluoroethyl, 2,2-difluoroethyl, 2-(methylsulfonyl)ethyl, 3-(methylsulfonyl)propyl , 2-(methylthio)ethyl, CH2 C(=O)OEt, CH2 C(=O)OH; R5 Represents hydrogen; and its non-image, isomer, metabolite, salt, solvate or salt solvate. Very particularly preferred are the following compounds: where R1 Represents chlorine or O-R4 ; R2 Representative 3-hydroxypropyl, 3-hydroxybutyl, 2-hydroxyethyl, 3-hydroxy-3-methylbutyl, 2,3-dihydroxypropyl, oxetan-3-ylmethyl, Tetrahydrofuran-3-ylmethyl, 3-oxobutyl, 3-methoxy-3-methylbutyl, 3-methoxypropyl, 2-methoxyethyl, 4,4,4 -trifluorobutyl, 2-(2-hydroxyethoxy)ethyl, CH2 CH2 OCH2 C(=O)OH, CH2 CH2 OCH2 C(=O)OEt; R3 Represents 1-(difluoromethyl)-1H-pyrazol-3-yl, 1-ethyl-1H-pyrazol-3-yl, 1-isopropyl-1H-pyrazol-3-yl, 2- (trifluoromethyl)-1,3-oxazol-4-yl, 2-(trifluoromethyl)-1,3-thiazol-4-yl, 2-cyclopropyl-1,3-oxazole- 4-yl, 2-methyl-1,3-oxazol-5-yl, 2-methyl-1,3-thiazol-4-yl, 4-(trifluoromethyl)-1,3-thiazole- 2-Based, 6-(1,1-difluoroethyl)pyridin-2-yl, 6-(2-hydroxypropan-2-yl)pyridin-2-yl, 6-(difluoromethyl)pyridine- 2-yl, 6-(pentafluoroethyl)pyridin-2-yl, 6-(trifluoromethyl)pyridin-2-yl, 6,7-dihydro-4H-pyrazolo[5,1-c ][1,4]oxazin-2-yl, 6-aminopyridin-2-yl, pyrazolo[1,5-a]pyrimidin-3-yl; R4 Represents methyl, ethyl, isopropyl, cyclopropylmethyl, oxetan-3-yl, oxetan-3-ylmethyl, tetrahydrofuran-3-yl, 2-hydroxyethyl, tri Fluoromethyl, 3-(methylsulfonyl)propyl, 2-(methylthio)ethyl, CH2 C(=O)OEt, CH2 C(=O)OH; and a solvate thereof, a non-image isomer, a mirror image isomer, a metabolite, a salt, a solvate or a salt. Also very popular are the following compounds: where R1 On behalf of O-R4 ; R2 Represents 3-hydroxy-3-methylbutyl; R3 Represents 1-(difluoromethyl)-1H-pyrazol-3-yl, 1-ethyl-1H-pyrazol-3-yl, 2-(trifluoromethyl)-1,3-oxazole-4 -yl, 2-(trifluoromethyl)-1,3-thiazol-4-yl, 2-cyclopropyl-1,3-oxazol-4-yl, 2-methyl-1,3-oxazole -5-yl, 2-methyl-1,3-thiazol-4-yl, 4-(trifluoromethyl)-1,3-thiazol-2-yl, 6-(1,1-difluoroethyl Pyridin-2-yl, 6-(2-hydroxypropan-2-yl)pyridin-2-yl, 6-(difluoromethyl)pyridin-2-yl, 6-(trifluoromethyl)pyridine-2 -yl,6-aminopyridin-2-yl, pyrazolo[1,5-a]pyrimidin-3-yl; R4 Represents methyl, ethyl, isopropyl, cyclopropylmethyl, oxetan-3-yl, oxetan-3-ylmethyl, 2-hydroxyethyl; and their non-image isomers a solvate of a mirror image isomer, a metabolite, a salt, a solvate or a salt. In particular, the invention provides the following compounds: Among the compounds mentioned above, the following compounds are preferred: 1-(difluoromethyl)-N-[2-(3-hydroxy-3-methylbutyl)-6-methoxy- 2H-carbazol-5-yl]-1H-pyrazole-3-carboxamide N-[6-ethoxy-2-(3-hydroxy-3-methylbutyl)-2H-indazole-5 -yl]-2-(trifluoromethyl)-1,3-thiazole-4-carboxamide 1-(difluoromethyl)-N-[6-ethoxy-2-(3-hydroxy-3) -Methylbutyl)-2H-indazol-5-yl]-1H-pyrazole-3-carboxamide N-[6-ethoxy-2-(3-hydroxy-3-methylbutyl) -2H-carbazol-5-yl]-2-methyl-1,3-thiazole-4-carboxamide 2-cyclopropyl-N-[6-ethoxy-2-(3-hydroxy-3) -Methylbutyl)-2H-indazol-5-yl]-1,3-oxazole-4-carboxamide 6-amino-N-[6-ethoxy-2-(3-hydroxy- 3-methylbutyl)-2H-indazol-5-yl]pyridine-2-carboxamide N-[6-ethoxy-2-(3-hydroxy-3-methylbutyl)-2H- Oxazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide N-[2-(3-hydroxy-3-methylbutyl)-6-(oxetan-3 -yloxy)-2H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide N-[2-(3-hydroxy-3-methylbutyl)-6 -(oxetan-3-yloxy)-2H-indazol-5-yl]-2-(trifluoromethyl)-1,3-thiazole-4-carboxamide 1-(difluoromethyl) Base)-N-[2-(3-hydroxy-3-甲Butyl)-6-(oxetan-3-ylmethoxy)-2H-indazol-5-yl]-1H-pyrazole-3-carboxamide 6-(difluoromethyl)- N-[2-(3-Hydroxy-3-methylbutyl)-6-(oxetan-3-ylmethoxy)-2H-indazol-5-yl]pyridine-2-carboxamide N-2-(3-hydroxy-3-methylbutyl)-6-[(3S)-tetrahydrofuran-3-yloxy]-2H-indazole-5-yl-2-methyl-1,3 -oxazol-5-carbamide N-2-(3-hydroxy-3-methylbutyl)-6-[(3S)-tetrahydrofuran-3-yloxy]-2H-indazole-5-yl -6-(trifluoromethyl)pyridine-2-carboxamide 6-amino-N-2-(3-hydroxy-3-methylbutyl)-6-[(3S)-tetrahydrofuran-3-yl Oxy]-2H-indazol-5-ylpyridin-2-carboxamide 6-(difluoromethyl)-N-[6-ethoxy-2-(3-hydroxy-3-methylbutyl) -2H-carbazol-5-yl]pyridine-2-carboxamide N-[2-(3-hydroxy-3-methylbutyl)-6-(oxetan-3-ylmethoxy) -2H-carbazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide N-[2-(3-hydroxy-3-methylbutyl)-6-methoxy -2H-carbazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide N-[6-(2-hydroxyethoxy)-2-(3-hydroxy-3-methyl Benzyl)-2H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide 6-(1,1-difluoroethyl)-N-[6-ethoxy Benzyl-2-(3-hydroxy-3-methylbutyrate -2H-carbazol-5-yl]pyridine-2-carboxamide N-[6-ethoxy-2-(3-hydroxy-3-methylbutyl)-2H-indazole-5- 1-ethyl-1H-pyrazole-3-carboxamide. The compounds of the invention act as inhibitors of IRAK4 kinase and have a surprisingly useful spectrum of pharmacological activity. Thus, in addition to the subject matter mentioned above, the invention also provides the use of a compound of the invention for the treatment and/or prevention of diseases in humans and animals. Particularly preferred is the use of the IRAK4 inhibitors of the invention to treat and/or prevent gynecological conditions, inflammatory skin conditions, cardiovascular disorders, pulmonary disorders, ocular disorders, autoimmune disorders, pain disorders, metabolic disorders, gout, liver Conditions, metabolic syndrome, insulin resistance and cancer. The compounds of the present invention are suitable for the prevention and/or treatment of various conditions and disease-related conditions, particularly those directly mediated by IRAK4, which are mediated by TLR (except TLR3) and/or the IL-1 receptor family and/or pathology. IRAK4-related conditions include multiple sclerosis, atherosclerosis, myocardial infarction, Alzheimer's disease, viral-induced myocarditis, gout, Gert-Koyanagi-Harada syndrome, lupus erythematosus, psoriasis, spondylarthritis And arthritis. The compounds of the invention are also useful for the prevention and/or treatment of conditions mediated by MyD88 and TLR (except TLR3). This includes multiple sclerosis, rheumatoid arthritis, spondylarthritis (especially psoriatic spondyloarthritis and Behdelev's disease), metabolic syndrome (including insulin resistance, diabetes), osteoarthritis, and Schüegen's syndrome , giant cell arteritis, sepsis, polymyositis and dermatomyositis, skin disorders (eg psoriasis, atopic dermatitis, alopecia areata, abnormal acne and acne vulgaris), lung disorders (eg pulmonary fibrosis, chronic Obstructive Pulmonary Disease (COPD), Acute Respiratory Syndrome (ARDS), Acute Lung Injury (ALI), Interstitial Lung Disease (ILD), Sarcoma-like Disease, and Pulmonary Hypertension). Due to the mechanism of action of the compounds of the invention, it is suitable for the prevention and/or treatment of TLR-mediated disorders such as Behcet's disease, gout, endometriosis and endometriosis-related pain and other endometrium Ectopic-related symptoms (such as dysmenorrhea, painful intercourse, difficulty urinating, and difficulty in defecation). In addition, the compounds of the invention are suitable for the prevention and/or treatment of transplant rejection, lupus erythematosus, adult onset Stil's disease and chronic inflammatory bowel disorders (eg, ulcerative colitis and Crohn's disease). In addition to the conditions listed, the use of the compounds of the invention is also suitable for the treatment and/or prevention of ocular conditions such as keratitis, allergic conjunctivitis, keratoconjunctivitis sicca, macular degeneration and uveitis; Vascular disorders such as atherosclerosis, myocardial reperfusion injury, myocardial infarction, hypertension, and neurological disorders (eg, Alzheimer's disease, stroke, and Parkinson's disease). The mechanism of action of the compounds of the invention also enables the prevention and/or treatment of liver disorders mediated by the TLR and IL-1 receptor families, particularly NAFLD, NASH, ASH, liver fibrosis and cirrhosis. The compounds of the invention may also prevent and/or treat pruritus and pain, particularly acute, chronic, inflammatory and neuropathic pain. Due to the mechanism of action of the compounds of the invention, they are suitable for the prevention and/or treatment of neoplastic disorders such as lymphoma, chronic lymphoid leukemia, melanoma and hepatocellular carcinoma, breast cancer and Ras dependent tumors. Furthermore, the compounds of the invention are suitable for the treatment and/or prevention of conditions mediated by the IL-1 receptor family. These conditions include CAPS (Hidden Heat Protein Related Cycle Syndrome) (including FCAS (familial cold autologous inflammatory syndrome), MWS (Mu-Wei's syndrome), NOMID (neonatal onset multi-system inflammatory disease) and CONCA (chronic infant, nerve, skin and joint syndrome), FMF (familial Mediterranean fever), HIDS (high IgD syndrome), TRAPS (tumor necrosis factor receptor 1 related cycle syndrome), juvenile idiopathic arthritis , adult onset Stil's disease, adalter-Bercher's disease, rheumatoid arthritis, psoriasis, arthritis, Behdejelev's disease, osteoarthritis, keratoconjunctivitis sicca and Sjogren's syndrome , multiple sclerosis, lupus erythematosus, alopecia areata, type 1 diabetes, type 2 diabetes, and sequelae of myocardial infarction. Pulmonary disorders (eg, asthma, COPD, idiopathic interstitial pneumonia and ARDS), gynecological conditions (eg endometriosis and endometriosis-related pain and other endometriosis) Symptoms (eg, dysmenorrhea, painful intercourse, dysuria, and difficulty in defecation), chronic-inflammatory bowel disorders (eg, Crohn's disease and ulcerative colitis) are associated with disorders of the IL-1 receptor family and are suitable for treatment and/or The compounds of the invention are used prophylactically. The compounds of the invention are also useful for the treatment and/or prevention of neurological disorders mediated by the IL1 receptor family (eg, stroke, Alzheimer's disease, traumatic brain injury) and dermatological conditions (eg, psoriasis, atopic dermatitis, Abnormal acne, alopecia areata and allergic contact dermatitis). Additionally, the compounds of the invention are suitable for the treatment and/or prevention of pain conditions, especially acute, chronic, inflammatory and neuropathic pain. This preferably includes hyperesthesia, touch pain, arthritic pain (eg osteoarthritis, rheumatoid arthritis and spinal arthritis), premenstrual pain, endometriosis-related pain, post-operative pain, interstitial Cystitis pain, CRPS (complex regional pain syndrome), trigeminal neuralgia, prostatitis pain, pain caused by spinal cord injury, inflammation-induced pain, lumbar pain, cancer pain, chemotherapy-related pain, HIV treatment induction Neuropathy, burn-induced pain, and chronic pain. The invention also further provides a method of treating and/or preventing a condition, particularly a condition as mentioned above, using an effective amount of at least one of the compounds of the invention. In the context of the present invention, the term "treatment" or "treating" includes inhibiting, delaying, suppressing, alleviating, attenuating, limiting, reducing, inhibiting, regressing or curing a disease, condition, condition, injury or health problem, or The occurrence, duration or progression of symptoms of such states and/or such states. The term "therapy" is understood herein to be synonymous with the term "treatment." The terms "prevention, prophylaxis and "prevention" are used synonymously in the context of the present invention and refer to avoiding or reducing infection, experience, suffering or suffering from a disease, condition, disorder, injury or health problem or such condition and/or Or the risk of the occurrence or progression of symptoms of such conditions. The disease, condition, condition, injury or health problem can be treated or prevented in part or in whole. The compounds of the invention may be used alone or in combination with other active compounds if desired. The invention further provides an agent comprising at least one of the compounds of the invention and one or more additional active compounds, in particular for use in the treatment and/or prevention of the conditions mentioned above. Preferred examples of suitable active compounds include: The following active compounds are generally mentioned: for example, antibacterial substances (for example, penicillins, vancomycin, ciprofloxacin), antiviral substances (eg aciclovir, oseltamivir) and antifungal substances (eg naftifin, nystatin) and gamma immunoglobulin), immunomodulation and immunity Inhibitory compounds (eg cyclosporin, Methotrexat®, TNF antagonists (eg Humira®, etanercept, infliximab), IL-1 inhibitors (eg anakinbine) Anakinra, Canakinumab, Rilonacept, phosphodiesterase inhibitors (eg Apremilast), Jak/STAT inhibitors (eg tofacitinib) (Tofacitinib), Baricitinib, GLPG0634), leflunomid, cyclophosphamide, rituximab, belimumab, tacrolimus (tacrolimus), rapamycin, mycophenolate mofetil, dry Interferon, corticosteroids (eg, prednisone, prednisolone, methylpresino, hydrocortisone, betamethasone, cyclophosphamide, Azathioprine and sulfasalazine; acetaminophen, non-steroidal anti-inflammatory substances (NSAIDS) (aspirin, ibuprofen, naproxen) , etodolac, celecoxib, colchicine. For tumor therapy, the following should be mentioned: immunotherapy (eg, aldesleukin, alemtuzumab, basiliximab, catusmaxomab, west) Celmoleukin, denileukin-diftitox, eculizumab, edrecolomab, gemtuzumab, temimumomab -tiuxetan), imiquimod, interferon-α, interferon-β, interferon-γ, ipilimumab, lenalidomid, lenograstim , mifamurtid, ofatumumab, oprelvekin, picibanil, plerixafor, polysaccharide-K, sagstatin Sargramostim), sipuleucel-T, tasonermin, teceleukin, tocilizumab, antiproliferative substances such as, but not limited to, ampoule Alpha (amsacrine), agrarabi (arglabin), arsenic trioxide, aspartate, bleomycin, busulfan An), actinomycin D, docetaxel, epirubicin, peplomycin, trastuzumab, rituximab, Obutuzuzumab, ofatumumab, tositumomab, aromatase inhibitors (eg exemestane, fadrozole, formamide) (formestane), letrozole, anastrozole, vorozole, antiestrogens (eg chlormadinone, fulvestrant, melamine) Mepitiostane), tamoxifen, raloxifen, toremifen, estrogen (eg estradiol, poly-estradiol phosphate), progesterone (eg methymidine) Progesterone (medroxyprogesteron), megestrol (megestrol), topoisomerase I inhibitor (eg irinotecan, topotecan), topoisomerase II inhibitor (eg amrubicin, daunorubicin, elliptinium acetate, etoposide, idarubi (idarubi) Cin), mitoxantrone, teniposide, microtubule actives (eg cabazitaxel, eribulin, paclitaxel, vinblastine) Vinblastine), vincristine, vindesine, vinorelbine, telomerase inhibitors (eg imetelstat), alkylated substances and tissue proteins Enzyme inhibitors (eg, bendamustine, carmustine, chlormethine, dacarbazine, estramustine, ifosfamid) , lomustine, mitobronitol, mitolactol, nimustine, prednimustine, procarbazine, Ramimustine, streptozotocine, temozolomide, thiotepa, treosulfan, trofosfamid, vorinostat ), romidepsin, panobinostat; affecting cells Substances of the differentiation process, such as abarelix, aminoglutethimide, bexarotene, MMP inhibitors (peptide mimetica, non-peptide mimetics, and tetracycline) For example, Marimastat, BAY 12-9566, BMS-275291, clodronic acid, prinomastat, doxycycline, and mTOR inhibitors (eg sirolimus) Sirolimus), everolimus, temsirolimus, zotarolimus, antimetabolites (eg clofarabine, deoxyuridine, amine methotrexate) (methotrexate), 5-fluorouracil, cladribine, cytarabine, fludarabine, sputum, pemetrexed, raltitrexed, Tegafur, thioguanine, platinum compounds (eg carboplatin, cisplatin, cisplatinum, eptaplatin, lobaplatin, Milli Platinum (miriplatin), nedaplatin, oxaliplatin; anti-angiogenesis Compound (such as bevacizumab), antiandrogen compound (such as bevacizumab, enzalutamide, flutamide, nilutamide, ratio Bicalutamide, cyproterone, cyproterone acetate, proteasome inhibitors (eg bortezomib, carfilzomib, opozomib) , ONYX0914), gonadotropin-releasing agonists and antagonists (eg, abarelix, buserelin, deslorelin, ganirelix, Gosher Goserelin, histrelin, triptorelin, degarelix, leuprorelin, methionine aminopeptidase inhibitor ( For example, benamide derivatives, TNP-470, PPI-2458), heparanase inhibitors (eg SST0001, PI-88); inhibitors of genetically modified Ras proteins (eg farnesyltransferase) Inhibitors, such as lonafarnib, tipifarnib, HSP90 inhibitors (eg, geldamycin derivatives, For example, 17-acrylamide geldanamycin, 17-desmethoxygeldanamycin (17AAG), 17-DMAG, retaspimycin hydrochloride, IPI-493, AUY922, BIIB028, STA -9090, KW-2478), spindle kinesin inhibitors (eg SB715992, SB743921, pentamidine/chlorpromazine), MEK (mitogen-activated protein kinase kinase) inhibitors (eg song Trametinib, BAY 86-9766 (refametinib), AZD6244), kinase inhibitors (eg, sorafenib, regorafenib, lapatinib) (lapatinib), sutent, dasatinib, cetuximab, BMS-908662, GSK2118436, AMG 706, erlotinib, gefitinib Gefitinib), imatinib, nilotinib, pazopanib, roniciclib, sunitinib, vandetanib, Vemurafenib), hedgehog signaling inhibitors (eg cyclopamine, vismodegib), BTK (brutton tyramine) Inhibitors of the kinase (Bruton's tyrosine kinase) (eg, ibrutinib), JAK/pan-JAK (Janus kinase) inhibitors (eg, SB-1578, baricitinib) , tofacitinib, pacitinib, momolotinib, ruxotinib, VX-509, AZD-1480, TG-101348), PI3K inhibitor ( For example, BAY 1082439, BAY 80-6946 (copanlisib), ATU-027, SF-1126, DS-7423, GSK-2126458, buparlisib, PF-4691502, BYL-719, XL- 147, XL-765, idelalisib), SYK (spleen tyrosine kinase) inhibitors (eg, fostamatinib, Excellair, PRT-062607), p53 gene therapy, double Phosphonates (eg etidronate, clodronate, tiludronate, pamidronate, alendronic acid, ibandronate) Ibandronate, risedronate, zoledronate. For the combination, the following active compounds should also be mentioned, such as, but not limited to, rituximab, cyclophosphamide, doxorubicin, a combination of doxorubicin and estrone, vincristine, Chlorambucil, fludarabine, dexamethasone, cladribine, praisson, 131I-chTNT, abirateron, aclarubicin, aqu Alitretinoin, bisantrene, leucovorin, calcium leucovorin, capecitabin, carmofur, clodronate, romiplostim, gram Crisantaspase, darbepoetin alfa, decitabin, denosumab, dibrospidium chloride, eltrombopag ), endostatin, epitiostanol, epoetin alfa, filgrastim, fotemustin, gallium nitrate, gemcitabine, gru Glutoxim, histamine dihydrochloride, hydroxyurea, improsulfan, ixabepilon, Lerectin, lentinan, levamisole, lisurid, lonidamin, masoprocol, methyl ketone, methoxsalen, amine Methyl ketone (methyl aminolevulinate), miltefosin, mitoguazon, mitomycin, mitotan, nelarabin, nimotuzumin Anti-nitricium, nitracilin, omeprazol, palifermin, panitumumab, parmozyme, PEG epotine beta (methoxy-PEG) Epoetin β), polyethylene glycol filgrastim, peginterferon alfa-2b, pentazocin, pentostatin, perfosfamid, pyrrole Pilarubicin, plicamycin, poliglusam, porfimer-sodium, pralatrexate, quinagolid, razori ( Razoxan), sizofiran, sobuzuxan, sodium glycididazole, tamibarroten, tegafur and gem Combination of gimeracil and orteracil, testosterone, tetrofosmin, thalidomide, thymalfasin, trabectedin, retinoic acid , trilostan, tryptophan, ubenimex, vapreotid, 钇-90 glass microbeads, zristatin, netstatin (zinostatin) Stimalamer). Also suitable for tumor therapy such as chemotherapy (eg, azacitidine, belototecan, enocitabine, melphalan, valrubicin) , vinflunin, zorubicin, radiation therapy (eg, I-125 grain, palladium-103 grain, radium chloride-223) or phototherapy (eg temoporfin) a combination of non-pharmacological therapies such as talaporfin, which is accompanied by a drug that utilizes the IRAK4 inhibitor of the present invention, or in a non-drug tumor therapy (eg, chemotherapy, radiation therapy, or phototherapy) After the end, it is supplemented by a drug treatment using the IRAK4 inhibitor of the present invention. In addition to those mentioned above, the IRAK4 inhibitors of the invention may also be combined with the following active compounds: active compounds for Alzheimer's therapy, such as acetylcholinesterase inhibitors (eg multiple Donepezil, rivastigmine, galantamin, tacrine, NMDA (N-methyl-D-aspartate) receptor antagonism Agent (eg memantine); L-DOPA/carbidopa (L-3,4-dihydroxyphenylalanine), COMT (catechol-O-A) for the treatment of Parkinson's disease Base transferase inhibitors (eg, entacapon), dopamine agonists (eg, ropinrol, pramipexol, bromocriptine), MAO-B (monoamine) Base oxidase-B) inhibitors (such as selegiline), anticholinergic agents (such as trihexyphenidyl), and NMDA antagonists (such as amantadin); Multiple sclerosis beta interferon (IFN-β) (eg IFN β-1b, IFN β-1a Avonex® and Betaferon®), glatiramer acetate, immunoglobulin, natalizumab ( Natalizumab), fingolimod and immunosuppressants (such as mitoxantrone, azathioprine and cyclophosphamide); substances used to treat pulmonary disorders, such as beta-2-sympathomimetics ( For example, salbutamol, anticholinergic agents (such as pyrrose), methylxanthine (such as theophylline), leukotriene receptor antagonists (such as montelukast), PDE-4 (4) Type phosphodiesterase inhibitors (eg, roflumilast), amine methotrexate, IgE antibodies, azathioprine and cyclophosphamide, cortisol-containing preparations; substances for the treatment of osteoarthritis, For example, non-steroidal anti-inflammatory substances (NSAIDs). In addition to the two therapies mentioned, for rheumatoid conditions (such as rheumatoid arthritis, spondylarthritis and juvenile idiopathic arthritis), mention should be made of methotrexate and for B cell and T cell therapy. Biological agents (eg rituximab, abatacept). Neurotrophic substances such as acetylcholinesterase inhibitors (eg, donepezil), MAO (monoamine oxidase) inhibitors (eg, selegiline), interferons, and anticonvulsants (eg gabapentin) Gabapentin)); an active compound for the treatment of cardiovascular disorders, such as beta blockers (eg metoprolol), ACE inhibitors (eg benazepril), vasoconstrictor receptor blockers Broken agents (such as losartan, valsartan), diuretics (such as hydrochlorothiazide), calcium channel blockers (such as nifedipine), statins (such as Simvastatin, fluvastatin; antidiabetic drugs such as metformin, glinide (eg nateglinide), DPP-4 (dipeptidyl peptidase) -4) Inhibitors (eg, linagliptin, saxagliptin, sitagliptin, vildagliptin), SGLT2 (sodium/glucose co-transporter 2) Inhibitor/gliflozin (eg dapagliflozin, empagliflozin), intestine Insulinotropic mimetics (hormone glucose-dependent insulinotropic peptide (GIP) and glycoside-like peptide 1 (GLP-1) analogues/agonists) (eg exenatide, liraglutide) ), lixisenatide, alpha-glucosidase inhibitors (eg, acarbose, miglitol, voglibiose) and sulfonylurea (eg Glibenclamide, tolbutamide, insulin sensitizers (eg pioglitazone) and insulin therapy (eg NPH insulin, insulin lispro) for the treatment of hypoglycemia A substance used to treat diabetes and metabolic syndrome. Lipid lowering agents, such as fibrates (eg, bezafibrate, etofibrate, fenofibrate, gemfibrozil), niacin-derived (eg niacin/laropiprant), ezetimib, statins (eg simvastatin, fluvastatin), anion exchangers (eg colestyramine) , colestipol, colesevelam). Active compounds for the treatment of chronic inflammatory bowel disorders, such as mesalazine, sulfasalazine, azathioprine, 6-oxime or amidoxime, probiotics (Mutaflor) , VSL#3®, Lactobacillus GG, Lactobacillus plantarum, L. acidophilus, L. casei, Bifidobacterium infantis 35624 , Enterococcus fecium SF68, Bifidobacterium longum, Escherichia coli Nissle 1917, antibiotics (eg ciprofloxacin and metronidazole), antibiotics Diarrhea (such as loperamide) or laxative (bisacodyl). Immunosuppressive agents for the treatment of lupus erythematosus, such as glucocorticoids and non-steroidal anti-inflammatory substances (NSAID), cortisone, chloroquin, cyclosporine, azathioprine, belimizumab, rituximab Infliximab, cyclophosphamide. Drugs for organ transplantation, such as, but not limited to, calcineurin inhibitors (such as tacrolimus and cyclosporine), cell division inhibitors (such as azathioprine, mycophenolate mofetil, mildew) Mycophenolic acid, everolimus or sirolimus, rapamycin, basiliximab, daclizumab, anti-CD3 antibody, anti-T-lymphocyte immunoglobulin /Anti-lymphocyte immunoglobulin. Vitamin D3 analogues for dermatological conditions, such as calcipotriol, tacalcitol or calcitriol, salicylic acid, urea, cyclosporine, methotrexate, legally Elemizumab. For the treatment and/or prophylaxis of the above-mentioned conditions, reference should also be made to inclusion of at least one of the compounds of the invention and one or more other active compounds (specifically EP4 inhibitors (prostaglandin E2 receptor 4 inhibition) Agent, P2X3 inhibitor (P2X嘌呤 receptor 3), PTGES inhibitor (prostaglandin E synthase inhibitor) or AKR1C3 inhibitor (aldosterone reductase family 1 member C3 inhibitor)). The compounds of the invention may act systemically and/or locally. For this purpose, it can be administered in a suitable manner, for example by oral, parenteral, transpulmonary, nasal, sublingual, translingual, buccal, rectal, transdermal, transdermal, or conjunctival routes. Through the ear or as an implant or stent. The compounds of the invention may be administered in a form suitable for administration in such administration routes. Suitable forms for oral administration include the following: working according to the prior art and releasing the compound according to the invention rapidly and/or in a modified manner and containing the invention in crystalline and/or amorphous and/or dissolved form A compound, such as a lozenge (uncoated or coated lozenge, for example, an anti-gastric solution or a delayed dissolution or insoluble coating having a release controlling the release of a compound of the invention), a tablet or film that is rapidly disintegrating in the oral cavity Agents, films/lyophiles, capsules (such as hard or soft gelatin capsules), sugar-coated lozenges, granules, pills, powders, emulsions, suspensions, aerosols or solutions. Parenteral administration may avoid reabsorption steps (eg, by intravenous, intraarterial, intracardiac, intraspinal or intralumbar route) or include resorption (eg, by intramuscular, subcutaneous, intradermal, transdermal or The intraperitoneal route) is achieved. Formulations suitable for parenteral administration include injection and infusion preparations in the form of solutions, suspensions, emulsions, lyophilizates or sterile powders. Suitable examples of other routes of administration are inhalation medicaments (including powder inhalers, nebulizers), nasal drops, solutions or sprays; for lozenges, sublingual or buccal administration of lozenges, films/blinds or Capsules, suppositories, ear or eye preparations, vaginal capsules, aqueous suspensions (lotions, oscillating mixtures), lipophilic suspensions, ointments, creams, transdermal therapeutic systems (eg patches), milk, pastes, foams, Powder, graft or stent. Preferred are administered orally or parenterally, especially by oral administration. The compounds of the invention can be converted to the stated dosage forms. This can be achieved in a manner known per se by mixing with inert, non-toxic, pharmaceutically suitable auxiliaries. Such excipients include carriers (eg, microcrystalline cellulose, lactose, mannitol), solvents (eg, liquid polyethylene glycol), emulsifiers, and dispersing or wetting agents (eg, sodium lauryl sulfate, poly Oxysorbol oleate), binders (eg polyvinylpyrrolidone), synthetic and natural polymers (eg albumin), stabilizers (eg oxidizing agents such as ascorbic acid), colorants (eg inorganic pigments, eg oxidation Iron) and flavoring agents and / or flavoring agents. The invention further provides medicaments comprising at least one of the compounds of the invention, usually together with one or more inert, non-toxic, pharmaceutically suitable excipients, and their use for the above mentioned purposes. In general, in the case of parenteral administration, it is advantageous to administer from about 0.001 mg/kg body weight to 1 mg/kg body weight, preferably from about 0.01 mg/kg body weight to 0.5 mg/kg body weight, to achieve effective results. . In the case of oral administration, the dosage is from about 0.01 mg/kg body weight to 100 mg/kg body weight, preferably from about 0.01 mg/kg body weight to 20 mg/kg body weight, and optimally from 0.1 mg/kg body weight to 10 mg. /kg body weight. However, deviations from such amounts may be necessary in some instances, particularly as a function of body weight, route of administration, reaction of the individual to the active compound, the nature of the formulation, and the time or interval at which the administration takes place. Therefore, in some cases, it may be sufficient to control less than the minimum amount mentioned above, while in other cases the upper limit mentioned must be exceeded. In the case of administration of larger amounts, these may be appropriately divided into individual doses and administered throughout the day. The following working examples illustrate the invention. The invention is not limited to the examples. Unless otherwise stated, the percentages in the following tests and examples are percentages by weight; parts are parts by weight. The solvent ratio, dilution ratio and concentration data of the liquid/liquid solution are in each case based on volume. The preparation of the substance of the formula (I) of the present invention is illustrated in the following Synthesis Schemes 1 to 6. As shown in Synthesis Scheme 1, 6-chloro-5-nitro-1H-carbazole (CAS 101420-98-8) can be converted to the intermediate 1 by an alkylation reaction or a Mitsunobu reaction. The reactants suitable for the alkylation reaction are optionally substituted alkyl chlorides, alkyl bromides, alkyl iodides or alkyl 4-toluenesulfonates. The alkyl halide or alkyl 4-toluenesulfonate used may be commercially available or may be prepared in a manner similar to that known from the literature (in the preparation of alkyl 4-toluenesulfonate, one An example is the reaction of a suitable alcohol with 4-toluenesulfonyl chloride in the presence of triethylamine or pyridine; see, for example, Bioorganic and Medicinal Chemistry, 2006, 14, 12 4277 - 4294). Optionally, an alkali metal iodide such as potassium iodide or sodium iodide may be added in the case of using an alkyl chloride or an alkyl bromide. The base used may be, for example, potassium carbonate, cesium carbonate or sodium hydride. In the case of a reactive alkyl halide, N-cyclohexyl-N-methylcyclohexylamine can also be used in some cases. Useful solvents include, for example, 1-methylpyrrolidin-2-one, DMF, DMSO or THF. Optionally, the alkyl halide or alkyl 4-toluenesulfonate used has a functional group which has been previously protected by a protecting group (see also P. G. M. Wuts, T. W. Greene,Greene's Protective Groups in Organic Synthesis , Fourth Edition, ISBN: 9780471697541). If, for example, an alkyl halide having one or more hydroxyl groups or an alkyl 4-toluenesulfonate is used, the hydroxyl groups may be familiar to the third butyl (dimethyl) oxime familiar to those skilled in the art. Protected by a base or similar hydrazine-containing protecting group. Alternatively, the hydroxy group can also be protected with a tetrahydro-2H-pyran (THP) group or with an acetamino group or a benzamidine group. The protecting group used can then be removed after synthesis of intermediate 1 or intermediate 2, or after synthesis of (I)-a. If, for example, a t-butyl(dimethylhydrazino) group is used as the protecting group, tetrabutylammonium fluoride can be used, for example, in a solvent such as THF to be liberated. The THP protecting group can be detached using, for example, 4-toluenesulfonic acid, optionally in the form of a monohydrate. The acetamino group or the benzamidine group can be detached by treatment with an aqueous solution of sodium hydroxide. The alkyl halide or alkyl 4-toluenesulfonate employed may optionally contain a functional group which can be converted to an alternative functional group by a reaction known to those skilled in the art. The examples mentioned are oxidation or reduction reactions or hydrolysis reactions of alkyl carboxylates (for example comparison)Science of Synthesis , Georg Thieme Verlag). Alternatively, 6-chloro-5-nitro-1H-indazole can be converted to a 6-chloro-5-nitro-1H-carbazole using an optionally substituted alkyl alcohol in a light delay reaction (see, for example, KCK Swamy et al. Chem. Rev. 2009, 109, 2551-2651). Intermediate 1. Various phosphines such as triphenylphosphine, tributylphosphine or 1,2-diphenylphosphinoethane can be used with diisopropyl azodicarboxylate (CAS 2446-83-5) or in the literature Combinations of other diazene derivatives mentioned (KCK Swamy et al. Chem. Rev. 2009, 109, 2551-2651). Preferred are triphenylphosphine and diisopropyl azodicarboxylate. If the alkyl alcohol has a functional group, a known protecting group strategy can be used (as in the case of the reaction with an alkyl halide as mentioned above) (for additional instructions see PGM Wuts, TW Greene, Greene's Protective Groups in Organic Synthesis, Fourth Edition, ISBN: 9780471697541) and an oxidation or reduction step can be carried out after synthesis of intermediate 1 or after synthesis of intermediate 2 or after synthesis of a compound of formula (I) according to the invention (as in the above) Reference is made to the case of reaction with an alkyl halide). Intermediate 1 can be converted to intermediate 2 by reduction of the nitro group. Iron and ammonium chloride in water and ethanol can be used for this purpose (see also, for example, Journal of the Chemical Society, 1955, 2412-2419). To prepare a subset (I)-a of the compound of formula (I) of the present invention from intermediate 2, it can be synthesized using indoleamine. Can be used in the literature (Amino Acids, Peptides and Proteins in Organic Chemistry, Vol. 3 - Building Blocks, Catalysis and Coupling Chemistry, Andrew B. Hughes, Wiley, Chapter 12 - Peptide-Coupling Reagents, 407-442; Chem. Soc. Rev., 2009, 38, 606) Various coupling agents are known. For example, 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride can be used with 1-hydroxy-1H-benzotriazole hydrate (HOBt, WO2012107475; Bioorg Med. Chem. Lett., 2008, 18, 2093), tetrafluoroboric acid (1H-benzotriazol-1-yloxy)(dimethylamino)-N,N-dimethylmethylammonium salt (TBTU, CAS 125700-67-6), hexafluorophosphoric acid (dimethylamino)-N,N-dimethyl (3H-[1,2,3]triazolo[4,5-b]pyridine -3-yloxy)methylammonium salt (HATU, CAS 148893-10-1), propanephosphonic anhydride (formed as a solution in ethyl acetate or DMF, CAS68957-94-8) or bis-1H-imidazole- A combination of 1-methyl ketone (CDI) is used as a coupling agent, and a base such as triethylamine or N-ethyl-N-isopropylpropan-2-amine can be added to the reaction mixture in each case. Preferred are HATU and N-ethyl-N-isopropylpropan-2-amine used in DMF.Synthetic Scheme 1 A subset (I)-a of a compound of formula (I) of the present invention (wherein R1 Preparation of = Cl). R2 And R3 It is as defined for the general formula (I). Another subset (I)-b of the compounds of formula (I) of the present invention can be obtained as shown in Synthetic Scheme 2. Here, 6-chloro-1H-indazole-5-amine (CAS 221681-75-0) was reacted with a carboxylic acid. The method of indoleamine synthesis described in Synthetic Scheme 1 is suitable for this purpose. Preferred are 1-hydroxy-1H-benzotriazole hydrate, 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride and triethylamine in THF. . This gave intermediate 3 which was then reacted with an alkyl halide in a similar manner to that of the scheme 1 to afford (I)-b.Synthetic Scheme 2 A subset (I)-b of a compound of formula (I) of the present invention (wherein R1 Preparation of = Cl). R2 And R3 It is as defined for the general formula (I). Another subset (I)-c of the compounds of formula (I) of the present invention can be obtained as shown in Synthetic Scheme 3. Here, 5-nitro-1H-indazole-6-ol (CAS 1082041-56-2) was converted to the intermediate 4. The preparation of some intermediates 4 is known (for example, WO2013174744, WO201574986, Bioorganic and Medicinal Chemistry, 2004, 12, 2115-2137). A preferred retardation reaction for the preparation of intermediate 4 from 5-nitro-1H-indazole-6-ol (Comparative Synthesis Scheme 1). More preferred are triphenylphosphine and diisopropyl azodicarboxylate in THF. Starting from intermediate 4, intermediate 5 is obtained by reduction reaction (comparative synthesis 1 for iron and ammonium chloride used in water and ethanol; however, carbon-supported palladium in a hydrogen atmosphere can also be used (WO2015/74986 Or tin (II) chloride in methanol (compare WO2013174744)). Indoleamine synthesis as described in Synthetic Scheme 1 yields Intermediate 6. Similar to the synthesis scheme 1, the alkylation reaction gives (I)-c.Synthetic Scheme 3 A subset (I)-c of a compound of formula (I) of the present invention (wherein R1 = OR4 Preparation of .R2 , R3 And R4 It is as defined for the general formula (I). Another subset (I)-d of the compounds of formula (I) of the present invention can be obtained as shown in Synthetic Scheme 4. Intermediate 4 as set forth in Scheme 1 is converted to Intermediate 7 in an alkylation reaction. Reduction of the nitro group (Comparative Synthesis Scheme 4) yielded intermediate 8, which was then converted to (I)-d by indoleamine synthesis (Comparative Scheme 1).Synthetic Scheme 4 A subset (I)-d of a compound of formula (I) of the present invention (wherein R1 = OR4 Preparation of. R2 , R3 And R4 It is as defined for the general formula (I). Intermediate 8 can be prepared from intermediate 5 by an alternative route as set forth in Scheme 5. The intermediate 5 was first protected with a third butoxycarbonyl group (comp. WO2015091426) and thereafter alkylated as described in Synthetic Scheme 1 and the protecting group was removed in the presence of trifluoroacetic acid in dichloromethane.Synthetic Scheme 5 Alternative Preparation of Intermediate 8. R2 And R4 It is as defined for the general formula (I). Substituent R can also be introduced by alkylation of an alkyl halide or an alkyl 4-toluenesulfonate starting from intermediates 11, 12 and 13 according to Scheme 6.4 This allows the intermediate 6 (subsets (I)-e) of the compound of the formula (I) of the invention and the intermediate 10 to be obtained. Preferred herein is the alkylation reaction using alkyl bromide and potassium carbonate in DMF. The alkyl halide used may optionally have a functional group which can be converted to other functional groups by a transformation known to those skilled in the art after the alkylation reaction. An example which may be mentioned is the reduction of the alkyl carboxylate group to an alcohol group using sodium borohydride or the hydrolysis of the alkyl carboxylate to a carboxylic acid group using lithium hydroxide in water and THF.Synthetic Scheme 6 A subset (I)-e of a compound of formula (I) of the invention (wherein R1 = OR4 Preparation of the intermediates and preparation of intermediates 6 and 10. R2 , R3 And R4 It is as defined for the general formula (I).Synthesis of example compounds Abbreviations and descriptions The term saturated brine means a saturated aqueous solution of sodium chloride. Chemical names for intermediates and examples were generated using ACD / LABS (Batch 12.01.) software.method In some cases, the compounds of the invention and their precursors and/or intermediates are analyzed by LC-MS. Method A1: UPLC (MeCN-HCOOH): Instrument: Waters Acquity UPLC-MS SQD 3001; Column: Acquity UPLC BEH C18 1.7 50 x 2.1 mm; Mobile phase A: Water + 0.1% by volume of formic acid (99%), flow Phase B: acetonitrile; gradient: 0-1.6 min 1-99% B, 1.6-2.0 min 99% B; flow rate 0.8 ml/min; temperature: 60 ° C; injection: 2 μl; DAD scan: 210-400 nm; ESI+, ESI-, scan range 160-1000 m/z; ELSD. Method A2: UPLC (MeCN-NH3 ): Instrument: Waters Acquity UPLC-MS SQD 3001; Column: Acquity UPLC BEH C18 1.7 50 x 2.1 mm; Mobile phase A: water + 0.2% by volume of formic acid (32%), mobile phase B: acetonitrile; Gradient: 0 -1.6 min 1-99% B, 1.6-2.0 min 99% B; flow rate 0.8 ml/min; temperature: 60 ° C; injection: 2 μl; DAD scan: 210-400 nm; MS ESI+, ESI-, scan range 160 -1000 m/z; ELSD. Method A3: (LC-MS) Instrument: Agilent 1290 Infinity LC; Column: Acquity UPLC BEH C18 1.7 50 x 2.1 mm; mobile phase A: water + 0.05 vol% formic acid, mobile phase B: acetonitrile + 0.05 vol% formic acid; Gradient: 0-1.7 min 2-90% B, 1.7-2.0 min 90% B; flow rate 1.2 ml/min; temperature: 60 ° C; injection: 2 μl; DAD scan: 190-390 nm; MS: Agilent TOF 6230. Method A4: (LC-MS) Instrument: Waters Acquity; column: Kinetex (Phenomenex), 50 x 2 mm; mobile phase A: water + 0.05 vol% formic acid, mobile phase B: acetonitrile + 0.05 vol% formic acid; gradient: 0-1.9 min 1-99% B, 1.9-2.1 min 99% B; flow rate 1.5 ml/min; temperature: 60 ° C; injection: 0.5 μl; DAD scan: 200-400 nm. In some instances, the compounds of the invention and their precursors and/or intermediate systems are purified by the following exemplary preparative HPLC method: Method P1: System: Waters Self-purification System: Pump 2545, Sample Tube Controller 2767, CFO , DAD 2996, ELSD 2424, SQD; column: XBridge C18 5μm 100×30 mm; mobile phase: A: water + 0.1% by volume of formic acid, mobile phase B: acetonitrile; gradient: 0-8 min 10-100% B, 8-10 min 100% B; flow rate: 50 ml/min; temperature: room temperature; solution: maximum 250 mg / maximum 2.5 ml DMSO or DMF; injection: 1 × 2.5 ml; detection: DAD scan range 210-400 nm; MS ESI+, ESI-, scanning range 160-1000 m/z. Method P2: System: Waters self-purification system: pump 254, sample controller 2767, CFO, DAD 2996, ELSD 2424, SQD 3100; column: XBridge C18 5μm 100×30 mm; mobile phase: A: water + 0.2 Volume % ammonia (32%), mobile phase B: methanol; gradient: 0-8 min 30-70% B; flow rate: 50 ml/min; temperature: room temperature; detection: DAD scan range 210-400 nm; MS ESI+ , ESI-, scanning range 160-1000 m / z; ELSD. Method P3: System: Labomatic, Pump: HD-5000, Fraction Collector: LABOCOL Vario-4000, UV Detector: Knauer UVD 2.1S; Column: XBridge C18 5μm 100×30 mm; Mobile Phase A: Water+ 0.2 vol% ammonia (25%), mobile phase B: acetonitrile; gradient: 0-1 min 15% B, 1-6.3 min 15-55% B, 6.3-6.4 min 55-100% B, 6.4-7.4 min 100 % B; flow rate: 60 ml/min; temperature: room temperature; solution: maximum 250 mg / 2 ml DMSO; injection: 2 × 2 ml; detection: UV 218 nm; software: SCPA PrepCon5. Method P4: System: Labomatic, Pump: HD-5000, Fraction Collector: LABOCOL Vario-4000, UV Detector: Knauer UVD 2.1S; Column: Chromatorex RP C18 10 μm 125×30 mm, Mobile Phase: A: Water + 0.1% by volume formic acid, mobile phase B: acetonitrile; Gradient: 0 - 15 min 65 – 100% B; Flow rate: 60 ml/min; Temperature: room temperature; Solution: Max 250 mg / 2 ml DMSO; Injection: 2 × 2 ml; detection: UV 254 nm; software: SCPA PrepCon5. Method P5: System: Sepiatec: Prep SFC100; column: Chiralpak IA 5 μm 250×20 mm; mobile phase A: carbon dioxide, mobile phase B: ethanol; gradient: isocratic 20% B; flow rate: 80 ml/min; 40 ° C; solution: maximum 250 mg / 2 ml DMSO; injection: 5 × 0.4 ml detection: UV 254 nm. Method P6: System: Agilent: Prep 1200, 2×Prep Pump, DLA, MWD, Gilson: Liquid Dispenser 215; Column: Chiralcel OJ-H 5 μm 250×20 mm; Mobile Phase A: Hexane, Mobile Phase B: Ethanol; Gradient: isocratic 30% B; flow rate: 25 ml/min; temperature: 25 ° C; solution: 187 mg / 8 ml ethanol/methanol; injection: 8 × 1.0 ml detection: UV 280 nm. Method P7: System: Labomatic, pump: HD-5000, fraction collector: LABOCOL Vario-4000, UV detector: Knauer UVD 2.1S; column: XBridge C18 5μm 100×30 mm; mobile phase A: water + 0.1% by volume formic acid, mobile phase B: acetonitrile; gradient: 0-3 min: 65% B isocratic, 3-13 min: 65-100% B; flow rate: 60 ml/min; temperature: room temperature; solution: max. 250 mg / 2 ml DMSO; injection: 2 × 2 ml; detection: UV 254 nm. Method P8: System: Agilent: Prep 1200, 2×Prep Pump, DLA, MWD, Gilson: Liquid Dispenser 215; Column: Chiralpak IF 5 μm 250×20 mm; Mobile Phase A: Ethanol, Mobile Phase B: Methanol; Gradient : isocratic 50% B; flow rate: 25 ml/min; temperature: 25 ° C; solution: 600 mg / 7 ml N,N-dimethylformamide; injection: 10 × 0.7 ml detection: UV 254 nm. For the preparation of some of the compounds of the invention and their precursors and/or intermediates, Isolera from Biotage is used on silicone® The device was subjected to column chromatography purification ("quick chromatography"). This is done using a column from Biotage (for example, "SNAP Cartridge, KP_SIL" columns of different sizes and "Interchim Puriflash Silica HP 15UM Boiling Tower" columns of different sizes from Interchim).Intermediate 1-1 6- chlorine -2-(3- Methoxy -3- Methyl butyl )-5- Nitro -2H- Carbazole First, 500 mg (2.53 mmol) of 6-chloro-5-nitro-1H-carbazole (CAS No.: 101420-98-8) and 390 μl (3.0 mmol) of 3-methoxy-3 were charged in 5 ml of THF. -Methylbutan-1-ol and 996 mg (3.80 mmol) of triphenylphosphine, 540 μl (2.8 mmol) of diisopropyl azodicarboxylate were slowly added dropwise and the mixture was stirred at 25 ° C for 21.5 hours. The reaction mixture was diluted with water and extracted three times with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was purified by flash chromatography (Interchim 15[mu]m column, KP-Sil, mobile phase: hexane/ethyl acetate). This gave 792 mg (crude) of the title compound. LC-MS (Method A1): Rt = 1.22 min (UV detector: TIC Smooth), mass measurement 297.001 H-NMR (500MHz, DMSO-d6, selected signal): δ = 2.10 - 2.16 (m, 1H), 3.12 (s, 1H), 4.50 - 4.55 (m, 1H), 8.00 (d, 1H), 8.67 (s, 1H), 8.82 (d, 1H).Intermediate 2-1 6- chlorine -2-(3- Methoxy -3- Methyl butyl )-2H- Carbazole -5- amine 791 mg of 6-chloro-2-(3-methoxy-3-methylbutyl)-5-nitro-2H-carbazole (crude) was dissolved in 8 ml of ethanol and 2 ml of water was added. Then 1.48 g (26.6 mmol) of iron powder and 71.1 mg (1.33 mmol) of ammonium chloride were added, and the mixture was stirred at 90 ° C for 1.5 hours. The reaction mixture was cooled and filtered through celite. The filtrate was washed three times with ethanol and concentrated. The aqueous phase was extracted three times with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, filtered (hydrophobic filter) and concentrated. This gave 715 mg of the title compound as a crude material. LC-MS (Method A1): Rt = 0.92 min (UV detector: TIC Smooth), mass measurement 267.00.Intermediate 3-1 N-(6- chlorine -1H- Carbazole -5- base )-6-( Difluoromethyl ) Pyridine -2- Formamide 2.00 g (11.9 mmol) of 6-chloro-1H-indazole-5-amine (CAS number: 221681-75-0), 2.48 g (14.3 mmol) of 6-(difluoromethyl)pyridine-2-carboxylic acid, 1.83 g (11.9 mmol) 1-hydroxy-1H-benzotriazole hydrate, 4.58 g (23.9 mmol) 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride A mixture of 5.0 ml (36 mmol) of triethylamine in 40 ml of THF was stirred at 25 ° C for 20.5 hours. The reaction mixture was diluted with water and concentrated. The solid was filtered off with suction and washed three times with water and three times with diethyl ether. Then ether was added and the mixture was stirred. The solid was filtered off with suction, washed three times with diethyl ether and dried under reduced pressure. This gave 3.45 g of the title compound as a crude product. LC-MS (Method A1): Rt = 1.09 min (UV detector: TIC Smooth), mass measurement 322.00.Intermediate 4-1 5- Nitro -6-( Oxetane -3- Baseoxy )-1H- Carbazole First, 4.00 g (22.3 mmol) of 5-nitro-1H-indazole-6-ol (CAS No.: 1082041-56-2), 1.74 g (23.4 mmol) of oxetane-3- was charged in 50 ml of THF. Alcohol and 8.79 g (33.5 mmol) of triphenylphosphine were slowly added dropwise 6.5 ml (33 mmol) of diisopropyl azodicarboxylate and the mixture was stirred at 25 ° C for 17.5 hours. The reaction mixture was diluted with water and extracted three times with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was purified by flash chromatography (Biotage SNAP cartridge (100 g; KP-Sil), mobile phase: hexane/ethyl acetate). This gave a crude product of 4.58 g of the title compound. LC-MS (Method A1): Rt = 0.78 min (UV detector: TIC Smooth), mass measurement 235.001 H NMR (500MHz, DMSO-d6): δ = 4.59 - 4.64 (m, 2H), 4.95 - 5.02 (m, 2H), 5.50 - 5.56 (m, 1H), 6.84 (s, 1H), 8.21 (s, 1H), 8.49 (s, 1H), 13.33 (s, 1H).Intermediate 4-2 5- Nitro -6-( Oxetane -3- Methoxy )-1H- Carbazole First, fill 2.50 g (11.2 mmol) of 5-nitro-1H-indazole-6-ol (CAS number: 1082041-56-2), 1.03 g (11.7 mmol) of oxetane-3- in 25 ml of THF. Methanol and 4.39 g (16.7 mmol) of triphenylphosphine were slowly added dropwise 3.2 ml (17 mmol) of diisopropyl azodicarboxylate and the mixture was stirred at 25 ° C for 18.5 hours. The reaction mixture was diluted with water and extracted three times with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was purified by flash chromatography (Biotage SNAP cartridge (100 g; KP-Sil), mobile phase: hexane/ethyl acetate). This gave 1.03 g of the crude title compound. LC-MS (Method A1): Rt = 0.79 min (UV detector: TIC Smooth), mass measurement 249.001 H NMR (500MHz, DMSO-d 6 ): δ = 3.41 - 3.48 (m, 1H), 4.40 (d, 2H), 4.48 (t, 2H), 4.70 (dd, 2H), 7.28 (s, 1H), 8.20 (s, 1H), 8.43 (s, 1H), 13.38 (s, 1H).Intermediate 4-3 5- Nitro -6-[(3S)- Tetrahydrofuran -3- Baseoxy ]-1H- Carbazole First, 2.50 g (11.2 mmol) of 5-nitro-1H-indazole-6-ol (CAS number: 1082041-56-2), 940 μl (12 mmol) (3S)-tetrahydrofuran-3 were charged in 25 ml of THF. - Alcohol and 4.39 g (16.7 mmol) of triphenylphosphine, 3.2 ml (17 mmol) of diisopropyl azodicarboxylate were slowly added dropwise and the mixture was stirred at 25 ° C for 67 hours. The reaction mixture was concentrated and purified by flash chromatography (EtOAc EtOAc EtOAc (EtOAc) This gave 3.08 g of the title compound. LC-MS (Method A1): Rt = 0.82 min (UV detector: TIC Smooth), mass measurement 249.00.Intermediate 6-1 N-(6- Isopropoxy -1H- Carbazole -5- base )-6-( Trifluoromethyl ) Pyridine -2- Formamide First, 7.02 g (36.73 mmol) of 6-(trifluoromethyl)pyridine-2-carboxylic acid and 13.48 g (41.98 mmol) of O-(benzotriazol-1-yl)-N,N were charged in 400 ml of THF. N', N'-tetramethyluronium tetrafluoroborate and 7.31 ml (41.98 mmol) of N,N-diisopropylethylamine. The mixture was stirred at 25 ° C for 30 minutes, and then 6.69 g (34.98 mmol) of 6-isopropoxy-1H-indazole-5-amine (CAS number: 1498329-80-8) was added. The reaction mixture was stirred at 25 ° C for 16 hours. The reaction mixture was concentrated and the residue was partitioned between water andEtOAc. The mixture was extracted twice with ethyl acetate and the combined organic phases were washed with saturated sodium chloride, filtered and concentrated with a The residue was purified by flash chromatography (Biotage SNAP cartridge (340 g; KP-Sil), mobile phase: hexane/ethyl acetate). The combined product fractions were concentrated, suspended in water and stirred vigorously for 20 min. The product was filtered off with suction and washed with water and diethyl ether. This gave 8.56 g of the title compound. LC-MS (Method A1): Rt = 1.23 min (UV detector: TIC Smooth), mass measurement 364.001 H NMR (400 MHz, DMSO-d6): δ = 1.41 (d, 6 H), 4.85 (spt, 1 H), 7.18 (s, 1 H), 8.01 (s, 1 H), 8.21 (dd, 1 H), 8.40 (t, 1 H), 8.47 (d, 1 H), 8.81 (s, 1 H), 10.67 (s, 1 H), 12.85 (s, 1 H).Intermediate 6-2 6-( Difluoromethyl )-N-(6- Ethoxy -1H- Carbazole -5- base ) Pyridine -2- Formamide 512 mg (2.89 mmol) 6-ethoxy-1H-indazole-5-amine, 600 mg (3.47 mmol) 6-(difluoromethyl)pyridine-2-carboxylic acid, 442 mg (2.89 mmol) 1- Hydroxy-1H-benzotriazole hydrate, 1.11 g (5.78 mmol) of 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride and 1.2 ml (8.7 mmol) of three The mixture of ethylamine in 10 ml of THF was stirred at 25 ° C for 20.5 hours. The reaction mixture was diluted with water and extracted three times with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. Diethyl ether was added to the residue, the mixture was stirred and the solid formed was filtered off with suction, washed three times with diethyl ether and dried. This gave 499 mg of the title compound. LC-MS (Method A1): Rt = 1.10 min (UV detector: TIC Smooth), mass measurement 332.001 </ RTI> <RTIgt; 8.26 - 8.36 (m, 2H), 8.76 (s, 1H), 10.71 (s, 1H), 12.86 (s, 1H).Intermediate 6-3 N-[6-( Benzyloxy )-1H- Carbazole -5- base ]-6-( Trifluoromethyl ) Pyridine -2- Formamide First, 5.40 g (22.57 mmol) of 6-(trifluoromethyl)pyridine-2-carboxylic acid and 10.87 g (33.85 mmol) of O-(benzotriazol-1-yl)-N,N were charged in 150 ml of THF. N', N'-tetramethyluronium tetrafluoroborate and 5.90 ml (33.85 mmol) of N,N-diisopropylethylamine. The mixture was stirred at 25 ° C for 30 minutes, and then 5.40 g (22.57 mmol) of 6-(benzyloxy)-1H-indazole-5-amine (CAS No.: 1499162-36-5) was added. The reaction mixture was stirred at 25 ° C for 16 hours. The reaction mixture was concentrated and the residue was partitioned between water andEtOAc. The mixture was extracted with EtOAc and EtOAc (EtOAc)EtOAc. Diethyl ether was added to the crude product, and the mixture was stirred vigorously for 10 minutes and the formed precipitate was filtered off with suction. This gave 6.72 g of the title compound. LC-MS (Method A1): Rt = 1.28 min (UV detector: TIC Smooth), mass measurement 412.001 H NMR (400 MHz, DMSO-d6): δ = 5.34 (s, 2 H), 7.31 (s, 1 H), 7.41 (d, 3 H), 7.58 (dd, 2 H), 8.04 (s, 1 H), 8.18 (d, 1 H), 8.40 (d, 1 H), 8.45 - 8.53 (m, 1 H), 8.85 (s, 1 H), 10.41 (s, 1 H), 12.97 (s, 1 H).Intermediate 6-4 N-[6-( Cyclopropylmethoxy )-1H- Carbazole -5- base ]-6-( Trifluoromethyl ) Pyridine -2- Formamide Dissolve 1.00 g of N-(6-hydroxy-1H-indazol-5-yl)-6-(trifluoromethyl)pyridine-2-carboxamide in 20 ml of DMF and add 1.72 g with stirring ( 12.41 mmol) potassium carbonate. The suspension was stirred at 25 ° C for 30 minutes and then 431 μl (4.65 mmol) of (bromomethyl)cyclopropane was added. The reaction mixture was stirred at 25 ° C for 16 hours. The reaction mixture was then diluted with water and the solid formed was filtered with suction, washed with diethyl ether and dried under reduced pressure. This gave 585 mg of the title compound. LC-MS (Method A1): Rt = 1.25 min (UV detector: TIC Smooth), mass measurement 376.001 H NMR (300 MHz, DMSO-d6): δ = 0.40 - 0.51 (m, 2 H), 0.59 - 0.71 (m, 2 H), 1.27 - 1.41 (m, 1 H), 4.05 (d, 2 H) , 7.08 (s, 1 H), 8.01 (s, 1 H), 8.16 - 8.25 (m, 1 H), 8.41 (t, 1 H), 8.48 (d, 1 H), 8.82 (s, 1 H) , 10.65 (s, 1 H), 12.90 (s, 1 H).Intermediate 6-5 N-[6-( Trifluoromethoxy )-1H- Carbazole -5- base ]-6-( Trifluoromethyl ) Pyridine -2- Formamide 600 mg of 6-(trifluoromethoxy)-1H-indazole-5-amine (CAS number: 1491 916-39-8, used as a crude product) and 673 mg (3.52 mmol) of 6-(trifluoromethyl) Pyridine-2-carboxylic acid was dissolved in 15 ml of THF, and 360 mg (2.35 mmol) of 1-hydroxy-1H-benzotriazole hydrate, 900 mg (4.70 mmol) 1-(3-di) was added at 25 °C. Methylaminopropyl)-3-ethylcarbodiimide hydrochloride and 980 μl (7.0 mmol) of triethylamine. The reaction mixture was stirred at RT for 24 hours and then diluted with water and a solid precipitated was filtered. The solid was washed with water and diethyl ether and dried under reduced pressure. This gave 406 mg of the title compound. LC-MS (Method A1): Rt = 1.26 min (UV detector: TIC Smooth) with a mass measurement of 390.00.Intermediate 6-6 2- Cyclopropyl -N-(6- Methoxy -1H- Carbazole -5- base )-1,3- Oxazole -4- Formamide 586 mg (3.59 mmol) of 6-methoxy-1H-indazole-5-amine (CAS number: 749223-61-8) and 500 mg (3.27 mmol) of 2-cyclopropyl-1,3-oxazole 4-carboxylic acid (CAS number: 1060816-04-7) was dissolved in 10 ml of THF, and 500 mg (3.3 mmol) of 1-hydroxy-1H-benzotriazole hydrate, 1.25 g (6.5) was added at 25 °C. Methyl) 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride and 1.37 ml (9.8 mmol) of triethylamine. Water was added, the solvent was partially removed and the solid was filtered, washed with water and diethyl ether and dried. This gave 758 mg of the title compound. LC-MS (Method A1): Rt = 1.05 min (UV detector: TIC Smooth), mass measurement 298.00.Intermediate 6-7 N-(6- Methoxy -1H- Carbazole -5- base )-6-( Pyrrolidine -1- base ) Pyridine -2- Formamide Add 692 mg HATU to 248 mg of 6-methoxy-1H-indazole-5-amine, 350 mg of 6-(pyrrolidin-1-yl)pyridine-2-carboxylic acid and 0.79 ml of N,N-diisopropyl The mixture was stirred in 5.3 ml of THF and the mixture was stirred at room temperature overnight. Water was added and the solid was filtered off with suction and dried under reduced pressure. This gave 503 mg of the title compound as a crude material.1 H-NMR (400MHz, DMSO-d6 ): δ [ppm] = 1.98 - 2.07 (m, 4H), 3.46 - 3.58 (m, 4H), 4.00 (s, 3H), 6.74 (d, 1H), 7.10 (s, 1H), 7.33 (d, 1H), 7.72 (dd, 1H), 7.98 (s, 1H), 8.74 (s, 1H), 10.85 (s, 1H), 12.88 (s, 1H).Intermediate 7-1 4-(6- Ethoxy -5- Nitro -2H- Carbazole -2- base )-2- Methyl butyl -2- alcohol First, 10 g (48 mmol) of 6-ethoxy-5-nitro-1H-carbazole (see WO2015091426) and 20 g of potassium carbonate were charged in 100 ml of DMF, and the mixture was stirred for 15 minutes. Then 8.8 ml (72.3 mmol) of 4-bromo-2-methylbutan-2-ol was added and the mixture was stirred at 70 ° C overnight. Another 2.9 ml (24 mmol) of 4-bromo-2-methylbutan-2-ol and 6.6 g of potassium carbonate were added and stirring was continued at 70 ° C for another night. The mixture was filtered off with suction and the filtrate was taken up in toluene and washed three times with water. The mixture was washed with a saturated sodium chloride solution and dried over magnesium sulfate and then concentrated. Rapid chromatography (Biotage, hexane/ethyl acetate gradient) gave 4. LC-MS (Method A1): Rt = 0.99 min (UV detector: TIC Smooth), mass measurement 293.00. 1H-NMR (400MHz, DMSO-d6): δ [ppm] = 1.15 (s, 6 H) 1.35 (t, 3H) 1.99 - 2.06 (m, 2 H) 4.14 – 4.21 (q, 2 H) 4.45 - 4.53 (m, 2 H) 4.54 (s, 1H) 7.23 (s, 1H) 8.37 ( s, 1 H) 8.60 (s, 1 H)Intermediate 7-2 2- methyl -4-[5- Nitro -6-( Oxetane -3- Baseoxy )-2H- Carbazole -2- base ] Ding -2- alcohol 4.58 g of 5-nitro-6-(oxetan-3-yloxy)-1H-indazole (crude product) was dissolved in 50 ml of DMF and 6.06 g (43.8 mmol) of carbonic acid was added with stirring. Potassium and 7.16 g (used as a crude product) 3-hydroxy-3-methylbutyl-4-toluenesulfonate (M. Shimizu et al.,Bioorganic and Medicinal Chemistry 2006 ,14, 4277 – 4294). The reaction mixture was stirred at 80 &lt;0&gt;C for 18 h, diluted with H~~~~ The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was purified by flash chromatography (Biotage SNAP cartridge, KP-Sil, mobile phase: hexane / ethyl acetate). This gave 627 mg of the title compound. LC-MS (Method A1): Rt = 0.87 min (UV detector: TIC Smooth), mass measurement 321.001 H NMR (500MHz, DMSO-d6): δ = 1.14 (s, 6H), 1.99 - 2.04 (m, 2H), 4.47 - 4.51 (m, 2H), 4.53 (s, 1H), 4.57 (dd, 2H) , 4.97 - 5.02 (m, 2H), 5.42 - 5.49 (m, 1H), 6.90 (s, 1H), 8.47 (s, 1H), 8.63 (d, 1H).Intermediate 7-3 2- methyl -4-[5- Nitro -6-( Oxetane -3- Methoxy )-2H- Carbazole -2- base ] Ding -2- alcohol Dissolve 1.03 g of 5-nitro-6-(oxetan-3-ylmethoxy)-1H-indazole (crude) in 7.5 ml DMF and add 777 mg (4.65 mmol) with stirring 4-bromo-2-methylbutan-2-ol and 1.29 g (9.30 mmol) of potassium carbonate. The reaction mixture was stirred at 100 ° C for 16.5 hours. The reaction mixture was diluted with water and extracted three times with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. The reaction mixture was partitioned between ethyl acetate and water. The organic phase was washed twice with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was purified by preparative HPLC. This gave 287 mg of the title compound. LC-MS (Method A1): Rt = 0.84 min; MS (ESIpos): m/z = 335 [M+H]+ 1 H NMR (500MHz, DMSO-d6): δ = 1.15 (s, 6H), 2.00 - 2.06 (m, 2H), 3.40 - 3.47 (m, 1H), 4.35 (d, 2H), 4.46 (t, 2H) , 4.48 - 4.52 (m, 2H), 4.70 (dd, 2H), 7.30 (s, 1H), 8.39 (s, 1H), 8.61 (d, 1H).Intermediate 7-4 2- methyl -4-{5- Nitro -6-[(3S)- Tetrahydrofuran -3- Baseoxy ]-2H- Carbazole -2- base } Ding -2- alcohol 3.08 g of 5-nitro-6-[(3S)-tetrahydrofuran-3-yloxy]-1H-indazole (crude) was dissolved in 50 ml of DMF and 3.84 g (27.8 mmol) was added with stirring. Potassium carbonate and 4.55 g (used as a crude product) 3-hydroxy-3-methylbutyl-4-toluenesulfonate. The reaction mixture was stirred at 80 ° C for 22 hours. The reaction mixture was diluted with water and extracted three times with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was purified by flash chromatography (Biotage SNAP cartridge (100 g; KP-Sil), mobile phase: hexane/ethyl acetate). This gave 618 mg of the title compound as a crude material. LC-MS (Method A1): Rt = 0.91 min (UV detector: TIC Smooth), mass measurement 335.00.Intermediate 8-1 [5- Amine -6-( Cyclopropylmethoxy )-2H- Carbazole -2- base ] Ethyl acetate Add 5.52 ml (71.6 mmol) of trifluoroacetic acid to 2.79 g (7.16 mmol) of {5-[(t-butoxycarbonyl)amino]-6-(cyclopropylmethoxy) in 50 ml of dichloromethane Ethyl 2-H-oxazol-2-yl}acetate, and the mixture was stirred at 25 ° C for 6 hours. The mixture was poured into saturated sodium bicarbonate solution and stirred for 10 minutes, the phases were separated and the aqueous phase was extracted twice with dichloromethane. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. This gave 2.03 g of the title compound. LC-MS (Method A1): Rt = 0.75 min (UV detector: TIC Smooth), mass 289.00 1H NMR (400 MHz, DMSO-d6): δ = 0.32 - 0.43 (m, 2 H), 0.53 - 0.64 (m, 2 H), 1.20 (t, 3 H), 1.29 (s, 1 H), 3.86 (d, 2 H), 4.14 (q, 2 H), 4.65 (br. s., 2 H), 5.17 (s, 2 H), 6.65 (s, 1 H), 6.76 (s, 1 H), 7.84 (d, 1 H).Intermediate 8-2 4-[5- Amine -6-( Benzyloxy )-2H- Carbazole -2- base ]-2- Methyl butyl -2- alcohol Add 2.06 ml (26.79 mmol) of trifluoroacetic acid to 1.14 g (2.68 mmol) of [6-(benzyloxy)-2-(3-hydroxy-3-methylbutyl) in 20 ml of dichloromethane -2H-carbazol-5-yl] carbamic acid tert-butyl ester, and the mixture was stirred at 25 ° C for 3 hours. The mixture was poured into a saturated sodium bicarbonate solution. The solid formed was filtered off with suction, washed with water and dried. This gave 840 mg of the title compound. LC-MS (Method A1): Rt = 0.82 min (UV detector: TIC Smooth), mass measurement 325.00 1H NMR (400 MHz, DMSO-d6): δ = 1.12 (s, 6 H), 1.90 - 2.02 (m, 2 H), 4.21 - 4.38 (m, 2 H), 4.46 (s, 1 H), 4.76 (br. s., 2 H), 5.17 (s, 2 H), 6.67 (s, 1 H), 6.91 (s, 1 H), 7.28 - 7.36 (m, 1 H), 7.36 - 7.45 (m, 2 H), 7.48 - 7.55 (m, 2 H), 7.85 (s, 1 H).Intermediate 8-3 4-(5- Amine -6- Methoxy -2H- Carbazole -2- base )-2- Methyl butyl -2- alcohol 610 mg of [2-(3-hydroxy-3-methylbutyl)-6-methoxy-2H-indazol-5-yl]carbamic acid tert-butyl ester in 10 ml of dichloromethane and 1.3 The mixture in ml (16 mmol) trifluoroacetic acid was stirred at 25 ° C for 19.5 hours. The mixture was poured into saturated sodium bicarbonate solution and stirred for 10 minutes, the phases were separated and dichloromethane was extracted twice. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. This gave 318 mg of the title compound as a crude material. LC-MS (Method A2): Rt = 0.72 min (UV detector: TIC Smooth), mass measurement 249.00.Intermediate 8-4 6- Methoxy -2-[3-( Tetrahydrogen -2H- Pyran -2- Baseoxy ) Propyl ]-2H- Carbazole -5- amine First, fill 789 mg (0.95 mmol) of {6-methoxy-2-[3-(tetrahydro-2H-pyran-2-yloxy)propyl]-2H-carbazole in 5 ml of dichloromethane. -5-yl} tert-butyl carbamic acid. 735 μl (9.53 mmol) of trifluoroacetic acid was added, and the mixture was stirred at 25 ° C for 22 hours. Another 735 μl (9.53 mmol) of trifluoroacetic acid was added, and the mixture was stirred at 25 ° C for 68 hours. The reaction mixture was extracted with water and washed with dichloromethane three times. The combined organic phases were washed with saturated sodium bicarbonate solution and saturated sodium chloride solution, filtered (hydrophobic filter) and concentrated. Once again, a saturated sodium bicarbonate solution (pH about equal to 9) was added to the aqueous solution, and the mixture was extracted three times with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, filtered (hydrophobic filter) and concentrated. Sodium chloride and ethyl acetate were added to the aqueous phase and the mixture was stirred for 30 minutes. The phases were separated and the aqueous phase was extracted twice more with ethyl acetate. The organic phase was combined by filtration (hydrophobic filter) and concentrated. This gave 199 mg of the title compound. LC-MS (Method A2): Rt = 0.55 min (UV detector: TIC Smooth), mass measurement 221.00.Intermediate 8-5 4-(5- Amine -6- Ethoxy -2H- Carbazole -2- base )-2- Methyl butyl -2- alcohol 4.87 g (16.6 mmol) of 4-(6-ethoxy-5-nitro-2H-indazol-2-yl)-2-methylbutan-2-ol was dissolved in 164 ml of THF and 70 ml of methanol And hydrogenation was carried out for 3 hours at 25 ° C in a hydrogen atmosphere using 1.46 g (0.1 mmol) of palladium on activated carbon. The reaction mixture was filtered through celite, and the filtrate was washed with ethyl acetate. This gave 4.37 g of the title compound. 1H-NMR (400MHz, DMSO-d6): δ = 1.13 (s, 6 H), 1.40 (t, 3H), 1.92 – 1.99 (m, 2 H), 4.00 – 4.07 (q, 2 H), 4.26 - 4.34 (m, 2 H), 4.49 (s, 1H), 4.57 (s, 2H), 6.62 (s, 1H), 6.80 ( s, 1 H), 7.83 (s, 1 H).Intermediate 8-6 4-[5- Amine -6-( Oxetane -3- Baseoxy )-2H- Carbazole -2- base ]-2- Methyl butyl -2- alcohol Dissolve 616 mg of 2-methyl-4-[5-nitro-6-(oxetan-3-yloxy)-2H-indazol-2-yl]butan-2-ol in 12 ml of ethanol Medium and add 3 ml of water. Then 963 mg (17.3 mmol) of iron powder and 46.1 mg (863 μmol) of ammonium chloride were added, and the mixture was stirred at 90 ° C for 17 hours. The reaction mixture was filtered with EtOAc (EtOAc)EtOAc. This gave 574 mg of the title compound as a crude material. LC-MS (Method A2): Rt = 0.68 min (UV detector: TIC Smooth), mass measurement 291.00.Intermediate 8-7 4-[5- Amine -6-( Oxetane -3- Methoxy )-2H- Carbazole -2- base ]-2- Methyl butyl -2- alcohol Dissolve 281 mg of 2-methyl-4-[5-nitro-6-(oxetan-3-ylmethoxy)-2H-indazol-2-yl]butan-2-ol in 7.5 ml Add in ethanol and add 2.5 ml of water. Then 426 mg (7.62 mmol) of iron powder and 20.4 mg (381 μmol) of ammonium chloride were added, and the mixture was stirred at 90 ° C for 21 hours. The reaction mixture was cooled and filtered. The filtrate was concentrated and the residue was taken up in EtOAc, filtered and concentrated. This gave 227 mg of the title compound as a crude material. LC-MS (Method A2): Rt = 0.70 min (UV detector: TIC Smooth), mass measurement 303.00.Intermediate 8-8 4-{5- Amine -6-[(3S)- Tetrahydrofuran -3- Baseoxy ]-2H- Carbazole -2- base }-2- Methyl butyl -2- alcohol 610 mg 2-methyl-4-{5-nitro-6-[(3S)-tetrahydrofuran-3-yloxy]-2H-indazol-2-yl}butan-2-ol (crude) Dissolve in 10 ml of ethanol and add 3 ml of water. Then 843 mg (15.1 mmol) of iron powder and 40 mg of ammonium chloride were added, and the mixture was stirred at 90 ° C for 5 hours. The reaction mixture was cooled and filtered through celite. The filtrate was concentrated and the residue was taken up in EtOAc, filtered and concentrated. The residue was taken up in 2 mL DMF and purified by preparative HPLC. This gave 348 mg of the title compound as a crude material. LC-MS (Method A4): Rt = 0.72 min; MS (ESIpos): m/z = 305 [M+H]+ .Intermediate 9-1 (6- Methoxy -1H- Carbazole -5- base ) Tert-butyl carbamic acid The synthesis is described in WO2015091426.Intermediate 10-1 [2-(3- Hydroxyl -3- Methyl butyl )-6- Methoxy -2H- Carbazole -5- base ] Tert-butyl carbamic acid 2.00 g (7.60 mmol) of (6-methoxy-1H-indazol-5-yl)carbamic acid tert-butyl ester was dissolved in 20 ml of 1-methyl-2-pyrrolidone and stirred 1.90 g (11.4 mmol) of 4-bromo-2-methylbutan-2-ol, 3.15 g (22.8 mmol) of potassium carbonate and 1.89 g (11.4 mmol) of potassium iodide were added. The suspension was stirred at 80 ° C for 21 hours. Another 525 mg (3.8 mmol) potassium carbonate and 630 mg (3.8 mmol) potassium iodide were added, and the mixture was stirred at 80 ° C for an additional 6 hours. The reaction mixture was diluted with water and extracted three times with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was purified by flash chromatography (Biotage EtOAc EtOAc (EtOAc) The combined product fractions were concentrated and dried. This gave 610 mg of the title compound. LC-MS (Method A1): Rt = 1.16 min (UV detector: TIC Smooth), mass measurement 349.001 H-NMR (500MHz, DMSO-d 6 ): δ = 1.13 (s, 6H), 1.46 (s), 1.96 - 2.03 (m, 2H), 3.85 (s, 3H), 4.36 - 4.41 (m, 2H) , 4.50 (s, 1H), 6.97 (s, 1H), 7.77 (s, 1H), 7.89 (br. s., 1H), 8.17 (d, 1H).Intermediate 10-2 {6- Methoxy -2-[3-( Tetrahydrogen -2H- Pyran -2- Baseoxy ) Propyl ]-2H- Carbazole -5- base } Tert-butyl carbamic acid Dissolve 1.00 g of (3-methoxy-1H-indazol-5-yl)carbamic acid tert-butyl ester in 15 ml of DMF and add 967 μl (5.7 mmol) 2-(3- Bromopropoxy)tetrahydro-2H-pyran, 1.58 g (11.4 mmol) potassium carbonate and 757 mg (4.6 mmol) potassium iodide. The reaction mixture was stirred at 100 ° C for 20 hours. Another 484 μl (2.9 mmol) of 2-(3-bromopropoxy)tetrahydro-2H-pyran was added, and the mixture was stirred at 100 ° C for 24 hours. The reaction mixture was then diluted with water and extracted with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution and the phases were separated and filtered thru a filtered filter. The residue was taken up in dichloromethane and adsorbed onto Isolute during concentration. The residue was purified by flash chromatography (Biotage SNAP cartridge (100 g; KP-Sil), mobile phase: hexane/ethyl acetate). The combined product fractions were concentrated and dried. This gave 790 mg of the title compound. LC-MS (Method A1): Rt = 1.33 min (UV detector: TIC Smooth) with a mass measurement of 405.00.Intermediate 10-3 [6-( Benzyloxy )-2-(3- Hydroxyl -3- Methyl butyl )-2H- Carbazole -5- base ] Tert-butyl carbamic acid 3.50 (10.3 mmol) of [6-(benzyloxy)-1H-indazol-5-yl]carbamic acid tert-butyl ester (CAS number: 1799835-17-8) was dissolved in 30 ml of DMF. 4.28 g (30.94 mmol) potassium carbonate and 1.71 g (10.31 mmol) potassium iodide were added with stirring. The suspension was stirred at 25 ° C for 30 minutes and then 2.58 g (15.47 mmol) of 4-bromo-2-methylbutan-2-ol was added. The reaction mixture was stirred at 100 ° C for 16 hours. Another 1.72 g (10.31 mmol) of 4-bromo-2-methylbutan-2-ol and 1.42 g (10.31 mmol) of potassium carbonate were added, and the mixture was stirred at 100 ° C for another 24 hours. The reaction mixture was then diluted with water and extracted twice with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was purified by flash chromatography (Biotage SNAP cartridge (340 g; KP-Sil), mobile phase: hexane/ethyl acetate). This gave 1.14 g of the title compound. LC-MS (Method A1): Rt = 1.35 min (UV detector: TIC Smooth), mass measurement 425.00 1H NMR (400 MHz, DMSO-d6): δ = 1.13 (s, 6 H), 1.45 (s, 9 H), 1.92 - 2.02 (m , 2 H), 4.34 - 4.42 (m, 2 H), 4.47 (s, 1 H), 5.20 (s, 2 H), 7.03 (s, 1 H), 7.29 - 7.37 (m, 1 H), 7.37 - 7.44 (m, 2 H), 7.53 (d, 2 H), 7.81 - 7.87 (m, 2 H), 8.18 (s, 1 H).Intermediate 10-4 {5-[( Third butoxycarbonyl ) Amine ]-6-( Cyclopropylmethoxy )-2H- Carbazole -2- base } Ethyl acetate 3.00 g (8.95 mmol) of {5-[(t-butoxycarbonyl)amino]-6-hydroxy-2H-indazol-2-yl}acetate was dissolved in 45 ml of DMF with stirring Add 3.71 g (26.84 mmol) of potassium carbonate. The suspension was stirred at 25 ° C for 10 minutes and then 995 μl (10.73 mmol) of (bromomethyl)cyclopropane was added. The reaction mixture was stirred at 25 ° C for 16 hours and at 80 ° C for 4 hours. The reaction mixture was partitioned between water and ethyl acetate and the phases were separated. The aqueous phase was extracted with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution and concentrated. The residue was purified by flash chromatography (Biotage SNAP cartridge (340 g; KP-Sil), mobile phase: hexane/ethyl acetate). This gave 2.79 g of the title compound. LC-MS (Method A1): Rt = 1.37 min (UV detector: TIC Smooth), mass 389.00 1H NMR (400 MHz, chloroform-d): δ = 0.36 - 0.44 (m, 2 H), 0.66 - 0.74 (m, 2 H), 1.28 (t, 3 H), 1.32 - 1.42 (m, 1 H), 1.57 (s, 9 H), 3.91 (d, 2 H), 4.25 (q, 2 H), 5.10 (s, 2 H), 6.92 (s, 1 H), 7.30 (s, 1 H), 7.81 (s, 1 H), 8.28 (br. s., 1 H).Intermediate 11-1 N-(6- Hydroxyl -1H- Carbazole -5- base )-6-( Trifluoromethyl ) Pyridine -2- Formamide 7.20 g (17.5 mmol) of N-[6-(benzyloxy)-1H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide was dissolved in 400 ml of methanol The flask was evacuated and then rinsed with nitrogen (this operation was repeated twice more). 9.29 g (8.73 mmol) of palladium on carbon was added and the flask was evacuated and rinsed with hydrogen. The reaction mixture was hydrogenated at 25 ° C for 3 hours under standard hydrogen pressure. The flask was then evacuated and filled with nitrogen. 5.50 g of ammonium formate was added and the mixture was stirred at 25 ° C for 1 hour. The reaction mixture was filtered through celite and concentrated. This gave 499 mg of the title compound. LC-MS (Method A1): Rt = 0.95 min (UV detector: TIC Smooth), mass 322.00 1H NMR (400 MHz, DMSO-d6): δ = 6.98 (s, 1 H), 7.95 (s, 1 H), 8.17 - 8.23 (m , 1 H), 8.40 (t, 1 H), 8.47 (d, 1 H), 8.72 (s, 1 H), 10.46 (s, 1 H), 10.76 (br. s., 1 H), 12.63 ( s, 1 H).Intermediate 12-1 N-[6- Hydroxyl -2-(3- Hydroxyl -3- Methyl butyl )-2H- Carbazole -5- base ]-6-( Trifluoromethyl ) Pyridine -2- Formamide step A :840 mg (2.58 mmol) 4-[5-Amino-6-(benzyloxy)-2H-indazol-2-yl]-2-methylbutan-2-ol and 542 mg (2.84 mmol) 6-(Trifluoromethyl)pyridine-2-carboxylic acid was dissolved in 50 ml of THF, and 395 mg (2.58 mmol) of 1-hydroxy-1H-benzotriazole hydrate, 990 mg (5.16 mmol) 1-( 3-Dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride and 1.1 ml of triethylamine and the mixture was stirred at 25 ° C for 2 hours. After concentrating the solution, water was added to the formed precipitate and the precipitate was filtered off with suction, washed with water and diethyl ether and dried under reduced pressure. This gave 1.02 g of N-[6-(benzyloxy)-2-(3-hydroxy-3-methylbutyl)-2H-indazol-5-yl]-6-(trifluoromethyl)pyridine 2-carbamamine. LC-MS (Method A1): Rt = 1.37 min (UV detector: TIC Smooth), mass determined 498.00 1H NMR (400 MHz, DMSO-d6): δ = 1.15 (s, 6H), 1.99 - 2.06 (m, 2H), 4.40 - 4.46 (m , 2H), 4.51 (s, 1H), 5.30 (s, 2H), 7.32 (s, 1H), 7.37 - 7.45 (m, 3H), 7.54 - 7.60 (m, 2H), 8.18 (dd, 1H), 8.33 (s, 1H), 8.38 (t, 1H), 8.46 (d, 1H), 8.77 (s, 1H), 10.46 (s, 1H).step B : 940 mg (1.89 mmol) N-[6-(benzyloxy)-2-(3-hydroxy-3-methylbutyl)-2H-indazol-5-yl]-6-(trifluoromethyl) The pyridyl-2-carboxamide was dissolved in 94 ml of methanol, and the flask was evacuated and then flushed with nitrogen (this operation was repeated twice more). 201 mg (0.20 mmol) of palladium on carbon was added and the flask was evacuated and rinsed with hydrogen. The reaction mixture was hydrogenated at 25 ° C for 3 hours under standard hydrogen pressure. The reaction mixture was filtered through celite and washed with methanol, and filtrate was concentrated. This gave 731 mg of N-[6-hydroxy-2-(3-hydroxy-3-methylbutyl)-2H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamidine. amine. LC-MS (Method A1): Rt = 1.03 min (UV detector: TIC Smooth), mass determined 408.00 1H NMR (400 MHz, DMSO-d6): δ = 1.15 (s, 6 H), 1.93 - 2.04 (m, 2 H), 4.28 - 4.44 (m, 2 H), 4.50 (s, 1 H), 6.93 (s, 1 H), 8.21 (d, 1 H), 8.26 (s, 1 H), 8.40 (t, 1 H), 8.47 (d , 1 H), 8.66 (s, 1 H), 10.53 (s, 1 H), 10.61 (s, 1 H).Intermediate 13-1 {5-[( Third butoxycarbonyl ) Amine ]-6- Hydroxyl -2H- Carbazole -2- base } Ethyl acetate 4.50 g (10.6 mmol) of {6-(benzyloxy)-5-[(t-butoxycarbonyl)amino]-2H-indazol-2-yl}acetate (CAS number: 1799835- 24-7) Dissolved in 225 ml of ethanol, and the flask was evacuated and then rinsed with nitrogen (this procedure was repeated twice more). 1.13 g (1.06 mmol) of palladium on carbon was added and the flask was evacuated and rinsed with hydrogen. The reaction mixture was hydrogenated at 25 ° C for 4 hours under standard hydrogen pressure. The reaction mixture was filtered through celite and concentrated. This gave 3.44 g of the title compound. LC-MS (Method A1): Rt = 1.05 min (UV detector: TIC Smooth), mass measurement 335.00 1H NMR (300 MHz, DMSO-d6): δ = 1.21 (t, 3 H), 1.47 (s, 9 H), 4.15 (q, 2 H), 5.24 (s, 2 H), 6.82 (s, 1 H), 7.64 (s, 1 H), 7.93 (s, 1 H), 8.10 - 8.16 (m, 1 H), 10.26 (s, 1 H). Under ExpNo UBOT8758-1Instance 1 1-( Difluoromethyl )-N-[2-(3- Hydroxyl -3- Methyl butyl )-6- Methoxy -2H- Carbazole -5- base ]-1H- Pyrazole -3- Formamide 158 mg (66% pure, 418 μmol) 4-(5-amino-6-methoxy-2H-indazol-2-yl)-2-methylbutan-2-ol (CAS number: 1799835- 12-3), 81.4 mg (502 μmol) 1-(difluoromethyl)-1H-pyrazole-3-carboxylic acid, 191 mg (502 μmol) HATU and 87 μl (500 μmol) N,N-diisopropyl The ethylamine was dissolved in 2 ml of THF. The mixture was stirred at 25 ° C for 23 hours. The reaction mixture was diluted with water and extracted twice with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was purified by preparative HPLC. The combined product fractions were lyophilized. This gave 95.2 mg of the title compound. LC-MS (Method A2): Rt = 1.01 min (UV detector: TIC Smooth), mass measurement 393.001 </ RTI> <RTIgt; 1H), 7.04 (d, 1H), 7.12 (s, 1H), 7.98 (t, 1H), 8.30 (s, 1H), 8.46 (d, 1H), 8.50 (s, 1H), 9.39 (s, 1H) ).Instance 2 N-[2-(3- Hydroxyl -3- Methyl butyl )-6- Methoxy -2H- Carbazole -5- base ]-2-( Trifluoromethyl )-1,3- Oxazole -4- Formamide 158 mg (66% pure, 418 μmol) 4-(5-Amino-6-methoxy-2H-indazol-2-yl)-2-methylbutan-2-ol, 90.9 mg (502 μmol) 2-(Trifluoromethyl)-1,3-oxazole-4-carboxylic acid, 191 mg (502 μmol) HATU and 87 μl (500 μmol) N,N-diisopropylethylamine dissolved in 2 ml THF in. The mixture was stirred at 25 ° C for 23 hours. The reaction mixture was diluted with water and extracted twice with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was purified by preparative HPLC. The combined product fractions were lyophilized. This gave 133 mg of the title compound. LC-MS (Method A3): Rt = 1.12 min (UV detector: TIC Smooth), mass measurement 412.001 </ RTI> <RTIgt; ), 7.13 (s, 1H), 8.32 (s, 1H), 8.44 (s, 1H), 9.23 (s, 1H), 9.41 (s, 1H).Instance 3 6-( Difluoromethyl )-N-[2-(3- Hydroxypropyl )-6- Methoxy -2H- Carbazole -5- base ] Pyridine -2- Formamide 100 mg (0.45 mmol) of 3-(5-amino-6-methoxy-2H-indazol-2-yl)propan-1-ol and 94 mg (0.54 mmol) of 6-(difluoromethyl) Pyridine-2-carboxylic acid was dissolved in 3.0 ml of DMF, and 69 mg (0.45 mmol) of 1-hydroxy-1H-benzotriazole hydrate, 173 mg (0.90 mmol) of 1-(3-dimethylaminopropyl) was added. -3-ethylcarbodiimide hydrochloride and 189 μl (1.36 mmol) of triethylamine and the mixture was stirred at 25 ° C for 27 hours. Water was added and the mixture was extracted three times with ethyl acetate. The combined organic phases were filtered through a hydrophobic filter and concentrated. The residue was purified by preparative HPLC (column: YMC-Triart 5[mu]m 100 x 30 mm). The fractions containing the product are lyophilized. This gave 18 mg of the title compound. LC-MS (Method A1): Rt = 1.02 min (UV detector: TIC Smooth), mass measurement 376.001 H NMR (500 MHz, DMSO-d6): δ = 2.04 (five peaks, 2 H), 3.37 - 3.43 (m, 2 H), 4.00 (s, 3 H), 4.40 (t, 2 H), 4.62 (t, 1 H), 7.15 (t, 1 H), 7.14 (s, 1 H), 7.95 - 8.02 (m, 1 H), 8.26 - 8.36 (m, 3 H), 8.69 (s, 1 H) , 10.55 (s, 1 H).Instance 4 N-[6- Ethoxy -2-(3- Hydroxyl -3- Methyl butyl )-2H- Carbazole -5- base ]-4-( Trifluoromethyl )-1,3- Thiazole -2- Formamide 80 mg (0.3 mmol) of 4-(5-amino-6-ethoxy-2H-indazol-2-yl)-2-methylbutan-2-ol, 90 mg in 2 ml DMF 0.45 mmol) 4-(Trifluoromethyl)-1,3-thiazole-2-carboxylic acid, 63 μl (0.45 mmol) of triethylamine and 174 mg (0.45 mmol) of HATU were stirred overnight at RT. Purification by preparative HPLC gave 95 mg (70%) of desired compound. LC-MS (Method A1): Rt = 1.29 min (UV detector: TIC Smooth), mass 442.00 1H-NMR (400MHz, DMSO-d6): δ = 1.15 (s, 6 H) 1.45 (t, 3H) 1.98 – 2.06 (m, 2 H 4.16 – 4.25 (q, 2 H) 4.38 - 4.48 (m, 2 H) 4.52 (s, 1H) 7.14 (s, 1H) 8.33 ( s, 1 H) 8.46 (s, 1 H) 8.90 (s, 1H) ) 9.92 (s, 1H).Instance 5 N-[6- Ethoxy -2-(3- Hydroxyl -3- Methyl butyl )-2H- Carbazole -5- base ]-2-( Trifluoromethyl )-1,3- Thiazole -4- Formamide 80 mg (0.3 mmol) of 4-(5-amino-6-ethoxy-2H-indazol-2-yl)-2-methylbutan-2-ol, 90 mg in 2 ml DMF 0.45 mmol) 2-(Trifluoromethyl)-1,3-thiazole-4-carboxylic acid, 63 μl (0.45 mmol) of triethylamine and 174 mg (0.45 mmol) of HATU were stirred overnight at RT. Purification by preparative HPLC gave 90 mg (67%) of desired compound. LC-MS (Method A1): Rt = 1.27 min (UV detector: TIC Smooth), mass 442.00 1H-NMR (400MHz, DMSO-d6): δ = 1.15 (s, 6 H) 1.47 (t, 3H) 1.98 – 2.05 (m, 2 H 4.15 – 4.24 (q, 2 H) 4.38 - 4.46 (m, 2 H) 4.53 (s, 1H) 7.11 (s, 1H) 8.31 ( s, 1 H) 8.57 (s, 1 H) 8.87 (s, 1H) ) 9.98 (s, 1H).Instance 6 N-[6- Ethoxy -2-(3- Hydroxyl -3- Methyl butyl )-2H- Carbazole -5- base ]-2- methyl -1,3- Oxazole -5- Formamide 80 mg (0.3 mmol) of 4-(5-amino-6-ethoxy-2H-indazol-2-yl)-2-methylbutan-2-ol, 58 mg (2 mg) in 2 ml DMF 0.45 mmol) 2-methyl-1,3-oxazole-5-carboxylic acid, 63 μl (0.45 mmol) of triethylamine and 174 mg (0.45 mmol) of HATU were stirred overnight at RT. Purification by preparative HPLC gave 80 mg (70%) of desired compound. LC-MS (Method A1): Rt = 0.91 min (UV detector: TIC Smooth), mass 372.00 1H-NMR (400MHz, DMSO-d6): δ = 1.14 (s, 6H), 1.43 (t, 3H), 1.98 – 2.05 (m, 2H ), 2.54 (s, 3H), 4.11 - 4.29 (q, 2H), 4.38 - 4.45 (m, 2H), 4.52 (s, 1H), 7.07 (s, 1H), 7.82 ( s, 1H), 8.17 ( s, 1H), 8.29 (s, 1H), 9.21 (s, 1H).Instance 7 1-( Difluoromethyl )-N-[6- Ethoxy -2-(3- Hydroxyl -3- Methyl butyl )-2H- Carbazole -5- base ]-1H- Pyrazole -3- Formamide 80 mg (0.3 mmol) of 4-(5-amino-6-ethoxy-2H-indazol-2-yl)-2-methylbutan-2-ol, 74 mg (2 mg) in 2 ml DMF 0.45 mmol) 1-(Difluoromethyl)-1H-pyrazole-3-carboxylic acid, 63 μl (0.45 mmol) of triethylamine and 174 mg (0.45 mmol) of HATU were stirred overnight at RT. Purification by preparative HPLC gave 20 mg (16%) of title compound. LC-MS (Method A1): Rt = 1.05 min (UV detector: TIC Smooth), mass 407.00 1H-NMR (400MHz, DMSO-d6): δ = 1.15 (s, 6 H) 1.46 (t, 3H), 1.97 – 2.05 (m, 2 H), 4.16 – 4.25 (q, 2H), 4.37 - 4.45 (m, 2H), 4.52 (s, 1H), 7.03 (m, 1H), 7.10 ( s, 1H), 7.96 (t, 1H), 8.29 (s, 1 H), 8.48 (m, 1H), 8.52 (s, 1H), 9.53 (s, 1H).Instance 8 N-[6- Ethoxy -2-(3- Hydroxyl -3- Methyl butyl )-2H- Carbazole -5- base ]-2- methyl -1,3- Thiazole -5- Formamide 80 mg (0.3 mmol) of 4-(5-amino-6-ethoxy-2H-indazol-2-yl)-2-methylbutan-2-ol, 65 mg (2 ml) in 2 ml DMF 0.45 mmol) 2-methyl-1,3-thiazole-5-carboxylic acid, 63 μl (0.45 mmol) of triethylamine and 174 mg (0.45 mmol) of HATU were stirred overnight at RT. Purification by preparative HPLC gave 70 mg (59%) of desired compound. LC-MS (Method A1): Rt = 1.12 min (UV detector: TIC Smooth), mass measurement 388.00 1H-NMR (400MHz, DMSO-d6): δ = 1.14 (s, 6 H), 1.49 (t, 3 H), 1.96 – 2.06 (m , 2 H), 2.75 (s, 3 H), 4.16 – 4.25 (q, 2 H), 4.36 - 4.45 (m, 2 H), 4.52 (s, 1 H), 7.09 (s, 1 H), 8.29 (m, 2 H), 8.61 (s, 1 H), 10.03 (s, 1 H).Instance 9 2- Cyclopropyl -N-[6- Ethoxy -2-(3- Hydroxyl -3- Methyl butyl )-2H- Carbazole -5- base ]-1,3- Oxazole -4- Formamide 80 mg (0.3 mmol) of 4-(5-amino-6-ethoxy-2H-indazol-2-yl)-2-methylbutan-2-ol, 70 mg (2 ml) in 2 ml DMF 0.45 mmol) 2-cyclopropyl-1,3-oxazole-4-carboxylic acid, 63 μl (0.45 mmol) of triethylamine and 174 mg (0.45 mmol) of HATU were stirred at RT overnight. Purification by preparative HPLC gave 65 mg (53%) of desired compound. LC-MS (Method A1): Rt = 1.15 min (UV detector: TIC Smooth), mass determined 398.00 1H-NMR (400MHz, DMSO-d6): δ = 1.00 – 1.06 (m, 2 H), 1.11 – 1.18 (m, 8 H), 1.48 (t, 3 H), 1.96 – 2.04 (m, 2 H), 2.17 – 2.26 (m, 1 H), 4.15 – 4.23 (q, 2 H), 4.36 - 4.44 (m, 2 H), 4.52 (s , 1 H), 7.09 (s, 1 H), 8.27 (s, 1 H), 8.54 (s, 1 H), 8.64 (s, 1 H), 9.55 (s, 1 H).Instance 10 6- Amine -N-[6- Ethoxy -2-(3- Hydroxyl -3- Methyl butyl )-2H- Carbazole -5- base ] Pyridine -2- Formamide 160 mg (0.6 mmol) of 4-(5-amino-6-ethoxy-2H-indazol-2-yl)-2-methylbutan-2-ol, 126 mg (4 mg) in 4 ml DMF 0.91 mmol) 6-Aminopyridine-4-carboxylic acid, 127 μl (0.91 mmol) of triethylamine and 347 mg (0.91 mmol) of HATU were stirred at RT overnight. Purification by preparative HPLC gave 100 mg (43%) of desired compound. LC-MS (Method A1): Rt = 0.94 min (UV detector: TIC Smooth), mass 383.00 1H-NMR (400MHz, DMSO-d6): δ = 1.15 (s, 6 H), 1.53 (t, 3 H), 1.96 – 2.06 (m , 2 H), 4.16 – 4.27 (q, 2 H), 4.35 - 4.45 (m, 2 H), 4.51 (s br, 1 H), 6.22 (s br, 2 H), 6.72 (d, 1 H) , 7.08 (s, 1 H), 7.31 (d, 1 H), 7.62 (t, 1 H), 8.26 (s, 1 H), 8.66 (s, 1 H), 10.58 (s, 1 H).Instance 11 N-[6- Ethoxy -2-(3- Hydroxyl -3- Methyl butyl )-2H- Carbazole -5- base ]-6-( Trifluoromethyl ) Pyridine -2- Formamide 80 mg (0.3 mmol) of 4-(5-amino-6-ethoxy-2H-indazol-2-yl)-2-methylbutan-2-ol, 87 mg (1.5 mg) in 1.5 ml DMF 0.45 mmol) 6-(Trifluoromethyl)pyridine-2-carboxylic acid, 63 μl (0.45 mmol) of triethylamine and 174 mg (0.45 mmol) of HATU were stirred overnight at RT. Purification by preparative HPLC gave 100 mg (75%) of title compound. LC-MS (Method A1): Rt = 1.25 min (UV detector: TIC Smooth), mass measurement 436.00 1H-NMR (400MHz, DMSO-d6): δ = 1.15 (s, 6 H), 1.51 (t, 3 H) 1.98 – 2.06 (m, 2 H) 4.16 – 4.25 (q, 2 H), 4.38 - 4.46 (m, 2 H), 4.53 (s, 1 H), 7.12 (s, 1 H), 8.22 (d, 1 H) 8.32 (s, 1 H) 8.37 – 8.49 (m, 2 H), 8.70 (s, 1 H), 10.74 (s, 1 H).Instance 12 N-[2-(3- Hydroxyl -3- Methyl butyl )-6-( Oxetane -3- Baseoxy )-2H- Carbazole -5- base ]-6-( Trifluoromethyl ) Pyridine -2- Formamide 150 mg (515 μmol) of 4-[5-amino-6-(oxetan-3-yloxy)-2H-indazol-2-yl]-2-methylbutan-2-ol, 118 mg (618 μmol) 6-(trifluoromethyl)pyridine-2-carboxylic acid, 198 mg (618 μmol) 2-(1H-benzotriazol-1-yl)-1,1,3,3-tetra Methylurea quinone tetrafluoroborate and 110 μl (620 μmol)N ,N - Diisopropylethylamine was dissolved in 3 ml of THF. The mixture was stirred at 25 ° C for 16 hours. The reaction mixture was diluted with water and extracted twice with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was purified by preparative HPLC. The combined product fractions were lyophilized. This gave 117 mg of the title compound. LC-MS (Method A1): Rt = 0.90 min (UV detector: TIC Smooth), mass measurement 464.001 </ RTI> <RTIgt; ), 5.10 (t, 2H), 5.50 (five peaks, 1H), 6.82 (s, 1H), 8.23 (dd, 1H), 8.34 (s, 1H), 8.39 - 8.45 (m, 1H), 8.46 - 8.50 (m, 1H), 8.73 (s, 1H), 10.70 (s, 1H).Instance 13 N-[2-(3- Hydroxyl -3- Methyl butyl )-6-( Oxetane -3- Baseoxy )-2H- Carbazole -5- base ]-2-( Trifluoromethyl )-1,3- Thiazole -4- Formamide 80.0 mg (275 μmol) of 4-[5-amino-6-(oxetan-3-yloxy)-2H-indazol-2-yl]-2-methylbutan-2-ol, 65.0 mg (329 μmol) 2-(trifluoromethyl)-1,3-thiazole-4-carboxylic acid, 125 mg (329 μmol) HATU and 57 μl (330 μmol)N ,N -Diisopropylethylamine was dissolved in 2 ml of THF. The mixture was stirred at 25 ° C for 16.5 hours. The reaction mixture was diluted with water and extracted twice with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was purified by preparative. The combined product fractions were lyophilized. This gave 64.1 mg of the title compound. LC-MS (Method A3): Rt = 0.90 min (UV detector: TIC Smooth), mass measurement 470.001 </ RTI> <RTIgt; ), 5.07 (t, 2H), 5.44 - 5.51 (m, 1H), 6.80 (s, 1H), 8.33 (s, 1H), 8.56 (s, 1H), 8.88 (s, 1H), 9.96 (s, 1H).Instance 14 N-[2-(3- Hydroxyl -3- Methyl butyl )-6-( Oxetane -3- Baseoxy )-2H- Carbazole -5- base ]-4-( Trifluoromethyl )-1,3- Thiazole -2- Formamide 80.0 mg (275 μmol) of 4-[5-amino-6-(oxetan-3-yloxy)-2H-indazol-2-yl]-2-methylbutan-2-ol, 65.0 mg (329 μmol) 4-(trifluoromethyl)-1,3-thiazole-2-carboxylic acid, 125 mg (329 μmol) HATU and 57 μl (330 μmol)N ,N -Diisopropylethylamine was dissolved in 2 ml of THF. The mixture was stirred at 25 ° C for 16.5 hours. The reaction mixture was diluted with water and extracted twice with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was purified by preparative HPLC. The combined product fractions were lyophilized. This gave 61 mg of the title compound. LC-MS (Method A3): Rt = 0.95 min (UV detector: TIC Smooth), mass measurement 470.001 </ RTI> <RTIgt; ,,,,,,,,,,,,,, 1H).Instance 15 1-( Difluoromethyl )-N-[2-(3- Hydroxyl -3- Methyl butyl )-6-( Oxetane -3- Methoxy )-2H- Carbazole -5- base ]-1H- Pyrazole -3- Formamide 113 mg (63% pure, 233 μmol) 4-[5-amino-6-(oxetan-3-ylmethoxy)-2H-indazol-2-yl]-2-methylbutyl -2-ol, 45.3 mg (280 μmol) 1-(difluoromethyl)-1H-pyrazole-3-carboxylic acid, 89.8 mg (280 μmol) 2-(1H-benzotriazol-1-yl)- 1,1,3,3-tetramethyluronium tetrafluoroborate and 49 μl (280 μmol)N ,N -Diisopropylethylamine was dissolved in 2 ml of THF. The mixture was stirred at 25 ° C for 67.5 hours. The reaction mixture was diluted with water and extracted three times with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was purified by preparative HPLC. The combined product fractions were lyophilized. This gave 66.5 mg of the title compound. LC-MS (Method A3): Rt = 0.95 min (UV detector: TIC Smooth), mass measurement 449.001 H NMR (400MHz, DMSO-d6): δ = 1.15 (s, 6H), 1.97 - 2.07 (m, 2H), 3.46 - 3.66 (m, 1H), 4.33 - 4.47 (m, 4H), 4.53 (s, 1H), 4.58 (t, 2H), 4.78 (dd, 2H), 7.02 (d, 1H), 7.17 (s, 1H), 7.90 (t, 1H), 8.31 (s, 1H), 8.45 (d, 1H) ), 8.50 (s, 1H), 9.58 (s, 1H).Instance 16 6-( Difluoromethyl )-N-[2-(3- Hydroxyl -3- Methyl butyl )-6-( Oxetane -3- Methoxy )-2H- Carbazole -5- base ] Pyridine -2- Formamide 113 mg (63% pure, 233 μmol) 4-[5-amino-6-(oxetan-3-ylmethoxy)-2H-indazol-2-yl]-2-methylbutyl -2-ol, 48.4 mg (280 μmol) 6-(difluoromethyl)-pyridine-2-carboxylic acid, 89.8 mg (280 μmol) 2-(1H-benzotriazol-1-yl)-1,1 , 3,3-tetramethyluronium tetrafluoroborate and 49 μl (280 μmol)N ,N -Diisopropylethylamine was dissolved in 2 ml of THF. The mixture was stirred at 25 ° C for 67.5 hours. The reaction mixture was diluted with water and extracted three times with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was purified by preparative HPLC. The combined product fractions were lyophilized. This gave 66.0 mg of the title compound. LC-MS (Method A3): Rt = 1.07 min (UV detector: TIC Smooth), mass measurement 460.001 H NMR (400MHz, DMSO-d6): δ = 1.15 (s, 6H), 1.98 - 2.07 (m, 2H), 3.53 - 3.66 (m, 1H), 4.37 (d, 2H), 4.39 - 4.48 (m, 2H), 4.53 (s, 1H), 4.65 (t, 2H), 4.82 (dd, 2H), 7.12 (t, 1H), 7.19 (s, 1H), 8.00 (dd, 1H), 8.26 - 8.40 (m , 3H), 8.76 (s, 1H), 10.65 (s, 1H).Instance 17 N-{2-(3- Hydroxyl -3- Methyl butyl )-6-[(3S)- Tetrahydrofuran -3- Baseoxy ]-2H- Carbazole -5- base }-2- methyl -1,3- Oxazole -5- Formamide 116 mg (379 μmol) 4-{5-amino-6-[(3S)-tetrahydrofuran-3-yloxy]-2H-indazol-2-yl}-2-methylbutan-2-ol 57.8 mg (455 μmol) 2-methyl-1,3-oxazole-5-carboxylic acid, 173 mg (455 μmol) HATU and 79 μl (460 μmol)N ,N - Diisopropylethylamine was dissolved in 2 ml. The mixture was stirred at 25 ° C for 16.5 hours. The reaction mixture was diluted with water and extracted twice with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was taken up in 2 mL DMF and purified by preparative HPLC. The combined product fractions were lyophilized. This gave 96.9 mg of the title compound. LC-MS (Method A3): Rt = 0.93 min (UV detector: TIC Smooth), mass measurement 414.001 </ RTI> <RTIgt; , 3 H), 3.79 - 3.92 (m, 2H), 3.95 - 4.04 (m, 2H), 4.38 - 4.46 (m, 2H), 4.52 (s, 1H), 5.23 (dd, 1H), 7.12 (s, 1H), 8.30 (s, 1H), 8.56 (s, 1H), 8.68 (s, 1H), 9.47 (s, 1H).Instance 18 N-{2-(3- Hydroxyl -3- Methyl butyl )-6-[(3S)- Tetrahydrofuran -3- Baseoxy ]-2H- Carbazole -5- base }-6-( Trifluoromethyl ) Pyridine -2- Formamide 116 mg (379 μmol) 4-{5-amino-6-[(3S)-tetrahydrofuran-3-yloxy]-2H-indazol-2-yl}-2-methylbutan-2-ol , 87.0 mg (455 μmol) of 6-(trifluoromethyl)pyridine-2-carboxylic acid, 173 mg (455 μmol) of HATU and 79 μl (460 μmol)N ,N - Diisopropylethylamine was dissolved in 2 ml. The mixture was stirred at 25 ° C for 16.5 hours. The reaction mixture was diluted with water and extracted three times with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was taken up in 2 mL DMF and purified by preparative HPLC. The combined product fractions were lyophilized. This gave 105 mg of the title compound. LC-MS (Method A3): Rt = 1.14 min (UV detector: TIC Smooth), mass measurement 478.001 </ RTI> <RTIgt; (m, 3H), 4.06 (dd, 1H), 4.39 - 4.46 (m, 2H), 4.54 (s, 1H), 7.12 (s, 1H), 8.22 (dd, 1H), 8.33 (s, 1H), 8.38 - 8.49 (m, 2H), 8.73 (s, 1H), 10.63 (s, 1H).Instance 19 6- Amine -N-{2-(3- Hydroxyl -3- Methyl butyl )-6-[(3S)- Tetrahydrofuran -3- Baseoxy ]-2H- Carbazole -5- base } Pyridine -2- Formamide 116 mg (379 μmol) 4-{5-amino-6-[(3S)-tetrahydrofuran-3-yloxy]-2H-indazol-2-yl}-2-methylbutan-2-ol 62.9 mg (455 μmol) 6-aminopyridine-2-carboxylic acid, 173 mg (455 μmol) HATU and 79 μl (460 μmol)N ,N - Diisopropylethylamine was dissolved in 2 ml. The mixture was stirred at 25 ° C for 16.5 hours. The reaction mixture was diluted with water and extracted three times with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was purified by preparative HPLC. The combined product fractions were lyophilized. This gave 18.9 mg of the title compound. LC-MS (Method A3): Rt = 0.89 min (UV detector: TIC Smooth), mass measurement 425.001 </ RTI> <RTIgt; , 3H), 4.36 - 4.47 (m, 2H), 4.53 (s, 1H), 5.25 (br d, 1H), 6.17 (s, 2H), 6.73 (dd, 1H), 7.12 (s, 1H), 7.31 (dd, 1H), 7.63 (dd, 1H), 8.29 (s, 1H), 8.66 (s, 1H), 10.61 (s, 1H).Instance 20 N-[6- chlorine -2-(3- Methoxy -3- Methyl butyl )-2H- Carbazole -5- base ]-6-(2- Hydroxypropyl -2- base ) Pyridine -2- Formamide 150 mg (20% pure, 112 μmol) of 6-chloro-2-(3-methoxy-3-methylbutyl)-2H-indazole-5-amine, 36.9 mg (168 μmol) 6-( Potassium 2-hydroxyprop-2-yl)pyridine-2-carboxylate (see WO2015091426), 46.9 mg (123 μmol) HATU and 21 μl (120 μmol)N ,N - Diisopropylethylamine was dissolved in 2 ml of DMF. The mixture was stirred at 25 ° C for 17.5 hours. The reaction mixture was diluted with water and extracted twice with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was purified by preparative HPLC. The combined product fractions were lyophilized. This gave 33.5 mg of the title compound. LC-MS (Method A3): Rt = 1.22 min (UV detector: TIC Smooth), mass measurement 430.001 H-NMR (400MHz, DMSO-d6): δ = 1.17 (s, 6H), 1.57 (s, 6H), 2.09 - 2.17 (m, 2H), 3.14 (s, 3H), 4.41 - 4.50 (m, 2H ), 5.49 (s, 1H), 7.92 - 7.98 (m, 2H), 8.01 - 8.12 (m, 2H), 8.53 (d, 1H), 8.71 (s, 1H), 10.88 (s, 1H).Instance twenty one N-[6- chlorine -2-(3- Methoxy -3- Methyl butyl )-2H- Carbazole -5- base ]-6-( Trifluoromethyl ) Pyridine -2- Formamide 150 mg (20% pure, 112 μmol) 6-chloro-2-(3-methoxy-3-methylbutyl)-2H-indazole-5-amine, 32.1 mg (168 μmol) 6-( Trifluoromethyl)pyridine-2-carboxylic acid, 46.9 mg (123 μmol) HATU and 21 μl (120 μmol)N ,N - Diisopropylethylamine was dissolved in 2 ml of DMF. The mixture was stirred at 25 ° C for 17.5 hours. The reaction mixture was diluted with water and extracted twice with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was purified by preparative HPLC. The combined product fractions were lyophilized. This gave 48 mg of the title compound. LC-MS (Method A3): Rt = 1.43 min (UV detector: TIC Smooth), mass measurement 440.00 1H-NMR (400MHz, DMSO-d6): δ = 1.17 (s, 6H), 2.10 - 2.17 (m, 2H), 3.14 (s, 3H ), 4.43 - 4.50 (m, 2H), 7.93 (s, 1H), 8.24 (dd, 1H), 8.39 - 8.45 (m, 1H), 8.45 - 8.50 (m, 1H), 8.54 (s, 1H), 8.63 (s, 1H), 10.52 (s, 1H).Instance twenty two N-[6- Isopropoxy -2-(2- Methoxyethyl )-2H- Carbazole -5- base ]-6-( Trifluoromethyl ) Pyridine -2- Formamide 150 mg (0.41 mmol) N-(6-isopropoxy-1H-indazol-5-yl)-6-(trifluoromethyl)pyridine-2-carboxamide, 171 mg (1.24 mmol) Potassium and 34 mg (0.21 mmol) of potassium iodide were suspended in 2.0 ml of DMF, and the mixture was stirred at 25 ° C for 30 minutes. Subsequently, 58 μl (0.62 mmol) of 2-bromoethyl methyl ether was added, and the mixture was stirred at 25 ° C for 16 hours and at 80 ° C for 48 hours. The reaction mixture was partitioned between ethyl acetate and water. Separate the phases. The organic phase was washed twice with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was purified by preparative HPLC. Freeze dry product fractions. This gave 41 mg of the title compound. LC-MS (Method A1): Rt = 1.31 min (UV detector: TIC Smooth), mass 422.00 1H NMR (400 MHz, DMSO-d6): δ = 1.41 (d, 6 H), 3.31 (s, 3 H), 3.81 (t, 2 H), 4.50 (t, 2 H), 4.84 (dt, 1 H), 7.19 (s, 1 H), 8.21 (dd, 1 H), 8.28 (s, 1 H), 8.37 - 8.43 (m, 1 H), 8.43 - 8.48 (m, 1 H), 8.72 (s, 1 H), 10.73 (s, 1 H).Instance twenty three N-[6- Isopropoxy -2-(3- Methoxypropyl )-2H- Carbazole -5- base ]-6-( Trifluoromethyl ) Pyridine -2- Formamide 150 mg (0.41 mmol) N-(6-isopropoxy-1H-indazol-5-yl)-6-(trifluoromethyl)pyridine-2-carboxamide, 170 mg (1.24 mmol) Potassium and 34 mg (0.21 mmol) of potassium iodide were suspended in 2.0 ml of DMF, and the mixture was stirred at 25 ° C for 30 minutes. Subsequently, 69 μl (0.62 mmol) of 1-bromo-3-methoxypropane was added, and the mixture was stirred at 25 ° C for 16 hours and at 80 ° C for 24 hours. The reaction mixture was partitioned between ethyl acetate and water. Separate the phases. The organic phase was washed twice with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was purified by preparative HPLC. Freeze dry product fractions. This gave 45 mg of the title compound. LC-MS (Method A1): Rt = 1.35 min (UV detector: TIC Smooth), mass measurement 436.00 1H NMR (400 MHz, DMSO-d6): δ = 1.41 (d, 6 H), 2.12 (five peaks, 2 H), 3.24 (s , 3 H), 3.32 - 3.35 (m, 2 H), 4.39 (t, 2 H), 4.84 (five peaks, 1 H), 7.19 (s, 1 H), 8.21 (dd, 1 H), 8.28 (s, 1 H), 8.40 (t, 1 H), 8.46 (d, 1 H), 8.72 (s, 1 H), 10.73 (s, 1 H).Instance twenty four 6-( Difluoromethyl )-N-[6- Ethoxy -2-(3- Hydroxyl -3- Methyl butyl )-2H- Carbazole -5- base ] Pyridine -2- Formamide Dissolve 150 mg (89% pure, 402 μmol) of 6-(difluoromethyl)-N-(6-ethoxy-1H-indazol-5-yl)pyridine-2-carboxamide in 4 ml DMF Medium and 101 mg (603 μmol) of 4-bromo-2-methylbutan-2-ol, 167 mg (1.21 mmol) of potassium carbonate and 100 mg (603 μmol) of potassium iodide were added under stirring. The suspension was stirred at 120 ° C for 6 hours. The reaction mixture was diluted with water and extracted three times with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was purified by preparative HPLC. The combined product fractions were lyophilized. This gave 36.6 mg of the title compound. LC-MS (Method A1): Rt = 1.19 min (UV detector: TIC Smooth), mass measurement 418.001 </ RTI> <RTIgt; 2H), 4.50 (s, 1H), 6.93 - 7.27 (m, 2H), 7.98 (dd, 1H), 8.27 - 8.36 (m, 3H), 8.69 (s, 1H), 10.78 (s, 1H).Instance 25 N-[6-( Cyclopropylmethoxy )-2-(3- Methoxypropyl )-2H- Carbazole -5- base ]-6-( Trifluoromethyl ) Pyridine -2- Formamide 200 mg (0.53 mmol) N-[6-(cyclopropylmethoxy)-1H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide, 220 mg ( 1.59 mmol) potassium carbonate and a spatula potassium iodide were suspended in 3.0 ml of DMF, and the mixture was stirred at 25 ° C for 30 minutes. Subsequently, 89 μl (0.80 mmol) of 1-bromo-3-methoxypropane was added, and the mixture was stirred at 25 ° C for 72 hours. The reaction mixture was partitioned between ethyl acetate and water. Separate the phases. The organic phase was washed twice with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was purified by preparative HPLC. Freeze dry product fractions. This gave 25 mg of the title compound. LC-MS (Method A1): Rt = 1.37 min (UV detector: TIC Smooth), mass measurement 448.001 H-NMR (400 MHz, DMSO-d6): δ = 0.41 - 0.47 (m, 2 H), 0.60 - 0.69 (m, 2 H), 1.30 - 1.40 (m, 1 H), 2.12 (five peaks, 2 H), 3.24 (s, 3 H), 3.30 (t, 2 H), 4.02 (d, 2 H), 4.38 (t, 2 H), 7.09 (s, 1 H), 8.21 (dd, 1 H ), 8.28 (s, 1 H), 8.41 (t, 1 H), 8.47 (d, 1 H), 8.74 (s, 1 H), 10.69 (s, 1 H).Instance 26 N-[6-( Cyclopropylmethoxy )-2-(2- Methoxyethyl )-2H- Carbazole -5- base ]-6-( Trifluoromethyl ) Pyridine -2- Formamide 280 mg (0.74 mmol) N-[6-(cyclopropylmethoxy)-1H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide, 308 mg ( 2.23 mmol) potassium carbonate and 148 mg (0.89 mmol) of potassium iodide were suspended in 5.0 ml of DMF, and the mixture was stirred at 25 ° C for 30 minutes. Subsequently, 105 μl (1.12 mmol) of 2-bromomethyl methyl ether was added, and the mixture was stirred at 25 ° C for 72 hours. The reaction mixture was partitioned between ethyl acetate and water. Separate the phases. The organic phase was washed twice with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was purified by preparative HPLC. Freeze dry product fractions. This gave 50 mg of the title compound. LC-MS (Method A1): Rt = 1.33 min (UV detector: TIC Smooth), mass 434.00 1H NMR (400 MHz, DMSO-d6): δ = 0.39 - 0.47 (m, 2 H), 0.63 - 0.69 (m, 2 H), 1.29 - 1.40 (m, 1 H), 3.23 (s, 3 H), 3.80 (t, 2 H), 4.02 (d, 2 H), 4.49 (t, 2 H), 7.08 (s, 1 H), 8.21 (dd, 1 H), 8.26 - 8.29 (m, 1 H), 8.38 - 8.44 (m, 1 H), 8.45 - 8.50 (m, 1 H), 8.74 (s, 1 H), 10.70 (s, 1 H).Instance 27 N-[6-( Cyclopropylmethoxy )-2-( Oxetane -3- Methyl )-2H- Carbazole -5- base ]-6-( Trifluoromethyl ) Pyridine -2- Formamide 200 mg (0.53 mmol) N-[6-(cyclopropylmethoxy)-1H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide, 220 mg ( 1.59 mmol) potassium carbonate and a spatula potassium iodide were suspended in 3.0 ml of DMF, and the mixture was stirred at 25 ° C for 30 minutes. Subsequently, 120 mg (0.80 mmol) of 3-bromomethyloxetane was added, and the mixture was stirred at 25 ° C for 72 hours. The reaction mixture was partitioned between ethyl acetate and water. Separate the phases. The organic phase was washed twice with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was purified by preparative HPLC according to Method P4. Freeze dry product fractions. Method P2 was used by preparative HPLC (column: YMC Triart C18 5 μm 100×30 mm, gradient: 0–0.5 min 25 ml/min to 70 ml/min 52% B; 0.5-5.5 min 52-61% B; Flow rate: 70 ml/min) The lyophilizate was repurified. Freeze dry product fractions. This gave 14 mg of the title compound. LC-MS (Method A1): Rt =1.28 min (UV detector: TIC Smooth), mass measurement 446.00 1H NMR (400 MHz, DMSO-d6): δ = 0.38 - 0.47 (m, 2 H), 0.59 - 0.70 (m, 2 H), 1.29 - 1.40 (m, 1 H), 3.44 - 3.58 (m, 1 H), 4.02 (d, 2 H), 4.47 (t, 2 H), 4.62 - 4.70 (m, 4 H), 7.08 (s, 1 H), 8.18 - 8.24 (m, 1 H), 8.33 (s, 1 H), 8.37 - 8.45 (m, 1 H), 8.45 - 8.50 (m, 1 H), 8.73 (s, 1 H), 10.69 (s, 1 H).Instance 28 N-[2-(3- Hydroxyl -3- Methyl butyl )-6-( Trifluoromethoxy )-2H- Carbazole -5- base ]-6-( Trifluoromethyl ) Pyridine -2- Formamide Dissolve 406 mg (94% pure, 978 μmol) of N-[6-(trifluoromethoxy)-1H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide 245 mg (1.47 mmol) of 4-bromo-2-methylbutan-2-ol, 405 mg (2.93 mmol) of potassium carbonate and 244 mg (1.47 mmol) of potassium iodide were added in 5 ml of DMF with stirring. The suspension was stirred at 120 ° C for 19 hours. The reaction mixture was diluted with water and extracted three times with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was purified by preparative HPLC. The combined product fractions were lyophilized. This gave 19 mg of the title compound. LC-MS (Method A1): Rt = 1.33 min (UV detector: TIC Smooth), mass measurement 476.001 </ RTI> <RTIgt; ), 8.39 - 8.43 (m, 1H), 8.46 (t, 1H), 8.56 (s, 1H), 8.69 (s, 1H), 10.38 (s, 1H).Instance 29 2- Cyclopropyl -N-[6- Methoxy -2-(3- Methoxypropyl )-2H- Carbazole -5- base ]-1,3- Oxazole -4- Formamide Dissolve 378 mg (0.63 mmol) of 2-cyclopropyl-N-(6-methoxy-1H-indazol-5-yl)-1,3-oxazol-4-carboxamide in 4 ml of DMF And, under stirring, 108 μl (0.95 mmol) of 1-bromo-3-methoxypropane, 263 mg (1.90 mmol) of potassium carbonate and 158 mg (0.95 mmol) of potassium iodide were added. The reaction mixture was stirred at 100 ° C for 23 hours. Subsequently, the mixture was diluted with water and extracted with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was purified by preparative HPLC. The fractions containing the product are lyophilized. This gave 36 mg of the title compound. LC-MS (Method A4): Rt = 1.12 min (UV detector: TIC Smooth), mass measurement 370.001 </ RTI> <RTIgt; (s, 3H), 3.27 - 3.30 (m, 2H), 3.97 (s, 3H), 4.38 (t, 2H), 7.11 (s, 1H), 8.24 (s, 1H), 8.54 (s, 1H), 8.60 (s, 1H), 9.34 (s, 1H).Instance 30 N-[6- chlorine -2-(3- Hydroxyl -3- Methyl butyl )-2H- Carbazole -5- base ]-6-( Difluoromethyl ) Pyridine -2- Formamide 3.15 g (51% pure, 4.98 mmol) of N-(6-chloro-1H-indazol-5-yl)-6-(difluoromethyl)pyridine-2-carboxamide was dissolved in 20 ml of DMF. 1.25 g (7.47 mmol) of 4-bromo-2-methylbutan-2-ol, 2.06 g (14.9 mmol) of potassium carbonate and 1.24 g (7.47 mmol) of potassium iodide were added with stirring. The suspension was stirred at 120 ° C for 16.5 hours. Another 1.03 mg (7.5 mmol) potassium carbonate and 620 mg (3.8 mmol) potassium iodide were added, and the mixture was stirred at 120 ° C for additional 24 hours. The reaction mixture was diluted with water and extracted three times with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was purified by flash chromatography (Biotage EtOAc EtOAc (EtOAc) The combined product fractions are concentrated. The solid was filtered off with suction, washed three times with diethyl ether and dried. The filtrate was concentrated and the residue was purified by preparative. The combined product fractions were lyophilized. This gave a total of 314 mg of the title compound. LC-MS (Method A1): Rt = 1.17 min (UV detector: TIC Smooth), mass measurement 408.001 </ RTI> <RTIgt; ), 8.03 (dd, 1H), 8.29 - 8.40 (m, 2H), 8.52 (s, 1H), 8.63 (s, 1H), 10.59 (s, 1H).Instance 31 N-[2-(2- Hydroxyethyl )-6- Isopropoxy -2H- Carbazole -5- base ]-6-( Trifluoromethyl ) Pyridine -2- Formamide Step A:Dissolve 200 mg (0.55 mmol) of N-(6-isopropoxy-1H-indazol-5-yl)-6-(trifluoromethyl)pyridine-2-carboxamide in 2 ml of DMF and 228 mg (1.65 mmol) potassium carbonate and 46 mg (0.27 mmol) potassium iodide were added with stirring. The suspension was stirred at 25 ° C for 30 minutes, and then 177 μl (0.82 mmol) of (2-bromoethoxy)(t-butyl)dimethyl decane was added. The reaction mixture was stirred at 25 ° C for 16 hours and at 80 ° C for 4 hours. The reaction mixture was then diluted with water and extracted twice with ethyl acetate. The combined organic phases were filtered through a hydrophobic filter and concentrated. The residue was purified by flash chromatography (EtOAc EtOAc: EtOAc (EtOAc) This gave 56 mg of N-[2-(2-{[t-butyl(dimethyl)indolyl]oxy}ethyl)-6-isopropoxy-2H-indazol-5-yl]- 6-(Trifluoromethyl)pyridine-2-carbamide. LC-MS (Method A1): Rt = 1.71 min (UV detector: TIC Smooth), mass 522.00 1H NMR (400 MHz, DMSO-d6): δ = -0.13 - -0.10 (m, 6 H), 0.76 - 0.79 (m, 9 H) , 1.41 (d, 6 H), 4.03 (t, 2 H), 4.42 (t, 2 H), 4.84 (dt, 1 H), 7.19 (s, 1 H), 8.21 (dd, 1 H), 8.25 (s, 1 H), 8.40 (t, 1 H), 8.46 (d, 1 H), 8.72 (s, 1 H), 10.73 (s, 1 H). Step B:53 mg (0.10 mmol) N-[2-(2-{[T-butyl(dimethyl)indenyl]oxy}ethyl)-6-isopropoxy-2H-indazole-5- The benzyl-6-(trifluoromethyl)pyridine-2-carboxamide was dissolved in 0.5 ml of THF and mixed with a solution of 304 μl (0.30 mmol) of tetrabutylammonium fluoride in THF in 1 M. The reaction mixture was stirred at 25 ° C for 2 hours and added to 10 ml of water, and the formed precipitate was filtered off with suction, washed with water and diethyl ether and dried under reduced pressure. This gave 29 mg of N-[2-(2-hydroxyethyl)-6-isopropoxy-2H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide. LC-MS (Method A1): Rt = 1.19 min (UV detector: TIC Smooth), mass determined 408.00 1H NMR (400 MHz, DMSO-d6): δ = 1.41 (d, 6 H), 3.85 (q, 2 H), 4.38 (t, 2 H), 4.84 (dt, 1 H), 4.94 (t, 1 H), 7.18 (s, 1 H), 8.21 (dd, 1 H), 8.27 (s, 1 H), 8.40 (t, 1 H) , 8.46 (d, 1 H), 8.73 (s, 1 H), 10.73 (s, 1 H).Instance 32 N-[2-(3- Hydroxypropyl )-6- Isopropoxy -2H- Carbazole -5- base ]-6-( Trifluoromethyl ) Pyridine -2- Formamide Step A:Dissolve 200 mg (0.55 mmol) of N-(6-isopropoxy-1H-indazol-5-yl)-6-(trifluoromethyl)pyridine-2-carboxamide in 2 ml of DMF and 228 mg (1.65 mmol) potassium carbonate and 46 mg (0.27 mmol) potassium iodide were added with stirring. The suspension was stirred at 25 ° C for 30 minutes, and then 191 μl (0.82 mmol) of (3-bromopropoxy) (t-butyl) dimethyl decane was added. The reaction mixture was stirred at 25 ° C for 16 hours and at 80 ° C for 48 hours. The reaction mixture was then diluted with water and extracted twice with ethyl acetate. The combined organic phases were filtered through a hydrophobic filter and concentrated. The residue was purified by flash chromatography (EtOAc EtOAc: EtOAc (EtOAc) The crude product obtained was purified by preparative HPLC. This gave 59 mg of N-[2-(3-{[t-butyl(dimethyl)indenyl]oxy}propyl)-6-isopropoxy-2H-indazol-5-yl]- 6-(Trifluoromethyl)pyridine-2-carbamide. LC-MS (Method A1): Rt = 1.75 min (UV detector: TIC Smooth), mass determined 536.00 1H NMR (400 MHz, DMSO-d6): δ = 0.03 (s, 6 H), 0.87 (s, 9 H), 1.41 (d, 6 H), 2.09 (five peaks, 2 H), 3.60 (t, 2 H), 4.40 (t, 2 H), 4.84 (five peaks, 1 H), 7.18 (s, 1 H), 8.21 (dd , 1 H), 8.26 (s, 1 H), 8.40 (t, 1 H), 8.46 (d, 1 H), 8.72 (s, 1 H), 10.73 (s, 1 H). Step B:59 mg (0.11 mmol) N-[2-(3-{[T-butyl(dimethyl)indenyl]oxy}propyl)-6-isopropoxy-2H-indazole-5- The group]-6-(trifluoromethyl)pyridine-2-carboxamide was dissolved in 0.5 ml of THF and mixed with 330 μl (0.33 mmol) of tetrabutylammonium fluoride in 1 M solution in THF. The reaction mixture was stirred at 25 ° C for 90 minutes and added to 10 ml of water, and the formed precipitate was filtered off with suction, washed with water and diethyl ether and dried under reduced pressure. This gave 15 mg of N-[2-(3-hydroxypropyl)-6-isopropoxy-2H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide. 1H NMR (400 MHz, DMSO-d6): δ = 1.41 (d, 6 H), 1.99 - 2.09 (m, 2 H), 3.41 (q, 2 H), 4.40 (t, 2 H), 4.62 (t , 1 H), 4.79 - 4.88 (m, 1 H), 7.18 (s, 1 H), 8.21 (dd, 1 H), 8.28 (s, 1 H), 8.40 (t, 1 H), 8.46 (d , 1 H), 8.72 (s, 1 H), 10.73 (s, 1 H).Instance 33 N-2-(3- Hydroxyl -3- Methyl butyl )-6-[3-( Methylsulfonyl ) Propoxy ]-2H- Carbazole -5- base -6-( Trifluoromethyl ) Pyridine -2- Formamide 50 mg (0.12 mmol) of N-[6-hydroxy-2-(3-hydroxy-3-methylbutyl)-2H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2 - Methionine was dissolved in 735 μl of DMF and 33 mg (0.24 mmol) of potassium carbonate and 29 mg (0.15 mmol) of 1-bromo-3-(methylsulfonyl)propane were added with stirring. The reaction mixture was stirred in a microwave at 100 ° C for 60 minutes. Water was added and the mixture was extracted with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was dissolved in 2 mL EtOAc and purified by preparative HPLC. Freeze dry product fractions. This gave 46 mg of the title compound. LC-MS (Method A1): Rt = 1.04 min (UV detector: TIC Smooth), mass determined 528.00 1H NMR (300 MHz, DMSO-d6): δ = -1.15 (s, 6 H), 1.95 - 2.07 (m, 2 H), 2.23 - 2.37 (m, 2 H), 3.01 (s, 3 H), 3.34 - 3.40 (m, 2 H), 4.30 (t, 2 H), 4.37 - 4.46 (m, 2 H), 4.51 (s, 1 H ), 7.13 (s, 1 H), 8.22 (d, 1 H), 8.33 (s, 1 H), 8.41 (t, 1 H), 8.47 (d, 1 H), 8.71 (s, 1 H), 10.55 (s, 1 H).Instance 34 N-{2-(3- Hydroxyl -3- Methyl butyl )-6-[2-( Methylthio ) Ethoxy ]-2H- Carbazole -5- base }-6-( Trifluoromethyl ) Pyridine -2- Formamide 50 mg (0.12 mmol) of N-[6-hydroxy-2-(3-hydroxy-3-methylbutyl)-2H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2 - Promethamine was dissolved in 1 ml of DMF and 33 mg (0.24 mmol) of potassium carbonate and 23 mg (0.15 mmol) of 2-bromoethylmethyl sulfide were added with stirring. The reaction mixture was stirred in a microwave at 100 ° C for 60 minutes. Water was added and the mixture was extracted with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was purified by preparative HPLC. Freeze dry product fractions. This gave 32 mg of the title compound. LC-MS (Method A1): Rt = 1.26 min (UV detector: TIC Smooth), mass 482.00 1H NMR (400 MHz, DMSO-d6): δ = 1.15 (s, 6 H), 1.97 - 2.06 (m, 2 H), 2.15 (s , 3 H), 2.98 (t, 2 H), 4.35 (t, 2 H), 4.39 - 4.46 (m, 2 H), 4.51 (s, 1 H), 7.19 (s, 1 H), 8.22 (dd , 1 H), 8.32 (s, 1 H), 8.40 (t, 1 H), 8.47 (d, 1 H), 8.71 (s, 1 H), 10.60 (s, 1 H).Instance 35 {[2-(3- Hydroxyl -3- Methyl butyl )-5-({[6-( Trifluoromethyl ) Pyridine -2- base ] Carbonyl } Amine )-2H- Carbazole -6- base ] Oxyl } Ethyl acetate 170 mg (0.42 mmol) of N-[6-hydroxy-2-(3-hydroxy-3-methylbutyl)-2H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2 - Methionine was dissolved in 2.5 ml of DMF and 115 mg (0.83 mmol) of potassium carbonate and 56 μl (0.50 mmol) of ethyl bromoacetate were added with stirring. The reaction mixture was stirred in a microwave at 100 ° C for 60 minutes. Water was added and the mixture was extracted with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was purified by preparative HPLC. Freeze dry product fractions. This gave 143 mg of the title compound. LC-MS (Method A1): Rt = 1.19 min (UV detector: TIC Smooth), mass 494.00 1H NMR (400 MHz, DMSO-d6): δ = 1.15 (s, 6 H), 1.21 (t, 3 H), 1.95 - 2.05 (m , 2 H), 4.20 (q, 2 H), 4.39 - 4.46 (m, 2 H), 4.50 (s, 1 H), 5.00 (s, 2 H), 7.18 (s, 1 H), 8.21 (dd , 1 H), 8.34 (s, 1 H), 8.40 (t, 1 H), 8.48 (d, 1 H), 8.72 (s, 1 H), 10.54 (s, 1 H).Instance 36 N-[2-(3- Hydroxyl -3- Methyl butyl )-6-( Oxetane -3- Methoxy )-2H- Carbazole -5- base ]-6-( Trifluoromethyl ) Pyridine -2- Formamide 75 mg (0.18 mmol) of N-[6-hydroxy-2-(3-hydroxy-3-methylbutyl)-2H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2 - Methionine was dissolved in 1.1 ml of DMF and 50 mg (0.37 mmol) of potassium carbonate and 42 mg (0.28 mmol) of 3-(bromomethyl)oxetane were added with stirring. The reaction mixture was stirred in a microwave at 100 ° C for 60 minutes. Water was added and the mixture was extracted with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was purified by preparative HPLC. Freeze dry product fractions. This gave 50 mg of the title compound. LC-MS (Method A1): Rt = 1.12 min (UV detector: TIC Smooth), mass determined 478.00 1H NMR (400 MHz, DMSO-d6): δ = 1.15 (s, 6 H), 1.98 - 2.06 (m, 2 H), 3.43 - 3.54 (m, 1 H), 4.33 - 4.56 (m), 4.79 (dd, 2 H), 7.20 (s, 1 H), 8.21 (dd, 1 H), 8.32 (s, 1 H), 8.36 - 8.44 ( m, 1 H), 8.44 - 8.48 (m, 1 H), 8.72 (s, 1 H), 10.44 (s, 1 H).Instance 37 N-[6-( Cyclopropylmethoxy )-2-(3- Hydroxyl -3- Methyl butyl )-2H- Carbazole -5- base ]-6-( Trifluoromethyl ) Pyridine -2- Formamide 68 mg (0.17 mmol) of N-[6-hydroxy-2-(3-hydroxy-3-methylbutyl)-2H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2 - Promethamine was dissolved in 1 ml of DMF and 46 mg (0.33 mmol) of potassium carbonate and 19 μl (0.20 mmol) of (bromomethyl)cyclopropane were added with stirring. The suspension was stirred in a microwave at 100 ° C for 60 minutes, and then 431 μl (4.65 mmol) of (bromomethyl)cyclopropane was added. The reaction mixture was stirred at 25 ° C for 16 hours. The reaction mixture was partitioned between ethyl acetate and water. Separate the phases. The organic phase was washed twice with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was purified by preparative HPLC. Freeze dry product fractions. This gave 45 mg of the title compound. LC-MS (Method A1): Rt = 1.33 min (UV detector: TIC Smooth), mass 462.00 1H NMR (400 MHz, DMSO-d6): δ = 0.39 - 0.47 (m, 2 H), 0.62 - 0.69 (m, 2 H), 1.15 (s, 6 H), 1.29 - 1.39 (m, 1 H), 1.98 - 2.04 (m, 2 H), 4.02 (d, 2 H), 4.38 - 4.45 (m, 2 H), 4.49 (s, 1 H), 7.07 (s, 1 H), 8.21 (dd, 1 H), 8.30 (s, 1 H), 8.40 (t, 1 H), 8.47 (d, 1 H), 8.73 (s, 1 H) , 10.69 (s, 1 H).Instance 38 N-[6-( Cyclopropylmethoxy )-2-(3- Hydroxypropyl )-2H- Carbazole -5- base ]-6-( Trifluoromethyl ) Pyridine -2- Formamide Step A:280 mg (0.74 mmol) of N-[6-(cyclopropylmethoxy)-1H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2-methyl in 4.2 ml DMF Indoleamine, 308 mg (2.23 mmol) of potassium carbonate and a spatula of potassium iodide were stirred at 25 ° C for 30 minutes. Subsequently, 253 μl (1.49 mmol) of 2-(3-bromopropoxy)tetrahydro-2H-pyran was added, and the mixture was stirred at 25 ° C for 72 hours. The reaction mixture was partitioned between ethyl acetate and water. Separate the phases. The organic phase was washed twice with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was purified by preparative HPLC. This gave 91 mg of N-{6-(cyclopropylmethoxy)-2-[3-(tetrahydro-2H-pyran-2-yloxy)propyl]-2H-indazol-5-yl }-6-(Trifluoromethyl)pyridine-2-carbamide. LC-MS (Method A1): Rt = 1.47 min (UV detector: TIC Smooth), mass determined 518.00 1H NMR (400 MHz, DMSO-d6, selected signal): δ = 0.40 - 0.48 (m, 2 H), 0.61 - 0.69 (m, 2 H), 1.30 - 1.39 (m, 1 H), 1.39 - 1.54 (m, 4 H), 1.56 - 1.67 (m, 1 H), 1.67 - 1.79 (m, 1 H), 2.15 (五重峰, 2 H), 3.36 - 3.44 (m, 1 H), 3.63 (dt, 1 H), 3.73 (ddd, 1 H), 3.98 - 4.07 (m, 2 H), 4.42 (t, 2 H), 4.53 (t , 1 H), 7.08 (s, 1 H), 8.17 - 8.24 (m, 1 H), 8.29 (s, 1 H), 8.41 (t, 1 H), 8.47 (d, 1 H), 8.74 (s , 1 H), 10.69 (s, 1 H). Step B:88 mg (0.17 mmol) N-{6-(cyclopropylmethoxy)-2-[3-(tetrahydro-2H-pyran-2-yloxy)propyl]-2H-carbazole- 5-Base}-6-(trifluoromethyl)pyridine-2-carboxamide was dissolved in 1.1 ml of dichloromethane, and 96 mg (0.51 mmol) of p-toluenesulfonic acid was added. The reaction mixture was stirred at 25 ° C for 19 hours. Saturated sodium bicarbonate solution was added and the mixture was extracted with ethyl acetate. The combined organic phases were filtered through a hydrophobic filter and concentrated. This gave 46 mg of N-[6-(cyclopropylmethoxy)-2-(3-hydroxypropyl)-2H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2- Formamide. LC-MS (Method A1): Rt = 1.22 min (UV detector: TIC Smooth), mass 434.00 1H NMR (400 MHz, DMSO-d6): δ = 0.39 - 0.47 (m, 2 H), 0.62 - 0.70 (m, 2 H), 1.29 - 1.41 (m, 1 H), 2.03 (five peaks, 2 H), 3.36 - 3.42 (m, 2 H), 4.02 (d, 2 H), 4.39 (t, 2 H), 4.63 (t, 1 H), 7.08 (s, 1 H), 8.22 (dd, 1 H), 8.28 (s, 1 H), 8.41 (t, 1 H), 8.48 (d, 1 H), 8.74 (s, 1 H) , 10.70 (s, 1 H).Instance 39 N-[6- Methoxy -2-(3- Oxidyl butyl )-2H- Carbazole -5- base ]-6-( Trifluoromethyl ) Pyridine -2- Formamide Step A:2.50 g (7.4 mmol) of N-(6-methoxy-1H-indazol-5-yl)-6-(trifluoromethyl)pyridine-2-carboxamide (CAS number: 1799836-45-5) Dissolved in 15 ml of DMF and added 1.63 ml (11.9 mmol) of 2-(2-bromoethyl)-2-methyl-1,3-dioxolane, 3.08 g (22.3 mmol) of carbonic acid with stirring. Potassium and 1.85 g (11.2 mmol) of potassium iodide. The reaction mixture was stirred at 100 ° C for 21 hours. Subsequently, the mixture was diluted with water and extracted with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, the phases separated, filtered and concentrated with a pad. Ethyl acetate was added to the residue and the solid formed was filtered. The filtrate was concentrated and purified by flash chromatography (Biotage SNAP cartridge (100 g; KP-Sil), mobile phase: hexane/ethyl acetate). This gave 860 mg of N-{6-methoxy-2-[2-(2-methyl-1,3-dioxolan-2-yl)ethyl]-2H-indazol-5-yl} -6-(Trifluoromethyl)pyridine-2-carboxamide. LC-MS (Method A1): Rt = 1.20 min (UV detector: TIC Smooth), mass measurement 450.00. Step B:855 mg (1.37 mmol) N-{6-methoxy-2-[2-(2-methyl-1,3-dioxolan-2-yl)ethyl]-2H-indazole-5 -yl}-6-(trifluoromethyl)pyridine-2-carboxamide dissolved in 20 ml of dichloromethane, adding 520 mg (2.73 mmol) of p-toluenesulfonic acid monohydrate and stirring the mixture at 25 ° C 67 hours. Another 260 mg (1.36 mmol) of p-toluenesulfonic acid monohydrate was added, and the mixture was stirred at 25 ° C for 24 hours. The solid formed was filtered off and washed three times with dichloromethane. A saturated sodium bicarbonate solution was added and the reaction mixture was stirred briefly. The phases were separated and the aqueous phase was extracted twice with dichloromethane. The combined organic phases were washed with a saturated sodium chloride solution, filtered (hydrophobic filter) and concentrated. Ethyl acetate was added to the residue and the mixture was stirred briefly. The solid was filtered off with suction and washed three times with ethyl acetate and dried. This gave 392 mg of N-[6-methoxy-2-(3-o-oxybutyl)-2H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide . LC-MS (Method A1): Rt = 1.13 min (UV detector: TIC Smooth), mass measurement 406.001 H NMR (400 MHz, DMSO-d6): δ = 2.14 (s, 3 H), 3.15 (t, 2 H), 3.98 (s, 3 H), 4.54 (t, 2 H), 7.13 (s, 1 H), 8.21 (dd, 1 H), 8.28 (s, 1 H), 8.40 (t, 1 H), 8.46 (d, 1 H), 8.67 (s, 1 H), 10.49 (s, 1 H) .Instance 40 N-[2-(2- Hydroxyethyl )-6- Methoxy -2H- Carbazole -5- base ]-6-( Trifluoromethyl ) Pyridine -2- Formamide Step A:7.00 g (20.8 mmol) of N-(6-methoxy-1H-indazol-5-yl)-6-(trifluoromethyl)pyridine-2-carboxamide, 6.60 ml (41.6 mmol) of bromoacetic acid A mixture of benzyl ester and 8.83 ml (41.6 mmol) of N,N-dicyclohexylmethylamine in 100 ml of THF was heated to 70 ° C for 5 hours. Another 3.30 ml (20.8 mmol) of benzyl bromoacetate and 4.42 ml (20.8 mmol) of N,N-dicyclohexylmethylamine were added, and the mixture was stirred at 65 ° C for 20 hours. The precipitated solid was filtered, washed twice with ethyl acetate, washed three times with water and th The filtrate of the reaction mixture was concentrated, ethyl acetate was added and the mixture was stirred for 15 min. The solid was filtered off with suction and washed twice with ethyl acetate and dried. The two batches of solids were combined. This gave 7.08 g of solid [6-methoxy-5-({[6-(trifluoromethyl)pyridin-2-yl]carbonyl}amino)-2H-indazol-2-yl]acetic acid benzyl ester. LC-MS (Method A1): Rt = 1.37 min (UV detector: TIC Smooth), mass measurement 484.001 H NMR (400 MHz, DMSO-d6): δ = 3.99 (s, 3 H), 5.21. (s, 2 H), 5.41 (s, 2 H), 7.14 (s, 1 H), 7.31 - 7.42 (m , 5 H), 8.22 (dd, 1 H), 8.35 (s, 1 H), 8.40 (t, 1 H), 8.47 (d, 1 H), 8.72 (s, 1 H), 10.51 (s, 1 H). Step B:7.08 g (14.6 mmol) of [6-methoxy-5-({[6-(trifluoromethyl)pyridin-2-yl]carbonyl}amino)-2H-indazol-2-yl]benzyl acetate The base ester was dissolved in 110 ml of THF/ethanol (10:1), and a total of 553 mg (14.6 mmol) of sodium borohydride was added a little at a time. The mixture was stirred at 25 ° C for 48 hours. Water was added to the reaction mixture and the aqueous phase was extracted once with ethyl acetate. The solid was filtered off with suction and washed twice with ethyl acetate. The filtrate phase was separated and the aqueous phase was extracted twice more with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution and concentrated. The crude product and the solid which had been filtered were combined, 60 ml diethyl ether was added and the mixture was stirred for 20 min. The solid was filtered off with suction, washed three times with diethyl ether and dried. This gave 4.56 g of N-[2-(2-hydroxyethyl)-6-methoxy-2H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide. LC-MS (Method A1): Rt = 1.07 min (UV detector: TIC Smooth), mass measurement 380.001 H NMR (400 MHz, DMSO-d6): δ = 3.85 (q, 2 H), 3.98 (s, 3 H), 4.38 (t, 2 H), 4.99 (t, 1 H), 7.14 (s, 1 H), 8.19 - 8.23 (m, 1 H), 8.28 (s, 1 H), 8.40 (t, 1 H), 8.46 (d, 1 H), 8.69 (s, 1 H), 10.50 (s, 1 H).Instance 41 Under ExpNo UBOT8717-1, BAY 1752003N-[2-(3- Hydroxypropyl )-6- Methoxy -2H- Carbazole -5- base ]-6-( Trifluoromethyl ) Pyridine -2- Formamide Step A:200 mg (0.60 mmol) of N-(6-methoxy-1H-indazol-5-yl)-6-(trifluoromethyl)pyridine-2-carboxamide (CAS number: 1799836-45-5) Dissolved in 4 ml of DMF and added 151 μl (0.89 mmol) of 2-(3-bromopropoxy)tetrahydro-2H-pyran, 247 mg (1.78 mmol) of potassium carbonate and 118 mg (0.71) with stirring. Methyl) potassium iodide. The reaction mixture was stirred at 100 ° C for 19 hours. The reaction mixture was diluted with water and extracted three times with ethyl acetate. The combined organic phases were filtered through a hydrophobic filter and concentrated. The residue was dissolved in 2 mL of dimethyl hydrazine and purified by preparative HPLC. This gave 50 mg of N-{6-methoxy-2-[3-(tetrahydro-2H-pyran-2-yloxy)propyl]-2H-indazol-5-yl}-6-( Trifluoromethyl)pyridine-2-carboxamide. LC-MS (Method A1): Rt = 1.46 min (UV detector: TIC Smooth), mass measurement 478.00. Step B:50 mg (0.10 mmol) of N-{6-methoxy-2-[3-(tetrahydro-2H-pyran-2-yloxy)propyl]-2H-indazol-5-yl}- 6-(Trifluoromethyl)pyridine-2-carboxamide was dissolved in 4 ml of dichloromethane, 10 mg (0.05 mmol) of p-toluenesulfonic acid monohydrate was added and the mixture was stirred at 25 ° C for 72 hours. Another 50 mg (0.25 mmol) of p-toluenesulfonic acid monohydrate was added, and the mixture was stirred at 25 ° C for 72 hours. The reaction mixture was mixed with a saturated sodium hydrogencarbonate solution and extracted thrice with ethyl acetate. The organic phase was combined by filtration (hydrophobic filter) and concentrated. The residue was dissolved in 2.5 mL of dimethyl hydrazine and purified by preparative HPLC. The fractions containing the product are lyophilized. This gave 30 mg of N-[2-(3-hydroxypropyl)-6-methoxy-2H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide. LC-MS (Method A1): Rt = 1.19 min (UV detector: TIC Smooth), mass measurement 394.001 </ RTI> <RTIgt; 7.14 (s, 1H), 8.20 (dd, 1H), 8.28 (s, 1H), 8.37 - 8.43 (m, 1H), 8.44 - 8.49 (m, 1H), 8.69 (s, 1H), 10.50 (s, 1H).Instance 42 N-{2-[(2S)-2,3- Dihydroxypropyl ]-6- Methoxy -2H- Carbazole -5- base }-6-( Trifluoromethyl ) Pyridine -2- Formamide Step A:Dissolve 200 mg (595 μmol) of N-(6-methoxy-1H-indazol-5-yl)-6-(trifluoromethyl)pyridine-2-carboxamide in 5 ml of DMF and Add 120 μl (890 μmol) of (4R)-4-(chloromethyl)-2,2-dimethyl-1,3-dioxolane, 247 mg (0.89 mmol) potassium carbonate and 148 mg (with stirring) 892 μmol) potassium iodide. The suspension was stirred at 120 ° C for 21 hours. Another 124 mg (4.85 mmol) potassium carbonate and 74 mg (446 μmol) potassium iodide were added, and the mixture was stirred at 120 ° C for an additional 23 hours. The reaction mixture was diluted with water and extracted three times with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was taken up in 3 mL of dimethyl hydrazine and purified by preparative HPLC. This gave 81 mg of N-(2-{[(4S)-2,2-dimethyl-1,3-dioxolan-4-yl]methyl}-6-methoxy-2H-carbazole -5-yl)-6-(trifluoromethyl)pyridine-2-carboxamide. LC-MS (Method A1): Rt = 1.27 min (UV detector: TIC Smooth), mass measurement 450.00. Step B:First, 81.3 mg (180 μmol) of N-(2-{[(4S)-2,2-dimethyl-1,3-dioxolan-4-yl]methyl} was charged in 4 ml of dichloromethane. -6-Methoxy-2H-indazol-5-yl)-6-(trifluoromethyl)pyridine-2-carboxamide. 51.5 mg (271 μmol) of p-toluenesulfonic acid monohydrate was added, and the mixture was stirred at 25 ° C for 19 hours. A saturated sodium bicarbonate solution was added to the reaction mixture, and the solid formed was filtered off with suction and washed three times with water and three times with diethyl ether. The solid was dried under reduced pressure. This gave 66.3 mg of N-{2-[(2S)-2,3-dihydroxypropyl]-6-methoxy-2H-indazol-5-yl}-6-(trifluoromethyl)pyridine- 2-Protonamine. LC-MS (Method A1): Rt = 1.02 min (UV detector: TIC Smooth), mass measurement 410.001 </ RTI> <RTIgt; ), 8.20 (dd, 1H), 8.24 (s, 1H), 8.36 - 8.43 (m, 1H), 8.44 - 8.48 (m, 1H), 8.68 (s, 1H), 10.49 (s, 1H).Instance 43 6-( Difluoromethyl )-N-[2-(2- Hydroxyethyl )-6- Methoxy -2H- Carbazole -5- base ] Pyridine -2- Formamide Step A:400 mg (1.29 mmol) of (5-amino-6-methoxy-2H-indazol-2-yl)acetic acid benzyl ester (CAS number: 1799835-89-4) and 245 mg (1.41 mmol) 6 -(Difluoromethyl)pyridine-2-carboxylic acid was dissolved in 10 ml of THF, and 197 mg (1.29 mmol) of 1-hydroxy-1H-benzotriazole hydrate, 493 mg (2.57 mmol) was added at 25 °C. 1-(3-Dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride and 537 μl (3.85 mmol) of triethylamine. After aqueous work-up, filtration and concentration, the residue was taken in diethyl ether and a little water and allowed to stir for 30 min. The solid was filtered off with suction, washed with diethyl ether and dried. This gave 401 mg of benzyl 5-[{-({[6-(difluoromethyl)pyridin-2-yl]carbonyl}amino)-6-methoxy-2H-indazol-2-yl]acetate. LC-MS (Method A1): Rt = 1.29 min (UV detector: TIC Smooth), mass measurement 466.00. Step B:128 mg (0.27 mmol) of [5-({[6-(difluoromethyl)pyridin-2-yl]carbonyl}amino)-6-methoxy-2H-indazol-2-yl]benzyl acetate The base ester was dissolved in 4.2 ml of THF/methanol (10:1) and 10 mg (0.27 mmol) of sodium borohydride was added. The mixture was stirred at 25 ° C for 21 hours. Water was added to the reaction mixture and the resulting solid was filtered with suction and washed three times with water and three times with diethyl ether. The crude product was dissolved in 2.5 mL of dimethyl hydrazine and purified by preparative HPLC. The fractions containing the product are lyophilized. This gave 37 mg of 6-(difluoromethyl)-N-[2-(2-hydroxyethyl)-6-methoxy-2H-indazol-5-yl]pyridine-2-carboxamide. LC-MS (Method A1): Rt = 0.98 min (UV detector: TIC Smooth), mass measurement 362.001 H NMR (400 MHz, DMSO-d6): δ = 3.85 (q, 2 H), 4.00 (s, 3 H), 4.38 (t, 2 H), 4.95 (t, 1 H), 7.15 (t, 1 H), 7.15 (d, 1 H), 7.94 - 8.02 (m, 1 H), 8.25 - 8.37 (m, 3 H), 8.69 (s, 1 H), 10.55 (s, 1 H).Instance 44 N-[6- chlorine -2-(2- Hydroxy ethane base )-2H- Carbazole -5- base ]-6-( Trifluoromethyl ) Pyridine -2- Formamide Step A:2.50 g (7.4 mmol) N-(6-chloro-1H-indazol-5-yl)-6-(trifluoromethyl)pyridine-2-carboxamide, 75 ml THF, 2.33 ml (14.7 mmol) A mixture of benzyl bromoacetate and 3.11 ml (14.7 mmol) of N,N-dicyclohexylmethylamine was stirred at 65 ° C for 27 hours. 2.33 ml (14.7 mmol) of benzyl bromoacetate and 3.11 ml (14.7 mmol) of N,N-dicyclohexylmethylamine were added, and the mixture was stirred at 65 ° C for 67 hours. The precipitated solid was filtered off and the filtrate was extracted twice with ethyl acetate. The combined organic phases were washed with aq. EtOAc EtOAc EtOAc. The residue was purified by flash chromatography (Biotage SNAP cartridge (100 g; KP-Sil), mobile phase: hexane/ethyl acetate). The crude product was then purified by preparative HPLC. This gave 1.08 g of [6-chloro-5-({[6-(trifluoromethyl)pyridin-2-yl]carbonyl}amino)-2H-indazol-2-yl]acetic acid benzyl ester. LC-MS (Method A1): Rt = 1.46 min (UV detector: TIC Smooth), mass measurement 488.001 H NMR (400 MHz, DMSO-d6): δ = 5.22 (s, 2 H), 5.53 (s, 2 H), 7.33 - 7.41 (m, 5 H), 7.95 (s, 1 H), 8.24 (dd , 1 H), 8.42 (d, 1 H), 8.45 - 8.49 (m, 1 H), 8.53 - 8.55 (m, 1 H), 8.66 (s, 1 H), 10.53 (s, 1 H). Step B:185 mg (0.38 mmol) of [6-chloro-5-({[6-(trifluoromethyl)pyridin-2-yl]carbonyl}amino)-2H-indazol-2-yl]acetic acid benzyl ester Dissolved in 5 ml of THF and added 14 mg (0.38 mmol) of sodium borohydride. The mixture was stirred at 25 ° C for 21.5 hours. Then 500 μl of methanol was added using a pipette, and the mixture was stirred at 25 ° C for 5 hours. Water is added to the reaction mixture. The solid was filtered off with suction, washed twice with water and three portions with diethyl ether and dried under reduced pressure. This gave 144 mg of N-[6-chloro-2-(2-hydroxyethyl)-2H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide. LC-MS (Method A1): Rt = 1.16 min (UV detector: TIC Smooth), mass measurement 384.001 </ RTI> <RTIgt; 8.44 (m, 1H), 8.45 - 8.50 (m, 2H), 8.65 (s, 1H), 10.52 (s, 1H).Instance 45 N-{6- chlorine -2-[(2R)-2,3- Dihydroxypropyl ]-2H- Carbazole -5- base }-6-( Trifluoromethyl ) Pyridine -2- Formamide Step A:Dissolve 300 mg (881 μmol) of N-(6-chloro-1H-indazol-5-yl)-6-(trifluoromethyl)pyridine-2-carboxamide in 4 ml of DMF with stirring Add 180 μl (1.3 mmol) of (4S)-4-(chloromethyl)-2,2-dimethyl-1,3-dioxolane, 365 mg (2.64 mmol) potassium carbonate and 219 mg (1.32 mmol) ) Potassium iodide. The suspension was stirred at 120 ° C for 17 hours. The reaction mixture was diluted with water and extracted three times with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was purified by preparative HPLC. This gave 67.3 mg of N-(6-chloro-2-{[(4R)-2,2-dimethyl-1,3-dioxolan-4-yl]methyl}-2H-indazole-5 -yl)-6-(trifluoromethyl)pyridine-2-carbamide. LC-MS (Method A1): Rt = 1.37 min (UV detector: TIC Smooth), mass measurement 454.00. Step B:67.3 mg (148 μmol) of N-(6-chloro-2-{[(4R)-2,2-dimethyl-1,3-dioxolan-4-yl]methyl}-2H-indole Zyrid-5-yl)-6-(trifluoromethyl)pyridine-2-carboxamide was dissolved in 3 ml of dichloromethane, 42.2 mg (222 μmol) of p-toluenesulfonic acid monohydrate was added and the mixture was at 25 Stir at ° C for 24 hours. A saturated sodium bicarbonate solution was added and the reaction mixture was stirred briefly. The solid was filtered off with suction, washed three times with water and three times with diethyl ether and dried overnight. This gave 57 mg of N-{6-chloro-2-[(2R)-2,3-dihydroxypropyl]-2H-indazol-5-yl}-6-(trifluoromethyl)pyridine-2- Formamide. LC-MS (Method A1): Rt = 1.07 min (UV detector: TIC Smooth), mass measurement 414.001 </ RTI> <RTIgt; , 5.10 (br s, 1H), 7.92 (s, 1H), 8.23 (dd, 1H), 8.36 - 8.51 (m, 3H), 8.62 (s, 1H), 10.51 (s, 1H).Instance 46 N-[2-(3- Hydroxyl -3- Methyl butyl )-6- Methoxy -2H- Carbazole -5- base ]-6-( Trifluoromethyl ) Pyridine -2- Formamide Dissolve 150 mg (446 μmol) of N-(6-methoxy-1H-indazol-5-yl)-6-(trifluoromethyl)pyridine-2-carboxamide in 4 ml of DMF and 112 mg (669 μmol) of 4-bromo-2-methylbutan-2-ol, 185 mg (1.34 mmol) of potassium carbonate and 111 mg (669 μmol) of potassium iodide were added with stirring. The suspension was stirred at 120 ° C for 5.5 hours. The reaction mixture was diluted with water and extracted three times with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was taken up in 2 mL of dimethyl hydrazine and purified by preparative HPLC. The combined product fractions were lyophilized. This gave 41.4 mg of the title compound. LC-MS (Method A1): Rt = 1.19 min (UV detector: TIC Smooth), mass measurement 422.001 </ RTI> <RTIgt; ), 7.15 (s, 1H), 8.22 (dd, 1H), 8.33 (s, 1H), 8.37 - 8.51 (m, 2H), 8.69 (s, 1H), 10.50 (s, 1H).Instance 47 N-[2-(3- Hydroxybutyl )-6- Methoxy -2H- Carbazole -5- base ]-6-( Trifluoromethyl ) Pyridine -2- Formamide ( Mirror isomer 1) Instance 48 N-[2-(3- Hydroxybutyl )-6- Methoxy -2H- Carbazole -5- base ]-6-( Trifluoromethyl ) Pyridine -2- Formamide ( Mirror isomer 2) 192 mg (0.47 mmol) of N-[6-methoxy-2-(3-oxobutyl)-2H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2- The methotrexate was dissolved in 5.5 ml of THF/methanol (10:1) and 18 mg (0.47 mmol) of sodium borohydride was added. The mixture was stirred at 25 ° C for 20 hours. Water was added to the reaction mixture and the aqueous phase was extracted three times with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated. The residue was dissolved in 8 ml of a mixture of ethanol and methanol and purified and purified by preparative chiral HPLC using method P6. The fractions containing the product are lyophilized. This gave 72 mg of the title compound (m.p. 1) and 75 mg of ss. LC-MS (Method A1): Rt = 1.16 min (UV detector: TIC Smooth), mass measurement 408.001 H NMR (300 MHz, DMSO-d6): δ = 1.02 - 1.11 (m, 3 H), 1.80 - 2.07 (m, 2 H), 3.48 - 3.63 (m, 1 H), 3.98 (s, 3 H) , 4.41 (t, 2 H), 4.68 (d, 1 H), 7.15 (s, 1 H), 8.22 (dd, 1 H), 8.30 (s, 1 H), 8.37 - 8.44 (m, 1 H) , 8.44 - 8.50 (m, 1 H), 8.68 (s, 1 H), 10.50 (s, 1 H).Instance 49 N-[6- Methoxy -2-(3- Methoxypropyl )-2H- Carbazole -5- base ]-6-( Trifluoromethyl ) Pyridine -2- Formamide Dissolve 300 mg (0.60 mmol) of N-(6-methoxy-1H-indazol-5-yl)-6-(trifluoromethyl)pyridine-2-carboxamide in 4 ml of DMF and 121 μl (1.07 mmol) of 1-bromo-3-methoxypropane, 370 mg (2.68 mmol) of potassium carbonate and 178 mg (1.07 mmol) of potassium iodide were added with stirring. The reaction mixture was stirred at 100 &lt;0&gt;C for 17 h, diluted with water andEtOAc. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was purified by preparative HPLC. The fractions containing the product are lyophilized. This gave 81 mg of the title compound. LC-MS (Method A1): Rt = 1.28 min (UV detector: TIC Smooth), mass measurement 408.001 H NMR (300 MHz, DMSO-d6): δ = 2.05 (five peaks, 2 H), 3.22 (s, 3 H), 3.27 (t, 2 H), 4.04 (s, 3 H), 4.42 (t , 2 H), 7.31 (s, 1 H), 8.01 (s, 1 H), 8.17 - 8.25 (m, 1 H), 8.40 (t, 1 H), 8.47 (d, 1 H), 8.72 (s , 1 H), 10.43 (s, 1 H).Instance 50 N-[6-(2- Hydroxyethoxy )-2-(3- Hydroxyl -3- Methyl butyl )-2H- Carbazole -5- base ]-6-( Trifluoromethyl ) Pyridine -2- Formamide 40 mg (0.08 mmol) of {[2-(3-hydroxy-3-methylbutyl)-5-({[6-(trifluoromethyl)pyridin-2-yl)carbonyl}amino)-2H Ethyl oxazol-6-yl]oxy}acetate was dissolved in 1.1 ml of THF/methanol (10:1) and 6 mg (0.16 mmol) of sodium borohydride was added. The reaction mixture was stirred at 25 ° C for 2 hours. Water was added and the reaction mixture was extracted with ethyl acetate. The combined organic phases were filtered through a hydrophobic filter and concentrated. This gave 28 mg of the title compound. LC-MS (Method A1): Rt = 1.02 min (UV detector: TIC Smooth), mass 452.00 1H NMR (400 MHz, DMSO-d6): δ = 1.15 (s, 6 H), 1.96 - 2.04 (m, 2 H), 3.88 (q , 2 H), 4.19 (t, 2 H), 4.38 - 4.45 (m, 2 H), 4.51 (s, 1 H), 4.87 (t, 1 H), 7.14 (s, 1 H), 8.21 (dd , 1 H), 8.31 (s, 1 H), 8.40 (t, 1 H), 8.46 (d, 1 H), 8.70 (s, 1 H), 10.65 (s, 1 H).Instance 51 N-[6-( Cyclopropylmethoxy )-2-(2- Hydroxyethyl )-2H- Carbazole -5- base ]-6-( Trifluoromethyl ) Pyridine -2- Formamide 3.00 g (6.49 mmol) of [6-(cyclopropylmethoxy)-5-({[6-(trifluoromethyl)pyridin-2-yl]carbonyl}amino)-2H-indazole-2 Ethyl acetate was dissolved in 33 ml of THF/methanol (10:1) and 245 mg (6.49 mmol) of sodium borohydride was added. The reaction mixture was stirred at 25 ° C for 3 hours. Another 245 mg (6.49 mmol) of sodium borohydride was added and the mixture was stirred at 25 ° C for another 2 hours. Water was added and the reaction mixture was extracted with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. The crude product was stirred with diethyl ether and the solid was filtered with suction. This gave 1.91 g of the title compound. LC-MS (Method A1): Rt = 1.20 min (UV detector: TIC Smooth), mass measurement 420.00 1H NMR (400 MHz, DMSO-d6): δ = 0.40 - 0.47 (m, 2 H), 0.61 - 0.71 (m, 2 H), 1.35 (br. s., 1 H), 3.85 (q, 2 H), 3.99 - 4.05 (m, 2 H), 4.37 (t, 2 H), 4.94 (t, 1 H), 7.08 (s, 1 H ), 8.21 (dd, 1 H), 8.26 (s, 1 H), 8.40 (t, 1 H), 8.47 (d, 1 H), 8.74 (s, 1 H), 10.69 (s, 1 H).Instance 52 N-[6- Methoxy -2-( Oxetane -3- Methyl )-2H- Carbazole -5- base ]-6-( Trifluoromethyl ) Pyridine -2- Formamide Dissolve 1.00 g (2.97 mmol) of N-(6-methoxy-1H-indazol-5-yl)-6-(trifluoromethyl)pyridine-2-carboxamide in 10 ml of DMF and 539 mg (3.57 mmol) of 3-(bromomethyl)oxetane, 1.23 g (8.92 mmol) of potassium carbonate and 592 mg (3.57 mmol) of potassium iodide were added with stirring. The reaction mixture was stirred at 100 ° C for 22.5 hours. Then, the mixture was diluted with water and extracted twice with ethyl acetate. The combined organic phases were filtered through a hydrophobic filter and concentrated. The residue was stirred with EtOAc (EtOAc)EtOAc. This gave 158 mg of the title compound. LC-MS (Method A1): Rt = 1.19 min (UV detector: TIC Smooth), mass measurement 406.001 </ RTI> <RTIgt; ), 8.21 (dd, 1H), 8.33 (d, 1H), 8.38 - 8.43 (m, 1H), 8.45 - 8.48 (m, 1H), 8.69 (s, 1H), 10.50 (s, 1H).Instance 53 N-[6- Methoxy -2-(4,4,4- Trifluorobutyl )-2H- Carbazole -5- base ]-6-( Trifluoromethyl ) Pyridine -2- Formamide Dissolve 120 mg (357 μmol) of N-(6-methoxy-1H-indazol-5-yl)-6-(trifluoromethyl)pyridine-2-carboxamide in 2 ml of DMF and 68 μl (540 μmol) of 3-(bromomethyl)oxetane and 148 mg (1.07 mmol) of potassium carbonate were added with stirring. The reaction mixture was stirred at 50 ° C for 4.5 hours and at 80 ° C for 16 hours. Then, the mixture was diluted with water and extracted twice with ethyl acetate. The combined organic phases were filtered through a hydrophobic filter and concentrated. The residue was purified by preparative HPLC. The combined product fractions were lyophilized. This gave 33 mg of the title compound. LC-MS (Method A3): Rt = 1.36 min (UV detector: TIC Smooth), mass measurement 446.001 </ RTI> <RTIgt; 8.34 (s, 1H), 8.38 - 8.44 (m, 1H), 8.45 - 8.49 (m, 1H), 8.70 (s, 1H), 10.51 (s, 1H).Instance 54 N-[6- chlorine -2-(3- Hydroxyl -3- Methyl butyl )-2H- Carbazole -5- base ]-6-( Trifluoromethyl ) Pyridine -2- Formamide Dissolve 1.10 g (3.23 mmol) of N-(6-chloro-1H-indazol-5-yl)-6-(trifluoromethyl)pyridine-2-carboxamide in 10 ml of DMF with stirring 809 mg (4.84 mmol) of 4-bromo-2-methylbutan-2-ol, 1.34 g (9.69 mmol) of potassium carbonate and 804 mg (4.84 mmol) of potassium iodide were added. The suspension was stirred at 120 ° C for 23 hours. Another 670 mg (4.85 mmol) potassium carbonate and 402 mg (2.42 mmol) potassium iodide were added, and the mixture was stirred at 120 ° C for additional 24 hours. The reaction mixture was diluted with water and extracted three times with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was purified by flash chromatography (mobile phase: hexane/ethyl acetate, EtOAc, EtOAc (EtOAc) The combined product fractions were concentrated, diethyl ether was added and the mixture was stirred 15 min. The solid was filtered off with suction, washed three times with diethyl ether and dried overnight in a dry cabinet. This gave 342 mg of the title compound. LC-MS (Method A1): Rt = 1.17 min (UV detector: TIC Smooth), mass measurement 408.001 </ RTI> <RTIgt; ), 8.03 (dd, 1H), 8.28 - 8.39 (m, 2H), 8.52 (s, 1H), 8.63 (s, 1H), 10.59 (s, 1H).Instance 55 N-[6- Methoxy -2-(2- Methoxyethyl )-2H- Carbazole -5- base ]-6-( Trifluoromethyl ) Pyridine -2- Formamide Dissolve 200 mg (0.60 mmol) of N-(6-methoxy-1H-indazol-5-yl)-6-(trifluoromethyl)pyridine-2-carboxamide in 4 ml of DMF and 84 μl (0.89 mmol) of 2-bromoethyl methyl ether, 247 mg (1.78 mmol) of potassium carbonate and 118 mg (0.71 mmol) of potassium iodide were added with stirring. The reaction mixture was stirred at 100 &lt;0&gt;C for 19 h, diluted with water andEtOAc. The combined organic phases were filtered through a hydrophobic filter and concentrated. The residue was purified by preparative HPLC. The fractions containing the product are lyophilized. This gave 52 mg of the title compound. LC-MS (Method A1): Rt = 1.28 min (UV detector: TIC Smooth), mass measurement 394.001 H-NMR (500MHz, DMSO-d6): δ = 3.24 (s, 3H), 3.81 (t, 2H), 3.99 (s, 3H), 4.51 (t, 2H), 7.15 (s, 1H), 8.21 ( Dd, 1H), 8.28 (s, 1H), 8.38 - 8.43 (m, 1H), 8.45 - 8.48 (m, 1H), 8.69 (s, 1H), 10.50 (s, 1H).Instance 56 N-{2-[2-(2- Hydroxyethoxy ) Ethyl ]-6- Methoxy -2H- Carbazole -5- base }-6-( Trifluoromethyl ) Pyridine -2- Formamide Dissolve 150 mg (446 μmol) of N-(6-methoxy-1H-indazol-5-yl)-6-(trifluoromethyl)pyridine-2-carboxamide in 2 ml of DMF and 71 μl (670 μmol) of 2-(2-chloroethoxy)ethanol, 185 mg (1.34 mmol) of potassium carbonate and 111 mg (669 μmol) of potassium iodide were added with stirring. The suspension was stirred at 100 ° C for 26 hours. The reaction mixture was diluted with water and extracted three times with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, filtered and concentrated with a pad. The residue was purified by flash chromatography (EtOAc EtOAc (EtOAc) This gave 38.9 mg of the title compound. LC-MS (Method A1): Rt = 1.08 min (UV detector: TIC Smooth), mass measurement 424.001 H-NMR (400MHz, DMSO-d6): δ = 3.40 - 3.50 (m, 4H), 3.90 (t, 2H), 3.99 (s, 3H), 4.51 (t, 2H), 4.62 (t, 1H), 7.16 (s, 1H), 8.22 (d, 1H), 8.32 (s, 1H), 8.38 - 8.45 (m, 1H), 8.45 - 8.50 (m, 1H), 8.70 (s, 1H), 10.51 (s, 1H).Instance 57 {2-[6- Methoxy -5-({[6-( Trifluoromethyl ) Pyridine -2- base ] Carbonyl } Amine )-2H- Carbazole -2- base ] Ethoxy } Ethyl acetate Dissolve 300 mg (0.89 mmol) of N-(6-methoxy-1H-indazol-5-yl)-6-methylpyridine-2-carboxamide in 5 ml of DMF and add 377 with stirring. Methyl (1.78 mmol) (2-bromoethoxy)ethyl acetate and 493 mg (3.57 mmol) of potassium carbonate. The reaction mixture was stirred at 25 ° C for 2 hours and at 65 ° C for 21 hours. The reaction mixture was diluted with water and extracted three times with ethyl acetate. The combined organic phases were washed with a saturated sodium chloride solution, the phases separated, filtered and concentrated with a pad. The residue was purified by preparative HPLC. The fractions containing the product are lyophilized. This gave 96 mg of the title compound. LC-MS (Method A1): Rt = 1.26 min (UV detector: TIC Smooth), mass measurement 466.001 </ RTI> <RTIgt; 7.40 (s, 1H), 8.02 (d, 1H), 8.21 (dd, 1H), 8.38 - 8.43 (m, 1H), 8.45 - 8.49 (m, 1H), 8.72 (s, 1H), 10.43 (s, 1H).Instance 58 {[2-(3- Hydroxyl -3- Methyl butyl )-5-({[6-( Trifluoromethyl ) Pyridine -2- base ] Carbonyl } Amine )-2H- Carbazole -6- base ] Oxyl } Acetic acid 50 mg (0.10 mmol) of {[2-(3-hydroxy-3-methylbutyl)-5-({[6-(trifluoromethyl)pyridin-2-yl)carbonyl}amino)-2H Ethyl acetate of carbazole-6-yl]oxy} was dissolved in 1.5 ml of THF, and 12 mg (0.51 mmol) of lithium hydroxide monohydrate and 150 μl of water were added. The reaction mixture was stirred at 25 ° C for 60 minutes. The mixture was acidified to pH = 3 with 1 M hydrochloric acid and the obtained residue was filtered, and washed with water and diethyl ether. This gave 35 mg of the title compound. LC-MS (Method A1): Rt = 1.00 min (UV detector: TIC Smooth), mass measurement 466.00 1H NMR (300 MHz, DMSO-d6): δ = 1.15 (s, 6 H), 1.96 - 2.06 (m, 2 H), 4.38 - 4.47 (m, 2 H), 4.51 (s, 1 H), 4.89 (s, 2 H), 7.13 (s, 1 H), 8.14 - 8.24 (m, 1 H), 8.34 (s, 1 H), 8.36 - 8.45 (m, 1 H), 8.45 - 8.50 (m, 1 H), 8.73 (s, 1 H), 10.57 (s, 1 H).Instance 59 {2-[6- Methoxy -5-({[6-( Trifluoromethyl ) Pyridine -2- base ] Carbonyl } Amine )-2H- Carbazole -2- base ] Ethoxy } Acetic acid 82 mg (0.18 mmol) of {2-[6-methoxy-5-({[6-(trifluoromethyl)pyridin-2-yl]carbonyl}amino)-2H-indazol-2-yl Ethyloxy}acetate was dissolved in 2 ml of THF and a solution of 300 μl of ethanol, 73 mg (1.76 mmol) of lithium hydroxide monohydrate in 300 μl of water was added, and the mixture was stirred at 50 ° C for 17 hours. The mixture was diluted with water and acidified to pH 3 using a 10% strength citric acid solution. The aqueous phase was extracted three times with ethyl acetate and the combined organics were filtered th Diethyl ether was added to the residue, and the mixture was stirred for 10 min, filtered, washed with diethyl ether and dried under reduced pressure. The residue was purified by preparative HPLC. The fractions containing the product are lyophilized. This gave 49 mg of the title compound. LC-MS (Method A1): Rt = 1.09 min (UV detector: TIC Smooth), mass measurement 438.001 H NMR (300 MHz, DMSO-d6): δ = 3.94 - 4.02 (m, 7 H), 4.53 (t, 2 H), 7.15 (s, 1 H), 8.21 (dd, 1 H), 8.33 (s , 1 H), 8.36 - 8.44 (m, 1 H), 8.44 - 8.49 (m, 1 H), 8.69 (s, 1 H), 10.50 (s, 1 H).Instance 60 N-[6- Ethoxy -2-(3- Hydroxyl -3- Methyl butyl )-2H- Carbazole -5- base ]-6-( Pentafluoroethyl ) Pyridine -2- Formamide 100 mg (0.38 mmol) 4-(5-Amino-6-ethoxy-2H-indazol-2-yl)-2-methylbutan-2-ol, 137 mg (0.57 mmol) 6-( A mixture of pentafluoroethyl)pyridine-2-carboxylic acid (CAS 1283717-85-0), 80 μl (0.57 mmol) of triethylamine and 217 mg (0.57 mmol) of HATU in 2 ml of DMF was stirred overnight at RT. The mixture was purified by preparative HPLC to give 150 mg of the title compound. LC-MS (Method A2): Rt = 1.32 min (UV detector: TIC Smooth), mass measurement 486.171 H-NMR (400MHz, DMSO-d6 ): δ = 1.14 (s, 6H), 1.47 (t, 3H), 1.97 – 2.05 (m, 2H), 4.15 – 4.24 (q, 2H), 4.37 - 4.48 (m, 2H), 4.52 (s, 1H ), 7.11 (s, 1H), 8.23 - 8.27 (m, 1H), 8.31 (s, 1H), 8.39 – 8.51 (m, 2H), 8.73 (s, 1H), 10.60 (s, 1H).Instance 61 6-(1,1- Difluoroethyl )-N-[6- Ethoxy -2-(3- Hydroxyl -3- Methyl butyl )-2H- Carbazole -5- base ] Pyridine -2- Formamide 100 mg (0.38 mmol) 4-(5-Amino-6-ethoxy-2H-indazol-2-yl)-2-methylbutan-2-ol, 106 mg (0.57 mmol) 6-( A mixture of 1,1-difluoroethyl)pyridine-2-carboxylic acid, 80 μl (0.57 mmol) of triethylamine and 217 mg (0.57 mmol) of HATU in 2 ml of DMF was stirred overnight at RT. The mixture was purified by preparative HPLC to give the title compound. LC-MS (Method A2): Rt = 1.23 min (UV detector: TIC Smooth), mass measurement 432.20.1 H-NMR (400MHz, DMSO-d6 ): δ = 1.15 (s, 6 H), 1.51 (t, 3H), 1.96 – 2.05 (m, 2H), 2.15 (t, 3H), 4.15 – 4.26 (m, 2 H), 4.37 - 4.45 (m , 2H), 4.52 (s, 1H), 7.11 (s, 1H), 7.97 - 8.04 (m, 1H), 8.24 – 8.37 (m, 3H), 8.72 (s, 1H), 10.70 (s, 1H).Instance 62 N-[6- Ethoxy -2-(3- Hydroxyl -3- Methyl butyl )-2H- Carbazole -5- base ]-6,7- Dihydrogen -4H- Pyrazole [5,1-c][1,4] Oxazine -2- Formamide 100 mg 4-(5-Amino-6-ethoxy-2H-indazol-2-yl)-2-methylbutan-2-ol, 67 mg 6,7-dihydro-4H-pyrazole And a mixture of [5,1-c][1,4]oxazine-2-carboxylic acid, 0.19 ml N-ethyl-N-isopropylpropan-2-amine and 165 mg HATU in 2 ml THF at RT Stir for 18 hours. The reaction mixture was diluted with water and extracted with EtOAc EtOAc. The mixture was purified by preparative HPLC to give 124 mg of the title compound. LC-MS (Method A1): Rt = 0.93 min (UV detector: TIC Smooth), mass measurement 413.61.1 H-NMR (400MHz, DMSO-d6 ): δ = 1.14 (s, 6H), 1.46 (t, 3H), 1.96 - 2.04 (m, 2H), 4.07 - 4.14 (m, 2H), 4.16 - 4.28 (m, 4H), 4.36 - 4.44 (m , 2H), 4.51 (br s, 1H), 4.84 (s, 2H), 6.59 (s, 1H), 7.08 (s, 1H), 8.25 (s, 1H), 8.55 (s, 1H), 9.46 (s , 1H).Instance 63 N-[6- Ethoxy -2-(3- Hydroxyl -3- Methyl butyl )-2H- Carbazole -5- base ]-1- Ethyl -1H- Pyrazole -3- Formamide 100 mg 4-(5-Amino-6-ethoxy-2H-indazol-2-yl)-2-methylbutan-2-ol, 56 mg 1-ethyl-1H-pyrazole-3 A mixture of formic acid, 0.19 ml of N-ethyl-N-isopropylpropan-2-amine and 165 mg of HATU in 2 ml of THF was stirred at RT for 18 h. The reaction mixture was diluted with water and extracted with EtOAc EtOAc. The mixture was purified by preparative HPLC to give 114 mg of the title compound. LC-MS (Method A1): Rt = 1.02 min (UV detector: TIC Smooth), mass measurement 385.21.1 H-NMR (400MHz, DMSO-d6 ): δ = 1.14 (s, 6H), 1.42 - 1.51 (m, 6H), 1.97 - 2.03 (m, 2H), 4.15 - 4.28 (m, 4H), 4.36 - 4.43 (m, 2H), 4.51 (s , 1H), 6.75 (d, 1H), 7.07 (s, 1H), 7.93 (d, 1H), 8.25 (s, 1H), 8.54 (s, 1H), 9.57 (s, 1H).Instance 64 N-[6- Ethoxy -2-(3- Hydroxyl -3- Methyl butyl )-2H- Carbazole -5- base ]-1- Isopropyl -1H- Pyrazole -3- Formamide 100 mg 4-(5-Amino-6-ethoxy-2H-indazol-2-yl)-2-methylbutan-2-ol, 61 mg 1-isopropyl-1H-pyrazole- A mixture of 3-carboxylic acid, 0.19 ml of N-ethyl-N-isopropylpropan-2-amine and 165 mg of HATU in 2 ml of THF was stirred at RT for 18 h. The reaction mixture was diluted with water and extracted with EtOAc EtOAc. The mixture was purified by preparative HPLC to give 127 mg of the title compound. LC-MS (Method A1): Rt = 1.10 min (UV detector: TIC Smooth) with a mass measurement of 399.23.1 H-NMR (400MHz, DMSO-d6 ): δ [ppm] = 1.14 (s, 6H), 1.44 - 1.53 (m, 9H), 1.97 - 2.03 (m, 2H), 4.18 (q, 2H), 4.36 - 4.44 (m, 2H), 4.51 ( s, 1H), 4.62 (spt, 1H), 6.74 (d, 1H), 7.07 (s, 1H), 7.96 (d, 1H), 8.25 (s, 1H), 8.54 (s, 1H), 9.65 (s , 1H).Instance 65 N-[6- Ethoxy -2-(3- Hydroxyl -3- Methyl butyl )-2H- Carbazole -5- base ] Pyrazole [1,5-a] Pyrimidine -3- Formamide 100 mg 4-(5-Amino-6-ethoxy-2H-indazol-2-yl)-2-methylbutan-2-ol, 65 mg 1-pyrazolo[1,5-a A mixture of pyrimidine-3-carboxylic acid, 0.19 ml of N-ethyl-N-isopropylpropan-2-amine and 165 mg of HATU in 2 ml of THF was stirred at RT for 18 h. The reaction mixture was diluted with water and extracted with EtOAc EtOAc. The mixture was purified by preparative HPLC to give 57 mg of the title compound. LC-MS (Method A1): Rt = 0.89 min (UV detector: TIC Smooth), mass measurement 408.191 </ RTI> <RTIgt; ), 4.51 (br s, 1H), 7.05 (s, 1H), 7.34 (dd, 1H), 8.25 (s, 1H), 8.71 (s, 2H), 8.86 (dd, 1H), 9.38 (dd, 1H) ), 10.64 (s, 1H).Instance 66 N-[6- Methoxy -2-( Tetrahydrofuran -3- Methyl )-2H- Carbazole -5- base ]-6-( Trifluoromethyl ) Pyridine -2- Formamide 400 mg of N-(6-methoxy-1H-indazol-5-yl)-6-(trifluoromethyl)pyridine-2-carboxamide (CAS number: 1799836-45-5) as crude product A mixture of 310 mg of 3-(bromomethyl)tetrahydrofuran and 519 mg of potassium carbonate in 5.0 ml of DMF was stirred at 100 ° C for 5 hours and at 80 ° C for 21 hours. Water was then added and the mixture was extracted three times with ethyl acetate. The combined organic phases were washed with saturated brine, filtered and concentrated with a sep. Purification by preparative HPLC gave 120 mg of the title compound.1 H-NMR (400MHz, DMSO-d6 ): δ = 1.59 - 1.72 (m, 1H), 1.88 - 2.00 (m, 1H), 2.79 - 2.92 (m, 1H), 3.49 - 3.57 (m, 1H), 3.60 - 3.73 (m, 2H), 3.79 (td, 1H), 3.99 (s, 3H), 4.35 (d, 2H), 7.17 (s, 1H), 8.22 (d, 1H), 8.34 - 8.49 (m, 3H), 8.69 (s, 1H), 10.51 (s, 1H).Instance 67 N-[2-(3- Hydroxyl -3- Methyl butyl )-6- Methoxy -2H- Carbazole -5- base ]-6-( Pyrrolidine -1- base ) Pyridine -2- Formamide 277 mg of potassium carbonate and 125 mg of potassium iodide were added to 169 mg of N-(6-methoxy-1H-indazol-5-yl)-6-(pyrrolidin-1-yl)pyridine-2-carboxamide and 126 mg of 4-bromo-2-methylbutan-2-ol in a mixture of 2.7 ml of DMF, and the mixture was heated at 100 ° C for 22 hours. 42 mg of 4-bromo-2-methylbutan-2-ol and 327 mg of cesium carbonate were added and the mixture was stirred at 100 ° C for 6 hours. The mixture was filtered off with suction, the residue was purified eluting eluting This gave 49 mg of the title compound.1 </ RTI> <RTIgt; - 4.45 (m, 2H), 4.52 (s, 1H), 6.73 (d, 1H), 7.09 (s, 1H), 7.33 (d, 1H), 7.67 - 7.75 (m, 1H), 8.26 (s, 1H ), 8.65 (s, 1H), 10.92 (s, 1H).Evaluation of physiological efficacy IRAK4 Kinase analysis The IRAK4-inhibitory activity of the substance of the invention was measured in an Irak4 TR-FRET assay (TR-FRET = time-resolved fluorescence resonance energy transfer) as set forth below. Use from baculovirus-infected insect cells (Hi5, BTI-TN-5B1-4, cell line purchased from Invitrogen, Cat. No. B855-02) and purified from affinity chromatography by N-terminal GST (Beth A recombinant fusion protein of the peptide S-transferase) and human Irak4 is used as an enzyme. The substrate used for the kinase reaction is biotinylated peptide biotin-Ahx-KKARFSRFAGSSPSQASFAEPG (C-terminal in the form of a guanamine), which is commercially available, for example, from Biosyntan GmbH (Berlin-Buch). For this analysis, 11 different concentrations ranging from 20 μM to 0.073 nM were prepared from the test substance in 2 mM solution in DMSO. 50 nl of each solution was pipetted into a black low volume 384-well microtiter plate (Greiner Bio-One, Frickenhausen, Germany) and 2 μl of Irak4 was added to the assay buffer [50 mM HEPES pH 7.5, 5 mM MgCl2, 1.0 mM Dithiothreitol, 30 μM activated sodium orthovanadate, 0.1% (w/v) bovine gamma-immunoglobulin (BGG) 0.04% (v/v) nonidet-P40 (Sigma)] solution, and The mixture was incubated for 15 minutes to allow the material to pre-adhere to the enzyme prior to the kinase reaction. Then by adding 3 μl adenosine triphosphate (ATP, 1.67 mM = final concentration in 5 μl assay volume: 1 mM) and peptide substrate (0.83 μM = final concentration in 5 μl assay volume: 0.5 μM) The kinase reaction was started by analyzing the solution in the buffer, and the resulting mixture was incubated at 22 ° C for a reaction time of 45 minutes. The concentration of Irak4 was adjusted according to the individual activities of the enzyme and was set such that the analysis was carried out in a linear range. Typical concentrations are on the order of about 0.2 nM. By adding 5 μl of TR-FRET detection reagent [0.1 μM streptavidin-XL665 (Cisbio Bioassays; France, Cat. No. 610SAXLG)] and 1.5 nM anti-phosphoric acid antibody [Merck Millipore, "STK Antibody", catalog number 35-002] and 0.6 nM LANCE EU-W1024-labeled anti-mouse IgG antibody (Perkin-Elmer, product number AD0077; alternatively, anti-chick compound resistance from Cisbio Bioassays can be used The mouse-IgG antibody was stopped in a solution of EDTA aqueous solution (100 mM EDTA in 0.4% [w/v] bovine serum albumin [BSA] in 25 mM HEPES (pH 7.5)). The resulting mixture was incubated at 22 ° C for 1 hour to allow formation of a complex of biotinylated phosphorylated substrate and detection reagent. The amount of phosphorylated host was then assessed by measuring the resonance energy transfer of the ruthenium chelate labeled anti-mouse-IgG antibody to streptavidin-XL665. To this end, fluorescence emission at 620 nm and 665 nm was measured after excitation at 350 nm in a TR-FRET measuring instrument (eg, Rubystar (BMG Labtechnologies, Offenburg, Germany) or Viewlux (Perkin-Elmer)). The ratio of emission at 665 nm and 622 nm is taken as a measure of the amount of phosphorylation. The data was normalized (enzyme reaction without test substance = 0% inhibition; all other analytical components but no enzyme = 100% inhibition). Typically, 11 different concentrations (20 μM, 5.7 μM, 1.6 μM, 0.47 μM, 0.13 μM, 38 nM, 11 nM, 3.1 nM, 0.89 nM, 0.25) in the range of 20 μM to 0.073 nM on the same microtiter plate. The test substance was tested under nM and 0.073 nM). A dilution series (2 mM to 7.3 nM in 100% DMSO) was prepared by serial dilution prior to analysis. Calculate IC by 4 parameter fitting50 value.table 1 : IC of an example compound in IRAK4 kinase assay50 value THP-1 In cells TNF-α secretion By means of this test, the ability of a substance to inhibit the secretion of TNF-α (tumor necrosis factor alpha) in THP-1 cells (human mononuclear acute leukemia cell line) can be tested. TNF-α is involved in the interleukins of the inflammatory process. In this test, TNF-α secretion was triggered by utilization with bacterial lipopolysaccharide (LPS). THP-1 cells were maintained in continuous suspension cell culture [no L-Glutamax (GE Healthcare, Cat. No. E15-039) and supplemented with fetal bovine serum (FCS) 10% (Invitrogen, Cat. No. 10082-147), 1% L - RPMI of branide (Sigma, Cat. No. G7513), 1% penicillin/streptomycin (PAA, Cat. No. P11-010) and 50 μM 2-mercaptoethanol (Gibco, Cat. No. 31350-010) 1460 medium] and should not exceed 1 × 106 Cell concentration of cells/ml. The analysis was carried out in a cell culture medium (RPMI 1460 medium supplemented with L-glutamic acid, penicillin, streptomycin and 2-mercaptoethanol). THP-1 cells at 2.5×105 The cell density of the cells/well was seeded in a 96-well plate. The compounds of the invention were subjected to serial dilutions in a constant volume of 100% DMSO and used in this assay at 8 different concentrations ranging from 10 μM to 3 nM such that the final DMSO concentration was 0.4% DMSO. Cells were pre-incubated with these for 30 minutes prior to actual stimulation. To induce interleukin secretion, cells were stimulated with 1 μg/ml LPS (Sigma, E. coli 0127: B8, Cat. No. L4516) for 6 hours. As a neutral control, cells were treated with 1 μg/ml LPS and 0.04% DMSO, and as inhibitor controls, cells were only treated with 0.04% DMSO. Cell viability was determined using CellTiter-Glo Luminescence Analysis (Promega, Cat. No. G7571 (G755/G756A)) according to the manufacturer's instructions. The amount of TNF-α secreted in the cell culture supernatant was determined using a human pro-inflammatory 9-Plex tissue culture kit (MSD, Cat. No. K15007B) according to the manufacturer's instructions. The activity of the substance is expressed as the ratio between the neutral and inhibitor controls (expressed as a percentage). Calculate the IC using a 4-parameter fit50 value.table 2 : Example Compounds for the secretion of TNF-α in THP-1 cells and having no effect on cell viability50 value Working examples of pharmaceutical compositions The compounds of the invention can be converted into the following pharmaceutical preparations:Lozenge : composition : 100 mg of the compound of Example 11 or the compound of Example 12, 50 mg of lactose (monohydrate), 50 mg of maize starch (natural), 10 mg of polyvinylpyrrolidone (PVP 25) (from BASF, Ludwigshafen, Germany) and 2 mg magnesium stearate. The tablet weight is 212 mg. It has a diameter of 8 mm and a radius of curvature of 12 mm.produce: A mixture of the compound of the present invention, lactose and starch was granulated using a 5% aqueous solution of PVP (w/w). The granules were dried and then mixed with magnesium stearate for 5 minutes. This mixture was compressed using a conventional tablet press (see above for the format of the tablet). The pressing force used for pressing is 15 kN.Suspension for oral administration composition : 1000 mg of the compound of Example 11 or the compound of Example 12, 1000 mg of ethanol (96%), 400 mg of Rhodigel® (xanthan gum, from FMC, Pennsylvania, USA) and 99 g of water. A 10 ml oral suspension corresponds to a single dose of 100 mg of a compound of the invention.produce: Rhodigel is suspended in ethanol; the compound of the invention is added to the suspension. Add water while stirring. The mixture was stirred for about 6 hours until the Rhodigel had finished expanding.For oral administration of solutions: composition 500 mg of the compound of Example 11 or the compound of Example 12, 2.5 g of polysorbate and 97 g of polyethylene glycol 400. The 20 g oral solution corresponds to a single dose of 100 mg of the compound of the invention.produce The compound of the invention is suspended in a mixture of polyethylene glycol and polysorbate with stirring. This stirring operation is continued until the compound of the present invention is completely dissolved.

Claims (17)

一種具有通式(I)之化合物,其中 R1 代表氯或O-R4 , R2 代表C1 -C8 -烷基, 其可視情況彼此獨立地經選自由以下組成之群之相同或不同取代基單取代或多取代:1至5個氟原子、1至3個羥基、氧雜環丁基、四氫呋喃基、吡喃基、側氧基及C1 -C6 -烷氧基, 其中該C1 -C6 -烷氧基 可經氟或 經羥基或 經C(=O)OH、C(=O)Me、C(=O)Et、C(=O)NH2 單取代至三取代; R3 代表選自以下之基團: , 其中R5 代表氫、C3 -C6 -環烷基或C1 -C6 -烷基,其中C1 -C6 -烷基可視情況經環丙基、氰基及羥基單取代至三取代,其中每一取代基可僅出現一次,或經氟原子單取代至五取代; R6 代表氫、氟或甲基, 或 R3 代表選自以下之基團:其中R7 代表氫、C3 -C6 -環烷基、氰基、NH2 、NH(C1 -C6 -烷基)、N(C1 -C6 -烷基)2 、吡咯啶-1-基、六氫吡啶-1-基、嗎啉-4-基、4-甲基六氫吡嗪-1-基 或代表C1 -C6 -烷基,其中C1 -C6 -烷基可視情況經環丙基、氰基及羥基單取代至三取代,其中每一取代基可僅出現一次,或經氟原子單取代至五取代; R8 、R9 、R10 代表氫、甲基或氟, 或 R3 代表其中R11 代表 C3 -C6 -環烷基或C1 -C6 -烷基,其中C1 -C6 -烷基可視情況經環丙基、氰基及羥基單取代至三取代,其中每一取代基可僅出現一次,或經氟原子單取代至五取代; 且R12 代表氫、氟或C1 -C6 -烷基; 或 R3 代表 6,7-二氫-4H-吡唑并[5,1-c][1,4]噁嗪-2-基、5-甲基-4,5,6,7-四氫吡唑并[1,5-a]吡嗪-2-基或4,5,6,7-四氫吡唑并[1,5-a]吡嗪-2-基 或代表 吡唑并[1,5-a]嘧啶-3-基、吡咯并[2,1-f][1,2,4]三嗪-7-基、吡咯并[1,2-b]嗒嗪-7-基、噻吩并[2,3-b]吡嗪-7-基、5-胺基吡唑并[1,5-a]嘧啶-3-基、2-胺基吡咯并[2,1-f][1,2,4]三嗪-7-基、2-胺基吡咯并[1,2-b]嗒嗪-7-基、2-胺基噻吩并[2,3-b]吡嗪-7-基; R4 代表飽和4員至7員雜環基或C3 -C7 -環烷基,其中該飽和雜環基及該C3 -C7 -環烷基可視情況經來自由以下組成之群之相同或不同取代基單取代或多取代:C1 -C6 -烷基、三氟甲基、2,2,2-三氟乙基、環丙基、C(=O)OH、C1 -C6 -烷氧基、三氟甲氧基、2,2,2-三氟乙氧基、氟、氯、氰基、羥基、NH2 、NHRa 、N(Ra )Rb , 或為 C1 -C6 -烷基,其中C1 -C6 -烷基可視情況經來自由以下組成之群之相同或不同取代基單取代或多取代:氟、氯、氰基、羥基、C(=O)OC1 -C6 -烷基、C(=O)OH、C(=O)NH2 、C(=O)N(H)Ra 、C(=O)N(Ra )Rb 、SC1 -C6 -烷基、S(=O)2 -C1 -C6 -烷基、S(=O)2 NH2 、C1 -C6 -烷氧基、三氟甲氧基、2,2,2-三氟乙氧基、NH2 、NHRa 、N(Ra )Rb 、C3 -C7 -環烷基、四唑及4員至7員飽和雜環基, 其中C3 -C7 -環烷基、雜芳基及4員至7員飽和雜環基可視情況經來自由以下組成之群之相同或不同取代基單取代至四取代:氟、C1 -C4 -烷基、羥基、C(=O)OH、三氟甲基、2,2,2-三氟乙基、甲氧基、乙氧基、三氟甲氧基、環丙基、環丙基甲基、甲基磺醯基、NH2 、NHRa 、N(Ra )Rb ; Ra 代表C1 -C6 -烷基或C3 -C7 -環烷基, 其中C1 -C6 -烷基及C3 -C7 -環烷基可視情況經來自由以下組成之群之相同或不同取代基單取代或多取代:氟、羥基、氰基、C1 -C4 -烷基、C1 -C4 -烷氧基及C3 -C7 -環烷基; Rb 代表C1 -C6 -烷基或C3 -C7 -環烷基; 及其非鏡像異構物、鏡像異構物、代謝物、鹽、溶劑合物或其鹽之溶劑合物。a compound of the formula (I), Wherein R 1 represents chloro or OR 4 , and R 2 represents C 1 -C 8 -alkyl, which may optionally be mono- or polysubstituted, respectively, via the same or different substituents selected from the group consisting of: 1 to 5 a fluorine atom, 1 to 3 hydroxyl groups, oxetanyl group, tetrahydrofuranyl group, pyranyl group, pendant oxy group, and C 1 -C 6 -alkoxy group, wherein the C 1 -C 6 -alkoxy group may be fluorine-containing Or mono-substituted to trisubstituted by hydroxyl group or via C(=O)OH, C(=O)Me, C(=O)Et, C(=O)NH 2 ; R 3 represents a group selected from the group consisting of: Wherein R 5 represents hydrogen, C 3 -C 6 -cycloalkyl or C 1 -C 6 -alkyl, wherein the C 1 -C 6 -alkyl group may optionally be monosubstituted by cyclopropyl, cyano and hydroxy to three Substituting wherein each substituent may occur only once, or may be mono-substituted to penta-substituted with a fluorine atom; R 6 represents hydrogen, fluorine or methyl, or R 3 represents a group selected from the group consisting of: Wherein R 7 represents hydrogen, C 3 -C 6 -cycloalkyl, cyano, NH 2 , NH(C 1 -C 6 -alkyl), N(C 1 -C 6 -alkyl) 2 , pyrrolidine- 1-yl, hexahydropyridin-1-yl, morpholin-4-yl, 4-methylhexahydropyrazin-1-yl or a C 1 -C 6 -alkyl group, wherein C 1 -C 6 -alkane The group may be mono-substituted to trisubstituted by cyclopropyl, cyano and hydroxy, wherein each substituent may occur only once, or may be mono-substituted to penta-substituted through a fluorine atom; R 8 , R 9 , R 10 represent hydrogen, A Base or fluorine, or R 3 represents Wherein R 11 represents a C 3 -C 6 -cycloalkyl group or a C 1 -C 6 -alkyl group, wherein the C 1 -C 6 -alkyl group may be monosubstituted to trisubstituted by a cyclopropyl group, a cyano group and a hydroxy group, wherein Each substituent may occur only once, or may be mono-substituted to penta-substituted with a fluorine atom; and R 12 represents hydrogen, fluorine or C 1 -C 6 -alkyl; or R 3 represents 6,7-dihydro-4H-pyridyl Zoxa[5,1-c][1,4]oxazin-2-yl, 5-methyl-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrazine-2 -yl or 4,5,6,7-tetrahydropyrazolo[1,5-a]pyrazin-2-yl or represents pyrazolo[1,5-a]pyrimidin-3-yl, pyrrole [ 2,1-f][1,2,4]triazin-7-yl, pyrrolo[1,2-b]pyridazin-7-yl, thieno[2,3-b]pyrazine-7- 5-Aminopyrazolo[1,5-a]pyrimidin-3-yl, 2-aminopyrrolo[2,1-f][1,2,4]triazin-7-yl, 2 -Aminopyrrolo[1,2-b]pyridazin-7-yl, 2-aminothieno[2,3-b]pyrazin-7-yl; R 4 represents a saturated 4- to 7-membered heterocyclic ring Or a C 3 -C 7 -cycloalkyl group, wherein the saturated heterocyclic group and the C 3 -C 7 -cycloalkyl group are optionally mono- or polysubstituted by the same or different substituents from the group consisting of: C 1 -C 6 -alkyl, trifluoromethyl, 2,2,2-trifluoroethyl, cyclopropyl , C(=O)OH, C 1 -C 6 -alkoxy, trifluoromethoxy, 2,2,2-trifluoroethoxy, fluoro, chloro, cyano, hydroxy, NH 2 , NHR a And N(R a )R b , or C 1 -C 6 -alkyl, wherein the C 1 -C 6 -alkyl group may be mono- or polysubstituted by the same or different substituents from the group consisting of: Fluorine, chlorine, cyano, hydroxy, C(=O)OC 1 -C 6 -alkyl, C(=O)OH, C(=O)NH 2 , C(=O)N(H)R a , C(=O)N(R a )R b , SC 1 -C 6 -alkyl, S(=O) 2 -C 1 -C 6 -alkyl, S(=O) 2 NH 2 , C 1 - C 6 -alkoxy, trifluoromethoxy, 2,2,2-trifluoroethoxy, NH 2 , NHR a , N(R a )R b , C 3 -C 7 -cycloalkyl, tetra An azole and a 4- to 7-membered saturated heterocyclic group, wherein the C 3 -C 7 -cycloalkyl group, the heteroaryl group and the 4 to 7 membered saturated heterocyclic group may be optionally substituted by the same or different groups from the group consisting of Substituted to tetrasubstituted: fluorine, C 1 -C 4 -alkyl, hydroxy, C(=O)OH, trifluoromethyl, 2,2,2-trifluoroethyl, methoxy, ethoxy , trifluoromethoxy, cyclopropyl, cyclopropylmethyl, methyl sulfonic acyl, NH 2, NHR a, N (R a) R b; R a representative of C 1 -C 6 - alkyl C 3 -C 7 - cycloalkyl, wherein C 1 -C 6 - alkyl and C 3 -C 7 - cycloalkyl group optionally substituted by the same or a different group of consisting of mono- or poly-substituted by the group consisting of the following: Fluorine, hydroxy, cyano, C 1 -C 4 -alkyl, C 1 -C 4 -alkoxy and C 3 -C 7 -cycloalkyl; R b represents C 1 -C 6 -alkyl or C 3 -C 7 -cycloalkyl; and solvates thereof and their non-image isomers, mirror image isomers, metabolites, salts, solvates or salts thereof. 如請求項1之式(I)化合物,其中 R1 代表氯或O-R4 , R2 代表C1 -C6 -烷基, 其可視情況彼此獨立地經選自由以下組成之群之相同或不同取代基單取代或多取代:1至3個氟原子、1至2個羥基、氧雜環丁基、四氫呋喃基及C1 -C3 -烷氧基, 其中該C1 -C3 -烷氧基 可經羥基或 經C(=O)OH、C(=O)Me、C(=O)Et取代; 或R2 代表3-側氧基丁基; R3 代表選自以下之基團: , 其中R5 代表氫、環丙基或C1 -C6 -烷基,其中C1 -C6 -烷基可視情況經環丙基、氰基及羥基單取代至三取代,其中每一取代基可僅出現一次,或經氟原子單取代至五取代; 且R6 代表氫; 或 R3 代表基團其中R7 代表氫、環丙基、氰基、NH2 、NH(C1-C6 -烷基)、N(C1 -C6 -烷基)2 、吡咯啶-1-基、六氫吡啶-1-基、嗎啉-4-基、4-甲基六氫吡嗪-1-基 或代表C1 -C6 -烷基,其中C1 -C6 -烷基可視情況經環丙基、氰基及羥基單取代至三取代,其中每一取代基可僅出現一次,或經氟原子單取代至五取代; 且R8 、R9 、R10 代表氫 或 R3 代表基團其中R11 代表 C3 -C6 -環烷基或C1 -C6 -烷基,其中C1 -C6 -烷基可視情況經環丙基、氰基及羥基單取代至三取代,其中每一取代基可僅出現一次,或經氟原子單取代至五取代; 且R12 代表氫 或 R3 代表 6,7-二氫-4H-吡唑并[5,1-c][1,4]噁嗪-2-基、5-甲基-4,5,6,7-四氫吡唑并[1,5-a]吡嗪-2-基或4,5,6,7-四氫吡唑并[1,5-a]吡嗪-2-基, 或代表 吡唑并[1,5-a]嘧啶-3-基、吡咯并[2,1-f][1,2,4]三嗪-7-基、吡咯并[1,2-b]嗒嗪-7-基、噻吩并[2,3-b]吡嗪-7-基、5-胺基吡唑并[1,5-a]嘧啶-3-基、2-胺基吡咯并[2,1-f][1,2,4]三嗪-7-基、2-胺基吡咯并[1,2-b]嗒嗪-7-基、2-胺基噻吩并[2,3-b]吡嗪-7-基; R4 代表環丙基、六氫吡啶-4-基、1-甲基六氫吡啶-4-基、吡咯啶-3-基、1-甲基吡咯啶-3-基、氮雜環丁-3-基、1-甲基氮雜環丁-3-基、氧雜環丁-3-基、四氫呋喃-3-基或四氫-2H-吡喃-4-基,其中氧雜環丁-3-基、四氫呋喃-3-基、四氫-2H-吡喃-4-基可視情況經甲基單取代或二取代, 或代表 C1 -C6 -烷基,其中C1 -C6 -烷基可視情況經以下取代:羥基、C(=O)OMe、C(=O)OEt、C(=O)OH、C(=O)NH2 、SMe、SEt、S(=O)2 Me、S(=O)2 Et、甲氧基、乙氧基、N(CH3 )2 、環丙基、環丁基、環戊基、氧雜環丁-3-基、四氫呋喃-3-基、四氫-2H-吡喃-4-基, 或代表 三氟甲基、2,2,2-三氟乙基或2,2-二氟乙基; 及其非鏡像異構物、鏡像異構物、代謝物、鹽、溶劑合物或其鹽之溶劑合物。A compound of formula (I), wherein R 1 represents chloro or OR 4 , and R 2 represents C 1 -C 6 -alkyl, which may optionally be substituted, independently of each other, by the same or different substituents selected from the group consisting of Mono- or poly-substituted: 1 to 3 fluorine atoms, 1 to 2 hydroxyl groups, oxetanyl groups, tetrahydrofuranyl groups, and C 1 -C 3 -alkoxy groups, wherein the C 1 -C 3 -alkoxy group It may be substituted by a hydroxyl group or by C(=O)OH, C(=O)Me, C(=O)Et; or R 2 represents a 3-sided oxybutyl group; R 3 represents a group selected from the group consisting of: Wherein R 5 represents hydrogen, cyclopropyl or C 1 -C 6 -alkyl, wherein the C 1 -C 6 -alkyl group may optionally be mono-substituted to trisubstituted with cyclopropyl, cyano and hydroxy, wherein each substitution The group may occur only once, or may be mono-substituted to penta-substituted with a fluorine atom; and R 6 represents hydrogen; or R 3 represents a group Wherein R 7 represents hydrogen, cyclopropyl, cyano, NH 2 , NH(C1-C 6 -alkyl), N(C 1 -C 6 -alkyl) 2 , pyrrolidin-1-yl, hexahydropyridine -1-yl, morpholin-4-yl, 4-methylhexahydropyrazin-1-yl or represents C 1 -C 6 -alkyl, wherein C 1 -C 6 -alkyl may optionally be cyclopropyl , cyano and hydroxy, mono- to tri-substituted, wherein each substituent may occur only once, or may be mono-substituted to penta-substituted through a fluorine atom; and R 8 , R 9 , R 10 represent hydrogen or R 3 represents a group Wherein R 11 represents a C 3 -C 6 -cycloalkyl group or a C 1 -C 6 -alkyl group, wherein the C 1 -C 6 -alkyl group may be monosubstituted to trisubstituted by a cyclopropyl group, a cyano group and a hydroxy group, wherein Each substituent may occur only once, or may be mono-substituted to penta-substituted with a fluorine atom; and R 12 represents hydrogen or R 3 represents 6,7-dihydro-4H-pyrazolo[5,1-c][1, 4]oxazin-2-yl, 5-methyl-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrazin-2-yl or 4,5,6,7-tetra Hydropyrazolo[1,5-a]pyrazin-2-yl, or represents pyrazolo[1,5-a]pyrimidin-3-yl, pyrrolo[2,1-f][1,2, 4] Triazin-7-yl, pyrrolo[1,2-b]pyridazin-7-yl, thieno[2,3-b]pyrazin-7-yl, 5-aminopyrazolo[1 , 5-a]pyrimidin-3-yl, 2-aminopyrrolo[2,1-f][1,2,4]triazin-7-yl, 2-aminopyrrolo[1,2-b Pyridazine-7-yl, 2-aminothieno[2,3-b]pyrazin-7-yl; R 4 represents cyclopropyl, hexahydropyridin-4-yl, 1-methylhexahydropyridine 4-yl, pyrrolidin-3-yl, 1-methylpyrrolidin-3-yl, azetidin-3-yl, 1-methylazetidin-3-yl, oxetane- 3-yl, tetrahydrofuran-3-yl or tetrahydro-2H-pyran-4-yl, wherein oxetan-3-yl, tetrahydrofuran-3-yl, tetrahydro-2H- Thiopyran-4-yl optionally substituted by mono or di-substituted by methyl, or represents C 1 -C 6 - alkyl, wherein C 1 -C 6 - alkyl optionally substituted by the following: hydroxy, C (= O) OMe , C(=O)OEt, C(=O)OH, C(=O)NH 2 , SMe, SEt, S(=O) 2 Me, S(=O) 2 Et, methoxy, ethoxy , N(CH 3 ) 2 , cyclopropyl, cyclobutyl, cyclopentyl, oxetan-3-yl, tetrahydrofuran-3-yl, tetrahydro-2H-pyran-4-yl, or represents three a solvent such as fluoromethyl, 2,2,2-trifluoroethyl or 2,2-difluoroethyl; and its non-image, isomer, metabolite, metabolite, salt, solvate or salt thereof Compound. 如請求項2之通式(I)化合物,其中 R1 代表氯或O-R4 ; R2 代表C2 -C5 -烷基,其 經1至2個羥基或 經氧雜環丁-3-基或經四氫呋喃-3-基或 經基團OCH2 C(=O)OH、OCH2 C(=O)OMe、OCH2 CH2 OH或 經C1 -C3 -烷氧基取代, 其中該C1 -C3 -烷氧基 可經羥基或 經C(=O)OH、C(=O)Me、C(=O)Et取代; 或R2 代表3,3,3-三氟丙基、4,4,4-三氟丁基或3-側氧基丁基; R3 代表1-(C1 -C6 -烷基)-1H-吡唑-3-基,其中該C1 -C6 -烷基取代基可視情況經環丙基單取代、經氟原子單取代至五取代且視情況經羥基單取代, 或 代表2-環丙基-1,3-噻唑-4-基或2-(C1 -C6 -烷基)-1,3-噻唑-4-基,其中該C1 -C6 -烷基取代基可視情況經環丙基單取代、經氟原子單取代至五取代且視情況經羥基單取代, 或 代表4-(C1 -C6 -烷基)-1,3-噻唑-2-基,其中該C1 -C6 -烷基取代基可視情況經環丙基單取代、經氟原子單取代至五取代且視情況經羥基單取代, 或 代表2-環丙基-1,3-噁唑-4-基或2-(C1 -C6 -烷基)-1,3-噁唑-4-基,其中該C1 -C6 -烷基取代基可視情況經環丙基單取代、經氟原子單取代至五取代且視情況經羥基單取代, 或 代表2-(C1 -C6 -烷基)-1,3-噁唑-5-基,其中該C1 -C6 -烷基取代基可視情況經環丙基單取代、經氟原子單取代至五取代且視情況經羥基單取代, 或 代表6-(C1 -C6 -烷基)吡啶-2-基,其中該C1 -C6 -烷基取代基可視情況 經環丙基單取代、經氟原子單取代至五取代且視情況經羥基單取代, 或代表6-胺基吡啶-2-基、6-((C1 -C4 -烷基)胺基)吡啶-2-基、6-(二-(C1 -C4 -烷基)胺基)吡啶-2-基、6-(吡咯啶-1-基)吡啶-2-基、6-(六氫吡啶-1-基)吡啶-2-基、6-(嗎啉-4-基)吡啶-2-基、6-(4-甲基六氫吡嗪-1-基)吡啶-2-基, 或 代表6,7-二氫-4H-吡唑并[5,1-c][1,4]噁嗪-2-基、5-甲基-4,5,6,7-四氫吡唑并[1,5-a]吡嗪-2-基或4,5,6,7-四氫吡唑并[1,5-a]吡嗪-2-基, 或為 吡唑并[1,5-a]嘧啶-3-基、吡咯并[2,1-f][1,2,4]三嗪-7-基、吡咯并[1,2-b]嗒嗪-7-基、噻吩并[2,3-b]吡嗪-7-基、5-胺基吡唑并[1,5-a]嘧啶-3-基、2-胺基吡咯并[2,1-f][1,2,4]三嗪-7-基、2-胺基吡咯并[1,2-b]嗒嗪-7-基、2-胺基噻吩并[2,3-b]吡嗪-7-基; R4 代表C1 -C4 -烷基,或 代表氧雜環丁-3-基、四氫呋喃-3-基或四氫-2H-吡喃-4-基 或代表 2-羥基乙基、3-羥基丙基、2-羥基丙基、2-羥基-2-甲基丙基 或代表 CH2 C(=O)OMe、CH2 C(=O)OEt、CH2 C(=O)OH、CH2 CH2 SMe、2-(甲基磺醯基)乙基、3-(甲基磺醯基)丙基、2-甲氧基乙基、2-乙氧基乙基、CH2 CH2 N(CH3 )2 、環丙基甲基、環丁基甲基、環戊基甲基、氧雜環丁-3-基甲基、四氫呋喃-3-基甲基、四氫-2H-吡喃-4-基甲基 或代表 三氟甲基、2,2,2-三氟乙基或2,2-二氟乙基; R5 代表氫或氟; 及其非鏡像異構物、鏡像異構物、代謝物、鹽、溶劑合物或其鹽之溶劑合物。A compound of the formula (I) according to claim 2, wherein R 1 represents chloro or OR 4 ; R 2 represents C 2 -C 5 -alkyl, which is 1 to 2 hydroxy or oxetan-3-yl Or substituted with tetrahydrofuran-3-yl or via the group OCH 2 C(=O)OH, OCH 2 C(=O)OMe, OCH 2 CH 2 OH or via C 1 -C 3 -alkoxy, where C 1 -C 3 -alkoxy may be substituted by hydroxy or by C(=O)OH, C(=O)Me, C(=O)Et; or R 2 represents 3,3,3-trifluoropropyl, 4,4,4-trifluorobutyl or 3-sided oxybutyl; R 3 represents 1-(C 1 -C 6 -alkyl)-1H-pyrazol-3-yl, wherein the C 1 -C The 6 -alkyl substituent may optionally be mono-substituted with a cyclopropyl group, monosubstituted to penta-substituted with a fluorine atom and optionally substituted by a hydroxy group, or represented by 2-cyclopropyl-1,3-thiazol-4-yl or 2 -(C 1 -C 6 -alkyl)-1,3-thiazol-4-yl, wherein the C 1 -C 6 -alkyl substituent may optionally be monosubstituted by cyclopropyl, monosubstituted by fluorine atom to five Substituting and optionally monosubstituted by a hydroxy group, or representing 4-(C 1 -C 6 -alkyl)-1,3-thiazol-2-yl, wherein the C 1 -C 6 -alkyl substituent may optionally be cyclic Propyl monosubstituted, monosubstituted to pentasubstituted by fluorine atom and optionally substituted by hydroxy group Or represents 2-propyl-1,3-oxazol-4-yl or 2- (C 1 -C 6 - alkyl) -1,3-oxazol-4-yl, wherein the C 1 -C 6 - an alkyl substituent may optionally be monosubstituted by cyclopropyl, monosubstituted to pentasubstituted by fluorine atom and optionally substituted by hydroxy, or represented by 2-(C 1 -C 6 -alkyl)-1,3- An oxazol-5-yl group, wherein the C 1 -C 6 -alkyl substituent is optionally monosubstituted by a cyclopropyl group, monosubstituted to pentasubstituted with a fluorine atom, and optionally substituted by a hydroxy group, or represents 6-(C 1 a -C 6 -alkyl)pyridin-2-yl group, wherein the C 1 -C 6 -alkyl substituent may optionally be monosubstituted by a cyclopropyl group, monosubstituted to pentasubstituted with a fluorine atom, and optionally monosubstituted by a hydroxy group, Or represents 6-aminopyridin-2-yl, 6-((C 1 -C 4 -alkyl)amino)pyridin-2-yl, 6-(di-(C 1 -C 4 -alkyl)amine Pyridin-2-yl, 6-(pyrrolidin-1-yl)pyridin-2-yl, 6-(hexahydropyridin-1-yl)pyridin-2-yl, 6-(morpholin-4-yl Pyridin-2-yl, 6-(4-methylhexahydropyrazin-1-yl)pyridin-2-yl, or 6,7-dihydro-4H-pyrazolo[5,1-c] [1,4]oxazin-2-yl, 5-methyl-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrazin-2-yl or 4,5,6, 7-tetrahydropyrazolo[1,5-a] Pyrazin-2-yl, or pyrazolo[1,5-a]pyrimidin-3-yl, pyrrolo[2,1-f][1,2,4]triazin-7-yl, pyrrolo[ 1,2-b]pyridazin-7-yl, thieno[2,3-b]pyrazin-7-yl, 5-aminopyrazolo[1,5-a]pyrimidin-3-yl, 2 -aminopyrrolo[2,1-f][1,2,4]triazin-7-yl, 2-aminopyrrolo[1,2-b]pyridazin-7-yl, 2-amino group Thieno[2,3-b]pyrazin-7-yl; R 4 represents C 1 -C 4 -alkyl, or represents oxetan-3-yl, tetrahydrofuran-3-yl or tetrahydro-2H- Pyran-4-yl or represents 2-hydroxyethyl, 3-hydroxypropyl, 2-hydroxypropyl, 2-hydroxy-2-methylpropyl or represents CH 2 C(=O)OMe, CH 2 C (=O)OEt, CH 2 C(=O)OH, CH 2 CH 2 SMe, 2-(methylsulfonyl)ethyl, 3-(methylsulfonyl)propyl, 2-methoxy Ethyl, 2-ethoxyethyl, CH 2 CH 2 N(CH 3 ) 2 , cyclopropylmethyl, cyclobutylmethyl, cyclopentylmethyl, oxetan-3-ylmethyl, tetrahydrofuran -3-ylmethyl, tetrahydro-2H-pyran-4-ylmethyl or represents trifluoromethyl, 2,2,2-trifluoroethyl or 2,2-difluoroethyl; R 5 represents Hydrogen or fluorine; and its non-image isomers, mirror image isomers, metabolites, salts, solvates or salts thereof Solvate. 如請求項3之通式(I)化合物,其中 R1 代表氯或O-R4 ; R2 代表3-羥基丙基、3-羥基丁基、2-羥基乙基、3-羥基-3-甲基丁基、4-羥基丁基、4-羥基戊基、2,3-二羥基丙基、2,3-二羥基-2-甲基丙基、2-羥基丙基, 氧雜環丁-3-基甲基、氧雜環丁-3-基乙基, 四氫呋喃-3-基甲基、四氫呋喃-3-基乙基, 3-側氧基丁基, 3-甲氧基-3-甲基丁基、3-甲氧基丙基、2-甲氧基乙基, 4,4,4-三氟丁基、3,3,3-三氟丙基, 2-(2-羥基乙氧基)乙基, CH2 CH2 OCH2 C(=O)OH、CH2 CH2 OCH2 C(=O)OEt; R3 代表1-(二氟甲基)-1H-吡唑-3-基、1-甲基-1H-吡唑-3-基、1-乙基-1H-吡唑-3-基、1-(2,2,2-三氟乙基)-1H-吡唑-3-基、1-異丙基-1H-吡唑-3-基、1-丙基-1H-吡唑-3-基、1-第三丁基-1H-吡唑-3-基、1-異丁基-1H-吡唑-3-基, 2-(三氟甲基)-1,3-噻唑-4-基、2-甲基-1,3-噻唑-4-基、2-乙基-1,3-噻唑-4-基、2-丙基-1,3-噻唑-4-基、2-異丙基-1,3-噻唑-4-基、2-第三丁基-1,3-噻唑-4-基、2-環丙基-1,3-噻唑-4-基, 4-(三氟甲基)-1,3-噻唑-2-基、4-甲基-1,3-噻唑-2-基、4-乙基-1,3-噻唑-2-基、4-異丙基-1,3-噻唑-2-基、4-第三丁基-1,3-噻唑-2-基、4-環丙基-1,3-噻唑-2-基, 2-甲基-1,3-噁唑-4-基、2-(三氟甲基)-1,3-噁唑-4-基、2-乙基-1,3-噁唑-4-基、2-(1,1-二氟乙基)-1,3-噁唑-4-基、2-(2,2,2-三氟乙基)-1,3-噁唑-4-基、2-異丙基-1,3-噁唑-4-基、2-第三丁基-1,3-噁唑-4-基、2-環丙基-1,3-噁唑-4-基、2-(環丙基甲基)-1,3-噁唑-4-基, 2-甲基-1,3-噁唑-5-基、2-乙基-1,3-噁唑-5-基、2-異丙基-1,3-噁唑-5-基, 6-甲基吡啶-2-基、6-(二氟甲基)吡啶-2-基、6-(三氟甲基)吡啶-2-基、6-乙基吡啶-2-基、6-(1,1-二氟乙基)吡啶-2-基、6-(五氟乙基)吡啶-2-基、6-(2,2,2-三氟乙基)吡啶-2-基、6-丙基吡啶-2-基、6-異丙基吡啶-2-基、6-(2-羥基丙-2-基)吡啶-2-基、6-第三丁基吡啶-2-基、6-環丙基吡啶-2-基、6-胺基吡啶-2-基、6-(甲基胺基)吡啶-2-基、6-(乙基胺基)吡啶-2-基、6-(二甲基胺基)吡啶-2-基, 6,7-二氫-4H-吡唑并[5,1-c][1,4]噁嗪-2-基、5-甲基-4,5,6,7-四氫吡唑并[1,5-a]吡嗪-2-基、4,5,6,7-四氫吡唑并[1,5-a]吡嗪-2-基, 吡唑并[1,5-a]嘧啶-3-基、吡咯并[2,1-f][1,2,4]三嗪-7-基、吡咯并[1,2-b]嗒嗪-7-基、噻吩并[2,3-b]吡嗪-7-基; R4 代表甲基、乙基、異丙基、丙基、環丙基甲基、氧雜環丁-3-基、氧雜環丁-3-基甲基、四氫呋喃-3-基、2-羥基乙基、三氟甲基、2,2,2-三氟乙基、2,2-二氟乙基、2-(甲基磺醯基)乙基、3-(甲基磺醯基)丙基、2-(甲基硫基)乙基、CH2 C(=O)OEt、CH2 C(=O)OH; R5 代表氫; 及其非鏡像異構物、鏡像異構物、代謝物、鹽、溶劑合物或其鹽之溶劑合物。A compound of the formula (I) according to claim 3, wherein R 1 represents chlorine or OR 4 ; R 2 represents 3-hydroxypropyl, 3-hydroxybutyl, 2-hydroxyethyl, 3-hydroxy-3-methyl Butyl, 4-hydroxybutyl, 4-hydroxypentyl, 2,3-dihydroxypropyl, 2,3-dihydroxy-2-methylpropyl, 2-hydroxypropyl, oxetane-3 -ylmethyl, oxetan-3-ylethyl, tetrahydrofuran-3-ylmethyl, tetrahydrofuran-3-ylethyl, 3-sided oxybutyl, 3-methoxy-3-methyl Butyl, 3-methoxypropyl, 2-methoxyethyl, 4,4,4-trifluorobutyl, 3,3,3-trifluoropropyl, 2-(2-hydroxyethoxy) Ethyl, CH 2 CH 2 OCH 2 C(=O)OH, CH 2 CH 2 OCH 2 C(=O)OEt; R 3 represents 1-(difluoromethyl)-1H-pyrazol-3-yl , 1-methyl-1H-pyrazol-3-yl, 1-ethyl-1H-pyrazol-3-yl, 1-(2,2,2-trifluoroethyl)-1H-pyrazole-3 -yl, 1-isopropyl-1H-pyrazol-3-yl, 1-propyl-1H-pyrazol-3-yl, 1-tert-butyl-1H-pyrazol-3-yl, 1- Isobutyl-1H-pyrazol-3-yl, 2-(trifluoromethyl)-1,3-thiazol-4-yl, 2-methyl-1,3-thiazol-4-yl, 2-B -1,3-thiazol-4-yl, 2-propyl-1,3-thiazol-4-yl, 2-isopropyl-1,3-thiazol-4-yl, 2- Third butyl-1,3-thiazol-4-yl, 2-cyclopropyl-1,3-thiazol-4-yl, 4-(trifluoromethyl)-1,3-thiazol-2-yl, 4-methyl-1,3-thiazol-2-yl, 4-ethyl-1,3-thiazol-2-yl, 4-isopropyl-1,3-thiazol-2-yl, 4-third Butyl-1,3-thiazol-2-yl, 4-cyclopropyl-1,3-thiazol-2-yl, 2-methyl-1,3-oxazol-4-yl, 2-(trifluoro Methyl)-1,3-oxazol-4-yl, 2-ethyl-1,3-oxazol-4-yl, 2-(1,1-difluoroethyl)-1,3-oxazole 4-yl, 2-(2,2,2-trifluoroethyl)-1,3-oxazol-4-yl, 2-isopropyl-1,3-oxazol-4-yl, 2- Third butyl-1,3-oxazol-4-yl, 2-cyclopropyl-1,3-oxazol-4-yl, 2-(cyclopropylmethyl)-1,3-oxazole- 4-yl, 2-methyl-1,3-oxazol-5-yl, 2-ethyl-1,3-oxazol-5-yl, 2-isopropyl-1,3-oxazolyl-5 -yl, 6-methylpyridin-2-yl, 6-(difluoromethyl)pyridin-2-yl, 6-(trifluoromethyl)pyridin-2-yl, 6-ethylpyridin-2-yl 6-(1,1-Difluoroethyl)pyridin-2-yl, 6-(pentafluoroethyl)pyridin-2-yl, 6-(2,2,2-trifluoroethyl)pyridine-2 -yl,6-propylpyridin-2-yl, 6-isopropylpyridin-2-yl, 6-(2-hydroxypropan-2-yl)pyridin-2-yl, 6-tert-butylpyridine- 2-based, 6-cyclopropylpyridin-2-yl 6-Aminopyridin-2-yl, 6-(methylamino)pyridin-2-yl, 6-(ethylamino)pyridin-2-yl, 6-(dimethylamino)pyridine-2 -yl, 6,7-dihydro-4H-pyrazolo[5,1-c][1,4]oxazin-2-yl, 5-methyl-4,5,6,7-tetrahydropyridyl Zizo[1,5-a]pyrazin-2-yl, 4,5,6,7-tetrahydropyrazolo[1,5-a]pyrazin-2-yl, pyrazolo[1,5 -a]pyrimidin-3-yl, pyrrolo[2,1-f][1,2,4]triazin-7-yl, pyrrolo[1,2-b]pyridazin-7-yl, thieno [2,3-b]pyrazin-7-yl; R 4 represents methyl, ethyl, isopropyl, propyl, cyclopropylmethyl, oxetan-3-yl, oxetane- 3-ylmethyl, tetrahydrofuran-3-yl, 2-hydroxyethyl, trifluoromethyl, 2,2,2-trifluoroethyl, 2,2-difluoroethyl, 2-(methylsulfonate Ethyl, 3-(methylsulfonyl)propyl, 2-(methylthio)ethyl, CH 2 C(=O)OEt, CH 2 C(=O)OH; R 5 represents hydrogen And solvates thereof, which are non-image isomers, mirror image isomers, metabolites, salts, solvates or salts thereof. 如請求項4之通式(I)化合物,其中 R1 代表氯或O-R4 ; R2 代表3-羥基丙基、3-羥基丁基、2-羥基乙基、3-羥基-3-甲基丁基、2,3-二羥基丙基, 氧雜環丁-3-基甲基、四氫呋喃-3-基甲基、3-側氧基丁基, 3-甲氧基-3-甲基丁基、3-甲氧基丙基、2-甲氧基乙基, 4,4,4-三氟丁基, 2-(2-羥基乙氧基)乙基, CH2 CH2 OCH2 C(=O)OH、CH2 CH2 OCH2 C(=O)OEt; R3 代表1-(二氟甲基)-1H-吡唑-3-基、1-乙基-1H-吡唑-3-基、1-異丙基-1H-吡唑-3-基、2-(三氟甲基)-1,3-噁唑-4-基、2-(三氟甲基)-1,3-噻唑-4-基、2-環丙基-1,3-噁唑-4-基、2-甲基-1,3-噁唑-5-基、2-甲基-1,3-噻唑-4-基、4-(三氟甲基)-1,3-噻唑-2-基、6-(1,1-二氟乙基)吡啶-2-基、6-(2-羥基丙-2-基)吡啶-2-基、6-(二氟甲基)吡啶-2-基、6-(五氟乙基)吡啶-2-基、6-(三氟甲基)吡啶-2-基、6,7-二氫-4H-吡唑并[5,1-c][1,4]噁嗪-2-基、6-胺基吡啶-2-基、吡唑并[1,5-a]嘧啶-3-基; R4 代表甲基、乙基、異丙基、環丙基甲基、氧雜環丁-3-基、氧雜環丁-3-基甲基、四氫呋喃-3-基、2-羥基乙基、三氟甲基、3-(甲基磺醯基)丙基、2-(甲基硫基)乙基、CH2 C(=O)OEt、CH2 C(=O)OH; 及其非鏡像異構物、鏡像異構物、代謝物、鹽、溶劑合物或其鹽之溶劑合物。A compound of the formula (I) according to claim 4, wherein R 1 represents chlorine or OR 4 ; R 2 represents 3-hydroxypropyl, 3-hydroxybutyl, 2-hydroxyethyl, 3-hydroxy-3-methyl Butyl, 2,3-dihydroxypropyl, oxetan-3-ylmethyl, tetrahydrofuran-3-ylmethyl, 3-sided oxybutyl, 3-methoxy-3-methylbutyl , 3-methoxypropyl, 2-methoxyethyl, 4,4,4-trifluorobutyl, 2-(2-hydroxyethoxy)ethyl, CH 2 CH 2 OCH 2 C ( =O)OH, CH 2 CH 2 OCH 2 C(=O)OEt; R 3 represents 1-(difluoromethyl)-1H-pyrazol-3-yl, 1-ethyl-1H-pyrazole-3 -yl, 1-isopropyl-1H-pyrazol-3-yl, 2-(trifluoromethyl)-1,3-oxazol-4-yl, 2-(trifluoromethyl)-1,3 -thiazol-4-yl, 2-cyclopropyl-1,3-oxazol-4-yl, 2-methyl-1,3-oxazol-5-yl, 2-methyl-1,3-thiazole 4-yl, 4-(trifluoromethyl)-1,3-thiazol-2-yl, 6-(1,1-difluoroethyl)pyridin-2-yl, 6-(2-hydroxypropyl- 2-yl)pyridin-2-yl, 6-(difluoromethyl)pyridin-2-yl, 6-(pentafluoroethyl)pyridin-2-yl, 6-(trifluoromethyl)pyridine-2- 6,6-dihydro-4H-pyrazolo[5,1-c][1,4]oxazin-2-yl, 6-aminopyridin-2-yl, pyrazolo[1,5 -a] pyrimidin-3-yl; R 4 generations Methyl, ethyl, isopropyl, cyclopropylmethyl, oxetan-3-yl, oxetan-3-ylmethyl, tetrahydrofuran-3-yl, 2-hydroxyethyl, trifluoro Methyl, 3-(methylsulfonyl)propyl, 2-(methylthio)ethyl, CH 2 C(=O)OEt, CH 2 C(=O)OH; a solvate of a substance, a mirror image isomer, a metabolite, a salt, a solvate or a salt thereof. 如請求項5之通式(I)化合物,其中 R1 代表O-R4 ; R2 代表3-羥基-3-甲基丁基; R3 代表1-(二氟甲基)-1H-吡唑-3-基、1-乙基-1H-吡唑-3-基、2-(三氟甲基)-1,3-噁唑-4-基、2-(三氟甲基)-1,3-噻唑-4-基、2-環丙基-1,3-噁唑-4-基、2-甲基-1,3-噁唑-5-基、2-甲基-1,3-噻唑-4-基、4-(三氟甲基)-1,3-噻唑-2-基、6-(1,1-二氟乙基)吡啶-2-基、6-(2-羥基丙-2-基)吡啶-2-基、6-(二氟甲基)吡啶-2-基、6-(三氟甲基)吡啶-2-基、6-胺基吡啶-2-基、吡唑并[1,5-a]嘧啶-3-基; R4 代表甲基、乙基、異丙基、環丙基甲基、氧雜環丁-3-基、氧雜環丁-3-基甲基、2-羥基乙基; 及其非鏡像異構物、鏡像異構物、代謝物、鹽、溶劑合物或其鹽之溶劑合物。A compound of the formula (I) according to claim 5, wherein R 1 represents OR 4 ; R 2 represents 3-hydroxy-3-methylbutyl; and R 3 represents 1-(difluoromethyl)-1H-pyrazole- 3-yl, 1-ethyl-1H-pyrazol-3-yl, 2-(trifluoromethyl)-1,3-oxazol-4-yl, 2-(trifluoromethyl)-1,3 -thiazol-4-yl, 2-cyclopropyl-1,3-oxazol-4-yl, 2-methyl-1,3-oxazol-5-yl, 2-methyl-1,3-thiazole 4-yl, 4-(trifluoromethyl)-1,3-thiazol-2-yl, 6-(1,1-difluoroethyl)pyridin-2-yl, 6-(2-hydroxypropyl- 2-yl)pyridin-2-yl, 6-(difluoromethyl)pyridin-2-yl, 6-(trifluoromethyl)pyridin-2-yl, 6-aminopyridin-2-yl, pyrazole And [1,5-a]pyrimidin-3-yl; R 4 represents methyl, ethyl, isopropyl, cyclopropylmethyl, oxetan-3-yl, oxetan-3-yl A solvate of a methyl group, a 2-hydroxyethyl group; and a non-image thereof, a mirror image isomer, a metabolite, a salt, a solvate or a salt thereof. 如請求項5之通式(I)化合物,其係 1-(二氟甲基)-N-[2-(3-羥基-3-甲基丁基)-6-甲氧基-2H-吲唑-5-基]-1H-吡唑-3-甲醯胺 N-[2-(3-羥基-3-甲基丁基)-6-甲氧基-2H-吲唑-5-基]-2-(三氟甲基)-1,3-噁唑-4-甲醯胺 6-(二氟甲基)-N-[2-(3-羥基丙基)-6-甲氧基-2H-吲唑-5-基]吡啶-2-甲醯胺 N-[6-乙氧基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]-4-(三氟甲基)-1,3-噻唑-2-甲醯胺 N-[6-乙氧基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]-2-(三氟甲基)-1,3-噻唑-4-甲醯胺 N-[6-乙氧基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]-2-甲基-1,3-噁唑-5-甲醯胺 1-(二氟甲基)-N-[6-乙氧基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]-1H-吡唑-3-甲醯胺 N-[6-乙氧基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]-2-甲基-1,3-噻唑-4-甲醯胺 2-環丙基-N-[6-乙氧基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]-1,3-噁唑-4-甲醯胺 6-胺基-N-[6-乙氧基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]吡啶-2-甲醯胺 N-[6-乙氧基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 N-[2-(3-羥基-3-甲基丁基)-6-(氧雜環丁-3-基氧基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 N-[2-(3-羥基-3-甲基丁基)-6-(氧雜環丁-3-基氧基)-2H-吲唑-5-基]-2-(三氟甲基)-1,3-噻唑-4-甲醯胺 N-[2-(3-羥基-3-甲基丁基)-6-(氧雜環丁-3-基氧基)-2H-吲唑-5-基]-4-(三氟甲基)-1,3-噻唑-2-甲醯胺 1-(二氟甲基)-N-[2-(3-羥基-3-甲基丁基)-6-(氧雜環丁-3-基甲氧基)-2H-吲唑-5-基]-1H-吡唑-3-甲醯胺 6-(二氟甲基)-N-[2-(3-羥基-3-甲基丁基)-6-(氧雜環丁-3-基甲氧基)-2H-吲唑-5-基]吡啶-2-甲醯胺 N-2-(3-羥基-3-甲基丁基)-6-[(3S)-四氫呋喃-3-基氧基]-2H-吲唑-5-基-2-甲基-1,3-噁唑-5-甲醯胺 N-2-(3-羥基-3-甲基丁基)-6-[(3S)-四氫呋喃-3-基氧基]-2H-吲唑-5-基-6-(三氟甲基)吡啶-2-甲醯胺 6-胺基-N-2-(3-羥基-3-甲基丁基)-6-[(3S)-四氫呋喃-3-基氧基]-2H-吲唑-5-基吡啶-2-甲醯胺 N-[6-氯-2-(3-甲氧基-3-甲基丁基)-2H-吲唑-5-基]-6-(2-羥基丙-2-基)吡啶-2-甲醯胺 N-[6-氯-2-(3-甲氧基-3-甲基丁基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 N-[6-異丙氧基-2-(2-甲氧基乙基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 N-[6-異丙氧基-2-(3-甲氧基丙基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 6-(二氟甲基)-N-[6-乙氧基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]吡啶-2-甲醯胺 N-[6-(環丙基甲氧基)-2-(3-甲氧基丙基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 N-[6-(環丙基甲氧基)-2-(2-甲氧基乙基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 N-[6-(環丙基甲氧基)-2-(氧雜環丁-3-基甲基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 N-[2-(3-羥基-3-甲基丁基)-6-(三氟甲氧基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 2-環丙基-N-[6-甲氧基-2-(3-甲氧基丙基)-2H-吲唑-5-基]-1,3-噁唑-4-甲醯胺 N-[6-氯-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]-6-(二氟甲基)吡啶-2-甲醯胺 N-[2-(2-羥基乙基)-6-異丙氧基-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 N-[2-(3-羥基丙基)-6-異丙氧基-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 N-2-(3-羥基-3-甲基丁基)-6-[3-(甲基磺醯基)丙氧基]-2H-吲唑-5-基-6-(三氟甲基)吡啶-2-甲醯胺 N-2-(3-羥基-3-甲基丁基)-6-[2-(甲基硫基)乙氧基]-2H-吲唑-5-基-6-(三氟甲基)吡啶-2-甲醯胺 [2-(3-羥基-3-甲基丁基)-5-([6-(三氟甲基)吡啶-2-基]羰基胺基)-2H-吲唑-6-基]氧基乙酸乙酯 N-[2-(3-羥基-3-甲基丁基)-6-(氧雜環丁-3-基甲氧基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 N-[6-(環丙基甲氧基)-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 N-[6-(環丙基甲氧基)-2-(3-羥基丙基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 N-[6-甲氧基-2-(3-側氧基丁基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 N-[2-(2-羥基乙基)-6-甲氧基-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 N-[2-(3-羥基丙基)-6-甲氧基-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 N-2-[(2S)-2,3-二羥基丙基]-6-甲氧基-2H-吲唑-5-基-6-(三氟甲基)吡啶-2-甲醯胺 6-(二氟甲基)-N-[2-(2-羥基乙基)-6-甲氧基-2H-吲唑-5-基]吡啶-2-甲醯胺 N-[6-氯-2-(2-羥基乙基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 N-6-氯-2-[(2R)-2,3-二羥基丙基]-2H-吲唑-5-基-6-(三氟甲基)吡啶-2-甲醯胺 N-[2-(3-羥基-3-甲基丁基)-6-甲氧基-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 N-[2-(3-羥基丁基)-6-甲氧基-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 N-[2-(3-羥基丁基)-6-甲氧基-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 N-[6-甲氧基-2-(3-甲氧基丙基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 N-[6-(2-羥基乙氧基)-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 N-[6-(環丙基甲氧基)-2-(2-羥基乙基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 N-[6-甲氧基-2-(氧雜環丁-3-基甲基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 N-[6-甲氧基-2-(4,4,4-三氟丁基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 N-[6-氯-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 N-[6-甲氧基-2-(2-甲氧基乙基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 N-2-[2-(2-羥基乙氧基)乙基]-6-甲氧基-2H-吲唑-5-基-6-(三氟甲基)吡啶-2-甲醯胺 2-[6-甲氧基-5-([6-(三氟甲基)吡啶-2-基]羰基胺基)-2H-吲唑-2-基]乙氧基乙酸乙酯 [2-(3-羥基-3-甲基丁基)-5-([6-(三氟甲基)吡啶-2-基]羰基胺基)-2H-吲唑-6-基]氧基乙酸 2-[6-甲氧基-5-([6-(三氟甲基)吡啶-2-基]羰基胺基)-2H-吲唑-2-基]乙氧基乙酸 N-[6-乙氧基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]-6-(五氟乙基)吡啶-2-甲醯胺 6-(1,1-二氟乙基)-N-[6-乙氧基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]吡啶-2-甲醯胺 N-[6-乙氧基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]-6,7-二氫-4H-吡唑并[5,1-c][1,4]噁嗪-2-甲醯胺 N-[6-乙氧基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]-1-乙基-1H-吡唑-3-甲醯胺 N-[6-乙氧基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]-1-異丙基-1H-吡唑-3-甲醯胺 N-[6-乙氧基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]吡唑并[1,5-a]嘧啶-3-甲醯胺 N-[6-甲氧基-2-(四氫呋喃-3-基甲基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 N-[2-(3-羥基-3-甲基丁基)-6-甲氧基-2H-吲唑-5-基]-6-(吡咯啶-1-基)吡啶-2-甲醯胺。A compound of the formula (I) according to claim 5 which is 1-(difluoromethyl)-N-[2-(3-hydroxy-3-methylbutyl)-6-methoxy-2H-indole Zyrid-5-yl]-1H-pyrazole-3-carboxamide N-[2-(3-hydroxy-3-methylbutyl)-6-methoxy-2H-indazol-5-yl] -2-(trifluoromethyl)-1,3-oxazole-4-carboxamide 6-(difluoromethyl)-N-[2-(3-hydroxypropyl)-6-methoxy- 2H-carbazol-5-yl]pyridine-2-carboxamide N-[6-ethoxy-2-(3-hydroxy-3-methylbutyl)-2H-indazol-5-yl]- 4-(Trifluoromethyl)-1,3-thiazol-2-carboxamide N-[6-ethoxy-2-(3-hydroxy-3-methylbutyl)-2H-indazole-5 -yl]-2-(trifluoromethyl)-1,3-thiazole-4-carboxamide N-[6-ethoxy-2-(3-hydroxy-3-methylbutyl)-2H- Oxazol-5-yl]-2-methyl-1,3-oxazol-5-carboxamide 1-(difluoromethyl)-N-[6-ethoxy-2-(3-hydroxy- 3-methylbutyl)-2H-indazol-5-yl]-1H-pyrazole-3-carboxamide N-[6-ethoxy-2-(3-hydroxy-3-methylbutyl) -2H-carbazol-5-yl]-2-methyl-1,3-thiazole-4-carboxamide 2-cyclopropyl-N-[6-ethoxy-2-(3-hydroxy- 3-methylbutyl)-2H-indazol-5-yl]-1,3-oxazole-4-carboxamide 6-amino-N-[6-ethoxy-2-(3-hydroxyl) -3-methylbutyl)-2H-indazol-5-yl]pyridine-2-carboxamide N-[6- Oxy-2-(3-hydroxy-3-methylbutyl)-2H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide N-[2-(3 -hydroxy-3-methylbutyl)-6-(oxetan-3-yloxy)-2H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamidine Amine N-[2-(3-hydroxy-3-methylbutyl)-6-(oxetan-3-yloxy)-2H-indazol-5-yl]-2-(trifluoromethyl) -1,3-1,3-thiazol-4-carboxamide N-[2-(3-hydroxy-3-methylbutyl)-6-(oxetan-3-yloxy)-2H-indole Zyrid-5-yl]-4-(trifluoromethyl)-1,3-thiazole-2-carboxamide 1-(difluoromethyl)-N-[2-(3-hydroxy-3-methyl Butyl)-6-(oxetan-3-ylmethoxy)-2H-indazol-5-yl]-1H-pyrazole-3-carboxamide 6-(difluoromethyl)-N -[2-(3-hydroxy-3-methylbutyl)-6-(oxetan-3-ylmethoxy)-2H-indazol-5-yl]pyridine-2-carboxamide N -2-(3-hydroxy-3-methylbutyl)-6-[(3S)-tetrahydrofuran-3-yloxy]-2H-indazol-5-yl-2-methyl-1,3- Oxazole-5-carbamamine N-2-(3-hydroxy-3-methylbutyl)-6-[(3S)-tetrahydrofuran-3-yloxy]-2H-indazol-5-yl- 6-(Trifluoromethyl)pyridine-2-carboxamide 6-amino-N-2-(3-hydroxy-3-methylbutyl)-6-[(3S)-tetrahydrofuran-3-yloxy ]]-2H-carbazole-5-ylpyridine-2-carboxamidine N-[6-Chloro-2-(3-methoxy-3-methylbutyl)-2H-indazol-5-yl]-6-(2-hydroxypropan-2-yl)pyridine-2- Methionamine N-[6-chloro-2-(3-methoxy-3-methylbutyl)-2H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2-methyl Indoleamine N-[6-isopropoxy-2-(2-methoxyethyl)-2H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide N -[6-Isopropoxy-2-(3-methoxypropyl)-2H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide 6-(two Fluoromethyl)-N-[6-ethoxy-2-(3-hydroxy-3-methylbutyl)-2H-indazol-5-yl]pyridine-2-carboxamide N-[6- (cyclopropylmethoxy)-2-(3-methoxypropyl)-2H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide N-[6 -(cyclopropylmethoxy)-2-(2-methoxyethyl)-2H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide N-[ 6-(cyclopropylmethoxy)-2-(oxetan-3-ylmethyl)-2H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamidine Amine N-[2-(3-hydroxy-3-methylbutyl)-6-(trifluoromethoxy)-2H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2 -Procarbamide 2-cyclopropyl-N-[6-methoxy-2-(3-methoxypropyl)-2H-indazol-5-yl]-1,3-oxazole-4- Methionamine N-[6-chloro-2-(3-hydroxy-3-methyl) -2H-carbazol-5-yl]-6-(difluoromethyl)pyridine-2-carboxamide N-[2-(2-hydroxyethyl)-6-isopropoxy-2H- Oxazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide N-[2-(3-hydroxypropyl)-6-isopropoxy-2H-indazole-5- -6-(trifluoromethyl)pyridine-2-carboxamide N-2-(3-hydroxy-3-methylbutyl)-6-[3-(methylsulfonyl)propoxy -2H-carbazol-5-yl-6-(trifluoromethyl)pyridine-2-carboxamide N-2-(3-hydroxy-3-methylbutyl)-6-[2-(A Ethylthio)ethoxy]-2H-indazol-5-yl-6-(trifluoromethyl)pyridine-2-carboxamide [2-(3-hydroxy-3-methylbutyl)-5 -([6-(Trifluoromethyl)pyridin-2-yl]carbonylamino)-2H-indazol-6-yl]oxyacetic acid ethyl N-[2-(3-hydroxy-3-methyl Butyl)-6-(oxetan-3-ylmethoxy)-2H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide N-[6- (cyclopropylmethoxy)-2-(3-hydroxy-3-methylbutyl)-2H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide N -[6-(cyclopropylmethoxy)-2-(3-hydroxypropyl)-2H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide N- [6-Methoxy-2-(3-o-oxybutyl)-2H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide N-[2-( 2-hydroxyethyl - 6-methoxy-2H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide N-[2-(3-hydroxypropyl)-6-methoxy -2H-carbazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide N-2-[(2S)-2,3-dihydroxypropyl]-6-methoxy -2H-indazole-5-yl-6-(trifluoromethyl)pyridine-2-carboxamide 6-(difluoromethyl)-N-[2-(2-hydroxyethyl)-6- Methoxy-2H-indazol-5-yl]pyridine-2-carboxamide N-[6-chloro-2-(2-hydroxyethyl)-2H-indazol-5-yl]-6-( Trifluoromethyl)pyridine-2-carboxamide N-6-chloro-2-[(2R)-2,3-dihydroxypropyl]-2H-indazol-5-yl-6-(trifluoromethyl) Pyridyl-2-carboxamide N-[2-(3-hydroxy-3-methylbutyl)-6-methoxy-2H-indazol-5-yl]-6-(trifluoromethyl Pyridine-2-carboxamide N-[2-(3-hydroxybutyl)-6-methoxy-2H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2-methyl Indoleamine N-[2-(3-hydroxybutyl)-6-methoxy-2H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide N-[6 -Methoxy-2-(3-methoxypropyl)-2H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide N-[6-(2- Hydroxyethoxy)-2-(3-hydroxy-3-methylbutyl)-2H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide N-[6 -(cyclopropylmethoxy)-2-(2-hydroxyl -2H-carbazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide N-[6-methoxy-2-(oxetan-3-ylmethyl) -2H-carbazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide N-[6-methoxy-2-(4,4,4-trifluorobutyl) -2H-carbazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide N-[6-chloro-2-(3-hydroxy-3-methylbutyl)-2H- Oxazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide N-[6-methoxy-2-(2-methoxyethyl)-2H-indazole-5 -yl]-6-(trifluoromethyl)pyridine-2-carboxamide N-2-[2-(2-hydroxyethoxy)ethyl]-6-methoxy-2H-indazole-5 -yl-6-(trifluoromethyl)pyridine-2-carboxamide 2-[6-methoxy-5-([6-(trifluoromethyl)pyridin-2-yl]carbonylamino)- Ethyl 2H-carbazol-2-yl]ethoxyacetate [2-(3-hydroxy-3-methylbutyl)-5-([6-(trifluoromethyl)pyridin-2-yl]carbonyl) Amino)-2H-indazol-6-yl]oxyacetic acid 2-[6-methoxy-5-([6-(trifluoromethyl)pyridin-2-yl]carbonylamino)-2H- Oxazol-2-yl]ethoxyacetic acid N-[6-ethoxy-2-(3-hydroxy-3-methylbutyl)-2H-indazol-5-yl]-6-(pentafluoro Ethyl)pyridine-2-carboxamide 6-(1,1-difluoroethyl)-N-[6-ethoxy-2-(3-hydroxy-3-methylbutyl)-2H-indole Oxazol-5-yl]pyridine- 2-Protonamine N-[6-ethoxy-2-(3-hydroxy-3-methylbutyl)-2H-indazol-5-yl]-6,7-dihydro-4H-pyrazole And [5,1-c][1,4]oxazine-2-carboxamide N-[6-ethoxy-2-(3-hydroxy-3-methylbutyl)-2H-carbazole- 5-yl]-1-ethyl-1H-pyrazole-3-carboxamide N-[6-ethoxy-2-(3-hydroxy-3-methylbutyl)-2H-indazole-5 -yl]-1-isopropyl-1H-pyrazole-3-carboxamide N-[6-ethoxy-2-(3-hydroxy-3-methylbutyl)-2H-indazole-5 -yl]pyrazolo[1,5-a]pyrimidine-3-carboxamide N-[6-methoxy-2-(tetrahydrofuran-3-ylmethyl)-2H-indazol-5-yl] -6-(trifluoromethyl)pyridine-2-carboxamide N-[2-(3-hydroxy-3-methylbutyl)-6-methoxy-2H-indazol-5-yl]- 6-(pyrrolidin-1-yl)pyridine-2-carboxamide. 如請求項6之通式(I)之化合物,其係 1-(二氟甲基)-N-[2-(3-羥基-3-甲基丁基)-6-甲氧基-2H-吲唑-5-基]-1H-吡唑-3-甲醯胺 N-[6-乙氧基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]-2-(三氟甲基)-1,3-噻唑-4-甲醯胺 1-(二氟甲基)-N-[6-乙氧基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]-1H-吡唑-3-甲醯胺 N-[6-乙氧基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]-2-甲基-1,3-噻唑-4-甲醯胺 2-環丙基-N-[6-乙氧基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]-1,3-噁唑-4-甲醯胺 6-胺基-N-[6-乙氧基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]吡啶-2-甲醯胺 N-[6-乙氧基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 N-[2-(3-羥基-3-甲基丁基)-6-(氧雜環丁-3-基氧基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 N-[2-(3-羥基-3-甲基丁基)-6-(氧雜環丁-3-基氧基)-2H-吲唑-5-基]-2-(三氟甲基)-1,3-噻唑-4-甲醯胺 1-(二氟甲基)-N-[2-(3-羥基-3-甲基丁基)-6-(氧雜環丁-3-基甲氧基)-2H-吲唑-5-基]-1H-吡唑-3-甲醯胺 6-(二氟甲基)-N-[2-(3-羥基-3-甲基丁基)-6-(氧雜環丁-3-基甲氧基)-2H-吲唑-5-基]吡啶-2-甲醯胺 N-2-(3-羥基-3-甲基丁基)-6-[(3S)-四氫呋喃-3-基氧基]-2H-吲唑-5-基-2-甲基-1,3-噁唑-5-甲醯胺 N-2-(3-羥基-3-甲基丁基)-6-[(3S)-四氫呋喃-3-基氧基]-2H-吲唑-5-基-6-(三氟甲基)吡啶-2-甲醯胺 6-胺基-N-2-(3-羥基-3-甲基丁基)-6-[(3S)-四氫呋喃-3-基氧基]-2H-吲唑-5-基吡啶-2-甲醯胺 6-(二氟甲基)-N-[6-乙氧基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]吡啶-2-甲醯胺 N-[2-(3-羥基-3-甲基丁基)-6-(氧雜環丁-3-基甲氧基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 N-[2-(3-羥基-3-甲基丁基)-6-甲氧基-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 N-[6-(2-羥基乙氧基)-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]-6-(三氟甲基)吡啶-2-甲醯胺 6-(1,1-二氟乙基)-N-[6-乙氧基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]吡啶-2-甲醯胺 N-[6-乙氧基-2-(3-羥基-3-甲基丁基)-2H-吲唑-5-基]-1-乙基-1H-吡唑-3-甲醯胺。A compound of the formula (I) according to claim 6 which is 1-(difluoromethyl)-N-[2-(3-hydroxy-3-methylbutyl)-6-methoxy-2H- Oxazol-5-yl]-1H-pyrazole-3-carboxamide N-[6-ethoxy-2-(3-hydroxy-3-methylbutyl)-2H-carbazole-5-yl ]-2-(trifluoromethyl)-1,3-thiazole-4-carboxamide 1-(difluoromethyl)-N-[6-ethoxy-2-(3-hydroxy-3-methyl) Benzyl)-2H-indazol-5-yl]-1H-pyrazole-3-carboxamide N-[6-ethoxy-2-(3-hydroxy-3-methylbutyl)-2H -carbazol-5-yl]-2-methyl-1,3-thiazole-4-carboxamide 2-cyclopropyl-N-[6-ethoxy-2-(3-hydroxy-3-methyl Butyl)-2H-carbazol-5-yl]-1,3-oxazole-4-carboxamide 6-amino-N-[6-ethoxy-2-(3-hydroxy-3- Methylbutyl)-2H-indazol-5-yl]pyridine-2-carboxamide N-[6-ethoxy-2-(3-hydroxy-3-methylbutyl)-2H-carbazole -5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide N-[2-(3-hydroxy-3-methylbutyl)-6-(oxetan-3-yl) Oxy)-2H-indazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide N-[2-(3-hydroxy-3-methylbutyl)-6-( Oxetidin-3-yloxy)-2H-indazol-5-yl]-2-(trifluoromethyl)-1,3-thiazole-4-carboxamide 1-(difluoromethyl) -N-[2-(3-hydroxy-3-methylbutyl)-6-(oxa) Cyclobut-3-ylmethoxy)-2H-indazol-5-yl]-1H-pyrazole-3-carboxamide 6-(difluoromethyl)-N-[2-(3-hydroxy- 3-methylbutyl)-6-(oxetan-3-ylmethoxy)-2H-indazol-5-yl]pyridine-2-carboxamide N-2-(3-hydroxy-3 -Methylbutyl)-6-[(3S)-tetrahydrofuran-3-yloxy]-2H-indazol-5-yl-2-methyl-1,3-oxazol-5-carboxamide N -2-(3-Hydroxy-3-methylbutyl)-6-[(3S)-tetrahydrofuran-3-yloxy]-2H-indazole-5-yl-6-(trifluoromethyl)pyridine 2-carbamamine 6-amino-N-2-(3-hydroxy-3-methylbutyl)-6-[(3S)-tetrahydrofuran-3-yloxy]-2H-indazole-5 -ylpyridine-2-carboxamide 6-(difluoromethyl)-N-[6-ethoxy-2-(3-hydroxy-3-methylbutyl)-2H-carbazole-5-yl Pyridine-2-carboxamide N-[2-(3-hydroxy-3-methylbutyl)-6-(oxetan-3-ylmethoxy)-2H-indazole-5-yl -6-(Trifluoromethyl)pyridine-2-carboxamide N-[2-(3-hydroxy-3-methylbutyl)-6-methoxy-2H-indazol-5-yl] -6-(trifluoromethyl)pyridine-2-carboxamide N-[6-(2-hydroxyethoxy)-2-(3-hydroxy-3-methylbutyl)-2H-carbazole- 5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide 6-(1,1-difluoroethyl)-N-[6-ethoxy-2-(3-hydroxy-3) -methylbutyl)-2H-carbazole-5-yl Pyridine-2-carboxamide N-[6-ethoxy-2-(3-hydroxy-3-methylbutyl)-2H-indazol-5-yl]-1-ethyl-1H-pyridyl Oxazole-3-carboxamide. 如請求項1至8中任一項之通式(I)化合物,其用於治療及/或預防疾病。A compound of the formula (I) according to any one of claims 1 to 8 for use in the treatment and/or prevention of a disease. 如請求項1至8中任一項之通式(I)化合物,其用於用以治療及/或預防以下疾病之方法中:贅瘤性病症、皮膚病學病症、婦科病症、心血管病症、肺部病症、眼科病症、神經病症、代謝失調、肝臟病症、發炎病症、自體免疫病症及疼痛。A compound of the formula (I) according to any one of claims 1 to 8 for use in a method for the treatment and/or prevention of a neoplastic disorder, a dermatological disorder, a gynecological disorder, a cardiovascular disorder , pulmonary disorders, ophthalmic disorders, neurological disorders, metabolic disorders, liver disorders, inflammatory conditions, autoimmune disorders and pain. 如請求項1至8中任一項之通式(I)化合物,其用於用以治療及/或預防以下疾病之方法中:淋巴瘤、黃斑部病變、牛皮癬、紅斑性狼瘡、多發性硬化、COPD、痛風、NASH、肝纖維化、胰島素抗性、代謝症候群、脊椎關節炎及類風濕性關節炎、子宮內膜異位症及子宮內膜異位症相關性疼痛及其他子宮內膜異位症相關性症狀(例如痛經、性交疼痛、排尿困難及排糞困難)。A compound of the formula (I) according to any one of claims 1 to 8 for use in a method for the treatment and/or prevention of lymphoma, macular degeneration, psoriasis, lupus erythematosus, multiple sclerosis , COPD, gout, NASH, liver fibrosis, insulin resistance, metabolic syndrome, spondylarthritis and rheumatoid arthritis, endometriosis and endometriosis-related pain and other endometriosis Symptoms associated with symptoms (such as dysmenorrhea, painful intercourse, difficulty urinating, and difficulty in defecation). 如請求項1至8中任一項之通式(I)化合物,其用於治療及/或預防疼痛之方法,該疼痛包括急性、慢性、發炎性及神經病性疼痛,較佳感覺過敏、觸摸痛、關節炎疼痛(例如骨關節炎、類風濕性關節炎及脊椎關節炎)、經前疼痛、子宮內膜異位症相關性疼痛、手術後疼痛、間質性膀胱炎疼痛、CRPS (複雜性區域疼痛症候群)、三叉神經痛、前列腺炎疼痛、由脊髓損傷造成之疼痛、發炎誘導之疼痛、腰部疼痛、癌性疼痛、化學療法相關性疼痛、HIV治療誘導之神經病變、燒傷誘導之疼痛及慢性疼痛。A compound of the formula (I) according to any one of claims 1 to 8, which is a method for the treatment and/or prevention of pain, which comprises acute, chronic, inflammatory and neuropathic pain, preferably hyperesthesia, touch Pain, arthritis pain (eg osteoarthritis, rheumatoid arthritis and spinal arthritis), premenstrual pain, endometriosis-related pain, post-operative pain, interstitial cystitis pain, CRPS (complex Sexual regional pain syndrome), trigeminal neuralgia, prostatitis pain, pain caused by spinal cord injury, inflammation-induced pain, lumbar pain, cancer pain, chemotherapy-related pain, HIV-induced neuropathy, burn-induced pain And chronic pain. 一種如請求項1至8中任一項之通式(I)化合物之用途,其用於製備藥劑。Use of a compound of formula (I) according to any one of claims 1 to 8 for the preparation of a medicament. 如請求項13之用途,其中該藥劑係用於治療及/或預防贅瘤性病症、皮膚病學病症、婦科病症、心血管病症、肺部病症、眼科病症、神經病症、代謝失調、肝臟病症、發炎病症、自體免疫病症及疼痛。The use of claim 13, wherein the agent is for treating and/or preventing a neoplastic disorder, a dermatological disorder, a gynecological disorder, a cardiovascular disorder, a pulmonary disorder, an ophthalmic disorder, a neurological disorder, a metabolic disorder, a liver disorder , inflammatory conditions, autoimmune disorders and pain. 如請求項13或14之用途,其用於治療及/或預防淋巴瘤、黃斑部病變、牛皮癬、紅斑性狼瘡、多發性硬化、COPD、痛風、NASH、肝纖維化、胰島素抗性、代謝症候群、脊椎關節炎及類風濕性關節炎、子宮內膜異位症及子宮內膜異位症相關性疼痛及其他子宮內膜異位症相關性症狀(例如痛經、性交疼痛、排尿困難及排糞困難)。The use of claim 13 or 14, for the treatment and/or prevention of lymphoma, macular degeneration, psoriasis, lupus erythematosus, multiple sclerosis, COPD, gout, NASH, liver fibrosis, insulin resistance, metabolic syndrome , spondylitis and rheumatoid arthritis, endometriosis and endometriosis-related pain and other endometriosis-related symptoms (such as dysmenorrhea, painful intercourse, dysuria, and feces) difficult). 如請求項13或14之用途,其用於治療及/或預防疼痛,該疼痛包括急性、慢性、發炎性及神經病性疼痛,較佳感覺過敏、觸摸痛、關節炎疼痛(例如骨關節炎、類風濕性關節炎及脊椎關節炎)、經前疼痛、子宮內膜異位症相關性疼痛、手術後疼痛、間質性膀胱炎疼痛、CRPS (複雜性區域疼痛症候群)、三叉神經痛、前列腺炎疼痛、由脊髓損傷造成之疼痛、發炎誘導之疼痛、腰部疼痛、癌性疼痛、化學療法相關性疼痛、HIV治療誘導之神經病變、燒傷誘導之疼痛及慢性疼痛。The use of claim 13 or 14, for the treatment and/or prevention of pain, including acute, chronic, inflammatory and neuropathic pain, preferably hyperesthesia, touch pain, arthritic pain (eg osteoarthritis, Rheumatoid arthritis and spondyloarthritis), premenstrual pain, endometriosis-related pain, postoperative pain, interstitial cystitis pain, CRPS (complex regional pain syndrome), trigeminal neuralgia, prostate Inflammation pain, pain caused by spinal cord injury, inflammation-induced pain, lumbar pain, cancer pain, chemotherapy-related pain, HIV-induced neuropathy, burn-induced pain, and chronic pain. 一種藥劑,其包含如請求項1至8中任一項之式(I)化合物與惰性無毒醫藥上適宜之賦形劑之組合。An agent comprising a combination of a compound of formula (I) according to any one of claims 1 to 8 with an inert, non-toxic pharmaceutically suitable excipient.
TW105142812A 2015-12-22 2016-12-22 Novel substituted indazoles, processes for preparation thereof, pharmaceutical preparations comprising them and use thereof for production of medicaments TW201725202A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
EP15202240 2015-12-22

Publications (1)

Publication Number Publication Date
TW201725202A true TW201725202A (en) 2017-07-16

Family

ID=55068837

Family Applications (1)

Application Number Title Priority Date Filing Date
TW105142812A TW201725202A (en) 2015-12-22 2016-12-22 Novel substituted indazoles, processes for preparation thereof, pharmaceutical preparations comprising them and use thereof for production of medicaments

Country Status (4)

Country Link
AR (1) AR107127A1 (en)
TW (1) TW201725202A (en)
UY (1) UY37048A (en)
WO (1) WO2017108744A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111386115A (en) * 2017-11-28 2020-07-07 艾格尔峰生物制药有限公司 Methods and pharmaceutical compositions for treating non-alcoholic steatohepatitis

Families Citing this family (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018049214A1 (en) 2016-09-09 2018-03-15 Incyte Corporation Pyrazolopyridine derivatives as hpk1 modulators and uses thereof for the treatment of cancer
WO2018049191A1 (en) 2016-09-09 2018-03-15 Incyte Corporation Pyrazolopyridone derivatives as hpk1 modulators and uses thereof for the treatment of cancer
TW201811799A (en) 2016-09-09 2018-04-01 美商英塞特公司 Pyrazolopyrimidine compounds and uses thereof
MX2019002750A (en) 2016-09-09 2019-07-04 Incyte Corp Pyrazolopyridine derivatives as hpk1 modulators and uses thereof for the treatment of cancer.
WO2018060174A1 (en) * 2016-09-29 2018-04-05 Bayer Pharma Aktiengesellschaft Substituted benzimidazoles, pharmaceutical preparations containing same, and use of same to produce drugs
US20180228786A1 (en) 2017-02-15 2018-08-16 Incyte Corporation Pyrazolopyridine compounds and uses thereof
JOP20180011A1 (en) 2017-02-16 2019-01-30 Gilead Sciences Inc PYRROLO[1,2-b] PYRIDAZINE DERIVATIVES
US10722495B2 (en) 2017-09-08 2020-07-28 Incyte Corporation Cyanoindazole compounds and uses thereof
CN108663441B (en) * 2017-12-30 2021-03-30 南京海融制药有限公司 Method for checking related substances of tacalcitol ointment
CA3091517A1 (en) 2018-02-20 2019-08-29 Incyte Corporation N-(phenyl)-2-(phenyl)pyrimidine-4-carboxamide derivatives and related compounds as hpk1 inhibitors for treating cancer
US10745388B2 (en) 2018-02-20 2020-08-18 Incyte Corporation Indazole compounds and uses thereof
WO2019164847A1 (en) 2018-02-20 2019-08-29 Incyte Corporation Indazole compounds and uses thereof
US11299473B2 (en) 2018-04-13 2022-04-12 Incyte Corporation Benzimidazole and indole compounds and uses thereof
TW202136268A (en) 2018-07-13 2021-10-01 美商基利科學股份有限公司 Pyrrolo[1,2-b]pyridazine derivatives
US10899755B2 (en) 2018-08-08 2021-01-26 Incyte Corporation Benzothiazole compounds and uses thereof
WO2020035019A1 (en) * 2018-08-17 2020-02-20 浙江海正药业股份有限公司 Indazole amine derivative, preparation method therefor and medical use thereof
MA53726A (en) 2018-09-25 2022-05-11 Incyte Corp PYRAZOLO[4,3-D]PYRIMIDINE COMPOUNDS AS ALK2 AND/OR FGFR MODULATORS
US20220204473A1 (en) * 2018-12-25 2022-06-30 Shanghai Meiyue Biotech Development Co., Ltd. Compound as irak inhibitor
CN111499612B (en) * 2019-01-30 2022-12-30 上海美悦生物科技发展有限公司 Compound as IRAK inhibitor and preparation method and application thereof
CN111560012B (en) * 2019-02-14 2023-06-23 上海美悦生物科技发展有限公司 Compound as IRAK inhibitor
CN111821297A (en) * 2019-04-16 2020-10-27 天津合美医药科技有限公司 Use of isoindoline derivatives for the treatment of immunoglobulin E (IgE) -mediated diseases
US20220227758A1 (en) * 2019-06-26 2022-07-21 Medshine Discovery Inc. Imidazopyridine compound as irak4 inhibitor
MX2021015995A (en) 2019-06-28 2022-03-11 Kymera Therapeutics Inc Irak degraders and uses thereof.
JP2022543155A (en) 2019-08-06 2022-10-07 インサイト・コーポレイション Solid forms of HPK1 inhibitors
JP7353474B2 (en) * 2019-09-24 2023-09-29 上海美悦生物科技発展有限公司 IRAK inhibitor, its manufacturing method and use
CN113278017B (en) * 2021-05-27 2023-03-28 上海应用技术大学 Substituted indazoles, preparation method, application and composition containing same
CN113402499B (en) 2021-06-21 2022-05-13 上海勋和医药科技有限公司 Sulfimide substituted indazole IRAK4 kinase inhibitor, preparation method and application

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI667233B (en) * 2013-12-19 2019-08-01 德商拜耳製藥公司 Novel indazolecarboxamides, processes for their preparation, pharmaceutical preparations comprising them and their use for producing medicaments
US10160753B2 (en) * 2014-01-10 2018-12-25 Aurigene Discovery Technologies Limited Indazole compounds as IRAK4 inhibitors

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111386115A (en) * 2017-11-28 2020-07-07 艾格尔峰生物制药有限公司 Methods and pharmaceutical compositions for treating non-alcoholic steatohepatitis

Also Published As

Publication number Publication date
AR107127A1 (en) 2018-03-21
UY37048A (en) 2017-07-31
WO2017108744A1 (en) 2017-06-29

Similar Documents

Publication Publication Date Title
TW201725202A (en) Novel substituted indazoles, processes for preparation thereof, pharmaceutical preparations comprising them and use thereof for production of medicaments
US20230174508A1 (en) Substituted indazoles, methods for the production thereof, pharmaceutical preparations that contain said new substituted indazoles, and use of said new substituted indazoles to produce drugs
US10435396B2 (en) 2-substituted indazoles, methods for producing same, pharmaceutical preparations that contain same, and use of same to produce drugs
AU2017257211B2 (en) Polymorphic form of N-{6-(2-Hydroxypropan-2-yl)-2-[2-(methylsulphonyl)ethyl]-2H-indazol-5-yl}-6-(trifluoromethyl)pyridine-2-carboxamide
IL262391A (en) Crystalline forms of n-[2-(3-hydroxy-3-methylbutyl)-6-(2-hydroxypropan-2-yl)-2h-indazol-5-yl]-6-(trifluoromethyl)pyridine-2-carboxamide
CA3079060A1 (en) Substituted pyrrolopyridine-derivatives as map4k1 modulators for the treatment of cancer diseases
WO2018060174A1 (en) Substituted benzimidazoles, pharmaceutical preparations containing same, and use of same to produce drugs
TW201803866A (en) Novel substituted benzimidazoles, processes for their preparation, pharmaceutical preparations comprising them and their use for preparing medicaments
EA042116B1 (en) POLYMORPHIC FORM of N-{6-(2-HYDROXYPROPAN-2-YL)-2-[2-(METHYLSULFONIL)ETHYL]-2H-INDAZOL-5-YL}-6-(TRIFLUOROMETHYL)PYRIDINE-2-CARBOXAMIDE