TW202019426A - Pharmaceutical composition comprising quinazoline compound as active ingredient - Google Patents

Pharmaceutical composition comprising quinazoline compound as active ingredient Download PDF

Info

Publication number
TW202019426A
TW202019426A TW108126915A TW108126915A TW202019426A TW 202019426 A TW202019426 A TW 202019426A TW 108126915 A TW108126915 A TW 108126915A TW 108126915 A TW108126915 A TW 108126915A TW 202019426 A TW202019426 A TW 202019426A
Authority
TW
Taiwan
Prior art keywords
compound
oxy
diazaspiro
methyl
indazol
Prior art date
Application number
TW108126915A
Other languages
Chinese (zh)
Inventor
長島建之
Original Assignee
日商安斯泰來製藥股份有限公司
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 日商安斯泰來製藥股份有限公司 filed Critical 日商安斯泰來製藥股份有限公司
Publication of TW202019426A publication Critical patent/TW202019426A/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Landscapes

  • Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

To provide a pharmaceutical composition for treating colon cancer, in particular, KRAS G12C mutation-positive colon cancer. The present inventors have examined compounds having G12C mutation KRAS inhibitory effect. As a result, the present inventors have confirmed that specific quinazoline compounds have G12C mutation KRAS inhibitory effect and a pharmaceutical composition comprising such a compound as an active ingredient has a therapeutic effect on colon cancer, in particular, KRAS G12C mutation-positive colon cancer to thereby complete the present invention.

Description

以喹唑啉化合物作為有效成分之醫藥組成物Pharmaceutical composition with quinazoline compound as active ingredient

本發明係關於以喹唑啉化合物或其製藥學上容許之鹽作為有效成分之大腸癌之治療用醫藥組成物。The present invention relates to a pharmaceutical composition for the treatment of colorectal cancer using a quinazoline compound or a pharmaceutically acceptable salt thereof as an active ingredient.

有報導指出大腸癌為全世界罹患率及致死率高的癌,世界中一年約100萬人新罹患(World Cancer Report 2014)。雖然大腸癌之治療中最有效的手段為外科手術,但近年來化學療法或放射線療法等之進步亦顯著。以歐美為中心進行大規模之臨床試驗的結果,明白對於大腸癌以組合多種類之抗癌劑的化學併用療法為有效,有助於腫瘤退縮或預後延長(J. Clin. Oncol., 22, 229, 2004)。進而化學療法之外,如抗血管內皮生長因子(VEGF)抗體或抗表皮生長因子受體(EGFR)抗體之分子標靶藥物亦被作為第一選擇藥物使用於與化學療法之併用中。已知關於抗EGFR抗體藥,RAS基因突變為負面之效果預測因子(Cancer Res. 66, 3992, 2006),現在抗EGFR抗體藥,僅可適應RAS基因野生型之大腸癌病患。It has been reported that colorectal cancer is a cancer with a high attack rate and fatality rate in the world, and about 1 million people are newly infected in the world each year (World Cancer Report 2014). Although the most effective method in the treatment of colorectal cancer is surgery, recent advances in chemotherapy or radiation therapy have also been remarkable. The results of large-scale clinical trials centered on Europe and the United States have shown that it is effective for colorectal cancer to combine multiple types of anti-cancer chemical combination therapy, which can help tumor regression or prognosis prolongation (J. Clin. Oncol., 22, 229, 2004). In addition to chemotherapy, molecular target drugs such as anti-vascular endothelial growth factor (VEGF) antibodies or anti-epidermal growth factor receptor (EGFR) antibodies are also used as the first-selected drugs in combination with chemotherapy. Regarding anti-EGFR antibody drugs, RAS gene mutations are known to be a predictor of negative effects (Cancer Res. 66, 3992, 2006), and now anti-EGFR antibody drugs can only be adapted to patients with colorectal cancer who have wild type RAS genes.

RAS蛋白質係由188-189個胺基酸而成之約21kDa的低分子鳥苷三磷酸(GTP)結合蛋白質,存在有由KRAS基因、NRAS基因、HRAS基因3個基因所生之4種類的主要蛋白質(KRAS (KRAS4A及KRAS4B)、NRAS、HRAS)。RAS蛋白質存在有活性型之GTP結合型,與不活性型之鳥苷二磷酸(GDP)結合型。RAS蛋白質,因配位體對EGFR等細胞膜受體之刺激等,藉由GDP與GTP交換而活化。活性型RAS,與RAF、PI3K、RALGDS等多達20種類之效應器蛋白質結合,活化下游之傳訊級聯反應。另一方面,活性型RAS藉由內在性之GTP水解(GTPase)活性,藉由將GTP變換成GDP成為不活性型。此GTPase活性因GAP (GTPase活化蛋白質)而增強。因此,RAS擔當了EGFR等之細胞內信號傳遞路徑中重要的「分子開關」之機能,在細胞的成長、增殖、血管生成等之過程中,發揮了重要的作用(Nature rev. cancer, 11, 761, 2011、Nature rev. drug discov., 13, 828, 2014、Nature rev. drug discov., 15, 771, 2016)。The RAS protein is a low-molecular-weight guanosine triphosphate (GTP) binding protein of approximately 21 kDa made from 188-189 amino acids. There are four main types derived from the three genes of KRAS gene, NRAS gene and HRAS gene. Proteins (KRAS (KRAS4A and KRAS4B), NRAS, HRAS). The RAS protein has an active GTP binding type and an inactive guanosine diphosphate (GDP) binding type. RAS protein is activated by the exchange of GDP and GTP due to ligand stimulation of cell membrane receptors such as EGFR. Active RAS combines with up to 20 types of effector proteins such as RAF, PI3K, and RALGDS to activate downstream communication cascade reactions. On the other hand, the active RAS becomes inactive by converting GTP to GDP by its inherent GTP hydrolysis activity (GTPase). This GTPase activity is enhanced by GAP (GTPase activating protein). Therefore, RAS acts as an important "molecular switch" in intracellular signal transmission pathways such as EGFR, and plays an important role in the process of cell growth, proliferation, and angiogenesis (Nature rev. cancer, 11, 761, 2011, Nature rev. drug discov., 13, 828, 2014, Nature rev. drug discov., 15, 771, 2016).

認為若因RAS基因之突變而發生胺基酸置換,則因作為RAS之GTPase機能降低或對GAP之親和性降低,而活性型之比例增加。起因於此的過度的信號傳遞,引發致癌或癌之增殖亢進。大腸癌中,30-40%中於KRAS基因發現有突變,特別是KRAS外顯子2(密碼子12,密碼子13)之點突變多(Ann. Oncol., 27, 1746, 2016)。現行之抗癌劑中對於具有KRAS突變之大腸癌未顯現有效性,對於此部分的未滿足的醫療需求高。It is believed that if amino acid substitution occurs due to mutation of the RAS gene, the function of GTPase as RAS decreases or the affinity for GAP decreases, and the proportion of active types increases. The excessive signal transmission caused by this causes carcinogenesis or hyperproliferation of cancer. 30-40% of colorectal cancers have mutations found in the KRAS gene, especially KRAS exon 2 (codon 12, codon 13) point mutations (Ann. Oncol., 27, 1746, 2016). The current anticancer agents have not shown effectiveness for colorectal cancer with KRAS mutations, and the unmet medical needs for this part are high.

近年來,對於G12C突變KRAS,顯示在稱為開關II(switch II)之區域附近有異位口袋(allosteric pocket)的存在(Nature, 503, 548, 2013),同時報導有藉由對突變半胱胺酸形成共價鍵,而不可逆地與G12C突變KRAS結合的化合物(Cancer Discov., 6, 316, 2016、Cell, 172, 578, 2018)。G12C突變KRAS選擇性的抑制劑,藉由對G12C突變KRAS形成共價鍵,抑制由不活性型向活性型的變換,藉阻斷下游信號而誘導癌細胞死。由於大腸癌中被發現之KRAS突變的3%為G12C突變,故認為具有本作用機制之化合物,對於KRAS G12C突變陽性大腸癌之治療為有用。In recent years, the G12C mutant KRAS has shown the presence of allosteric pockets near the area called switch II (Nature, 503, 548, 2013), and it has been reported that The amino acid forms a covalent bond and is a compound that irreversibly binds to the G12C mutant KRAS (Cancer Discov., 6, 316, 2016, Cell, 172, 578, 2018). The G12C mutant KRAS selective inhibitor, by forming a covalent bond to the G12C mutant KRAS, inhibits the conversion from inactive to active form, and induces cancer cell death by blocking downstream signals. Since 3% of the KRAS mutations found in colorectal cancer are G12C mutations, it is believed that compounds with this mechanism of action are useful for the treatment of KRAS G12C mutation-positive colorectal cancer.

下述之式(A)及式(B)所示之化合物,被報導對於G12C突變KRAS具有結合能力(專利文獻1、2及3)。The compounds represented by the following formula (A) and formula (B) have been reported to have binding ability to the G12C mutant KRAS (Patent Documents 1, 2 and 3).

Figure 02_image001
(式中記號的意思參照該公報)
Figure 02_image001
(See the bulletin for the meaning of the symbols in the formula)

本案之最早的優先權日後公開之文獻(專利文獻4)中,揭示下述之式(C)所示之化合物或其鹽具有G12C突變KRAS抑制活性,對於作為肺癌,特別是KRAS G12C突變陽性肺癌之治療劑為有用。又,該文獻中雖具體揭示本發明之醫藥組成物之有效成分的化合物作為實施例化合物,但未揭示關於對於大腸癌,特別是KRAS G12C突變陽性大腸癌的作用。The earliest priority document published in the present case (Patent Document 4) discloses that the compound represented by the following formula (C) or a salt thereof has a G12C mutation KRAS inhibitory activity, and is a lung cancer, especially KRAS G12C mutation positive lung cancer The therapeutic agent is useful. In this document, although the compound of the active ingredient of the pharmaceutical composition of the present invention is specifically disclosed as an example compound, it does not disclose the effect on colorectal cancer, especially KRAS G12C mutation-positive colorectal cancer.

Figure 02_image003
(式中記號的意思參照該公報)
Figure 02_image003
(See the bulletin for the meaning of the symbols in the formula)

又,非專利文獻1中報導有專利文獻1所揭示之實施例353的化合物(亦稱為ARS-1620),對於人類KRAS G12C突變陽性大腸癌株SW1463具有細胞增殖抑制作用。 [先前技術文獻] [專利文獻]In addition, Non-Patent Document 1 reports that the compound of Example 353 disclosed in Patent Document 1 (also referred to as ARS-1620) has a cell proliferation inhibitory effect on human KRAS G12C mutation-positive colorectal cancer strain SW1463. [Prior Technical Literature] [Patent Literature]

[專利文獻1]國際公開第2015/054572號 [專利文獻2]國際公開第2016/164675號 [專利文獻3]國際公開第2017/087528號 [專利文獻4]國際公開第2018/143315號 [非專利文獻][Patent Literature 1] International Publication No. 2015/054572 [Patent Document 2] International Publication No. 2016/164675 [Patent Literature 3] International Publication No. 2017/087528 [Patent Document 4] International Publication No. 2018/143315 [Non-patent literature]

[非專利文獻1]Cell、2018年、第172卷、第3號、p.578-589[Non-Patent Document 1] Cell, 2018, Volume 172, No. 3, p.578-589

[發明所欲解決之課題][Problems to be solved by the invention]

提供一種大腸癌之治療用醫藥組成物,作為某態樣提供一種KRAS G12C突變陽性大腸癌治療用醫藥組成物。 [解決課題之手段]To provide a medical composition for the treatment of colorectal cancer, and as a certain aspect, to provide a KRAS G12C mutation-positive medical composition for the treatment of colorectal cancer. [Means to solve the problem]

本發明者們,以創製大腸癌之治療用醫藥組成物,特別是KRAS G12C突變陽性大腸癌治療用醫藥組成物為目的,深入研究具有G12C突變KRAS抑制作用之化合物的結果,發現本發明之喹唑啉化合物或其製藥學上容許之鹽具有優異之G12C突變KRAS抑制作用,以此等之化合物作為有效成分的醫藥組成物作為大腸癌之治療用醫藥組成物為有用而完成本發明。 即,本發明係關於一種大腸癌之治療用醫藥組成物,其含有選自由 (+)-1-{7-[6-環丙基-2-{[1-(3-甲氧基丙基)哌啶-4-基]氧基}-7-(5-甲基-1H-吲唑-4-基)-8-(2,2,2-三氟乙氧基)喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬-2-基}丙-2-烯-1-酮(以下,有時記載為「化合物A」)、 (+)-1-{7-[6-環丙基-2-{[1-(2-甲氧基乙基)哌啶-4-基]氧基}-7-(5-甲基-1H-吲唑-4-基)-8-(2,2,2-三氟乙氧基)喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬-2-基}丙-2-烯-1-酮(以下,有時記載為「化合物B」)、 (+)-1-{7-[2-{[1-(2-甲氧基乙基)哌啶-4-基]氧基}-7-(5-甲基-1H-吲唑-4-基)-8-(2,2,2-三氟乙氧基)-6-乙烯基喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬-2-基}丙-2-烯-1-酮(以下,有時記載為「化合物C」),及 (+)-1-(7-{8-乙氧基-7-(5-甲基-1H-吲唑-4-基)-2-[(1-甲基哌啶-4-基)氧基]-6-乙烯基喹唑啉-4-基}-2,7-二氮雜螺[3.5]壬-2-基)丙-2-烯-1-酮(以下,有時記載為「化合物D」) 所成之群組之化合物,或其製藥學上容許之鹽,及製藥學上容許之賦形劑。The inventors of the present invention, for the purpose of creating a pharmaceutical composition for the treatment of colorectal cancer, in particular, a pharmaceutical composition for the treatment of KRAS G12C mutation-positive colorectal cancer, as a result of in-depth study of compounds having an inhibitory effect of G12C mutation KRAS, discovered The oxazoline compound or its pharmaceutically acceptable salt has an excellent G12C mutation KRAS inhibitory effect, and the pharmaceutical composition using these compounds as an active ingredient is useful as a pharmaceutical composition for the treatment of colorectal cancer and completed the present invention. That is, the present invention relates to a pharmaceutical composition for the treatment of colorectal cancer, which contains (+)-1-{7-[6-cyclopropyl-2-{[1-(3-methoxypropyl)piperidin-4-yl]oxy}-7-(5-methyl- 1H-indazol-4-yl)-8-(2,2,2-trifluoroethoxy)quinazolin-4-yl]-2,7-diazaspiro[3.5]non-2-yl }Prop-2-en-1-one (hereinafter sometimes referred to as "compound A"), (+)-1-{7-[6-cyclopropyl-2-{[1-(2-methoxyethyl)piperidin-4-yl]oxy}-7-(5-methyl- 1H-indazol-4-yl)-8-(2,2,2-trifluoroethoxy)quinazolin-4-yl]-2,7-diazaspiro[3.5]non-2-yl }Prop-2-en-1-one (hereinafter sometimes referred to as "compound B"), (+)-1-{7-[2-{[1-(2-methoxyethyl)piperidin-4-yl]oxy}-7-(5-methyl-1H-indazole-4 -Yl)-8-(2,2,2-trifluoroethoxy)-6-vinylquinazolin-4-yl]-2,7-diazaspiro[3.5]non-2-yl} Prop-2-en-1-one (hereinafter sometimes referred to as "compound C"), and (+)-1-(7-{8-ethoxy-7-(5-methyl-1H-indazol-4-yl)-2-[(1-methylpiperidin-4-yl)oxy Group]-6-vinylquinazolin-4-yl}-2,7-diazaspiro[3.5]non-2-yl)prop-2-en-1-one (hereinafter, sometimes described as " Compound D'') The formed group of compounds, or their pharmaceutically acceptable salts, and pharmaceutically acceptable excipients.

又,本發明係關於含有選自由化合物A、化合物B、化合物C及化合物D所成之群組之化合物,或其製藥學上容許之鹽的大腸癌之治療劑,作為一態樣係關於一種KRAS G12C突變陽性大腸癌之治療劑,其含有選自由化合物A、化合物B、化合物C及化合物D所成之群組之化合物,或其製藥學上容許之鹽。In addition, the present invention relates to a therapeutic agent for colorectal cancer containing a compound selected from the group consisting of compound A, compound B, compound C, and compound D, or a pharmaceutically acceptable salt thereof. A therapeutic agent for KRAS G12C mutation-positive colorectal cancer, which contains a compound selected from the group consisting of compound A, compound B, compound C, and compound D, or a pharmaceutically acceptable salt thereof.

又,本發明係關於一種用於製造大腸癌之治療用醫藥組成物的選自由化合物A、化合物B、化合物C及化合物D所成之群組之化合物,或其製藥學上容許之鹽之用途,作為某態樣係關於一種用於製造KRAS G12C突變陽性大腸癌治療用醫藥組成物的選自由化合物A、化合物B、化合物C及化合物D所成之群組之化合物,或其製藥學上容許之鹽之用途;一種用於治療大腸癌的選自由化合物A、化合物B、化合物C及化合物D所成之群組之化合物,或其製藥學上容許之鹽之用途,作為某態樣係關於一種用於治療KRAS G12C突變陽性大腸癌的選自由化合物A、化合物B、化合物C及化合物D所成之群組之化合物,或其製藥學上容許之鹽之用途;一種用於治療大腸癌之用途的選自由化合物A、化合物B、化合物C及化合物D所成之群組之化合物,或其製藥學上容許之鹽,作為某態樣係關於一種用於治療KRAS G12C突變陽性大腸癌之用途的選自由化合物A、化合物B、化合物C及化合物D所成之群組之化合物,或其製藥學上容許之鹽;及一種包含將選自由化合物A、化合物B、化合物C及化合物D所成之群組之化合物,或其製藥學上容許之鹽的有效量投予至對象而成之大腸癌之治療方法,作為某態樣係關於一種包含將選自由化合物A、化合物B、化合物C及化合物D所成之群組之化合物,或其製藥學上容許之鹽的有效量投予至對象而成之KRAS G12C突變陽性大腸癌之治療方法。此外,所謂「對象」,為需要該治療之人類或其他動物,作為某態樣,為需要該治療之人類。 [發明效果]Furthermore, the present invention relates to the use of a compound selected from the group consisting of compound A, compound B, compound C, and compound D, or a pharmaceutically acceptable salt thereof, for manufacturing a pharmaceutical composition for the treatment of colorectal cancer , As an aspect, it relates to a compound selected from the group consisting of compound A, compound B, compound C, and compound D for the manufacture of a pharmaceutical composition for the treatment of KRAS G12C mutation-positive colorectal cancer, or its pharmaceutically acceptable Use of a salt; a compound selected from the group consisting of compound A, compound B, compound C, and compound D, or a pharmaceutically acceptable salt thereof for the treatment of colorectal cancer, as a form of A compound selected from the group consisting of Compound A, Compound B, Compound C and Compound D, or a pharmaceutically acceptable salt thereof for the treatment of KRAS G12C mutation-positive colorectal cancer; A compound selected from the group consisting of compound A, compound B, compound C, and compound D, or a pharmaceutically acceptable salt thereof, as a form of use for treating KRAS G12C mutation-positive colorectal cancer A compound selected from the group consisting of compound A, compound B, compound C and compound D, or a pharmaceutically acceptable salt thereof; and one containing a compound selected from the group consisting of compound A, compound B, compound C and compound D The compound of the group, or a pharmaceutically acceptable salt thereof, is administered to the subject in an effective amount of colorectal cancer treatment method. As an aspect, it relates to a group consisting of compound A, compound B, compound C and A therapeutic method for KRAS G12C mutation-positive colorectal cancer in which an effective amount of a compound formed by Compound D, or a pharmaceutically acceptable salt thereof is administered to a subject. In addition, the so-called "subject" is a human or other animal in need of the treatment, as a certain form, a human in need of the treatment. [Effect of invention]

本發明之醫藥組成物之有效成分之選自由化合物A、化合物B、化合物C及化合物D所成之群組之化合物,或其製藥學上容許之鹽,具有G12C突變KRAS抑制作用,可使用作為大腸癌之治療用醫藥組成物,作為一態樣,可使用作為KRAS G12C突變陽性大腸癌之治療用醫藥組成物的有效成分。The active ingredient of the pharmaceutical composition of the present invention is a compound selected from the group consisting of compound A, compound B, compound C, and compound D, or a pharmaceutically acceptable salt thereof, has a G12C mutation KRAS inhibitory effect, and can be used as As an aspect, the pharmaceutical composition for the treatment of colorectal cancer can be used as the active ingredient of the pharmaceutical composition for the treatment of KRAS G12C mutation-positive colorectal cancer.

以下,詳細說明本發明。Hereinafter, the present invention will be described in detail.

所謂「G12C突變」,係表示在基因突變之中,對應於野生型蛋白質中之第12個的殘基由甘胺酸變換成半胱胺酸之突變。The so-called "G12C mutation" refers to a mutation in which a residue corresponding to the twelfth residue in the wild-type protein is changed from glycine to cysteine.

所謂「G12C突變KRAS」,係表示在編碼KRAS之基因中,具有上述「G12C突變」的KRAS。The so-called "G12C mutant KRAS" refers to the KRAS with the above-mentioned "G12C mutation" in the gene encoding KRAS.

本發明之某態樣表示於下。 (1-1)含有化合物A或其製藥學上容許之鹽,及製藥學上容許之賦形劑的大腸癌之治療用醫藥組成物。作為其他態樣,為含有化合物A或其製藥學上容許之鹽,及製藥學上容許之賦形劑的KRAS G12C突變陽性大腸癌治療用醫藥組成物。 (1-2)含有化合物A或其製藥學上容許之鹽的大腸癌之治療劑。作為其他態樣,為含有化合物A或其製藥學上容許之鹽的KRAS G12C突變陽性大腸癌之治療劑。 (1-3)用於製造大腸癌之治療用醫藥組成物的化合物A或其製藥學上容許之鹽之用途。作為其他態樣,為用於製造KRAS G12C突變陽性大腸癌治療用醫藥組成物的化合物A或其製藥學上容許之鹽之用途。 (1-4)用於治療大腸癌的化合物A或其製藥學上容許之鹽之用途。作為其他態樣,為用於治療KRAS G12C突變陽性大腸癌的化合物A或其製藥學上容許之鹽之用途。 (1-5)用於治療大腸癌之用途的化合物A或其製藥學上容許之鹽。作為其他態樣,用於治療KRAS G12C突變陽性大腸癌之用途的化合物A或其製藥學上容許之鹽。 (1-6)含有將化合物A或其製藥學上容許之鹽的有效量投予至對象而成之大腸癌之治療方法。作為其他態樣,為含有將化合物A或其製藥學上容許之鹽的有效量投予至對象而成之KRAS G12C突變陽性大腸癌之治療方法。An aspect of the present invention is shown below. (1-1) A pharmaceutical composition for the treatment of colorectal cancer containing Compound A or a pharmaceutically acceptable salt thereof and pharmaceutically acceptable excipients. As another aspect, it is a pharmaceutical composition for the treatment of KRAS G12C mutation-positive colorectal cancer containing Compound A or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient. (1-2) A therapeutic agent for colorectal cancer containing Compound A or a pharmaceutically acceptable salt thereof. As another aspect, it is a therapeutic agent for KRAS G12C mutation-positive colorectal cancer containing Compound A or a pharmaceutically acceptable salt thereof. (1-3) Use of Compound A or a pharmaceutically acceptable salt thereof for the manufacture of a pharmaceutical composition for the treatment of colorectal cancer. As another aspect, it is the use of Compound A or a pharmaceutically acceptable salt thereof for the manufacture of a pharmaceutical composition for the treatment of KRAS G12C mutation-positive colorectal cancer. (1-4) Use of Compound A or a pharmaceutically acceptable salt thereof for the treatment of colorectal cancer. As another aspect, it is the use of Compound A or a pharmaceutically acceptable salt thereof for the treatment of KRAS G12C mutation-positive colorectal cancer. (1-5) Compound A or a pharmaceutically acceptable salt thereof for the treatment of colorectal cancer. As another aspect, Compound A or a pharmaceutically acceptable salt thereof for the treatment of KRAS G12C mutation-positive colorectal cancer. (1-6) A method for treating colorectal cancer containing an effective amount of Compound A or a pharmaceutically acceptable salt thereof administered to a subject. As another aspect, it is a therapeutic method containing KRAS G12C mutation-positive colorectal cancer in which an effective amount of Compound A or a pharmaceutically acceptable salt thereof is administered to a subject.

(2-1)含有化合物B或其製藥學上容許之鹽,及製藥學上容許之賦形劑的大腸癌之治療用醫藥組成物。作為其他態樣,為含有化合物B或其製藥學上容許之鹽,及製藥學上容許之賦形劑的KRAS G12C突變陽性大腸癌治療用醫藥組成物。 (2-2)含有化合物B或其製藥學上容許之鹽的大腸癌之治療劑。作為其他態樣,為含有化合物B或其製藥學上容許之鹽的KRAS G12C突變陽性大腸癌之治療劑。 (2-3)用於製造大腸癌之治療用醫藥組成物的化合物B或其製藥學上容許之鹽之用途。作為其他態樣,為用於製造KRAS G12C突變陽性大腸癌治療用醫藥組成物的化合物B或其製藥學上容許之鹽之用途。 (2-4)用於治療大腸癌的化合物B或其製藥學上容許之鹽之用途。作為其他態樣,為用於治療KRAS G12C突變陽性大腸癌的化合物B或其製藥學上容許之鹽之用途。 (2-5)用於治療大腸癌之用途的化合物B或其製藥學上容許之鹽。作為其他態樣,為用於治療KRAS G12C突變陽性大腸癌之用途的化合物B或其製藥學上容許之鹽。 (2-6)含有將化合物B或其製藥學上容許之鹽的有效量投予至對象而成之大腸癌之治療方法。作為其他態樣,為含有將化合物B或其製藥學上容許之鹽的有效量投予至對象而成之KRAS G12C突變陽性大腸癌之治療方法。(2-1) A pharmaceutical composition for the treatment of colorectal cancer containing Compound B or a pharmaceutically acceptable salt thereof and pharmaceutically acceptable excipients. As another aspect, it is a pharmaceutical composition for the treatment of KRAS G12C mutation-positive colorectal cancer containing Compound B or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient. (2-2) A therapeutic agent for colorectal cancer containing Compound B or a pharmaceutically acceptable salt thereof. As another aspect, it is a therapeutic agent for KRAS G12C mutation-positive colorectal cancer containing Compound B or a pharmaceutically acceptable salt thereof. (2-3) Use of Compound B or a pharmaceutically acceptable salt thereof for the manufacture of a pharmaceutical composition for the treatment of colorectal cancer. As another aspect, it is the use of Compound B or a pharmaceutically acceptable salt thereof for the manufacture of a pharmaceutical composition for the treatment of KRAS G12C mutation-positive colorectal cancer. (2-4) Use of Compound B or its pharmaceutically acceptable salts for the treatment of colorectal cancer. As another aspect, it is a compound B or a pharmaceutically acceptable salt thereof for treating KRAS G12C mutation-positive colorectal cancer. (2-5) Compound B or a pharmaceutically acceptable salt thereof for the treatment of colorectal cancer. As another aspect, Compound B or a pharmaceutically acceptable salt thereof is used for the treatment of KRAS G12C mutation-positive colorectal cancer. (2-6) A treatment method for colorectal cancer containing an effective amount of Compound B or a pharmaceutically acceptable salt thereof administered to a subject. As another aspect, it is a therapeutic method containing KRAS G12C mutation-positive colorectal cancer in which an effective amount of Compound B or a pharmaceutically acceptable salt thereof is administered to a subject.

(3-1)含有化合物C或其製藥學上容許之鹽,及製藥學上容許之賦形劑的大腸癌之治療用醫藥組成物。作為其他態樣,為含有化合物C或其製藥學上容許之鹽,及製藥學上容許之賦形劑的KRAS G12C突變陽性大腸癌治療用醫藥組成物。 (3-2)含有化合物C或其製藥學上容許之鹽的大腸癌之治療劑。作為其他態樣,為含有化合物C或其製藥學上容許之鹽的KRAS G12C突變陽性大腸癌之治療劑。 (3-3)用於製造大腸癌之治療用醫藥組成物的化合物C或其製藥學上容許之鹽之用途。作為其他態樣,為用於製造KRAS G12C突變陽性大腸癌治療用醫藥組成物的化合物C或其製藥學上容許之鹽之用途。 (3-4)用於治療大腸癌的化合物C或其製藥學上容許之鹽之用途。作為其他態樣,為用於治療KRAS G12C突變陽性大腸癌的化合物C或其製藥學上容許之鹽之用途。 (3-5)用於治療大腸癌之用途的化合物C或其製藥學上容許之鹽。作為其他態樣,為用於治療KRAS G12C突變陽性大腸癌之用途的化合物C或其製藥學上容許之鹽。 (3-6)含有將化合物C或其製藥學上容許之鹽的有效量投予至對象而成之大腸癌之治療方法。作為其他態樣,含有將化合物C或其製藥學上容許之鹽的有效量投予至對象而成之KRAS G12C突變陽性大腸癌之治療方法。(3-1) A pharmaceutical composition for the treatment of colorectal cancer containing Compound C or a pharmaceutically acceptable salt thereof and pharmaceutically acceptable excipients. As another aspect, it is a pharmaceutical composition for the treatment of KRAS G12C mutation-positive colorectal cancer containing Compound C or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient. (3-2) A therapeutic agent for colorectal cancer containing Compound C or a pharmaceutically acceptable salt thereof. As another aspect, it is a therapeutic agent for KRAS G12C mutation-positive colorectal cancer containing Compound C or a pharmaceutically acceptable salt thereof. (3-3) Use of Compound C or a pharmaceutically acceptable salt thereof for the manufacture of a pharmaceutical composition for treatment of colorectal cancer. As another aspect, it is the use of compound C or a pharmaceutically acceptable salt thereof for the manufacture of a pharmaceutical composition for the treatment of KRAS G12C mutation-positive colorectal cancer. (3-4) Use of Compound C or its pharmaceutically acceptable salts for the treatment of colorectal cancer. As another aspect, it is a compound C or a pharmaceutically acceptable salt thereof for treating KRAS G12C mutation-positive colorectal cancer. (3-5) Compound C or its pharmaceutically acceptable salt for the treatment of colorectal cancer. As another aspect, Compound C or a pharmaceutically acceptable salt thereof is used for the treatment of KRAS G12C mutation-positive colorectal cancer. (3-6) A method for treating colorectal cancer containing an effective amount of Compound C or a pharmaceutically acceptable salt thereof administered to a subject. As another aspect, it contains a therapeutic method of KRAS G12C mutation-positive colorectal cancer in which an effective amount of Compound C or a pharmaceutically acceptable salt thereof is administered to a subject.

(4-1)含有化合物D或其製藥學上容許之鹽,及製藥學上容許之賦形劑的大腸癌之治療用醫藥組成物。作為其他態樣,含有化合物D或其製藥學上容許之鹽,及製藥學上容許之賦形劑的KRAS G12C突變陽性大腸癌治療用醫藥組成物。 (4-2)含有化合物D或其製藥學上容許之鹽的大腸癌之治療劑。作為其他態樣,含有化合物D或其製藥學上容許之鹽的KRAS G12C突變陽性大腸癌之治療劑。 (4-3)用於製造大腸癌之治療用醫藥組成物的化合物D或其製藥學上容許之鹽之用途。作為其他態樣,用於製造KRAS G12C突變陽性大腸癌治療用醫藥組成物的化合物D或其製藥學上容許之鹽之用途。 (4-4)用於治療大腸癌的化合物D或其製藥學上容許之鹽之用途。作為其他態樣,用於治療KRAS G12C突變陽性大腸癌的化合物D或其製藥學上容許之鹽之用途。 (4-5)用於治療大腸癌之用途的化合物D或其製藥學上容許之鹽。作為其他態樣,用於治療KRAS G12C突變陽性大腸癌之用途的化合物D或其製藥學上容許之鹽。 (4-6)含有將化合物D或其製藥學上容許之鹽的有效量投予至對象而成之大腸癌之治療方法。作為其他態樣,含有將化合物D或其製藥學上容許之鹽的有效量投予至對象而成之KRAS G12C突變陽性大腸癌之治療方法。(4-1) A pharmaceutical composition for the treatment of colorectal cancer containing Compound D or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable excipient. As another aspect, a pharmaceutical composition for the treatment of KRAS G12C mutation-positive colorectal cancer containing Compound D or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient. (4-2) A therapeutic agent for colorectal cancer containing Compound D or a pharmaceutically acceptable salt thereof. As another aspect, a therapeutic agent for KRAS G12C mutation-positive colorectal cancer containing Compound D or a pharmaceutically acceptable salt thereof. (4-3) Use of Compound D or a pharmaceutically acceptable salt thereof for the manufacture of a pharmaceutical composition for the treatment of colorectal cancer. As another aspect, it is used for the manufacture of Compound D or a pharmaceutically acceptable salt thereof for the pharmaceutical composition for the treatment of KRAS G12C mutation-positive colorectal cancer. (4-4) The use of Compound D or its pharmaceutically acceptable salts for the treatment of colorectal cancer. As another aspect, the use of Compound D or a pharmaceutically acceptable salt thereof for the treatment of KRAS G12C mutation-positive colorectal cancer. (4-5) Compound D or its pharmaceutically acceptable salt for the treatment of colorectal cancer. As another aspect, Compound D or a pharmaceutically acceptable salt thereof for the treatment of KRAS G12C mutation-positive colorectal cancer. (4-6) A treatment method for colorectal cancer containing an effective amount of Compound D or a pharmaceutically acceptable salt thereof administered to a subject. As another aspect, it contains a therapeutic method of KRAS G12C mutation-positive colorectal cancer in which an effective amount of Compound D or a pharmaceutically acceptable salt thereof is administered to a subject.

本說明書中,所謂「選自由化合物A、化合物B、化合物C及化合物D所成之群組之化合物的製藥學上容許之鹽」,係指化合物A、化合物B、化合物C或化合物D之酸加成鹽的意思,具體而言,可舉例與鹽酸、氫溴酸、碘化氫酸、硫酸、硝酸、磷酸等之無機酸,或甲酸、乙酸、丙酸、草酸、丙二酸、琥珀酸、富馬酸、馬來酸、乳酸、蘋果酸、苦杏仁酸、酒石酸、二苯甲醯基酒石酸、二甲苯甲醯基酒石酸、檸檬酸、甲磺酸(Mesylic acid)、乙磺酸、苯磺酸、p-甲苯磺酸(對甲苯磺酸)、天冬胺酸、麩胺酸等之有機酸的酸加成鹽。 此外,「選自由化合物A、化合物B、化合物C及化合物D所成之群組之化合物」中,包含化合物A、化合物B、化合物C或化合物D之各種溶劑合物,具體而言,包含水合物或乙醇合物。進而,「製藥學上容許之鹽」中亦包含此等之溶劑合物的酸加成鹽。 又,作為「選自由化合物A、化合物B、化合物C及化合物D所成之群組之化合物,或其製藥學上容許之鹽」之某態樣,可舉例未形成鹽之自由鹼,即,化合物A、化合物B、化合物C或化合物D,作為其他態樣,為化合物A,進而作為其他態樣,為化合物B,進而作為其他態樣,為化合物C,又進而作為其他態樣,為化合物D。In this specification, "a pharmaceutically acceptable salt of a compound selected from the group consisting of compound A, compound B, compound C, and compound D" refers to the acid of compound A, compound B, compound C, or compound D The meaning of the addition salt can be specifically exemplified by inorganic acids such as hydrochloric acid, hydrobromic acid, hydrogen iodide acid, sulfuric acid, nitric acid, and phosphoric acid, or formic acid, acetic acid, propionic acid, oxalic acid, malonic acid, and succinic acid , Fumaric acid, maleic acid, lactic acid, malic acid, amygdalic acid, tartaric acid, dibenzoyl tartaric acid, xylene methyl tartaric acid, citric acid, mesylic acid, ethanesulfonic acid, benzene Acid addition salts of organic acids such as sulfonic acid, p-toluenesulfonic acid (p-toluenesulfonic acid), aspartic acid, and glutamic acid. In addition, "a compound selected from the group consisting of compound A, compound B, compound C, and compound D" includes various solvates of compound A, compound B, compound C, or compound D, specifically, including hydration Or ethanolate. Furthermore, "additionally acceptable salts of pharmacy" also include acid addition salts of these solvates. In addition, as a form of "a compound selected from the group consisting of compound A, compound B, compound C, and compound D, or a pharmaceutically acceptable salt thereof", a free base that does not form a salt can be exemplified, that is, Compound A, Compound B, Compound C, or Compound D, as other aspects, are Compound A, further as other aspects, as Compound B, further as other aspects, as Compound C, and further as other aspects, as Compounds D.

含有選自由化合物A、化合物B、化合物C及化合物D所成之群組之化合物,或其製藥學上容許之鹽作為有效成分的醫藥組成物,可使用該領域中常用之賦形劑,即藥劑用賦形劑或藥劑用載體等,藉由一般常用方法來調製。 投予可為錠劑、丸劑、膠囊劑、顆粒劑、散劑、液劑等之口服投予,或,關節內、靜脈內、肌肉內等之注射劑,栓劑、點眼劑、眼軟膏、經皮用液劑、軟膏劑、經皮用貼附劑、經黏膜液劑、經黏膜貼附劑、吸入劑等之非口服投予之任一形態。A pharmaceutical composition containing a compound selected from the group consisting of compound A, compound B, compound C, and compound D, or a pharmaceutically acceptable salt thereof as an active ingredient can use excipients commonly used in the field, namely The excipient for pharmaceuticals, the carrier for pharmaceuticals, etc. are prepared by a commonly used method. The administration may be oral administration of tablets, pills, capsules, granules, powders, liquids, etc., or intra-articular, intravenous, intramuscular injections, suppositories, eye drops, eye ointment, transdermal, etc. Any form of non-oral administration with liquid, ointment, transdermal patch, transmucosal liquid, transmucosal patch, inhalation, etc.

作為用以口服投予的固體組成物,使用錠劑、散劑、顆粒劑等。如此之固體組成物中,將1種或2種以上之有效成分,與至少1種的惰性賦形劑混合。組成物,亦可依循常規方法含有惰性添加劑,例如潤滑劑或崩解劑、穩定化劑、溶解輔助劑。錠劑或丸劑視需要亦可以糖衣或胃溶性或腸溶性物質的薄膜進行被膜。 用於口服投予的液體組成物,包含藥劑上容許之乳濁劑、溶液劑、懸濁劑、糖漿劑或酏劑等,包含一般使用的惰性稀釋劑,例如純化水或乙醇。該液體組成物在惰性稀釋劑以外亦可含有如助溶劑、濕潤劑、懸濁劑之輔助劑,甜味劑、風味劑、芳香劑、防腐劑。As a solid composition for oral administration, lozenges, powders, granules and the like are used. In such a solid composition, one or more active ingredients are mixed with at least one inert excipient. The composition may also contain inert additives such as lubricants or disintegrants, stabilizers, and dissolution aids in accordance with conventional methods. Lozenges or pills can be coated with sugar coating or a film of stomach-soluble or enteric-substance, if necessary. Liquid compositions for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, or elixirs, and generally used inert diluents, such as purified water or ethanol. The liquid composition may contain auxiliary agents such as co-solvents, wetting agents, suspending agents, sweeteners, flavoring agents, fragrances, preservatives in addition to the inert diluent.

用於非口服投予的注射劑,含有無菌之水性或非水性的溶液劑、懸濁劑或乳濁劑。作為水性之溶劑,包含例如注射用蒸餾水或生理食鹽液。作為非水性之溶劑,例如有如乙醇之醇類。如此之組成物,亦可進一步包含等張化劑、防腐劑、濕潤劑、乳化劑、分散劑、穩定化劑,或助溶劑。此等藉由例如通過細菌保留過濾器進行過濾,殺菌劑的摻合或照射而無菌化。又,此等可製造無菌之固體組成物,使用前溶解或懸濁於無菌水或無菌之注射用溶劑中來使用。Injections for non-oral administration contain sterile aqueous or non-aqueous solutions, suspensions or emulsions. Examples of the aqueous solvent include distilled water for injection or physiological saline solution. Examples of non-aqueous solvents include alcohols such as ethanol. Such a composition may further contain isotonic agents, preservatives, wetting agents, emulsifiers, dispersants, stabilizers, or co-solvents. These are sterilized by, for example, filtering through a bacteria-retaining filter, blending or irradiation of a bactericide. In addition, these can produce sterile solid compositions, which are dissolved or suspended in sterile water or sterile injectable solvents before use.

一般口服投予之情形中,1日之投予量為約0.001~100mg/kg體重,較佳為0.1~30mg/kg,更佳為0.1~10mg/kg為適當,將此1次或分成2次~4次投予。靜脈內投予之情形中,1日之投予量為約0.0001~10mg/kg體重為適當,1日1次~分成複數次投予。In the case of general oral administration, the daily dose is about 0.001~100mg/kg body weight, preferably 0.1~30mg/kg, more preferably 0.1~10mg/kg is appropriate, this one or divided into 2 From 4 to 4 times. In the case of intravenous administration, the amount of administration per day is about 0.0001 to 10 mg/kg body weight, and it is once per day to be divided into multiple administrations.

雖依投予路徑、劑形、投予部位、賦形劑或添加劑的種類而不同,但本發明之醫藥組成物含有0.01~100重量%,作為某態樣為0.01~50重量%之有效成分的選自由化合物A、化合物B、化合物C及化合物D所成之群組之化合物,或其製藥學上容許之鹽。Although it differs depending on the administration route, dosage form, administration site, excipient or additive, the pharmaceutical composition of the present invention contains 0.01 to 100% by weight, as an active ingredient of 0.01 to 50% by weight Compound selected from the group consisting of compound A, compound B, compound C and compound D, or a pharmaceutically acceptable salt thereof.

本發明之醫藥組成物,可與認為在大腸癌,特別是KRAS G12C突變陽性大腸癌顯示有效性的各種治療劑併用。該併用,可同時投予,或可個別連續地,或在期望之時間間隔進行投予。同時投予之情形中,可為配合劑亦可個別製劑化。The pharmaceutical composition of the present invention can be used in combination with various therapeutic agents believed to show effectiveness in colorectal cancer, especially KRAS G12C mutation-positive colorectal cancer. The combination can be administered at the same time, or it can be administered individually continuously, or at a desired time interval. In the case of simultaneous administration, it may be a compounding agent or may be formulated separately.

以下以化合物A、化合物B、化合物C及化合物D之製造法作為參考例詳細地進行說明。又,該等原料化合物之製法表示於製造例。又,化合物A、化合物B、化合物C及化合物D之製造法,並不限定於僅以下所示之具體參考例之製造法,亦可藉由此等製造法的其他組合,或是該業者不言自明的方法來製造。Hereinafter, the production methods of compound A, compound B, compound C, and compound D will be described in detail as reference examples. In addition, the manufacturing method of these raw material compounds is shown in a manufacturing example. In addition, the production methods of compound A, compound B, compound C and compound D are not limited to the production methods of the specific reference examples shown below, and other combinations of these production methods may be used, or the industry is self-explanatory Method.

本說明書中,化合物之命名有時使用ACD/Name(註冊商標、Advanced Chemistry Development, Inc.)等之命名軟體。In this specification, naming software such as ACD/Name (registered trademark, Advanced Chemistry Development, Inc.) is sometimes used for naming compounds.

又,為了方便,濃度mol/L以M表示。例如,1M氫氧化鈉水溶液係指1mol/L之氫氧化鈉水溶液的意思。Also, for convenience, the concentration mol/L is represented by M. For example, 1M sodium hydroxide aqueous solution means 1mol/L sodium hydroxide aqueous solution.

製造例1 將2-胺基-4-溴-3-氟苯甲酸(4.0g)、N-碘琥珀酸醯亞胺(4.0g)、N,N-二甲基甲醯胺(以下,簡寫為DMF)(40mL)之混合物,在氬氣流下,於50℃攪拌2小時。於反應混合物中,於50℃加入N-碘琥珀酸醯亞胺(1.5g),同溫度下攪拌1.5小時。於反應混合物中,於50℃加入N-碘琥珀酸醯亞胺(1.5g),同溫度下攪拌一整夜。將反應混合物放置冷卻至室溫後,加入水,於室溫攪拌5小時。過濾取出析出之固體,於室溫通風乾燥。將所得之固體懸濁於水,於室溫攪拌1小時。過濾取出固體,以水洗淨後,減壓下,於50℃進行乾燥,得到固體之2-胺基-4-溴-3-氟-5-碘苯甲酸(5.6g)。Manufacturing Example 1 2-Amino-4-bromo-3-fluorobenzoic acid (4.0g), N-iodosuccinate amide imide (4.0g), N,N-dimethylformamide (hereinafter abbreviated as DMF) (40 mL) of the mixture was stirred at 50°C for 2 hours under a stream of argon. To the reaction mixture, N-iodosuccinic acid imide (1.5 g) was added at 50°C, and stirred at the same temperature for 1.5 hours. To the reaction mixture, N-iodosuccinate amide imine (1.5 g) was added at 50°C, and stirred at the same temperature overnight. After the reaction mixture was allowed to cool to room temperature, water was added and stirred at room temperature for 5 hours. The precipitated solid was taken out by filtration and dried at room temperature with ventilation. The obtained solid was suspended in water and stirred at room temperature for 1 hour. The solid was taken out by filtration, washed with water, and dried under reduced pressure at 50° C. to obtain solid 2-amino-4-bromo-3-fluoro-5-iodobenzoic acid (5.6 g).

製造例2 將2-胺基-4-溴-3-氟-5-碘苯甲酸(5.6g)、尿素(4.7g)之混合物,於200℃攪拌3小時。將反應混合物放置冷卻至室溫後,加入水,於室溫攪拌15分鐘。過濾取出固體,以水洗淨後,減壓下,於50℃進行乾燥。將所得之固體磨碎成粉末後,與磷醯氯(80mL)混合進行冰冷,在氮氣流下,滴加N,N-二異丙基乙胺(以下,簡寫成DIPEA)(8.0mL)。將反應混合物於150℃攪拌2.5小時。將反應混合物放置冷卻至室溫後,減壓下,進行濃縮,於所得之殘渣中加入乙酸乙酯。將反應混合物注入至冰水後,濾掉不溶物,以乙酸乙酯萃取濾液。將有機層以飽和氯化鈉水溶液洗淨後,以無水硫酸鎂進行乾燥,減壓下,濃縮溶液。以二氧化矽凝膠管柱色層分析(己烷/乙酸乙酯)純化所得之殘渣,得到固體之7-溴-2,4-二氯-8-氟-6-碘喹唑啉(3.6g)。Manufacturing Example 2 A mixture of 2-amino-4-bromo-3-fluoro-5-iodobenzoic acid (5.6 g) and urea (4.7 g) was stirred at 200°C for 3 hours. After the reaction mixture was left to cool to room temperature, water was added and stirred at room temperature for 15 minutes. The solid was taken out by filtration, washed with water, and then dried under reduced pressure at 50°C. After grinding the obtained solid into a powder, it was mixed with phosphazene chloride (80 mL) and ice-cooled. Under a nitrogen stream, N,N-diisopropylethylamine (hereinafter, abbreviated as DIPEA) (8.0 mL) was added dropwise. The reaction mixture was stirred at 150°C for 2.5 hours. After the reaction mixture was left to cool to room temperature, it was concentrated under reduced pressure, and ethyl acetate was added to the resulting residue. After the reaction mixture was poured into ice water, insoluble materials were filtered off, and the filtrate was extracted with ethyl acetate. After the organic layer was washed with a saturated sodium chloride aqueous solution, it was dried over anhydrous magnesium sulfate, and the solution was concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (hexane/ethyl acetate) to obtain a solid 7-bromo-2,4-dichloro-8-fluoro-6-iodoquinazoline (3.6 g).

製造例3 將7-溴-2,4-二氯-8-氟-6-碘喹唑啉(3.6g)、1,4-二

Figure 108126915-A0304-12-01
烷(35mL)之混合物進行冰冷後,在氮氣流下,加入DIPEA(8.0mL)、2,7-二氮雜螺[3.5]壬烷-2-羧酸三級丁酯(2.0g),於室溫攪拌1.5小時。於反應混合物中加入水,以氯仿進行萃取。將有機層以無水硫酸鎂進行乾燥後,減壓下,濃縮溶液。以二氧化矽凝膠管柱色層分析(氯仿/乙酸乙酯)純化所得之殘渣,得到固體之7-(7-溴-2-氯-8-氟-6-碘喹唑啉-4-基)-2,7-二氮雜螺[3.5]壬烷-2-羧酸三級丁酯(4.6g)。Production Example 3 7-Bromo-2,4-dichloro-8-fluoro-6-iodoquinazoline (3.6 g), 1,4-bis
Figure 108126915-A0304-12-01
After the mixture of alkane (35mL) was ice-cooled, DIPEA (8.0mL) and 2,7-diazaspiro[3.5]nonane-2-carboxylic acid tertiary butyl ester (2.0g) were added under a nitrogen stream. Warm for 1.5 hours. Water was added to the reaction mixture, and extraction was performed with chloroform. After the organic layer was dried over anhydrous magnesium sulfate, the solution was concentrated under reduced pressure. The residue obtained was purified by silica gel column chromatography (chloroform/ethyl acetate) to obtain solid 7-(7-bromo-2-chloro-8-fluoro-6-iodoquinazoline-4- Radical)-2,7-diazaspiro[3.5]nonane-2-carboxylic acid tertiary butyl ester (4.6 g).

製造例4 將7-(7-溴-2-氯-8-氟-6-碘喹唑啉-4-基)-2,7-二氮雜螺[3.5]壬烷-2-羧酸三級丁酯(9.5g)、1-(3-甲氧基丙基)哌啶-4-醇(6.8g)、碳酸銫(15g)、1,4-二吖雙環[2.2.2]辛烷(以下,簡寫成DABCO)(260mg)、DMF(76mL)、四氫呋喃(以下,簡寫成THF)(76mL)之混合物,在氬氛圍下,於室溫攪拌一整夜。於反應混合物中加入水,以乙酸乙酯進行萃取。將有機層以飽和氯化鈉水溶液洗淨後,以無水硫酸鎂進行乾燥。減壓下,濃縮溶液後,於所得之固體中加入二異丙基醚,於室溫攪拌。過濾取出固體,減壓下,於50℃進行乾燥,得到固體之7-(7-溴-8-氟-6-碘-2-{[1-(3-甲氧基丙基)哌啶-4-基]氧基}喹唑啉-4-基)-2,7-二氮雜螺[3.5]壬烷-2-羧酸三級丁酯(9.1g)。Manufacturing Example 4 7-(7-Bromo-2-chloro-8-fluoro-6-iodoquinazolin-4-yl)-2,7-diazaspiro[3.5]nonane-2-carboxylic acid tertiary butyl ester (9.5g), 1-(3-methoxypropyl) piperidin-4-ol (6.8g), cesium carbonate (15g), 1,4-diazepine [2.2.2]octane (hereinafter, Abbreviated as a mixture of DABCO) (260 mg), DMF (76 mL), and tetrahydrofuran (hereinafter abbreviated as THF) (76 mL), and stirred at room temperature overnight under an argon atmosphere. Water was added to the reaction mixture, and extraction was performed with ethyl acetate. After the organic layer was washed with a saturated sodium chloride aqueous solution, it was dried over anhydrous magnesium sulfate. After concentrating the solution under reduced pressure, diisopropyl ether was added to the resulting solid and stirred at room temperature. The solid was taken out by filtration, and dried at 50°C under reduced pressure to obtain 7-(7-bromo-8-fluoro-6-iodo-2-{[1-(3-methoxypropyl)piperidine- 4-yl]oxy}quinazolin-4-yl)-2,7-diazaspiro[3.5]nonane-2-carboxylic acid tertiary butyl ester (9.1 g).

製造例5 將7-(7-溴-8-氟-6-碘-2-{[1-(3-甲氧基丙基)哌啶-4-基]氧基}喹唑啉-4-基)-2,7-二氮雜螺[3.5]壬烷-2-羧酸三級丁酯(5.0g)、2,2,2-三氟乙醇(1.4mL)、碳酸銫(6.5g)、DMF(50mL)之混合物,於室溫攪拌5小時。將反應混合物,於50℃攪拌1.5小時。將反應混合物放置冷卻至室溫後,加入水,以乙酸乙酯進行萃取。將有機層以水、飽和氯化鈉水溶液洗淨後,以無水硫酸鎂進行乾燥。減壓下,濃縮溶液後,以二氧化矽凝膠管柱色層分析(鹼性二氧化矽凝膠、己烷/乙酸乙酯)純化所得之殘渣,得到7-[7-溴-6-碘-2-{[1-(3-甲氧基丙基)哌啶-4-基]氧基}-8-(2,2,2-三氟乙氧基)喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬烷-2-羧酸三級丁酯(5.5g)。Manufacturing Example 5 7-(7-Bromo-8-fluoro-6-iodo-2-{[1-(3-methoxypropyl)piperidin-4-yl]oxy}quinazolin-4-yl)- 2,7-diazaspiro[3.5]nonane-2-carboxylic acid tertiary butyl ester (5.0g), 2,2,2-trifluoroethanol (1.4mL), cesium carbonate (6.5g), DMF ( 50 mL) of the mixture was stirred at room temperature for 5 hours. The reaction mixture was stirred at 50°C for 1.5 hours. After the reaction mixture was left to cool to room temperature, water was added, and extraction was performed with ethyl acetate. After washing the organic layer with water and a saturated sodium chloride aqueous solution, it was dried over anhydrous magnesium sulfate. After concentrating the solution under reduced pressure, the resulting residue was purified by silica gel column chromatography (alkaline silica gel, hexane/ethyl acetate) to obtain 7-[7-bromo-6- Iodo-2-{[1-(3-methoxypropyl)piperidin-4-yl]oxy}-8-(2,2,2-trifluoroethoxy)quinazolin-4-yl ]-2,7-diazaspiro[3.5]nonane-2-carboxylic acid tertiary butyl ester (5.5g).

製造例6 將7-[7-溴-6-碘-2-{[1-(3-甲氧基丙基)哌啶-4-基]氧基}-8-(2,2,2-三氟乙氧基)喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬烷-2-羧酸三級丁酯(2.7g)、環丙基硼酸(560mg)、磷酸三鉀(2.5g)、[1,1'-雙(二苯基膦基)二茂鐵]鈀(II)二氯化物 二氯甲烷加成物(以下,簡寫成PdCl2 (dppf)・CH2 Cl2 )(270mg)、1,4-二

Figure 108126915-A0304-12-01
烷(20mL)、乙腈(20mL)、水(8.6mL)之混合物,在氬氛圍下,於100℃攪拌4小時。將反應混合物放置冷卻至室溫後,加入飽和氯化鈉水溶液,以乙酸乙酯進行萃取。將有機層以無水硫酸鎂進行乾燥後,減壓下,濃縮溶液。以二氧化矽凝膠管柱色層分析(鹼性二氧化矽凝膠、己烷/乙酸乙酯)純化所得之殘渣,得到7-[7-溴-6-環丙基-2-{[1-(3-甲氧基丙基)哌啶-4-基]氧基}-8-(2,2,2-三氟乙氧基)喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬烷-2-羧酸三級丁酯(1.9g)。Production Example 6 7-[7-Bromo-6-iodo-2-{[1-(3-methoxypropyl)piperidin-4-yl]oxy}-8-(2,2,2- Trifluoroethoxy)quinazolin-4-yl]-2,7-diazaspiro[3.5]nonane-2-carboxylic acid tertiary butyl ester (2.7g), cyclopropylboronic acid (560mg), Tripotassium phosphate (2.5g), [1,1'-bis(diphenylphosphino)ferrocene] palladium (II) dichloride dichloromethane adduct (hereinafter, abbreviated as PdCl 2 (dppf) ・CH 2 Cl 2 ) (270mg), 1,4-di
Figure 108126915-A0304-12-01
A mixture of alkane (20 mL), acetonitrile (20 mL) and water (8.6 mL) was stirred at 100° C. for 4 hours under argon atmosphere. After the reaction mixture was left to cool to room temperature, a saturated aqueous sodium chloride solution was added, and extraction was performed with ethyl acetate. After the organic layer was dried over anhydrous magnesium sulfate, the solution was concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (alkaline silica gel, hexane/ethyl acetate) to obtain 7-[7-bromo-6-cyclopropyl-2-{[ 1-(3-methoxypropyl)piperidin-4-yl)oxy)-8-(2,2,2-trifluoroethoxy)quinazolin-4-yl)-2,7- Diazaspiro[3.5]nonane-2-carboxylic acid tertiary butyl ester (1.9 g).

製造例7 在氬氛圍下,混合7-[7-溴-6-環丙基-2-{[1-(3-甲氧基丙基)哌啶-4-基]氧基}-8-(2,2,2-三氟乙氧基)喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬烷-2-羧酸三級丁酯(1.9g)、(5-甲基-1H-吲唑-4-基)硼酸(710mg)、(1E,4E)-1,5-二苯基五-1,4-二烯-3-酮/鈀(3:2)(以下,簡寫成Pd2 (dba)3 )(230mg)、二環己基(2',6'-二甲氧基聯苯-2-基)膦(以下,簡寫成SPhos)(210mg)、磷酸三鉀(2.2g)、1,4-二

Figure 108126915-A0304-12-01
烷(15mL)、水(2.8mL),在微波照射下,於120℃攪拌70分鐘。將反應混合物在減壓下,進行濃縮,以二氧化矽凝膠管柱色層分析(鹼性二氧化矽凝膠、己烷/乙酸乙酯)純化所得之殘渣。於所得之純化物中加入乙腈,於室溫攪拌30分鐘。過濾取出析出之固體,得到固體之7-[6-環丙基-2-{[1-(3-甲氧基丙基)哌啶-4-基]氧基}-7-(5-甲基-1H-吲唑-4-基)-8-(2,2,2-三氟乙氧基)喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬烷-2-羧酸三級丁酯(630mg)。Production Example 7 Under argon atmosphere, 7-[7-bromo-6-cyclopropyl-2-{[1-(3-methoxypropyl)piperidin-4-yl]oxy}-8- (2,2,2-trifluoroethoxy)quinazolin-4-yl]-2,7-diazaspiro[3.5]nonane-2-carboxylic acid tertiary butyl ester (1.9g), ( 5-methyl-1H-indazol-4-yl)boronic acid (710mg), (1E,4E)-1,5-diphenylpenta-1,4-dien-3-one/palladium (3:2 ) (Hereinafter abbreviated as Pd 2 (dba) 3 ) (230mg), dicyclohexyl (2', 6'-dimethoxybiphenyl-2-yl) phosphine (hereinafter abbreviated as SPhos) (210mg), Tripotassium phosphate (2.2g), 1,4-di
Figure 108126915-A0304-12-01
Alkane (15 mL) and water (2.8 mL) were stirred at 120°C for 70 minutes under microwave irradiation. The reaction mixture was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography (basic silica gel, hexane/ethyl acetate). Acetonitrile was added to the resulting purified product, and stirred at room temperature for 30 minutes. The precipitated solid was removed by filtration to obtain 7-[6-cyclopropyl-2-{[1-(3-methoxypropyl)piperidin-4-yl]oxy}-7-(5-methyl Yl-1H-indazol-4-yl)-8-(2,2,2-trifluoroethoxy)quinazolin-4-yl]-2,7-diazaspiro[3.5]nonane- Tertiary butyl 2-carboxylate (630 mg).

製造例8 將7-[6-環丙基-2-{[1-(3-甲氧基丙基)哌啶-4-基]氧基}-7-(5-甲基-1H-吲唑-4-基)-8-(2,2,2-三氟乙氧基)喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬烷-2-羧酸三級丁酯(630mg)、1M鹽酸(5.5mL)之混合物,在氬氛圍下,於50℃攪拌1小時。將反應混合物進行冰冷後,加入二氯甲烷(3.2mL)、3-氯丙醯氯(0.17mL)、碳酸氫鈉(670mg),同溫度下攪拌30分鐘。於反應混合物中,同溫下加入異丙醇(以下,簡寫成IPA)(9.3mL)、4M氫氧化鈉水溶液(3.0mL),於室溫攪拌3小時。將反應混合物進行冰冷後,加入1M鹽酸(10mL)、飽和碳酸氫鈉水溶液、乙酸乙酯,於室溫攪拌5分鐘。將反應混合物以乙酸乙酯進行萃取,將有機層以飽和氯化鈉水溶液洗淨。將有機層以無水硫酸鎂進行乾燥後,減壓下,濃縮溶液。以二氧化矽凝膠管柱色層分析(氯仿/甲醇)純化所得之殘渣。所得之純化物中加入乙腈,於室溫攪拌1小時。過濾取出析出之固體,得到固體之1-{7-[6-環丙基-2-{[1-(3-甲氧基丙基)哌啶-4-基]氧基}-7-(5-甲基-1H-吲唑-4-基)-8-(2,2,2-三氟乙氧基)喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬-2-基}丙-2-烯-1-酮(230mg)。Manufacturing Example 8 7-[6-Cyclopropyl-2-{[1-(3-methoxypropyl)piperidin-4-yl]oxy}-7-(5-methyl-1H-indazole-4 -Yl)-8-(2,2,2-trifluoroethoxy)quinazolin-4-yl]-2,7-diazaspiro[3.5]nonane-2-carboxylic acid tertiary butyl ester A mixture of (630 mg) and 1M hydrochloric acid (5.5 mL) was stirred at 50°C for 1 hour under an argon atmosphere. After the reaction mixture was ice-cooled, dichloromethane (3.2 mL), 3-chloropropionyl chloride (0.17 mL), and sodium bicarbonate (670 mg) were added, and the mixture was stirred at the same temperature for 30 minutes. To the reaction mixture, isopropanol (hereinafter abbreviated as IPA) (9.3 mL) and 4M aqueous sodium hydroxide solution (3.0 mL) were added at the same temperature, and stirred at room temperature for 3 hours. After the reaction mixture was ice-cooled, 1M hydrochloric acid (10 mL), saturated aqueous sodium bicarbonate solution, and ethyl acetate were added, and stirred at room temperature for 5 minutes. The reaction mixture was extracted with ethyl acetate, and the organic layer was washed with saturated aqueous sodium chloride solution. After the organic layer was dried over anhydrous magnesium sulfate, the solution was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (chloroform/methanol). Acetonitrile was added to the resulting purified product, and stirred at room temperature for 1 hour. The precipitated solid was removed by filtration to obtain 1-{7-[6-cyclopropyl-2-{[1-(3-methoxypropyl)piperidin-4-yl]oxy}-7-( 5-methyl-1H-indazol-4-yl)-8-(2,2,2-trifluoroethoxy)quinazolin-4-yl]-2,7-diazaspiro[3.5] Non-2-yl}prop-2-en-1-one (230 mg).

製造例9 將7-(7-溴-2-氯-8-氟-6-碘喹唑啉-4-基)-2,7-二氮雜螺[3.5]壬烷-2-羧酸三級丁酯(8.1g)、1-(2-甲氧基乙基)哌啶-4-醇(5.3g)、碳酸銫(13g)、DABCO(220mg)、DMF (65mL)、THF(65mL)之混合物,在氬氛圍下,於室溫攪拌3日。於反應混合物中加入水,以乙酸乙酯進行萃取。將有機層以飽和氯化鈉水溶液洗淨後,以無水硫酸鎂進行乾燥。減壓下,濃縮溶液後,以二氧化矽凝膠管柱色層分析(鹼性二氧化矽凝膠、己烷/乙酸乙酯)純化所得之殘渣,得到7-(7-溴-8-氟-6-碘-2-{[1-(2-甲氧基乙基)哌啶-4-基]氧基}喹唑啉-4-基)-2,7-二氮雜螺[3.5]壬烷-2-羧酸三級丁酯(7.9g)。Manufacturing Example 9 7-(7-Bromo-2-chloro-8-fluoro-6-iodoquinazolin-4-yl)-2,7-diazaspiro[3.5]nonane-2-carboxylic acid tertiary butyl ester (8.1g), a mixture of 1-(2-methoxyethyl) piperidin-4-ol (5.3g), cesium carbonate (13g), DABCO (220mg), DMF (65mL), THF (65mL), Stir at room temperature for 3 days under argon atmosphere. Water was added to the reaction mixture, and extraction was performed with ethyl acetate. After the organic layer was washed with a saturated sodium chloride aqueous solution, it was dried over anhydrous magnesium sulfate. After concentrating the solution under reduced pressure, the resulting residue was purified by silica gel column chromatography (alkaline silica gel, hexane/ethyl acetate) to obtain 7-(7-bromo-8- Fluoro-6-iodo-2-{[1-(2-methoxyethyl)piperidin-4-yl]oxy}quinazolin-4-yl)-2,7-diazaspiro[3.5 ] Nonane-2-carboxylic acid tertiary butyl ester (7.9g).

製造例10 將7-(7-溴-8-氟-6-碘-2-{[1-(2-甲氧基乙基)哌啶-4-基]氧基}喹唑啉-4-基)-2,7-二氮雜螺[3.5]壬烷-2-羧酸三級丁酯(3.8g)、2,2,2-三氟乙醇(0.75mL)、碳酸銫(3.4g)、DMF(40mL)之混合物,在氬氛圍下,於室溫攪拌20小時。於反應混合物中加入2,2,2-三氟乙醇(0.40mL)、碳酸銫(1.7g),於室溫攪拌24小時。於反應混合物中加入水,於室溫攪拌10分鐘。過濾取出析出之固體後,加入氯仿/甲醇(9:1),將所得之溶液以無水硫酸鎂進行乾燥。減壓下,濃縮溶液後,以二氧化矽凝膠管柱色層分析(鹼性二氧化矽凝膠、己烷/乙酸乙酯)純化所得之殘渣,得到固體之7-[7-溴-6-碘-2-{[1-(2-甲氧基乙基)哌啶-4-基]氧基}-8-(2,2,2-三氟乙氧基)喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬烷-2-羧酸三級丁酯(3.1g)。Manufacturing Example 10 7-(7-Bromo-8-fluoro-6-iodo-2-{[1-(2-methoxyethyl)piperidin-4-yl]oxy}quinazolin-4-yl)- 2,7-diazaspiro[3.5]nonane-2-carboxylic acid tertiary butyl ester (3.8g), 2,2,2-trifluoroethanol (0.75mL), cesium carbonate (3.4g), DMF ( 40 mL) of the mixture was stirred at room temperature for 20 hours under an argon atmosphere. 2,2,2-Trifluoroethanol (0.40 mL) and cesium carbonate (1.7 g) were added to the reaction mixture, and stirred at room temperature for 24 hours. Water was added to the reaction mixture and stirred at room temperature for 10 minutes. After removing the precipitated solid by filtration, chloroform/methanol (9:1) was added, and the resulting solution was dried over anhydrous magnesium sulfate. After concentrating the solution under reduced pressure, the resulting residue was purified by silica gel column chromatography (alkaline silica gel, hexane/ethyl acetate) to obtain 7-[7-bromo- 6-iodo-2-{[1-(2-methoxyethyl)piperidin-4-yl]oxy}-8-(2,2,2-trifluoroethoxy)quinazoline-4 -Yl]-2,7-diazaspiro[3.5]nonane-2-carboxylic acid tertiary butyl ester (3.1 g).

製造例13 將7-[6-環丙基-2-{[1-(2-甲氧基乙基)哌啶-4-基]氧基}-7-(5-甲基-1H-吲唑-4-基)-8-(2,2,2-三氟乙氧基)喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬烷-2-羧酸三級丁酯(800mg)、二氯甲烷(4.0mL)之混合物進行冰冷後,加入三氟乙酸(以下,簡寫成TFA)(1.2mL),於室溫攪拌6小時。於反應混合物中加入氯仿/IPA(4:1)、2M碳酸鉀水溶液(11mL)。將水層以氯仿/IPA(4:1)進行萃取,將有機層以無水硫酸鎂進行乾燥後,減壓下,濃縮溶液(殘渣A)。將DIPEA(0.45mL)、THF(16mL)之混合物以冰-甲醇浴進行冷卻後,加入丙烯醯氯(0.20mL)。於此混合物中,滴加殘渣A之THF(12mL)溶液,同溫度下攪拌20分鐘。於反應混合物中,同溫度下滴加1M氫氧化鈉水溶液(5.0mL),於室溫攪拌4小時。於反應混合物中加入飽和碳酸氫鈉水溶液,以乙酸乙酯進行萃取。將有機層以無水硫酸鎂進行乾燥後,減壓下,濃縮溶液。以二氧化矽凝膠管柱色層分析(氯仿/甲醇/28%氨水)純化所得之殘渣。於所得之純化物中加入乙腈,進行超音波振盪處理。過濾取出析出之固體,得到固體之1-{7-[6-環丙基-2-{[1-(2-甲氧基乙基)哌啶-4-基]氧基}-7-(5-甲基-1H-吲唑-4-基)-8-(2,2,2-三氟乙氧基)喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬-2-基}丙-2-烯-1-酮(410mg)。Manufacturing Example 13 7-[6-Cyclopropyl-2-{[1-(2-methoxyethyl)piperidin-4-yl]oxy}-7-(5-methyl-1H-indazole-4 -Yl)-8-(2,2,2-trifluoroethoxy)quinazolin-4-yl]-2,7-diazaspiro[3.5]nonane-2-carboxylic acid tertiary butyl ester After ice-cooling a mixture of (800 mg) and dichloromethane (4.0 mL), trifluoroacetic acid (hereinafter abbreviated as TFA) (1.2 mL) was added, and the mixture was stirred at room temperature for 6 hours. Chloroform/IPA (4:1) and 2M aqueous potassium carbonate solution (11 mL) were added to the reaction mixture. The aqueous layer was extracted with chloroform/IPA (4:1), the organic layer was dried over anhydrous magnesium sulfate, and the solution (residue A) was concentrated under reduced pressure. After cooling a mixture of DIPEA (0.45 mL) and THF (16 mL) with an ice-methanol bath, propylene acetyl chloride (0.20 mL) was added. To this mixture, a solution of residue A in THF (12 mL) was added dropwise and stirred at the same temperature for 20 minutes. To the reaction mixture, 1M aqueous sodium hydroxide solution (5.0 mL) was added dropwise at the same temperature, and stirred at room temperature for 4 hours. A saturated sodium bicarbonate aqueous solution was added to the reaction mixture, and extraction was performed with ethyl acetate. After the organic layer was dried over anhydrous magnesium sulfate, the solution was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (chloroform/methanol/28% ammonia). Acetonitrile was added to the obtained purified product and subjected to ultrasonic oscillation treatment. The precipitated solid was taken out by filtration to obtain 1-{7-[6-cyclopropyl-2-{[1-(2-methoxyethyl)piperidin-4-yl]oxy}-7-( 5-methyl-1H-indazol-4-yl)-8-(2,2,2-trifluoroethoxy)quinazolin-4-yl]-2,7-diazaspiro[3.5] Non-2-yl}prop-2-en-1-one (410 mg).

製造例14 於7-[7-溴-6-碘-2-{[1-(2-甲氧基乙基)哌啶-4-基]氧基}-8-(2,2,2-三氟乙氧基)喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬烷-2-羧酸三級丁酯(3.0g)、乙烯基三氟硼酸鉀(640mg)、1,4-二

Figure 108126915-A0304-12-01
烷(30mL)、水(3.0mL)之混合物中加入碳酸鉀(1.5g)、PdCl2 (dppf)・CH2 Cl2 (30mg),在氮氛圍下,於50℃攪拌2小時。於反應混合物中加入PdCl2 (dppf)・CH2 Cl2 (150mg),在氮氛圍下,於60℃攪拌4小時。將反應混合物放置冷卻至室溫後,加入乙酸乙酯、水。將不溶物濾掉後,以乙酸乙酯萃取濾液。將有機層以無水硫酸鎂進行乾燥後,減壓下,濃縮溶液。以二氧化矽凝膠管柱色層分析(鹼性二氧化矽凝膠、己烷/乙酸乙酯)純化所得之殘渣,得到7-[7-溴-2-{[1-(2-甲氧基乙基)哌啶-4-基]氧基}-8-(2,2,2-三氟乙氧基)-6-乙烯基喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬烷-2-羧酸三級丁酯(2.2g)。Production Example 14 In 7-[7-bromo-6-iodo-2-{[1-(2-methoxyethyl)piperidin-4-yl]oxy}-8-(2,2,2- Trifluoroethoxy)quinazolin-4-yl]-2,7-diazaspiro[3.5]nonane-2-carboxylic acid tertiary butyl ester (3.0g), potassium trifluoroborate (640mg) ), 1,4-two
Figure 108126915-A0304-12-01
To a mixture of alkane (30 mL) and water (3.0 mL) was added potassium carbonate (1.5 g), PdCl 2 (dppf)·CH 2 Cl 2 (30 mg), and stirred at 50° C. for 2 hours under a nitrogen atmosphere. PdCl 2 (dppf)·CH 2 Cl 2 (150 mg) was added to the reaction mixture, and stirred at 60° C. for 4 hours under a nitrogen atmosphere. After the reaction mixture was allowed to cool to room temperature, ethyl acetate and water were added. After filtering off the insoluble matter, the filtrate was extracted with ethyl acetate. After the organic layer was dried over anhydrous magnesium sulfate, the solution was concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (alkaline silica gel, hexane/ethyl acetate) to obtain 7-[7-bromo-2-{[1-(2-methyl Oxyethyl)piperidin-4-yl]oxy)-8-(2,2,2-trifluoroethoxy)-6-vinylquinazolin-4-yl]-2,7-di Azaspiro[3.5]nonane-2-carboxylic acid tertiary butyl ester (2.2g).

製造例15 於7-[7-溴-2-{[1-(2-甲氧基乙基)哌啶-4-基]氧基}-8-(2,2,2-三氟乙氧基)-6-乙烯基喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬烷-2-羧酸三級丁酯(1.0g)、磷酸三鉀(900mg)、1,4-二

Figure 108126915-A0304-12-01
烷(10mL)、水(2.0mL)之混合物中加入(5-甲基-1H-吲唑-4-基)硼酸(490mg)、Pd2 (dba)3( 130mg)、SPhos(120mg),在氬氛圍下,於130℃攪拌4小時。將反應混合物放置冷卻至室溫後,加入乙酸乙酯、水。將不溶物濾掉後,以乙酸乙酯萃取濾液。將有機層以無水硫酸鎂進行乾燥後,減壓下,濃縮溶液。以二氧化矽凝膠管柱色層分析(氯仿/甲醇/28%氨水)純化所得之殘渣。以二氧化矽凝膠管柱色層分析(鹼性二氧化矽凝膠、己烷/乙酸乙酯)純化所得之純化物。於所得之固體中加入乙腈,於室溫攪拌。過濾取出析出之固體,得到固體之7-[2-{[1-(2-甲氧基乙基)哌啶-4-基]氧基}-7-(5-甲基-1H-吲唑-4-基)-8-(2,2,2-三氟乙氧基)-6-乙烯基喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬烷-2-羧酸三級丁酯(630mg)。Production Example 15 In 7-[7-bromo-2-{[1-(2-methoxyethyl)piperidin-4-yl]oxy}-8-(2,2,2-trifluoroethoxy Yl)-6-vinylquinazolin-4-yl]-2,7-diazaspiro[3.5]nonane-2-carboxylic acid tertiary butyl ester (1.0g), tripotassium phosphate (900mg), 1,4-two
Figure 108126915-A0304-12-01
(5-methyl-1H-indazol-4-yl)boronic acid (490mg), Pd 2 (dba) 3 ( 130mg), SPhos (120mg) was added to the mixture of alkane (10mL) and water (2.0mL). Under an argon atmosphere, the mixture was stirred at 130°C for 4 hours. After the reaction mixture was allowed to cool to room temperature, ethyl acetate and water were added. After filtering off the insoluble matter, the filtrate was extracted with ethyl acetate. After the organic layer was dried over anhydrous magnesium sulfate, the solution was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (chloroform/methanol/28% ammonia). The purified product was purified by silica gel column chromatography (alkaline silica gel, hexane/ethyl acetate). Acetonitrile was added to the obtained solid and stirred at room temperature. The precipitated solid was removed by filtration to obtain 7-[2-{[1-(2-methoxyethyl)piperidin-4-yl]oxy}-7-(5-methyl-1H-indazole -4-yl)-8-(2,2,2-trifluoroethoxy)-6-vinylquinazolin-4-yl]-2,7-diazaspiro[3.5]nonane-2 -Tertiary butyl carboxylate (630 mg).

製造例16 將7-[2-{[1-(2-甲氧基乙基)哌啶-4-基]氧基}-7-(5-甲基-1H-吲唑-4-基)-8-(2,2,2-三氟乙氧基)-6-乙烯基喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬烷-2-羧酸三級丁酯(470mg)、1M鹽酸(4.1mL)之混合物,於50℃攪拌1小時。將反應混合物進行冰冷後,加入二氯甲烷(2.3mL)、3-氯丙醯氯(0.13mL)、碳酸氫鈉(520mg),同溫度下攪拌1小時。於反應混合物中,同溫度下加入IPA(4.7mL)、4M氫氧化鈉水溶液(2.3mL),於室溫攪拌2小時。於反應混合物中加入水,以乙酸乙酯進行萃取。將有機層以飽和氯化鈉水溶液洗淨後,以無水硫酸鎂進行乾燥。減壓下,將溶液進行濃縮,以二氧化矽凝膠管柱色層分析(氯仿/甲醇/28%氨水)純化所得之殘渣。於所得之純化物中加入乙腈,於室溫攪拌5分鐘。過濾取出析出之固體後,減壓下,於30℃進行乾燥,得到固體之1-{7-[2-{[1-(2-甲氧基乙基)哌啶-4-基]氧基}-7-(5-甲基-1H-吲唑-4-基)-8-(2,2,2-三氟乙氧基)-6-乙烯基喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬-2-基}丙-2-烯-1-酮(320mg)。Manufacturing Example 16 7-[2-{[1-(2-methoxyethyl)piperidin-4-yl]oxy}-7-(5-methyl-1H-indazol-4-yl)-8- (2,2,2-trifluoroethoxy)-6-vinylquinazolin-4-yl]-2,7-diazaspiro[3.5]nonane-2-carboxylic acid tertiary butyl ester ( 470 mg), 1M hydrochloric acid (4.1 mL), and stirred at 50°C for 1 hour. After the reaction mixture was ice-cooled, dichloromethane (2.3 mL), 3-chloropropionyl chloride (0.13 mL), and sodium bicarbonate (520 mg) were added, and the mixture was stirred at the same temperature for 1 hour. To the reaction mixture, IPA (4.7 mL) and 4M aqueous sodium hydroxide solution (2.3 mL) were added at the same temperature, and stirred at room temperature for 2 hours. Water was added to the reaction mixture, and extraction was performed with ethyl acetate. After the organic layer was washed with a saturated sodium chloride aqueous solution, it was dried over anhydrous magnesium sulfate. Under reduced pressure, the solution was concentrated, and the resulting residue was purified by silica gel column chromatography (chloroform/methanol/28% ammonia). Acetonitrile was added to the obtained purified product, and stirred at room temperature for 5 minutes. After filtering out the precipitated solid, it was dried under reduced pressure at 30°C to obtain 1-{7-[2-{[1-(2-methoxyethyl)piperidin-4-yl]oxy of the solid }-7-(5-methyl-1H-indazol-4-yl)-8-(2,2,2-trifluoroethoxy)-6-vinylquinazolin-4-yl)-2 ,7-diazaspiro[3.5]non-2-yl}prop-2-en-1-one (320mg).

製造例17 於7-(7-溴-2-氯-8-氟-6-碘喹唑啉-4-基)-2,7-二氮雜螺[3.5]壬烷-2-羧酸三級丁酯(6.0g)、DMF(60mL)、THF(60mL)之混合物中,在氬氛圍下,加入1-甲基哌啶-4-醇(3.5mL)、碳酸銫(9.6g)、DABCO(220mg),於室溫攪拌14小時。於反應混合物中加入水,以乙酸乙酯進行萃取。將有機層以飽和氯化鈉水溶液洗淨後,以無水硫酸鈉進行乾燥。減壓下,濃縮溶液後,以二氧化矽凝膠管柱色層分析(氯仿/甲醇/28%氨水)純化所得之殘渣,得到7-{7-溴-8-氟-6-碘-2-[(1-甲基哌啶-4-基)氧基]喹唑啉-4-基}-2,7-二氮雜螺[3.5]壬烷-2-羧酸三級丁酯(6.3g)。Manufacturing Example 17 7-(7-Bromo-2-chloro-8-fluoro-6-iodoquinazolin-4-yl)-2,7-diazaspiro[3.5]nonane-2-carboxylic acid tertiary butyl ester (6.0g), DMF (60mL), THF (60mL) mixture, under argon atmosphere, add 1-methylpiperidin-4-ol (3.5mL), cesium carbonate (9.6g), DABCO (220mg) , Stir at room temperature for 14 hours. Water was added to the reaction mixture, and extraction was performed with ethyl acetate. After washing the organic layer with a saturated sodium chloride aqueous solution, it was dried over anhydrous sodium sulfate. After concentrating the solution under reduced pressure, the resulting residue was purified by silica gel column chromatography (chloroform/methanol/28% ammonia) to obtain 7-{7-bromo-8-fluoro-6-iodine-2 -[(1-methylpiperidin-4-yl)oxy]quinazolin-4-yl}-2,7-diazaspiro[3.5]nonane-2-carboxylic acid tertiary butyl ester (6.3 g).

製造例18 於7-{7-溴-8-氟-6-碘-2-[(1-甲基哌啶-4-基)氧基]喹唑啉-4-基}-2,7-二氮雜螺[3.5]壬烷-2-羧酸三級丁酯(3.3g)、THF(65mL)之混合物中加入乙氧化鈉(390mg),於40℃攪拌一整夜。於反應混合物中加入乙氧化鈉(390mg),於40℃攪拌一整夜。於反應混合物中加入飽和氯化銨水溶液,以乙酸乙酯進行萃取。將有機層以飽和氯化鈉水溶液洗淨後,以無水硫酸鎂進行乾燥。減壓下,濃縮溶液後,以二氧化矽凝膠管柱色層分析(鹼性二氧化矽凝膠、己烷/乙酸乙酯)純化所得之殘渣,得到固體之7-{7-溴-8-乙氧基-6-碘-2-[(1-甲基哌啶-4-基)氧基]喹唑啉-4-基}-2,7-二氮雜螺[3.5]壬烷-2-羧酸三級丁酯(1.3g)。Manufacturing Example 18 7-{7-Bromo-8-fluoro-6-iodo-2-[(1-methylpiperidin-4-yl)oxy]quinazolin-4-yl}-2,7-diaza Sodium ethoxide (390 mg) was added to a mixture of spiro[3.5]nonane-2-carboxylic acid tertiary butyl ester (3.3 g) and THF (65 mL), and stirred overnight at 40°C. Sodium ethoxide (390 mg) was added to the reaction mixture, and stirred at 40°C overnight. A saturated aqueous ammonium chloride solution was added to the reaction mixture, and extraction was performed with ethyl acetate. After the organic layer was washed with a saturated sodium chloride aqueous solution, it was dried over anhydrous magnesium sulfate. After concentrating the solution under reduced pressure, the resulting residue was purified by silica gel column chromatography (alkaline silica gel, hexane/ethyl acetate) to obtain 7-{7-bromo- 8-ethoxy-6-iodo-2-[(1-methylpiperidin-4-yl)oxy]quinazolin-4-yl}-2,7-diazaspiro[3.5]nonane -2-carboxylic acid tertiary butyl ester (1.3 g).

製造例19 於7-{7-溴-8-乙氧基-6-碘-2-[(1-甲基哌啶-4-基)氧基]喹唑啉-4-基}-2,7-二氮雜螺[3.5]壬烷-2-羧酸三級丁酯(9.5g)、4,4,5,5-四甲基-2-乙烯基-1,3,2-二氧環戊硼烷(4.5mL)、1,4-二

Figure 108126915-A0304-12-01
烷(100mL)、水(10mL)之混合物中加入碳酸鉀(5.5g)、PdCl2 (dppf)・CH2 Cl2 (1.1g),在氬氛圍下,於80℃攪拌1小時。將反應混合物放置冷卻至室溫後,加入乙酸乙酯、飽和氯化鈉水溶液、矽藻土,於室溫攪拌10分鐘。將不溶物濾掉後,以乙酸乙酯萃取濾液,將有機層以無水硫酸鎂進行乾燥。減壓下,濃縮溶液後,以二氧化矽凝膠管柱色層分析(鹼性二氧化矽凝膠、己烷/乙酸乙酯)純化所得之殘渣,得到固體之7-{7-溴-8-乙氧基-2-[(1-甲基哌啶-4-基)氧基]-6-乙烯基喹唑啉-4-基}-2,7-二氮雜螺[3.5]壬烷-2-羧酸三級丁酯(6.8g)。Production Example 19 In 7-{7-bromo-8-ethoxy-6-iodo-2-[(1-methylpiperidin-4-yl)oxy]quinazolin-4-yl}-2, 7-Diazaspiro[3.5]nonane-2-carboxylic acid tertiary butyl ester (9.5g), 4,4,5,5-tetramethyl-2-vinyl-1,3,2-diox Cyclopentane (4.5mL), 1,4-di
Figure 108126915-A0304-12-01
To a mixture of alkane (100 mL) and water (10 mL), potassium carbonate (5.5 g), PdCl 2 (dppf)·CH 2 Cl 2 (1.1 g) were added, and the mixture was stirred at 80° C. for 1 hour under an argon atmosphere. After the reaction mixture was left to cool to room temperature, ethyl acetate, saturated aqueous sodium chloride solution, and diatomaceous earth were added, and stirred at room temperature for 10 minutes. After filtering off the insoluble matter, the filtrate was extracted with ethyl acetate, and the organic layer was dried over anhydrous magnesium sulfate. After concentrating the solution under reduced pressure, the resulting residue was purified by silica gel column chromatography (alkaline silica gel, hexane/ethyl acetate) to obtain 7-{7-bromo- 8-ethoxy-2-[(1-methylpiperidin-4-yl)oxy]-6-vinylquinazolin-4-yl}-2,7-diazaspiro[3.5]non Alkane-2-carboxylic acid tertiary butyl ester (6.8 g).

製造例20 將7-{7-溴-8-乙氧基-2-[(1-甲基哌啶-4-基)氧基]-6-乙烯基喹唑啉-4-基}-2,7-二氮雜螺[3.5]壬烷-2-羧酸三級丁酯(5.7g)、(5-甲基-1H-吲唑-4-基)硼酸(2.4g)、Pd2 (dba)3( 850mg)、SPhos(760mg)、1,4-二

Figure 108126915-A0304-12-01
烷(60mL)、磷酸三鉀(10g)、水(12mL)之混合物,在氬氛圍下,於120℃攪拌4小時。將反應混合物放置冷卻至室溫後,加入乙酸乙酯、飽和氯化鈉水溶液,將不溶物濾掉。以乙酸乙酯萃取濾液。將有機層以無水硫酸鎂進行乾燥。減壓下,濃縮溶液後,以二氧化矽凝膠管柱色層分析(氯仿/甲醇/28%氨水)純化所得之殘渣。於所得之純化物中加入乙腈,於室溫攪拌6小時。過濾取出析出之固體,得到固體之7-{8-乙氧基-7-(5-甲基-1H-吲唑-4-基)-2-[(1-甲基哌啶-4-基)氧基]-6-乙烯基喹唑啉-4-基}-2,7-二氮雜螺[3.5]壬烷-2-羧酸三級丁酯(2.2g)。Production Example 20 7-{7-Bromo-8-ethoxy-2-[(1-methylpiperidin-4-yl)oxy]-6-vinylquinazolin-4-yl}-2 ,7-diazaspiro[3.5]nonane-2-carboxylic acid tertiary butyl ester (5.7g), (5-methyl-1H-indazol-4-yl)boronic acid (2.4g), Pd 2 ( dba) 3 ( 850mg), SPhos (760mg), 1,4-di
Figure 108126915-A0304-12-01
A mixture of alkane (60 mL), tripotassium phosphate (10 g), and water (12 mL) was stirred at 120° C. for 4 hours under an argon atmosphere. After the reaction mixture was left to cool to room temperature, ethyl acetate and saturated aqueous sodium chloride solution were added, and the insoluble matter was filtered off. The filtrate was extracted with ethyl acetate. The organic layer was dried over anhydrous magnesium sulfate. After concentrating the solution under reduced pressure, the resulting residue was purified by silica gel column chromatography (chloroform/methanol/28% ammonia). Acetonitrile was added to the obtained purified product, and stirred at room temperature for 6 hours. The precipitated solid was taken out by filtration to obtain 7-{8-ethoxy-7-(5-methyl-1H-indazol-4-yl)-2-[(1-methylpiperidin-4-yl )Oxy]-6-vinylquinazolin-4-yl}-2,7-diazaspiro[3.5]nonane-2-carboxylic acid tertiary butyl ester (2.2 g).

製造例21 將7-{8-乙氧基-7-(5-甲基-1H-吲唑-4-基)-2-[(1-甲基哌啶-4-基)氧基]-6-乙烯基喹唑啉-4-基}-2,7-二氮雜螺[3.5]壬烷-2-羧酸三級丁酯(1.3g)、1M鹽酸(13mL)之混合物,於室溫攪拌18小時。將反應混合物進行冰冷後,加入二氯甲烷(6.5mL)、3-氯丙醯氯(0.41mL)、碳酸氫鈉(1.6g),同溫度下攪拌30分鐘。於反應混合物中,同溫度下加入IPA(13mL)、4M氫氧化鈉水溶液(4.9mL),於室溫攪拌4小時。將反應混合物進行冰冷後,加入1M鹽酸(6.4mL)、5%碳酸氫鈉水溶液(26mL)、氯仿(39mL),於室溫攪拌10分鐘。將反應混合物進行矽藻土過濾,將濾液以氯仿進行萃取。將有機層以無水硫酸鎂進行乾燥後,減壓下,濃縮溶液。以二氧化矽凝膠管柱色層分析(氯仿/甲醇/28%氨水)純化所得之殘渣。於所得之純化物中加入乙腈,過濾取出析出之固體。將所得之固體在減壓下,於50℃進行乾燥,得到固體之1-(7-{8-乙氧基-7-(5-甲基-1H-吲唑-4-基)-2-[(1-甲基哌啶-4-基)氧基]-6-乙烯基喹唑啉-4-基}-2,7-二氮雜螺[3.5]壬-2-基)丙-2-烯-1-酮(980mg)。Production Example 21 7-{8-ethoxy-7-(5-methyl-1H-indazol-4-yl)-2-[(1-methylpiperidin-4-yl)oxy]-6-ethylene Quinazolin-4-yl}-2,7-diazaspiro[3.5]nonane-2-carboxylic acid tertiary butyl ester (1.3g), 1M hydrochloric acid (13mL) mixture, stirred at room temperature 18 hour. After the reaction mixture was ice-cooled, dichloromethane (6.5 mL), 3-chloropropionyl chloride (0.41 mL), and sodium bicarbonate (1.6 g) were added, and the mixture was stirred at the same temperature for 30 minutes. To the reaction mixture, IPA (13 mL) and 4M aqueous sodium hydroxide solution (4.9 mL) were added at the same temperature, and stirred at room temperature for 4 hours. After the reaction mixture was ice-cooled, 1M hydrochloric acid (6.4 mL), 5% sodium bicarbonate aqueous solution (26 mL), and chloroform (39 mL) were added, and the mixture was stirred at room temperature for 10 minutes. The reaction mixture was filtered through celite, and the filtrate was extracted with chloroform. After the organic layer was dried over anhydrous magnesium sulfate, the solution was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (chloroform/methanol/28% ammonia). Acetonitrile was added to the obtained purified product, and the precipitated solid was filtered out. The obtained solid was dried under reduced pressure at 50°C to obtain 1-(7-{8-ethoxy-7-(5-methyl-1H-indazol-4-yl)-2- [(1-methylpiperidin-4-yl)oxy]-6-vinylquinazolin-4-yl}-2,7-diazaspiro[3.5]non-2-yl)propan-2 -En-1-one (980 mg).

以上述所示製造例之製造方法及同樣的方法,製造後述表4~表10所示之化合物。又,各製造例化合物之製造法、結構及物理化學性數據表示於表4~表10。The compounds shown in Tables 4 to 10 described below were produced by the production method shown in the above production example and the same method. In addition, the manufacturing method, structure, and physicochemical data of the compounds of each manufacturing example are shown in Table 4 to Table 10.

參考例1 將1-{7-[6-環丙基-2-{[1-(3-甲氧基丙基)哌啶-4-基]氧基}-7-(5-甲基-1H-吲唑-4-基)-8-(2,2,2-三氟乙氧基)喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬-2-基}丙-2-烯-1-酮(2.2g)以超臨界流體色層分析(CHIRALPAK AS-H(DAICEL公司製),二氧化碳/乙醇/三乙胺)進行分劃。以二氧化矽凝膠管柱色層分析(氯仿/甲醇)純化所得之分劃物,得到(+)-1-{7-[6-環丙基-2-{[1-(3-甲氧基丙基)哌啶-4-基]氧基}-7-(5-甲基-1H-吲唑-4-基)-8-(2,2,2-三氟乙氧基)喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬-2-基}丙-2-烯-1-酮(化合物A)(1.1g)。Reference example 1 1-{7-[6-cyclopropyl-2-{[1-(3-methoxypropyl)piperidin-4-yl]oxy}-7-(5-methyl-1H-ind Azole-4-yl)-8-(2,2,2-trifluoroethoxy)quinazolin-4-yl]-2,7-diazaspiro[3.5]non-2-yl}propane- 2-En-1-one (2.2 g) was divided by supercritical fluid chromatography (CHIRALPAK AS-H (manufactured by DAICEL), carbon dioxide/ethanol/triethylamine). The obtained fraction was purified by silica gel column chromatography (chloroform/methanol) to obtain (+)-1-{7-[6-cyclopropyl-2-{[1-(3-methyl Oxypropyl)piperidin-4-yl]oxy)-7-(5-methyl-1H-indazol-4-yl)-8-(2,2,2-trifluoroethoxy)quin Oxazolin-4-yl]-2,7-diazaspiro[3.5]non-2-yl}prop-2-en-1-one (compound A) (1.1 g).

參考例3 將1-{7-[2-{[1-(2-甲氧基乙基)哌啶-4-基]氧基}-7-(5-甲基-1H-吲唑-4-基)-8-(2,2,2-三氟乙氧基)-6-乙烯基喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬-2-基}丙-2-烯-1-酮(2.0g)以超臨界流體色層分析(CHIRALPAK AS-H(DAICEL公司製),二氧化碳/甲醇/三乙胺、二氧化碳/乙醇/三乙胺)進行分劃。以二氧化矽凝膠管柱色層分析(氯仿/甲醇/28%氨水)純化所得之分劃物,得到(+)-1-{7-[2-{[1-(2-甲氧基乙基)哌啶-4-基]氧基}-7-(5-甲基-1H-吲唑-4-基)-8-(2,2,2-三氟乙氧基)-6-乙烯基喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬-2-基}丙-2-烯-1-酮(化合物C)(810mg)。Reference Example 3 Put 1-{7-[2-{[1-(2-methoxyethyl)piperidin-4-yl]oxy}-7-(5-methyl-1H-indazol-4-yl) -8-(2,2,2-trifluoroethoxy)-6-vinylquinazolin-4-yl]-2,7-diazaspiro[3.5]non-2-yl}propan-2 -En-1-one (2.0 g) was divided by supercritical fluid chromatography (CHIRALPAK AS-H (manufactured by DAICEL), carbon dioxide/methanol/triethylamine, carbon dioxide/ethanol/triethylamine). The obtained fraction was purified by silica gel column chromatography (chloroform/methanol/28% ammonia) to obtain (+)-1-{7-[2-{[1-(2-methoxy Ethyl)piperidin-4-yl)oxy)-7-(5-methyl-1H-indazol-4-yl)-8-(2,2,2-trifluoroethoxy)-6- Vinylquinazolin-4-yl]-2,7-diazaspiro[3.5]non-2-yl}prop-2-en-1-one (Compound C) (810 mg).

參考例4 將1-(7-{8-乙氧基-7-(5-甲基-1H-吲唑-4-基)-2-[(1-甲基哌啶-4-基)氧基]-6-乙烯基喹唑啉-4-基}-2,7-二氮雜螺[3.5]壬-2-基)丙-2-烯-1-酮(1.5g)以超臨界流體色層分析(CHIRALPAK AS-H(DAICEL公司製),二氧化碳/乙醇/三乙胺)進行分劃。於所得之分劃物中加入己烷、乙酸乙酯,進行研製。過濾取出析出之固體,得到固體之(+)-1-(7-{8-乙氧基-7-(5-甲基-1H-吲唑-4-基)-2-[(1-甲基哌啶-4-基)氧基]-6-乙烯基喹唑啉-4-基}-2,7-二氮雜螺[3.5]壬-2-基)丙-2-烯-1-酮(670mg)。 以二氧化矽凝膠管柱色層分析(ODS二氧化矽凝膠、水/甲醇)純化藉由進行上述方法複數次而得之固體(3.0g)。於所得之純化物中加入乙腈,於室溫攪拌。過濾取出析出之固體,將所得之固體在減壓下,於40℃進行乾燥,得到結晶之(+)-1-(7-{8-乙氧基-7-(5-甲基-1H-吲唑-4-基)-2-[(1-甲基哌啶-4-基)氧基]-6-乙烯基喹唑啉-4-基}-2,7-二氮雜螺[3.5]壬-2-基)丙-2-烯-1-酮(化合物D)(2.0g)。Reference Example 4 1-(7-{8-ethoxy-7-(5-methyl-1H-indazol-4-yl)-2-[(1-methylpiperidin-4-yl)oxy]- 6-vinylquinazolin-4-yl}-2,7-diazaspiro[3.5]non-2-yl)prop-2-en-1-one (1.5g) by supercritical fluid chromatography (CHIRALPAK AS-H (manufactured by DAICEL), carbon dioxide/ethanol/triethylamine). Hexane and ethyl acetate were added to the obtained division to conduct trituration. The precipitated solid was taken out by filtration to obtain (+)-1-(7-{8-ethoxy-7-(5-methyl-1H-indazol-4-yl)-2-[(1-methyl Piperidin-4-yl)oxy]-6-vinylquinazolin-4-yl}-2,7-diazaspiro[3.5]non-2-yl)prop-2-ene-1- Ketone (670mg). The solid (3.0 g) obtained by performing the above method a plurality of times was purified by silica gel column chromatography (ODS silica gel, water/methanol). Acetonitrile was added to the obtained purified product and stirred at room temperature. The precipitated solid was taken out by filtration, and the obtained solid was dried under reduced pressure at 40°C to obtain crystalline (+)-1-(7-{8-ethoxy-7-(5-methyl-1H- Indazol-4-yl)-2-[(1-methylpiperidin-4-yl)oxy]-6-vinylquinazolin-4-yl}-2,7-diazaspiro[3.5 ] Non-2-yl)prop-2-en-1-one (Compound D) (2.0 g).

以與上述所示參考例4之製造方法相同的方法,製造參考例2(化合物B)。又,各參考例化合物之結構表示於後述表11~表12,各參考例化合物之製造法及物理化學性數據表示於表13。Reference Example 2 (Compound B) was produced in the same manner as the production method of Reference Example 4 shown above. In addition, the structures of the compounds of each reference example are shown in Tables 11 to 12 described later, and the manufacturing method and physicochemical data of each reference example compound are shown in Table 13.

又,後述表中,有時使用以下之縮寫。 PEx:製造例編號、REx:參考例編號、PSyn:以同樣的方法製造之製造例編號、Syn:以同樣的方法製造之參考例編號(例如,R1表示參考例1)、Str:化學結構式(表示Me:甲基、Et:乙基、Boc:tert-丁氧基羰基。此外,化學結構式中附加有「#」之化合物,表示基於軸不對稱之單一的光學異構物。)、Dat:物理化學性數據、ESI+:質量分析中之m/z值(離子化法ESI,無特別說明時為[M+H]+ )、ESI-:質量分析中之m/z值(離子化法ESI,無特別說明時為[M-H]- )、CI+:質量分析中之m/z值(離子化法CI,無特別說明時為[M+H]+ )、NMR:DMSO-d6中之1 H-NMR中之信號的δ值(ppm)、s:單線(譜)、d:雙線(譜)、t:三線(譜)、m:多重線(譜)、[α]D 20 :20℃下之比旋光度、c:測定比旋光度時的濃度(g/100mL)。 [實施例]In the following table, the following abbreviations may be used. PEx: manufacturing example number, REx: reference example number, PSyn: manufacturing example number made by the same method, Syn: reference example number made by the same method (for example, R1 means reference example 1), Str: chemical structure formula (Indicates Me: methyl, Et: ethyl, Boc: tert-butoxycarbonyl. In addition, compounds with "#" added to the chemical structural formula represent single optical isomers based on axis asymmetry.), Dat: physicochemical data, ESI+: m/z value in mass analysis (ionization method ESI, [M+H] + unless otherwise specified), ESI-: m/z value in mass analysis (ionization Method ESI, [MH] - ) without special instructions, CI+: m/z value in mass analysis (ionization method CI, [M+H] + without special instructions), NMR: DMSO-d6 Δ value (ppm) of signal in 1 H-NMR, s: single line (spectrum), d: double line (spectrum), t: triple line (spectrum), m: multiple line (spectrum), [α] D 20 : Specific optical rotation at 20°C, c: concentration when measuring the specific optical rotation (g/100 mL). [Example]

本發明之醫藥組成物之藥理性效果,藉由以下實施例確認。The pharmacological effect of the pharmaceutical composition of the present invention is confirmed by the following examples.

實施例1 KRAS G12C/SOS/c-Raf複合體形成抑制作用之評估 使用人類重組KRAS G12C、SOS及c-Raf蛋白質以時間解析螢光-螢光共振能量轉移(TR-FRET)法探討被驗化合物對此等蛋白質之複合體形成的抑制作用。 將溶解於assay buffer(50mM HEPES, 150mM NaCl, 5mM MgCl2 , 0.05% Tween 20, pH7.4)之生物素化AviTag-KRAS G12C(胺基酸區域1-185, GDP) (2.5μL; 400nM)與被驗化合物以4,000nM至4nM之2.5μL的液量添加至384孔盤(Corning公司)中。於其中加入Son of Sevenless (SOS) (胺基酸區域564-1049, 2.5μL; 1.3μM)、包含GTP (Sigma-Aldrich公司)之c-Raf(胺基酸區域51-131)GST(2.5μL; 分別為4μM及130nM)於室溫靜置1小時。之後加入LANCE Ulight-anti-GST(120nM、PerkinElmer公司)及LANCE Eu-W1024 labeled Streptoavidin (100ng/mL、PerkinElmer公司)的混合液(10μL),使用EnVision 2103 Multilabel Reader (PerkinElmer公司)在激發波長337nm之條件下,測定620nm及665nm中之螢光強度。以參考波長620nm之螢光強度將值進行標準化後,將溶劑處理之信號值定為0%抑制、未添加GTP之信號值定為100%抑制,以Sigmoid-Emax模型非線性回歸分析算出50%抑制濃度(IC50 )。作為被驗化合物使用化合物A、化合物B、化合物C及化合物D之試驗結果表示於表1。Example 1 Evaluation of KRAS G12C/SOS/c-Raf complex formation inhibition using human recombinant KRAS G12C, SOS and c-Raf proteins to investigate the time-lapse fluorescence-fluorescence resonance energy transfer (TR-FRET) method Compounds inhibit the formation of complexes of these proteins. Biotinylated AviTag-KRAS G12C (amino acid region 1-185, GDP) (2.5μL; 400nM) dissolved in assay buffer (50mM HEPES, 150mM NaCl, 5mM MgCl 2 , 0.05% Tween 20, pH7.4) The test compound was added to a 384-well dish (Corning) in a volume of 2.5 μL of 4,000 nM to 4 nM. Add Son of Sevenless (SOS) (amino acid region 564-1049, 2.5 μL; 1.3 μM), c-Raf (amino acid region 51-131) containing GTP (Sigma-Aldrich) GST (2.5 μL ; Respectively 4μM and 130nM) at room temperature for 1 hour. Then add a mixture (10μL) of LANCE Ulight-anti-GST (120nM, PerkinElmer) and LANCE Eu-W1024 labeled Streptoavidin (100ng/mL, PerkinElmer), and use EnVision 2103 Multilabel Reader (PerkinElmer) at an excitation wavelength of 337nm Under the conditions, the fluorescence intensity at 620 nm and 665 nm was measured. After normalizing the value with the fluorescence intensity at the reference wavelength of 620nm, the signal value of the solvent treatment is set to 0% suppression, and the signal value without GTP is set to 100% suppression, and the 50% is calculated by the nonlinear regression analysis of the Sigmoid-Emax model Inhibition concentration (IC 50 ). The test results of using compound A, compound B, compound C, and compound D as test compounds are shown in Table 1.

Figure 02_image005
Figure 02_image005

實施例2 腫瘤內pERK抑制作用之評估 使用pERK測定套組(Advanced ERK phospho-T202/Y204 kit, Cisbio公司)以TR-FRET法探討被驗化合物投予後之腫瘤樣本中之ERK的磷酸化量。 以含有10%牛胎兒血清(GE Life Sciences公司)之Leibovitz’s L-15培養基(Life technologies公司)於37℃培養SW1463細胞(ATCC, CCL-234),使用於PBS添加等量之基質膠(Becton, Dickinson and Company公司)而成的溶液調製2.0x106 個。將該細胞懸濁液以100μL之用量注射植入至4週齡之雄性SCID小鼠(CB17/Icr-Prkdcscid/CrlCrlj, 日本Charles River公司)皮下,25日後使用於試驗中。試驗以溶劑組及被驗化合物投予組各3隻來進行,使用溶劑以成為表2記載之投予量之方式調製被驗化合物。被驗化合物為口服投予,使用6%之2-羥基丙基-β-環糊精(Sigma-Aldrich公司)水溶液作為溶劑。投予6小時後由在異氟烷麻醉下經頸椎脫臼之小鼠摘出腫瘤,一部分放入2mL微量離心管(eppendorf),使用液體氮進行冷凍。腫瘤樣本至進行pERK測定試驗為止,皆在-80℃之低溫冷凍器內保存。 腫瘤樣本中添加500μL之lysis buffer1 (Phospho lysis buffer [Cisbio公司], Complete EDTA free [Roche公司], Phosphatase inhibitor cocktail2 [Sigma-Aldrich公司])及小珠1個(YTZ球 5mm [NIKKATO公司]),使用Tissue Lyser II (QIAGEN公司)進行破碎(frequency 25/s, 3分鐘)。將全部量移至新的管中,使用微量高速冷卻離心機進行離心(20,400xg, 10分鐘, 4℃)得到上清即腫瘤溶胞產物(lysate)。使用蛋白定量套組(Pierce 660 nm Protein Assay Kit [Thermo Fisher公司])進行腫瘤溶胞產物的蛋白定量,以最終成為1.0μg/μL之濃度的方式使用lysis buffer 2 (Phospho lysis buffer [Cisbio公司], Blocking Agent [pERK測定套組所附]稀釋各樣本。 將pERK1/2 Cryptate抗體及pERK1/2 d2抗體(pERK測定套組所附)以detection buffer (pERK測定套組所附)稀釋40倍,製作此等2種抗體之混合溶液。將抗體之混合溶液以4μL/孔之液量添加至384孔盤。進而,以16μL/孔之液量添加稀釋成1.0μg/μL之腫瘤溶胞產物。在濕箱內室溫靜置約17小時後,使用EnVision 2103 Multilabel Reader (PerkinElmer公司),以激發波長337nm之條件,測定620nm及665nm中之螢光強度。以參考波長620nm之螢光強度將值進行標準化後,將vehicle投予組之計數定為0%抑制,未添加溶胞產物之計數定為100%抑制,以%抑制率算出被驗化合物投予樣本之抑制值。作為被驗化合物使用化合物A、化合物B、化合物C及化合物D的試驗結果表示於表2。Example 2 Evaluation of pERK inhibition in tumors Using the pERK assay kit (Advanced ERK phospho-T202/Y204 kit, Cisbio), TR-FRET method was used to investigate the phosphorylation of ERK in tumor samples after administration of test compounds. SW1463 cells (ATCC, CCL-234) were cultured in Leibovitz's L-15 medium (Life technologies) containing 10% bovine fetal serum (GE Life Sciences) at 37°C, and the same amount of Matrigel (Becton, Dickinson and Company) prepared 2.0x10 6 solutions. The cell suspension was injected into a 4-week-old male SCID mouse (CB17/Icr-Prkdcscid/CrlCrlj, Charles River Corporation, Japan) at a dose of 100 μL and implanted subcutaneously and used in the test 25 days later. The test was carried out in three groups each of the solvent group and the test compound administration group, and the test compound was prepared using the solvent so as to be the administration amount described in Table 2. The test compound was administered orally, and 6% aqueous solution of 2-hydroxypropyl-β-cyclodextrin (Sigma-Aldrich) was used as a solvent. Six hours after administration, tumors were removed from mice undergoing cervical dislocation under isoflurane anesthesia, and a part was placed in a 2 mL microcentrifuge tube (eppendorf) and frozen with liquid nitrogen. Tumor samples were stored in a cryogenic freezer at -80°C until the pERK measurement test. Add 500 μL of lysis buffer1 (Phospho lysis buffer [Cisbio], Complete EDTA free [Roche], Phosphatase inhibitor cocktail2 [Sigma-Aldrich]] and 1 bead (YTZ ball 5mm [NIKKATO]) to the tumor sample, Tissue Lyser II (QIAGEN) was used for crushing (frequency 25/s, 3 minutes). The entire amount was transferred to a new tube, and centrifuged using a micro high-speed cooling centrifuge (20,400xg, 10 minutes, 4°C) to obtain the supernatant, that is, tumor lysate. The protein quantification kit (Pierce 660 nm Protein Assay Kit [Thermo Fisher]) was used to quantify the protein of the tumor lysate, and lysis buffer 2 (Phospho lysis buffer [Cisbio]) was used in such a way that the concentration became 1.0 μg/μL , Blocking Agent [attached to pERK assay kit] Dilute each sample. Dilute pERK1/2 Cryptate antibody and pERK1/2 d2 antibody (attached to pERK assay kit) with detection buffer (attached to pERK assay kit) 40 times, A mixed solution of these two kinds of antibodies was prepared. The mixed solution of antibodies was added to a 384-well dish at a liquid volume of 4 μL/well. Furthermore, a tumor lysate diluted to 1.0 μg/μL was added at a liquid volume of 16 μL/well. After standing at room temperature for about 17 hours in a wet box, use EnVision 2103 Multilabel Reader (PerkinElmer) to measure the fluorescence intensity at 620nm and 665nm under the excitation wavelength of 337nm. The value is based on the fluorescence intensity at the reference wavelength of 620nm After standardization, the count of the vehicle-administered group was set to 0% inhibition, and the count of no added lysate was set to 100% inhibition, and the inhibition value of the test compound administered to the sample was calculated as the% inhibition rate. Used as the test compound The test results of Compound A, Compound B, Compound C and Compound D are shown in Table 2.

Figure 02_image007
Figure 02_image007

實施例3 人類KRAS G12C突變陽性大腸癌株SW1463罹癌小鼠中之抗腫瘤作用之評估 使用於PBS添加等量之基質膠(Becton, Dickinson and Company公司)而成的溶液調製SW1463細胞2.0x106 個,以100μL之用量注射植入至4週齡之雄性SCID小鼠(CB17/Icr-Prkdcscid/CrlCrlj, 日本Charles River公司)皮下。植入約3週後,以各組間之腫瘤體積及體重幾乎同等的方式進行分組,自翌日開始被驗化合物的投予。試驗為以溶劑組及被驗化合物投予組各5隻來進行,於溶劑組口服投予6%之2-羥基丙基-β-環糊精(Sigma-Aldrich公司)水溶液,於被驗化合物投予組口服投予於6%之2-羥基丙基-β-環糊精水溶液中混合被驗化合物(10或40mg/kg)者。投予為1日1次進行14日,1週測定2次腫瘤徑及體重。腫瘤體積的計算使用以下之式。 [腫瘤體積(mm3 )]=[腫瘤之長徑(mm)]×[腫瘤之短徑(mm)]2 ×0.5 被驗化合物所致之腫瘤增殖抑制率(%)係將投予開始前日之被驗化合物投予組的腫瘤體積定為100%抑制,投予結束日之溶劑組的腫瘤體積定為0%抑制來算出。又,被驗化合物投予組的腫瘤體積較投予開始前日的腫瘤體積減少時,以投予開始前日的腫瘤體積定為0%退縮,腫瘤體積0定為100%退縮,來算出被驗化合物之腫瘤退縮率(%)。作為被驗化合物使用化合物A、化合物B、化合物C及化合物D之試驗結果表示於表3。Example 3 Evaluation of anti-tumor effect in human KRAS G12C mutation-positive colorectal cancer strain SW1463 cancer-bearing mice SW1463 cells 2.0x10 6 were prepared by adding an equal amount of Matrigel (Becton, Dickinson and Company) in PBS A 100-L injection was implanted subcutaneously into 4-week-old male SCID mice (CB17/Icr-Prkdcscid/CrlCrlj, Charles River, Japan). About 3 weeks after implantation, the tumor volume and body weight of each group were almost the same, and the test compound was administered from the next day. The test was conducted with 5 animals each in the solvent group and the test compound administration group, and 6% 2-hydroxypropyl-β-cyclodextrin (Sigma-Aldrich) aqueous solution was orally administered in the solvent group to the test compound In the administration group, the test compound (10 or 40 mg/kg) was mixed with 6% 2-hydroxypropyl-β-cyclodextrin aqueous solution orally. The administration was performed once a day for 14 days, and the tumor diameter and body weight were measured twice a week. The calculation of tumor volume uses the following formula. [Tumor volume (mm 3 )]=[Long diameter of tumor (mm)]×[Small diameter of tumor (mm)] 2 ×0.5 The tumor growth inhibition rate (%) caused by the test compound will be the day before the start of administration The tumor volume of the test compound administration group was determined to be 100% inhibition, and the tumor volume of the solvent group at the end of administration was determined to be 0% inhibition. In addition, when the tumor volume of the test compound administration group decreased compared with the tumor volume on the day before the start of administration, the tumor volume on the day before the start of administration was regarded as 0% retreat and the tumor volume 0 as 100% retreat to calculate the test compound The tumor shrinkage rate (%). The test results of using compound A, compound B, compound C, and compound D as test compounds are shown in Table 3.

Figure 02_image009
Figure 02_image009

由以上之結果,確認了本發明之醫藥組成物的有效成分即化合物A、化合物B、化合物C及化合物D抑制G12C突變KRAS。又,確認了此等之化合物對於KRAS G12C突變陽性大腸癌細胞移植罹癌小鼠具有抗腫瘤作用。From the above results, it was confirmed that Compound A, Compound B, Compound C, and Compound D, which are active ingredients of the pharmaceutical composition of the present invention, inhibit G12C mutation KRAS. Furthermore, it was confirmed that these compounds have an antitumor effect on mice transplanted with KRAS G12C mutation-positive colorectal cancer cells.

因此,選自由化合物A、化合物B、化合物C及化合物D所成之群組之化合物或其製藥學上容許之鹽,可使用於大腸癌,特別是KRAS G12C突變陽性大腸癌的治療。Therefore, a compound selected from the group consisting of compound A, compound B, compound C, and compound D or a pharmaceutically acceptable salt thereof can be used for the treatment of colorectal cancer, especially KRAS G12C mutation-positive colorectal cancer.

Figure 02_image011
Figure 02_image011

Figure 02_image013
Figure 02_image013

Figure 02_image015
Figure 02_image015

Figure 02_image017
Figure 02_image017

Figure 02_image019
Figure 02_image019

Figure 02_image021
Figure 02_image021

Figure 02_image023
Figure 02_image023

Figure 02_image025
Figure 02_image025

Figure 02_image027
Figure 02_image027

Figure 02_image029
[產業上之可利用性]
Figure 02_image029
[Industry availability]

本發明之醫藥組成物之有效成分即選自由化合物A、化合物B、化合物C及化合物D所成之群組之化合物,或其製藥學上容許之鹽,具有G12C突變KRAS抑制作用,大腸癌之治療用醫藥組成物,作為一態樣,可使用作為KRAS G12C突變陽性大腸癌之治療用醫藥組成物的有效成分。The active ingredient of the pharmaceutical composition of the present invention is a compound selected from the group consisting of compound A, compound B, compound C, and compound D, or a pharmaceutically acceptable salt thereof, which has the G12C mutation KRAS inhibitory effect and is The therapeutic pharmaceutical composition can be used as an active ingredient of the therapeutic pharmaceutical composition for KRAS G12C mutation-positive colorectal cancer.

Claims (18)

一種大腸癌之治療用醫藥組成物,其含有選自由(+)-1-{7-[6-環丙基-2-{[1-(3-甲氧基丙基)哌啶-4-基]氧基}-7-(5-甲基-1H-吲唑-4-基)-8-(2,2,2-三氟乙氧基)喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬-2-基}丙-2-烯-1-酮、(+)-1-{7-[6-環丙基-2-{[1-(2-甲氧基乙基)哌啶-4-基]氧基}-7-(5-甲基-1H-吲唑-4-基)-8-(2,2,2-三氟乙氧基)喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬-2-基}丙-2-烯-1-酮、(+)-1-{7-[2-{[1-(2-甲氧基乙基)哌啶-4-基]氧基}-7-(5-甲基-1H-吲唑-4-基)-8-(2,2,2-三氟乙氧基)-6-乙烯基喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬-2-基}丙-2-烯-1-酮,及(+)-1-(7-{8-乙氧基-7-(5-甲基-1H-吲唑-4-基)-2-[(1-甲基哌啶-4-基)氧基]-6-乙烯基喹唑啉-4-基}-2,7-二氮雜螺[3.5]壬-2-基)丙-2-烯-1-酮所成之群組之化合物,或其製藥學上容許之鹽,及製藥學上容許之賦形劑。A medical composition for the treatment of colorectal cancer, which is selected from (+)-1-{7-[6-cyclopropyl-2-{[1-(3-methoxypropyl)piperidine-4- Yl]oxy}-7-(5-methyl-1H-indazol-4-yl)-8-(2,2,2-trifluoroethoxy)quinazolin-4-yl]-2, 7-diazaspiro[3.5]non-2-yl}prop-2-en-1-one, (+)-1-{7-[6-cyclopropyl-2-{[1-(2- (Methoxyethyl) piperidin-4-yl)oxy)-7-(5-methyl-1H-indazol-4-yl)-8-(2,2,2-trifluoroethoxy) Quinazolin-4-yl]-2,7-diazaspiro[3.5]non-2-yl}prop-2-en-1-one, (+)-1-{7-[2-{[ 1-(2-methoxyethyl)piperidin-4-yl)oxy)-7-(5-methyl-1H-indazol-4-yl)-8-(2,2,2-tri Fluoroethoxy)-6-vinylquinazolin-4-yl]-2,7-diazaspiro[3.5]non-2-yl}prop-2-en-1-one, and (+) -1-(7-{8-ethoxy-7-(5-methyl-1H-indazol-4-yl)-2-[(1-methylpiperidin-4-yl)oxy]- 6-vinylquinazolin-4-yl}-2,7-diazaspiro[3.5]non-2-yl)prop-2-en-1-one, or a compound thereof Pharmaceutically acceptable salts and pharmaceutically acceptable excipients. 如請求項1之醫藥組成物,其中化合物為(+)-1-{7-[6-環丙基-2-{[1-(3-甲氧基丙基)哌啶-4-基]氧基}-7-(5-甲基-1H-吲唑-4-基)-8-(2,2,2-三氟乙氧基)喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬-2-基}丙-2-烯-1-酮。The pharmaceutical composition according to claim 1, wherein the compound is (+)-1-{7-[6-cyclopropyl-2-{[1-(3-methoxypropyl)piperidin-4-yl] Oxy)-7-(5-methyl-1H-indazol-4-yl)-8-(2,2,2-trifluoroethoxy)quinazolin-4-yl)-2,7- Diazaspiro[3.5]non-2-yl}prop-2-en-1-one. 如請求項1之醫藥組成物,其中化合物為(+)-1-{7-[6-環丙基-2-{[1-(2-甲氧基乙基)哌啶-4-基]氧基}-7-(5-甲基-1H-吲唑-4-基)-8-(2,2,2-三氟乙氧基)喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬-2-基}丙-2-烯-1-酮。The pharmaceutical composition according to claim 1, wherein the compound is (+)-1-{7-[6-cyclopropyl-2-{[1-(2-methoxyethyl)piperidin-4-yl] Oxy)-7-(5-methyl-1H-indazol-4-yl)-8-(2,2,2-trifluoroethoxy)quinazolin-4-yl)-2,7- Diazaspiro[3.5]non-2-yl}prop-2-en-1-one. 如請求項1之醫藥組成物,其中化合物為(+)-1-{7-[2-{[1-(2-甲氧基乙基)哌啶-4-基]氧基}-7-(5-甲基-1H-吲唑-4-基)-8-(2,2,2-三氟乙氧基)-6-乙烯基喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬-2-基}丙-2-烯-1-酮。The pharmaceutical composition according to claim 1, wherein the compound is (+)-1-{7-[2-{[1-(2-methoxyethyl)piperidin-4-yl]oxy}-7- (5-methyl-1H-indazol-4-yl)-8-(2,2,2-trifluoroethoxy)-6-vinylquinazolin-4-yl)-2,7-di Azaspiro[3.5]non-2-yl}prop-2-en-1-one. 如請求項1之醫藥組成物,其中化合物為(+)-1-(7-{8-乙氧基-7-(5-甲基-1H-吲唑-4-基)-2-[(1-甲基哌啶-4-基)氧基]-6-乙烯基喹唑啉-4-基}-2,7-二氮雜螺[3.5]壬-2-基)丙-2-烯-1-酮。The pharmaceutical composition according to claim 1, wherein the compound is (+)-1-(7-{8-ethoxy-7-(5-methyl-1H-indazol-4-yl)-2-[( 1-methylpiperidin-4-yl)oxy]-6-vinylquinazolin-4-yl}-2,7-diazaspiro[3.5]non-2-yl)prop-2-ene -1-one. 如請求項1~請求項5中任一項之醫藥組成物,其中大腸癌為KRAS G12C突變陽性大腸癌。The pharmaceutical composition according to any one of claim 1 to claim 5, wherein the colorectal cancer is KRAS G12C mutation-positive colorectal cancer. 一種用於治療大腸癌之用途的選自由(+)-1-{7-[6-環丙基-2-{[1-(3-甲氧基丙基)哌啶-4-基]氧基}-7-(5-甲基-1H-吲唑-4-基)-8-(2,2,2-三氟乙氧基)喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬-2-基}丙-2-烯-1-酮、(+)-1-{7-[6-環丙基-2-{[1-(2-甲氧基乙基)哌啶-4-基]氧基}-7-(5-甲基-1H-吲唑-4-基)-8-(2,2,2-三氟乙氧基)喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬-2-基}丙-2-烯-1-酮、(+)-1-{7-[2-{[1-(2-甲氧基乙基)哌啶-4-基]氧基}-7-(5-甲基-1H-吲唑-4-基)-8-(2,2,2-三氟乙氧基)-6-乙烯基喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬-2-基}丙-2-烯-1-酮,及(+)-1-(7-{8-乙氧基-7-(5-甲基-1H-吲唑-4-基)-2-[(1-甲基哌啶-4-基)氧基]-6-乙烯基喹唑啉-4-基}-2,7-二氮雜螺[3.5]壬-2-基)丙-2-烯-1-酮所成之群組之化合物,或其製藥學上容許之鹽。A use for treating colorectal cancer is selected from (+)-1-{7-[6-cyclopropyl-2-{[1-(3-methoxypropyl)piperidin-4-yl]oxygen Yl)-7-(5-methyl-1H-indazol-4-yl)-8-(2,2,2-trifluoroethoxy)quinazolin-4-yl)-2,7-di Azaspiro[3.5]non-2-yl}prop-2-en-1-one, (+)-1-{7-[6-cyclopropyl-2-{[1-(2-methoxy Ethyl)piperidin-4-yl)oxy)-7-(5-methyl-1H-indazol-4-yl)-8-(2,2,2-trifluoroethoxy)quinazoline -4-yl]-2,7-diazaspiro[3.5]non-2-yl}prop-2-en-1-one, (+)-1-{7-[2-{[1-( 2-methoxyethyl)piperidin-4-yl)oxy)-7-(5-methyl-1H-indazol-4-yl)-8-(2,2,2-trifluoroethoxy Group)-6-vinylquinazolin-4-yl]-2,7-diazaspiro[3.5]non-2-yl}prop-2-en-1-one, and (+)-1- (7-{8-ethoxy-7-(5-methyl-1H-indazol-4-yl)-2-[(1-methylpiperidin-4-yl)oxy]-6-ethylene Quinazolin-4-yl}-2,7-diazaspiro[3.5]non-2-yl)prop-2-en-1-one, or its pharmacologically acceptable Salt. 如請求項7之化合物或其製藥學上容許之鹽,其中化合物為(+)-1-{7-[6-環丙基-2-{[1-(3-甲氧基丙基)哌啶-4-基]氧基}-7-(5-甲基-1H-吲唑-4-基)-8-(2,2,2-三氟乙氧基)喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬-2-基}丙-2-烯-1-酮。If the compound of claim 7 or a pharmaceutically acceptable salt thereof, the compound is (+)-1-{7-[6-cyclopropyl-2-{[1-(3-methoxypropyl)piper Pyridin-4-yl]oxy}-7-(5-methyl-1H-indazol-4-yl)-8-(2,2,2-trifluoroethoxy)quinazolin-4-yl ]-2,7-diazaspiro[3.5]non-2-yl}prop-2-en-1-one. 如請求項7之化合物或其製藥學上容許之鹽,其中化合物為(+)-1-{7-[6-環丙基-2-{[1-(2-甲氧基乙基)哌啶-4-基]氧基}-7-(5-甲基-1H-吲唑-4-基)-8-(2,2,2-三氟乙氧基)喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬-2-基}丙-2-烯-1-酮。If the compound of claim 7 or a pharmaceutically acceptable salt thereof, the compound is (+)-1-{7-[6-cyclopropyl-2-{[1-(2-methoxyethyl)piper Pyridin-4-yl]oxy}-7-(5-methyl-1H-indazol-4-yl)-8-(2,2,2-trifluoroethoxy)quinazolin-4-yl ]-2,7-diazaspiro[3.5]non-2-yl}prop-2-en-1-one. 如請求項7之化合物或其製藥學上容許之鹽,其中化合物為(+)-1-{7-[2-{[1-(2-甲氧基乙基)哌啶-4-基]氧基}-7-(5-甲基-1H-吲唑-4-基)-8-(2,2,2-三氟乙氧基)-6-乙烯基喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬-2-基}丙-2-烯-1-酮。If the compound of claim 7 or a pharmaceutically acceptable salt thereof, the compound is (+)-1-{7-[2-{[1-(2-methoxyethyl)piperidin-4-yl] Oxy)-7-(5-methyl-1H-indazol-4-yl)-8-(2,2,2-trifluoroethoxy)-6-vinylquinazolin-4-yl] -2,7-diazaspiro[3.5]non-2-yl}prop-2-en-1-one. 如請求項7之化合物或其製藥學上容許之鹽,其中化合物為(+)-1-(7-{8-乙氧基-7-(5-甲基-1H-吲唑-4-基)-2-[(1-甲基哌啶-4-基)氧基]-6-乙烯基喹唑啉-4-基}-2,7-二氮雜螺[3.5]壬-2-基)丙-2-烯-1-酮。The compound of claim 7 or a pharmaceutically acceptable salt thereof, wherein the compound is (+)-1-(7-{8-ethoxy-7-(5-methyl-1H-indazol-4-yl )-2-[(1-methylpiperidin-4-yl)oxy]-6-vinylquinazolin-4-yl}-2,7-diazaspiro[3.5]non-2-yl ) Prop-2-en-1-one. 如請求項7~請求項11中任一項之化合物或其製藥學上容許之鹽,其中大腸癌為KRAS G12C突變陽性大腸癌。If the compound of any one of claim 7 to claim 11 or a pharmaceutically acceptable salt thereof, the colorectal cancer is KRAS G12C mutation-positive colorectal cancer. 一種大腸癌之治療方法,其包含將選自由(+)-1-{7-[6-環丙基-2-{[1-(3-甲氧基丙基)哌啶-4-基]氧基}-7-(5-甲基-1H-吲唑-4-基)-8-(2,2,2-三氟乙氧基)喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬-2-基}丙-2-烯-1-酮、(+)-1-{7-[6-環丙基-2-{[1-(2-甲氧基乙基)哌啶-4-基]氧基}-7-(5-甲基-1H-吲唑-4-基)-8-(2,2,2-三氟乙氧基)喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬-2-基}丙-2-烯-1-酮、(+)-1-{7-[2-{[1-(2-甲氧基乙基)哌啶-4-基]氧基}-7-(5-甲基-1H-吲唑-4-基)-8-(2,2,2-三氟乙氧基)-6-乙烯基喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬-2-基}丙-2-烯-1-酮,及(+)-1-(7-{8-乙氧基-7-(5-甲基-1H-吲唑-4-基)-2-[(1-甲基哌啶-4-基)氧基]-6-乙烯基喹唑啉-4-基}-2,7-二氮雜螺[3.5]壬-2-基)丙-2-烯-1-酮所成之群組之化合物或其製藥學上容許之鹽的有效量投予至對象而成。A treatment method for colorectal cancer, which will be selected from (+)-1-{7-[6-cyclopropyl-2-{[1-(3-methoxypropyl)piperidin-4-yl] Oxy)-7-(5-methyl-1H-indazol-4-yl)-8-(2,2,2-trifluoroethoxy)quinazolin-4-yl)-2,7- Diazaspiro[3.5]non-2-yl}prop-2-en-1-one, (+)-1-{7-[6-cyclopropyl-2-{[1-(2-methoxy Ethyl)piperidin-4-yl)oxy)-7-(5-methyl-1H-indazol-4-yl)-8-(2,2,2-trifluoroethoxy)quinazol Lin-4-yl]-2,7-diazaspiro[3.5]non-2-yl}prop-2-en-1-one, (+)-1-{7-[2-{[1- (2-Methoxyethyl)piperidin-4-yl)oxy)-7-(5-methyl-1H-indazol-4-yl)-8-(2,2,2-trifluoroethane Oxy)-6-vinylquinazolin-4-yl]-2,7-diazaspiro[3.5]non-2-yl}prop-2-en-1-one, and (+)-1 -(7-{8-ethoxy-7-(5-methyl-1H-indazol-4-yl)-2-[(1-methylpiperidin-4-yl)oxy]-6- Vinylquinazolin-4-yl}-2,7-diazaspiro[3.5]non-2-yl)prop-2-en-1-one group of compounds or pharmaceutically acceptable The effective amount of the salt is administered to the target. 如請求項13之大腸癌之治療方法,其中化合物為(+)-1-{7-[6-環丙基-2-{[1-(3-甲氧基丙基)哌啶-4-基]氧基}-7-(5-甲基-1H-吲唑-4-基)-8-(2,2,2-三氟乙氧基)喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬-2-基}丙-2-烯-1-酮。The treatment method for colorectal cancer according to claim 13, wherein the compound is (+)-1-{7-[6-cyclopropyl-2-{[1-(3-methoxypropyl)piperidine-4- Yl]oxy}-7-(5-methyl-1H-indazol-4-yl)-8-(2,2,2-trifluoroethoxy)quinazolin-4-yl]-2, 7-diazaspiro[3.5]non-2-yl}prop-2-en-1-one. 如請求項13之大腸癌之治療方法,其中化合物為(+)-1-{7-[6-環丙基-2-{[1-(2-甲氧基乙基)哌啶-4-基]氧基}-7-(5-甲基-1H-吲唑-4-基)-8-(2,2,2-三氟乙氧基)喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬-2-基}丙-2-烯-1-酮。The treatment method for colorectal cancer according to claim 13, wherein the compound is (+)-1-{7-[6-cyclopropyl-2-{[1-(2-methoxyethyl)piperidine-4- Yl]oxy}-7-(5-methyl-1H-indazol-4-yl)-8-(2,2,2-trifluoroethoxy)quinazolin-4-yl]-2, 7-diazaspiro[3.5]non-2-yl}prop-2-en-1-one. 如請求項13之大腸癌之治療方法,其中化合物為(+)-1-{7-[2-{[1-(2-甲氧基乙基)哌啶-4-基]氧基}-7-(5-甲基-1H-吲唑-4-基)-8-(2,2,2-三氟乙氧基)-6-乙烯基喹唑啉-4-基]-2,7-二氮雜螺[3.5]壬-2-基}丙-2-烯-1-酮。The treatment method for colorectal cancer as claimed in claim 13, wherein the compound is (+)-1-{7-[2-{[1-(2-methoxyethyl)piperidin-4-yl]oxy}- 7-(5-methyl-1H-indazol-4-yl)-8-(2,2,2-trifluoroethoxy)-6-vinylquinazolin-4-yl)-2,7 -Diazaspiro[3.5]non-2-yl}prop-2-en-1-one. 如請求項13之大腸癌之治療方法,其中化合物為(+)-1-(7-{8-乙氧基-7-(5-甲基-1H-吲唑-4-基)-2-[(1-甲基哌啶-4-基)氧基]-6-乙烯基喹唑啉-4-基}-2,7-二氮雜螺[3.5]壬-2-基)丙-2-烯-1-酮。The treatment method for colorectal cancer according to claim 13, wherein the compound is (+)-1-(7-{8-ethoxy-7-(5-methyl-1H-indazol-4-yl)-2- [(1-methylpiperidin-4-yl)oxy]-6-vinylquinazolin-4-yl}-2,7-diazaspiro[3.5]non-2-yl)propan-2 -En-1-one. 如請求項13~請求項17中任一項之大腸癌之治療方法,其中大腸癌為KRAS G12C突變陽性大腸癌。The treatment method for colorectal cancer according to any one of claim 13 to claim 17, wherein the colorectal cancer is KRAS G12C mutation-positive colorectal cancer.
TW108126915A 2018-07-31 2019-07-30 Pharmaceutical composition comprising quinazoline compound as active ingredient TW202019426A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2018-144066 2018-07-31
JP2018144066A JP2021176819A (en) 2018-07-31 2018-07-31 Pharmaceutical composition comprising quinazoline compound as active ingredient

Publications (1)

Publication Number Publication Date
TW202019426A true TW202019426A (en) 2020-06-01

Family

ID=69231889

Family Applications (1)

Application Number Title Priority Date Filing Date
TW108126915A TW202019426A (en) 2018-07-31 2019-07-30 Pharmaceutical composition comprising quinazoline compound as active ingredient

Country Status (3)

Country Link
JP (1) JP2021176819A (en)
TW (1) TW202019426A (en)
WO (1) WO2020027083A1 (en)

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020177629A1 (en) * 2019-03-01 2020-09-10 劲方医药科技(上海)有限公司 Spiro-substituted pyrimidine-fused cyclic compound, preparation method therefor and medical use thereof
BR112022010267A2 (en) * 2019-12-27 2022-08-16 Wigen Biomedicine Tech Shanghai Co Ltd COMPOUND, PHARMACEUTICAL COMPOSITION, AND USE OF COMPOUNDS
CN116390728B (en) * 2020-09-27 2024-03-29 微境生物医药科技(上海)有限公司 Quinazoline derivative, preparation method and application thereof
WO2022133345A1 (en) 2020-12-18 2022-06-23 Erasca, Inc. Tricyclic pyridones and pyrimidones
US20220331324A1 (en) * 2021-04-08 2022-10-20 Mirati Therapeutics, Inc. Combination Therapies Using PRMT5 Inhibitors for the Treatment of Cancer
WO2022266206A1 (en) 2021-06-16 2022-12-22 Erasca, Inc. Kras inhibitor conjugates
EP4389751A1 (en) 2021-09-03 2024-06-26 Kumquat Biosciences Inc. Heterocyclic compounds and uses thereof
WO2023205701A1 (en) 2022-04-20 2023-10-26 Kumquat Biosciences Inc. Macrocyclic heterocycles and uses thereof
WO2024022471A1 (en) * 2022-07-28 2024-02-01 上海湃隆生物科技有限公司 Kras inhibitor compound

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106488910B (en) * 2013-10-10 2020-07-31 亚瑞克西斯制药公司 Inhibitors of KRAS G12C
AU2016245864C1 (en) * 2015-04-10 2021-09-09 Araxes Pharma Llc Substituted quinazoline compounds and methods of use thereof
WO2017087528A1 (en) * 2015-11-16 2017-05-26 Araxes Pharma Llc 2-substituted quinazoline compounds comprising a substituted heterocyclic group and methods of use thereof
JOP20190186A1 (en) * 2017-02-02 2019-08-01 Astellas Pharma Inc Quinazoline compound

Also Published As

Publication number Publication date
WO2020027083A1 (en) 2020-02-06
JP2021176819A (en) 2021-11-11

Similar Documents

Publication Publication Date Title
TW202019426A (en) Pharmaceutical composition comprising quinazoline compound as active ingredient
JP6587116B2 (en) Quinazoline compounds
JP7072519B2 (en) BET proteolytic agent
US10238652B2 (en) Compounds for treatment of cancer
TW202019427A (en) Pharmaceutical composition comprising quinazoline compound as active ingredient
JP6911031B2 (en) Heterocyclic compounds as immunomodulators
JP6139782B2 (en) Substituted pyrazolopyrimidine compounds, and pharmaceutically acceptable salts thereof, and solvates, stereoisomers, and tautomers thereof, and pharmaceutical compositions containing them
JP2022519772A (en) Cyclin-dependent kinase 2 biomarker and its use
CN108341813B (en) Substituted 1- (isoxazol-3-yl) -3- (3-fluoro-4-phenyl) urea derivative and preparation method and application thereof
WO2015158310A1 (en) Tyrosine kinase inhibitor and uses thereof
KR20220133258A (en) Compounds and uses thereof
JP6609308B2 (en) Substitutional macrocycles as kinase inhibitors
TW202344253A (en) Erbb receptor inhibitors
JP2018508563A (en) USP7 inhibitor compounds and methods of use
WO2013041038A1 (en) Pyridine compounds as inhibitors of kinase
US20220135592A1 (en) Substituted macrocycles useful as kinase inhibitors
JP2020529465A (en) Substituted pyrazolopyrimidine useful as a kinase inhibitor
TW202340209A (en) Annulated 2-amino-3-cyano thiophenes and derivatives for the treatment of cancer
WO2020238179A1 (en) Pyrrolopyrimidine compound as selective jak2 inhibitor, synthesis method therefor and use thereof
EA028545B1 (en) Bicyclic nitrogen-containing aromatic heterocyclic amide compound
KR102502046B1 (en) CXCR7 Receptor Agonist (3S,4S)-1-Cyclopropylmethyl-4-{[5-(2,4-difluoro-phenyl)-isoxazole-3-carbonyl]-amino}-piperidine- Crystalline form of 3-carboxylic acid (1-pyrimidin-2-yl-cyclopropyl)-amide
WO2020232401A1 (en) Combination therapies with ire1 small molecule inhibitors
JP7036332B2 (en) Quinoline or quinazoline compounds and their applications
TW202028195A (en) Compound as TGF-[beta] R1 inhibitor and application thereof
CN117304157A (en) Chromene derivatives as inhibitors of TCR-NCK interactions