TW202416963A - Egfr inhibitors and uses thereof - Google Patents

Egfr inhibitors and uses thereof Download PDF

Info

Publication number
TW202416963A
TW202416963A TW112129923A TW112129923A TW202416963A TW 202416963 A TW202416963 A TW 202416963A TW 112129923 A TW112129923 A TW 112129923A TW 112129923 A TW112129923 A TW 112129923A TW 202416963 A TW202416963 A TW 202416963A
Authority
TW
Taiwan
Prior art keywords
group
alkyl
compound
pharmaceutically acceptable
acceptable salt
Prior art date
Application number
TW112129923A
Other languages
Chinese (zh)
Inventor
慶北 曾
漢忠 徐
楊振帆
小林 張
Original Assignee
大陸商迪哲(江蘇)醫藥股份有限公司
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 大陸商迪哲(江蘇)醫藥股份有限公司 filed Critical 大陸商迪哲(江蘇)醫藥股份有限公司
Publication of TW202416963A publication Critical patent/TW202416963A/en

Links

Abstract

Disclosed herein are compounds or pharmaceutically acceptable salts thereof that are useful as EGFR inhibitors. Also disclosed are pharmaceutical compositions comprising such compounds, and methods of using such compounds or compositions to treat EGFR-related disorder (e.g., cancers).

Description

EGFR抑制劑及其用途EGFR inhibitor and its use

本公開總體上涉及抑制表皮生長因數受體(EGFR)的新型化合物和其藥學上可接受的鹽。本公開還涉及包含所述化合物作為活性成分的藥物組合物以及所述化合物在治療EGFR相關病症(包括癌症)中的用途。The present disclosure generally relates to novel compounds and pharmaceutically acceptable salts thereof that inhibit epidermal growth factor receptor (EGFR). The present disclosure also relates to pharmaceutical compositions comprising the compounds as active ingredients and the use of the compounds in treating EGFR-related diseases (including cancer).

表皮生長因數受體(EGFR)是一種跨膜蛋白,其是胞外蛋白配體的表皮生長因數家族(EGF家族)成員的受體。受體酪氨酸激酶EGFR家族調節細胞增殖、存活、黏附、遷移和分化。EGFR活性的抑制已證明在廣泛的病理條件下具有潛在的治療適用性。某些癌症的特徵在於EGFR突變,其導致細胞增殖增加。 目前可用的EGFR抑制劑包括例如作為第1代EGFR抑制劑的吉非替尼(gefitinib)和埃羅替尼(erlotinib)以及作為第2代共價EGFR抑制劑的阿法替尼(afatinib)。最近,已報告如WZ4002等第3代保留野生型EGFR抑制劑和第4代EGFR抑制劑的開發。 仍然需要抑制EGFR的新型化合物,其可以用作藥理學工具並且作為用於治療如癌症等EGFR相關病症的藥物具有相當大的關注。 Epidermal growth factor receptor (EGFR) is a transmembrane protein that is a receptor for members of the epidermal growth factor family (EGF family) of extracellular protein ligands. The EGFR family of receptor tyrosine kinases regulates cell proliferation, survival, adhesion, migration, and differentiation. Inhibition of EGFR activity has demonstrated potential therapeutic applicability in a wide range of pathological conditions. Certain cancers are characterized by EGFR mutations that result in increased cell proliferation. Currently available EGFR inhibitors include, for example, gefitinib and erlotinib as first-generation EGFR inhibitors and afatinib as a second-generation covalent EGFR inhibitor. Recently, the development of third-generation wild-type-retaining EGFR inhibitors such as WZ4002 and fourth-generation EGFR inhibitors has been reported. There remains a need for novel compounds that inhibit EGFR, which can be used as pharmacological tools and are of considerable interest as drugs for the treatment of EGFR-related disorders such as cancer.

本文公開了能夠抑制EGFR的新型化合物。因此,本公開的化合物可用於治療如癌症等EGFR相關疾病。 一方面,本公開提供了一種式(I)的化合物: (I) 或其藥學上可接受的鹽,其中: 環A選自由以下組成的組:環烷基、雜環基、芳基和雜芳基; 環B選自由以下組成的組:環烷基、雜環基、芳基和雜芳基; L 1選自由以下組成的組:鍵、烷基、烯基、炔基、雜烷基、雜烯基和雜炔基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基和所述雜炔基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基和氨基; L 2選自由以下組成的組:鍵、N(R A)、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個R B取代; 其中R A選自由以下組成的組:氫、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基、烷基和鹵代烷基; 其中每個R B獨立地選自由以下組成的組:羥基、鹵素、氰基、氨基、烷基、烯基、炔基、雜烷基、雜烯基、環烷基、雜環基、芳基和雜芳基; L 3選自由以下組成的組:鍵、烷基、烯基、炔基、雜烷基、雜烯基和雜炔基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基和所述雜炔基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基和氨基; L 4選自O、S或N(R C); 其中R C選自由以下組成的組:氫、烷基、烯基、炔基、鹵代烷基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基; 每個R 1獨立地選自由以下組成的組:羥基、鹵素、氰基、氨基、-N(R D) 2、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個R E取代; 其中每個R D獨立地選自由以下組成的組:氫、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、  -N(R F) 2或-OR G; 其中每個R E獨立地選自由以下組成的組:氫、羥基、鹵素、氰基、氨基、-N(R F) 2、-烷基-N(R F) 2、-C(O)OR G、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基和烷基; 其中R F和R G中的每一個獨立地選自由以下組成的組:烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基; 每個R 2獨立地選自由以下組成的組:氫、羥基、鹵素、氰基、氨基、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基、烷基和鹵代烷基; R 3選自由以下組成的組:氫、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基和雜環基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基和所述雜環基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基、烷基和鹵代烷基; m是0至5的整數;並且 n是0至4的整數。 另一方面,本公開提供了一種化合物或其藥學上可接受的鹽,其中所述化合物選自由以下組成的組: 。 另一方面,本公開提供了一種式(II)的化合物: (II) 或其藥學上可接受的鹽,其中: 環A 1是7-12元環烷基、7-12元雜環基、7-12元芳基或7-12元雜芳基; 環B選自由以下組成的組:環烷基、雜環基、芳基和雜芳基; L 1選自由以下組成的組:鍵、烷基、烯基、炔基、雜烷基、雜烯基和雜炔基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基和所述雜炔基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基和氨基; L 2選自由以下組成的組:鍵、N(R A)、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個R B取代; 其中R A選自由以下組成的組:氫、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基、烷基和鹵代烷基; 其中每個R B獨立地選自由以下組成的組:羥基、鹵素、氰基、氨基、烷基、烯基、炔基、雜烷基、雜烯基、環烷基、雜環基、芳基和雜芳基; L 3選自由以下組成的組:鍵、烷基、烯基、炔基、雜烷基、雜烯基和雜炔基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基和所述雜炔基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基和氨基; L 4選自O、S或N(R C); 其中R C選自由以下組成的組:氫、烷基、烯基、炔基、鹵代烷基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基; 每個R 1獨立地選自由以下組成的組:羥基、鹵素、氰基、氨基、-N(R D) 2、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個R E取代; 其中每個R D獨立地選自由以下組成的組:氫、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、  -N(R F) 2或-OR G; 其中每個R E獨立地選自由以下組成的組:氫、羥基、鹵素、氰基、氨基、-N(R F) 2、-烷基-N(R F) 2、-C(O)OR G、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基和烷基; 其中R F和R G中的每一個獨立地選自由以下組成的組:烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基; 每個R 2獨立地選自由以下組成的組:氫、羥基、鹵素、氰基、氨基、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基、烷基和鹵代烷基; 每個R 3選自由以下組成的組:氫、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基和雜環基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基和所述雜環基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基、烷基和鹵代烷基; m是0至5的整數; n是0至4的整數;並且 p是0至3的整數。 另一方面,本公開提供了一種式(III)的化合物 (III) 或其藥學上可接受的鹽,其中: 環A選自由以下組成的組:環烷基、雜環基、芳基和雜芳基; 環B 1是7-12元環烷基、7-12元雜環基、7-12元芳基或7-12元雜芳基; L 1選自由以下組成的組:鍵、烷基、烯基、炔基、雜烷基、雜烯基和雜炔基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基和所述雜炔基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基和氨基; L 2選自由以下組成的組:鍵、N(R A)、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個R B取代; 其中R A選自由以下組成的組:氫、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基、烷基和鹵代烷基; 其中每個R B獨立地選自由以下組成的組:羥基、鹵素、氰基、氨基、烷基、烯基、炔基、雜烷基、雜烯基、環烷基、雜環基、芳基和雜芳基; L 3選自由以下組成的組:鍵、烷基、烯基、炔基、雜烷基、雜烯基和雜炔基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基和所述雜炔基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基和氨基; L 4選自O、S或N(R C); 其中R C選自由以下組成的組:氫、烷基、烯基、炔基、鹵代烷基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基; 每個R 1獨立地選自由以下組成的組:羥基、鹵素、氰基、氨基、-N(R D) 2、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個R E取代; 其中每個R D獨立地選自由以下組成的組:氫、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、  -N(R F) 2或-OR G; 其中每個R E獨立地選自由以下組成的組:氫、羥基、鹵素、氰基、氨基、-N(R F) 2、-烷基-N(R F) 2、-C(O)OR G、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基和烷基; 其中R F和R G中的每一個獨立地選自由以下組成的組:烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基; 每個R 2獨立地選自由以下組成的組:氫、羥基、鹵素、氰基、氨基、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基、烷基和鹵代烷基; 每個R 3選自由以下組成的組:氫、鹵素、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基和雜環基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基和所述雜環基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基、烷基和鹵代烷基; m是0至5的整數; n是0至4的整數;並且 p是0至3的整數。 另一方面,本公開提供了一種式(IV)的化合物: (IV) 或其藥學上可接受的鹽,其中: 環A選自由以下組成的組:環烷基、雜環基、芳基和雜芳基; 環B選自由以下組成的組:環烷基、雜環基、芳基和雜芳基; L 1選自由以下組成的組:鍵、烷基、烯基、炔基、雜烷基、雜烯基和雜炔基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基和所述雜炔基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基和氨基; L 21是7-12元環烷基或7-12元雜環基,其中所述環烷基和雜環基任選地被一個或多個R B取代; 其中每個R B獨立地選自由以下組成的組:羥基、鹵素、氰基、氨基、烷基、烯基、炔基、雜烷基、雜烯基、環烷基、雜環基、芳基和雜芳基; L 3選自由以下組成的組:鍵、烷基、烯基、炔基、雜烷基、雜烯基和雜炔基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基和所述雜炔基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基和氨基; L 4選自O、S或N(R C); 其中R C選自由以下組成的組:氫、烷基、烯基、炔基、鹵代烷基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基; 每個R 1獨立地選自由以下組成的組:羥基、鹵素、氰基、氨基、-N(R D) 2、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個R E取代; 其中每個R D獨立地選自由以下組成的組:氫、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、  -N(R F) 2或-OR G; 其中每個R E獨立地選自由以下組成的組:氫、羥基、鹵素、氰基、氨基、-N(R F) 2、-烷基-N(R F) 2、-C(O)OR G、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基和烷基; 其中R F和R G中的每一個獨立地選自由以下組成的組:烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基; 每個R 2獨立地選自由以下組成的組:氫、羥基、鹵素、氰基、氨基、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基、烷基和鹵代烷基; R 3選自由以下組成的組:氫、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基和雜環基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基和所述雜環基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基、烷基和鹵代烷基; m是0至5的整數; n是0至4的整數;並且 p是0至3的整數。 另一方面,本公開提供了一種化合物或其藥學上可接受的鹽,其中所述化合物選自由以下組成的組:       另一方面,本公開提供了一種藥物組合物,所述藥物組合物包含本公開的化合物或其藥學上可接受的鹽以及藥學上可接受的載體。 在另外的方面,本公開提供了一種用於抑制有需要的受試者體內的EGFR活性的方法,所述方法包括向所述受試者施用有效量的本公開的化合物或其藥學上可接受的鹽或本公開的藥物組合物。 在另外的方面,本公開提供了一種用於治療EGFR相關病症的方法,所述方法包括向有需要的受試者施用有效量的本公開的化合物或其藥學上可接受的鹽或本公開的藥物組合物。 另一方面,本公開提供了本公開的化合物或其藥學上可接受的鹽或本公開的藥物組合物在製備用於治療EGFR相關病症的藥物中的用途。 另一方面,本公開提供了用於治療EGFR相關病症的本公開的化合物或其藥學上可接受的鹽或本公開的藥物組合物。 Disclosed herein are novel compounds capable of inhibiting EGFR. Therefore, the compounds disclosed herein can be used to treat EGFR-related diseases such as cancer. In one aspect, the disclosure provides a compound of formula (I): (I) or a pharmaceutically acceptable salt thereof, wherein: Ring A is selected from the group consisting of cycloalkyl, heterocyclic, aryl and heteroaryl; Ring B is selected from the group consisting of cycloalkyl, heterocyclic, aryl and heteroaryl; L1 is selected from the group consisting of a bond, an alkyl, an alkenyl, an alkynyl, a heteroalkyl, a heteroalkenyl and a heteroalkynyl, wherein the alkyl, the alkenyl, the alkynyl, the heteroalkyl, the heteroalkenyl and the heteroalkynyl are optionally substituted with one or more groups independently selected from the group consisting of a hydroxyl, a halogen, a cyano and an amino; L2 is selected from the group consisting of a bond, N(R A ), alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more RB ; wherein R A is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the group consisting of hydroxyl, halogen, cyano, amino, alkyl and halogenated alkyl; wherein each RB is independently selected from the group consisting of hydroxyl, halogen, cyano, amino, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, cycloalkyl, heterocyclo, aryl and heteroaryl; L wherein L is selected from the group consisting of: a bond, an alkyl, an alkenyl, an alkynyl, a heteroalkyl, a heteroalkenyl, and a heteroalkynyl, wherein the alkyl, the alkenyl, the alkynyl, the heteroalkyl, the heteroalkenyl, and the heteroalkynyl are optionally substituted with one or more groups independently selected from the group consisting of: a hydroxyl, a halogen, a cyano, and an amino group; L is selected from O, S, or N(R C ); wherein R C is selected from the group consisting of: hydrogen, an alkyl, an alkenyl, an alkynyl, a halogenated alkyl, a heteroalkyl, a heteroalkenyl, a heteroalkynyl, a cycloalkyl, a heterocyclo, an aryl, and a heteroaryl group; each R 1 is independently selected from the group consisting of: a hydroxyl, a halogen, a cyano, an amino group, -N(R D ) 2 , alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more R E ; wherein each R D is independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the group consisting of hydroxyl, halogen, cyano, -N( RF ) 2 or -ORG ; wherein each RE is independently selected from the group consisting of hydrogen, hydroxyl, halogen, cyano, amino, -N( RF ) 2 , -alkyl-N( RF ) 2 , -C(O) ORG , alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the following: hydroxyl, halogen, cyano, amino and alkyl; wherein each of RF and RG is independently selected from the group consisting of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl; each R R is independently selected from the group consisting of hydrogen, hydroxyl, halogen, cyano, amino, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the group consisting of hydroxyl, halogen, cyano, amino, alkyl and halogenated alkyl; 3 is selected from the group consisting of: hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl and heterocyclo, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl and heterocyclo are optionally substituted with one or more groups independently selected from the group consisting of: hydroxyl, halogen, cyano, amino, alkyl and halogenated alkyl; m is an integer from 0 to 5; and n is an integer from 0 to 4. On the other hand, the present disclosure provides a compound or a pharmaceutically acceptable salt thereof, wherein the compound is selected from the group consisting of: and On the other hand, the present disclosure provides a compound of formula (II): (II) or a pharmaceutically acceptable salt thereof, wherein: Ring A 1 is a 7-12 membered cycloalkyl, a 7-12 membered heterocyclic group, a 7-12 membered aryl or a 7-12 membered heteroaryl; Ring B is selected from the group consisting of a cycloalkyl, a heterocyclic group, an aryl and a heteroaryl; L 1 is selected from the group consisting of a bond, an alkyl, an alkenyl, an alkynyl, a heteroalkyl, a heteroalkenyl and a heteroalkynyl, wherein the alkyl, the alkenyl, the alkynyl, the heteroalkyl, the heteroalkenyl and the heteroalkynyl are optionally substituted with one or more groups independently selected from the group consisting of a hydroxyl, a halogen, a cyano and an amino group; L 2 is selected from the group consisting of a bond, an N(R A ), alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more RB ; wherein R A is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the group consisting of hydroxyl, halogen, cyano, amino, alkyl and halogenated alkyl; wherein each RB is independently selected from the group consisting of hydroxyl, halogen, cyano, amino, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, cycloalkyl, heterocyclo, aryl and heteroaryl; L wherein L is selected from the group consisting of: a bond, an alkyl, an alkenyl, an alkynyl, a heteroalkyl, a heteroalkenyl, and a heteroalkynyl, wherein the alkyl, the alkenyl, the alkynyl, the heteroalkyl, the heteroalkenyl, and the heteroalkynyl are optionally substituted with one or more groups independently selected from the group consisting of: a hydroxyl, a halogen, a cyano, and an amino group; L is selected from O, S, or N(R C ); wherein R C is selected from the group consisting of: hydrogen, an alkyl, an alkenyl, an alkynyl, a halogenated alkyl, a heteroalkyl, a heteroalkenyl, a heteroalkynyl, a cycloalkyl, a heterocyclo, an aryl, and a heteroaryl group; each R 1 is independently selected from the group consisting of: a hydroxyl, a halogen, a cyano, an amino group, -N(R D ) 2 , alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more R E ; wherein each R D is independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the group consisting of hydroxyl, halogen, cyano, -N( RF ) 2 or -ORG ; wherein each RE is independently selected from the group consisting of hydrogen, hydroxyl, halogen, cyano, amino, -N( RF ) 2 , -alkyl-N( RF ) 2 , -C(O) ORG , alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the following: hydroxyl, halogen, cyano, amino and alkyl; wherein each of RF and RG is independently selected from the group consisting of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl; each R R is independently selected from the group consisting of hydrogen, hydroxyl, halogen, cyano, amino, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the group consisting of hydroxyl, halogen, cyano, amino, alkyl and halogenated alkyl; each R 3 is selected from the group consisting of: hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl and heterocyclo, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl and heterocyclo are optionally substituted with one or more groups independently selected from the following: hydroxyl, halogen, cyano, amino, alkyl and halogenated alkyl; m is an integer from 0 to 5; n is an integer from 0 to 4; and p is an integer from 0 to 3. On the other hand, the present disclosure provides a compound of formula (III) (III) or a pharmaceutically acceptable salt thereof, wherein: Ring A is selected from the group consisting of cycloalkyl, heterocyclic, aryl and heteroaryl; Ring B is 7-12 membered cycloalkyl, 7-12 membered heterocyclic, 7-12 membered aryl or 7-12 membered heteroaryl; L is selected from the group consisting of a bond, an alkyl, an alkenyl, an alkynyl, a heteroalkyl, a heteroalkenyl and a heteroalkynyl, wherein the alkyl, the alkenyl, the alkynyl, the heteroalkyl, the heteroalkenyl and the heteroalkynyl are optionally substituted with one or more groups independently selected from the group consisting of a hydroxyl, a halogen, a cyano and an amino; L is selected from the group consisting of a bond, N(R A ), alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more RB ; wherein R A is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the group consisting of hydroxyl, halogen, cyano, amino, alkyl and halogenated alkyl; wherein each RB is independently selected from the group consisting of hydroxyl, halogen, cyano, amino, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, cycloalkyl, heterocyclo, aryl and heteroaryl; L wherein L is selected from the group consisting of: a bond, an alkyl, an alkenyl, an alkynyl, a heteroalkyl, a heteroalkenyl, and a heteroalkynyl, wherein the alkyl, the alkenyl, the alkynyl, the heteroalkyl, the heteroalkenyl, and the heteroalkynyl are optionally substituted with one or more groups independently selected from the group consisting of: a hydroxyl, a halogen, a cyano, and an amino group; L is selected from O, S, or N(R C ); wherein R C is selected from the group consisting of: hydrogen, an alkyl, an alkenyl, an alkynyl, a halogenated alkyl, a heteroalkyl, a heteroalkenyl, a heteroalkynyl, a cycloalkyl, a heterocyclo, an aryl, and a heteroaryl group; each R 1 is independently selected from the group consisting of: a hydroxyl, a halogen, a cyano, an amino group, -N(R D ) 2 , alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more R E ; wherein each R D is independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the group consisting of hydroxyl, halogen, cyano, -N( RF ) 2 or -ORG ; wherein each RE is independently selected from the group consisting of hydrogen, hydroxyl, halogen, cyano, amino, -N( RF ) 2 , -alkyl-N( RF ) 2 , -C(O) ORG , alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the following: hydroxyl, halogen, cyano, amino and alkyl; wherein each of RF and RG is independently selected from the group consisting of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl; each R R is independently selected from the group consisting of hydrogen, hydroxyl, halogen, cyano, amino, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the group consisting of hydroxyl, halogen, cyano, amino, alkyl and halogenated alkyl; each R 3 is selected from the group consisting of: hydrogen, halogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl and heterocyclo, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl and heterocyclo are optionally substituted with one or more groups independently selected from the following: hydroxyl, halogen, cyano, amino, alkyl and halogenated alkyl; m is an integer from 0 to 5; n is an integer from 0 to 4; and p is an integer from 0 to 3. On the other hand, the disclosure provides a compound of formula (IV): (IV) or a pharmaceutically acceptable salt thereof, wherein: Ring A is selected from the group consisting of cycloalkyl, heterocyclic, aryl and heteroaryl; Ring B is selected from the group consisting of cycloalkyl, heterocyclic, aryl and heteroaryl; L 1 is selected from the group consisting of a bond, an alkyl, an alkenyl, an alkynyl, a heteroalkyl, a heteroalkenyl and a heteroalkynyl, wherein the alkyl, the alkenyl, the alkynyl, the heteroalkyl, the heteroalkenyl and the heteroalkynyl are optionally substituted with one or more groups independently selected from the following: hydroxyl, halogen, cyano and amino; L 21 is a 7-12 membered cycloalkyl or a 7-12 membered heterocyclic, wherein the cycloalkyl and heterocyclic are optionally substituted with one or more RB ; wherein each R B is independently selected from the group consisting of: hydroxyl, halogen, cyano, amino, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, cycloalkyl, heterocyclo, aryl and heteroaryl; L3 is selected from the group consisting of: bond, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl and heteroalkynyl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl and heteroalkynyl are optionally substituted with one or more groups independently selected from the group consisting of: hydroxyl, halogen, cyano and amino; L4 is selected from O, S or N (R C ); wherein R C is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, halogenated alkyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl; each R 1 is independently selected from the group consisting of hydroxyl, halogen, cyano, amino, -N( RD ) 2 , alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more RE ; wherein each R D is independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the group consisting of hydroxyl, halogen, cyano, -N( RF ) 2 or -ORG ; wherein each RE is independently selected from the group consisting of hydrogen, hydroxyl, halogen, cyano, amino, -N( RF ) 2 , -alkyl-N( RF ) 2 , -C(O) ORG , alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the following: hydroxyl, halogen, cyano, amino and alkyl; wherein each of RF and RG is independently selected from the group consisting of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl; each R R is independently selected from the group consisting of hydrogen, hydroxyl, halogen, cyano, amino, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the group consisting of hydroxyl, halogen, cyano, amino, alkyl and halogenated alkyl; 3 is selected from the group consisting of: hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl and heterocyclo, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl and heterocyclo are optionally substituted with one or more groups independently selected from the group consisting of: hydroxyl, halogen, cyano, amino, alkyl and halogenated alkyl; m is an integer from 0 to 5; n is an integer from 0 to 4; and p is an integer from 0 to 3. On the other hand, the present disclosure provides a compound or a pharmaceutically acceptable salt thereof, wherein the compound is selected from the group consisting of: , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , and . On the other hand, the disclosure provides a kind of pharmaceutical composition, the pharmaceutical composition comprises the compound disclosed herein or its pharmaceutically acceptable salt and a pharmaceutically acceptable carrier. In other aspects, the disclosure provides a kind of method for suppressing the EGFR activity in the subject in need, the method comprising applying an effective amount of the compound disclosed herein or its pharmaceutically acceptable salt or the pharmaceutical composition disclosed herein to the subject. In other aspects, the disclosure provides a kind of method for treating EGFR related conditions, the method comprising applying an effective amount of the compound disclosed herein or its pharmaceutically acceptable salt or the pharmaceutical composition disclosed herein to the subject in need. On the other hand, the disclosure provides the use of the compound disclosed herein or its pharmaceutically acceptable salt or the pharmaceutical composition disclosed herein in the preparation of a medicine for treating EGFR related conditions. In another aspect, the present disclosure provides a compound or a pharmaceutically acceptable salt thereof or a pharmaceutical composition of the present disclosure for treating EGFR-related diseases.

現在將詳細參考本公開的某些實施方式,在所附結構和式中展示了所述實施方式的實施例。雖然將結合所列舉的實施方式描述本公開,但應理解,所述實施方式不旨在將本公開限於那些實施方式。相反,本公開旨在涵蓋所有替代方案、修改和等效物,所述替代方案、修改和等效物可以包括在如權利要求書所定義的本公開的範圍內。本領域技術人員將認識到與本文所描述的方法和材料類似或等效的可以在實踐本公開時使用的許多方法和材料。本公開決不限於所描述的方法和材料。在所併入的參考文獻和類似材料中的一個或多個(包括但不限於所定義的術語、術語用法、所描述的技術等)與本申請不同或相矛盾的情況下,以本公開為準。本公開中所引用的所有參考文獻、專利、專利申請均特此通過引用整體併入。 應理解,為清楚起見而在單獨實施方式的上下文中描述的本公開的某些特徵也可以在單個實施方式中組合提供。相反,為簡潔起見而在單個實施方式的上下文中描述的本公開的各種特徵也可以單獨提供或以任何合適的子組合提供。必須指出,除非上下文另外明確指示,否則如在說明書和所附權利要求中所使用的,單數形式“一個/一種(a或an)”和“所述(the)”包括其複數形式。因此,例如,對“一種化合物”的提及包括多種化合物。 定義 下文更詳細地描述了具體官能團和化學術語的定義。出於本公開的目的,化學元素是根據元素週期表(Periodic Table of the Elements), CAS版, 《物理和化學手冊(Handbook of Chemistry and Physics)》, 第75版, 內封面來標識的,並且具體的官能團總體上是如本文所描述那樣定義的。另外,有機化學的一般原理以及特定的功能部分和反應性描述於以下文獻中:《有機化學(Organic Chemistry)》, Thomas Sorrell, 索薩利托科學書籍出版社(University Science Books, Sausalito), 1999;Smith和March, 《三月先進有機化學(March's Advanced Organic Chemistry)》, 第5版, 紐約的約翰威利父子出版公司(John Wiley & Sons, Inc., New York), 2001;LaEGFR, 《複雜有機轉換(Comprehensive Organic Transformations)》, 紐約的VCH出版社有限公司(VCH Publishers, Inc., New York), 1989;Carruthers, 《一些現代有機合成方法(Some Modern Methods of Organic Synthesis)》, 第3版, 劍橋的劍橋大學出版社(Cambridge University Press, Cambridge), 1987;上述文獻中的每個文獻的全部內容通過引用併入本文。 在本公開的各處,描述了連接取代基。在結構明確需要連接基團的情況下,關於所述基團所列的馬庫什變數(Markush variable)應理解為連接基團。例如,如果結構需要連接基團且所述變數的馬庫什組定義列出“烷基”,則應理解“烷基”表示連接亞烷基。 在任何變數(例如,R i)在化合物的任何構成或式中出現多於一次時,其在每次出現時的定義獨立於其在其它每次出現時的定義。因此,例如,如果顯示基團被0至2個R i部分取代,則所述基團可以任選地被至多兩個R i部分取代,並且R i在每次出現時各自獨立地選自R i的定義。而且,取代基和/或變數的組合是允許的,但僅在此類組合產生穩定化合物時才允許。 如本文所使用的,為了方便起見,在化學基團的前面或末端使用破折號“-”來表示取代基的連接點。例如,-OH通過氧原子連接;化學基團可以在不丟失其一般含義的情況下用一個或多個破折號或不用一個或多個破折號描繪。通過結構中的線繪製的波浪線指示基團的連接點。除非化學上或結構上需要,否則方向性不指示或暗示化學基團所書寫或命名的順序。如本文所使用的,從環中心出來的實線指示環上取代基的連接點可以在任何環原子處。在列出取代基,但未指定此類取代基通過哪個原子與給定式的化合物的剩餘部分鍵合時,此類取代基可以通過此式中的任何原子鍵合。取代基和/或變數的組合是允許的,但僅在此類組合產生穩定化合物時才允許。 除非在本文中另外指示,否則對數值範圍的敘述僅旨在充當個別提及屬於所述範圍的每一單獨值的速記方法,並且每一單獨值併入到本說明書中,如同在本文中單獨地敘述一般。除非另有說明,否則本文所使用的範圍包括所述範圍的兩個極限。例如,表達“n是介於1與6之間的整數”和“n是1至6的整數”均意指“n是1、2、3、4、5或6”。 如本文所使用的,術語“本文所提供的化合物”、或“本文所公開的化合物”或“本公開的化合物”是指式(I)、(II)、(III)和(IV)的化合物以及本文所公開的特定化合物。 如本文所使用的,術語“C i-j”指示碳原子數量的範圍,其中i和j是整數,並且碳原子數量的範圍包括端點(即,i和j)和其間的每個整數點,並且其中j大於i。例如,C 1-6指示一至六個碳原子的範圍,包括一個碳原子、兩個碳原子、三個碳原子、四個碳原子、五個碳原子和六個碳原子。在一些實施方式中,術語“C 1-12”指示1至12個碳原子,尤其是1至10個碳原子,尤其是1至8個碳原子,尤其是1至6個碳原子,尤其是1至5個碳原子,尤其是1至4個碳原子,尤其是1至3個碳原子或尤其是1至2個碳原子。 如本文所使用的,術語“烷基”,不論作為另一個術語的一部分還是獨立地使用,是指可以任選地獨立地被一個或多個下文所描述的取代基取代的飽和直鏈或支鏈烴基。術語“C i-j烷基”是指具有i至j個碳原子的烷基。在一些實施方式中,烷基含有1至10個碳原子。在一些實施方式中,烷基含有1至9個碳原子。在一些實施方式中,烷基含有1至8個碳原子、1至7個碳原子、1至6個碳原子、1至5個碳原子、1至4個碳原子、1至3個碳原子或1至2個碳原子。“C 1-10烷基”的實例包括但不限於甲基、乙基、丙基、丁基、戊基、己基、庚基、辛基、壬基和癸基。“C 1-6烷基”的實例為甲基、乙基、丙基、異丙基、正丁基、異丁基、仲丁基、叔丁基、正戊基、2-戊基、3-戊基、2-甲基-2-丁基、3-甲基-2-丁基、3-甲基-1-丁基、2-甲基-1-丁基、1-己基、2-己基、3-己基、2-甲基-2-戊基、3-甲基-2-戊基、4-甲基-2-戊基、3-甲基-3-戊基、2-甲基-3-戊基、2,3-二甲基-2-丁基、3,3-二甲基-2-丁基等。 如本文所使用的,術語“烯基”,不論作為另一個術語的一部分還是獨立地使用,是指可以任選地獨立地被一個或多個本文所描述的取代基取代的具有至少一個碳-碳雙鍵的直鏈或支鏈烴基並且包括具有“順式”取向和“反式”取向或可替代地“E”取向和“Z”取向的基團。在一些實施方式中,烯基含有2至12個碳原子。在一些實施方式中,烯基含有2至11個碳原子。在一些實施方式中,烯基含有2至11個碳原子、2至10個碳原子、2至9個碳原子、2至8個碳原子、2至7個碳原子、2至6個碳原子、2至5個碳原子、2至4個碳原子、2至3個碳原子,並且在一些實施方式中,烯基包括2個碳原子。烯基的實例包括但不限於乙烯基(ethylenyl或vinyl)、丙烯基(烯丙基)、丁烯基、戊烯基、1-甲基-2-丁烯-1-基、5-己烯基等。 如本文所使用的,術語“炔基”,不論作為另一個術語的一部分還是獨立地使用,是指可以任選地獨立地被一個或多個本文所描述的取代基取代的具有至少一個碳-碳三鍵的直鏈或支鏈烴基。在一些實施方式中,烯基含有2至12個碳原子。在一些實施方式中,炔基含有2至11個碳原子。在一些實施方式中,炔基含有2至11個碳原子、2至10個碳原子、2至9個碳原子、2至8個碳原子、2至7個碳原子、2至6個碳原子、2至5個碳原子、2至4個碳原子、2至3個碳原子,並且在一些實施方式中,炔基包括2個碳原子。炔基的實例包括但不限於乙炔基、1-丙炔基、2-丙炔基等。 如本文所使用的,術語“氨基”是指-NH 2基團。氨基還可以被一個或多個如烷基、烯基、炔基、芳基、羰基或其它氨基等基團取代。 如本文所使用的,術語“芳基”,不論作為另一術語的一部分還是獨立地使用,是指具有總共5至20個環成員的單環和多環體系,其中體系中的至少一個環是芳香族的,並且其中體系中的每個環包括3至12個環成員。“芳基”的實例包括但不限於可以帶有一個或多個取代基的苯基、聯苯基、萘基、蒽基等。如本文所使用的,術語“芳基”的範圍還包括芳香族環與一個或多個另外的環稠合的基團。在多環體系的情況下,僅需要一個環是芳香族的(例如,2,3-二氫吲哚),但所有環都可以是芳香族的(例如,喹啉)。第二環也可以是稠合的或橋連的。多環芳基的實例包括但不限於苯並呋喃基、茚滿基、鄰苯二甲醯亞胺基、萘二甲醯亞胺基、菲啶基或四氫萘基等。芳基可以在一個或多個環位置處被如上文所描述的取代基取代。 如本文所使用的,術語“氰基”是指-CN。 如本文所使用的,術語“環烷基”,不論作為另一個術語的一部分還是獨立地使用,是指單價非芳香族飽和或部分不飽和單環和多環體系,其中所有環原子均為碳,並且所述體系包括至少三個成環碳原子。在一些實施方式中,環烷基可以含有3至12個成環碳原子、3至10個成環碳原子、3至9個成環碳原子、3至8個成環碳原子、3至7個成環碳原子、3至6個成環碳原子、3至5個成環碳原子、4至12個成環碳原子、4至10個成環碳原子、4至9個成環碳原子、4至8個成環碳原子、4至7個成環碳原子、4至6個成環碳原子、4至5個成環碳原子。環烷基可以為飽和的或部分不飽和的。環烷基可以為經取代的。在一些實施方式中,環烷基可以為飽和環狀烷基。在一些實施方式中,環烷基可以為在其環體系中包括至少一個雙鍵或三鍵的部分不飽和環狀烷基。在一些實施方式中,環烷基可以為單環的或多環的。在多環體系的情況下,稠合、螺和橋連環體系包括在本定義的範圍內。單環環烷基的實例包括但不限於環丙基、環丁基、環戊基、1-環戊-1-烯基、1-環戊-2-烯基、1-環戊-3-烯基、環己基、1-環己-1-烯基、1-環己-2-烯基、1-環己-3-烯基、環己二烯基、環庚基、環辛基、環壬基、環癸基、環十一烷基和環十二烷基。多環環烷基的實例包括但不限於金剛烷基、降冰片烷基、芴基、螺-戊二烯基、螺[3.6]-癸烷基、雙環[1,1,1]戊烯基、雙環[2,2,1]庚烯基等。 如本文所使用的,術語“鹵素”是指選自氟(fluorine或fluoro)、氯(chlorine或chloro)、溴(bromine或bromo)和碘(iodine或iodo)的原子。 如本文所使用的,術語“雜原子”是指氮、氧、硫、磷,並且包括氮、硫或磷的任何氧化形式以及鹼性氮(包括N-氧化物)的任何季銨化形式。 如本文所使用的,術語“雜烷基”是指其碳原子中的至少一個被選自N、O或S的雜原子替代的烷基。雜烷基可以是碳基或雜原子基(即,雜原子可以出現在基團的中間中或末端處),並且可以任選地獨立地被本文所描述的一個或多個取代基取代。術語“雜烷基”涵蓋烷氧基和雜烷氧基。 如本文所使用的,術語“雜烯基”是指其碳原子中的至少一個被選自N、O或S的雜原子替代的烯基。雜烯基可以為碳基或雜原子基(即,雜原子可以出現在基團的中間中或末端處),並且可以任選地獨立地被本文所描述的一個或多個取代基取代。 如本文所使用的,術語“雜炔基”是指其碳原子中的至少一個被選自N、O或S的雜原子替代的炔基。雜炔基可以為碳基或雜原子基(即,雜原子可以出現在基團的中間中或末端處),並且可以任選地獨立地被本文所描述的一個或多個取代基取代。 如本文所使用的,術語“雜芳基”,不論作為另一個術語的一部分還是獨立地使用,是指除碳原子之外還具有一個或多個雜原子的芳基。雜芳基可以為單環的。單環雜芳基的實例包括但不限於噻吩基、呋喃基、吡咯基、咪唑基、吡唑基、三唑基、四唑基、噁唑基、異噁唑基、噁二唑基、噻唑基、異噻唑基、噻二唑基、吡啶基、噠嗪基、嘧啶基、吡嗪基、吲哚嗪基、嘌呤基、萘啶基、苯並呋喃基和蝶啶基。雜芳基還包括雜芳香族環與一個或多個芳基、環烷基或雜環基環稠合的多環基團,其中連接基團或連接點位於雜芳香族環上。多環雜芳基的實例包括但不限於吲哚基、異吲哚基、苯並噻吩基、苯並呋喃基、苯並[1,3]二氧雜環戊烯基、二苯並呋喃基、吲唑基、苯並咪唑基、苯並噻唑基、喹啉基、異喹啉基、二氫喹啉基、二氫異喹啉基、四氫喹啉基、四氫異喹啉基、噌啉基、酞嗪基、喹唑啉基、喹喔啉基、4H-喹嗪基、哢唑基、吖啶基、吩嗪基、吩噻嗪基、吩噁嗪基、四氫喹啉基、四氫異喹啉基等。 如本文所使用的,術語“雜環基”是指飽和或部分不飽和碳環基,其中一個或多個環原子為獨立地選自氧、硫、氮、磷等的雜原子,剩餘環原子為碳,其中一個或多個環原子可以任選地獨立地被一個或多個取代基取代。在一些實施方式中,雜環基為飽和雜環基。在一些實施方式中,雜環基為在其環體系中具有一個或多個雙鍵的部分不飽和雜環基。在一些實施方式中,雜環基可以包括碳、氮、硫或磷的任何氧化形式以及鹼性氮的任何季銨化形式。“雜環基”還包括其中雜環基與飽和、部分不飽和或完全不飽和(即,芳香族)碳環或雜環稠合的基團。在可能的情況下,雜環基可以為碳連接的或氮連接的。在一些實施方式中,雜環為碳連接的。在一些實施方式中,雜環為氮連接的。例如,衍生自吡咯的基團可以為吡咯-1-基(氮連接的)或吡咯-3-基(碳連接的)。進一步地,衍生自咪唑的基團可以為咪唑-1-基(氮連接的)或咪唑-3-基(碳連接的)。 在一些實施方式中,術語“3元至12元雜環基”是指具有1個至3個獨立地選自氮、氧、硫或磷的雜原子的3元至12元飽和或部分不飽和單環或多環雜環體系。在多環體系的情況下,稠合、螺和橋連環體系還包括在本定義的範圍內。單環雜環基的實例包括但不限於氧雜環丁烷基、1,1-二氧代硫雜環丁烷基吡咯烷基、四氫呋喃基、四氫噻吩基、吡咯基、呋喃基、噻吩基、吡唑基、咪唑基、三唑基、噁唑基、噻唑基、哌啶基、哌嗪基、哌啶基、嗎啉基、吡啶基、吡嗪基、嘧啶基、噠嗪基、三嗪基、吡啶酮基、嘧啶酮基、吡嗪酮基、嘧啶酮基、噠嗪酮基、吡咯烷基、三嗪酮基等。稠合雜環基的實例包括但不限於喹啉基、異喹啉基、四氫喹啉基、四氫異喹啉基、喹喔啉基、喹嗪基、喹唑啉基、氮雜吲哚嗪基、蝶啶基、色烯基、異色烯基、吲哚基、異吲哚基、吲哚嗪基、吲唑基、嘌呤基、苯並呋喃基、異苯並呋喃基、苯並咪唑基、苯並噻吩基、苯並噻唑基、哢唑基、吩嗪基、吩噻嗪基、啡啶基、六氫-1H-吡咯裡嗪基、咪唑並[1,2-a]吡啶基、[1,2,4]三唑並[4,3-a]吡啶基、[1,2,3]三唑並[4,3-a]吡啶基、八氫吡咯並[3,4-b]吡咯基、八氫吡咯並[3,4-c]吡咯基等。螺雜環基的實例包括但不限於螺吡喃基、螺噁嗪基、2,6-二氮雜螺[3.3]庚烷基、2,5-二氮雜螺[3.4]辛烷基、2,6-二氮雜螺[3.4]辛烷基、2,7-二氮雜螺[3.5]壬烷基、5-氧雜-2,8-二氮雜螺[3.5]壬烷基、2,7-二氮雜螺[4.4]壬烷基、1,7-二氮雜螺[3.5]壬烷基、2,8-二氮雜螺[4.5]癸烷基、2,8-二氮雜螺[4.5]癸烷基等。橋連雜環基的實例包括但不限於嗎啡烷基、六亞甲基四胺基、3-氮雜-雙環[3.1.0]己烷、3,6-二氮雜雙環[3.1.1]庚烷、2,5-二氮雜雙環[2.2.1]庚烷、8-氮雜-雙環[3.2.1]辛烷、3-氮雜雙環[3.2.1]辛烷、1-氮雜-雙環[2.2.2]辛烷、1,4-二氮雜雙環[2.2.2]辛烷、3-氮雜雙環[3.2.2]壬烷等。 如本文所使用的,術語“羥基(hydroxyl或hydroxy)”是指-OH。 如本文所使用的,術語“部分不飽和的”是指包括至少一個雙鍵或三鍵的基團。術語“部分不飽和的”旨在涵蓋具有多個不飽和位點的環,但並不旨在包括芳香族(即,完全不飽和的)部分。 如本文所使用的,術語“任選的”或“任選地”意味著,隨後描述的事件或情形可能發生或可能不發生,並且所述描述包括其中所述事件或情況發生的實例以及其中所述事件或情況不發生的實例。如本文所使用的,術語“經取代的”,無論前面是否有術語“任選地”,都意味著指定部分的一個或多個氫被合適的取代基替代。應當理解,“取代”或“被…取代”包括隱含前提條件,即此類取代與經取代的原子的允許化合價一致,並且取代產生穩定的或化學上可行的化合物,例如,不會自發地經歷如重排、環化、消除等轉化的化合物。除非另有說明,否則“任選地經取代的”基團可以在基團的每個可取代位置處具有適當的取代基,並且在任何給定結構中的多於一個位置可以被多於一個選自指定組的取代基取代時,在每個位置處,取代基可以相同或不同。取代可以包括但不限於烷基、烯基、炔基、烷氧基、醯基、氨基、醯氨基、脒基、芳基、疊氮基、氨基甲醯基、羧基、羧基酯、氰基、胍基、鹵代、鹵代烷基、雜烷基、雜芳基、雜環基、羥基、肼基、亞氨基、氧代、硝基、烷基亞磺醯基、磺酸、烷基磺醯基、硫氰酸鹽、硫醇、硫酮或其組合。本領域技術人員應理解,如果合適的話,取代基本身可以被取代。除非特別陳述為“未經取代的”,否則對本文中的化學部分的提及應理解為包括經取代的變體。例如,對“芳基”基團或部分的提及隱含地包括經取代的變體和未經取代的變體兩者。 如本文所使用的,術語“經取代的”,無論前面是否有術語“任選地”,都意味著指定部分的一個或多個氫被合適的取代基替代。應當理解,“取代”或“被…取代”包括隱含前提條件,即此類取代與經取代的原子的允許化合價一致,並且取代產生穩定的或化學上可行的化合物,例如,不會自發地經歷如重排、環化、消除等轉化的化合物。除非另有說明,否則“任選地經取代的”基團可以在基團的每個可取代位置處具有適當的取代基,並且在任何給定結構中的多於一個位置可以被多於一個選自指定組的取代基取代時,在每個位置處,取代基可以相同或不同。本領域技術人員應理解,如果合適的話,取代基本身可以被取代。除非特別陳述為“未經取代的”,否則對本文中的化學部分的提及應理解為包括經取代的變體。例如,對“芳基”基團或部分的提及隱含地包括經取代的變體和未經取代的變體兩者。 符號“R”和“S”表示手性碳原子周圍的取代基的構型。異構體描述符“R”和“S”如本文所描述的用於指示相對於核心分子的原子構型,並且旨在如文獻(IUPAC推薦1996, 《純粹與應用化學(Pure and Applied Chemistry)》, 68: 2193-2222 (1996))中所定義的那樣使用。 化合物 一方面,本公開提供了式(I)的化合物: 式(I) 或其藥學上可接受的鹽, 其中, 環A選自由以下組成的組:環烷基、雜環基、芳基和雜芳基; 環B選自由以下組成的組:環烷基、雜環基、芳基和雜芳基; L 1選自由以下組成的組:鍵、烷基、烯基、炔基、雜烷基、雜烯基和雜炔基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基和所述雜炔基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基和氨基; L 2選自由以下組成的組:鍵、N(R A)、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個R B取代; 其中R A選自由以下組成的組:氫、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基、烷基和鹵代烷基; 其中每個R B獨立地選自由以下組成的組:羥基、鹵素、氰基、氨基、烷基、烯基、炔基、雜烷基、雜烯基、環烷基、雜環基、芳基和雜芳基; L 3選自由以下組成的組:鍵、烷基、烯基、炔基、雜烷基、雜烯基和雜炔基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基和所述雜炔基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基和氨基; L 4選自O、S或N(R C); 其中R C選自由以下組成的組:氫、烷基、烯基、炔基、鹵代烷基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基; 每個R 1獨立地選自由以下組成的組:羥基、鹵素、氰基、氨基、-N(R D) 2、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個R E取代; 其中每個R D獨立地選自由以下組成的組:氫、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、  -N(R F) 2或-OR G; 其中每個R E獨立地選自由以下組成的組:氫、羥基、鹵素、氰基、氨基、-N(R F) 2、-烷基-N(R F) 2、-C(O)OR G、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基和烷基; 其中R F和R G中的每一個獨立地選自由以下組成的組:烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基; 每個R 2獨立地選自由以下組成的組:氫、羥基、鹵素、氰基、氨基、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基、烷基和鹵代烷基; R 3選自由以下組成的組:氫、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基和雜環基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基和所述雜環基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基、烷基和鹵代烷基; m是0至5的整數;並且 n是0至4的整數。 在一些實施方式中,環A是芳基。在某些實施方式中,環A是C 6-12芳基、C 6-11芳基、C 6-10芳基、C 6-9芳基或C 6-8芳基。在某些實施方式中,環A是苯基。 在一些實施方式中,環A是雜芳基。在某些實施方式中,環A是5-12元雜芳基、5-11元雜芳基、5-10元雜芳基、5-9元雜芳基、5-8元雜芳基、5-7元雜芳基或5-6元雜芳基。 在某些實施方式中,環A選自由以下組成的組:呋喃基、苯硫基、吡咯基、吡啶基、吡喃基、嘧啶基、噠嗪基、吡嗪基和四氫異喹啉基。 在某些實施方式中,環A選自由以下組成的組: 。 在一些實施方式中,環B是芳基。在某些實施方式中,環B是C 6-12芳基、C 6-11芳基、C 6-10芳基、C 6-9芳基或C 6-8芳基。在某些實施方式中,環B是苯基。 在一些實施方式中,環B是雜芳基。在某些實施方式中,環B是5-12元雜芳基、5-11元雜芳基、5-10元雜芳基、5-9元雜芳基、5-8元雜芳基、5-7元雜芳基或5-6元雜芳基。在某些實施方式中,環B是吡啶基或吡唑基。 在某些實施方式中,環B選自由以下組成的組: 。 在一些實施方式中,L 1是鍵。 在一些實施方式中,L 1是烷基。在某些實施方式中,L 1是C 1-6烷基、C 1-5烷基、C 1-4烷基、C 1-3烷基或C 1-2烷基。 在某些實施方式中,L 1選自由以下組成的組: ,其中L 1的*端與L 2連接。 在一些實施方式中,L 2是鍵。 在一些實施方式中,L 2是N(R A),並且R A選自烷基或雜環基,其中所述烷基或所述雜環基任選地被一個或多個鹵素或烷基取代。在某些實施方式中,L 2是N(R A),並且R A是乙基、二氟乙基、三氟乙基或氧雜環丁烷基。 在一些實施方式中,L 2是任選地被一個或多個R B取代的環烷基。在某些實施方式中,L 2是C 3-10環烷基、C 3-9環烷基、C 3-8環烷基、C 3-7環烷基、C 3-6環烷基或C 3-5環烷基,其各自任選地被一個或多個R B取代。 在某些實施方式中,L 2是任選地被一個或多個R B取代的 。 在一些實施方式中,L 2是任選地被一個或多個R B取代的雜環基。在某些實施方式中,L 2是含有一個或多個選自N、O或S的雜原子的雜環基。在某些實施方式中,L 2是3-12元雜環基、3-11元雜環基、3-10元雜環基或3-9元雜環基,其各自任選地被一個或多個R B取代。 在某些實施方式中,L 2是選自由以下組成的組的雜環基: ,其中的每一個任選地被一個或多個R B取代,其中L 2的*端與L 3連接。 在一些實施方式中,L 2是被一個或多個R B取代的環烷基或雜環基,並且R B是烷基。在某些實施方式中,R B是C 1-6烷基、C 1-5烷基、C 1-4烷基、C 1-3烷基或C 1-2烷基。在某些實施方式中,R B是甲基。 在一些實施方式中,L 2選自由以下組成的組: ,其中L 2的*端與L 3連接。 在一些實施方式中,L 3是烷基。在一些實施方式中,L 3是C 1-6烷基、C 1-5烷基、C 1-4烷基、C 1-3烷基或C 1-2烷基。 在某些實施方式中,L 3是乙基。 在一些實施方式中,L 4是O或NH。 在一些實施方式中,L 1是鍵,並且L 2是鍵或任選地被一個或多個R B取代的雜環基。 在一些實施方式中,L 1是烷基,並且L 2是鍵、N(R A)或任選地被一個或多個R B取代的環烷基。 在一些實施方式中,-L 1-L 2-L 3-L 4-是-烷基-O-。在某些實施方式中,-L 1-L 2-L 3-L 4-是-(C 3-8烷基)-O-。在某些實施方式中,-L 1-L 2-L 3-L 4-是-CH 2CH(CH 3)(CH 2) 3-O-或    -CH 2CH(CH 2CH 3)(CH 2) 3-O-。 在一些實施方式中,-L 1-L 2-L 3-L 4-是-雜環基-烷基-O-,其中所述雜環基任選地被一個或多個R B取代。在某些實施方式中,-L 1-L 2-L 3-L 4-是-(5-10元雜環基)-(C 1-6烷基)-O-,其中所述5-10元雜環基任選地被一個或多個R B取代。在某些實施方式中,-L 1-L 2-L 3-L 4-選自由以下組成的組: 以及 。 在一些實施方式中,-L 1-L 2-L 3-L 4-是-烷基-N(R A)-烷基-O-,其中R A選自烷基或雜環基,其中所述烷基或所述雜環基任選地被一個或多個鹵素或烷基取代。在某些實施方式中,-L 1-L 2-L 3-L 4-是-(C 1-6烷基)-N(R A)-(C 1-6烷基)-O-,其中R A選自C 1-6烷基或3元至6元雜環基,其中所述烷基或雜環基任選地被一個或多個鹵素或C 1-6烷基取代。在某些實施方式中,-L 1-L 2-L 3-L 4-是 。 在一些實施方式中,-L 1-L 2-L 3-L 4-是-烷基-環烷基-烷基-O-,其中所述環烷基任選地被一個或多個R B取代。在某些實施方式中,-L 1-L 2-L 3-L 4-是-(C 1-6烷基)-(C 3-6環烷基)-(C 1-6烷基)-O-,其中所述C 3-6環烷基任選地被一個或多個R B取代。在某些實施方式中,-L 1-L 2-L 3-L 4-是 。 在一些實施方式中,R 1是羥基並且m是1。 在一些實施方式中,R 1是鹵素並且m是1。在某些實施方式中,R 1是溴或氟,並且m是1。 在一些實施方式中,R 1是-N(R D) 2,並且m是1。 在一些實施方式中,R 1是-N(R D) 2,並且每個R D獨立地是氫或任選地被一個或多個獨立地選自-N(R F) 2或-OR G的基團取代的烷基。在某些實施方式中,R 1是-N(R D) 2,並且每個R D是任選地被一個或多個獨立地選自-N(R F) 2或 -OR G的基團取代的C 1-6烷基、C 1-5烷基、C 1-4烷基、C 1-3烷基或C 1-2烷基。在某些實施方式中,R 1是-N(R D) 2,並且每個R D獨立地是甲基、甲氧基乙基、N,N-二甲基氨基乙基、羥乙基或N,N-二甲基氨基丙基。 在一些實施方式中,R 1是任選地被一個或多個R E取代的雜環基,並且m是1。在某些實施方式中,R 1是任選地被一個或多個R E取代的3元至10元雜環基,並且m是1。 在某些實施方式中,R 1選自由以下組成的組: ,其中的每一個任選地被一個或多個R E取代。 在一些實施方式中,R E是鹵素。 在某些實施方式中,R E是F。 在某些實施方式中,每個R E獨立地是鹵素、-N(R F) 2、-烷基-N(R F) 2、-C(O)OR G或任選地被一個或多個鹵素取代的烷基。 在一些實施方式中,R F和R G中的每一個獨立地是烷基。在某些實施方式中,R F和R G中的每一個獨立地是C 1-6烷基、C 1-5烷基、C 1-4烷基、C 1-3烷基或C 1-2烷基。 在某些實施方式中,R F和R G中的每一個獨立地是C 1-3烷基。 在某些實施方式中,每個R E獨立地選自由以下組成的組F、-N(CH 3) 2、-C 1-3烷基-N(CH 3) 2、-C(O)O(叔丁基)、甲基、乙基或三氟乙基。 在某些實施方式中,R E是環烷基或雜環基,其中所述環烷基和雜環基任選地被一個或多個烷基和/或鹵素取代。 在某些實施方式中,R E選自環丙基、嗎啉、哌嗪、氧雜環丁烯基或氮雜環丁烷基,其中的每一個任選地被一個或多個烷基和/或鹵素取代。 在某些實施方式中,R E選自環丙基、嗎啉基、哌嗪基、氧雜環丁烯基或氮雜環丁烷基,其中的每一個任選地被一個或多個烷基和/或鹵素取代。 在一些實施方式中,m是2,R 1中的一個是鹵素,並且另一個R 1是任選地被一個或多個R E取代的雜環基。 在某些實施方式中,m是2,R 1中的一個是鹵素,並且另一個R 1是選自由以下組成的組的雜環基: , 其中的每一個任選地被一個或多個R E取代。 在某些實施方式中,每個R E獨立地選自鹵素、     -N(R F) 2、-烷基-N(R F) 2、-C(O)OR G或任選地被一個或多個鹵素取代的烷基。 在一些實施方式中,R 1選自由以下組成的組: OH、F、Br、 。 在某些實施方式中,R 2是鹵素,並且n是1或2。 在某些實施方式中,R 2是F。 在一些實施方式中,R 2是烷基或環烷基,其中所述烷基和環烷基任選地被一個或多個或鹵素取代。在某些實施方式中,R 2是C 1-6烷基或C 3-6環烷基,其中所述C 1-6烷基和C 3-6環烷基任選地被一個或多個鹵素取代。在某些實施方式中,R 2是甲基、乙基、環丙基或三氟乙基。 在一些實施方式中,R 3是烷基或環烷基。在某些實施方式中,R 3是C 1-6烷基或C 3-6環烷基。在某些實施方式中,R 3是甲基、乙基或環丙基。 在一些實施方式中,m是0、1或2。 在一些實施方式中,n是0或1。 下文闡述了式(I)的示例性化合物。 。 一方面,本公開提供了一種式(II)的化合物: (II) 或其藥學上可接受的鹽,其中: 環A 1是7-12元環烷基、7-12元雜環基、7-12元芳基或7-12元雜芳基; 環B選自由以下組成的組:環烷基、雜環基、芳基和雜芳基; L 1選自由以下組成的組:鍵、烷基、烯基、炔基、雜烷基、雜烯基和雜炔基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基和所述雜炔基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基和氨基; L 2選自由以下組成的組:鍵、N(R A)、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個R B取代; 其中R A選自由以下組成的組:氫、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基、烷基和鹵代烷基; 其中每個R B獨立地選自由以下組成的組:羥基、鹵素、氰基、氨基、烷基、烯基、炔基、雜烷基、雜烯基、環烷基、雜環基、芳基和雜芳基; L 3選自由以下組成的組:鍵、烷基、烯基、炔基、雜烷基、雜烯基和雜炔基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基和所述雜炔基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基和氨基; L 4選自O、S或N(R C); 其中R C選自由以下組成的組:氫、烷基、烯基、炔基、鹵代烷基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基; 每個R 1獨立地選自由以下組成的組:羥基、鹵素、氰基、氨基、-N(R D) 2、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個R E取代; 其中每個R D獨立地選自由以下組成的組:氫、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、  -N(R F) 2或-OR G; 其中每個R E獨立地選自由以下組成的組:氫、羥基、鹵素、氰基、氨基、-N(R F) 2、-烷基-N(R F) 2、-C(O)OR G、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基和烷基; 其中R F和R G中的每一個獨立地選自由以下組成的組:烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基; 每個R 2獨立地選自由以下組成的組:氫、羥基、鹵素、氰基、氨基、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基、烷基和鹵代烷基; 每個R 3選自由以下組成的組:氫、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基和雜環基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基和所述雜環基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基、烷基和鹵代烷基; m是0至5的整數; n是0至4的整數;並且 p是0至3的整數。 在一些實施方式中,環A 1是7-12元雜芳基。 在某些實施方式中,環A 1是四氫異喹啉基。 在某些實施方式中,環A 1。 在一些實施方式中,環B是芳基。在某些實施方式中,環B是C 6-12芳基、C 6-11芳基、C 6-10芳基、C 6-9芳基或C 6-8芳基。在某些實施方式中,環B是苯基。 在一些實施方式中,環B是雜芳基。在某些實施方式中,環B是5-12元雜芳基、5-11元雜芳基、5-10元雜芳基、5-9元雜芳基、5-8元雜芳基、5-7元雜芳基或5-12元雜芳基。在某些實施方式中,環B是吡啶基或吡唑基。 在某些實施方式中,環B選自由以下組成的組: 。 在一些實施方式中,L 1是烷基。在某些實施方式中,L 1是C 1-6烷基、C 1-5烷基、C 1-4烷基、C 1-3烷基或C 1-2烷基。 在某些實施方式中,其中L 1,其中L 1的*端與L 2連接。 在一些實施方式中,L 2是鍵。 在一些實施方式中,L 1是烷基,並且L 2是鍵。 在一些實施方式中,L 3是烷基。在某些實施方式中,L 3是C 1-6烷基、C 1-5烷基、C 1-4烷基、C 1-3烷基或C 1-2烷基。在某些實施方式中,L 3是乙基。 在一些實施方式中,L 4是O或NH。 在一些實施方式中,-L 1-L 2-L 3-L 4-是-烷基-O-。在某些實施方式中,-L 1-L 2-L 3-L 4-是-(C 3-8烷基)-O-。在某些實施方式中,-L 1-L 2-L 3-L 4-是-(CH 2) 5-O-、 -CH 2CH(CH 3)(CH 2) 3-O-或-CH 2CH(CH 2CH 3)(CH 2) 3-O-。 在某個實施方式中,m是0。 在一些實施方式中,m是1,並且R 1是任選地被一個或多個R E取代的烷基。在某些實施方式中,m是1,並且R 1是任選地被一個或多個R E取代的C 1-6烷基、C 1-5烷基、C 1-4烷基、C 1-3烷基或C 1-2烷基。在某些實施方式中,R 1是任選地被一個或多個R E取代的乙基。 在一些實施方式中,R E是-N(R F) 2,並且R F是烷基。在某些實施方式中,R E是-N(R F) 2,並且R F是C 1-6烷基、 C 1-5烷基、C 1-4烷基、C 1-3烷基或C 1-2烷基。在某些實施方式中,R E是-N(CH 3) 2。 在一些實施方式中,m是1,並且R 1是任選地被一個或多個R E取代的雜環基。在某些實施方式中,m是1,並且R 1是3元至10元雜環基、3元至9元雜環基、3元至8元雜環基、3元至7元雜環基、3元至6元雜環基或3元至5元雜環基,其各自任選地被一個或多個R E取代。在某些實施方式中,R 1是氧雜環丁烷基。在某些實施方式中,R 1。 在一些實施方式中,n是1。 在一些實施方式中,R 2是烷基。在某些實施方式中,R 2是C 1-6烷基、C 1-5烷基、C 1-4烷基、C 1-3烷基或C 1-2烷基。在某些實施方式中,R 2是甲基。 在一些實施方式中,n是1,並且R 2是烷基。在某些實施方式中,n是1,並且R 2是C 1-6烷基、C 1-5烷基、C 1-4烷基、C 1-3烷基或C 1-2烷基。在某些實施方式中,n是1,並且R 2是甲基。 在一些實施方式中, 。 在一些實施方式中,p是1。 在一些實施方式中,R 3是烷基。在某些實施方式中,R 3是C 1-6烷基、C 1-5烷基、C 1-4烷基、C 1-3烷基或C 1-2烷基。在某些實施方式中,R 3是甲基。 在一些實施方式中,p是1,並且R 3是烷基。在某些實施方式中,p是1,並且R 3是C 1-6烷基、C 1-5烷基、C 1-4烷基、C 1-3烷基或C 1-2烷基。在某些實施方式中,p是1,並且R 3是甲基。 在一些實施方式中, 。在某些實施方式中, 。 在一些實施方式中,m是0或1。 在一些實施方式中,n是1。 在一些實施方式中,p是1。 另一方面,本公開提供了一種式(III)的化合物: (III) 或其藥學上可接受的鹽,其中: 環A選自由以下組成的組:環烷基、雜環基、芳基和雜芳基; 環B 1是7-12元環烷基、7-12元雜環基、7-12元芳基或7-12元雜芳基; L 1選自由以下組成的組:鍵、烷基、烯基、炔基、雜烷基、雜烯基和雜炔基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基和所述雜炔基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基和氨基; L 2選自由以下組成的組:鍵、N(R A)、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個R B取代; 其中R A選自由以下組成的組:氫、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基、烷基和鹵代烷基; 其中每個R B獨立地選自由以下組成的組:羥基、鹵素、氰基、氨基、烷基、烯基、炔基、雜烷基、雜烯基、環烷基、雜環基、芳基和雜芳基; L 3選自由以下組成的組:鍵、烷基、烯基、炔基、雜烷基、雜烯基和雜炔基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基和所述雜炔基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基和氨基; L 4選自O、S或N(R C); 其中R C選自由以下組成的組:氫、烷基、烯基、炔基、鹵代烷基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基; 每個R 1獨立地選自由以下組成的組:羥基、鹵素、氰基、氨基、-N(R D) 2、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個R E取代; 其中每個R D獨立地選自由以下組成的組:氫、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、  -N(R F) 2或-OR G; 其中每個R E獨立地選自由以下組成的組:氫、羥基、鹵素、氰基、氨基、-N(R F) 2、-烷基-N(R F) 2、-C(O)OR G、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基和烷基; 其中R F和R G中的每一個獨立地選自由以下組成的組:烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基; 每個R 2獨立地選自由以下組成的組:氫、羥基、鹵素、氰基、氨基、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基、烷基和鹵代烷基; 每個R 3選自由以下組成的組:氫、鹵素、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基和雜環基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基和所述雜環基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基、烷基和鹵代烷基; m是0至5的整數; n是0至4的整數;並且 p是0至3的整數。 在一些實施方式中,環A是芳基。在某些實施方式中,環A是C 6-12芳基、C 6-11芳基、C 6-10芳基、C 6-9芳基或C 6-8芳基。在某些實施方式中,環A是苯基。 在一些實施方式中,環A是雜芳基。在某些實施方式中,環A是5-12元雜芳基、5-11元雜芳基、5-10元雜芳基、5-9元雜芳基、5-8元雜芳基、5-7元雜芳基或5-6元雜芳基。在某些實施方式中,環A選自由以下組成的組:呋喃基、苯硫基、吡咯基、吡啶基、吡喃基、嘧啶基、噠嗪基、吡嗪基和四氫異喹啉基。 在一些實施方式中,環B 1是7-12元雜芳基。 在某些實施方式中,環B 1是吡唑並吡啶基。 在某些實施方式中,環B 1。 在一些實施方式中,L 1是鍵。 在一些實施方式中,L 1是烷基。在某些實施方式中,L 1是C 1-6烷基、C 1-5烷基、C 1-4烷基、C 1-3烷基或C 1-2烷基。 在某些實施方式中,L 1選自由以下組成的組: ,其中L 1的*端與L 2連接。 在一些實施方式中,L 2是鍵。 在一些實施方式中,L 2是N(R A),並且R A選自烷基或雜環基,其中所述烷基或所述雜環基任選地被一個或多個鹵素或烷基取代。在某些實施方式中,L 2是N(R A),並且R A是乙基、二氟乙基、三氟乙基或氧雜環丁烷基。 在一些實施方式中,L 2是任選地被一個或多個R B取代的環烷基。在某些實施方式中,L 2是C 3-10環烷基、C 3-9環烷基、C 3-8環烷基、C 3-7環烷基、C 3-6環烷基或C 3-5環烷基,其各自任選地被一個或多個R B取代。 在某些實施方式中,L 2是任選地被一個或多個R B取代的 。 在一些實施方式中,L 2是任選地被一個或多個R B取代的雜環基。在某些實施方式中,L 2是含有一個或多個選自N、O或S的雜原子的雜環基。在某些實施方式中,L 2是3-12元雜環基、3-11元雜環基、3-10元雜環基或3-9元雜環基,其各自任選地被一個或多個R B取代。 在某些實施方式中,L 2是選自由以下組成的組的雜環基: ,其中的每一個任選地被一個或多個R B取代,其中L 2的*端與L 3連接。 在一些實施方式中,L 2是被一個或多個R B取代的環烷基或雜環基,並且R B是烷基。在某些實施方式中,R B是C 1-6烷基、C 1-5烷基、C 1-4烷基、C 1-3烷基或C 1-2烷基。在某些實施方式中,R B是甲基。 在一些實施方式中,L 2選自由以下組成的組: ,其中L 2的*端與L 3連接。 在一些實施方式中,L 3是烷基。在一些實施方式中,L 3是C 1-6烷基、C 1-5烷基、C 1-4烷基、C 1-3烷基或C 1-2烷基。 在某些實施方式中,L 3是乙基。 在一些實施方式中,L 4是O或NH。 在一些實施方式中,L 1是鍵,並且L 2是鍵或任選地被一個或多個R B取代的雜環基。 在一些實施方式中,L 1是烷基,並且L 2是鍵、N(R A)或任選地被一個或多個R B取代的環烷基。 在一些實施方式中,-L 1-L 2-L 3-L 4-是-烷基-O-。在某些實施方式中,-L 1-L 2-L 3-L 4-是-(C 3-8烷基)-O-。在某些實施方式中,-L 1-L 2-L 3-L 4-是-(CH 2) 5-O-、 -CH 2CH(CH 3)(CH 2) 3-O-或-CH 2CH(CH 2CH 3)(CH 2) 3-O-。 在一些實施方式中,-L 1-L 2-L 3-L 4-是-雜環基-烷基   -O-,其中所述雜環基任選地被一個或多個R B取代。在某些實施方式中,-L 1-L 2-L 3-L 4-是-(5-10元雜環基)-(C 1-6烷基)-O-,其中所述5-10元雜環基任選地被一個或多個R B取代。在某些實施方式中,-L 1-L 2-L 3-L 4-選自由以下組成的組: 以及 。 在一些實施方式中,-L 1-L 2-L 3-L 4-是-烷基-N(R A)-烷基-O-,其中R A選自烷基或雜環基,其中所述烷基或所述雜環基任選地被一個或多個鹵素或烷基取代。在某些實施方式中,-L 1-L 2-L 3-L 4-是-(C 1-6烷基)-N(R A)-(C 1-6烷基)-O-,其中RA選自C 1-6烷基或3元至6元雜環基,其中所述烷基或雜環基任選地被一個或多個鹵素或C 1-6烷基取代。在某些實施方式中,-L 1-L 2-L 3-L 4-是 。 在一些實施方式中,-L 1-L 2-L 3-L 4-是-烷基-環烷基-烷基-O-,其中所述環烷基任選地被一個或多個R B取代。在某些實施方式中,-L 1-L 2-L 3-L 4-是-(C 1-6烷基)-(C 3-6環烷基)-(C 1-6烷基)-O-,其中所述C 3-6環烷基任選地被一個或多個R B取代。在某些實施方式中,-L 1-L 2-L 3-L 4-是 。 在一些實施方式中,R 1是鹵素並且m是1。 在一些實施方式中,R 1是-N(R D) 2,並且m是1。 在一些實施方式中,R 1是-N(R D) 2,並且每個R D獨立地是任選地被一個或多個獨立地選自-N(R F) 2或-OR G的基團取代的烷基。在某些實施方式中,R 1是-N(R D) 2,並且每個R D是任選地被一個或多個獨立地選自-N(R F) 2或-OR G的基團取代的C 1-6烷基、C 1-5烷基、C 1-4烷基、C 1-3烷基或C 1-2烷基。在某些實施方式中,R 1是-N(R D) 2,並且每個R D獨立地是甲基、甲氧基乙基、N,N-二甲基氨基乙基或N,N-二甲基氨基丙基。 在一些實施方式中,R 1是任選地被一個或多個R E取代的雜環基,並且m是1。在某些實施方式中,R 1是任選地被一個或多個R E取代的3元至10元雜環基,並且m是1。 在某些實施方式中,R 1選自由以下組成的組: ,其中的每一個任選地被一個或多個R E取代。 在一些實施方式中,R E是鹵素。 在某些實施方式中,R E是F。 在某些實施方式中,R E是-N(R F) 2、-烷基-N(R F) 2、  -C(O)OR G或任選地被一個或多個鹵素取代的烷基。 在一些實施方式中,R F和R G中的每一個獨立地是烷基。在某些實施方式中,R F和R G中的每一個獨立地是C 1-6烷基、C 1-5烷基、C 1-4烷基、C 1-3烷基或C 1-2烷基。 在某些實施方式中,R F和R G中的每一個獨立地是C 1-3烷基。 在某些實施方式中,R E選自由以下組成的組:     -N(CH 3) 2、-C 1-3烷基-N(CH 3) 2、-C(O)O(叔丁基)、甲基、乙基和三氟乙基。 在某些實施方式中,R E是環烷基或雜環基,其中所述環烷基和雜環基任選地被一個或多個烷基和/或鹵素取代。 在某些實施方式中,R E選自環丙基、嗎啉基、哌嗪基、氧雜環丁烷基或氮雜環丁烷基,其中的每一個任選地被一個或多個烷基和/或鹵素取代。 在某些實施方式中,R E選自環丙基、嗎啉基、哌嗪基、氧雜環丁烷基或氮雜環丁烷基,其中的每一個任選地被一個或多個烷基和/或鹵素取代。 在某些實施方式中,R 2是鹵素,並且n是1或2。 在某些實施方式中,R 2是F。 在一些實施方式中,R 2是烷基或環烷基,其中所述烷基和環烷基任選地被一個或多個或鹵素取代。在某些實施方式中,R 2是C 1-6烷基或C 3-6環烷基,其中所述C 1-6烷基和C 3-6環烷基任選地被一個或多個鹵素取代。在某些實施方式中,R 2是甲基、乙基、環丙基或三氟乙基。 在一些實施方式中, 。 在一些實施方式中,R 3是烷基或環烷基。在某些實施方式中,R 3是C 1-6烷基或C 3-6環烷基。在某些實施方式中,R 3是甲基、乙基或環丙基。 在一些實施方式中,p是1,並且R 3是烷基。在某些實施方式中,p是1,並且R 3是C 1-6烷基、C 1-5烷基、C 1-4烷基、C 1-3烷基或C 1-2烷基。在某些實施方式中,p是1,並且R 3是甲基。 在一些實施方式中, 。在某些實施方式中, 。 在一些實施方式中,m是0、1或2。 在一些實施方式中,n是0或1。 在一些實施方式中,p是1。 另一方面,本公開提供了一種式(IV)的化合物: (IV) 或其藥學上可接受的鹽,其中: 環A選自由以下組成的組:環烷基、雜環基、芳基和雜芳基; 環B選自由以下組成的組:環烷基、雜環基、芳基和雜芳基; L 1選自由以下組成的組:鍵、烷基、烯基、炔基、雜烷基、雜烯基和雜炔基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基和所述雜炔基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基和氨基; L 21是7-12元環烷基或7-12元雜環基,其中所述環烷基和雜環基任選地被一個或多個R B取代; 其中每個R B獨立地選自由以下組成的組:羥基、鹵素、氰基、氨基、烷基、烯基、炔基、雜烷基、雜烯基、環烷基、雜環基、芳基和雜芳基; L 3選自由以下組成的組:鍵、烷基、烯基、炔基、雜烷基、雜烯基和雜炔基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基和所述雜炔基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基和氨基; L 4選自O、S或N(R C); 其中R C選自由以下組成的組:氫、烷基、烯基、炔基、鹵代烷基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基; 每個R 1獨立地選自由以下組成的組:羥基、鹵素、氰基、氨基、-N(R D) 2、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個R E取代; 其中每個R D獨立地選自由以下組成的組:氫、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、  -N(R F) 2或-OR G; 其中每個R E獨立地選自由以下組成的組:氫、羥基、鹵素、氰基、氨基、-N(R F) 2、-烷基-N(R F) 2、-C(O)OR G、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基和烷基; 其中R F和R G中的每一個獨立地選自由以下組成的組:烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基; 每個R 2獨立地選自由以下組成的組:氫、羥基、鹵素、氰基、氨基、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基、烷基和鹵代烷基; R 3選自由以下組成的組:氫、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基和雜環基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基和所述雜環基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基、烷基和鹵代烷基; m是0至5的整數; n是0至4的整數;並且 p是0至3的整數。 在一些實施方式中,環A是芳基。在某些實施方式中,環A是C 6-12芳基、C 6-11芳基、C 6-10芳基、C 6-9芳基或C 6-8芳基。在某些實施方式中,環A是苯基。 在一些實施方式中,環A是雜芳基。在某些實施方式中,環A是5-12元雜芳基、5-11元雜芳基、5-10元雜芳基、5-9元雜芳基、5-8元雜芳基、5-7元雜芳基或5-6元雜芳基。在某些實施方式中,環A選自由以下組成的組:呋喃基、苯硫基、吡咯基、吡啶基、吡喃基、嘧啶基、噠嗪基、吡嗪基和四氫異喹啉基。 在一些實施方式中,環B是芳基。在某些實施方式中,環B是C 6-12芳基、C 6-11芳基、C 6-10芳基、C 6-9芳基或C 6-8芳基。在某些實施方式中,環B是苯基。 在一些實施方式中,環B是雜芳基。在某些實施方式中,環B是5-12元雜芳基、5-11元雜芳基、5-10元雜芳基、5-9元雜芳基、5-8元雜芳基、5-7元雜芳基或5-6元雜芳基。在某些實施方式中,環B是吡啶基或吡唑基。 在某些實施方式中,環B選自由以下組成的組: 。 在一些實施方式中,L 1是鍵。 在一些實施方式中,L 21是含有一個或多個選自N或O的雜原子的7-10元雜環基。 在某些實施方式中,L 21選自由以下組成的組: ,其中L 21的*端與L 3連接。 在一些實施方式中,L 3是烷基。在某些實施方式中,L 3是C 1-6烷基、C 1-5烷基、C 1-4烷基、C 1-3烷基或C 1-2烷基。 在某些實施方式中,L 3是乙基。 在一些實施方式中,L 4是O或NH。 在一些實施方式中,-L 1-L 21-L 3-L 4-是-雜環基-烷基  -O-,其中所述雜環基任選地被一個或多個R B取代。在某些實施方式中,-L 1-L 21-L 3-L 4-是-(5-10元雜環基)-(C 1-6烷基)-O-,其中所述5-10元雜環基任選地被一個或多個R B取代。 在某些實施方式中,-L 1-L 21-L 3-L 4-選自由以下組成的組: 。 在一些實施方式中,R 1是鹵素並且m是1。 在一些實施方式中,R 1是-N(R D) 2,並且m是1。 在一些實施方式中,R 1是-N(R D) 2,並且每個R D獨立地是任選地被一個或多個獨立地選自-N(R F) 2或-OR G的基團取代的烷基。在某些實施方式中,R 1是-N(R D) 2,並且每個R D是任選地被一個或多個獨立地選自-N(R F) 2或-OR G的基團取代的C 1-6烷基、C 1-5烷基、C 1-4烷基、C 1-3烷基或C 1-2烷基。在某些實施方式中,R 1是-N(R D) 2,並且每個R D獨立地是甲基、甲氧基乙基或N,N-二甲基氨基丙基。 在一些實施方式中,R 1是任選地被一個或多個R E取代的雜環基,並且m是1。在某些實施方式中,R 1是任選地被一個或多個R E取代的3元至10元雜環基,並且m是1。 在某些實施方式中,R 1選自由以下組成的組: ,其中的每一個任選地被一個或多個R E取代。 在一些實施方式中,R E是鹵素。 在某些實施方式中,R E是F。 在某些實施方式中,R E是-N(R F) 2、-烷基-N(R F) 2、  -C(O)OR G或任選地被一個或多個鹵素取代的烷基。 在一些實施方式中,R F和R G中的每一個獨立地是烷基。在某些實施方式中,R F和R G中的每一個獨立地是C 1-6烷基、C 1-5烷基、C 1-4烷基、C 1-3烷基或C 1-2烷基。 在某些實施方式中,R F和R G中的每一個獨立地是C 1-3烷基。 在某些實施方式中,R E選自由以下組成的組:     -N(CH 3) 2、-C 1-3烷基-N(CH 3) 2、-C(O)O(叔丁基)、甲基、乙基和三氟乙基。 在某些實施方式中,R E是環烷基或雜環基,其中所述環烷基和雜環基任選地被一個或多個烷基和/或鹵素取代。 在某些實施方式中,R E選自環丙基、嗎啉基、哌嗪基、氧雜環丁烷基或氮雜環丁烷基,其中的每一個任選地被一個或多個烷基和/或鹵素取代。 在某些實施方式中,R E選自環丙基、嗎啉基、哌嗪基、氧雜環丁烯基或氮雜環丁烷基,其中的每一個任選地被一個或多個烷基和/或鹵素取代。 在某些實施方式中,R 1選自由以下組成的組: 。 在某些實施方式中,R 2是鹵素,並且n是1或2。 在某些實施方式中,R 2是F。 在一些實施方式中,R 2是烷基或環烷基,其中所述烷基和環烷基任選地被一個或多個或鹵素取代。在某些實施方式中,R 2是C 1-6烷基或C 3-6環烷基,其中所述C 1-6烷基和C 3-6環烷基任選地被一個或多個鹵素取代。在某些實施方式中,R 2是甲基、乙基、環丙基或三氟乙基。 在某些實施方式中,R 2是甲基。 在一些實施方式中,R 3是烷基。在某些實施方式中,R 3中的每個獨立地是C 1-6烷基、C 1-5烷基、C 1-4烷基、C 1-3烷基或C 1-2烷基。在某些實施方式中,R 3是甲基。 在一些實施方式中,p是1,並且R 3是烷基。在某些實施方式中,p是1,並且R 3是C 1-6烷基、C 1-5烷基、C 1-4烷基、C 1-3烷基或C 1-2烷基。在某些實施方式中,p是1,並且R 3是甲基。 在一些實施方式中, 。在某些實施方式中, 。 在一些實施方式中,m是1。 在一些實施方式中,n是1。 在一些實施方式中,p是1。 下文闡述了本公開的示例性化合物。          本文所提供的化合物是參考通式和具體化合物兩者來描述的。另外,本公開的化合物可以以多種不同的形式或衍生物存在,包括但不限於前藥、活性代謝衍生物(活性代謝物)、溶劑化物、藥學上可接受的鹽或同位素衍生物,所有這些都在本公開的範圍內。 如本文所使用的,術語“前藥”是指在生理條件下代謝或通過溶劑分解轉化時產生期望的活性化合物的化合物或其藥學上可接受的鹽。前藥包括但不限於活性化合物的酯、醯胺、氨基甲酸酯、碳酸酯、醯脲、溶劑化物或水合物。典型地,前藥無活性或活性低於活性化合物,但是可以提供一種或多種有利的處理、施用和/或代謝性質。例如,一些前藥是活性化合物的酯;在代謝期間中,酯基被裂解以產生活性藥物。此外,一些前藥被酶促啟動以產生活性化合物或在進一步化學反應後產生活性化合物的化合物。前藥可以在單個步驟中由前藥形式發展為活性形式,或者可以具有一種或多種本身可以具有活性或可以無活性的中間體形式。在以下參考文獻中討論了前藥的製備和用途:T. Higuchi和V. Stella, “作為新型遞送系統的前藥(Pro-drugs as Novel Delivery Systems)”, A.C.S.研討會系列(A.C.S. Symposium Series)的第14卷, 《藥物設計中的生物可逆載體(Bioreversible Carriers in Drug Design)》, 編輯Edward B. Roche, 美國製藥協會(American Pharmaceutical Association)和培格曼出版社(Pergamon Press), 1987;《前藥:挑戰與回報(Prodrugs: Challenges and Rewards)》, 編輯V. Stella, R. Borchardt, M. Hageman, R. Oliyai, H. Maag, J. Tilley, 紐約施普林格出版社(Springer Verlag New York), 2007,所述參考文獻全都據此通過引用整體併入。 如本文所使用的,術語“代謝物”,例如活性代謝物,與如上文所描述的前藥重疊。因此,此類代謝物是藥理學活性化合物或進一步代謝為藥理學活性化合物的化合物,所述藥理學活性化合物是由受試者體內的代謝過程產生的衍生物。例如,此類代謝物可以由所施用的化合物或鹽或前藥的氧化、還原、水解、醯胺化、脫醯胺化、酯化、脫酯化、酶促裂解等產生。其中,活性代謝物是此類藥理學活性衍生化合物。對於前藥,前藥化合物通常無活性或活性低於代謝產物。對於活性代謝物,母體化合物可以是活性化合物或者可以是無活性前藥。 可以使用本領域已知的常規技術來鑒定前藥和活性代謝物。參見例如Bertolini等人, 1997, 《藥物化學雜誌(J Med Chem)》40:2011-2016;Shan等人, 《藥物科學雜誌(J Pharm Sci)》86:756-757;Bagshawe, 1995, 《藥物開發研究(DrugDev Res)》 34:220-230;Wermuth, 同上。 如本文所使用的,術語“藥學上可接受的”表明物質或組合物在化學和/或毒理學上與構成調配物的其它成分和/或被治療的受試者相容。 如本文所使用的,除非另外指示,否則術語“藥學上可接受的鹽”包括保留指定化合物的游離酸和鹼的生物有效性並且在生物學上或其它方面並非不理想的鹽。所考慮的藥學上可接受的鹽形式包括但不限於單鹽、雙鹽、三鹽、四鹽等。藥學上可接受的鹽在其被施用的量和濃度下是無毒的。此類鹽的製備可以通過改變化合物的物理特性而不妨礙其發揮其生理作用來促進藥理學使用。物理性質的有用改變包括降低熔點以促進經黏膜施用以及增加溶解度以促進施用更高濃度的藥物。 藥學上可接受的鹽包括酸加成鹽,如包括以下的酸加成鹽:硫酸鹽、氯化物、鹽酸鹽、富馬酸鹽、馬來酸鹽、磷酸鹽、氨基磺酸鹽、乙酸鹽、檸檬酸鹽、乳酸鹽、酒石酸鹽、甲磺酸鹽、乙磺酸鹽、苯磺酸鹽、對甲苯磺酸鹽、環己基氨基磺酸鹽和奎尼酸鹽。藥學上可接受的鹽可以由如以下等酸獲得:鹽酸、馬來酸、硫酸、磷酸、氨基磺酸、乙酸、檸檬酸、乳酸、酒石酸、丙二酸、甲磺酸、乙磺酸、苯磺酸、對甲苯磺酸、環己基氨基磺酸、富馬酸和奎寧酸。 當存在如羧酸或苯酚等酸性官能團時,藥學上可接受的鹽還包括鹼加成鹽,如包括以下的鹼加成鹽:苄星青黴素(benzathine)、氯普魯卡因(chloroprocaine)、膽鹼、二乙醇胺、乙醇胺、叔丁胺、乙二胺、葡甲胺、普魯卡因(procaine)、鋁、鈣、鋰、鎂、鉀、鈉、銨、烷基胺和鋅。例如參見《雷明頓氏藥物科學(Remington's Pharmaceutical Sciences)》, 第19版, 賓夕法尼亞州伊斯頓的馬克出版公司(Mack Publishing Co., Easton, PA), 第2卷, 第1457頁, 1995;《藥用鹽手冊:性質、選擇和用途(Handbook of Pharmaceutical Salts: Properties, Selection, and Use)》, Stahl和Wermuth, 德國魏因海姆的Wiley-VCH出版社(Wiley-VCH, Weinheim, Germany), 2002。此類鹽可以使用適當的對應鹼來製備。 藥學上可接受的鹽可以通過標準技術製備。例如,化合物的游離鹼形式可以溶解在合適的溶劑(如含有適當的酸的水溶液或水-醇溶液)中,並且然後通過蒸發溶液來分離。因此,如果特定化合物是鹼,則期望的藥學上可接受的鹽可以通過本領域中可用的任何合適的方法來製備,例如,用以下酸處理游離鹼:如鹽酸、氫溴酸、硫酸、硝酸、磷酸等無機酸;或如乙酸、馬來酸、琥珀酸、杏仁酸、富馬酸、丙二酸、丙酮酸、草酸、乙醇酸、水楊酸等有機酸;如葡糖醛酸或半乳糖醛酸等吡喃糖基酸;如檸檬酸或酒石酸等α-羥基酸;如天冬氨酸或谷氨酸等氨基酸;如苯甲酸或肉桂酸等芳香族酸;如對甲苯磺酸或乙磺酸等磺酸;等等。 類似地,如果特定化合物為酸,則期望的藥學上可接受的鹽可以通過任何合適的方法來製備,例如,用如胺(伯、仲或叔)等無機或有機鹼、鹼金屬氫氧化物或鹼土金屬氫氧化物等處理游離酸。合適的鹽的說明性實例包括衍生自如L-甘氨酸、L-賴氨酸和L-精氨酸等氨基酸;氨、伯胺、仲胺和叔胺;以及如羥乙基吡咯烷、哌啶、嗎啉或哌嗪等環胺的有機鹽;以及衍生自鈉、鈣、鉀、鎂、錳、鐵、銅、鋅、鋁和鋰的無機鹽。 還應理解,本公開的化合物可以以非溶劑化形式、溶劑化形式(例如,水合形式)和固體形式(例如,晶體形式或多晶形式)存在,並且本公開旨在涵蓋所有此類形式。 如本文所使用的,術語“溶劑化物”或“溶劑化形式”是指包括化學計量或非化學計量量的溶劑的溶劑加成形式。一些化合物具有在結晶固態中捕獲固定摩爾比的溶劑分子,從而形成溶劑化物的趨勢。如果溶劑是水,則所形成的溶劑化物是水合物,並且如果溶劑是醇,所形成的溶劑化物是醇化物。水合物是通過將一個或多個水分子與其中水保持其作為H 2O的分子狀態的物質的一個分子組合來形成的。形成溶劑化物的溶劑的實例包括但不限於水、異丙醇、乙醇、甲醇、DMSO、乙酸乙酯、乙酸和乙醇胺。 本公開還旨在包括本文所提供的化合物的所有同位素形式。原子的同位素包括具有相同原子序數但具有不同質量數的原子。例如,除非另外說明,否則本公開的化合物中的氫、碳、氮、氧、磷、硫、氟、氯、溴或碘意指還包括其同位素,如但不限於 1H、 2H、 3H、 11C、 12C、 13C、 14C、 14N、 15N、 16O、 17O、 18O、 31P、 32P、 32S、 33S、 34S、 36S、 17F、 18F、 19F、 35Cl、 37Cl、 79Br、 81Br、 124I、 127I和 131I。在一些實施方式中,氫包括氕、氘和氚。在一些實施方式中,碳包括 12C和 13C。 本文所提供的化合物或其藥學上可接受的鹽可以含有一個或多個不對稱中心,因此產生對映異構體、非對映異構體和其它立體異構形式,其可以根據絕對立體化學定義為氨基酸的(R)-或(S)-或(D)-或(L)-,或者根據相對構型定義為rel-(R)-或rel-(S)-。本公開包括所有此類可能異構體,以及其外消旋和光學純形式。光學活性的(+)和(-)、(R)-和(S)或(D)-和(L)-異構體可以通過手性合成子或手性試劑製備或使用常規技術解析,如色譜法和分步結晶法。用於製備、分離單獨的對映異構體的傳統技術包括從合適的光學純前體進行手性合成或使用例如手性高壓液相色譜法(HPLC)拆分外消旋體(或鹽或衍生物的外消旋體)。當化合物以其手性形式表示時,應當理解所述實施方式包括但不限於特定的非對映體或對映體富集形式。在未指定手性但存在手性的情況下,應當理解所述實施方式旨在包括特定的非對映體或對映體富集形式;或此類化合物的外消旋或非外消混合物(scalemic mixture)。 術語“立體異構體”是指含有通過相同鍵連接的但具有不同三維結構的相同原子的化合物,其不可互換。本公開考慮了各種立體異構體和其混合物並且包括“對映異構體”,所述對映異構體是指兩種立體異構體,所述立體異構體的分子是彼此不可重疊的鏡像。 術語“對映異構體”表示一對彼此不可重疊鏡像的立體異構體。一對對映異構體的1:1混合物是“外消旋”混合物。比率不是1:1的對映體混合物是“非外消旋”混合物。 術語“非對映異構體”表示具有至少兩個不對稱原子但不是彼此鏡像的立體異構體。 術語“互變異構體”或“互變異構形式”是指可通過低能壘相互轉化的不同能量的結構異構體。異構形式的存在和濃度將取決於化合物所處的環境並且可以因例如化合物是否為固體或是否在有機或水溶液中而不同。舉例來說,質子互變異構體(也稱為質子異變互變異構體)包括通過質子遷移進行的相互轉化,如酮-烯醇、醯胺-亞胺酸、內醯胺-內醯亞胺、亞胺-烯胺異構化,以及質子可以佔據雜環體系的兩個或更多個位置的環狀形式。化合價互變異構體包括通過一些成鍵電子的重組而進行的相互轉化。互變異構體可以處於平衡狀態或通過適當的取代而空間鎖定成一種形式。除非另外說明,否則本公開的按名稱或結構標識為一種特定互變異構形式的化合物旨在包括其它互變異構形式。 當本文所提供的這些化合物含有烯烴雙鍵或幾何不對稱的其它中心時,並且除非另外指明,否則這些化合物包括E幾何異構體和Z幾何異構體兩者。 合成方法 本文所提供的化合物可以使用任何已知的有機合成技術來製備並且可以根據多種可能的合成途徑中的任何一種來合成。 可以在有機合成領域的技術人員可以容易地選擇的合適的溶劑中進行用於製備本公開的化合物的反應。合適的溶劑可以在進行反應的溫度,例如範圍可以為溶劑的凍結溫度到溶劑的沸騰溫度的溫度下與起始材料(反應物)、中間體或產物基本上無反應性。給定反應可以在一種溶劑或多於一種溶劑的混合物中進行。根據特定反應步驟,特定反應步驟的合適溶劑可以由本領域技術人員選擇。 本公開的化合物的製備可以涉及各種化學基團的保護和脫保護。對保護和脫保護的需求和對適當的保護基團的選擇可以由所屬領域的技術人員容易地確定。可以在例如以下參考文獻中找到保護基團的化學:T. W. Greene和P. G. M. Wuts, 《有機合成中的保護基團(Protective Groups in Organic Synthesis)》, 第3版, 紐約的約翰威利父子出版公司, (1999);P. Kocienski, 《保護基團(Protecting Groups)》, 喬治泰米出版社(Georg Thieme Verlag), 2003;和Peter G.M. Wuts, 《格林氏有機合成中的保護基團(Greene's Protective Groups in Organic Synthesis)》, 第5版, 威利出版社(Wiley), 2014,所述參考文獻通過引用整體併入本文。 可以根據本領域已知的任何合適的方法監測反應。例如,可以通過如核磁共振波譜法(例如, 1H或 13C)、紅外光譜法、分光光度法(例如,UV可見)、質譜法等光譜學手段或通過如高效液相色譜法(HPLC)、液相色譜法-質譜法(LCMS)或薄層色譜法(TLC)等色譜方法來監測產物形成。本領域技術人員可以通過各種方法對化合物進行純化,所述方法包括高效液相色譜法(HPLC)(“製備型LC-MS純化:改進的化合物特定方法優化(Preparative LC-MS Purification: Improved Compound Specific Method Optimization)” Karl F. Blom, Brian Glass, Richard Sparks, Andrew P. Combs 《組合化學雜誌(J. Combi. Chem.)》2004, 6(6), 874-883,所述參考文獻通過引用整體併入本文)和正相矽膠色譜法。 藥物組合物 在另外的方面,提供了藥物組合物,其包含一種或多種本公開的化合物或其藥學上可接受的鹽。在一些實施方式中,本公開的藥物組合物包含本文所提供的第一化合物或其藥學上可接受的鹽和一種或多種相同式的另外的化合物,但所述第一化合物和另外的化合物不是相同的分子。 另一方面,提供了一種藥物組合物,其包含一種或多種本公開的化合物或其藥學上可接受的鹽以及至少一種藥學上可接受的賦形劑。 在一些實施方式中,本公開的藥物組合物包含治療有效量的一種或多種本公開的化合物或其藥學上可接受的鹽。 在一些實施方式中,本公開的藥物組合物包含治療有效量的一種或多種本公開的化合物或其藥學上可接受的鹽以及至少一種藥學上可接受的賦形劑。 如本文所使用的,術語“治療有效量”是指治療、改善或預防鑒別出的疾病或病狀或顯示出可檢測的治療或抑制作用的分子、化合物或包含所述分子或化合物的組合物的量。效應可以通過本領域已知的任何測定方法來檢測。受試者的精確有效量將取決於受試者的體重、體型和健康狀況;病狀的性質和程度;施用速率;選擇用於施用的治療或治療組合;以及處方醫師的判斷。對於給定情況的治療有效量可以通過在臨床醫生的技能和判斷內的常規實驗來確定。 如本文所使用的,術語“藥物組合物”是指呈適於向受試者施用的形式的包括本公開的分子或化合物的調配物。藥物組合物包含適合於口服施用、直腸施用、局部施用、腸胃外(包括皮下、肌肉內和靜脈內)施用、舌下施用、眼部施用、透皮施用或鼻腔施用的組合物,但是在任何給定情況下最合適的途徑將取決於特定宿主和施用活性成分的病狀的性質和嚴重程度。藥物組合物可以方便地以單位劑型呈現並且可以通過藥學領域熟知的任何方法製備。 如本文所使用的,術語“藥學上可接受的賦形劑”意指可用於製備通常安全、無毒並且在生物學上和其它方面均合乎需要的藥物組合物的賦形劑,並且包括對於獸醫用途以及人類藥物用途而言是可接受的賦形劑。如本文所使用的,“藥學上可接受的賦形劑”包括一種和多於一種此類賦形劑。術語“藥學上可接受的賦形劑”還涵蓋“藥學上可接受的載體”和“藥學上可接受的稀釋劑”。 所使用的特定賦形劑將取決於應用本公開的化合物的手段和目的。溶劑通常是基於要向包括人在內的哺乳動物施用的本領域技術人員認為安全的溶劑來選擇的。通常,安全溶劑為無毒水性溶劑,如水和可溶於或可混溶於水中的其它無毒溶劑。合適的水性溶劑包括水、乙醇、丙二醇、聚乙二醇(例如,PEG 400、PEG 300)等以及其混合物。 在一些實施方式中,合適的賦形劑可以包括緩衝劑,如磷酸鹽、檸檬酸鹽和其它有機酸;抗氧化劑,包括抗壞血酸和甲硫氨酸;防腐劑(如氯化十八烷基二甲基苄基銨;氯化六甲銨;苯紮氯銨;苄索氯銨;苯酚、丁醇或苯甲醇;對羥苯甲酸烷基酯,如對羥苯甲酸甲酯或丙酯;鄰苯二酚;間苯二酚;環己醇;3-戊醇;以及間甲酚);低分子量(少於約10個殘基)多肽;蛋白質,如血清白蛋白、明膠或免疫球蛋白;親水性聚合物,如聚乙烯吡咯烷酮;氨基酸,如甘氨酸、穀氨醯胺、天冬醯胺、組氨酸、精氨酸或賴氨酸;單糖、二糖和其它碳水化合物,包括葡萄糖、甘露糖或葡聚糖;螯合劑,如EDTA;糖,如蔗糖、甘露醇、海藻糖或山梨糖醇;成鹽反離子,如鈉;金屬絡合物(例如,Zn蛋白質絡合物);和/或非離子型表面活性劑,如TWEEN™、PLURONICS™或聚乙二醇(PEG)。 在一些實施方式中,合適的賦形劑可以包括一種或多種穩定劑、表面活性劑、潤濕劑、潤滑劑、乳化劑、懸浮劑、防腐劑、抗氧化劑、遮光劑、助流劑、加工助劑、著色劑、甜味劑、芳香劑、調味劑和其它已知添加劑以提供藥物(即,本公開的化合物或其藥物組合物)的最佳呈現形式或説明製造藥物產品(即,藥物)。活性藥物成分還可以包埋在例如通過凝聚技術或通過介面聚合製備的微膠囊中,所述微膠囊例如分別在膠體藥物遞送系統(例如,脂質體、白蛋白微球、微乳液、納米顆粒以及納米膠囊)或在粗乳液中的羥甲基纖維素或明膠微膠囊以及聚-(甲基丙烯酸甲酯)微膠囊。此類技術公開於《雷明頓氏藥物科學》第16版, Osol, A.編輯(1980)中。“脂質體”為可用於將藥物(如本文中所公開的化合物和任選地化療劑)遞送到包括人在內的哺乳動物包括各種類型的脂質、磷脂和/或表面活性劑的小囊泡。脂質體的組分通常以雙層形式排列,類似於生物膜的脂質排列。 本文所提供的藥物組合物可以呈允許向受試者,包括但不限於人,施用組合物並且允許將組合物調配成與預期施用途徑相容的任何形式。 針對本文所提供的藥物組合物考慮了多種途徑,並且因此本文所提供的藥物組合物可以取決於預期施用途徑而以散裝或單位劑型供應。例如,對於口服、經頰和舌下施用,粉劑、懸浮液、顆粒、片劑、丸劑、膠囊、軟膠囊和囊片作為固體劑型可以為可接受的,並且乳液、糖漿、酏劑、懸浮液和溶液作為液體劑型可以為可接受的。對於注射施用,乳液和懸浮液作為液體劑型可以為可接受的,並且適於用合適溶液復原的粉劑作為固體劑型可以為可接受的。對於吸入施用,溶液、噴霧劑、乾粉和氣霧劑可以為可接受的劑型。對於局部(包括經頰和舌下)或經皮施用,粉劑、噴霧劑、軟膏、糊劑、乳膏、洗劑、凝膠、溶液和貼片可以為可接受的劑型。對於陰道施用,陰道栓劑、棉塞、乳膏、凝膠、糊劑、泡沫和噴霧劑可以為可接受的劑型。 在一些實施方式中,本公開的藥物組合物可以呈用於口服施用的調配物的形式。 在某些實施方式中,本公開的藥物組合物可以呈片劑調配物形式。適於片劑調配物的藥學上可接受的賦形劑包括例如惰性稀釋劑,如乳糖、碳酸鈉、磷酸鈣或碳酸鈣;造粒劑和崩解劑,如玉米澱粉或海藻酸;黏合劑,如澱粉;潤滑劑,如硬脂酸鎂、硬脂酸或滑石;防腐劑,如對羥基苯甲酸乙酯或對羥基苯甲酸丙酯;以及抗氧化劑,如抗壞血酸。片劑調配物可以是未包衣的或包衣的,以調節其崩解和後續活性成分在胃腸道內的吸收,或改進其穩定性和/或外觀,在任一種情況下,均使用本領域中熟知的常規包衣劑和程式。 在某些實施方式中,本公開的藥物組合物可以呈硬明膠膠囊形式,其中活性成分與惰性固體稀釋劑,例如碳酸鈣、磷酸鈣或高嶺土混合;或呈軟明膠膠囊形式,其中活性成分與水或油,如花生油、液體石蠟或橄欖油混合。 在某些實施方式中,本公開的藥物組合物可以呈水性懸浮液形式,所述水性懸浮液通常包括呈細粉形式的活性成分以及一種或多種懸浮劑,如羧甲基纖維素鈉、甲基纖維素、羥丙基甲基纖維素、海藻酸鈉、聚乙烯吡咯烷酮、黃蓍膠和阿拉伯樹膠;分散劑或潤濕劑,如卵磷脂或烯化氧與脂肪酸的縮合產物(例如,聚氧乙烯硬脂酸酯);或環氧乙烷與長鏈脂肪醇的縮合產物,例如十七亞乙基氧基鯨蠟醇;或環氧乙烷與衍生自脂肪酸和己糖醇的偏酯的縮合產物,如聚氧乙烯山梨醇單油酸酯;或環氧乙烷與衍生自脂肪酸和己糖醇酸酐的偏酯的縮合產物,例如聚乙烯脫水山梨醇單油酸酯。水性懸浮液還可以包括一種或多種防腐劑(如對羥基苯甲酸乙酯或對羥基苯甲酸丙酯)、抗氧化劑(如抗壞血酸)、著色劑、調味劑和/或甜味劑(如蔗糖、糖精或阿斯巴甜)。 在某些實施方式中,本公開的藥物組合物可以呈油性懸浮液形式,所述油性懸浮液通常包括於植物油(如花生油、蓖麻油、橄欖油、芝麻油或椰子油)中或礦物油(如液體石蠟)中的懸浮活性成分。油性懸浮液還可以包括增稠劑,如蜂蠟、硬石蠟或鯨蠟醇。可以添加甜味劑(如上文陳述的甜味劑)和調味劑以提供可口的口服製劑。這些組合物可以通過添加抗氧化劑(如抗壞血酸)來保存。 在某些實施方式中,本公開的藥物組合物可以呈水包油乳液形式。油相可以是植物油,如橄欖油或花生油;或礦物油,例如液體石蠟;或任何這些油的混合物。合適的乳化劑可以是例如天然存在的膠,如阿拉伯膠或黃芪膠;天然存在的磷脂,如大豆、卵磷脂、衍生自脂肪酸和己糖醇酸酐的酯或偏酯(例如,脫水山梨醇單油酸酯)和所述偏酯與環氧乙烷的縮合產物,如聚氧乙烯脫水山梨醇單油酸酯。乳液還可以包括甜味劑、調味劑和防腐劑。 在某些實施方式中,本文所提供的藥物組合物可以呈糖漿和酏劑形式,所述糖漿和酏劑可以包括甜味劑,如甘油、丙二醇、山梨醇、阿斯巴甜或蔗糖;緩和劑;防腐劑;調味劑和/或著色劑。 在一些實施方式中,本公開的藥物組合物可以呈用於注射施用的調配物的形式。 在某些實施方式中,本公開的藥物組合物可以呈無菌可注射製劑形式,如無菌可注射水性或油性懸浮液。這種懸浮液可以根據已知技術使用上文提到的那些合適的分散劑或潤濕劑和懸浮劑來調配。無菌可注射製劑還可以為在無毒腸胃外可接受的稀釋劑或溶劑中的無菌可注射溶液或懸浮液,如在1,3-丁二醇中的溶液或製備為凍幹粉劑。可以採用的可接受的媒劑和溶劑中包括水、林格氏溶液(Ringer's solution)和等滲氯化鈉溶液。另外,無菌不揮發性油常規地可以用作溶劑或懸浮介質。出於此目的,可以採用任何溫和的固定油,包括合成的甘油單酯或甘油二酯。另外,如油酸等脂肪酸同樣可以用於製備可注射物。 在一些實施方式中,本公開的藥物組合物可以呈用於吸入施用的調配物的形式。 在某些實施方式中,本公開的藥物組合物可以呈水性和非水性(例如,於碳氟化合物推進劑中)氣溶膠形式,所述水性和非水性氣溶膠包括任何合適的溶劑和任選地其它化合物,如但不限於穩定劑、抗微生物劑、抗氧化劑、pH調節劑、表面活性劑、生物可用性調節劑以及其組合。載體和穩定劑隨具體化合物的要求而變化,但通常包括非離子型表面活性劑(吐溫(Tween)、普朗尼克(Pluronic)或聚乙二醇)、無害蛋白質(如血清白蛋白)、脫水山梨醇酯、油酸、卵磷脂、如甘氨酸等氨基酸、緩衝劑、鹽、糖或糖醇。 在一些實施方式中,本公開的藥物組合物可以呈用於局部或經皮施用的調配物的形式。在某些實施方式中,本文所提供的藥物組合物可以呈乳膏、軟膏、凝膠和水性或油性溶液或懸浮液形式,所述乳膏、軟膏、凝膠和水性或油性溶液或懸浮液通常可以通過將活性成分與常規的局部可接受的賦形劑,如動物脂肪和植物脂肪、油、蠟、石蠟、澱粉、黃蓍膠、纖維素衍生物、聚乙二醇、矽酮、膨潤土、矽酸、滑石和氧化鋅或其混合物,一起調配而獲得。 在某些實施方式中,本文所提供的藥物組合物可以調配用於眼部施用。在某些實施方式中,本文所提供的藥物組合物可以呈眼用調配物的形式,如眼藥膏、粉末、溶液等。在某些實施方式中,眼用調配物是在舒適的pH和適當的緩衝液系統下製備的。 除上文所描述的那些代表性劑型以外,藥學上可接受的賦形劑和載體對於本領域技術人員而言通常是已知的並且因此包括在本公開中。在例如以下參考文獻中描述了此類賦形劑和載體:《雷明頓氏藥物科學(Remingtons Pharmaceutical Sciences)》, 新澤西州的馬克出版公司(Mack Pub.Co.), 新澤西州(New Jersey) (1991);《雷明頓:藥學科學與實踐(Remington: The Science and Practice of Pharmacy)》, 編輯,費城科學大學(University of the Sciences in Philadelphia), 第21版, LWW (2005),所述參考文獻通過引用併入本文。 本文所提供的化合物的劑量方案將根據已知因素而變化,如特定藥劑的藥效學特徵和其施用方式和途徑;接受者的物種、年齡、性別、健康狀況、身體狀況和重量;症狀的性質和程度;並行治療的種類;治療頻率;施用途徑、患者的腎功能和肝功能以及期望的效果。醫生或獸醫可以確定和開具預防、對抗或阻止病症進展所需的藥物的有效量。 在一些實施方式中,本公開的藥物組合物可以被調配成使得可以施用0.001 mg/kg體重/天至1000 mg/kg體重/天,例如0.01 mg/kg體重/天至800 mg/kg體重/天、0.01 mg/kg體重/天至700 mg/kg體重/天、0.01 mg/kg體重/天至600 mg/kg體重/天、0.01 mg/kg體重/天至500 mg/kg體重/天、0.01 mg/kg體重/天至400 mg/kg體重/天、0.01 mg/kg體重/天至300 mg/kg體重/天、0.1 mg/kg體重/天至200 mg/kg體重/天、0.1 mg/kg體重/天至150 mg/kg體重/天、0.1 mg/kg體重/天至100 mg/kg體重/天、0.5 mg/kg體重/天至100 mg/kg體重/天、0.5 mg/kg體重/天至80 mg/kg體重/天、0.5 mg/kg體重/天至60 mg/kg體重/天、0.5 mg/kg體重/天至50 mg/kg體重/天、1 mg/kg體重/天至50 mg/kg體重/天、1 mg/kg體重/天至45 mg/kg體重/天、1 mg/kg體重/天至40 mg/kg體重/天、1 mg/kg體重/天至35 mg/kg體重/天、1 mg/kg體重/天至30 mg/kg體重/天、1 mg/kg體重/天至25 mg/kg體重/天的劑量的本文所提供的化合物或其藥學上可接受的鹽。在一些情況下,低於前述範圍的下限的劑量水準可能綽綽有餘,而在其它情況下,可以採用較大劑量而不會引起任何有害副作用,條件是此類較大劑量首先分成若干小劑量以供全天施用。有關施用途徑和劑量方案的另外的資訊,參見《綜合藥物化學(Comprehensive Medicinal Chemistry)》第5卷 第25.3章(Corwin Hansch;編輯委員會主席), 培格曼出版社 1990,所述參考文獻特別地通過引用併入本文。 在一些實施方式中,本公開的藥物組合物可以被調配成單一劑型。本文所提供的化合物在單一劑型中的量將根據所治療的受試者和具體的施用模式而變化。 在一些實施方式中,適合施用的劑型每劑量單位可以含有約1 mg至約1000 mg的活性成分。在這些藥物組合物中,基於組合物的總重量,活性成分將通常以約0.1-95重量%的量存在。 在一些實施方式中,本公開的藥物組合物可以被調配成短效、快速釋放、長效和持續釋放形式。因此,本公開的藥物調配物也可以被調配成用於控制釋放或緩慢釋放。 在一些實施方式中,每天、每隔一天、每兩天、每三天、一週一次、一周兩次、一週三次或每兩週一次向受試者施用一定劑量的本文所提供的化合物或本文所提供的藥物組合物。如果需要的話,活性化合物的有效日劑量可以作為在一天中以適當的間隔單獨施用,任選地以單位劑型施用的兩個、三個、四個、五個、六個或更多個亞劑量施用。在一些實施方式中,施用一定劑量的本文所提供的化合物或本文所提供的藥物組合物,持續2天、3天、5天、7天、14天、21天、1個月、2個月、2.5個月、3個月、4個月、5個月、6個月或更長時間。 在另外的方面,還提供了獸用組合物,所述獸用組合物包含一種或多種本公開的分子或化合物或其藥學上可接受的鹽和獸用載體。獸用載體是可用於施用組合物的目的的材料,並且可以是在其它方面呈惰性或在獸醫學領域中可接受並且與活性成分相容的固體、液體或氣態材料。這些獸用組合物可以腸胃外、口服或通過任何其它期望的途徑來施用。 藥物組合物或獸用組合物可以取決於用於施用藥物的方法而以各種方式包裝。例如,用於分配的製品可以包括存放有適當形式的組合物的容器。合適的容器為本領域技術人員所熟知,並且包括如瓶子(塑膠和玻璃)、藥囊、安瓿瓶、塑膠袋、金屬筒等材料。容器還可以包括防拆組合件以防止輕易獲取包裝的內含物。另外,容器具有放置其上的描述容器的內含物的標籤。標籤還可以包括適當的警告。組合物還可以包裝於單位劑量或多劑量容器中,例如密封的安瓿瓶和小瓶中,並且可以儲存在僅需要緊接著在使用之前添加無菌液體載體,例如注射用水的冷凍乾燥(凍幹)條件下。臨時注射溶液和懸浮液由先前所描述的種類的無菌粉末、顆粒和片劑製備。 在一些實施方式中,包含一種或多種本文所提供的化合物或其藥學上可接受的鹽的本公開的藥物組合物進一步包含一種或多種另外的治療活性劑。 所述另外的治療活性劑具有與本文所提供的化合物互補的活性,使得其不會對彼此有不利影響。此類藥劑以對預期目的有效的量適當地組合在一起存在。 在某些實施方式中,所述另外的治療劑選自:EGFR TKI、EGFR抗體、MEK抑制劑、c-MET抑制劑、有絲分裂激酶抑制劑、免疫治療劑、抗血管生成劑、凋亡誘導劑、mTOR抑制劑、組蛋白去乙醯化酶抑制劑、IL6抑制劑、JAK抑制劑。 EGFR TKI的實例包括但不限於例如阿法替尼、埃羅替尼、吉非替尼、拉帕替尼(lapatinib)、達可替尼(dacomitinib)、奧希替尼(osimertinib)、奧莫替尼(olmutinib)、那紮替尼(nazartinib)和AC0010。 EGFR抗體的實例包括例如西妥昔單抗(cetuximab)、帕尼單抗(panitumumab)和耐昔妥珠單抗(necitumumab)。 MEK抑制劑的實例包括曲美替尼(trametinib)、考比替尼(cobimetinib)、貝美替尼(binimetinib)、司美替尼(selumetinib)和瑞法替尼(refametinib)。 c-MET抑制劑的實例包括例如賽沃替尼(savolitinib)、卡博替尼(cabozantinib)、福瑞替尼(foretinib)和MET抗體(如依瑪妥珠單抗(emibetuzumab))。 有絲分裂激酶抑制劑的實例包括CDK4/6抑制劑,如帕博西尼(palbociclib)、瑞博西尼(ribociclib)、阿貝西利(abemaciclib)。 免疫治療劑的實例包括免疫檢查點抑制劑,如抗CTLA4 mAb、抗PD1 mAb、抗PD-L1 mAb、抗PD-L2 mAb、抗LAG3 mAb、抗TM3 mAb,優選的抗PD1 mAb、伊匹單抗(ipilimumab)、納武單抗(nivolumab)、派姆單抗(pembrolizumab)、阿特朱單抗(atezolizumab)、阿維單抗(avelumab)、德瓦魯單抗(durvalumab)、匹地利珠單抗(pidilizumab)、PDR-001和免疫調節劑,如CD73抑制劑或CD73抑制性抗體。 抗血管生成劑的實例包括例如貝伐單抗(bevacizumab)、尼達尼布(nintedanib)。 細胞凋亡誘導劑的實例包括Bcl-2抑制劑(如維奈托克(venetoclax)、奧巴托克斯(obatoclax)、納威托克斯(navitoclax))、Mcl-1抑制劑(如AZD-5991、AMG-176、S-64315)。 mTOR抑制劑的實例包括例如雷帕黴素(rapamycin)、替西羅莫司(temsirolimus)、依維莫司(everolimus)、利達氟奧司(ridaforolimus)。 組蛋白脫乙醯酶抑制劑的實例包括例如帕比司他(panobinostat)、恩替諾特(entinostat)、羅米地辛(romidepsin)和伏立諾他(vorinostat)。 IL6抑制劑的實例包括例如托珠單抗(tocilizumab)、司妥昔單抗(siltuximab)、奧洛組單抗(olokizumab)、艾西莫單抗(elsilimomab)、克拉紮珠單抗(clazakizumab)、西魯庫單抗(sirukumab)、樂維利單抗(levilimab)、ARGX-109、FE301、FM101。 JAK抑制劑的實例包括例如巴瑞克替尼(baricitinib)、盧梭替尼(ruxolitinib)、托法替尼(tofacitinib)、奧拉替尼(oclacitinib)、巴瑞克替尼、培菲替尼(peficitinib)、菲達替尼(fedratinib)、烏帕替尼(upadacitinib)、非戈替尼(filgotinib)、迪高替尼(delgocitinib)、阿布西替尼(abrocitinib)。 一種或多種另外的治療劑可以與本文所提供的化合物同時或依次施用。依次施用包括在本文所提供的化合物之前或之後施用。在一些實施方式中,所述一種或多種另外的治療劑可以在與本文所提供的化合物相同的組合物中施用。在其它實施方式中,在施用另外的治療劑和本文所提供的化合物之間可以存在時間間隔。 在一些實施方式中,將另外的治療劑與本文所提供的化合物一起施用可以使得能夠降低其它治療劑的劑量和/或以較低頻率的間隔施用。 用於治療的方法 本公開的化合物和包含所述化合物的藥物組合物能夠抑制EGFR,因此可以用於抑制有需要的受試者中的EGFR活性,以及用於預防或治療EGFR相關病症。 在另外的方面,本公開提供了一種治療EGFR相關病症的方法,所述方法包括向有需要的受試者施用有效量的本文所提供的化合物或其藥學上可接受的鹽或藥物組合物。 如本文所使用的,術語“治療(treating/treatment)”或“療法”旨在具有其正常含義,即,處理疾病,以便完全或部分地緩解其症狀中的一種、一些或所有症狀,或糾正或補償潛在病理,由此實現有益的或期望的臨床結果。出於本公開的目的,有益的或期望的臨床結果包括但不限於症狀的緩解、疾病程度的減輕、疾病狀態的穩定(即,不惡化)、疾病進展的延緩或減慢、疾病狀態的改善或緩和以及緩解(無論是部分的還是全部的),無論是可檢測的還是不可檢測的。“治療”還可以意指與未接受療法的情況下的預期存活期相比,存活期延長。需要療法的情況包括已患有病狀或病症的情況以及易於患上病狀或病症的情況或要預防病狀或病症的情況。 如本文所使用的,術語“預防(preventing、prevention或prophylaxis)”旨在具有其正常含義並且包括用於預防疾病發展的一級預防和疾病已經發展並且暫時地或永久地保護患者以防疾病加劇或惡化或罹患與疾病相關的新症狀的二級預防。 在一些實施方式中,本文所提供的化合物或其藥學上可接受的鹽和組合物可以用於治療多種EGFR相關病症,包括癌症、自身免疫性疾病等。 在某些實施方式中,本文所提供的化合物或其藥學上可接受的鹽和組合物可以用於治療癌症,包括肺癌(例如非小細胞肺癌)、腦癌、結直腸癌、膀胱癌、尿路上皮癌、乳腺癌、前列腺癌、卵巢癌、頭頸癌、胰腺癌、胃癌和間皮瘤,包括轉移(特別是腦轉移)等。 在某些實施方式中,本文所提供的化合物或其藥學上可接受的鹽和組合物可以用於治療自身免疫性疾病,包括類風濕性關節炎、移植物抗宿主病、系統性紅斑狼瘡(SLE)、硬皮病、多發性硬化症、糖尿病、器官排斥、炎性腸病、牛皮癬以及其它的痛苦。 受試者的施用濃度和施用途徑將因要治療的EGFR相關病症而異。在某些實施方式中,所述施用通過選自由以下組成的組的途徑進行:腸胃外、腹膜內、皮內、心內、心室內、顱內、腦脊髓內、滑膜內、鞘內施用、肌肉內注射、玻璃體內注射、靜脈內注射、動脈內注射、口服、口腔、舌下、經皮、局部、氣管內、直腸內、皮下和眼部施用。 本公開提供的各個方面的實施方式也在以下段落中的任何段落中描述。 實施方式1:一種式(I)的化合物: (I) 或其藥學上可接受的鹽,其中: 環A選自由以下組成的組:環烷基、雜環基、芳基和雜芳基; 環B選自由以下組成的組:環烷基、雜環基、芳基和雜芳基; L 1選自由以下組成的組:鍵、烷基、烯基、炔基、雜烷基、雜烯基和雜炔基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基和所述雜炔基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基和氨基; L 2選自由以下組成的組:鍵、N(R A)、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個R B取代; 其中R A選自由以下組成的組:氫、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基、烷基和鹵代烷基; 其中每個R B獨立地選自由以下組成的組:羥基、鹵素、氰基、氨基、烷基、烯基、炔基、雜烷基、雜烯基、環烷基、雜環基、芳基和雜芳基; L 3選自由以下組成的組:鍵、烷基、烯基、炔基、雜烷基、雜烯基和雜炔基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基和所述雜炔基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基和氨基; L 4選自O、S或N(R C); 其中R C選自由以下組成的組:氫、烷基、烯基、炔基、鹵代烷基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基; 每個R 1獨立地選自由以下組成的組:羥基、鹵素、氰基、氨基、-N(R D) 2、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個R E取代; 其中每個R D獨立地選自由以下組成的組:氫、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、  -N(R F) 2或-OR G; 其中每個R E獨立地選自由以下組成的組:氫、羥基、鹵素、氰基、氨基、-N(R F) 2、-烷基-N(R F) 2、-C(O)OR G、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基和烷基; 其中R F和R G中的每一個獨立地選自由以下組成的組:烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基; 每個R 2獨立地選自由以下組成的組:氫、羥基、鹵素、氰基、氨基、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基、烷基和鹵代烷基; R 3選自由以下組成的組:氫、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基和雜環基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基和所述雜環基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基、烷基和鹵代烷基; m是0至5的整數;並且 n是0至4的整數。 實施方式2. 根據實施方式1所述的化合物或其藥學上可接受的鹽,其中環A是芳基或雜芳基。 實施方式3. 根據實施方式2所述的化合物或其藥學上可接受的鹽,其中環A選自由以下組成的組:呋喃基、苯硫基、吡咯基、苯基、吡啶基、吡喃基、嘧啶基、噠嗪基、吡嗪基和四氫異喹啉基。 實施方式4. 根據實施方式3所述的化合物或其藥學上可接受的鹽,其中環A選自由以下組成的組: 。 實施方式5. 根據實施方式1所述的化合物或其藥學上可接受的鹽,其中環B是芳基或雜芳基。 實施方式6. 根據實施方式5所述的化合物或其藥學上可接受的鹽,其中環B是苯基或吡唑基。 實施方式7. 根據實施方式6所述的化合物或其藥學上可接受的鹽,其中環B是 。 實施方式8. 根據實施方式1所述的化合物或其藥學上可接受的鹽,其中L 1是鍵。 實施方式9. 根據實施方式8所述的化合物或其藥學上可接受的鹽,其中L 1是烷基。 實施方式10. 根據實施方式9所述的化合物或其藥學上可接受的鹽,其中L 1選自由以下組成的組: ,其中L 1的*端與L 2連接。 實施方式11. 根據實施方式1所述的化合物或其藥學上可接受的鹽,其中L 2是鍵。 實施方式12. 根據實施方式1所述的化合物或其藥學上可接受的鹽,其中L 2是N(R A),並且R A選自烷基或雜環基,其中所述烷基或所述雜環基任選地被一個或多個鹵素或烷基取代。 實施方式13. 根據實施方式12所述的化合物或其藥學上可接受的鹽,其中R A是乙基、二氟乙基、三氟乙基或氧雜環丁烷基。 實施方式14. 根據實施方式1所述的化合物或其藥學上可接受的鹽,其中L 2是任選地被一個或多個R B取代的環烷基。 實施方式15. 根據實施方式14所述的化合物或其藥學上可接受的鹽,其中L 2,其任選地被一個或多個R B取代。 實施方式16. 根據實施方式1所述的化合物或其藥學上可接受的鹽,其中L 2是任選地被一個或多個R B取代的雜環基。 實施方式17. 根據實施方式16所述的化合物或其藥學上可接受的鹽,其中所述雜環基選自由以下組成的組: , 其中的每一個任選地被一個或多個R B取代,其中L 2的*端與L 3連接。 實施方式18. 根據實施方式16或17所述的化合物或其藥學上可接受的鹽,其中R B是烷基。 實施方式19. 根據實施方式19所述的化合物或其藥學上可接受的鹽,其中R B是甲基。 實施方式20. 根據實施方式1所述的化合物或其藥學上可接受的鹽,其中L 2選自由以下組成的組: ,其中L 2的*端與L 3連接。 實施方式21. 根據實施方式1所述的化合物或其藥學上可接受的鹽,其中L 3是烷基。 實施方式22. 根據實施方式21所述的化合物或其藥學上可接受的鹽,其中L 3是乙基。 實施方式23. 根據實施方式1所述的化合物或其藥學上可接受的鹽,其中L 4是O或NH。 實施方式24. 根據實施方式1所述的化合物或其藥學上可接受的鹽,其中L 1是鍵,並且L 2是鍵或任選地被一個或多個R B取代的雜環基。 實施方式25. 根據實施方式1所述的化合物或其藥學上可接受的鹽,其中L 1是烷基,並且L 2是鍵、N(R A)或任選地被一個或多個R B取代的環烷基。 實施方式26. 根據實施方式1所述的化合物或其藥學上可接受的鹽,其中R 1是鹵素,並且m是1。 實施方式27. 根據實施方式26所述的化合物或其藥學上可接受的鹽,其中R 1是溴。 實施方式28. 根據實施方式1所述的化合物或其藥學上可接受的鹽,其中R 1是-N(R D) 2,並且m是1。 實施方式29. 根據實施方式28所述的化合物或其藥學上可接受的鹽,其中每個R D獨立地是任選地被一個或多個獨立地選自-N(R F) 2或-OR G的基團取代的烷基。 實施方式30. 根據實施方式29所述的化合物或其藥學上可接受的鹽,其中每個R D獨立地是甲基、甲氧基乙基或N,N-二甲基氨基丙基。 實施方式31. 根據實施方式1所述的化合物或其藥學上可接受的鹽,其中R 1是任選地被一個或多個R E取代的雜環基,並且m是1。 實施方式32. 根據實施方式31所述的化合物或其藥學上可接受的鹽,其中所述雜環基選自由以下組成的組: , 其中的每一個任選地被一個或多個R E取代。 實施方式33. 根據實施方式31所述的化合物或其藥學上可接受的鹽,其中R E是鹵素。 實施方式34. 根據實施方式32所述的化合物或其藥學上可接受的鹽,其中R E是F。 實施方式35. 根據實施方式31所述的化合物或其藥學上可接受的鹽,其中R E是-N(R F) 2、-烷基-N(R F) 2、      -C(O)OR G或任選地被一個或多個鹵素取代的烷基。 實施方式36. 根據實施方式35所述的化合物或其藥學上可接受的鹽,其中R F和R G中的每一個是烷基。 實施方式37. 根據實施方式35所述的化合物或其藥學上可接受的鹽,其中R F和R G中每一個是C 1-3烷基。 實施方式38. 根據實施方式35所述的化合物或其藥學上可接受的鹽,其中R E選自由以下組成的組:-N(CH 3) 2、-C 1-3烷基-N(CH 3) 2、-C(O)O(叔丁基)、甲基、乙基或三氟乙基。 實施方式39. 根據實施方式31所述的化合物或其藥學上可接受的鹽,其中R E是環烷基或雜環基,其中所述環烷基和所述雜環基任選地被一個或多個烷基或鹵素取代。 實施方式40. 根據實施方式39所述的化合物或其藥學上可接受的鹽,其中R E選自環丙基、嗎啉基、哌嗪基、氧雜環丁烯基或氮雜環丁烷基,其中的每一個任選地被一個或多個烷基或鹵素取代。 實施方式41. 根據實施方式1所述的化合物或其藥學上可接受的鹽,其中R 1選自由以下組成的組: 。 實施方式42. 根據實施方式1所述的化合物或其藥學上可接受的鹽,其中R 2是鹵素,並且n是1或2。 實施方式43. 根據實施方式42所述的化合物或其藥學上可接受的鹽,其中R 2是F。 實施方式44. 根據實施方式1所述的化合物或其藥學上可接受的鹽,其中R 2是烷基或環烷基,其中所述烷基和環烷基任選地被一個或多個鹵素取代。 實施方式45. 根據實施方式44所述的化合物或其藥學上可接受的鹽,其中R 2是甲基、乙基、環丙基或三氟乙基。 實施方式46. 根據實施方式1所述的化合物或其藥學上可接受的鹽,其中R 3是烷基或環烷基。 實施方式47. 根據實施方式46所述的化合物或其藥學上可接受的鹽,其中R 3是甲基、乙基或環丙基。 實施方式48. 根據實施方式1所述的化合物或其藥學上可接受的鹽,其中m是0、1或2。 實施方式49. 根據實施方式1所述的化合物或其藥學上可接受的鹽,其中n是0或1。 實施方式50. 根據實施方式1所述的化合物或其藥學上可接受的鹽,其中所述化合物選自由以下組成的組: 。 實施方式51. 一種藥物組合物,其包含根據實施方式1至50中任一項所述的化合物或其藥學上可接受的鹽以及藥學上可接受的載體。 實施方式52. 一種抑制有需要的受試者中EGFR活性的方法,所述方法包括向所述受試者施用有效量的根據實施方式1至50中任一項所述的化合物或其藥學上可接受的鹽或根據實施方式51所述的藥物組合物。 實施方式53. 一種治療有需要的受試者中EGFR相關病症的方法,所述方法包括向所述受試者施用有效量的根據實施方式1至50中任一項所述的化合物或其藥學上可接受的鹽或向所述受試者施用根據實施方式51所述的藥物組合物。 實施方式54. 根據實施方式53所述的方法,其中所述EGFR相關病症是自身免疫性疾病或癌症。 實施方式55. 根據實施方式54所述的方法,其中所述癌症選自由以下組成的組:肺癌、腦癌、結直腸癌、膀胱癌、尿路上皮癌、乳腺癌、前列腺癌、卵巢癌、頭頸癌、胰腺癌、胃癌和間皮瘤,包括轉移(特別是腦轉移)等。 實施方式56. 根據實施例53至55中任一項所述的方法,其中所述化合物與一種或多種另外的治療劑同時、單獨或依次施用。 實施方式57. 根據實施方式56所述的方法,其中所述一種或多種另外的治療劑選自由以下組成的組:EGFR TKI、EGFR抗體、MEK抑制劑、c-MET抑制劑、有絲分裂激酶抑制劑、免疫治療劑、抗血管生成劑、凋亡誘導劑、mTOR抑制劑、組蛋白去乙醯化酶抑制劑、IL6抑制劑和JAK抑制劑。 此外,本公開提供的各個方面的實施方式也在以下段落中的任何段落中描述。 項目1. 一種式(II)化合物 (II) 或其藥學上可接受的鹽,其中: 環A 1是7-12元環烷基、7-12元雜環基、7-12元芳基或7-12元雜芳基; 環B選自由以下組成的組:環烷基、雜環基、芳基和雜芳基; L 1選自由以下組成的組:鍵、烷基、烯基、炔基、雜烷基、雜烯基和雜炔基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基和所述雜炔基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基和氨基; L 2選自由以下組成的組:鍵、N(R A)、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個R B取代; 其中R A選自由以下組成的組:氫、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基、烷基和鹵代烷基; 其中每個R B獨立地選自由以下組成的組:羥基、鹵素、氰基、氨基、烷基、烯基、炔基、雜烷基、雜烯基、環烷基、雜環基、芳基和雜芳基; L 3選自由以下組成的組:鍵、烷基、烯基、炔基、雜烷基、雜烯基和雜炔基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基和所述雜炔基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基和氨基; L 4選自O、S或N(R C); 其中R C選自由以下組成的組:氫、烷基、烯基、炔基、鹵代烷基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基; 每個R 1獨立地選自由以下組成的組:羥基、鹵素、氰基、氨基、-N(R D) 2、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個R E取代; 其中每個R D獨立地選自由以下組成的組:氫、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、  -N(R F) 2或-OR G; 其中每個R E獨立地選自由以下組成的組:氫、羥基、鹵素、氰基、氨基、-N(R F) 2、-烷基-N(R F) 2、-C(O)OR G、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基和烷基; 其中R F和R G中的每一個獨立地選自由以下組成的組:烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基; 每個R 2獨立地選自由以下組成的組:氫、羥基、鹵素、氰基、氨基、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基、烷基和鹵代烷基; 每個R 3選自由以下組成的組:氫、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基和雜環基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基和所述雜環基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基、烷基和鹵代烷基; m是0至5的整數; n是0至4的整數;並且 p是0至3的整數。 項目2. 根據專案1所述的化合物或其藥學上可接受的鹽,其中環A 1是7-12元雜芳基。 項目3. 根據專案2所述的化合物或其藥學上可接受的鹽,其中環A 1是四氫異喹啉基。 項目4. 根據專案3所述的化合物或其藥學上可接受的鹽,其中環A 1。 項目5. 根據前述專案中任一項所述的化合物或其藥學上可接受的鹽,其中m是0。 項目6. 根據專案1所述的化合物或其藥學上可接受的鹽,其中m是1。 項目7. 根據專案6所述的化合物或其藥學上可接受的鹽,其中R 1是任選地被一個或多個N(R F) 2取代的烷基。 項目8. 根據專案7所述的化合物或其藥學上可接受的鹽,其中R 1是任選地被一個或多個N(R F) 2取代的乙基。 項目9. 根據專案7或8所述的化合物或其藥學上可接受的鹽,其中R F是烷基。 項目10. 根據專案9所述的化合物或其藥學上可接受的鹽,其中R F是甲基。 項目11. 根據專案6所述的化合物或其藥學上可接受的鹽,其中R 1是雜環基。 項目12. 根據專案11所述的化合物或其藥學上可接受的鹽,其中R 1是氧雜環丁烷基。 項目13. 根據專案12所述的化合物或其藥學上可接受的鹽,其中R 1。 項目14. 根據專案1所述的化合物或其藥學上可接受的鹽,其中L 1是烷基。 項目15. 根據專案14所述的化合物或其藥學上可接受的鹽,其中L 1,其中L 1的*端與L 2連接。 項目16. 根據專案1所述的化合物或其藥學上可接受的鹽,其中L 2是鍵。 項目17. 根據專案1所述的化合物或其藥學上可接受的鹽,其中L 3是烷基。 項目18. 根據專案11所述的化合物或其藥學上可接受的鹽,其中L 3是乙基。 項目19. 根據專案1所述的化合物或其藥學上可接受的鹽,其中L 4是O或NH。 項目20. 根據專案1所述的化合物或其藥學上可接受的鹽,其中環B是芳基或雜芳基。 項目21. 根據專案20所述的化合物或其藥學上可接受的鹽,其中環B是苯基、吡啶基 或吡唑基。 項目22. 根據專案21所述的化合物或其藥學上可接受的鹽,其中環B選自由以下組成的組: 。 項目23. 根據專案1所述的化合物或其藥學上可接受的鹽,其中n是1。 項目24. 根據專案1所述的化合物或其藥學上可接受的鹽,其中R 2是烷基。 項目25. 根據專案24所述的化合物或其藥學上可接受的鹽,其中R 2是甲基。 項目26. 根據專案1所述的化合物或其藥學上可接受的鹽,其中R 3是烷基。 項目27. 根據專案26所述的化合物或其藥學上可接受的鹽,其中R 3是甲基。 項目28. 根據專案1所述的化合物或其藥學上可接受的鹽,其中m是0或1。 項目29. 根據專案1所述的化合物或其藥學上可接受的鹽,其中n是1。 項目30. 根據專案1所述的化合物或其藥學上可接受的鹽,其中p是1。 項目31. 一種式的(III)的化合物: (III) 或其藥學上可接受的鹽,其中: 環A選自由以下組成的組:環烷基、雜環基、芳基和雜芳基; 環B 1是7-12元環烷基、7-12元雜環基、7-12元芳基或7-12元雜芳基; L 1選自由以下組成的組:鍵、烷基、烯基、炔基、雜烷基、雜烯基和雜炔基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基和所述雜炔基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基和氨基; L 2選自由以下組成的組:鍵、N(R A)、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個R B取代; 其中R A選自由以下組成的組:氫、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基、烷基和鹵代烷基; 其中每個R B獨立地選自由以下組成的組:羥基、鹵素、氰基、氨基、烷基、烯基、炔基、雜烷基、雜烯基、環烷基、雜環基、芳基和雜芳基; L 3選自由以下組成的組:鍵、烷基、烯基、炔基、雜烷基、雜烯基和雜炔基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基和所述雜炔基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基和氨基; L 4選自O、S或N(R C); 其中R C選自由以下組成的組:氫、烷基、烯基、炔基、鹵代烷基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基; 每個R 1獨立地選自由以下組成的組:羥基、鹵素、氰基、氨基、-N(R D) 2、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個R E取代; 其中每個R D獨立地選自由以下組成的組:氫、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、  -N(R F) 2或-OR G; 其中每個R E獨立地選自由以下組成的組:氫、羥基、鹵素、氰基、氨基、-N(R F) 2、-烷基-N(R F) 2、-C(O)OR G、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基和烷基; 其中R F和R G中的每一個獨立地選自由以下組成的組:烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基; 每個R 2獨立地選自由以下組成的組:氫、羥基、鹵素、氰基、氨基、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基、烷基和鹵代烷基; 每個R 3選自由以下組成的組:氫、鹵素、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基和雜環基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基和所述雜環基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基、烷基和鹵代烷基; m是0至5的整數; n是0至4的整數;並且 p是0至3的整數。 項目32. 根據專案31所述的化合物或其藥學上可接受的鹽,其中環A是芳基或雜芳基。 項目33. 根據專案32所述的化合物或其藥學上可接受的鹽,其中環A選自由以下組成的組:呋喃基、苯硫基、吡咯基、苯基、吡啶基、吡喃基、嘧啶基、噠嗪基、吡嗪基和四氫異喹啉基。 項目34. 根據專案31所述的化合物或其藥學上可接受的鹽,其中m是0。 項目35. 根據專案31所述的化合物或其藥學上可接受的鹽,其中R 1是鹵素,並且m是1。 項目36. 根據專案35所述的化合物或其藥學上可接受的鹽,其中R 1是溴。 項目37. 根據專案31所述的化合物或其藥學上可接受的鹽,其中R 1是-N(R D) 2,並且m是1。 項目38. 根據專案37所述的化合物或其藥學上可接受的鹽,其中每個R D獨立地是任選地被一個或多個獨立地選自-N(R F) 2或-OR G的基團取代的烷基。 項目39. 根據專案38所述的化合物或其藥學上可接受的鹽,其中每個R D獨立地是甲基、甲氧基乙基或N,N-二甲基氨基丙基。 項目40. 根據專案31所述的化合物或其藥學上可接受的鹽,其中R 1是任選地被一個或多個R E取代的雜環基,並且m是1。 項目41. 根據專案40所述的化合物或其藥學上可接受的鹽,其中所述雜環基選自由以下組成的組: , 其中的每一個任選地被一個或多個R E取代。 項目42. 根據專案40所述的化合物或其藥學上可接受的鹽,其中R E是鹵素。 項目43. 根據專案42所述的化合物或其藥學上可接受的鹽,其中R E是F。 項目44. 根據專案40所述的化合物或其藥學上可接受的鹽,其中R E是-N(R F) 2、-烷基-N(R F) 2、-C(O)OR G或任選地被一個或多個鹵素取代的烷基。 項目45. 根據專案44所述的化合物或其藥學上可接受的鹽,其中R F和R G中的每一個是烷基。 項目46. 根據專案45所述的化合物或其藥學上可接受的鹽,其中R F和R G中每一個是C 1-3烷基。 項目47. 根據專案44所述的化合物或其藥學上可接受的鹽,其中R E選自由以下組成的組:-N(CH 3) 2、-C 1-3烷基-N(CH 3) 2、-C(O)O(叔丁基)、甲基、乙基或三氟乙基。 項目48. 根據專案40所述的化合物或其藥學上可接受的鹽,其中R E是環烷基或雜環基,其中所述環烷基和所述雜環基任選地被一個或多個烷基或鹵素取代。 項目49. 根據專案48所述的化合物或其藥學上可接受的鹽,其中R E選自環丙基、嗎啉、哌嗪、氧雜環丁烯基或氮雜環丁烷基,其中的每一個任選地被一個或多個烷基或鹵素取代。 項目50. 根據專案31所述的化合物或其藥學上可接受的鹽,其中R 1選自由以下組成的組 。 項目51. 根據專案31所述的化合物或其藥學上可接受的鹽,其中環B 1是7-12元雜芳基。 項目52. 根據專案51所述的化合物或其藥學上可接受的鹽,其中環B 1。 項目53. 根據專案31所述的化合物或其藥學上可接受的鹽,其中R 2是鹵素,並且n是1或2。 項目54. 根據專案53所述的化合物或其藥學上可接受的鹽,其中R 2是F。 項目55. 根據專案31所述的化合物或其藥學上可接受的鹽,其中R 2是烷基或環烷基,其中所述烷基和環烷基任選地被一個或多個鹵素取代。 項目56. 根據專案55所述的化合物或其藥學上可接受的鹽,其中R 2是甲基、乙基、環丙基或三氟乙基。 項目57. 根據專案31所述的化合物或其藥學上可接受的鹽,其中L 1是烷基。 項目58. 根據專案57所述的化合物或其藥學上可接受的鹽,其中L 1是丙基。 項目59. 根據專案31所述的化合物或其藥學上可接受的鹽,其中L 2是鍵。 項目60. 根據專案31所述的化合物或其藥學上可接受的鹽,其中L 3是烷基。 項目61. 根據專案60所述的化合物或其藥學上可接受的鹽,其中L 3是乙基。 項目62. 根據專案31所述的化合物或其藥學上可接受的鹽,其中L 4是O或NH。 項目63. 根據專案31所述的化合物或其藥學上可接受的鹽,其中R 3是烷基。 項目64. 根據專案63所述的化合物或其藥學上可接受的鹽,其中R 3是甲基。 項目65. 根據專案31所述的化合物或其藥學上可接受的鹽,其中R 3是鹵素。 項目66. 根據專案65所述的化合物或其藥學上可接受的鹽,其中R 3是Cl。 項目67. 根據專案31所述的化合物或其藥學上可接受的鹽,其中m是0。 項目68. 根據專案31所述的化合物或其藥學上可接受的鹽,其中n是0。 項目69. 根據專案31所述的化合物或其藥學上可接受的鹽,其中p是1。 項目70. 一種式(IV)的化合物: (IV) 或其藥學上可接受的鹽,其中: 環A選自由以下組成的組:環烷基、雜環基、芳基和雜芳基; 環B選自由以下組成的組:環烷基、雜環基、芳基和雜芳基; L 1選自由以下組成的組:鍵、烷基、烯基、炔基、雜烷基、雜烯基和雜炔基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基和所述雜炔基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基和氨基; L 21是7-12元環烷基或7-12元雜環基,其中所述環烷基和雜環基任選地被一個或多個R B取代; 其中每個R B獨立地選自由以下組成的組:羥基、鹵素、氰基、氨基、烷基、烯基、炔基、雜烷基、雜烯基、環烷基、雜環基、芳基和雜芳基; L 3選自由以下組成的組:鍵、烷基、烯基、炔基、雜烷基、雜烯基和雜炔基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基和所述雜炔基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基和氨基; L 4選自O、S或N(R C); 其中R C選自由以下組成的組:氫、烷基、烯基、炔基、鹵代烷基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基; 每個R 1獨立地選自由以下組成的組:羥基、鹵素、氰基、氨基、-N(R D) 2、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個R E取代; 其中每個R D獨立地選自由以下組成的組:氫、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、  -N(R F) 2或-OR G; 其中每個R E獨立地選自由以下組成的組:氫、羥基、鹵素、氰基、氨基、-N(R F) 2、-烷基-N(R F) 2、-C(O)OR G、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基和烷基; 其中R F和R G中的每一個獨立地選自由以下組成的組:烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基; 每個R 2獨立地選自由以下組成的組:氫、羥基、鹵素、氰基、氨基、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基、烷基和鹵代烷基; R 3選自由以下組成的組:氫、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基和雜環基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基和所述雜環基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基、烷基和鹵代烷基; m是0至5的整數; n是0至4的整數;並且 p是0至3的整數。 項目71. 根據專案70所述的化合物或其藥學上可接受的鹽,其中環A是芳基。 項目72. 根據專案71所述的化合物或其藥學上可接受的鹽,其中環A是苯基。 項目73. 根據專案70所述的化合物或其藥學上可接受的鹽,其中R 1是鹵素,並且m是1。 項目74. 根據專案73所述的化合物或其藥學上可接受的鹽,其中R 1是溴。 項目75. 根據專案70所述的化合物或其藥學上可接受的鹽,其中R 1是-N(R D) 2,並且m是1。 項目76. 根據專案75所述的化合物或其藥學上可接受的鹽,其中每個R D獨立地是任選地被一個或多個獨立地選自-N(R F) 2或-OR G的基團取代的烷基。 項目77. 根據專案76所述的化合物或其藥學上可接受的鹽,其中每個R D獨立地是甲基、甲氧基乙基或N,N-二甲基氨基丙基。 項目78. 根據專案70所述的化合物或其藥學上可接受的鹽,其中R 1是任選地被一個或多個R E取代的雜環基,並且m是1。 項目79. 根據專案78所述的化合物或其藥學上可接受的鹽,其中所述雜環基選自由以下組成的組: , 其中的每一個任選地被一個或多個R E取代。 項目80. 根據專案78所述的化合物或其藥學上可接受的鹽,其中R E是鹵素。 項目81. 根據專案80所述的化合物或其藥學上可接受的鹽,其中R E是F。 項目82. 根據專案78所述的化合物或其藥學上可接受的鹽,其中R E是-N(R F) 2、-烷基-N(R F) 2、-C(O)OR G或任選地被一個或多個鹵素取代的烷基。 項目83. 根據專案82所述的化合物或其藥學上可接受的鹽,其中R F和R G中的每一個是烷基。 項目84. 根據專案83所述的化合物或其藥學上可接受的鹽,其中R F和R G中每一個是C 1-3烷基。 項目85. 根據專案82所述的化合物或其藥學上可接受的鹽,其中R E選自由以下組成的組:-N(CH 3) 2、-C 1-3烷基-N(CH 3) 2、-C(O)O(叔丁基)、甲基、乙基或三氟乙基。 項目86. 根據專案78所述的化合物或其藥學上可接受的鹽,其中R E是環烷基或雜環基,其中所述環烷基和所述雜環基任選地被一個或多個烷基或鹵素取代。 項目87. 根據專案86所述的化合物或其藥學上可接受的鹽,其中R E選自環丙基、嗎啉、哌嗪、氧雜環丁烯基或氮雜環丁烷基,其中的每一個任選地被一個或多個烷基或鹵素取代。 專案88 根據專案70所述的化合物或其藥學上可接受的鹽,其中R 1選自由以下組成的組 。 項目89. 根據專案70所述的化合物或其藥學上可接受的鹽,其中環B是芳基或雜芳基。 項目90. 根據專案89所述的化合物或其藥學上可接受的鹽,其中環B是苯基、吡啶基或吡唑基。 項目91. 根據專案90所述的化合物或其藥學上可接受的鹽,其中環B選自由以下組成的組: 。 項目92. 根據專案70所述的化合物或其藥學上可接受的鹽,其中L 1是鍵。 項目93. 根據專案70所述的化合物或其藥學上可接受的鹽,其中L 21是是含有一個或多個選自N或O的雜原子的7-10元雜環基。 項目94. 根據專案93所述的化合物或其藥學上可接受的鹽,其中L 21選自由以下組成的組 ,其中L 21的*端與L 3連接。 項目95. 根據專案70所述的化合物或其藥學上可接受的鹽,其中L 3是烷基。 項目96. 根據專案95所述的化合物或其藥學上可接受的鹽,其中L 3是乙基。 項目97. 根據專案70所述的化合物或其藥學上可接受的鹽,其中L 4是O或NH。 項目98. 根據專案70所述的化合物或其藥學上可接受的鹽,其中R 1是任選地被一個或多個R E取代的雜環基,並且m是1。 項目99. 根據專案70所述的化合物或其藥學上可接受的鹽,其中R 3是烷基。 項目100. 根據專案99所述的化合物或其藥學上可接受的鹽,其中R 3是甲基。 項目101. 根據專案70所述的化合物或其藥學上可接受的鹽,其中m是0或1。 項目102. 根據專案70所述的化合物或其藥學上可接受的鹽,其中n是1。 項目103. 根據專案70所述的化合物或其藥學上可接受的鹽,其中p是1。 項目104. 一種化合物或其藥學上可接受的鹽,其中所述化合物選自由以下組成的組:  、       項目105. 一種藥物組合物,其包含根據專案1至104中任一項所述的化合物或其藥學上可接受的鹽以及藥學上可接受的載體。 項目106. 一種抑制有需要的受試者中EGFR活性的方法,所述方法包括向所述受試者施用有效量的根據專案1至104中任一項所述的化合物或其藥學上可接受的鹽或根據專案105所述的藥物組合物。 項目107. 一種治療有需要的受試者中EGFR相關病症的方法,所述方法包括向所述受試者施用有效量的根據專案1至104中任一項所述的化合物或其藥學上可接受的鹽或向所述受試者施用根據專案105所述的藥物組合物。 項目108. 根據專案107所述的方法,其中所述EGFR相關病症是自身免疫性疾病或癌症。 項目109. 根據專案108所述的方法,其中所述癌症選自由以下組成的組:肺癌、腦癌、結直腸癌、膀胱癌、尿路上皮癌、乳腺癌、前列腺癌、卵巢癌、頭頸癌、胰腺癌、胃癌和間皮瘤,包括轉移(特別是腦轉移)等。 項目110. 根據專案107至109中任一項所述的方法,其中所述化合物與一種或多種另外的治療劑同時、單獨或依次施用。 項目111. 根據專案110所述的方法,其中所述一種或多種另外的治療劑選自由以下組成的組:EGFR TKI、EGFR抗體、MEK抑制劑、c-MET抑制劑、有絲分裂激酶抑制劑、免疫治療劑、抗血管生成劑、凋亡誘導劑、mTOR抑制劑、組蛋白去乙醯化酶抑制劑、IL6抑制劑、JAK抑制劑。 實施例 以下進一步解釋本公開的通用方法。本公開的化合物可以通過本領域已知的方法製備。以下說明了本公開的優選化合物的詳細製備方法。然而,這些決不限制本公開的化合物的製備方法。 合成實施例 出於說明的目的,包括了以下實施例。本文所提供的實施例描述了本文所公開的化合物的合成以及用於製備所述化合物的中間體。然而,應理解,這些實施例並不限制本公開,並且僅意在表明實踐本公開的方法。本領域技術人員將認識到,可以容易地使所描述的化學反應適於製備本公開的多種其它化合物,並且用於製備本公開的化合物的可替代方法被視作處於本公開的範圍內。例如,通過對本領域技術人員而言顯而易見的修飾,例如通過適當地保護干擾基團、通過利用本領域已知的除所描述的試劑和構建塊之外的其它合適的試劑和構建塊和/或通過對反應條件進行常規修改,可以成功地合成根據本公開的非示例性化合物。此外,本領域技術人員還將理解,本文所描述的或化合物的單獨批次中的各個步驟可以組合。可替代地,本文所公開或本領域中已知的其它反應將被認為適用於製備本公開的其它化合物。因此,以下描述並非旨在限制本公開的範圍,而是由所附權利要求來指定。 化學、材料和通用方法 除非另有說明,否則所有試劑和溶劑都來自如藥石科技公司(PharmaBlock)、畢得醫藥科技公司(Bide Pharmatech)、上海升德醫藥公司(Shanghai Send Pharm)、奧德里奇公司(Aldrich)、西格瑪公司(Sigma)等商業供應商,並且無需進一步純化即可使用。乾燥有機溶劑(THF、DMF、1,4-二噁烷等)購自安徽澤升科技公司(Anhui Senrise Technology)、北京伊諾凱科技公司(Beijing innochem science & Technology)和百靈威科技公司(J & K Scientific),在氮氣下包裝在Sure/Seal瓶中。所有涉及空氣或濕氣敏感試劑的反應都在氮氣氣氛下進行。 1H NMR光譜在環境溫度下在布魯克(Bruker)AV III HD 400 MHz、布魯克AV NEO 400 MHz光譜儀上記錄。以百萬分率(ppm,δ單位)報告化學位移。資料包告如下:化學位移、質子數和多重性(s=單峰,d=雙重峰,dd=雙二重峰,dt=雙三重峰,t=三重峰,q=四重峰,br=寬,m=多重峰)。使用LCMS(島津(Shimadzu)20AD)通過254 nm處的UV檢測和低共振電噴霧模式(ESI)監測反應。如通過LCMS(3分鐘)測定的,大多數最終化合物純化到> 95%的純度。LCMS(3分鐘)方法使用以下:島津20AD光譜儀,Shim-pack Scepter C18-120 3.3 * 33 mm,3.0 um,30℃,其中流速為1.5毫升/分鐘;溶劑A為水+6.5 mM NH 4HCO 3+氫氧化氨(pH=10),溶劑B為乙腈;0.0-1.7分鐘,30% B-70% B;1.7-2.3分鐘,70% B-95% B;2.3-2.8分鐘,95% B;2.8-3.0分鐘,10% B。來自UV檢測器的流量被分流(1:3)到MS檢測器,所述檢測器配置有ESI作為電離源。CHIRAL方法使用以下:柱:CHIRALPAK IC‐3,0.46 * 5 cm,3 μm;在25℃下,流速為1.0毫升/分鐘;甲基叔丁基醚(0.2%二乙胺)/(乙醇/二氯甲烷=1/1)=20/80。SFC方法使用以下:柱:(R, R) -WHELK-01-Kromasil,5 * 25 cm,5 μm;流動相A:CO 2,流動相B:甲醇/乙腈= 1/1。 下文列出了本文所提供的化合物的合成中使用的縮寫: 中間體的合成 中間體M1 2-(5-羥基-1-甲基-1H-吡唑-4-基)-6-甲基異煙酸甲酯 步驟1. 2-甲基-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-3-酮(M1-1)的製備。在氮氣氣氛下在0℃下向2-甲基-1H-吡唑-3-酮(100 g,1019 mmol,1 equiv)和K 2CO 3(352 g,2548 mmol,2.5 equiv)於乙腈(1000 mL)中的攪拌溶液中逐滴添加2-(三甲基甲矽烷基)乙氧基甲基氯(254 g,1528 mmol,1.5 equiv)。將所得混合物在室溫下攪拌3 h。將反應在0℃下用水(200 mL)淬滅,並且將混合物用乙酸乙酯(3×500 mL)萃取。將合併的有機層用鹽水(3×500 mL)洗滌,並且經無水Na 2SO 4乾燥。過濾後,將濾液在減壓下濃縮。將殘餘物通過與石油醚(200 mL)一起研磨進行純化,以得到呈黃色固體的130 g 2-甲基-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-3-酮(M1-1)(55%)。LCMS: m/z (ESI), [2M+H] +=457.25。 1H NMR (DMSO - d 6, 400 MHz) δ 0.00 (9H, s), 0.80-0.85 (2H, m), 3.28 (3H, s), 3.40-3.50 (2H, m), 5.15 (2H, s), 5.29 (1H, d), 7.89 (1H, d)。 步驟2. 4-碘-2-甲基-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-3-酮(M1-2)的製備。在氮氣氣氛下在0℃下向2-甲基-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-3-酮(47 g,205.80 mmol,1 equiv)於乙腈(400 mL)中的攪拌混合物中添加 N-碘代丁二醯亞胺(50.93 g,226.39 mmol,1.1 equiv)。將所得混合物在氮氣氣氛下在0℃下攪拌1 h。將反應在0℃下用硫代硫酸鈉水溶液(400 mL)淬滅,並且將混合物用二氯甲烷(3×300 mL)萃取。將合併的有機層用鹽水(400 mL)洗滌,並經無水Na 2SO 4乾燥。過濾後,將濾液在減壓下濃縮。將殘餘物通過與石油醚(300 mL)一起研磨進行純化,以得到呈黃色固體的72 g 4-碘-2-甲基-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-3-酮(M1-2)(97%)。LCMS: m/z (ESI), [M+H] +=354.95。 1H NMR (DMSO-d 6, 400 MHz) δ -0.04 (9H, s), 0.75-0.86 (2H, m), 3.32 (3H, s), 3.39-3.48 (2H, m), 5.13 (2H, s), 8.13 (1H, s)。 步驟3. (6-氯-4-(甲氧基羰基)吡啶-2-基)硼酸(M1-3)的製備。將雙(頻哪醇合)二硼(11.5 g,453.7 mmol,1.3 equiv)於甲基叔丁基醚(100 mL)中的混合物升溫到80℃,並且攪拌0.5 h,然後冷卻到室溫,並且添加雙(1,5-環辛二烯)二-μ-甲氧基二銥(I)(1.16 g,17 mmol,0.05 equiv)和4-叔丁基-2-(4-叔丁基吡啶-2-基)吡啶(1.4 g,52 mmol,0.15 equiv)。將混合物在25℃下攪拌0.5 h,然後添加2-氯吡啶-4-甲酸甲酯(60 g,349 mmol,1.00 equiv),並且在80℃下攪拌16 h。將混合物冷卻到室溫,並且在減壓下濃縮,以得到呈棕色油的104 g 2-氯-6-(4,4,5,5-四甲基-1,3,2-二氧雜環戊硼烷-2-基)吡啶-4-甲酸甲酯(M1-3)(粗製物)。將此粗材料在不進行純化的情況下用於下一步驟。LCMS: m/z (ESI), [M+H] +=216.05。 步驟4. 2-氯-6-(2-甲基-3-氧代-1-((2-(三甲基甲矽烷基)乙氧基)甲基)-2,3-二氫-1H-吡唑-4-基)異煙酸甲酯(M1-4)的製備。將新鮮製備的2-氯-6-(4,4,5,5-四甲基-1,3,2-二氧雜環戊硼烷-2-基)吡啶-4-甲酸甲酯(104 g粗製物,約349 mmol,約1 equiv)、4-碘-2-甲基-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-3-酮(M1-2,124.30 g,350.877 mmol,1 equiv)、雙(金剛烷-1-基)(丁基)膦(12.58 g,35.088 mmol,0.1 equiv)、Cs 2CO 3(228.65 g,701.754 mmol,2 equiv)和Pd 2(dba) 3(9.6 g,10.4 mmol,0.03 equiv)於甲苯(600 mL)和水(150 mL)中的混合物在氮氣氣氛下在80℃下攪拌2 h。將混合物冷卻到室溫。將反應用水(800 mL)淬滅,並且將混合物用乙酸乙酯(3×600 mL)萃取。將合併的有機層用鹽水(3×500 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上使用石油醚/乙酸乙酯(1/4)洗脫進行純化,以得到呈黃色固體的38 g 2-氯-6-(2-甲基-3-氧代-1-((2-(三甲基甲矽烷基)乙氧基)甲基)-2,3-二氫-1H-吡唑-4-基)異煙酸甲酯(M1-4)(25%)。LCMS: m/z (ESI), [M+H] +=398.05。 步驟5. 2-甲基-6-(2-甲基-3-氧代-1-((2-(三甲基甲矽烷基)乙氧基)甲基)-2,3-二氫-1H-吡唑-4-基)異煙酸甲酯(M1-5)的製備。在氮氣氣氛下在室溫下向2-氯-6-(2-甲基-3-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-4-基)吡啶-4-甲酸甲酯(38 g,95 mmol,1.00 equiv)、Pd(dppf)Cl 2(3677 mg,5.02 mmol,0.05 equiv)、PCy 3 .HBF 4(10.5 g,28.64 mmol,0.3 equiv)和K 2CO 3(26.4 g,190 mmol,2 equiv)於1,4-二噁烷(500 mL)中的攪拌混合物中添加三甲基-1,3,5,2,4,6-三氧雜三硼環己烷(23.9 g,190 mmol,2 equiv)。將混合物在100℃下攪拌2 h,然後冷卻到室溫。將反應用水(300 mL)淬滅,並且將混合物用乙酸乙酯(3×200 mL)萃取。將合併的有機層用鹽水(3×300 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上使用石油醚/乙酸乙酯(1/3)洗脫進行純化,以得到呈黃色固體的34 g 2-甲基-6-(2-甲基-3-氧代-1-((2-(三甲基甲矽烷基)乙氧基)甲基)-2,3-二氫-1H-吡唑-4-基)異煙酸甲酯(M1-5)(92%)。LCMS: m/z (ESI), [M+H] +=378.15。 1H NMR (DMSO-d 6, 400 MHz) δ 0.05 (9H, s), 0.80-0.90 (2H, m), 2.53 (3H, s), 3.41 (3H, s), 3.50 (2H, t), 3.89 (3H, s), 5.37 (2H, s), 7.44 (1H, d), 8.52-8.60 (2H, m)。 步驟6. 2-(5-羥基-1-甲基-1H-吡唑-4-基)-6-甲基異煙酸甲酯(M1)的製備。將2-甲基-6-(2-甲基-3-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-4-基)吡啶-4-甲酸甲酯(34 g,90 mmol,1.00 equiv)和HCl於1,4-二噁烷(340 mL)中的溶液在氮氣氣氛下在室溫下攪拌2 h。將所得混合物在減壓下濃縮。將殘餘物用飽和NaHCO 3(水溶液)溶液(300 mL)處理,並且用乙酸乙酯(3×200 mL)萃取。將合併的有機層用鹽水(3×500 mL)洗滌,經無水Na 2SO 4乾燥,並且在減壓下濃縮,以得到呈紅色固體的20 g 2-(5-羥基-1-甲基-1H-吡唑-4-基)-6-甲基異煙酸甲酯(M1)(80%)。LCMS: m/z (ESI), [M+H] +=248.05。 1H NMR (DMSO-d 6, 400 MHz) δ 2.56 (3H, s), 3.48 (3H, s), 3.91 (3H, s), 7.31 (1H, s), 7.90-7.80 (2H, m)。 中間體M2 5-(5-羥基-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯 步驟1. 5-溴-1-甲基-6-氧代吡啶-3-甲酸甲酯(M2-1)的製備。在0℃下向5-溴-6-羥基吡啶-3-甲酸甲酯(20 g,86.19 mmol,1當量)於 N, N-二甲基甲醯胺(100 mL)中的混合物中添加NaH(60%)(4.14 g,172.39 mmol,2當量)。將混合物在氮氣氣氛下在0℃下攪拌60分鐘。緩慢添加CH 3I(13.46 g,94.81 mmol,1.1當量),並且將混合物在0℃下攪拌1 h。將反應在0℃下用水(1000 mL)淬滅,並且將混合物用乙酸乙酯(3×500 mL)萃取。將合併的有機層用鹽水(500 mL)洗滌,並且經無水Na 2SO 4乾燥。過濾後,將濾液在減壓下濃縮。將殘餘物通過與石油醚(200 mL)一起研磨進行純化,以得到呈白色固體的19 g 5-溴-1-甲基-6-氧代吡啶-3-甲酸甲酯(M2-1)(84%)。LCMS: m/z (ESI), [M+H] += 245.95。 1H NMR (DMSO-d 6, 400 MHz) δ 3.59 (3H, s), 3.81 (3H, s), 8.18 (1H, d), 8.62 (1H, d)。 步驟2. (5-(甲氧基羰基)-1-甲基-2-氧代-1,2-二氫吡啶-3-基)硼酸(M2-2)的製備。將5-溴-1-甲基-6-氧代吡啶-3-甲酸甲酯(30 g,121.92 mmol,1當量)、Pd(dppf)Cl 2 .CH 2Cl 2(8.92 g,12.2 mmol,0.1當量)、雙(頻哪醇)二硼(46.44 g,182.88 mmol,1.5當量)和KOAc(35.90 g,365.76 mmol,3當量)於1,4-二噁烷(600 mL)中的混合物在氮氣氣氛下在80℃下攪拌過夜。將混合物冷卻到室溫。將反應混合物在不進行後處理的情況下直接用於下一步驟。LCMS: m/z (ESI), [M+H] +=212.00。 步驟3. 1-甲基-5-(2-甲基-3-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-4-基)-6-氧代吡啶-3-甲酸甲酯(M2-3)的製備。在室溫下向通過上文所描述的反應新鮮製備的混合物中添加4-碘-2-甲基-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-3-酮(43 g,121.37 mmol,1.00當量)、Pd(dppf)Cl 2.CH 2Cl 2(7.91 g,9.71 mmol,0.08當量)、K 2CO 3(50.33 g,364.14 mmol,3當量)和H 2O(150 mL)。將混合物在氮氣氣氛下在80℃下攪拌2 h。將所得混合物冷卻到室溫,用水(200 mL)處理,並且用二氯甲烷(3×300 mL)萃取。將合併的有機層用鹽水(500 mL)洗滌,並且經無水Na 2SO 4乾燥。過濾後,將濾液在減壓下濃縮。將殘餘物在矽膠柱上使用石油醚/乙酸乙酯(1/8)洗脫進行純化,以得到呈棕色固體的30 g 1-甲基-5-(2-甲基-3-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-4-基)-6-氧代吡啶-3-甲酸甲酯(M2-3)(62%)。LCMS: m/z (ESI), [M+H] += 394.10。 1H NMR (DMSO-d 6, 400 MHz) δ -0.06 (9H, s), 0.82 (2H, t), 3.37 (3H, s), 3.47 (2H, t), 3.60 (3H, s), 3.81 (3H, s), 5.33 (2H, s), 8.41 (1H, d), 8.85 (1H, s), 9.21 (1H, d)。 步驟4. 5-(5-羥基-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M2)的製備。將1-甲基-5-(2-甲基-3-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-4-基)-6-氧代吡啶-3-甲酸甲酯(30 g,76.23 mmol,1當量)和HCl於1,4-二噁烷(50 mL)中的混合物在室溫下攪拌過夜。將所得混合物在減壓下濃縮。將殘餘物用二氯甲烷(50 mL)溶解,並且用K 2CO 3鹼化到pH 9。將所得混合物過濾,並且將濾餅用二氯甲烷(5×50 mL)洗滌。將所得溶液在減壓下濃縮。將殘餘物在矽膠柱上使用二氯甲烷/甲醇(10/1)洗脫進行純化,以得到呈棕色固體的15 g 5-(5-羥基-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M2)(74%)。LCMS: m/z (ESI), [M+H] +=264.10。 1H NMR (DMSO-d 6, 400 MHz) δ 3.52 (3H, s), 3.65 (3H, s), 3.83 (3H, s), 7.76 (1H, d), 8.09 (1H, s), 8.45 (1H, s), 13.34 (1H, s)。 中間體M3 (11 R)-16-溴-5,11,26-三甲基-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮 步驟1. (4 S)-4-苄基-3-[(2 R)-2-甲基戊-4-烯醯]-1,3-噁唑烷-2-酮(M3-1)的製備。在氮氣氣氛下在-78℃下向(4 S)-4-苄基-3-丙醯基-1,3-噁唑烷-2-酮(100 g,428.69 mmol,1當量)於四氫呋喃(640 mL)中的攪拌混合物中依次逐滴添加LiHMDS(471 mL,471.56 mmol,1.1當量)和丙烯基溴(207.4 g,1714.77 mmol,4當量)。將所得混合物在氮氣氣氛下在-50℃下攪拌2 h,並且升溫到室溫。將反應混合物用飽和NH 4Cl(水溶液)溶液(1000 mL)處理,並且用CH 2Cl 2(3×400 mL)萃取。將合併的有機層用鹽水(3×500 mL)洗滌,並且經無水Na 2SO 4乾燥。過濾後,將濾液在減壓下濃縮。將殘餘物通過矽膠柱色譜法使用石油醚/乙酸乙酯(40/1)洗脫來進行純化,以得到呈無色油的100 g (4 S)-4-苄基-3-[(2 R)-2-甲基戊-4-烯醯]-1,3-噁唑烷-2-酮(M3-1)(85%)。LCMS: m/z (ESI), [M+H] +=274.00。 1H NMR (DMSO-d 6, 400 MHz) δ 1.00-1.12 (3H, t), 2.08-2.49 (2H, m), 2.84-3.06 (2H, m), 3.64-3.78 (1H, m), 4.11-4.26 (1H, m), 4.29-4.39 (1H, m), 4.59-4.75 (1H, m), 4.98-5.18 (2H, m), 5.70-5.87 (1H, m), 7.13-7.38 (5H, m)。 步驟2. (2 R)-2-甲基戊-4-烯酸(M3-2)的製備。在0℃下向(4 S)-4-苄基-3-[(2 R)-2-甲基戊-4-烯醯]-1,3-噁唑烷-2-酮(M3-1,100 g,365.85 mmol,1當量)於四氫呋喃(800 mL)和H 2O(200 mL)中的攪拌混合物中添加LiOH .H 2O(46.05 g,1097.56 mmol,3當量)和H 2O 2(30%)(114.30 mL,4906 mmol,13.4當量)。將所得混合物在氮氣氣氛下在0℃下攪拌3 h。將反應用Na 2S 2O 3溶液(250 mL)淬滅,用HCl溶液酸化到pH=1,並且用CH 2Cl 2(3×400 mL)萃取。將合併的有機層用鹽水(3×500 mL)洗滌,並且經無水Na 2SO 4乾燥。過濾後,將濾液在減壓下濃縮。將粗產物在不進行進一步純化的情況下直接用於下一步驟。 步驟3. (2 R)- N,N-二苄基-2-甲基戊-4-烯醯胺(M3-3)的製備。在氮氣氣氛下在室溫下向(2 R)-2-甲基戊-4-烯酸(50 g,219.02 mmol,1當量)和二苄胺(43.2 g,219.022 mmol,1當量)於二噁烷(200 mL)中的攪拌溶液中添加 N,N-二異丙基乙胺(56.6 g,438.04 mmol,2當量)和N,N,N,N-四甲基-O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(99.94 g,262.82 mmol,1.2當量)。將所得混合物攪拌2 h。將反應用水(1000 mL)淬滅,並且將混合物用CH 2Cl 2(3×300 mL)萃取。將合併的有機層用鹽水(3×500 mL)洗滌,並且經無水Na 2SO 4乾燥。過濾後,將濾液在減壓下濃縮。將殘餘物在矽膠柱上使用石油醚/乙酸乙酯(40/1)洗脫進行純化,以得到呈黃色油的55.6 g (2 R)- N,N-二苄基-2-甲基戊-4-烯醯胺(M3-3)(86%)。LCMS: m/z (ESI), [M+H] +=294.15。 1H NMR (DMSO-d 6, 400 MHz) δ 1.01 (3H, d), 1.94-2.39 (2H, m), 2.76-2.90 (1H, m), 4.35-4.67 (4H, m), 4.89-5.11 (2H, m), 5.58-5.81 (1H, m), 7.08-7.43 (10H, m)。 步驟4. (2 R)- N,N-二苄基-5-羥基-2-甲基戊醯胺(M3-4)的製備。在0℃下向(2 R)- N,N-二苄基-2-甲基戊-4-烯醯胺(50 g,170.41 mmol,1當量)和9-硼雜雙環[3.3.1]壬烷(852.05 mL,426.02 mmol,2.5當量)於四氫呋喃(60 mL)中的攪拌混合物中逐滴添加NaOH(10 M)(220 mL)和H 2O 2(30%)(151 mL)。將所得混合物在室溫下攪拌2 h。將反應用Na 2S 2O 4(水溶液)溶液(500 mL)淬滅。將混合物用CH 2Cl 2(3×500 mL)萃取,並且將合併的有機層經無水Na 2SO 4乾燥。過濾後,將濾液在減壓下濃縮。將殘餘物在矽膠柱上使用石油醚/乙酸乙酯(1/1)洗脫進行純化,以得到呈黃色油的42.5 g (2 R)- N,N-二苄基-5-羥基-2-甲基戊醯胺(M3-4)(68%)。LCMS: m/z (ESI), [M+H] +=312.10。 1H NMR (DMSO-d 6, 400 MHz) δ 1.01 (3H, d), 1.24-1.43 (3H, m), 1.56-1.70 (1H, m), 2.60-2.80 (1H, m), 3.26-3.33 (2H, m), 4.30-4.42 (1H, m), 4.44-4.64 (4H, m), 7.15-7.23 (4H,m), 7.23-7.43 (6H, m)。 步驟5. (4 R)-5-(二苄基氨基)-4-甲基戊-1-醇(M3-5)的製備。在0℃下向LiAlH 4(6.73 g,177.41 mmol,1.3當量)於四氫呋喃(600 mL)中的攪拌混合物中添加(2 R)- N,N-二苄基-5-羥基-2-甲基戊醯胺(42.5 g,136.46 mmol,1當量)。將所得混合物在室溫下攪拌2 h。將反應在0℃下用水(7 mL)和15 mL NaOH(w/w,30%)淬滅,並且經無水Na 2SO 4乾燥。過濾後,將濾液在減壓下濃縮。將殘餘物在矽膠柱上使用石油醚/乙酸乙酯(10/1)洗脫進行純化,以得到呈黃色油的27.9 g (4 R)-5-(二苄基氨基)-4-甲基戊-1-醇(M3-5)(68%)。LCMS: m/z (ESI), [M+H] +=298.15。 1H NMR (DMSO-d 6, 400 MHz) δ 0.81 (3H, d), 0.88-1.00 (1H, m), 1.13-1.46 (3H, m), 1.64-1.78 (1H, m), 2.03-2.27 (2H, m), 3.27-3.39 (2H, m), 3.44 (2H, d), 3.49-3.59 (2H, d), 4.32 (1H, t), 7.18-7.26 (2H, m), 7.30-7.35 (8H, m)。 步驟6. (4 R)-5-氨基-4-甲基戊-1-醇(M3-6)的製備。將(4 R)-5-(二苄基氨基)-4-甲基戊-1-醇(18 g,60.51 mmol,1當量)和Pd/C(1.29 g,12.10 mmol,0.2當量)於甲醇(200 mL)中的混合物在氫氣氣氛下在室溫下攪拌6 h。將所得混合物過濾,並且將濾餅用甲醇(3×70 mL)洗滌。將所得溶液在減壓下濃縮,以得到呈黃色油的6.5 g (4 R)-5-氨基-4-甲基戊-1-醇(M3-6)(91%)。 1H NMR (DMSO-d 6, 400 MHz) δ 0.83 (3H, d), 0.94-1.09 (2H, m), 1.25-1.54 (3H, m),2.26-2.49 (2H, m), 3.17 (1H, s), 3.37 (2H, t)。 步驟7. (4 R)-5-[(5-溴-2-硝基苯基)氨基]-4-甲基戊-1-醇(M3-7)的製備。將4-溴-2-氟-1-硝基苯(10.00 g,45.46 mmol,1當量)、K 2CO 3(12.56 g,90.91 mmol,2當量)和(4 R)-5-氨基-4-甲基戊-1-醇(6.39 g,54.55 mmol,1.2當量)於乙腈(60 mL)中的混合物在氮氣氣氛下在60℃下攪拌2 h。將混合物冷卻到室溫,用水(500 mL)處理,並且用二氯甲烷(3×500 mL)萃取。將合併的有機層用鹽水(3×500 mL)洗滌,並且經無水Na 2SO 4乾燥。過濾後,將濾液在減壓下濃縮,並且通過prep-TLC使用石油醚/乙酸乙酯(1/3)洗脫進行純化,以得到呈黃色固體的11.7 g (4 R)-5-[(5-溴-2-硝基苯基)氨基]-4-甲基戊-1-醇(M3-7)(81%)。LCMS: m/z (ESI), [M+H] +=319.00。 1H NMR (400 MHz, DMSO-d 6 ) δ 0.95 (3H, d), 1.14-1.27 (1H, m), 1.37-1.59 (3H, m), 1.77-1.88 (1H, m), 3.18-3.22 (1H, m), 3.24-3.34 (1H, m), 3.35-3.44 (2H, m), 4.39 (1H, t), 6.83-6.87 (1H, m), 7.26 (1H, d), 7.99 (1H, d), 8.20 (1H, t)。 步驟8. 5-(5-{[(4 R)-5-[(5-溴-2-硝基苯基)氨基]-4-甲基戊基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M3-8)的製備。在氮氣氣氛下在0℃下向5-(5-羥基-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M2,2.67 g,10.09 mmol,1當量)和三苯基膦(6.62 g,25.22 mmol,2.5當量)於四氫呋喃(60 mL)中的攪拌混合物中添加偶氮二甲酸二異丙酯(4.08 g,20.18 mmol,2當量)和(4 R)-5-[(5-溴-2-硝基苯基)氨基]-4-甲基戊-1-醇(3.21 g,10.09 mmol,1當量)。在室溫下攪拌5 h後,將混合物用水(500 mL)處理,並且用二氯甲烷(3×500 mL)萃取。將合併的有機層用鹽水(3×500 mL)洗滌,並且經無水Na 2SO 4乾燥。過濾後,將濾液在減壓下濃縮。將殘餘物通過prep-TLC使用石油醚/乙酸乙酯(1/4)洗脫進行純化,以得到呈黃色固體的2.72 g 5-(5-{[(4 R)-5-[(5-溴-2-硝基苯基)氨基]-4-甲基戊基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M3-8)(48%)。LCMS: m/z (ESI), [M+H] +=564.10。 1H NMR (400 MHz, DMSO-d 6 ) δ 0.97 (3H, d), 1.45-1.29 (1H, m), 1.60-1.64 (1H, m), 1.78-1.82 (1H, m), 1.86 (2H, s), 3.38-3.15 (2H, m), 3.58 (3H, s), 3.68 (3H, s), 3.78 (3H, s), 3.96 (2H, t), 6.83-6.88 (1H, m), 7.26 (1H, d), 8.02-7.95 (2H, m), 8.14 (1H, d), 8.21 (1H, t), 8.43 (1H, d)。 步驟9. 5-(5-{[(4 R)-5-[(2-氨基-5-溴苯基)氨基]-4-甲基戊基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M3-9)的製備。在氮氣氣氛下在0℃下向5-(5-{[(4 R)-5-[(5-溴-2-硝基苯基)氨基]-4-甲基戊基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(4.00 g,7.11 mmol,1當量)和雷尼鎳(200 mg,2.33 mmol,0.33當量)於甲醇(40 mL)中的攪拌混合物中添加NH 2NH 2 .H 2O(712 mg,14.22 mmol,2當量)。在室溫下攪拌3 h後,將混合物過濾,並且將濾餅用甲醇(3×200 mL)洗滌。將有機溶液在減壓下濃縮,並且將殘餘物通過prep-TLC使用二氯甲烷/甲醇(30/1)洗脫進行純化,以得到呈黃色固體的3.65 g 5-(5-{[(4 R)-5-[(2-氨基-5-溴苯基)氨基]-4-甲基戊基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M3-9)(96%)。LCMS: m/z (ESI), [M+H] +=533.95。 步驟10. 5-(5-{[(4 R)-4-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]戊基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M3-10)的製備。在室溫下向5-(5-{[(4 R)-5-[(2-氨基-5-溴苯基)氨基]-4-甲基戊基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(3.00 g,5.63 mmol,1當量)於二氯甲烷(40 mL)中的攪拌混合物中添加溴化氰(895 mg,8.45 mmol,1.5當量)。在室溫下攪拌2 h後,將混合物在減壓下濃縮。將殘餘物通過prep-TLC使用二氯甲烷/甲醇(20/1)洗脫進行純化,以得到呈紅棕色固體的2.6 g 5-(5-{[(4 R)-4-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]戊基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M3-10)(83%)。LCMS: m/z (ESI), [M+H] +=559.05。 步驟11. 5-(5-{[(4 R)-4-{[2-氨基-6-(4-甲基哌嗪-1-基)-1,3-苯並二氮唑-1-基]甲基}戊基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(M3-11)的製備。在室溫下向5-(5-{[(4 R)-4-{[2-氨基-6-(4-甲基哌嗪-1-基)-1,3-苯並二氮唑-1-基]甲基}戊基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(3.71 g,6.42 mmol,1當量)於四氫呋喃(100 mL)中的攪拌混合物中添加含LiOH(307 mg,12.83 mmol,2當量)的H 2O(20 mL)。在室溫下攪拌1 h後,將反應混合物在減壓下濃縮。將殘餘物通過反向快速色譜法(10%至40%乙腈/水)進行純化,以得到呈白色固體的3.5 g 5-(5-{[(4 R)-4-{[2-氨基-6-(4-甲基哌嗪-1-基)-1,3-苯並二氮唑-1-基]甲基}戊基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(M3-11)(97%)。LCMS: m/z (ESI), [M+H] +=545.15。 步驟12. (11 R)-16-溴-5,11,26-三甲基-7-氧雜-4,5,13, 20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(M3)的製備。在室溫下向5-(5-{[(4 R)-4-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]戊基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(1.2 g,2.20 mmol,1當量)於二噁烷(18 mL)中的攪拌混合物中添加 N, N-二異丙基乙胺(0.85 g,6.62 mmol,3當量)和N,N,N,N-四甲基-O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(1.26 g,3.31 mmol,1.5當量)。在氮氣氣氛下在60℃下攪拌3 h後,將反應混合物冷卻到室溫,用水(300 mL)處理,並且用CH 2Cl 2(2×200 mL)萃取。將合併的有機層用鹽水(3×300 mL)洗滌,並且經無水Na 2SO 4乾燥。過濾後,將濾液在減壓下濃縮。將殘餘物通過prep-TLC使用CH 2Cl 2/甲醇(40/1)洗脫進行純化,以得到呈白色固體的1.14 g (11 R)-16-溴-5,11,26-三甲基-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(M3)(98%)。LCMS: m/z (ESI), [M+H] +=525.15。 1H NMR (DMSO-d 6, 400 MHz) δ 0.81 (3H, d), 1.43 (1H, q), 1.91 (2H, d), 2.06-2.26 (1H, m), 2.78 (1H, br s), 3.62 (3H, s), 3.72 (3H, s), 3.89-4.00 (2H, m), 4.07-4.15 (1H, m), 4.31-4.38 (1H, m), 7.36 (1H, dd), 7.46 (1H, d), 7.88 (1H, d), 8.30 (1H, d), 8.35 (1H, s), 8.79 (1H, d), 12.67 (1H, s)。 中間體M4A和M4B 5-溴-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮,異構體1(M4A)和異構體2(M4B) 步驟1. 苄基雙(乙氧基甲基)胺(M4-1)的製備。將苄胺(500 g,4666 mmol,1當量)、POM(840.62 g,9332 mmol,2當量)和K 2CO 3(644.88 g,4666. mmol,1當量)於乙醇(1200 mL)中的混合物在氮氣氣氛下在室溫下攪拌16 h。將所得混合物過濾。將濾餅用二氯甲烷(3×200 mL)洗滌,並且將有機溶液在減壓下濃縮,以得到呈黃色油的1200 g苄基雙(乙氧基甲基)胺(M4-1)(80%)。此粗材料在不進行進一步純化的情況下用於步驟3。 1H NMR (DMSO- d 6, 400 MHz) δ 1.01-1.19 (6H, m), 3.39-3.57 (3H, m), 3.90 (2H, t), 4.55-4.82 (4H, m), 7.27-7.39 (5H, m)。 步驟2. 2-氧代環戊烷-1-甲酸異丙酯(M4-2)的製備。將2-氧代環戊烷-1-甲酸乙酯(400 g,2561 mmol,1當量)和DMAP(31.29 g,256.11 mmol,0.1當量)於 i-PrOH(500 mL)中的混合物在氮氣氣氛下在80℃下攪拌過夜。將混合物冷卻到室溫,並且在減壓下濃縮。將殘餘物在用石油醚/乙酸乙酯(10/1)洗脫的矽膠柱上進行純化,以得到呈黃色油的400 g 2-氧代環戊烷-1-甲酸異丙酯(M4-2)(92%)。LCMS: m/z (ESI), [M - H] +=168.85。 1H NMR (DMSO- d 6, 400 MHz) δ 1.26 (6H, t), 1.80-1.92 (1H, m), 2.07-2.17 (1H, m), 2.25-2.35 (4H, m), 3.11 (1H, t), 5.00-5.10 (1H, m)。 步驟3. 3-苄基-8-氧代-3-氮雜雙環[3.2.1]辛烷-1-甲酸異丙酯(M4-3)的製備。將苄基雙(甲氧基甲基)胺(M4-1,601.1 g,3078.5 mmol,2.62當量)於DMF(1.5 L)中的溶液用甲基三氯矽烷(400.4 g,2679 mmol,2.28當量)在氮氣氣氛下在0℃下處理10分鐘,隨後在0℃下緩慢添加2-氧代環戊烷-1-甲酸異丙酯(M4-2,200 g,1175 mmol,1當量)。將所得混合物在氮氣氣氛下在室溫下攪拌過夜。將反應用水(3 L)淬滅,用飽和NaHCO 3(水溶液)溶液(3 L)鹼化到pH 7,並且將混合物用乙酸乙酯(3×2 L)萃取。將合併的有機層用鹽水(3×2 L)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上使用石油醚:乙酸乙酯(40/1)洗脫進行純化,並且在真空下濃縮。將殘餘物通過Prep-HPLC通過反向快速色譜法使用C18矽膠柱進行進一步純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃色油的110 g 3-苄基-8-氧代-3-氮雜雙環[3.2.1]辛烷-1-甲酸異丙酯(M4-3)(31%)。LCMS: m/z (ESI), [M+H] +=470.15。 1H NMR (DMSO- d 6, 400 MHz) δ 1.04-1.26 (6H, m), 1.84-1.96 (2H, m), 2.16-2.37 (3H, m), 2.45 (1H, d), 2.62 (1H, d), 2.94 (1H, d), 3.04 (1H, d), 3.65 (2H, s), 4.75-5.02 (1H, m), 7.13-7.48 (5H, m) 步驟4. 3-苄基-8-[(4-甲基苯磺醯胺)亞氨基]-3-氮雜雙環[3.2.1]辛烷-1-甲酸異丙酯(M4-4)的製備。3-苄基-8-氧代-3-氮雜雙環[3.2.1]辛烷-1-甲酸異丙酯(100 g,331.8 mmol,1當量)於甲醇(1 L)中的溶液在氮氣氣氛下,隨後在0℃下分批添加TsNHNH 2(123.58 g,663.5 mmol,2.00當量)。將所得混合物在氮氣氣氛下在室溫下攪拌過夜。將所得混合物在減壓下濃縮。將殘餘物在矽膠柱上進行純化並且使用石油醚/乙酸乙酯(5/1)洗脫,以得到呈白色固體的80 g 3-苄基-8-[(4-甲基苯磺醯胺)亞氨基]-3-氮雜雙環[3.2.1]辛烷-1-甲酸異丙酯(M4-4)(51%)。 1H NMR (DMSO- d 6, 400 MHz) δ 0.99-1.15 (6H, m), 1.57-1.85 (2H, m), 1.87-2.03 (1H, ddd), 2.02-2.13 (2H, m), 2.25 (1H, d), 2.39 (3H, s), 2.62-2.73 (1H, m),2.84(1H, d) 3.14-3.19 (1H, m), 3.06-3.10 (1H, m), 3.50 (2H, s), 4.75-4.91 (1H, m), 7.26-7.43 (7H, m), 7.67 (2H, d), 10.38 (1H, s)。 步驟5. 3-苄基-3-氮雜雙環[3.2.1]辛烷-1-甲酸酯(M4-5)的製備。在氮氣氣氛下在0℃下向3-苄基-8-[(4-甲基苯磺醯胺)亞氨基]-3-氮雜雙環[3.2.1]辛烷-1-甲酸異丙酯(100 g,212.95 mmol,1當量)於甲醇(2 L)中的攪拌溶液中添加NaBH 4(120.8 g,3194.20 mmol,15當量)。將混合物在80℃下攪拌過夜,然後冷卻到室溫。將反應用水(1 L)淬滅,並且將混合物用乙酸乙酯(3×1 L)萃取。將合併的有機層用鹽水(3×1 L)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法進行純化並且使用石油醚/乙酸乙酯(5/1)洗脫,以得到呈無色油的33.0 g 3-苄基-3-氮雜雙環[3.2.1]辛烷-1-甲酸異丙酯(M4-5)(54%)。LCMS: m/z (ESI), [M+H] +=288.05。 1H NMR (DMSO-d 6, 400 MHz) δ 1.14 (6H, d), 1.41 (1H, d), 1.62-1.69 (2H, m), 1.70-1.90 (3H, m), 1.9-2.04 (1H, m), 2.12 (1H, d), 2.18 (1H, d), 2.58 (1H, d), 2.82 (1H, d), 3.50 (2H, d), 4.81-4.90 (1H, m), 7.06-7.51 (5H, m)。 步驟6. 3-苄基-3-氮雜雙環[3.2.1]辛烷-1-羧酸(M4-6)的製備。將3-苄基-3-氮雜雙環[3.2.1]辛烷-1-甲酸異丙酯(33.0 g,114.82 mmol,1當量)和LiOH(3.0 g,126.30 mmol,1.1當量)於乙醇(400 mL)中的混合物在氮氣氣氛下在80℃下攪拌2天。將混合物冷卻到室溫,並且在減壓下濃縮,以得到呈黃色油的30 g 3-苄基-3-氮雜雙環[3.2.1]辛烷-1-羧酸(M4-6)(96%)。LCMS: m/z (ESI), [M+H] +=246.05。 步驟7. N-{3-苄基-3-氮雜雙環[3.2.1]辛-1-基}氨基甲酸叔丁酯(M4-7)的製備。將3-苄基-3-氮雜雙環[3.2.1]辛烷-1-羧酸(30 g,110.06 mmol,1當量,90%)、DPPA(36.35 g,132.07 mmol,1.2當量)和三乙胺(33.41 g,330.18 mmol,3.0當量)於四氫呋喃(300 mL)中的混合物在氮氣氣氛下在80℃下攪拌2 h。將混合物冷卻到室溫,並且在減壓下濃縮。在0℃下添加甲苯(500 mL)和 t-BuOK(18.53 g,165.09 mmol,1.5當量)。將混合物在氮氣氣氛下在100℃下攪拌2 h,然後冷卻到室溫。將反應用水(1 L)淬滅,並且將混合物用乙酸乙酯(3×1 L)萃取。將合併的有機層用鹽水(3×1 L)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法使用石油醚/乙酸乙酯(5/1)洗脫來進行純化,以得到呈白色固體的15 g N-{3-苄基-3-氮雜雙環[3.2.1]辛-1-基}氨基甲酸叔丁酯(M4-7)(43%)。LCMS: m/z (ESI), [M+H] +=317.15。 1H NMR (DMSO-d 6, 400 MHz) δ 1.23 (9H, s), 1.38-1.60 (3H, m), 1.59-1.76 (2H, m), 1.75-2.25 (5H, m), 2.87 (1H, d), 3.38-3.56 (2H, m), 6.88 (1H, s), 7.17-7.37 (5H, m) 步驟8. N-{3-氮雜雙環[3.2.1]辛-1-基}氨基甲酸叔丁酯(M4-8)的製備。將 N-{3-苄基-3-氮雜雙環[3.2.1]辛-1-基}氨基甲酸叔丁酯(15 g,47.40 mmol,1當量)和Pd/C(7.57 g,71.13 mmol,1.50當量)於甲醇(300 mL)中的混合物在氫氣氣氛下在室溫下攪拌2 h。將所得混合物過濾,並且將濾餅用甲醇(3×200 mL)洗滌。將有機溶液在減壓下濃縮,以得到呈白色固體的10 g N-{3-氮雜雙環[3.2.1]辛-1-基}氨基甲酸叔丁酯(M4-8)(93%)。此材料在不經進一步純化的情況下用於下一步驟中。LCMS: m/z (ESI), [M+H] +=227.10。 1H NMR (DMSO-d 6, 400 MHz) δ 1.36 (9H, d), 1.45-1.60 (2H, m), 1.60-1.75 (1H, m), 1.73-1.91 (1H, m), 1.93-2.00 (2H, m), 2.45 (2H, d), 2.76-2.89 (1H, m), 3.14-3.20 (1H, m), 6.73 (1H, d)。 步驟9. N-(3-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-3-氮雜雙環[3.2.1]辛-1-基)氨基甲酸叔丁酯(M4-9)的製備。將新鮮製備的 N-{3-氮雜雙環[3.2.1]辛-1-基}氨基甲酸叔丁酯(10 g,約44 mmol,約1當量)、(2-溴乙氧基)(叔丁基)二甲基矽烷(21.14 g,88.37 mmol,2當量)、NaI(13.25 g,88.37 mmol,2當量)和K 2CO 3(12.21 g,88.37 mmol,2當量)於DMF(160 mL)中的混合物在氮氣氣氛下在60℃下攪拌2 h。將混合物冷卻到室溫。將反應用水(300 mL)淬滅,並且將混合物用乙酸乙酯(3×200 mL)萃取。將合併的有機層用鹽水(3×300 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-HPLC使用含有0.1% NH 4HCO 3的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈無色油的12 g N-(3-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-3-氮雜雙環[3.2.1]辛-1-基)氨基甲酸叔丁酯(M4-9)(70%)。LCMS: m/z (ESI), [M+H] += 385.25。 1H NMR (DMSO-d 6, 400 MHz) δ 0.00 (6H, s), 0.82 (9H, s), 0.85 (1H, s), 1.32 (9H, s), 1.37-1.48 (2H, m), 1.53-1.64 (2H, m), 1.91-2.05 (4H, m), 2.36-2.43 (2H, m), 2.54-2.57 (1H, m), 2.86-2.92 (1H, m), 3.61 (2H, t), 6.80 (1H, s)。 步驟10. 2-(1-氨基-3-氮雜雙環[3.2.1]辛-3-基)乙-1-醇(M4-10)的製備。將 N-(3-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-3-氮雜雙環[3.2.1]辛-1-基)氨基甲酸叔丁酯(12 g,31.19 mmol,1當量)於三氟乙酸(28 mL)和二氯甲烷(84 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將所得混合物在減壓下濃縮。將呈橙色油的粗產物2-(1-氨基-3-氮雜雙環[3.2.1]辛-3-基)乙-1-醇(M4-10)在不進行進一步純化的情況下直接用於下一步驟。LCMS: m/z (ESI), [M+H] += 285.15。 步驟11. 2-[1-[(5-溴-2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基]乙醇(M4-11)的製備。將2-(1-氨基-3-氮雜雙環[3.2.1]辛-3-基)乙-1-醇(粗製物,M4-10)、K 2CO 3(21.56 g,156.25 mmol,5當量)和4-溴-2-氟-1-硝基苯(20.53 g,93.73 mmol,3當量)於DMSO(160 mL)中的混合物在氮氣氣氛下在120℃下攪拌2 h。將混合物冷卻到室溫。將反應用水(300 mL)淬滅,並且將混合物用乙酸乙酯(3×200 mL)萃取。將合併的有機層用鹽水(3×300 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將粗產物通過prep-HPLC使用含有0.1% NH 4HCO 3的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈橙色半固體的12 g 2-[1-[(5-溴-2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基]乙醇(M4-11)(76%)。LCMS: m/z (ESI), [M+H] += 371.95。 1H NMR (DMSO-d 6, 400 MHz) δ 1.68 (3H, d), 1.82-1.85 (1H, m), 1.94 (1H, s), 2.12 (1H, d), 2.17 (1H, d), 2.21 (1H, s), 2.36 (1H, s), 2.49 (2H, d), 2.65-2.73 (1H, m), 3.10-3.20 (1H, m), 3.47-3.57 (2H, m), 4.36 (1H, t), 6.86-3.89 (1H, m), 7.21 (1H, d), 8.01 (1H, d), 8.31 (1H, s)。 步驟12. 5-[5-(2-{1-[(5-溴-2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(M4-12)的製備。在氮氣氣氛下在0℃下向2-{1-[(5-溴-2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙醇(8 g,21.61 mmol,1當量)和三苯基膦(17.00 g,64.82 mmol,3當量)於四氫呋喃(500 mL)中的攪拌混合物中緩慢添加偶氮二甲酸二異丙酯(12.23 g,60.50 mmol,2.8當量)和5-(5-羥基-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(5.69 g,21.61 mmol,1當量)。將所得混合物在氮氣氣氛下在0℃下攪拌2 h。將反應用水(300 mL)淬滅,並且將混合物用乙酸乙酯(3×200 mL)萃取。將合併的有機層用鹽水(3×300 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上使用石油醚/乙酸乙酯(1/3)洗脫進行純化,以得到呈橙色油的9.5 g 5-[5-(2-{1-[(5-溴-2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(M4-12)(71%)。LCMS: m/z (ESI), [M+H] +=617.10。 步驟13. 5-[5-(2-{1-[(2-氨基-5-溴苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(M4-13)的製備。向5-[5-(2-{1-[(5-溴-2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(10.5 g,17.06 mmol,1當量)和雷尼鎳(2 g)於甲醇(150 mL)中的混合物中添加水合肼(2.19 g,68.24 mmol,1.5當量),並且在氮氣氣氛下在0℃下攪拌2 h。將所得混合物過濾,並且將濾餅用二氯甲烷(3×200 mL)洗滌。將有機溶液在減壓下濃縮。將殘餘物在矽膠柱上使用二氯甲烷/甲醇(10/1)洗脫進行純化,以得到呈棕色油的9.5 g 5-[5-(2-{1-[(2-氨基-5-溴苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(M4-13)(91%)。LCMS: m/z (ESI), [M+H] +=585.20。 1H NMR (DMSO-d 6, 400 MHz) δ 1.45-1.60 (3H, m), 1.70-1.85 (2H, m), 1.95-2.17 (4H, m), 2.65-2.80 (3H, m), 3.09-3.15 (1H, m), 3.17 (2H, d), 3.57 (3H, s), 3.74 (3H, s), 3.79 (3H, s), 4.00-4.10 (3H, m), 4.52 (1H, s), 4.75 (2H, s), 6.44-6.50 (1H, m), 6.50-6.56 (1H, m), 6.64 (1H, d), 7.97 (1H, s), 8.10 (1H, d), 8.43 (1H, d)。 步驟14. 5-(5-{2-[1-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M4-14)的製備。將5-[5-(2-{1-[(2-氨基-5-溴苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(10 g,17.08 mmol,1當量)和溴化氰(2.17 g,20.49 mmol,1.20當量)於乙醇(200 mL)中的溶液在氮氣氣氛下在室溫下攪拌過夜。將所得混合物在減壓下濃縮,並且將殘餘物在矽膠柱上使用二氯甲烷/甲醇(20/1)洗脫進行純化,以得到呈棕色固體的9.5 g 5-(5-{2-[1-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M4-14)(91%)。LCMS: m/z (ESI), [M+H] +=612.05。 步驟15. 5-(5-{2-[1-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯,異構體1(M4-14A)和異構體2(M4-14B)的製備。通過手性色譜法使用(R, R)-WHELK-01-Kromasil柱(5 * 25 cm,5 μm)、CO 2作為流動相A以及甲醇/乙腈(1/1)作為流動相B來分離M4-14(9 g,14.74 mmol)的外消旋混合物,以得到呈棕色固體的3.6 g異構體1(M4-14A,41%)和3.9 g異構體2(M4-14B,43%)。SFC-HPLC,Rt(異構體1)=2.246分鐘,Rt(異構體2)=3.898分鐘。 步驟16. 5-(5-(2-(1-(2-氨基-6-溴-1H-苯並[d]咪唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基)乙氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-羧酸(異構體1,M4-15A)的製備。向5-(5-(2-(1-(2-氨基-6-溴-1H-苯並[d]咪唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基)乙氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-甲酸甲酯(異構體1,M4-14A,4 g,6.55 mmol,1當量)於四氫呋喃(8 mL)中的攪拌溶液中添加含LiOH(0.31 g,13.10 mmol,2當量)的H 2O(2 mL)。將所得混合物在室溫下攪拌2 h。將反應在減壓下濃縮,並且將殘餘物通過Prep-HPLC通過反向快速色譜法使用C18矽膠柱使用水和乙腈洗脫來進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的3.6 g 5-(5-(2-(1-(2-氨基-6-溴-1H-苯並[d]咪唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基)乙氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-羧酸(異構體1,M4-15A) (92%)。LCMS: m/z (ESI), [M+H] +=598.05。 步驟17. 5-溴-15,21-二甲基-23-氧雜-2,9,11,15,20,21, 26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33), 18(22),19-八烯-12,16-二酮(M4A)的製備。將5-(5-{2-[1-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(M4-15A,3.6 g,6.03 mmol,1當量)、 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(3.44 g,9.05 mmol,1.5當量)和 N,N-二異丙基乙胺(2.34 g,18.10 mmol,3當量)於1,4-二噁烷(80 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將反應用水(300 mL)淬滅,並且將混合物用乙酸乙酯(3×200 mL)萃取。將合併的有機層用鹽水(3×300 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上使用二氯甲烷/甲醇(20/1)洗脫進行純化,以得到呈棕色固體的3.3 g 5-溴-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮(異構體1,M4A)(94%)。LCMS: m/z (ESI), [M+H] += 580.00。SFC-HPLC, Rt=5.498, 1H NMR (DMSO-d 6, 400 MHz) δ 1.56-1.81 (3H, m), 2.01 (1H, d), 2.42 (2H, d), 2.54 (1H, d), 2.63 (1H, s), 2.69-2.75 (1H, m), 3.09-3.20 (2H, m), 3.19-3.30 (1H, m), 3.63 (3H, s), 3.72 (3H, s), 4.54 (1H, t), 4.63 (1H, d), 7.36 (1H, d), 7.51 (1H, d), 7.82 (1H, d), 8.16 (1H, s), 8.30 (1H, d), 8.41 (1H, s), 8.81 (1H, d), 12.84 (1H, s)。 步驟18. 5-(5-(2-(1-(2-氨基-6-溴-1H-苯並[d]咪唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基)乙氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-羧酸(異構體2,M4-15B)的製備。向5-(5-(2-(1-(2-氨基-6-溴-1H-苯並[d]咪唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基)乙氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-甲酸甲酯(異構體2,M4-14B,4.2 g,6.88 mmol,1當量)於四氫呋喃(80 mL)中的攪拌溶液中添加含LiOH(0.33 g,13.76 mmol,2當量)的H 2O(20 mL)。將所得混合物在室溫下攪拌2 h。將反應在減壓下濃縮,並且將殘餘物通過Prep-HPLC通過反向快速色譜法使用C18矽膠柱使用水和乙腈洗脫來進行進一步純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的3.8 g 5-(5-(2-(1-(2-氨基-6-溴-1H-苯並[d]咪唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基)乙氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-羧酸(異構體2,M4-15B)(92%)。LCMS: m/z (ESI), [M+H] +=598.05。 步驟19. 5-溴-15,21-二甲基-23-氧雜-2,9,11,15,20,21, 26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮(異構體2,M4B)的製備。將5-(5-{2-[1-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(M4-15B,3.8 g,6.37 mmol,1當量)、 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(3.63 g,9.56 mmol,1.5當量)和 N,N-二異丙基乙胺(2.47 g,19.11 mmol,3當量)於1,4-二噁烷(80 mL)中的溶液在氮氣氣氛下在室溫下攪拌2 h。將反應用水(300 mL)淬滅,並且將混合物用乙酸乙酯(3×200 mL)萃取。將合併的有機層用鹽水(3×300 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上使用二氯甲烷/甲醇(20/1)洗脫進行純化,以得到呈棕色固體的3.5 g 5-溴-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮(異構體2,M4B)(94%)。LCMS: m/z (ESI), [M+H] += 580.00。SFC-HPLC, Rt=3.186, 1H NMR (DMSO-d 6, 400 MHz) δ 1.55-1.76 (2H, m), 2.01 (1H, d), 2.39-2.47 (2H, m), 2.51-2.57 (1H, m), 2.61-2.65 (1H, m), 2.70-2.77 (2H, m), 3.09-3.18 (2H, m), 3.19-3.29 (1H, m), 3.63 (3H, s), 3.72 (3H, s), 4.54 (1H, t), 4.63 (1H, d), 7.36 (1H, dd), 7.51 (1H, d), 7.82 (1H, d), 8.16 (1H, s), 8.30 (1H, d), 8.41 (1H, s), 8.81 (1H, d), 12.84 (1H, s) 中間體M5 5-溴-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十四-3,5,7,9,13,17(34),18(22),19-八烯-12,16-二酮 步驟1. 4-亞甲基環己烷-1-甲酸乙酯(M5-1)的製備。在0℃下向甲基三苯基溴化膦(314.81 g,881.27 mmol,1.5當量)和叔丁醇鉀(131.85 g,1175.03 mmol,2當量)於四氫呋喃(500 mL)中的混合物中添加4-氧代環己烷-1-甲酸乙酯(100 g,587.51 mmol,1當量)。將混合物在氮氣氣氛下在室溫下攪拌2 h。將反應用水(300 mL)淬滅,並且將混合物用乙酸乙酯(3×200 mL)萃取。將合併的有機層用鹽水(3×300 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法使用石油醚/乙酸乙酯(5/1)洗脫進行純化,以得到呈黃色油的80 g 4-亞甲基環己烷-1-甲酸乙酯(M5-1)(85%)。 1H NMR (DMSO-d 6, 400 MHz) δ 1.18 (3H, t), 1.30-1.50 (2H, m), 1.80-1.95 (2H, m), 1.96-2.15 (2H, m), 2.15-2.35 (2H, m), 2.42-2.49 (1H, m), 4.04-4.08 (2H, m), 4.64 (2H, t)。 步驟2. 1-(4-甲基苯磺醯基)-1-氮雜螺[2.5]辛烷-6-甲酸乙酯(M5-2)的製備。在0℃下向4-亞甲基環己烷-1-甲酸乙酯(80 g,475.52 mmol,1當量)和氯胺-T(216.50 g,951.04 mmol,2當量)於乙腈(800 mL)中的混合物中添加 N,N,N-三甲基苯銨溴化二溴烷(17.88 g,47.55 mmol,0.1當量)。將混合物在氮氣氣氛下在室溫下攪拌過夜。將所得混合物在減壓下濃縮。將殘餘物通過矽膠色譜法使用石油醚/乙酸乙酯(5/1)洗脫進行純化,以得到呈白色固體的60 g 1-(4-甲基苯磺醯基)-1-氮雜螺[2.5]辛烷-6-甲酸乙酯(M5-2) (33%)。LCMS: m/z (ESI), [M+H] +=338.05。 1H NMR (DMSO-d 6, 400 MHz) δ 1.18 (3H, t), 1.54-1.58 (1H, m), 1.58-1.72 (1H, m), 1.72-1.93 (4H, m), 2.40 (3H, s), 2.50-2.53(2H, m), 4.05-4.09 (2H, m), 7.43 (2H, d), 7.73-7.79 (2H, m)。 步驟3. 4-[(苄基氨基)甲基]-4-(4-甲基苯磺醯胺)環己烷-1-甲酸乙酯(M5-3)的製備。將1-(4-甲基苯磺醯基)-1-氮雜螺[2.5]辛烷-6-甲酸乙酯(60 g,177.81 mmol,1當量)和苄胺(28.58 g,266.72 mmol,1.5當量)於四氫呋喃(600 mL)中的溶液在氮氣氣氛下在80℃下攪拌2天。將混合物冷卻到室溫,並且在減壓下濃縮。將殘餘物通過矽膠色譜法使用石油醚/乙酸乙酯(1/1)洗脫進行純化,以得到呈白色油的60 g 4-[(苄基氨基)甲基]-4-(4-甲基苯磺醯胺)環己烷-1-甲酸乙酯(M5-3)(75%)。LCMS: m/z (ESI), [M+H] +=445.20。 1H NMR (DMSO-d 6, 400 MHz) δ 1.15 (3H, t), 1.31-1.44 (2H, m), 1.48-1.76 (4H, m), 2.00 (1H, m), 2.21-2.30 (1H, m), 2.30 (2H, s), 2.33-2.37 (2H, m), 3.43 (2H, s), 4.02 (2H, m), 7.15-7.25 (3H, m), 7.25-7.36 (4H, m), 7.67-7.75 (2H, m)。 步驟4. 4-((苄基氨基)甲基)-4-((4-甲基苯基)磺醯氨基)環己烷-1-甲酸鋰(M5-4)的製備。將4-[(苄基氨基)甲基]-4-(4-甲基苯磺醯胺)環己烷-1-甲酸乙酯(60 g,134.95 mmol,1當量)和LiOH(6.46 g,269.91 mmol,2當量)於乙醇(500 mL)中的混合物在氮氣氣氛下在80℃下攪拌16 h。將所得混合物在減壓下濃縮,以得到呈白色固體的57 g 4-((苄基氨基)甲基)-4-((4-甲基苯基)磺醯氨基)環己烷-1-甲酸鋰(M5-4)。此材料在不經進一步純化的情況下用於下一步驟中。LCMS: m/z (ESI), [M+H] +=417.10。 步驟5. N-{3-苄基-4-氧代-3-氮雜雙環[3.2.2]壬-1-基}-4-甲苯磺醯胺(M5-5)的製備。將 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(77.03 g,202.60 mmol,1.5當量)、4-((苄基氨基)甲基)-4-((4-甲基苯基)磺醯氨基)環己烷-1-甲酸鋰(57 g,約135 mmol,約1當量)和 N,N-二異丙基乙胺(52.37 g,405.21 mmol,3當量)於二氯甲烷(150 mL)中的混合物在氮氣氣氛下在室溫下攪拌過夜。將所得混合物在減壓下濃縮,並且將殘餘物通過矽膠柱色譜法使用石油醚/乙酸乙酯(1/1)洗脫來進行純化,以得到呈白色固體的50 g N-{3-苄基-4-氧代-3-氮雜雙環[3.2.2]壬-1-基}-4-甲苯磺醯胺(M5-5)(93%)。LCMS: m/z (ESI), [M+H] += 399.15。 1H NMR (DMSO-d 6, 400 MHz) δ 1.64-1.74 (8H, m), 2.38 (3H, s), 3.18 (2H, s), 4.37 (2H, s), 7.01-7.19 (2H, m), 7.21-7.41 (5H, m), 7.48-7.56 (2H, m), 7.73 (1H, s) 步驟6. N-{3-苄基-3-氮雜雙環[3.2.2]壬-1-基}-4-甲苯磺醯胺甲基(M5-6)的製備。在0℃下向氫化鋰鋁(7.62 g,200.74 mmol,2當量)於四氫呋喃(400 mL)中的混合物中添加 N-{3-苄基-4-氧代-3-氮雜雙環[3.2.2]壬-1-基}-4-甲苯磺醯胺(40 g,100.37 mmol,1當量),並且將混合物在氮氣氣氛下攪拌2 h。將反應在0℃下用水(7 mL)和15 mL NaOH(w/w,30%)淬滅。將混合物經無水Na 2SO 4乾燥,並且在減壓下濃縮。將殘餘物在矽膠柱上使用石油醚/乙酸乙酯(1/1)洗脫來進行純化,以得到呈白色固體的20 g N-{3-苄基-3-氮雜雙環[3.2.2]壬-1-基}-4-甲苯磺醯胺(M5-6)(46%)。LCMS: m/z (ESI), [M+H] +=385.15。 1H NMR (DMSO-d 6, 400 MHz) δ 1.39-1.63 (6H, m), 1.71-1.77 (1H, m), 1.90-2.02 (2H, m), 2.36 (3H, s), 2.40 (2H, d), 2.64 (2H, s), 3.44 (2H, s), 7.18-7.31 (3H, m), 7.31-7.40 (4H, m), 7.39 (1H, s), 7.55-7.62 (2H, m)。 步驟7. N-{3-氮雜雙環[3.2.2]壬-1-基}-4-甲苯磺醯胺(M5-7)的製備。將Pd/C(10%,500 mg)和 N-{3-苄基-3-氮雜雙環[3.2.2]壬-1-基}-4甲苯磺醯胺(20 g,52.01 mmol,1當量)於甲醇(200 mL)中的混合物在氫氣壓力下在室溫下攪拌4 h。將所得混合物過濾。將濾餅用甲醇(3×70 mL)洗滌,並且將溶液在減壓下濃縮。將殘餘物在矽膠柱上使用二氯甲烷/甲醇(10/1)洗脫來進行純化,以得到呈白色固體的15 g N-{3-氮雜雙環[3.2.2]壬-1-基}-4-甲苯磺醯胺(M5-7) (93%)。LCMS: m/z (ESI), [M+H] +=295.00。 1H NMR (DMSO-d 6, 400 MHz) δ 1.35-1.60 (6H, m), 1.63-1.69 (1H, m), 1.79-1.83 (2H, m), 2.37 (3H, s), 2.59 (2H, d), 2.80 (2H, s), 3.17 (1H, d), 7.29-7.41 (3H, m), 7.51-7.79 (2H, m)。 步驟8. N-(3-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-3-氮雜雙環[3.2.2]壬-1-基)-4-甲苯磺醯胺(M5-8)的製備。將 N-{3-氮雜雙環[3.2.2]壬-1-基}-4-甲苯磺醯胺(15 g,50.94 mmol,1當量)、NaI(7.64 g,50.94 mmol,1當量)、K 2CO 3(21.12 g,152.84 mmol,3當量)和(2-溴乙氧基)(叔丁基)二甲基矽烷(14.63 g,61.13 mmol,1.2當量)於二甲基甲醯胺(150 mL)中的混合物在氮氣氣氛下在60℃下攪拌16 h。將混合物冷卻到室溫。將反應用水(800 mL)淬滅,並且將混合物用乙酸乙酯(3×600 mL)萃取。將合併的有機層用鹽水(3×500 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上使用二氯甲烷/甲醇(10/1)洗脫來進行純化,以得到呈黃色固體的20 g N-(3-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-3-氮雜雙環[3.2.2]壬-1-基)-4-甲苯磺醯胺(M5-8)(78%)。LCMS: m/z (ESI), [M+H] += 453.30。 1H NMR (DMSO-d 6, 400 MHz) δ 0.00 (6H, s), 0.83 (9H, s), 1.32-1.42 (2H, m), 1.44-1.54 (4H, m), 1.65-1.71 (1H, m), 1.80-1.90 (2H, m), 2.34 (3H, s), 2.38 (2H, t), 2.44 (2H, d), 2.65 (2H, s), 3.54 (2H, t), 7.32 (2H, d), 7.37 (1H, s), 7.59-7.76 (2H, m)。 步驟9. 3-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-3-氮雜雙環[3.2.2]壬-1-胺(M5-9)的製備。將 N-(3-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-3-氮雜雙環[3.2.2]壬-1-基)-4-甲苯磺醯胺(5 g,11.04 mmol,1當量)和Mg(5.37 g,220.88 mmol,20當量)於甲醇(200 mL)中的混合物在氮氣氣氛下在70℃下攪拌過夜。將混合物冷卻到室溫。將反應通過添加飽和NH 4Cl(水溶液)溶液(300 mL)淬滅,並且將混合物用乙酸乙酯(3×300 mL)萃取。將合併的有機層用鹽水(3×400 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上使用二氯甲烷/甲醇(5/1)洗脫來進行純化,以得到呈黃色油的2.3 g 3-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-3-氮雜雙環[3.2.2]壬-1-胺(M5-9)(60%)。 1H NMR (DMSO-d 6, 400 MHz) δ 0.00 (6H, s), 0.82 (9H, s), 1.54-1.64 (4H, m), 1.79-1.83 (1H, m), 1.85-1.91 (2H, m), 2.42-2.52 (3H, m), 2.54 (2H, d), 2.60 (2H, s), 3.63 (2H, t)。 步驟10. N-(5-溴-2-硝基苯基)-3-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-3-氮雜雙環[3.2.2]壬-1-胺(M5-10)的製備。將3-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-3-氮雜雙環[3.2.2]壬-1-胺(1 g,3.35 mmol,1當量)、4-溴-2-氟-1-硝基苯(1.84 g,8.37 mmol,2.5當量)和K 2CO 3(1.39 g,10.05 mmol,3當量)於二甲基亞碸(10 mL)中的混合物在氮氣氣氛下在120℃下攪拌4 h。將混合物冷卻到室溫。將反應用水(50 mL)淬滅,並且將混合物用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法使用石油醚/乙酸乙酯(5/1)洗脫來進行純化,以得到呈黃色油的1.5 g N-(5-溴-2-硝基苯基)-3-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-3-氮雜雙環[3.2.2]壬-1-胺(M5-10)(85%)。LCMS: m/z (ESI), [M+H]+=498.15。 步驟11. 2-{1-[(5-溴-2-硝基苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙醇(M5-11)的製備。將 N-(5-溴-2-硝基苯基)-3-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-3-氮雜雙環[3.2.2]壬-1-胺(1.5 g,3.00 mmol,1當量)和四丁基氟化銨(1.57 g,6.01 mmol,2當量)於四氫呋喃(15 mL)中的溶液在氮氣氣氛下在室溫下攪拌2 h。將反應用飽和NaHCO 3(水溶液)溶液(100 mL)淬滅,並且將混合物用二氯甲烷(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法使用二氯甲烷/甲醇(10/1)洗脫來進行純化,以得到呈黃色油的1.2 g 2-{1-[(5-溴-2-硝基苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙醇(M5-11)(93%)。LCMS: m/z (ESI), [M+H] += 384.00。 步驟12. 5-[5-(2-{1-[(5-溴-2-硝基苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(M5-12)的製備。在0℃下向5-(5-羥基-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M2,0.66 g,2.49 mmol,0.8當量)和三苯基膦(2.46 g,9.369 mmol,3當量)於四氫呋喃(10 mL)中的攪拌混合物中添加偶氮二甲酸二異丙酯(1.89 g,9.36 mmol,3當量)和5-(5-羥基-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(0.66 g,2.49 mmol,0.8當量)。將混合物在氮氣氣氛下在0℃下攪拌2 h。將反應用水(50 mL)淬滅,並且將混合物用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC進行純化並且使用石油醚/乙酸乙酯(1/1)洗脫,以得到呈黃色油的1.2 g 5-[5-(2-{1-[(5-溴-2-硝基苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(M5-12)(57%)。LCMS: m/z (ESI), [M+H] +=629.25。 1H NMR (DMSO-d 6, 400 MHz) δ 1.58-1.76 (9H, m), 2.56-2.79 (2H, m), 2.90 (4H, q), 3.56 (3H, s), 3.72 (6H, d), 4.13 (2H, t), 6.70-6.85 (1H, m), 7.01 (1H, d), 7.95-8.12 (4H, m), 8.28 (1H, d)。 步驟13. 5-[5-(2-{1-[(2-氨基-5-溴苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(M5-13)的製備。在氮氣氣氛下在0℃下向雷尼鎳(0.90 g)和5-[5-(2-{1-[(5-溴-2-硝基苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(1.1 g,1.74 mmol,1當量)於甲醇(8 mL)中的攪拌混合物中添加NH 2NH 2.H 2O(47 mg,0.95 mmol,2當量)。將混合物在0℃下攪拌1 h。將所得混合物過濾。將濾餅用甲醇(2×20 mL)洗滌,並且將溶液在減壓下濃縮。將殘餘物通過Prep-TLC進行純化並且使用二氯甲烷/甲醇(10/1)洗脫,以得到呈淡黃色固體的910 mg 5-[5-(2-{1-[(2-氨基-5-溴苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(M5-13)(76%)。LCMS: m/z (ESI), [M+H] +=599.20。 步驟14. 5-(5-{2-[1-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.2]壬-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M5-14)的製備。將5-[5-(2-{1-[(2-氨基-5-溴苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(900 mg,1.50 mmol,1當量)和BrCN(174 mg,1.65 mmol,1.1當量)於乙醇(10 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將混合物在真空下濃縮。將殘餘物通過Prep-TLC進行純化並且使用二氯甲烷/甲醇(12/1)洗脫,以得到呈淡黃色固體的640 mg 5-(5-{2-[1-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.2]壬-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M5-14)(68%)。LCMS: m/z (ESI), [M+H] +=624.10。 1H NMR (DMSO-d 6, 400 MHz) δ 1.24 (1H, s), 1.78-1.95 (8H, m), 2.60-2.74 (2H, m), 2.85-2.95 (4H, m), 3.57 (3H, s), 3.71 (6H, d), 4.08-4.17 (2H, m), 5.95 (2H, s), 7.04 (2H, s), 7.47 (1H, s), 7.96 (1H, s), 8.12 (1H, ), 8.39 (1H, d)。 步驟15. 5-(5-{2-[1-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.2]壬-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(M5-15)的製備。將5-(5-{2-[1-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.2]壬-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(620 mg,0.99 mmol,1當量)和LiOH(28 mg,1.19 mmol,1.2當量)於四氫呋喃/H 2O(4 mL/1 mL)中的混合物在氮氣氣氛下在80℃下攪拌2 h。將混合物冷卻到室溫,並且在真空下濃縮,以得到呈白色固體的550 mg 5-(5-{2-[1-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.2]壬-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(M5-15)(90%)。LCMS: m/z (ESI), [M+H] +=610.10。 步驟16. 5-溴-15,21-二甲基-23-氧雜-2,9,11,15,20,21, 26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]三十四-3,5,7,9,13,17(34),18(22),19-八烯-12,16-二酮(M5)的製備。將5-(5-{2-[1-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.2]壬-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(550 mg,0.90 mmol,1當量)、 N,N,N,N-四甲基-O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(1027 mg,2.70 mmol,3當量)和 N,N-二異丙基乙胺(174 mg,1.35 mmol,1.5當量)於二噁烷(6 mL)中的混合物在氮氣氣氛下在60℃下攪拌2 h。將混合物冷卻到室溫,並且在真空下濃縮。將殘餘物通過Prep-HPLC用XBridge Prep OBD C18柱,使用含有10 mmol/L NH 4HCO 3的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃色固體的500 mg 5-溴-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十四-3,5,7,9,13,17(34),18(22),19-八烯-12,16-二酮(M5) (88%)。LCMS: m/z (ESI), [M+H] +=592.15。 合成實施例 實施例A1 5,26-二甲基-10-(2,2,2-三氟乙基)-7-氧雜-4,5,10,13,20,22, 26-七氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮 步驟1. 2-((2-((2-硝基苯基)氨基)乙基)氨基)乙-1-醇(INT-A1-1)的製備。將鄰氟硝基苯(10 g,70.871 mmol,1當量)、K 2CO 3(19.59 g,141.742 mmol,2當量)和氨乙基乙醇胺(14.76 g,141.742 mmol,2當量)於ACN(300 mL)中的混合物在氮氣氣氛下在60℃下攪拌2 h。將混合物冷卻到室溫。將反應用水(800 mL)淬滅,並且將混合物用乙酸乙酯(3×600 mL)萃取。將合併的有機層用鹽水(3×500 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法進行純化並且使用PE/EA(1:1)洗脫,以得到呈黃色固體的8.5 g 2-({2-[(2-硝基苯基)氨基]乙基}氨基)乙醇(INT-A1-1)(53%)。 1H NMR (DMSO-d 6, 400 MHz) δ 1.99 (1H, s), 2.62 (2H, t), 2.84 (2H, t), 3.36-3.40 (2H, m), 3.44-3.49 (2H, m), 4.50 (1H, t), 6.65-3.71 (1H, m), 7.04-7.08 (1H, m), 7.54 (1H, m), 8.34-8.38 (1H, t), 8.04-8.08 (1H, m)。 步驟2. 2-({2-[(2-硝基苯基)氨基]乙基}(2,2,2-三氟乙基)氨基)乙醇(INT-A1-2)的製備。將2-({2-[(2-硝基苯基)氨基]乙基}氨基)乙醇(INT-A1-1,2 g,8.879 mmol,1當量)、 N,N-二異丙基乙胺(2.30 g,17.758 mmol,2當量)和三氟甲磺酸2,2,2-三氟乙酯(2.47 g,10.655 mmol,1.2當量)於 N,N-二甲基甲醯胺(20 mL)中的混合物在氮氣氣氛下在60℃下攪拌2 h。將混合物冷卻到室溫。將反應用水(200 mL)淬滅,並且將混合物用二氯甲烷(3×200 mL)萃取。將合併的有機層用鹽水(3×200 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法使用石油醚/乙酸乙酯(1/1)洗脫進行純化,以得到呈黃色固體的2.19 g 2-({2-[(2-硝基苯基)氨基]乙基}(2,2,2-三氟乙基)氨基)乙醇(INT-A1-2)(80%)。LCMS: m/z (ESI), [M+H] +=308.10。 步驟3. 1-甲基-5-{1-甲基-5-[2-({2-[(2-硝基苯基)氨基]乙基}(2,2,2-三氟乙基)氨基)乙氧基]吡唑-4-基}-6-氧代吡啶-3-甲酸甲酯(INT-A1-3)的製備。在氮氣氣氛下在0℃下向5-(5-羥基-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M2,0.56 g,2.14 mmol,1當量)和三苯基膦(1.69 g,6.44 mmol,3當量)於四氫呋喃(20 mL)中的攪拌混合物中添加偶氮二甲酸二異丙酯(1.30 g,6.44 mmol,3當量)和2-({2-[(2-硝基苯基)氨基]乙基}(2,2,2-三氟乙基)氨基)乙醇(0.66 g,2.15 mmol,1當量)。將混合物在氮氣氣氛下在0℃下攪拌2 h。將反應用水(150 mL)淬滅,並且將混合物用二氯甲烷(3×150 mL)萃取。將合併的有機層用鹽水(3×150 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在Prep-TLC上使用石油醚/乙酸乙酯(1/3)洗脫進行純化,以得到呈橙色油的1.15 g 1-甲基-5-{1-甲基-5-[2-({2-[(2-硝基苯基)氨基]乙基}(2,2,2-三氟乙基)氨基)乙氧基]吡唑-4-基}-6-氧代吡啶-3-甲酸甲酯(INT-A1-3)(96%)。LCMS: m/z (ESI), [M+H] +=553.25。 1H NMR (DMSO-d 6 , 400 MHz) δ 3.02 (2H, t), 3.12-3.19 (2H, m), 3.38-3.44 (2H, m), 3.50 (1H, s), 3.56 (3H, s), 3.67 (3H, s), 3.76 (3H, s), 4.03 (2H, t), 6.63-6.72 (1H, m), 6.97-7.05 (1H, m), 7.50-7.58 (1H, m), 7.62-7.68 (1H, m), 7.92 (1H, s), 8.01-8.06 (2H, m), 8.20 (1H, t), 8.37 (1H, d) 步驟4. 5-{5-[2-({2-[(2-氨基苯基)氨基]乙基}(2,2,2-三氟乙基)氨基)乙氧基]-1-甲基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A1-4)的製備。在0℃下向1-甲基-5-{1-甲基-5-[2-({2-[(2-硝基苯基)氨基]乙基}(2,2,2-三氟乙基)氨基)乙氧基]吡唑-4-基}-6-氧代吡啶-3-甲酸甲酯(1.1 g,1.99 mmol,1當量)和雷尼鎳(0.85 g,9.95 mmol,5當量)於甲醇(30 mL)中的攪拌混合物中添加水合肼(0.15 g,2.98 mmol,1.5當量)。將混合物在氮氣氣氛下在0℃下攪拌30分鐘。將所得混合物過濾。將濾餅用甲醇(3×20mL)洗滌,並且將溶液在減壓下濃縮,以得到呈棕色固體的1 g 5-{5-[2-({2-[(2-氨基苯基)氨基]乙基}(2,2,2-三氟乙基)氨基)乙氧基]-1-甲基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A1-4)(96%)。此材料在不經進一步純化的情況下用於下一反應中。LCMS: m/z (ESI), [M+H] +=523.10。 1H NMR (DMSO-d 6 , 400 MHz) δ 2.95 (2H, t), 3.12 (2H, t), 3.17 (2H, d), 3.48 (2H, q), 3.57 (3H, s), 3.68 (3H, s), 3.79 (3H, s), 4.03 (2H, t), 4.36 (2H, s), 6.37-6.46 (2H, m), 7.51-7.59 (2H, m), 7.64-7.67 (1H, m), 7.94 (1H, s), 8.10 (1H, d), 8.45 (1H, d)。 步驟5. 5-[5-(2-{[2-(2-氨基-1,3-苯並二氮唑-1-基)乙基](2,2,2-三氟乙基)氨基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A1-5)的製備。將5-{5-[2-({2-[(2-氨基苯基)氨基]乙基}(2,2,2-三氟乙基)氨基)乙氧基]-1-甲基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸甲酯(1 g,約1.9 mmol,約1當量)和BrCN(0.24 g,2.29 mmol,1.2當量)於CH 2CI 2(20 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將混合物在減壓下濃縮。將殘餘物通過Prep-TLC使用CH 2Cl 2/甲醇(40/1)洗脫進行純化,以得到呈棕色固體的1 g 5-[5-(2-{[2-(2-氨基-1,3-苯並二氮唑-1-基)乙基] (2,2,2-三氟乙基)氨基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A01-5)(95%)。LCMS: m/z (ESI), [M+H] +=548.25。 1H NMR (DMSO-d 6 , 400 MHz) δ 3.04 (2H, t), 3.16 (2H, t), 3.50 (2H, q), 3.58 (3H, s), 3.65 (3H, s), 3.77 (3H, s), 3.98 (2H, t), 4.18 (2H, t), 7.07-7.21 (2H, m), 7.32 (1H, d), 7.40 (1H, d), 7.93 (1H, s), 8.04 (1H, d), 8.11 (2H, s), 8.46 (1H, d)。 步驟6. 5-[5-(2-{[2-(2-氨基-1,3-苯並二氮唑-1-基)乙基](2,2,2-三氟乙基)氨基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-羧酸(INT-A1-6)的製備。將5-[5-(2-{[2-(2-氨基-1,3-苯並二氮唑-1-基)乙基](2,2,2-三氟乙基)氨基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(200 mg,0.36 mmol,1當量)和LiOH.H 2O(23 mg,0.55 mmol,1.5當量)於四氫呋喃(4 mL)和H 2O(1 mL)中的混合物在氮氣氣氛下在室溫下攪拌過夜。將反應混合物在減壓下濃縮。將殘餘物通過C18FLASH使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的180 mg 5-[5-(2-{[2-(2-氨基-1,3-苯並二氮唑-1-基)乙基](2,2,2-三氟乙基)氨基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-羧酸(INT-A1-6)(92%)。LCMS: m/z (ESI), [M+H] +=534.10。 1H NMR (DMSO-d 6 , 400 MHz) δ 2.99 (2H, t), 3.28 (2H, t), 3.52 (6H, s), 3.67 (3H, s), 3.99 (2H, t), 4.18 (2H, t), 6.80-7.01 (4H, m), 7.05-7.14 (2H, m), 8.02 (1H, s), 8.09 (1H, d), 8.37 (1H, d) 步驟7. 5,26-二甲基-10-(2,2,2-三氟乙基)-7-氧雜-4,5, 10,13,20,22,26-七氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(實施例A1)的製備。將5-[5-(2-{[2-(2-氨基-1,3-苯並二氮唑-1-基)乙基](2,2,2-三氟乙基)氨基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-羧酸(150 mg,0.28 mmol,1當量)、N,N,N,N-四甲基-O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(160 mg,0.42 mmol,1.5當量)和 N,N-二異丙基乙胺(109 mg,0.84 mmol,3當量)於1,4-二噁烷(5 mL)中的混合物在氮氣氣氛下在室溫下攪拌過夜。將反應用水(50 mL)淬滅,並且將混合物用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×150 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-HPLC用XBridge Shield RP18 OBD柱,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3.H 2O的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈粉紅色固體的29.5 mg 5,26-二甲基-10-(2,2,2-三氟乙基)-7-氧雜-4,5,10,13,20,22, 26-七氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(實施例A1)(20%)。LCMS: m/z (ESI), [M+H] +=516.20。 1H NMR (DMSO, 400 MHz) δ 3.10 (2H, t), 3.27 (2H, t), 3.63 (3H, s), 3.59-3.69 (2H, m), 3.71 (3H, s), 4.19 (2H, t), 4.33 (2H, t), 7.18-7.31 (2H, m), 7.49-7.56 (1H, m), 7.70 (1H, d), 8.25 (1H, s), 8.29 (1H, d), 8.89 (1H, d), 12.55 (1H, s)。 19F NMR (DMSO-d 6 , 376 MHz) δ 69.23 (s)。 實施例A2 5-環丙基-26-甲基-16-(4-甲基哌嗪-1-基)-10-(2,2,2-三氟乙基)-7-氧雜-4,5,10,13,20,22,26-七氮雜五環[22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮 步驟1. 2-({2-[(5-溴-2-硝基苯基)氨基]乙基}氨基)乙醇(INT-A2-1)的製備。將4-溴-2-氟-1-硝基苯(2 g,9.09 mmol,1當量)、氨乙基乙醇胺(1.42 g,13.63 mmol,1.5當量)和K 2CO 3(3 g,27.27 mmol,3當量)於乙腈(20 mL)中的混合物在氮氣氣氛下在60℃下攪拌2 h。將混合物冷卻到室溫。將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(10/1)洗脫來進行純化,以得到呈黃色固體的2.1 g 2-({2-[(5-溴-2-硝基苯基)氨基]乙基}氨基)乙醇(INT-A2-1)(72%)。LCMS: m/z (ESI), [M+H] += 306.00。 1H NMR (DMSO-d 6 , 400 MHz) δ 2.64 (2H, t), 2.83 (2H, t), 3.18 (1H, d), 3.37-3.44 (2H, m), 3.48 (2H, q), 4.53 (1H, t), 6.83 (1H, d), 7.28 (1H, d), 7.99 (1H, d), 8.42 (1H, t)。 步驟2. 2-({2-[(5-溴-2-硝基苯基)氨基]乙基}(2,2,2-三氟乙基)氨基)乙醇(INT-A2-2)的製備。向2-({2-[(5-溴-2-硝基苯基)氨基]乙基}氨基)乙醇(INT-A2-1,10 g,32.87 mmol,1當量)和 N,N-二異丙基乙胺(12 g,98.63 mmol,3當量)於二甲基甲醯胺(100 mL)中的攪拌混合物中添加三氟甲磺酸2,2,2-三氟乙酯(11 g,49.31 mmol,1.5當量)。將混合物在氮氣氣氛下在60℃下攪拌2 h。將混合物冷卻到室溫。將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-TLC使用石油醚/乙酸乙酯(5/1)洗脫進行純化,以得到呈黃色油的3 g 2-({2-[(5-溴-2-硝基苯基)氨基]乙基}(2,2,2-三氟乙基)氨基)乙醇(INT-A2-2)(21%)。LCMS: m/z (ESI) , [M+H] +=385.90 1H NMR (DMSO-d 6, 400 MHz) δ 2.76 (2H, t), 2.95 (2H, t), 3.40 (4H, d), 3.57-3.45 (2H, m), 4.55 (1H, t), 6.84 (1H, d), 7.27 (1H, d), 8.00 (1H, d), 8.30 (1H, t)。 步驟3. 2-環丙基吡唑-3-醇(INT-A2-3)的製備。將(2E)-3-甲氧基丙-2-烯酸甲酯(1 g,8.61 mmol,1當量)和環丙基肼(621 mg,8.61 mmol,1當量)於甲醇(4 mL)中的混合物在空氣氣氛下在80℃下攪拌16 h。在冷卻到室溫後,將反應用飽和NH 4Cl溶液(300 mL)淬滅,並且將混合物用二氯甲烷(3×200 mL)萃取。將合併的有機層用鹽水(3×300 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過反向快速色譜法使用0%至15%乙腈/水洗脫進行純化,以得到呈黃色油的200 mg 2-環丙基吡唑-3-醇(INT-A2-3)(18%)。LCMS: m/z (ESI), [M+H] +=125.15。 1H NMR (DMSO-d 6, 400 MHz) δ 1.08-0.93 (4H, m), 3.44-3.31 (1H, m), 5.64 (1H, d), 7.62 (1H, d)。 步驟4. 2-環丙基-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-3-酮(INT-A2-4)的製備。在氮氣氣氛下在室溫下向2-環丙基吡唑-3-醇(1.1 g,8.86 mmol,1當量)和2-(三甲基甲矽烷基)乙氧基甲基氯(2.66 g,15.95 mmol,1.8當量)於 N,N-二甲基甲醯胺(10 mL)中的攪拌混合物中分批添加氫化鈉(0.85 g,17.72 mmol,2當量,60%)。將混合物攪拌2 h。將反應用飽和NH 4Cl溶液(300 mL)淬滅,並且將混合物用二氯甲烷(3×200 mL)萃取。將合併的有機層用鹽水(3×300 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上使用二氯甲烷/甲醇(10:1)洗脫進行純化。將產物通過反向快速色譜法使用0%至100%乙腈/水洗脫進行進一步純化,以得到呈棕色固體的330 mg 2-環丙基-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-3-酮(INT-A2-4)(15%)。LCMS: m/z (ESI), [M+H] +=255.20 步驟5. 2-環丙基-4-碘-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-3-酮(INT-A2-5)的製備。將2-環丙基-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-3-酮(4.5 g,17.69 mmol,1當量)和NIS(5.97 g,26.53 mmol,1.5當量於乙腈(50 mL)中的混合物在氮氣氣氛下在0℃下攪拌2 h。將反應通過添加飽和硫代硫酸鈉(水溶液)(100 mL)淬滅,並且將混合物用CH 2Cl 2(3×50 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥,並且在減壓下濃縮,以得到呈黃色固體的5.5 g 2-環丙基-4-碘-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-3-酮(INT-A2-5)(82%)。LCMS: m/z (ESI), [M+H] +=380.95 步驟6. 5-(2-環丙基-3-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A2-6)的製備。將新鮮製備的5-(甲氧基羰基)-1-甲基-2-氧代吡啶-3-基硼酸(M2-2,800 mg,3.8 mmol,1當量)、K 2CO 3(1.31 g,9.48 mmol,2.5當量)、Pd(dppf)Cl 2(554 mg,0.76 mmol,0.2當量)和2-環丙基-4-碘-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-3-酮(1.73 g,4.55 mmol,1.2當量)於1,4-二噁烷(8 mL)和H 2O(2 mL)中的混合物在氮氣氣氛下在80℃下攪拌2 h。在冷卻到室溫後,將反應用水(300 mL)淬滅,並且將混合物用二氯甲烷(3×200 mL)萃取。將合併的有機層用鹽水(3×300 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上使用石油醚/乙酸乙酯(1/1)洗脫進行純化,以得到呈棕色固體的1 g 5-(2-環丙基-3-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A22-4)(92%)。LCMS: m/z (ESI), [M+H] +=420.05。 步驟7. 5-(1-環丙基-5-羥基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A2-7)的製備。將5-(2-環丙基-3-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(750 mg,1.79 mmol,1當量)於含HCl的1,4-二噁烷(10 mL)中的溶液在氮氣氣氛下在60℃下攪拌5 h。將所得混合物冷卻到室溫,並且在減壓下濃縮。將殘餘物溶解於甲醇(10 mL)中,用K 2CO 3中和到pH 7。將混合物過濾。將濾餅用甲醇(2×5 mL)洗滌。將溶液在減壓下濃縮,以得到呈棕色固體的500 mg 5-(1-環丙基-5-羥基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A2-7)(96%)。此材料在不經進一步純化的情況下用於下一步驟中。LCMS: m/z (ESI), [M+H] +=290.05 步驟8. 5-{5-[2-({2-[(5-溴-2-硝基苯基)氨基]乙基} (2,2,2-三氟乙基)氨基)乙氧基]-1-環丙基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A2-8)的製備。在氮氣氣氛下在0℃下向5-(1-環丙基-5-羥基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(0.7 g,2.42 mmol,1當量)和三苯基膦(1.9 g,7.26 mmol,3當量)於四氫呋喃(20 mL)中的混合物中添加偶氮二甲酸二異丙酯(1.4 g,7.26 mmol,3當量)和2-({2-[(5-溴-2-硝基苯基)氨基]乙基}(2,2,2-三氟乙基)氨基)乙醇(INT-A2-2,0.9 g,2.42 mmol,1當量)。將混合物在0℃下攪拌2 h。將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過C18柱,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3 .H 2O的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈橙色固體的400 mg 5-{5-[2-({2-[(5-溴-2-硝基苯基)氨基]乙基}(2,2,2-三氟乙基)氨基)乙氧基]-1-環丙基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A2-8)(25%)。LCMS: m/z (ESI), [M+H] +=658.95。 1H NMR (DMSO-d 6, 400 MHz) δ 0.92-1.03 (2H, m), 0.99-1.10 (2H, m), 2.51 (3H, d), 3.04 (2H, t), 3.17 (2H, t), 3.44 (2H, q), 3.55-3.60 (3H, m), 3.77 (3H, s), 4.12-4.15 (2H, m), 6.80-6.83(1H, m), 7.23 (1H, d), 7.88 (1H, s), 7.94 (1H, d), 8.06 (1H, d), 8.25 (1H, t), 8.38 (1H, d)。 步驟9. 5-{5-[2-({2-[(2-氨基-5-溴苯基)氨基]乙基} (2,2,2-三氟乙基)氨基)乙氧基]-1-環丙基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A2-9)的製備。在氮氣氣氛下在0℃下向5-{5-[2-({2-[(5-溴-2-硝基苯基)氨基]乙基} (2,2,2-三氟乙基)氨基)乙氧基]-1-環丙基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸甲酯(350 mg,0.53 mmol,1當量)和雷尼鎳(365 mg,4.26 mmol,8當量)於甲醇(5 mL)中的混合物中添加水合肼(98%)(40 mg,0.80 mmol,1.5當量)。將所得混合物攪拌2 h,並且過濾。將濾餅用甲醇(3×50 mL)洗滌,並且將溶液在減壓下濃縮,以得到呈棕色固體的300 mg 5-{5-[2-({2-[(2-氨基-5-溴苯基)氨基]乙基}(2,2,2-三氟乙基)氨基)乙氧基]-1-環丙基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A2-9)(約90%)。LCMS: m/z (ESI), [M+H] +=627.15。 1H NMR (DMSO-d 6, 400 MHz) δ 0.91-1.03 (2H, m), 0.99-1.10 (2H, m), 2.98 (2H, t), 3.13-3.19 (4H, m), 3.51 (2H, q), 3.57 (3H, s), 3.79 (3H, s), 4.01-4.07 (3H, m), 4.12 (2H, t), 5.76 (1H, s), 6.38-6.58 (3H, m), 7.91 (1H, d), 8.13 (1H, d), 8.45 (1H, d)。 步驟10. 5-[5-(2-{[2-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)乙基](2,2,2-三氟乙基)氨基}乙氧基)-1-環丙基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A2-10)的製備。將5-{5-[2-({2-[(2-氨基-5-溴苯基)氨基]乙基}(2,2,2-三氟乙基)氨基)乙氧基]-1-環丙基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸甲酯(300 mg,0.48 mmol,1當量)和BrCN(60 mg,0.57 mmol,1.2當量)於二氯甲烷(5 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h,並且在真空下濃縮。將殘餘物在prep-TLC上使用二氯甲烷/甲醇(10/1)洗脫進行純化,以得到呈棕色固體的300 mg 5-[5-(2-{[2-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)乙基](2,2,2-三氟乙基)氨基}乙氧基)-1-環丙基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A2-10)(96%)。LCMS: m/z (ESI), [M+H] +=654.10。 步驟11. 5-[5-(2-{[2-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)乙基](2,2,2-三氟乙基)氨基}乙氧基)-1-環丙基吡唑-4-基]-1-甲基-6-氧代吡啶-3-羧酸(INT-A2-11)的製備。將5-[5-(2-{[2-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)乙基](2,2,2-三氟乙基)氨基}乙氧基)-1-環丙基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(280 mg,0.43 mmol,1當量)和LiOH .H 2O (27 mg,0.64 mmol,1.5當量)於THF(4 mL)和H 2O(1 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h,並且在減壓下濃縮。將殘餘物通過prep-HPLC用XBridge Shield RP18 OBD柱,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3 .H 2O的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈棕色固體的135 mg 5-[5-(2-{[2-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)乙基](2,2,2-三氟乙基)氨基}乙氧基)-1-環丙基吡唑-4-基]-1-甲基-6-氧代吡啶-3-羧酸(INT-A2-11)(49%)。LCMS: m/z (ESI), [M+H] +=638.10。 1H NMR (DMSO-d 6, 400 MHz) δ 0.90-1.03 (2H, m), 0.99-1.09 (2H, m), 3.01 (2H, t), 3.51 (3H, s), 3.53-3.63 (3H, m), 4.08 (2H, t), 4.23 (2H, t), 6.80-7.13 (3H, m), 7.19 (2H, s), 7.31 (1H, d), 7.97-8.05 (2H, m), 8.43 (1H, d) 步驟12. 16-溴-5-環丙基-26-甲基-10-(2,2,2-三氟乙基)-7-氧雜-4,5,10,13,20,22,26-七氮雜五環[22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(INT-A2-12)的製備。將5-[5-(2-{[2-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)乙基](2,2,2-三氟乙基)氨基}乙氧基)-1-環丙基吡唑-4-基]-1-甲基-6-氧代吡啶-3-羧酸(110 mg,0.17 mmol,1當量)、 N,N-二異丙基乙胺(67 mg,0.52 mmol,3當量)和 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(98 mg,0.26 mmol,1.5當量)於1,4-二噁烷(5 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將反應用水(50 mL)淬滅,並且將混合物用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-HPLC用XBridge Shield RP18 OBD柱,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3 .H 2O的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈棕色固體的60 mg 16-溴-5-環丙基-26-甲基-10-(2,2,2-三氟乙基)-7-氧雜-4,5,10,13,20,22,26-七氮雜五環[22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(INT-A2-12)(56%)。LCMS: m/z (ESI), [M+H] +=622.05。 1H NMR (DMSO-d 6, 400 MHz) δ 0.96-1.04 (2H, m), 1.08-1.35 (2H, m), 3.08-3.12 (2H, m), 3.28 (2H, q), 3.49-3.60 (1H, m), 3.62 (3H, s), 3.64-3.77 (2H, m), 4.21-4.33 (2H, m), 4.29 (2H, s), 7.36-7.38 (1H, m), 7.43 (1H, d), 8.09 (1H, d), 8.21 (1H, s), 8.30 (1H, d), 8.88 (1H, d), 12.63 (1H, s)。 步驟13. 5-環丙基-26-甲基-16-(4-甲基哌嗪-1-基)-10-(2,2,2-三氟乙基)-7-氧雜-4,5,10,13,20,22,26-七氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3, 14,16,18,20,24-八烯-23,27-二酮(實施例A2)的製備。在氮氣氣氛下在室溫下向16-溴-5-環丙基-26-甲基-10-(2,2,2-三氟乙基)-7-氧雜-4,5,10,13,20,22,26-七氮雜五環[22.3.1.0^ {2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3, 14,16,18, 20,24-八烯-23,27-二酮(40 mg,0.06 mmol,1當量)、1-甲基哌嗪(26 mg,0.256 mmol,4當量)和BrettPhos Pd G 3(18 mg,0.02 mmol,0.3當量)於1,4-二噁烷(5 mL)中的混合物中添加LiHMDS(65 mg,0.38 mmol,6當量)。將混合物在60℃下攪拌2 h,然後冷卻到室溫。將反應混合物用NH 4Cl(水溶液)溶液(50 mL)處理,並用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-HPLC用Shield RP18 OBD柱,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3 .H 2O的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的2.7 mg 5-環丙基-26-甲基-16-(4-甲基哌嗪-1-基)-10-(2,2,2-三氟乙基)-7-氧雜-4,5,10,13,20,22,26-七氮雜五環[22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(實施例A2)(6%)。LCMS: m/z (ESI), [M+H] +=640.35。 1H NMR (DMSO-d 6, 400 MHz) δ 0.96-1.04 (2H, m), 1.08-1.35 (2H, m), 2.25 (3H, s), 2.48-2.50 (4H, m), 3.04-3.15 (3H, m), 3.18 (4H, t), 3.57-3.64 (4H, m), 3.62 (3H, s), 4.24-4.29 (4H, m), 6.87 (1H, d), 7.23 (1H, s), 7.35 (1H, d), 8.21 (1H, s), 8.27 (1H, s), 8.89 (1H, s), 12.33 (1H, s)。 19F NMR (DMSO-d 6, 376 MHz) δ 69.26 (s)。 實施例A3 5,26-二甲基-16-(4-甲基哌嗪-1-基)-10-(2,2,2-三氟乙基)-7-氧雜-4,5,10,13,20,22,26-七氮雜五環[22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮 步驟1. 甲基-5-{5-[2-({2-[(5-溴-2-硝基苯基)氨基]乙基}(2,2,2-三氟乙基)氨基)乙氧基]-1-甲基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸酯(INT-A3-1)的製備。在0℃下向三苯基膦(1324 mg,5.04 mmol,3當量)和5-(5-羥基-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M2,354 mg,1.34 mmol,0.8當量)於四氫呋喃(9 mL)中的攪拌混合物中添加2-({2-[(5-溴-2-硝基苯基)氨基]乙基}(2,2,2-三氟乙基)氨基)乙醇(650 mg,1.68 mmol,1當量)和偶氮二甲酸二異丙酯(1021 mg,5.04 mmol,3當量)。將混合物在氮氣氣氛下在0℃下攪拌1 h。將反應用水(50 mL)淬滅,並且將混合物用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(10/1)洗脫進行純化,以得到呈黃色油的550 mg 5-{5-[2-({2-[(5-溴-2-硝基苯基)氨基]乙基}(2,2,2-三氟乙基)氨基)乙氧基]-1-甲基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A3-1)(36%)。LCMS: m/z (ESI), [M+H] +=630.85。 1H NMR (DMSO-d 6, 400 MHz) δ 3.01 (2H, t), 3.11 (2H, t), 3.31-3.45 (2H, m)3.49-3.54 (4H, m), 3.66 (3H, s), 3.76 (3H, s), 4.01 (2H, t), 5.73 (1H, s), 6.78-6.92 (1H, m), 7.20 (1H, d), 7.96-7.88 (2H, m), 8.00 (1H, d), 8.23 (1H, t), 8.33 (1H, d)。 步驟2. 5-{5-[2-({2-[(2-氨基-5-溴苯基)氨基]乙基} (2,2,2-三氟乙基)氨基)乙氧基]-1-甲基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A3-2)的製備。向5-{5-[2-({2-[(5-溴-2-硝基苯基)氨基]乙基}(2,2,2-三氟乙基)氨基)乙氧基]-1-甲基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸甲酯(570 mg,0.90 mmol,1當量)和雷尼鎳(30 mg)於甲醇(6 mL)中的攪拌混合物中添加NH 2NH 2 .H 2O(57 mg,1.80 mmol,2當量)。將混合物在氮氣氣氛下在0℃下攪拌30分鐘。將混合物過濾。將濾餅用甲醇(3×30 mL)洗滌,並且將溶液在減壓下濃縮,以得到呈黃色油的270 mg 5-{5-[2-({2-[(2-氨基-5-溴苯基)氨基]乙基}(2,2,2-三氟乙基)氨基)乙氧基]-1-甲基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A3-2)(42%)。LCMS: m/z (ESI) , [M+H] +=601.05。 步驟3. 5-[5-(2-{[2-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)乙基](2,2,2-三氟乙基)氨基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A3-3)的製備。將5-{5-[2-({2-[(2-氨基-5-溴苯基)氨基]乙基}(2,2,2-三氟乙基)氨基)乙氧基]-1-甲基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸甲酯(240 mg,0.39 mmol,1當量)和BrCN(42 mg,0.39 mmol,1當量)於乙醇(6 mL)中的混合物在氮氣氣氛下在0℃下攪拌1 h。將混合物在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(10/1)洗脫進行純化,以得到呈棕色固體的120 mg 5-[5-(2-{[2-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)乙基](2,2,2-三氟乙基)氨基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A3-3)(43%)。LCMS: m/z (ESI), [M+H] +=626.05 步驟4. 5-[5-(2-{[2-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)乙基](2,2,2-三氟乙基)氨基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-羧酸(INT-A3-4)的製備。向5-[5-(2-{[2-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)乙基](2,2,2-三氟乙基)氨基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(100 mg,0.16 mmol,1當量)於四氫呋喃(2 mL)中的攪拌混合物中添加含LiOH .H 2O(7.6 mg,0.32 mmol,2當量)的H 2O(0.5 mL)。將所得混合物在氮氣氣氛下在室溫下攪拌2 h。將混合物在減壓下濃縮。將殘餘物通過C18柱使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的70 mg 5-[5-(2-{[2-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)乙基](2,2,2-三氟乙基)氨基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-羧酸(INT-A3-4)(68%)。LCMS: m/z (ESI), [M+H] +=612.00。 1H NMR (DMSO-d 6, 400 MHz) δ 2.98 (2H, t), 3.51 (3H, s), 3.57 (5H, t), 3.68 (3H, s), 3.99 (2H, t), 4.25 (2H, t), 7.06-6.97 (2H, m), 7.28 (2H, d), 7.33 (3H, s ), 7.98 (1H, d), 8.09 (1H, s), 8.47 (1H, d,)。 步驟5. 16-溴-5,26-二甲基-10-(2,2,2-三氟乙基)-7-氧雜-4,5,10,13,20,22,26-七氮雜五環[22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(INT-A3-5)的製備。向5-[5-(2-{[2-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)乙基](2,2,2-三氟乙基)氨基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-羧酸(65 mg,0.10 mmol,1當量)和 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(60 mg,0.15 mmol,1.5當量)於1,4-二噁烷(2 mL)中的攪拌混合物中添加 N,N-二異丙基乙胺(41 mg,0.31 mmol,3當量),並且將混合物在氮氣氣氛下在60℃下攪拌2 h。冷卻到室溫後,將混合物在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(10/1)洗脫進行純化,以得到呈棕色固體的45 mg 16-溴-5,26-二甲基-10-(2,2,2-三氟乙基)-7-氧雜-4,5,10,13,20,22,26-七氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6), 3,14,16,18,20,24-八烯-23,27-二酮(INT-A3-5)(58%)。LCMS : m/z (ESI), [M+H] +=594.00。 步驟6. 5,26-二甲基-16-(4-甲基哌嗪-1-基)-10-(2,2,2-三氟乙基)-7-氧雜-4,5,10,13,20,22,26-七氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3, 14,16,18,20,24-八烯-23,27-二酮(實施例A3)的製備。在氮氣氣氛下在室溫下向16-溴-5,26-二甲基-10-(2,2,2-三氟乙基)-7-氧雜-4,5,10,13,20,22,26-七氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3, 14,16,18,20,24-八烯-23,27-二酮(50 mg,0.08 mmol,1當量)、BrettPhos Pd G 3(22 mg,0.02 mmol,0.3當量)和哌嗪、1-甲基-(33 mg,0.33 mmol,4當量)於1,4-二噁烷(2 mL)中的攪拌混合物中逐滴添加LiHMDS(0.48 mL,0.48 mmol,6當量)。將混合物在氮氣氣氛下在60℃下攪拌40分鐘。將混合物冷卻到室溫。將反應用水(30 mL)淬滅,並且將混合物用乙酸乙酯(3×20 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-HPLC用XBridge Shield RP18 OBD柱,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3 .H 2O的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃色固體的3.1 mg 5,26-二甲基-16-(4-甲基哌嗪-1-基)-10-(2,2,2-三氟乙基)-7-氧雜-4,5,10,13,20,22,26-七氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(實施例A3)(5%)。LCMS: m/z (ESI), [M+H] +=614.25。 1H NMR (DMSO-d 6, 400 MHz) δ 2.26 (3H, s), 3.04-3.10 (2H, m), 3.18 (4H, t), 3.15-3.27 (5H, m), 3.58-3.64 (4H, m), 3.64-3.78 (5H, m), 4.17 (2H, t), 4.29 (2H, t), 6.85-6.89 (1H, m), 7.23 (1H, d), 7.35 (1H, d), 8.26 (2H, d), 8.88 (1H, d), 12.34 (1H, s)。 19F NMR (DMSO-d 6, 376 MHz) δ -69.31 (s)。 實施例A4 10-(2,2-二氟乙基)-16-{[2-(二甲基氨基)乙基](甲基)氨基}-5,26-二甲基-7-氧雜-4,5,10,13,20,22,26-七氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3, 14,16,18,20,24-八烯-23,27-二酮 步驟1. 2-({2-[(5-溴-2-硝基苯基)氨基]乙基}(2,2,-二氟乙基)氨基)乙醇(INT-A4-1)的製備。將2-({2-[(5-溴-2-硝基苯基)氨基]乙基}氨基)乙醇(2 g,6.57 mmol,1當量)、 N,N-二異丙基乙胺(2.55 g,19.72 mmol,3當量)和三氟甲磺酸2,2-二氟乙酯(1.69 g,7.89 mmol,1.2當量)於二甲基甲醯胺(20 mL)中的混合物在氮氣氣氛下在60℃下攪拌2 h。將混合物冷卻到室溫。將反應用水(80 mL)淬滅,並且將混合物用乙酸乙酯(3×60 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(10/1)洗脫來進行純化,以得到呈黃色油的1.1 g 2-({2-[(5-溴-2-硝基苯基)氨基]乙基}(2,2-二氟乙基)氨基)乙醇(INT-A4-1) (43%)。LCMS: m/z (ESI), [M+H] +=366.00。 1H NMR (DMSO-d 6 , 400 MHz) δ 2.71 (2H, t), 2.90 (2H, t), 2.99 (2H, t), 3.35-3.43 (2H, m), 3.46-3.64 (2H, m), 4.51 (1H, t), 5.87-6.18 (1H, m), 6.83-6.86 (1H, m), 7.27 (1H, d), 7.99 (1H, d), 8.33 (1H, t)。 步驟2. 5-{5-[2-({2-[(5-溴-2-硝基苯基)氨基]乙基} (2,2-二氟乙基)氨基)乙氧基]-1-甲基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A4-2)的製備。在0℃下向三苯基膦(1.38 g,5.29 mmol,3當量)和5-(5-羥基-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M2,836 mg,3.17 mmol,1.8當量)於四氫呋喃(10 mL)中的混合物中添加2-({2-[(5-溴-2-硝基苯基)氨基]乙基}(2,2-二氟乙基)氨基)乙醇(650 mg,1.76 mmol,1當量)和偶氮二甲酸二異丙酯(1.07 g,5.29 mmol,3當量)。將混合物在氮氣氣氛下在0℃下攪拌1 h。將反應用水(80 mL)淬滅,並且將混合物用乙酸乙酯(3×60 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用石油醚/乙酸乙酯(1/3)洗脫進行純化,以得到呈黃色固體的400 mg 5-{5-[2-({2-[(5-溴-2-硝基苯基)氨基]乙基}(2,2-二氟乙基)氨基)乙氧基]-1-甲基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A4-2)(33%)。LCMS: m/z (ESI), [M+H] +=613.00。 步驟3. 5-{5-[2-({2-[(2-氨基-5-溴苯基)氨基]乙基} (2,2-二氟乙基)氨基)乙氧基]-1-甲基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A4-3)的製備。在0℃下向5-{5-[2-({2-[(5-溴-2-硝基苯基)氨基]乙基}(2,2-二氟乙基)氨基)乙氧基]-1-甲基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸甲酯(400 mg,0.65 mmol,1當量)和雷尼鎳(72 mg)於甲醇(5 mL)中的攪拌混合物中添加肼(41 mg,1.30 mmol,2當量)。將混合物在氮氣氣氛下在0℃下攪拌30分鐘。將所得混合物過濾。將濾餅用甲醇(3×10 mL)洗滌,並且將溶液在減壓下濃縮,以得到呈棕色固體的300 mg甲基-5-{5-[2-({2-[(2-氨基-5-溴苯基)氨基]乙基}(2,2-二氟乙基)氨基)乙氧基]-1-甲基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸酯(INT-A4-3)(55%)。LCMS: m/z (ESI) , [M+H]+=583.05。 步驟4. 5-[5-(2-{[2-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)乙基](2,2-二氟乙基)氨基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A4-4)的製備。將5-{5-[2-({2-[(2-氨基-5-溴苯基)氨基]乙基}(2,2-二氟乙基)氨基)乙氧基]-1-甲基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸甲酯(300 mg,0.51 mmol,1當量)和BrCN(54 mg,0.51 mmol,1當量)於乙醇(5 mL)中的混合物在氮氣氣氛下在室溫下攪拌1 h。將混合物在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(10/1)洗脫進行純化,以得到呈棕色固體的250 mg 5-[5-(2-{[2-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)乙基](2,2-二氟乙基)氨基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A4-4)(71%)。LCMS: m/z (ESI) , [M+H]+=608.05。 步驟5. 5-[5-(2-{[2-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)乙基](2,2-二氟乙基)氨基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-羧酸(INT-A4-5)的製備。向5-[5-(2-{[2-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)乙基](2,2-二氟乙基)氨基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(200 mg,0.32 mmol,1當量)於四氫呋喃(4 mL)中的攪拌混合物中添加含LiOH .H 2O(19.6 mg,0.82 mmol,2.5當量)的H 2O(1 mL)。將所得混合物在室溫下攪拌2 h。將混合物在減壓下濃縮。將殘餘物通過C18FLASH,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3 .H 2O的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈棕色固體的175 mg 5-[5-(2-{[2-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)乙基](2,2-二氟乙基)氨基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-羧酸(INT-A4-5) (80%)。LCMS: m/z (ESI), [M+H] +=594.10。 1H NMR (DMSO-d 6 , 400 MHz) δ 2.90 (4H, t), 3.05 (4H, t), 3.27 (2H, d), 3.51 (6H, s), 3.68 (6H, s), 3.97 (4H, t), 4.24 (4H, t), 5.85-6.20 (1H, m), 7.06-6.97 (4H, m), 7.28 (4H, s), 7.32 (2H, d), 7.97 (2H, d), 8.08 (2H, s), 8.46 (1H, s), 8.47 (1H, s)。 步驟6. 16-溴-10-(2,2-二氟乙基)-5,26-二甲基-7-氧雜-4,5,10,13,20,22,26-七氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(INT-A4-6)的製備。將5-[5-(2-{[2-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)乙基](2,2-二氟乙基)氨基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-羧酸(160 mg,0.26 mmol,1當量)、N,N,N,N-四甲基-O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(153 mg,0.40 mmol,1.5當量)和 N,N-二異丙基乙胺(104 mg,0.80 mmol,3當量)於1,4-二噁烷(6 mL)中的混合物在氮氣氣氛下在60℃下攪拌2 h。將混合物冷卻到室溫。將反應用水(30 mL)淬滅,並且將混合物用乙酸乙酯(3×20 mL)萃取。將合併的有機層用鹽水(3×20 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(10/1)洗脫進行純化,以得到呈棕色固體的130 mg 16-溴-10-(2,2-二氟乙基)-5,26-二甲基-7-氧雜-4,5,10,13,20,22,26-七氮雜五環[22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(INT-A4-6)(79%)。LCMS(EB237475-165): m/z (ESI), [M+H]+=576.00; 步驟7. 10-(2,2-二氟乙基)-16-{[2-(二甲基氨基)乙基](甲基)氨基}-5,26-二甲基-7-氧雜-4,5,10,13,20,22,26-七氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(實施例A4)的製備。在氮氣氣氛下在室溫下向16-溴-10-(2,2-二氟乙基)-5,26-二甲基-7-氧雜-4,5,10,13,20,22,26-七氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3, 14,16,18,20,24-八烯-23,27-二酮(100 mg,0.17 mmol,1當量)、BrettPhos Pd G 3(47 mg,0.05 mmol,0.3當量)和[2-(二甲基氨基)乙基](甲基)胺(70 mg,0.69 mmol,4當量)於1,4-二噁烷(3 mL)中的混合物中逐滴添加LiHMDS(0.7 mL,1.02 mmol,6當量)。將混合物在氮氣氣氛下在60℃下攪拌2 h,然後冷卻到室溫。將反應用飽和NH 4Cl(水溶液)溶液(5 mL)淬滅,並且將混合物用乙酸乙酯(3×200 mL)萃取。將合併的有機層用鹽水(3×300 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-HPLC用XBridge Shield RP18 OBD柱,使用含有10 mmol/L NH 4HCO 3+0.1%NH 3 .H 2O的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃色固體的1.2 mg 10-(2,2-二氟乙基)-16-{[2-(二甲基氨基)乙基](甲基)氨基}-5,26-二甲基-7-氧雜-4,5,10,13,20,22,26-七氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(實施例A4) (1%)。LCMS: m/z (ESI), [M+H] +=598.25。 1H NMR (DMSO-d 6, 400 MHz) δ 2.19 (6H, s), 2.39 (2H, t), 2.96 (3H, s), 3.03 (2H, t), 3.11-3.52 (6H, m), 3.61 (3H, s), 3.73 (3H, s), 4.19 (2H, t), 4.28 (2H, t), 6.21 (1H, t), 6.64 (1H, d), 6.84 (1H, s), 7.31 (1H, d), 8.25 (2H, s), 8.87 (1H, s), 12.20 (1H, s)。 19F NMR (DMSO-d 6, 376 MHz) δ 119.603 (s)。 實施例A5 5,26-二甲基-10-(氧雜環丁-3-基)-7-氧雜-4,5,10,13,20,22,26-七氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮 步驟1. 2-({2-[(2-硝基苯基)氨基]乙基}(氧雜環丁-3-基)氨基)乙醇(INT-A5-1)的製備。在室溫下向2-({2-[(2-硝基苯基)氨基]乙基}氨基)乙醇(1 g,4.44 mmol,1當量)、3-氧雜環丁烷酮(383 mg,5.33 mmol,1.2當量)和分子篩(20 mg)於二氯甲烷(30 mL)中的攪拌混合物中添加三乙醯氧基硼氫化鈉(1.88 g,8.88 mmol,2當量)。將混合物在氮氣氣氛下在室溫下攪拌12 h。將反應用水(80 mL)淬滅,並且將混合物用乙酸乙酯(3×60 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用石油醚/乙酸乙酯(1/1)洗脫進行純化,以得到呈紅色油的1.2 g 2-({2-[(2-硝基苯基)氨基]乙基}(氧雜環丁-3-基)氨基)乙醇(INT-A5-1)(95%)。LCMS: m/z (ESI), [M+H] +=282.10。 1H NMR (DMSO-d 6, 400 MHz) δ 2.56-2.65(2H, m), 2.74-2.81(2H, m), 3.31-3.40(2H, m), 3.41-3.49(2H, m), 4.00-4.06(1H, m), 4.42-4.48(2H, m), 4.51-4.58(2H, m), 6.64-6.74(1H, m), 7.01-7.08(1H, m), 7.50-7.59(1H, m), 8.03-8.11(1H, m), 8.32(1H, t)。 步驟2. 1-甲基-5-{1-甲基-5-[2-({2-[(2-硝基苯基)氨基]乙基}(氧雜環丁-3-基)氨基)乙氧基]吡唑-4-基}-6-氧代吡啶-3-甲酸甲酯(INT-A5-2)的製備。在0℃下向5-(5-羥基-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(308 mg,1.17 mmol,1.1當量)和三苯基膦(839 mg,3.20 mmol,3當量)於四氫呋喃(2 mL)中的攪拌混合物中添加偶氮二甲酸二異丙酯(647 mg,3.20 mmol,3當量)和2-({2-[(2-硝基苯基)氨基]乙基}(氧雜環丁-3-基)氨基)乙醇(300 mg,1.07 mmol,1當量)。將混合物在氮氣氣氛下在室溫下攪拌12 h。將反應用水(50 mL)淬滅,並且將混合物用乙酸乙酯(3×30 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用石油醚/乙酸乙酯(1/1)洗脫進行純化,以得到呈黃色固體的200 mg 1-甲基-5-{1-甲基-5-[2-({2-[(2-硝基苯基)氨基]乙基}(氧雜環丁-3-基)氨基)乙氧基]吡唑-4-基}-6-氧代吡啶-3-甲酸甲酯(INT-A5-2)(30%)。LCMS: m/z (ESI), [M+H] += 527.20。 1H NMR (DMSO-d 6, 400 MHz) δ 2.79-2.87(2H, m), 2.94-3.00(2H, m), 3.29-3.40(5H, m), 3.78(3H, s), 3.94-4.03(2H, m), 4.04-4.16(1H, m), 4.42-4.51(2H, m), 4.52-4.60(2H, m), 6.64-6.73(1H, m), 6.94-7.01(1H, m), 7.45-7.53(1H, m), 7.91(1H, s), 8.01-8.08(2H, m), 8.25(1H, t), 8.39(1H, d)。 步驟3. 5-{5-[2-({2-[(2-氨基苯基)氨基]乙基}(氧雜環丁-3-基)氨基)乙氧基]-1-甲基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A5-3)的製備。在0℃下向1-甲基-5-{1-甲基-5-[2-({2-[(2-硝基苯基)氨基]乙基}(氧雜環丁-3-基)氨基)乙氧基]吡唑-4-基}-6-氧代吡啶-3-甲酸甲酯(150 mg,0.29 mmol,1當量)和雷尼鎳(30 mg,0.35 mmol,1.2當量)於甲醇(30 mL)中的攪拌混合物中添加NH 2NH 2.H 2O(90 mg)。將混合物在氮氣氣氛下在0℃下攪拌1 h。將所得混合物過濾。將濾餅用甲醇(2×10 mL)洗滌,並且將溶液在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(10/1)洗脫進行純化,以得到呈黃色固體的120 mg 5-{5-[2-({2-[(2-氨基苯基)氨基]乙基}(氧雜環丁-3-基)氨基)乙氧基]-1-甲基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A5-3)(73%)。LCMS: m/z (ESI), [M+H] +=497.30。 步驟4. 5-[5-(2-{[2-(2-氨基-1,3-苯並二氮唑-1-基)乙基](氧雜環丁-3-基)氨基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A5-4)的製備。將5-{5-[2-({2-[(2-氨基苯基)氨基]乙基}(氧雜環丁-3-基)氨基)乙氧基]-1-甲基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸甲酯(120 mg,0.24 mmol,1當量)和溴化氰(38 mg,0.36 mmol,1.5當量)於乙醇(10 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將混合物在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(5/1)洗脫進行純化,以得到呈黃色固體的82 mg 5-[5-(2-{[2-(2-氨基-1,3-苯並二氮唑-1-基)乙基](氧雜環丁-3-基)氨基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A5-4)(65%)。LCMS: m/z (ESI), [M+H] +=522.25。 1H NMR (DMSO-d 6, 400 MHz) δ 2.77-2.86(2H, m), 2.99-3.07(2H, m), 3.57(3H, s), 3.68(3H, s), 3.77(3H, s), 3.91-3.98(2H, m), 3.98-4.11(3H, m), 4.29-4.36(2H, m), 4.43-4.50(2H, m), 6.39(2H, s), 6.78-6.85(1H, m), 6.87-6.95(1H, m), 7.04-7.14(1H, m), 7.93(1H, s), 8.08(1H, d), 8.45(1H, d)。 步驟5. 5-[5-(2-{[2-(2-氨基-1,3-苯並二氮唑-1-基)乙基](氧雜環丁-3-基)氨基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-羧酸(INT-A5-5)的製備。將5-[5-(2-{[2-(2-氨基-1,3-苯並二氮唑-1-基)乙基](氧雜環丁-3-基)氨基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(82 mg,0.16 mmol,1當量)和氫氧化鋰(13 mg,0.31 mmol,2當量)於四氫呋喃(10 mL)和水(2.5 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將混合物在減壓下濃縮。將殘餘物通過C18FLASH,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3 .H 2O的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈紅色固體的20 mg 5-[5-(2-{[2-(2-氨基-1,3-苯並二氮唑-1-基)乙基](氧雜環丁-3-基)氨基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-羧酸(INT-A5-5)(24%)。LCMS: m/z (ESI), [M+H] +=508.25。 步驟6. 5,26-二甲基-10-(氧雜環丁-3-基)-7-氧雜-4,5,10,13,20,22,26-七氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(實施例A5)的製備。將5-[5-(2-{[2-(2-氨基-1,3-苯並二氮唑-1-基)乙基](氧雜環丁-3-基)氨基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-羧酸(20 mg,0.04 mmol,1當量)、 N, N-二異丙基乙胺(15 mg,0.12 mmol,3當量)和 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(22 mg,0.06 mmol,1.5當量)於1,4-二噁烷(4 mL)中的混合物在氮氣氣氛下在室溫下攪拌3 h。將混合物在減壓下濃縮。將殘餘物通過prep-HPLC用XBridge Prep OBD C18柱,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3 .H 2O的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的8.8 mg 5,26-二甲基-10-(氧雜環丁-3-基)-7-氧雜-4,5,10,13,20,22,26-七氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3, 14,16,18,20,24-八烯-23,27-二酮(實施例A5)(45%)。LCMS: m/z (ESI), [M+H] +=490.20。 1H NMR (DMSO-d 6, 400 MHz) δ 2.95-3.03(2H, m), 3.17-3.24(2H, m), 3.62(3H, s), 3.74(3H, s), 4.13-4.30(5H, m), 4.59-4.64(2H, m), 4.66-4.72(2H, m), 7.19-7.29(2H, m), 7.49-7.54(1H, m), 7.58-7.64(1H, m), 8.24(1H, s), 8.29(1H, d), 8.80(1H, d), 11.69(1H, s)。 實施例A6 10-乙基-5,12,26-三甲基-7-氧雜-4,5,10,13,20,22,26-七氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮 步驟1. N-{1-[乙基(2-羥乙基)氨基]丙-2-基}氨基甲酸叔丁酯(INT-A6-1)的製備。在室溫下向乙基乙醇胺(1 g,11.21 mmol,1當量)和 N-(1-溴丙-2-基)氨基甲酸叔丁酯(3.2 g,13.46 mmol,1.2當量)於乙腈(40 mL)中的攪拌混合物中添加K 2CO 3(4.6 g,33.65 mmol,3當量)。將混合物在60℃下攪拌過夜並且冷卻到室溫。將反應用水(50 mL)淬滅,並且將混合物用CH 2Cl 2(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(10/1)洗脫來進行純化,以得到呈無色油的1 g N-{1-[乙基(2-羥乙基)氨基]丙-2-基}氨基甲酸叔丁酯(INT-A6-1)(36%)。 1H NMR (DMSO-d 6, 400 MHz) δ 1.04 (3H, t), 1.14 (3H, d), 1.46 (9H, s), 2.32-2.47 (2H, m), 2.52-2.70 (4H, m), 3.50-3.63 (2H, m), 3.72-3.80 (1H, m), 4.45 (1H, brs)。 步驟2. 2-[(2-氨基丙基)(乙基)氨基]乙醇(INT-A6-2)的製備。在室溫下向20 mL小瓶中添加 N-{1-[乙基(2--羥乙基)氨基]丙-2-基}氨基甲酸叔丁酯(1 g,4.05 mmol,1當量)和含HCl的1,4-二噁烷(10 mL)。將混合物在室溫下攪拌10 h,並且在減壓下濃縮。將殘餘物用飽和NaHCO 3(水溶液)溶液(30 mL)處理,並且用乙酸乙酯(3×30 mL)萃取。將合併的有機層用鹽水(3×40 mL)洗滌,經無水Na 2SO 4乾燥,並且在減壓下濃縮,以得到呈黃色固體的0.59 g 2-[(2-氨基丙基)(乙基)氨基]乙醇(INT-A6-2)。此材料在不經進一步純化的情況下用於下一步驟中。LCMS: m/z (ESI), [M+H] +=146.15 步驟3. 2-[乙基({2-[(2-硝基苯基)氨基]丙基})氨基]乙醇(INT-A6-3)的製備。在室下向2-[(2-氨基丙基)(乙基)氨基]乙醇(590 mg,約4 mmol,約1當量)和鄰氟硝基苯(569 mg,4.03 mmol,1當量)於乙腈(20 mL)中的攪拌混合物中添加K 2CO 3(1115 mg,8.07 mmol,2當量)。將所得混合物在60℃下攪拌2 h,並且冷卻到室溫。將反應用水(20 mL)淬滅,並且將混合物用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(12/1)洗脫來進行純化,以得到呈黃色固體的640 mg 2-[乙基({2-[(2-硝基苯基)氨基]丙基})氨基]乙醇(INT-A6-3) (59%)。LCMS: m/z (ESI), [M+H] +=268.10。 1H NMR (CDCl 3, 400 MHz) δ 1.16 (3H, t), 1.35 (3H, d), 2.34-3.01 (7H, m), 3.64-3.68 (2H, m), 6.68 (1H, t), 6.96 (1H, s), 7.47 (1H, t), 8.18-8.21 (2H, m)。 步驟4. 5-(5-{2-[乙基({2-[(2-硝基苯基)氨基]丙基})氨基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A6-4)的製備。向三苯基膦(1766 mg,6.73 mmol,3當量)和5-(5-羥基-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M2,590 mg,2.24 mmol,1當量)於四氫呋喃(20 mL)中的攪拌混合物中添加偶氮二甲酸二異丙酯(1361 mg,6.73 mmol,3當量)。將混合物在氮氣氣氛下在0℃下攪拌0.5 h。在0℃下在2分鐘內向上述混合物中添加2-[乙基({2-[(2-硝基苯基)氨基]丙基})氨基]乙醇(600 mg,2.24 mmol,1當量)於四氫呋喃(2 mL)。將混合物在0℃下另外攪拌2 h。將反應用水(20 mL)淬滅,並且將混合物用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用石油醚/乙酸乙酯(1/3)洗脫進行純化,以得到呈黃色油的400 mg 5-(5-{2-[乙基({2-[(2-硝基苯基)氨基]丙基})氨基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A6-4)(34%)。LCMS: m/z (ESI), [M+H] +=513.20。 1H NMR (甲醇-d 4, 400 MHz) δ 1.09 (3H, t), 1.23-1.31 (3H, m), 2.60-2.75 (3H, m), 2.84-2.95 (1H, m), 2.96-3.03 (1H, m), 3.66 (3H, s), 3.68 (3H, s), 3.80-3.92 (4H, m), 4.01 (2H, t), 6.42-6.62 (1H, m), 6.99 (1H, d), 7.39 (1H, d), 7.91 (1H, s), 8.05 (1H, d), 8.16 (1H, d), 8.35 (1H, d)。 步驟5. 5-{5-[2-({2-[(2-氨基苯基)氨基]丙基}(乙基)氨基)乙氧基]-1-甲基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A6-5)的製備。在0℃下向5-(5-{2-[乙基({2-[(2-硝基苯基)氨基]丙基})氨基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(450 mg,0.87 mmol,1當量)和雷尼鎳(50 mg)於甲醇(5 mL)中的攪拌混合物中添加水合肼(140 mg,4.39 mmol,5當量)。將混合物在氮氣氣氛下在室溫下攪拌1 h並且過濾。將濾餅用甲醇(3×20 mL)洗滌,並且將溶液在減壓下濃縮,以得到呈黃色油的260 mg 5-{5-[2-({2-[(2-氨基苯基)氨基]丙基}(乙基)氨基)乙氧基]-1-甲基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A6-5)(61%)。LCMS: m/z (ESI), [M+H] +=483.10。 步驟6. 5-[5-(2-{[2-(2-氨基-1,3-苯並二氮唑-1-基)丙基](乙基)氨基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸二甲酯(INT-A6-6)的製備。將5-{5-[2-({2-[(2-氨基苯基)氨基]丙基}(乙基)氨基)乙氧基]-1-甲基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸甲酯(260 mg,0.53 mmol,1當量)和BrCN(114 mg,1.07 mmol,2當量)於乙醇(5 mL)中的混合物在氮氣氣氛下在室溫下攪拌1 h,並且在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(20/1)洗脫進行純化,以得到呈黃色固體的250 mg 5-[5-(2-{[2-(2-氨基-1,3-苯並二氮唑-1-基)丙基](乙基)氨基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸二甲酯(INT-A6-6)(91%)。LCMS: m/z (ESI), [M+H] +=508.40。 步驟7. 5-[5-(2-{[2-(2-氨基-1,3-苯並二氮唑-1-基)丙基](乙基)氨基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-羧酸(INT-A6-7)的製備。向5-[5-(2-{[2-(2-氨基-1,3-苯並二氮唑-1-基)丙基](乙基)氨基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(240 mg,0.47 mmol,1當量)於四氫呋喃(6 mL)中的攪拌溶液中添加含LiOH.H 2O(33 mg,1.41 mmol,3當量)的H 2O(1.5 mL)。將所得混合物在室溫下攪拌2 h,並且在減壓下濃縮。將殘餘物通過反向快速色譜法使用C18矽膠柱使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃色固體的200 mg [5-(2-{[2-(2-氨基-1,3-苯並二氮唑-1-基)丙基](乙基)氨基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-羧酸(INT-A6-7)(85%)。LCMS: m/z (ESI), [M+H] +=494.40。 步驟8. 10-乙基-5,12,26-三甲基-7-氧雜-4,5,10,13,20, 22,26-七氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(實施例A6)的製備。在空氣氣氛下在室溫下向5-[5-(2-{[2-(2-氨基-1,3-苯並二氮唑-1-基)丙基](乙基)氨基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-羧酸(110 mg,0.22 mmol,1當量)和 N,N-二異丙基乙胺(115 mg,0.89 mmol,4當量)於1,4-二噁烷(6 mL)中的攪拌混合物中添加 N, N, N, N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(169 mg,0.44 mmol,2當量)。將所得混合物在60℃下攪拌2 h,冷卻到室溫,並且在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(12/1)洗脫進行純化,並且通過Prep-HPLC使用XBridge Shield RP18 OBD柱使用含有0.1% NH 4HCO 3和0.1% NH 3.H 2O的水和乙腈進行進一步純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的28.8 mg 10-乙基-5,12,26-三甲基-7-氧雜-4,5,10,13,20,22,26-七氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6), 3,14,16,18,20,24-八烯-23,27-二酮(實施例A6)(27%)。LCMS: m/z (ESI), [M+H] +=476.30。 1H NMR (DMSO-d 6, 400 MHz) δ 1.07 (3H, t), 1.62 (3H, d), 2.53-2.65 (2H, m), 2.75-2.82 (2H, m), 3.33-3.58 (2H, m), 3.62 (3H, s), 3.75 (3H, s), 4.09 (1H, s), 4.28-4.37 (1H, m), 5.15 (1H, s), 7.14-7.29 (2H, m), 7.47-7.60 (1H, m), 7.60-7.71 (1H, m), 8.17 (1H, s), 8.27 (1H, d), 8.88 (1H, d), 12.54 (1H, s)。 實施例A7 10-乙基-5,12,12,26-四甲基-7-氧雜-4,5,10,13,20,22,26-七氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮 步驟1. N-{1-[乙基(2-羥乙基)氨基]-2-甲基丙-2-基}氨基甲酸叔丁酯(INT-A7-1)的製備。將 N-(2-甲基-1-氧代丙-2-基)氨基甲酸叔丁酯(5 g,26.70 mmol,1當量)、三乙胺(8 g,80.11 mmol,3當量)和乙基乙醇胺(2.4 g,26.70 mmol,1.00當量)於二氯甲烷(50 mL)中的混合物在氮氣氣氛下在室溫下攪拌1 h。然後添加NaBH(OAc) 3(17 g,80.11 mmol,3當量),並且將混合物在氮氣氣氛下在室溫下攪拌過夜。將反應用水(300 mL)淬滅,並且將混合物用乙酸乙酯(3×200 mL)萃取。將合併的有機層用鹽水(3×300 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過C18柱使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈無色油的5 g N-{1-[乙基(2-羥乙基)氨基]-2-甲基丙-2-基}氨基甲酸叔丁酯(INT-A7-1)(72%)。LCMS: m/z (ESI), [M+H] +=261.10。 1H NMR (DMSO- d 6, 400 MHz) δ 0.93 (3H, t), 1.13-1.14 (6H, s), 1.36-1.39 (9H, d), 2.49-2.53 (3H, m), 2.53-2.56 (3H, m), 3.42-3.43 (2H, m), 4.31 (1H, s), 6.25 (1H, s)。 步驟2. 2-((2-氨基-2-甲基丙基)(乙基)氨基)乙-1-醇(INT-A7-2)的製備。 將 N-{1-[乙基(2-羥乙基)氨基]-2-甲基丙-2-基}氨基甲酸叔丁酯(2 g,3.84 mmol,1當量)於三氟乙酸(5 mL)和二氯甲烷(15 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將所得混合物在減壓下濃縮。將殘餘物用飽和NaHCO 3(水溶液)溶液(100 mL)處理,並且用二氯甲烷(3×100 mL)萃取。將合併的有機層用鹽水(3×200 mL)洗滌,經無水Na 2SO 4乾燥,並且在減壓下濃縮,以得到呈無色油的1.23 g 2-((2-氨基-2-甲基丙基)(乙基)氨基)乙-1-醇(INT-A7-2)。此材料在不經進一步純化的情況下用於下一步驟中。LCMS: m/z (ESI), [M+H] +=161.10。 步驟3. 2-[乙基({2-甲基-2-[(2-硝基苯基)氨基]丙基})氨基]乙醇(INT-A7-3)的製備。將2-[(2-氨基-2-甲基丙基) (乙基)氨基]乙醇(2 g,12.48 mmol,1當量)、K 2CO 3(5 g,37.44 mmol,3當量)和鄰氟硝基苯(1.7 g,12.48 mmol,1當量)於乙腈(20 mL)中的混合物在氮氣氣氛下在80℃下攪拌過夜。將混合物冷卻到室溫。將反應用水(100 mL)淬滅,並且將混合物用二氯甲烷(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-TLC使用二氯甲烷/甲醇(20/1)洗脫來進行純化,以得到呈黃色油的1.2 g 2-[乙基({2-甲基-2-[(2-硝基苯基)氨基]丙基})氨基]乙醇(INT-A7-3)(34%)。LCMS: m/z (ESI), [M+H] +=282.05。 1H NMR (DMSO- d 6, 400 MHz) δ 0.97 (3H, t), 1.41 (6H, s), 2.62-2.66 (3H, m), 2.69 (2H, s), 3.17 (1H, d), 3.44-3.50 (2H, m), 4.37 (1H, t), 6.62-6.68 (1H, m), 7.26-7.33 (1H, m), 7.44-7.52 (1H, m), 8.04-8.10 (1H, m), 8.62-8.68 (1H, s)。 步驟4. 5-(5-(2-(乙基(2-甲基-2-((2-硝基苯基)氨基)丙基)氨基)乙氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-甲酸甲酯(INT-A7-4)的製備。在氮氣氣氛下在0℃下向5-(5-羥基-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M2,281 mg,1.06 mmol,1當量)、三苯基膦(839 mg,3.19 mmol,3當量)於四氫呋喃(10 mL)中的攪拌混合物中添加偶氮二甲酸二異丙酯(646 mg,3.19 mmol,3當量)和2-[乙基({2-甲基-2-[(2-硝基苯基)氨基]丙基})氨基]乙醇(300 mg,1.06 mmol,1當量)。將混合物攪拌1 h,用水(100 mL)處理,並且用二氯甲烷(2×100 mL)萃取。將合併的有機層用鹽水(2×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-TLC使用二氯甲烷/甲醇(12/1)洗脫進行純化,以得到呈黃色固體的230 mg 5-(5-(2-(乙基(2-甲基-2-((2-硝基苯基)氨基)丙基)氨基)乙氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-甲酸甲酯(INT-A7-4)(41%)。LCMS: m/z (ESI), [M+H] +=527.20。 1H NMR (DMSO- d 6, 400 MHz) δ 0.98 (3H, t), 1.39 (6H, s), 2.67-2.69 (2H, m), 2.74 (2H, s), 2.98 (2H, t), 3.56 (3H, s), 3.65 (3H, s), 3.77 (3H, s), 4.00 (2H, t), 5.77 (1H, s), 6.62-6.67 (1H, m), 7.23-7.28 (1H, d), 7.41-7.48 (1H, m), 7.90-7.93 (1H, s), 8.00-8.04 (2H, m), 8.40 (1H, d), 8.58 (1H, s) 步驟5. 5-{5-[2-({2-[(2-氨基苯基)氨基]-2-甲基丙基} (乙基)氨基)乙氧基]-1-甲基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A7-5)的製備。在氮氣氣氛下在0℃下向5-(5-{2-[乙基({2-甲基-2-[(2-硝基苯基)氨基]丙基})氨基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(230 mg,0.44 mmol,1當量)和雷尼鎳(50 mg)於甲醇(2 mL)中的攪拌混合物中添加NH 2NH 2 .H 2O(22 mg,0.44 mmol,1當量)。將混合物在0℃下攪拌1 h。將反應用水(50 mL)淬滅,並且將混合物用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-TLC使用二氯甲烷/甲醇(10/1)洗脫進行純化,以得到呈黃色固體的110 mg 5-{5-[2-({2-[(2-氨基苯基)氨基]-2-甲基丙基}(乙基)氨基)乙氧基]-1-甲基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A7-5)(51%)。LCMS: m/z (ESI), [M+H] +=497.40。 1H NMR (DMSO- d 6, 400 MHz) δ 0.97 (3H, t), 1.14 (6H, s), 2.62-2.69 (2H, m), 2.94 (2H, t), 3.18 (2H, d), 3.58 (3H, s), 3.67 (4H, s), 3.80 (3H, s), 4.00 (2H, t), 4.38 (5H, s), 4.47 (2H, s), 6.35-6.39 (2H, m), 6.47-6.54 (4H, m), 6.72 (1H, d), 7.96 (1H, s), 8.11 (1H, d), 8.46 (1H, d)。 步驟6. 5-(5-(2-((2-(2-氨基-1H-苯並[d]咪唑-1-基)-2-甲基丙基)(乙基)氨基)乙氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-甲酸甲酯(INT-A7-6)的製備。將5-{5-[2-({2-[(2-氨基苯基)氨基]-2-甲基丙基}(乙基)氨基)乙氧基]-1-甲基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸甲酯(200 mg,0.40 mmol,1當量)和BrCN(51 mg,0.48 mmol,1.2當量)於二氯甲烷(5 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h,並且在真空下濃縮。將殘餘物通過prep-TLC使用二氯甲烷/甲醇(12/1)洗脫進行純化,以得到呈黃色固體的120 mg 5-(5-(2-((2-(2-氨基-1H-苯並[d]咪唑-1-基)-2-甲基丙基)(乙基)氨基)乙氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-甲酸甲酯(INT-A7-6) (61%)。LCMS: m/z (ESI), [M+H] +=522.30。 1H NMR (DMSO- d 6, 400 MHz) δ 0.82 (3H, t), 1.81 (6H, s), 2.44-2.48 (3H, m), 2.79 (2H, t), 3.02 (2H, s), 3.58 (6H, d), 3.78-3.82 (5H, m), 6.88 (1H, t), 6.99 (1H, t), 7.19 (1H, s), 7.55 (1H, d), 7.95 (1H, s), 8.05 (1H, d), 8.47 (1H, d) 步驟7. 5-(5-(2-((2-(2-氨基-1H-苯並[d]咪唑-1-基)-2-甲基丙基)(乙基)氨基)乙氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-羧酸(INT-A7-7)的製備。將5-[5-(2-{[2-(2-氨基-1,3-苯並二氮唑-1-基)-2-甲基丙基](乙基)氨基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(120 mg,0.23 mmol,1當量)和LiOH .H 2O(39 mg,0.92 mmol,4當量)於四氫呋喃/H 2O(2 mL/0.5 mL)中的混合物在氮氣氣氛下在40℃下攪拌2 h。將混合物冷卻到室溫,並且在減壓下濃縮。將殘餘物通過C18柱使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的65 mg 5-(5-(2-((2-(2-氨基-1H-苯並[d]咪唑-1-基)-2-甲基丙基)(乙基)氨基)乙氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-羧酸(INT-A7-7)(56%)。LCMS: m/z (ESI), [M+H] +=508.15。 1H NMR (DMSO- d 6, 400 MHz) δ 0.77 (3H, t), 1.79 (6H, s), 2.35-2,42 (3H, m), 2.67-2.69 (1H, m), 2.81 (2H, t), 2.99 (2H, s), 3.48 (3H, s), 3.56 (3H, s), 3.82 (2H, t), 6.30 (2H, s), 6.78 (1H, t), 6.86-6.93 (1H, m), 7.06-7.11 (1H, m), 7.46 (1H, d), 7.86 (1H, s), 7.96 (1H, s), 8.17 (1H, d) 步驟8. 10-乙基-5,12,12,26-四甲基-7-氧雜-4,5,10,13,20,22,26-七氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(實施例A7)的製備。將5-[5-(2-{[2-(2-氨基-1,3-苯並二氮唑-1-基)-2-甲基丙基](乙基)氨基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-羧酸(65 mg,0.13 mmol,1當量)、N,N,N,N-四甲基-O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(97 mg,0.25 mmol,2當量)和 N,N-二異丙基乙胺(33 mg,0.25 mmol,2當量)於1,4-二噁烷(2 mL)中的混合物在氮氣氣氛下在60℃下攪拌2 h。將混合物冷卻到室溫。將反應用水(50 mL)淬滅,並且將混合物用二氯甲烷(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-HPLC用XBridge Shield RP18 OBD柱,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3 .H 2O的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的13.6 mg 10-乙基-5,12,12,26-四甲基-7-氧雜-4,5,10,13,20,22,26-七氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(實施例A7)(22%)。LCMS: m/z (ESI), [M+H] += 490.20。 1H NMR (DMSO- d 6, 400 MHz) δ 0.99 (3H, t), 1.94 (6H, s), 2.68 (2H, d), 3.08-3.22 (2H, m), 3.60-3.64 (5H, m), 3.73 (3H, s), 4.28 (2H, d), 7.17 (2H, t), 7.56-7.63 (1H, m), 7.83 (1H, d), 8.13 (1H, d), 8.29 (1H, d), 8.67 (1H, d), 12.93 (1H, s)。 實施例A8 15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜六環[24.4.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十二-3,5,7,9,13,17(32),18(22),19-八烯-12,16-二酮 步驟1. 3-((2-硝基苯基)氨基)氮雜環庚烷-1-甲酸叔丁酯(INT-A8-1)的製備。向鄰氟硝基苯(5 g,35.43 mmol,1當量)和3-氨基氮雜環庚烷-1-甲酸叔丁酯(7.9 g,37.20 mmol,1.05當量)於乙腈(80 mL)中的攪拌混合物中添加K 2CO 3(9.7 g,70.87 mmol,2當量)。將混合物在氮氣氣氛下在80℃下攪拌3 h,並且冷卻到室溫。將反應用水(100 mL)淬滅,並且將混合物用CH 2Cl 2(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上進行純化並且使用石油醚/乙酸乙酯(10:1)洗脫,以得到呈棕色油的10.6 g 3-[(2-硝基苯基)氨基]氮雜環庚烷-1-甲酸叔丁酯(INT-A8-1) (89%)。LCMS: m/z (ESI), [M+H] +=336.15。 1H NMR (DMSO-d 6, 400 MHz) δ 1.37 (9H, d), 1.47-2.03 (6H, m), 3.34-3.49 (3H, m), 3.51-3.62 (1H, m), 3.93 (1H, s), 6.61-6.75 (1H, m), 7.20 (1H, m), 7.47-7.55 (1H, m), 7.97-8.22 (2H, m)。 步驟2. N-(2-硝基苯基)氮雜環庚-3-胺(INT-A8-2)的製備。將3-[(2-硝基苯基)氨基]氮雜環庚烷-1-甲酸叔丁酯(12 g,35.77 mmol,1當量)和HCl於1,4-二噁烷(100 mL)中的混合物在室溫下攪拌1.5 h。將混合物在減壓下濃縮,並且用CH 2Cl 2(60 mL)處理。將混合物用飽和NaHCO 3(水溶液)溶液(50 mL)處理,並且用CH 2Cl 2(3×100 mL)萃取。將合併的有機層經無水Na 2SO 4乾燥,並且在減壓下濃縮,以得到呈黃色油的8 g N-(2-硝基苯基)氮雜環庚-3-胺(INT-A8-2)。此材料在不經進一步純化的情況下用於下一反應中。LCMS: m/z (ESI), [M+H] +=236.20。 步驟3. 1-(2-((叔丁基二甲基甲矽烷基)氧基)乙基)- N-(2-硝基苯基)氮雜環庚-3-胺(INT-A8-3)的製備。向 N-(2-硝基苯基)氮雜環庚-3-胺(1 g,4.25 mmol,1當量)和(2-溴乙氧基)(叔丁基)二甲基矽烷(2.03 g,8.50 mmol,2當量)於 N, N-二甲基甲醯胺(15 mL)中的攪拌混合物中添加K 2CO 3(1.17 g,8.50 mmol,2當量)和KI(1.41 g,8.50 mmol,2當量)。將所得混合物在氮氣氣氛下在60℃下攪拌2 h。將混合物冷卻到室溫。將反應用水(100 mL)淬滅,並且將混合物用CH 2Cl 2(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上進行純化並且使用乙酸乙酯/石油醚(1/10)洗脫,以得到呈黃色油的819 mg 1-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}- N-(2-硝基苯基)氮雜環庚-3-胺(INT-A8-3)(48%)。LCMS: m/z (ESI), [M+H] +=394.20。 1H NMR (DMSO-d 6, 400 MHz) δ -0.02 (6H, d), 0.81 (9H, s), 1.35-1.65 (3H, m), 1.65-1.85 (3H, m), 2.57-2.80 (3H, m), 2.81-3.03 (3H, m), 3.61-3.82 (2H, m), 3.93 (1H, s), 6.64-6.66 (1H, m), 7.04 (1H, d), 7.45-7.55 (1H, m), 8.02-8.09 (1H, m), 8.73 (1H, d)。 步驟4. 2-(3-((2-硝基苯基)氨基)氮雜環庚-1-基)乙-1-醇(INT-A8-4)的製備。向1-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}- N-(2-硝基苯基)氮雜環庚-3-胺(447 mg,1.13 mmol,1當量)於四氫呋喃(10 mL)中的溶液中添加四丁基氟化銨(593 mg,2.272 mmol,2當量)。將混合物在室溫下攪拌2 h。將反應用水(50 mL)淬滅,並且將混合物用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥,並且在減壓下濃縮,以得到呈黃色油的447 mg 2-{3-[(2-硝基苯基)氨基]氮雜環庚-1-基}乙醇(INT-A8-4)。此材料在不經進一步純化的情況下用於下一反應中。LCMS: m/z (ESI), [M+H] +=280.05。 步驟5. 1-甲基-5-(1-甲基-5-(2-(3-((2-硝基苯基)氨基)氮雜環庚-1-基)乙氧基)-1H-吡唑-4-基)-6-氧代-1,6-二氫吡啶-3-甲酸甲酯(INT-A8-5)的製備。在氮氣氣氛下在0℃下向5-(5-羥基-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M2,207 mg,0.78 mmol,1.1當量)和三苯基膦(563 mg,2.14 mmol,3當量)於四氫呋喃(10 mL)中的攪拌混合物中添加偶氮二甲酸二異丙酯(434 mg,2.14 mmol,3當量)和2-{3-[(2-硝基苯基)氨基]氮雜環庚-1-基}乙醇(200 mg,0.71 mmol,1當量)。將所得混合物在室溫下攪拌過夜。將反應用水(50 mL)淬滅,並且將混合物用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用乙酸乙酯洗脫進行純化,以得到呈橙色固體的301 mg 1-甲基-5-[1-甲基-5-(2-{3-[(2-硝基苯基)氨基]氮雜環庚-1-基}乙氧基)吡唑-4-基]-6-氧代吡啶-3-甲酸甲酯(INT-A8-5)(80%)。LCMS: m/z (ESI), [M+H] +=525.30 1H NMR (DMSO-d 6, 400 MHz) δ 1.22-1.63 (7H, m), 2.45-2.68 (1H, m), 2.79-2.89 (2H, m), 2.95-3.05 (3H, m), 3.55 (3H, s), 3.68 (3H, s), 3.73 (3H, s), 4.05-4.10 (2H, m), 6.60-6.70 (1H, m), 6.95-7.09 (1H, m), 7.40-7.53 (1H,m), 7.94 (1H, s), 7.99-8.05 (2H, m), 8.32 (1H, d), 8.61 (1H, d) 步驟6. 5-(5-(2-(3-((2-氨基苯基)氨基)氮雜環庚-1-基)乙氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-甲酸甲酯(INT-A8-6)的製備。在0℃下向雷尼鎳(100 mg)和1-甲基-5-[1-甲基-5-(2-{3-[(2-硝基苯基)氨基]氮雜環庚-1-基}乙氧基)吡唑-4-基]-6-氧代吡啶-3-甲酸甲酯(300 mg,0.54 mmol,1當量)於甲醇(10 mL)中的攪拌混合物中添加NH 2NH 2 .H 2O(84 mg,0.28 mmol,3當量)。將混合物在室溫下攪拌2 h並且過濾。將濾餅用甲醇(3×5 mL)洗滌,並且將溶液在減壓下濃縮。將殘餘物通過Prep-TLC進行純化並且使用二氯甲烷/甲醇(10/1)洗脫,以得到呈黃色油的89 mg 5-[5-(2-{3-[(2-氨基苯基)氨基]氮雜環庚-1-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A8-6)(44%)。LCMS: m/z (ESI), [M+H] +=495.30 步驟7. 5-(5-(2-(3-(2-氨基-1H-苯並[d]咪唑-1-基)氮雜環庚-1-基)乙氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-甲酸甲酯(INT-A8-7)的製備。將5-[5-(2-{3-[(2-氨基苯基)氨基]氮雜環庚-1-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(89 mg,0.18 mmol,1當量)和BrCN(20 mg,0.19 mmol,1.10當量)於CH 2Cl 2(5 mL)中的混合物在室溫下攪拌5 h,並且在減壓下濃縮。將殘餘物通過Prep-TLC進行純化並且使用二氯甲烷/甲醇(10/1)洗脫,以得到呈白色固體的21 mg 5-(5-{2-[3-(2-氨基-1,3-苯並二氮唑-1-基)氮雜環庚-1-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸二甲酯(INT-A8-7)(22%)。LCMS: m/z (ESI), [M+H] +=520.25 步驟8. 5-(5-(2-(3-(2-氨基-1H-苯並[d]咪唑-1-基)氮雜環庚-1-基)乙氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-羧酸(INT-A8-8)的製備。向5-(5-{2-[3-(2-氨基-1,3-苯並二氮唑-1-基)氮雜環庚-1-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(60 mg,0.11 mmol,1當量)於四氫呋喃(4 mL)和H 2O(1 mL)中的攪拌混合物中添加LiOH.H 2O(8 mg,0.34 mmol,3當量)。將所得混合物在室溫下攪拌8 h,並且在減壓下濃縮。將混合物通過反向快速色譜法使用C18矽膠柱使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的16.1 mg 5-(5-{2-[3-(2-氨基-1,3-苯並二氮唑-1-基)氮雜環庚-1-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A8-8)(42%)。LCMS: m/z (ESI), [M+H] +=506.20 步驟9. 15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜六環[24.4.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十二-3,5,7,9,13,17(32),18(22),19-八烯-12,16-二酮(實施例A8)的製備。向5-(5-{2-[3-(2-氨基-1,3-苯並二氮唑-1-基)氮雜環庚-1-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(15 mg,0.03 mmol,1當量)和N,N,N,N-四甲基-O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(33 mg,0.09 mmol,3當量)於1,4-二噁烷(2 mL)中的攪拌混合物中添加 N,N-二異丙基乙胺(11 mg,0.09 mmol,3當量)。將所得混合物在室溫下攪拌3 h,並且濃縮。將殘餘物通過Prep-HPLC用XBridge Shield RP18 OBD柱,使用含有0.1% NH 4HCO 3的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的4 mg 15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜六環[24.4.1.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十二-3,5,7,9,13,17(32), 18(22),19-八烯-12,16-二酮(實施例A8)(27%)。LCMS: m/z (ESI), [M+H] +=488.15。 1H NMR (DMSO-d 6, 400 MHz) δ 1.65-2.03 (5H, m), 2.54-2.72 (1H, m), 2.72-3.03 (5H, m), 3.08-3.22 (1H, m), 3.63 (3H, s), 3.73 (3H, s), 3.80-3.94 (1H, m), 4.34-4.60 (1H, m), 5.14 (1H, s), 7.16-7.26 (2H, m), 7.50-7.57 (1H, m), 7.62 (1H, d), 8.27 (1H, d), 8.31 (1H, s), 8.98 (1H, s), 12.50 (1H, s)。 實施例A9 15,21-二甲基-5-(嗎啉-4-基)-23-氧雜-2,9,11,15,20,21, 26-七氮雜七環[24.4.1.1^{1,27}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]三十三-3,5,7,9,13,17(33), 18(22),19-八烯-12,16-二酮 步驟1. 1-[(5-溴-2-硝基苯基)氨基]-6-氮雜雙環[3.2.1]辛烷-6-甲酸叔丁酯(INT-A9-1)的製備。將1-氨基-6-氮雜雙環[3.2.1]辛烷-6-甲酸叔丁酯鹽酸鹽(1 g,3.81 mmol,1當量)、4-溴-2-氟-1-硝基苯(2.51 g,11.42 mmol,3當量)和K 2CO 3(2.10 g,15.22 mmol,4當量)於乙腈(20 mL)中的混合物在60℃下攪拌2 h。冷卻到室溫後,將反應用水(200 mL)處理,並且將混合物用乙酸乙酯(3×200 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法使用石油醚/乙酸乙酯(4/1)洗脫來進行純化,以得到呈黃色固體的1.25 g 1-[(5-溴-2-硝基苯基)氨基]-6-氮雜雙環[3.2.1]辛烷-6-甲酸叔丁酯(INT-A9-1)(77%)。LCMS: m/z (ESI), [M+H - 56] +=371.85。 1H NMR (DMSO-d 6, 400 MHz) δ 1.40-1.44 (10H, m), 1.48-1.75 (2H, m), 1.79-1.82 (1H, m), 1.98-2.00 (1H, m), 2.13-2.29 (3H, m), 3.23-3.33 (1H, m), 4.01-4.12 (2H, m), 6.92 (1H, d), 7.20 (1H, s), 8.03 (1H, d), 8.28 (1H, s)。 步驟2. N-(5-溴-2-硝基苯基)-6-氮雜雙環[3.2.1]辛-1-胺(INT-A9-2)的製備。在室溫下,向1-[(5-溴-2-硝基苯基)氨基]-6-氮雜雙環[3.2.1]辛烷-6-甲酸叔丁酯(1.28 g,3.00 mmol,1當量)於二氯甲烷(10 mL)中的攪拌混合物中添加TFA(10 mL)。將所得混合物在室溫下攪拌2 h,並且在減壓下濃縮。將殘餘物用飽和NaHCO 3(水溶液)溶液(200 mL)處理,並且用二氯甲烷(3×200 mL)萃取。將合併的有機層經無水Na 2SO 4乾燥,並且在減壓下濃縮,以得到呈淡黃色固體的950 mg N-(5-溴-2-硝基苯基)-6-氮雜雙環[3.2.1]辛-1-胺(INT-A9-2)(97%)。LCMS: m/z (ESI), [M+H] += 327.90。 1H NMR (CD 3OD, 400 MHz) δ 1.51-1.64 (1H, m), 1.72-1.76 (2H, m), 1.80-1.95 (2H, m), 1.97 (1H, d), 2.21-2.33 (2H, m), 3.02-3.06 (1H, m), 3.54-3.65 (2H, m), 6.83-6.86 (1H, m), 7.14 (1H, d), 8.07 (1H, d)。 步驟3. N-(5-溴-2-硝基苯基)-6-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-6-氮雜雙環[3.2.1]辛-1-胺(INT-A9-3)的製備。將 N-(5-溴-2-硝基苯基)-6-氮雜雙環[3.2.1]辛-1-胺(580 mg,1.78 mmol,1當量)、(2-溴乙氧基)(叔丁基)二甲基矽烷(850 mg,3.56 mmol,2當量)、NaI(533 mg,3.56 mmol,2當量)和K 2CO 3(491 mg,3.56 mmol,2當量)於 N,N-二甲基甲醯胺(10 mL)中的混合物在氮氣氣氛下在60℃下攪拌2 h。在冷卻到室溫後,將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過反向快速色譜法使用20%至100%乙腈/水洗脫進行進一步純化,以得到呈淡黃色固體的600 mg N-(5-溴-2-硝基苯基)-6-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-6-氮雜雙環[3.2.1]辛-1-胺(INT-A9-3)(69%)。LCMS: m/z (ESI), [M+H] +=484.51。 1H NMR (CD 3OD, 400 MHz) δ 0.11 (6H, d), 0.93 (9H, s), 1.38-1.57 (2H, m), 1.62-1.78 (2H, m), 1.83-1.86 (2H, m), 2.00 (1H, d), 2.28 (1H, d), 2.33-2.42 (1H, m), 2.91 (2H, t), 3.36-3.42 (2H, m), 3.79-3.83 (2H, m), 6.82-6.86 (1H, m), 7.09 (1H, d), 8.07 (1H, d)。 步驟4. 2-{1-[(5-溴-2-硝基苯基)氨基]-6-氮雜雙環[3.2.1]辛-6-基}乙醇(INT-A9-4)的製備。在室溫下向 N-(5-溴-2-硝基苯基)-6-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-6-氮雜雙環[3.2.1]辛-1-胺(598 mg,1.23 mmol,1當量)於四氫呋喃(9 mL)中的攪拌混合物中添加四丁基氟化銨(2 mL,四氫呋喃中1 M)。在室溫下攪拌2 h後,將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(5×100 mL)洗滌,經無水Na 2SO 4乾燥,並且在減壓下濃縮,以得到呈黃色固體的360 mg 2-{1-[(5-溴-2-硝基苯基)氨基]-6-氮雜雙環[3.2.1]辛-6-基}乙醇(INT-A9-4)(78%)。LCMS: m/z (ESI), [M+H] += 370.24。 步驟5. 5-[5-(2-{1-[(5-溴-2-硝基苯基)氨基]-6-氮雜雙環[3.2.1]辛-6-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A9-5)的製備。在氮氣氣氛下在0℃下向5-(5-羥基-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M2,268 mg,1.02 mmol,1當量)和三苯基膦(714 mg,2.72 mmol,2.8當量)於四氫呋喃(10 mL)中的攪拌混合物中添加偶氮二甲酸二異丙酯(491 mg,2.43 mmol,2.5當量),隨後添加含2-{1-[(5-溴-2-硝基苯基)氨基]-6-氮雜雙環[3.2.1]辛-6-基}乙醇(360 mg,0.97 mmol,1當量)的四氫呋喃(3 mL)。在0℃下攪拌30分鐘後,將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法使用石油醚/乙酸乙酯(1/9)洗脫進行純化,以得到呈淡黃色固體的470 mg 5-[5-(2-{1-[(5-溴-2-硝基苯基)氨基]-6-氮雜雙環[3.2.1]辛-6-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A9-5)(78%)。LCMS: m/z (ESI), [M+H] +=617.15。 1H NMR (CD 3OD, 400 MHz) δ 1.39-1.51 (1H, m), 1.62-1.74 (2H, m), 1.85 (3H, q), 2.24-2.28 (2H, m), 3.12-3.16 (2H, m), 3.18-3.25 (3H, m), 3.64 (3H, s), 3.79 (3H, s), 3.83 (3H, s), 4.12-4.16 (2H, m), 6.82-6.86 (1H, m), 7.06 (1H, d), 7.96 (1H, s), 8.08 (1H, d), 8.25-8.33 (2H, m) 步驟6. 5-[5-(2-{1-[(2-氨基-5-溴苯基)氨基]-6-氮雜雙環[3.2.1]辛-6-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A9-6)的製備。在0℃下向5-[5-(2-{1-[(5-溴-2-硝基苯基)氨基]-6-氮雜雙環[3.2.1]辛-6-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(460 mg,0.75 mmol,1當量)和雷尼鎳(128 mg,1.49 mmol,2當量)於甲醇(20 mL)中的攪拌混合物中添加NH 2NH 2 .H 2O(187 mg,3.74 mmol,5當量)。在室溫下攪拌2 h後,將反應混合物通過添加水(100 mL)淬滅,並且用二氯甲烷(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法使用二氯甲烷/甲醇(20/1)洗脫進行純化,以得到呈紅棕色固體的370 mg 5-[5-(2-{1-[(2-氨基-5-溴苯基)氨基]-6-氮雜雙環[3.2.1]辛-6-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A9-6)(84%)。LCMS: m/z (ESI), [M+H] +=586.95。 步驟7. 5-(5-{2-[1-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-6-氮雜雙環[3.2.1]辛-6-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A9-7)的製備。將5-[5-(2-{1-[(2-氨基-5-溴苯基)氨基]-6-氮雜雙環[3.2.1]辛-6-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(340 mg,0.58 mmol,1當量)和BrCN(92 mg,0.87 mmol,1.5當量)於EtOH(5 mL)中的混合物在室溫下攪拌4 h。將反應通過飽和NaHCO 3溶液(100 mL)淬滅,並且用二氯甲烷(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上使用二氯甲烷/甲醇(20/1)洗脫進行純化,以得到呈紅棕色固體的120 mg 5-(5-{2-[1-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-6-氮雜雙環[3.2.1]辛-6-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A9-7)(33%)。LCMS: m/z (ESI), [M+H] +=612.15。 步驟8. 5-(5-{2-[1-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-6-氮雜雙環[3.2.1]辛-6-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A9-8)的製備。在室溫下向5-(5-{2-[1-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-6-氮雜雙環[3.2.1]辛-6-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(120 mg,0.20 mmol,1當量)於THF(4 mL)中的攪拌溶液中添加含LiOH(28 mg,1.18 mmol,6當量)的H 2O(1 mL)。將所得混合物在室溫下攪拌2 h,並且在減壓下濃縮。將殘餘物通過反向快速色譜法使用20%至100%乙腈/水洗脫進行純化,以得到呈白色固體的75 mg 5-(5-{2-[1-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-6-氮雜雙環[3.2.1]辛-6-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A9-8)(63%)。LCMS: m/z (ESI), [M+H] += 598.20。 步驟9. 5-溴-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,27}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33), 18(22),19-八烯-12,16-二酮(INT-A9-9)的製備。在室溫下向5-(5-{2-[1-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-6-氮雜雙環[3.2.1]辛-6-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(70 mg,0.12 mmol,1當量)於二噁烷(2 mL)中的攪拌溶液中添加 N,N-二異丙基乙胺(45 mg,0.35 mmol,3當量)和 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(66 mg,0.17 mmol,1.5當量)。在室溫下攪拌2 h後,將反應用水(80 mL)淬滅,並且將混合物用二氯甲烷(2×80 mL)萃取。將合併的有機層用鹽水(3×80 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-TLC使用二氯甲烷/甲醇(20/1)洗脫進行純化,以得到呈棕色固體的65 mg 5-溴-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,27}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33), 18(22),19-八烯-12,16-二酮(INT-A9-9)(95%)。LCMS: m/z (ESI), [M+H] += 580.00。 步驟10. 15,21-二甲基-5-(嗎啉-4-基)-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,27}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33), 18(22),19-八烯-12,16-二酮(實施例A9)的製備。在氮氣氣氛下在室溫下向5-溴-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,27}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33), 18(22),19-八烯-12,16-二酮(50 mg,0.09 mmol,1當量)、嗎啉(30 mg,0.34 mmol,4當量)和BrettPhos Pd G 3(23 mg,0.03 mmol,0.3當量)於二噁烷(3 mL)中的攪拌混合物中逐滴添加LiHMDS(0.34 mL,0.34 mmol,4當量)。將反應混合物在60℃下攪拌30分鐘。在冷卻到室溫後,將反應用水(50 mL)淬滅,並且將混合物用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-HPLC用XBridge Shield RP18 OBD柱,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3 .H 2O的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的13 mg 15,21-二甲基-5-(嗎啉-4-基)-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,27}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33), 18(22),19-八烯-12,16-二酮(實施例A9)(25%)。LCMS: m/z (ESI), [M+H] +=585.35。 1H NMR (CD 3OD, 400 MHz) δ 1.63-1.69 (1H, m), 1.85-1.92 (2H, m), 2.02-2.08 (1H, m), 2.15 (1H, d), 2.26 (1H, q), 2.60-2.65 (1H, m), 2.82 (1H, d), 3.15-3.23 (4H, m), 3.39 (1H, m), 3.47-3.58 (2H, m), 3.68 (3H, s), 3.85 (3H, s), 3.90 (4H, t), 4.41-4.49 (3H, m), 4.61-4.68 (1H, m), 7.00 (1H, m), 7.25 (1H, d), 7.38 (1H, d), 8.20 (1H, s), 8.49 (1H, s), 9.05 (1H, d)。 實施例A10 1,15,21-三甲基-5-(嗎啉-4-基)-23-氧雜-2,9,11,15, 20,21,26-七氮雜六環[24.3.1.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]三十一-3,5,7,9,13,17(31),18(22),19-八烯-12,16-二酮 步驟1. 3-((5-溴-2-硝基苯基)氨基)-3-甲基哌啶-1-甲酸叔丁酯(INT-A10-1)的製備。將4-溴-2-氟-1-硝基苯(6.16 g,27.99 mmol,3當量)、K 2CO 3(3.8 g,27.99 mmol,3當量)和3-氨基-3-甲基哌啶-1-甲酸叔丁酯(2 g,9.33 mmol,1.00當量)於 N, N-二甲基乙醯胺(40 mL)中的混合物在氮氣氣氛下在100℃下攪拌12 h,然後冷卻到25℃。將反應用水(300 mL)淬滅,並且將混合物用乙酸乙酯(3×200 mL)萃取。將合併的有機層用鹽水(3×300 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-TLC使用石油醚/乙酸乙酯(3/1)洗脫來進行純化,以得到呈黃色油的3.5 g 3-[(5-溴-2-硝基苯基)氨基]-3-甲基哌啶-1-甲酸叔丁酯(INT-A10-1)(91%)。 1H NMR (DMSO-d 6, 400 MHz) δ 1.23 (9H, s), 1.44 (3H, s), 1.54 (2H, s), 1.64 (1H, s), 2.15 (1H, s), 2.78 (1H, s), 2.98 (1H, s), 3.88 (1H, s), 4.16-4.14 (1H, d), 6.86-6.93 (1H, m), 7.30 (1H, d), 8.04-7.98 (1H, d), 8.24 (1H, s)。 步驟2. N-(5-溴-2-硝基苯基)-3-甲基哌啶-3-胺(INT-A10-2)的製備。將三氟乙酸(5 mL,67.31 mmol,7當量)和3-[(5-溴-2-硝基苯基)氨基]-3-甲基哌啶-1-甲酸叔丁酯(3.5 g,8.44 mmol,1當量)於CH 2Cl 2(15 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h,並且在減壓下濃縮。將殘餘物用飽和NaHCO 3(水溶液)(100 mL)處理,並且用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥,並且在減壓下濃縮,以得到呈黃色固體的2.4 g N-(5-溴-2-硝基苯基)-3-甲基哌啶-3-胺(INT-A10-2)(89%)。 1H NMR (DMSO-d 6, 400 MHz) δ 1.51 (3H, s), 1.85-1.83 (3H, m), 2.03-2.0 (1H, t), 2.92-2.89 (1H, m), 3.09-3.05 (1H, d), 3.17-3.14 (1H, m), 3.85-3.82 (1H, d), 6.96-6.95 (1H, m), 7.34-7.31 (1H, d), 8.07-8.05 (1H, d), 8.18 (1H, s) 步驟3. N-(5-溴-2-硝基苯基)-1-(2-((叔丁基二甲基甲矽烷基)氧基)乙基)-3-甲基哌啶-3-胺(INT-A10-3)的製備。在氮氣氣氛下在0℃下向 N-(5-溴-2-硝基苯基)-3-甲基哌啶-3-胺(1 g,3.81 mmol,1當量)、2-[(叔丁基二甲基甲矽烷基)氧基]乙醛(2 g,15.27 mmol,4當量)和三甲基胺(1.5 g,15.27 mmol,4當量)於CH 2Cl 2(50 mL)中的攪拌混合物中添加NaBH(OAc) 3(1.6 g,7.63 mmol,2當量)。將混合物在室溫下攪拌2 h。將反應用水(100 mL)淬滅,並且將混合物用二氯甲烷(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法使用石油醚/乙酸乙酯(5/2)洗脫來進行純化,以得到呈黃色油的840 mg N-(5-溴-2-硝基苯基)-1-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-3-甲基哌啶-3-胺(INT-A10-3)(46%)。 1H NMR (DMSO-d 6, 400 MHz) δ 0.06 (6H, s), 0.86 (9H, s), 1.28-1.25 (1H, td), 1.39 (5H, s), 2.00-2.17 (2H, m), 2.23-2.20 (1H, d), 2.83-2.81 (1H, d), 2.93-2.9 (1H, d), 3.77-3.74(2H, td), 6.82-6.80 (1H, m), 7.22-7.19 (1H, d), 8.01-7.98 (1H, d), 8.78 (1H, s)。 步驟4. 2-(3-((5-溴-2-硝基苯基)氨基)-3-甲基哌啶-1-基)乙-1-醇(INT-A10-4)的製備。將 N-(5-溴-2-硝基苯基)-1-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-3-甲基哌啶-3-胺(700 mg,1.48 mmol,1當量)和四丁基氟化銨(774.7 mg,2.96 mmol,2.0當量)於四氫呋喃(7 mL)中的混合物在氮氣氣氛下在室溫下攪拌4 h。將反應混合物用水(30 mL)處理,並且用二氯甲烷(3×30 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-TLC使用CH 2Cl 2/甲醇(12/1)洗脫來進行純化,以得到呈棕色固體的500 mg 2-{3-[(5-溴-2-硝基苯基)氨基]-3-甲基哌啶-1-基}乙醇(INT-A10-4)(94%)。 1H NMR (DMSO-d 6, 400 MHz) δ 1.19-1.16 (1H, t), 1.29-1.26 (1H, t), 1.41 (4H, s), 1.98-2.13 (3H, m), 2.25-2.23 (1H, d), 2.85-2.83 (2H, d), 3.59-3.57 (2H, t), 4.40-4.36 (1H, t), 6.83-6.80 (1H, d), 7.23-7.21 (1H, d), 8.03-8.01 (1H, d), 8.80 (1H, s)。 步驟5. 5-[5-(2-{3-[(5-溴-2-硝基苯基)氨基]-3-甲基哌啶-1-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A10-5)的製備。在氮氣氣氛下在0℃下向5-(5-羥基-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M2,350 mg,1.33 mmol,1當量)和三苯基膦(1046 mg,3.99 mmol,3當量)於四氫呋喃(10 mL)中的攪拌混合物中添加偶氮二甲酸二異丙酯(806 mg,3.99 mmol,3當量)和2-{3-[(5-溴-2-硝基苯基)氨基]-3-甲基哌啶-1-基}乙醇(523 mg,1.463 mmol,1.1當量)。將混合物攪拌2 h。將反應用水(100 mL)淬滅,並且將混合物用二氯甲烷(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過C18 FLASH使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃色固體的450 mg 5-[5-(2-{3-[(5-溴-2-硝基苯基)氨基]-3-甲基哌啶-1-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A10-5)(56%)。 1H NMR (DMSO-d 6, 400 MHz) δ 1.41 (6H, s), 2.06-2.29 (3H, m), 2.74-2.94 (3H, m), 3.58 (3H, s), 3.69 (3H, s), 3.76 (3H, s), 4.10-4.06 (2H, m), 6.83-6.78 (1H, d), 7.23-7.20 (1H, d), 7.95 (1H, s), 8.01-7.98 (1H, d), 8.11-8.02 (1H, d), 8.44 (1H, d), 8.74 (1H, s)。 步驟6. 5-(5-(2-(3-((2-氨基-5-溴苯基)氨基)-3-甲基哌啶-1-基)乙氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-甲酸甲酯(INT-A10-6)的製備。將5-[5-(2-{3-[(5-溴-2-硝基苯基)氨基]-3-甲基哌啶-1-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A10-5,430 mg,0.71 mmol,1當量)、雷尼鎳(61 mg,0.71 mmol,1當量)和NH 2NH 2 .H 2O(71 mg,1.42 mmol,2當量)於甲醇(6 mL)中的混合物在氮氣氣氛下在0℃下攪拌0.5 h。將所得混合物過濾。將濾餅用甲醇(50 mL)洗滌。將溶液在減壓下濃縮,以得到呈黃色固體的340 mg 5-[5-(2-{3-[(2-氨基-5-溴苯基)氨基]-3-甲基哌啶-1-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A10-6)(83%)。LCMS: m/z (ESI), [M+H] +=573.20。 步驟7. 5-(5-(2-(3-(2-氨基-6-溴-1H-苯並[d]咪唑-1-基)-3-甲基哌啶-1-基)乙氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-甲酸甲酯(INT-A10-7)的製備。將5-[5-(2-{3-[(2-氨基-5-溴苯基)氨基]-3-甲基哌啶-1-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(340 mg,0.59 mmol,1當量)和BrCN(69 mg,0.65 mmol,1.1當量)於EtOH(5 mL)中的混合物在氮氣氣氛下在室溫下攪拌過夜。將所得混合物在減壓下濃縮。將殘餘物通過prep-TLC使用CH 2Cl 2/甲醇(12/1)洗脫進行純化,以得到呈棕色固體的300 mg 5-(5-{2-[3-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-甲基哌啶-1-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A10-7)(84%)。 1H NMR (DMSO-d 6, 400 MHz) δ 0.85-0.84 (1H, m), 0.98-1.09 (1H, m), 1.25-1.23 (4H, m), 1.49-1.47 (1H, m), 1.59 (4H, s), 2.20-2.07 (1H, m), 2.44-2.40 (1H, d), 2.79-2.67 (2H, m), 2.94-2.90 (1H, m), 3.15-3.12 (2H, m), 3.59 (3H, s), 3.71 (3H, s), 3.76 (3H, s), 4.21-4.06 (2H, m), 7.03-7.12 (2H, m), 7.35 (2H, s), 7.50-7.45 (1H, d), 7.95 (1H, s), 8.11-8.05 (1H, d), 8.47-8.45 (1H, d)。 步驟8. 5-(5-(2-(3-(2-氨基-6-溴-1H-苯並[d]咪唑-1-基)-3-甲基哌啶-1-基)乙氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-羧酸(INT-A10-8)的製備。將5-(5-{2-[3-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-甲基哌啶-1-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(290 mg,0.48 mmol,1當量)和LiOH .H 2O(40 mg,0.970 mmol,2當量)於THF/H 2O(2 mL/0.5 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將所得混合物在減壓下濃縮。將殘餘物通過C18FLASH使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈棕色固體的230 mg 5-(5-{2-[3-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-甲基哌啶-1-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A10-8)(81%)。 1H NMR (400 MHz, DMSO-d 6) δ 1.21-1.31 (1H, m), 1.49 (1H, t), 1.58 (4H, s), 2.14 (1H, t), 2.42 (1H, d), 2.70-2.84 (2H, m), 2.87-2.98 (1H, m), 3.13 (1H, d), 3.50 (3H, s), 3.69 (3H, s), 4.04-4.17 (2H, m), 7.08 (2H, d), 7.48 (1H, s), 7.54 (2H, s), 7.88 (1H, s), 7.99 (1H, t), 8.18 (1H, d)。 步驟9. 5-溴-1,15,21-三甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜六環[24.3.1.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]三十一-3,5,7,9,13,17(31),18(22),19-八烯-12,16-二酮(INT-A10-9)的製備。將5-(5-{2-[3-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-甲基哌啶-1-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(230 mg,0.39 mmol,1當量)、 N,N-二異丙基乙胺(152.58 mg,1.182mmol,3當量)和 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(224.44 mg,0.59 mmol,1.5當量)於二噁烷(5 mL)中的混合物在氮氣氣氛下在60℃下攪拌2 h。將混合物冷卻到室溫,並且在減壓下濃縮。將殘餘物通過P\prep-TLC使用二氯甲烷/甲醇(10/1)洗脫進行純化,以得到呈棕色固體的180 mg 5-溴-1,15,21-三甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜六環[24.3.1.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]三十一-3,5,7,9,13,17(31),18(22),19-八烯-12,16-二酮(INT-A10-9)(80%)。LCMS: m/z (ESI), [M+H] +=566.10。 步驟10. 1,15,21-三甲基-5-(嗎啉-4-基)-23-氧雜-2,9,11,15,20,21,26-七氮雜六環[24.3.1.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]三十一-3,5,7,9,13,17(31),18(22),19-八烯-12,16-二酮(實施例A10)的製備。在氮氣氣氛下在室溫下向5-溴-1,15,21-三甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜六環[24.3.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十一-3,5,7,9,13,17(31),18(22),19-八烯-12,16-二酮(90 mg,0.16 mmol,1當量)、BrettPhos Pd G 3(43 mg,0.05 mmol,0.3當量)和嗎啉(55 mg,0.63 mmol,4當量)於二噁烷(4 mL)中的攪拌混合物中添加LiHMDS(0.95 mL,0.95 mmol,6.0當量)。將混合物在60℃下攪拌1 h,冷卻到室溫,用水(30 mL)處理,並且用乙酸乙酯(3×30 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-HPLC用XBridge Shield RP18 OBD柱,使用含有10 mmol/L NH 4HCO 3+ 0.1%NH 3 .H 2O的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的12 mg 1,15,21-三甲基-5-(嗎啉-4-基)-23-氧雜-2,9,11,15,20,21,26-七氮雜六環[24.3.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十一-3,5,7,9,13,17(31),18(22),19-八烯-12,16-二酮(實施例A10)(13%)。LCMS: m/z (ESI), [M+H] +=573.50。 1H NMR (DMSO-d 6, 400 MHz) δ 1.89-1.85 (5H, m), 2.12-2.08 (1H, m), 2.30 (1H, s), 2.72-2.69 (1H, m), 2.92 (1H, d), 3.08-3.06 (3H, m), 3.29 (2H, s), 3.62 (3H, s), 3.72 (2H, s), 3.76-3.74 (3H, m), 4.62 (1H, t), 5.42-5.40 (1H, d), 6.93-6.90 (1H, m), 7.32 (1H, d), 7.48 (1H, d), 8.21-8.37 (1H, m), 8.74 (1H, d), 12.80 (1H, s)。 實施例A11 5,13,27-三甲基-17-(嗎啉-4-基)-7-氧雜-4,5,10,14,21,23,27-七氮雜六環[23.3.1.1^{10,13}.0^{2,6}.0^{14,22}.0^{15, 20}]三十-1(29),2(6),3,15,17,19,21,25-八烯-24,28-二酮 步驟1. 3-[(5-溴-2-硝基苯基)氨基]-3-甲基吡咯烷-1-甲酸叔丁酯(INT-A11-1)的製備。將4-溴-2-氟-1-硝基苯(2 g,9.1 mmol,3當量)、K 2CO 3(1.26 g,9.09 mmol,3當量)和3-氨基-3-甲基吡咯烷-1-甲酸叔丁酯(0.61 g,3.03 mmol,1當量)於乙腈(20 mL)中的混合物在氮氣氣氛下在100℃下攪拌2 h,並且冷卻到室溫。將反應用水(200 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用石油醚/乙酸乙酯(4/1)洗脫來進行純化,以得到呈黃色固體的1.2 g 3-[(5-溴-2-硝基苯基)氨基]-3-甲基吡咯烷-1-甲酸叔丁酯(INT-A11-1)(80%)。LCMS: m/z (ES+), [M+H - 56] +=344.00。 步驟2. N-(5-溴-2-硝基苯基)-3-甲基吡咯烷-3-胺(INT-A11-2)的製備。將3-[(5-溴-2-硝基苯基)氨基]-3-甲基吡咯烷-1-甲酸叔丁酯(2.1 g,5.25 mmol,1當量)於三氟乙酸(4 mL)和二氯甲烷(16 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將混合物在減壓下濃縮,並且用飽和NaHCO 3(水溶液)溶液(100 mL)處理。將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(10/1)洗脫來進行純化,以得到呈黃色固體的1.3 g N-(5-溴-2-硝基苯基)-3-甲基吡咯烷-3-胺(INT-A11-2)(74%)。LCMS: m/z (ES+), [M+H] +=300.20。 步驟3. N-(5-溴-2-硝基苯基)-1-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-3-甲基吡咯烷-3-胺(INT-A11-3)的製備。在氮氣氣氛下在室溫下向 N-(5-溴-2-硝基苯基)-3-甲基吡咯烷-3-胺(1.2 g,4 mmol,1當量)和2-[(叔丁基二甲基甲矽烷基)氧基]乙醛(2.8 g,16 mmol,4當量)於二氯甲烷(10 mL)中的攪拌溶液中添加三甲基胺(1.62 g,16 mmol,4當量)和NaBH(OAc) 3(1.69 g,8 mmol,2當量)。將所得混合物在60℃下攪拌過夜並且冷卻到室溫。將反應用水(10 mL)淬滅,並且將混合物用乙酸乙酯(3×10 mL)萃取。將合併的有機層用鹽水(3×10 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-HPLC用XBridge Shield RP18 OBD柱,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3 .H 2O的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃色油的1.24 g N-(5-溴-2-硝基苯基)-1-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-3-甲基吡咯烷-3-胺(INT-A11-3)(61%)。LCMS: m/z (ES+), [M+H] +=458.50 步驟4. 2-{3-[(5-溴-2-硝基苯基)氨基]-3-甲基吡咯烷-1-基}乙醇(INT-A11-4)的製備。將 N-(5-溴-2-硝基苯基)-1-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-3-甲基吡咯烷-3-胺(1.24 g,2.71 mmol,1當量)和四丁基氟化銨(1.41 g,5.41 mmol,2當量)於四氫呋喃(12 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將反應用飽和NaHCO 3(水溶液)溶液(50 mL)在室溫下淬滅,並且將混合物用二氯甲烷(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(10/1)洗脫來進行純化,以得到呈黃色油的800 mg 2-{3-[(5-溴-2-硝基苯基)氨基]-3-甲基吡咯烷-1-基}乙醇(INT-A11-4)(69%)。LCMS: m/z (ES+), [M+H] += 344.00。 步驟5. 5-[5-(2-{3-[(5-溴-2-硝基苯基)氨基]-3-甲基吡咯烷-1-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A11-5)的製備。在氮氣氣氛下在室溫下向2-{3-[(5-溴-2-硝基苯基)氨基]-3-甲基吡咯烷-1-基}乙醇(800 mg,2.32 mmol,1當量)和三苯基膦(1829 mg,7 mmol,3當量)於四氫呋喃(15 mL)中的攪拌混合物中添加5-(5-羥基-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M2,611 mg,2.32 mmol,1.0當量)和偶氮二甲酸二異丙酯(1410 mg,7 mmol,3當量)。將所得混合物在室溫下攪拌2 h。將反應用水(100 mL)淬滅,並且將混合物用二氯甲烷(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(10/1)洗脫進行純化,以得到呈黃色油的380 mg 5-[5-(2-{3-[(5-溴-2-硝基苯基)氨基]-3-甲基吡咯烷-1-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A11-5)(25%)。LCMS: m/z (ES+), [M+H] +=589.00。 步驟6. 5-[5-(2-{3-[(2-氨基-5-溴苯基)氨基]-3-甲基吡咯烷-1-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A11-6)的製備。在0℃下向5-[5-(2-{3-[(5-溴-2-硝基苯基)氨基]-3-甲基吡咯烷-1-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(380 mg,0.65 mmol,1當量)和雷尼鎳(55.23 mg,0.65 mmol,1當量)於甲醇(6 mL)中的攪拌混合物中逐滴添加NH 2NH 2 .H 2O (129.09 mg,2.58 mmol,4當量)。將所得混合物在室溫下攪拌1.5 h。將反應用水(30 mL)淬滅,並且將混合物用二氯甲烷(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法使用二氯甲烷/甲醇(25/1)洗脫進行純化,以得到呈棕色固體的240 mg 5-[5-(2-{3-[(2-氨基-5-溴苯基)氨基]-3-甲基吡咯烷-1-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A11-6)(67%)。LCMS: m/z (ES+), [M+H] +=559.05。 步驟7. 5-(5-{2-[3-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-甲基吡咯烷-1-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A11-7)的製備。在室溫下向5-[5-(2-{3-[(2-氨基-5-溴苯基)氨基]-3-甲基吡咯烷-1-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(193 mg,0.35 mmol,1當量)於乙醇(10 mL)中的攪拌混合物中分批添加BrCN(40.19 mg,0.38 mmol,1.1當量)。將所得混合物在40℃下攪拌1 h。將混合物冷卻到室溫。將反應用水(30 mL)淬滅,並且將混合物用二氯甲烷(3×30 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(12/1)洗脫進行純化,以得到呈棕色固體的140 mg 5-(5-{2-[3-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-甲基吡咯烷-1-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A11-7)(69%)。LCMS: m/z (ES+), [M+H] +=586.00。 1H NMR (400 MHz, DMSO- d 6) δ 1.24 (1H, s), 1.65 (3H, s), 2.35-2.48 (2H, m), 2.69-2.73 (1H, m), 2.80-2.90 (2H, m), 2.94-2.98 (1H, m), 3.18-3.22 (1H, m), 3.58 (3H, s), 3.75 (6H, d), 4.08-4.15 (2H, m), 6.52 (1H, s), 7.07 (1H, s), 7.43 (1H, s), 7.97 (1H, s), 8.13 (1H, d), 8.44 (1H, d)。 步驟8. 5-(5-{2-[3-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-甲基吡咯烷-1-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A11-8)的製備。將5-(5-{2-[3-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-甲基吡咯烷-1-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(170 mg,0.29 mmol,1當量)和LiOH .H 2O(50 mg,1.16 mmol,4當量)於四氫呋喃(4 mL)和水(1 mL)中的混合物在氮氣氣氛下在室溫下攪拌過夜,並且在減壓下濃縮。將殘餘物通過prep-HPLC用XBridge Shield RP18 OBD柱,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3 .H 2O的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的90 mg 5-(5-{2-[3-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-甲基吡咯烷-1-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A11-8)(54%)。LCMS: m/z (ES+), [M+H] +=572.35。 步驟9. 17-溴-5,13,27-三甲基-7-氧雜-4,5,10,14,21, 23,27-七氮雜六環[23.3.1.1^{10,13}.0^{2,6}.0^{14,22}. 0^{15,20}]三十-1(29),2(6),3,15,17,19,21,25-八烯-24,28-二酮(INT-A11-9)的製備。將5-(5-{2-[3-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-甲基吡咯烷-1-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(90 mg,0.18 mmol,1當量)、 N,N-二異丙基乙胺(45 mg,0.35 mmol,2當量)和 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(133 mg,0.35 mmol,2當量)於二噁烷(3 mL)中的混合物在氮氣氣氛下在室溫下攪拌過夜。將反應用水(30 mL)淬滅,並且將混合物用二氯甲烷(3×30 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(12/1)洗脫進行純化,以得到呈白色固體的70 mg 17-溴-5,13,27-三甲基-7-氧雜-4,5,10,14,21,23,27-七氮雜六環[23.3.1.1^{10, 13}.0^{2,6}.0^{14,22}.0^{15,20}]三十-1(29),2(6),3,15,17, 19,21,25-八烯-24,28-二酮(INT-A11-9)(72%)。LCMS: m/z (ES+), [M+H] +=554.00。 步驟10. 5,13,27-三甲基-17-(嗎啉-4-基)-7-氧雜-4,5, 10,14,21,23,27-七氮雜六環[23.3.1.1^{10,13}.0^{2,6}. 0^{14,22}.0^{15,20}]三十-1(29),2(6),3,15,17,19,21,25-八烯-24,28-二酮(實施例A11)的製備。在氮氣氣氛下在室溫下向17-溴-5,13,27-三甲基-7-氧雜-4,5,10,14,21,23,27-七氮雜六環[23.3.1.1^{10,13}.0^{2,6}.0^{14,22}.0^{15,20}]三十-1(29),2(6),3,15,17,19,21,25-八烯-24,28-二酮(70 mg,0.13 mmol,1當量)、BrettPhos(35 mg,0.065 mmol,0.5當量)和嗎啉(35 mg,0.40 mmol,3.2當量)於二噁烷(5 mL)中的攪拌混合物中逐滴添加LiHMDS(2.5 mL,2.5 mmol,19當量)。將混合物在氮氣氣氛下在60℃下攪拌1 h,然後冷卻到室溫。將反應用水(5 mL)淬滅,並且將混合物用二氯甲烷(3×20 mL)萃取。將合併的有機層用鹽水(3×20 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(10/1)洗脫進行純化,以得到呈灰白色固體的10.8 mg 5,13,27-三甲基-17-(嗎啉-4-基)-7-氧雜-4,5,10,14,21,23,27-七氮雜六環[23.3.1.1^{10,13}. 0^{2,6}.0^{14,22}.0^{15,20}]三十-1(29),2(6),3,15,17,19, 21,25-八烯-24,28-二酮(實施例A11)(15%)。LCMS: m/z (ES+), [M+H] +=559.20。 1H NMR (DMSO- d 6, 400 MHz) δ 1.65 (3H, s), 2.65-2.84 (3H, m), 2.97-3.03 (2H, m), 3.01-3.24 (4H, m), 3.31-3.37 (1H, m), 3.62 (3H, s), 3.75 (3H, s), 3.77-3.83 (5H, m), 4.14-4.20 (1H, m), 4.35-4.40 (1H, m), 5.16 (1H, d), 6.93 (1H, dd), 6.98 (1H, s), 7.45 (1H, d), 8.22 (1H, d), 8.31 (1H, s), 8.89 (1H, d), 12.63 (1H, s)。 實施例A12 15,21,27,27-四甲基-5-(嗎啉-4-基)-23-氧雜-2,9,11,15, 20,21,26-七氮雜六環[24.3.1.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]三十一-3,5,7,9,13,17(31),18(22),19-八烯-12,16-二酮 步驟1. N-(6,6-二甲基哌啶-3-基)氨基甲酸叔丁酯(INT-A12-1)的製備。將 N-(6-氧代哌啶-3-基)氨基甲酸叔丁酯(4 g,18.66 mmol,1當量)於THF(50 mL)中的溶液在氮氣氣氛下在10℃下用四氯化鋯(10.44 g,44.80 mmol,2.4當量)處理0.5 h,隨後在0℃下逐滴添加CH 3MgBr(28.94 g,242.68 mmol,13當量)。將所得混合物在氮氣氣氛下在室溫下攪拌16 h。將反應用水(500 mL)淬滅,並且將混合物用乙酸乙酯(3×400 mL)萃取。將合併的有機層用鹽水(3×400 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法使用二氯甲烷/甲醇(30/1)洗脫來進行純化,以得到呈黃色固體的3.74 g N-(6,6-二甲基哌啶-3-基)氨基甲酸叔丁酯(INT-A12-1)(87%)。LCMS: m/z (ESI), [M+H] +=229.05。 步驟2. N-(1-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-6,6-二甲基哌啶-3-基)氨基甲酸叔丁酯(INT-A12-2)的製備。在室溫下向 N-(6,6-二甲基哌啶-3-基)氨基甲酸叔丁酯(INT-A12-1,234 mg,1.02 mmol,1當量)和K 2CO 3(354 mg,2.56 mmol,2.5當量)於DMSO(10 mL)中的攪拌混合物中逐滴添加(2-溴甲氧基)(叔丁基)二甲基矽烷(294 mg,1.23 mmol,1.2當量)。將反應混合物在60℃下攪拌3 h。冷卻到室溫後,將反應混合物用水(100 mL)處理並用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法使用石油醚/乙酸乙酯(10/1)洗脫來進行純化,以得到呈黃色油的128 mg N-(1-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-6,6-二甲基哌啶-3-基)氨基甲酸叔丁酯(INT-A12-2)(32%)。LCMS: m/z (ESI), [M+H] +=387.15。 步驟3. 2-(5-氨基-2,2-二甲基哌啶-1-基)乙醇(INT-A12-3)的製備。將 N-(1-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-6,6-二甲基哌啶-3-基)氨基甲酸叔丁酯(INT-A12-2,20 g,50 mmol,1當量)和三氟乙酸(60 mL)於二氯甲烷(200 mL)中的溶液在室溫下攪拌2 h。將所得混合物在減壓下濃縮。將呈黃色油的粗產物2-(5-氨基-2,2-二甲基哌啶-1-基)乙醇(INT-A12-3)在不進行進一步純化的情況下直接用於下一步驟。LCMS: m/z (ESI), [M+H] +=173.00。 步驟4. 2-{5-[(5-溴-2-硝基苯基)氨基]-2,2-二甲基哌啶-1-基}乙醇(INT-A12-4)的製備。將2-(5-氨基-2,2-二甲基哌啶-1-基)乙醇(粗產物,INT-A12-3)、4-溴-2-氟-1-硝基苯(10.33 g,60 mmol,1.2當量)和K 2CO 3(20.67 g,52.24 mmol,3當量)於乙腈(150 mL)中的混合物在60℃下攪拌2 h。冷卻到室溫後,將反應用水(800 mL)淬滅,並且將混合物用乙酸乙酯(3×600 mL)萃取。將合併的有機層用鹽水(3×600 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠柱色譜法使用石油醚/乙酸乙酯(5/1)洗脫來進行純化,以得到呈黃色固體的9.3 g 2-{5-[(5-溴-2-硝基苯基)氨基]-2,2-二甲基哌啶-1-基}乙醇(INT-A12-4) (41%)。LCMS: m/z (ESI), [M+H] +=374.05。 1H NMR (DMSO-d 6, 400 MHz) δ 0.99 (3H, s), 1.05 (3H, s), 1.26-1.46 (2H, m), 1.63-1.70 (2H, m), 2.17-2.25 (1H, m), 2.50-2.60 (3H, m), 3.42 (2H, q), 3.86-3.92 (1H, m), 4.32 (1H, t), 6.76-7.82 (1H, m), 7.27 (1H, d), 8.00 (1H, d), 8.60 (1H, d)。 步驟5. 5-[5-(2-{5-[(5-溴-2-硝基苯基)氨基]-2,2-二甲基哌啶-1-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A12-5)的製備。將2-{5-[(5-溴-2-硝基苯基)氨基]-2,2-二甲基哌啶-1-基}乙醇(INT-A12-4,350 mg,0.94 mmol,1當量)、5-(5-羥基-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M2,247 mg,0.94 mmol,1當量)和2-(三丁基-l^[5]-膦亞基)乙腈(1.13 g,4.70 mmol,5當量)於甲苯(8 mL)中的混合物在100℃下攪拌3 h。冷卻到室溫後,將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過反向快速色譜法使用10%-50%乙腈/水洗脫進行純化,以得到呈黃色固體的350 mg 5-[5-(2-{5-[(5-溴-2-硝基苯基)氨基]-2,2-二甲基哌啶-1-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A12-5)(60%)。LCMS: m/z (ESI), [M+H] +=617.20。 步驟6. 5-[5-(2-{5-[(2-氨基-5-溴苯基)氨基]-2,2-二甲基哌啶-1-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A12-6)的製備。將5-[5-(2-{5-[(5-溴-2-硝基苯基)氨基]-2,2-二甲基哌啶-1-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A12-5,400 mg,0.64 mmol,1當量)、雷尼鎳(5 mg,0.15 mmol,0.5當量)和肼(62 mg,1.94 mmol,3當量)於甲醇(10 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-TLC使用二氯甲烷/甲醇(20/1)洗脫進行純化,以得到呈黃色固體的350 mg 5-[5-(2-{5-[(2-氨基-5-溴苯基)氨基]-2,2-二甲基哌啶-1-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A12-6)(91%)。LCMS: m/z (ESI), [M+H] +=589.35。 步驟7. 5-(5-{2-[5-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-2,2-二甲基哌啶-1-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A12-7)的製備。將5-[5-(2-{5-[(2-氨基-5-溴苯基)氨基]-2,2-二甲基哌啶-1-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A12-6,350 mg,0.59 mmol,1當量)和溴化氰(69 mg,0.65 mmol,1.1當量)於EtOH(10 mL)中的混合物在氮氣氣氛下在室溫下攪拌3 h。將反應混合物用飽和NaHCO 3溶液(100 mL)處理,並且用乙酸乙酯(3×100 mL)萃取。將合併的有機層經無水Na 2SO 4乾燥,並且在減壓下濃縮。將殘餘物通過prep-TLC使用二氯甲烷/甲醇(20/1)洗脫進行純化,以得到呈黃色固體的350 mg 5-(5-{2-[5-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-2,2-二甲基哌啶-1-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A12-7)(95%)。LCMS: m/z (ESI), [M+H] +=614.05。 步驟8. 5-(5-{2-[5-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-2,2-二甲基哌啶-1-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A12-8)的製備。向5-(5-{2-[5-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-2,2-二甲基哌啶-1-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A12-7,240 mg,0.39 mmol,1當量)於四氫呋喃(4 mL)中的攪拌溶液中添加含氫氧化鋰(28 mg,1.17 mmol,3當量)的H 2O(1 mL)。將所得混合物在室溫下攪拌2 h。將反應混合物在減壓下濃縮,並且將殘餘物通過反向快速色譜法使用10%至40%乙腈/水洗脫進行純化,以得到呈白色固體的190 mg 5-(5-{2-[5-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-2,2-二甲基哌啶-1-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A12-8)(81%)。LCMS: m/z (ESI), [M+H] +=598.30。 步驟9. 5-溴-15,21,27,27-四甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜六環[24.3.1.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]三十一-3,5,7,9,13,17(31),18(22),19-八烯-12,16-二酮(INT-A12-9)的製備。將5-(5-{2-[5-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-2,2-二甲基哌啶-1-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A12-8,190 mg,0.31 mmol,1當量)、 N,N-二異丙基乙胺(82 mg,0.63 mmol,2當量)和 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(181 mg,0.47 mmol,1.5當量)於二噁烷(5 mL)中的溶液在氮氣氣氛下在室溫下攪拌2 h。將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-TLC使用二氯甲烷/甲醇(20/1)洗脫進行純化,以得到以得到呈白色固體的100 mg 5-溴-15,21,27,27-四甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜六環[24.3.1.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]三十一-3,5,7,9,13,17(31),18(22),19-八烯-12,16-二酮(INT-A12-9)(27%)。LCMS: m/z (ESI), [M+H] +=580.35。 步驟10. 15,21,27,27-四甲基-5-(嗎啉-4-基)-23-氧雜-2,9,11,15,20,21,26-七氮雜六環[24.3.1.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]三十一-3,5,7,9,13,17(31),18(22),19-八烯-12,16-二酮(實施例A12)的製備。在氮氣氣氛下在室溫下向5-溴-15,21,27,27-四甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜六環[24.3.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十一-3,5,7,9,13,17(31),18(22),19-八烯-12,16-二酮(INT-A12-9,60 mg,0.10 mmol,1當量)、嗎啉(36 mg,0.41 mmol,4當量)和BrettPhos Pd G 3(46 mg,0.05 mmol,0.5當量)於二噁烷(3 mL)中的攪拌混合物中逐滴添加LiHMDS(0.5 mL,0.51 mmol,5當量)。將反應混合物在氮氣氣氛下在60℃下攪拌1 h。在冷卻到室溫後,將反應用飽和NH 4Cl溶液(50 mL)淬滅,並且將混合物用二氯甲烷(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-TLC使用二氯甲烷/甲醇(18/1)洗脫進行純化。將產物通過prep-HPLC用XBridge Shield RP18 OBD柱,使用含有10 mmol/L NH 4HCO和0.1% NH 3 .H 2O的水和乙腈作為流動相進行進一步純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的8.9 mg 515,21,27,27-四甲基-5-(嗎啉-4-基)-23-氧雜-2,9,11,15,20,21,26-七氮雜六環[24.3.1.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十一-3,5,7,9,13,17(31), 18(22),19-八烯-12,16-二酮(實施例A12)(14%)。LCMS: m/z (ESI), [M+H] +=587.40。 1H NMR (DMSO-d 6, 400 MHz) δ 1.12 (3H, s), 1.20-1.28 (1H, m), 1.30 (3H, s), 1.61-1.85 (2H, m), 2.17-2.29 (2H, m), 2.42 (1H, t), 3.05-3.18 (4H, m), 3.39-3.60 (3H, m), 3.62 (3H, s), 3.69 (3H, s), 3.78 (4H, t), 4.48-4.69 (2H, m), 6.87-6.94 (1H, m), 7.18 (1H, s), 7.43 (1H, d), 8.29 (1H, s), 8.37 (1H, s), 8.80 (1H, d), 11.81-12.75 (1H, m) 實施例A13 1,15,21-三甲基-5-(嗎啉-4-基)-23-氧雜-2,9,11,15,20, 21,26七氮雜六環[24.4.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18, 22}]三十二-3,5,7,9,13,17(32),18(22),19-八烯-12,16-二酮 步驟1. 1-苄基-3-甲基氮雜環庚-3-醇(INT-A13-1)的製備。在氮氣氣氛下在-78℃下向1-苄基氮雜環庚-3-酮(3.0 g,14.75 mmol,1當量)於四氫呋喃(30 mL)中的攪拌混合物中逐滴添加MeMgBr(10.5 g,88.54 mmol,6當量)。將混合物在60℃下攪拌2小時。將反應用飽和NH 4Cl(水溶液)溶液(300 mL)淬滅,並且將混合物用二氯甲烷(3×300 mL)萃取。將合併的有機層用鹽水(3×300 mL)洗滌,經無水Na 2SO 4乾燥,並且在減壓下濃縮,以得到呈棕色油的3.1 g 1-苄基-3-甲基氮雜環庚-3-醇(INT-A13-1)。此材料在不經純化的情況下用於下一反應中。LCMS: m/z (ESI), [M+H] +=220.05。 步驟2. N-(1-苄基-3-甲基氮雜環庚-3-基)乙醯胺(INT-A13-2)的製備。在氮氣氣氛下在0℃下向1-苄基-3-甲基氮雜環庚-3-醇(3.1 g,約14 mmol,約1當量)於乙腈(30 mL)中的攪拌混合物中逐滴添加煙化的H 2SO 4(2.5 g,14.13 mmol,1當量)。將混合物攪拌2 h。在室溫下將反應用水(300 mL)淬滅。將混合物用K 2CO 3調節到pH 9,並且用二氯甲烷(3×300 mL)萃取。將合併的有機層用鹽水(3×300 mL)洗滌,經無水Na 2SO 4乾燥,並且在減壓下濃縮,以得到呈棕色固體的1.1 g N-(1-苄基-3-甲基氮雜環庚-3-基)乙醯胺(INT-A13-2)。 1H NMR (400 MHz, DMSO-d 6) δ 1.18 (3H, s), 1.35-1.67 (5H, m), 1.72 (3H, s), 1.90-2.00 (1H, m), 2.61 (1H, d), 2.87 (1H, d), 3.61 (2H, d), 7.20-7.37 (5H, m)。 步驟3. N-(3-甲基氮雜環庚-3-基)乙醯胺(INT-A13-3)的製備。將 N-(1-苄基-3-甲基氮雜環庚-3-基)乙醯胺(INT-A13-2,1.1 g,4.22 mmol,1當量)和Pd/C(0.4 g,4.22 mmol,1當量)於甲醇(10 mL)中的混合物在氮氣氣氛下在室溫下攪拌過夜。將混合物過濾。將濾餅用甲醇(3×100 mL)洗滌。將所得溶液在減壓下濃縮。將粗產物在不進行進一步純化的情況下直接用於下一步驟。 1H NMR (400 MHz, DMSO-d 6) δ 1.20 (3H, s), 1.32-1.58 (5H, m), 1.78 (3H, s), 1.90-2.10 (1H, m), 2.61 (1H, d), 2.65-2.77 (2H, m), 2.85 (1H, d), 7.23 (1H, s)。 步驟4. N-(1-(2-((叔丁基二甲基甲矽烷基)氧基)乙基)-3-甲基氮雜環庚-3-基)乙醯胺(INT-A13-4)的製備。向 N-(3-甲基氮雜環庚-3-基)乙醯胺(400 mg,約2.3 mmol,1當量)和(2-溴乙氧基)(叔丁基)二甲基矽烷(1124 mg,4.69 mmol,2當量)於 N,N-二甲基甲醯胺(8 mL)中的攪拌混合物中添加K 2CO 3(649.4 mg,4.69 mmol,2當量)和NaI(704 mg,4.69 mmol,2當量)。將混合物在60℃下攪拌2 h,然後冷卻到室溫。將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過C18FLASH使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈棕色油的750 mg N-(1-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-3-甲基氮雜環庚-3-基)乙醯胺(INT-A13-4)(96%)。 1H NMR (400 MHz, DMSO-d 6) δ 0.01 (6H, s), 0.83 (9H, s), 1.19 (3H, s), 1.28-1.62 (6H, m), 1.73 (3H, s), 1.86 (1H, d), 2.51-2.64 (5H, m), 2.72-2.80 (1H, m), 3.55-3.63 (2H, m)。 步驟5. 2-(3-氨基-3-甲基氮雜環庚-1-基)乙醇(INT-A13-5)的製備。將 N-(1-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-3-甲基氮雜環庚-3-基)乙醯胺(1.4 g,4.26 mmol,1當量)和HCl(6 M)(140 mL,460.77 mmol,108當量)的混合物在氮氣氣氛下在100℃下攪拌過夜。將所得混合物在減壓下濃縮,以得到呈棕色油的500 mg 2-(3-氨基-3-甲基氮雜環庚-1-基)乙醇(INT-A13-5)。 1H NMR (400 MHz, DMSO-d 6) δ 1.11-1.39 (3H, m), 1.48 (2H, d), 1.70 (1H, d), 1.76-2.08 (6H, m), 2.19 (1H, t), 3.18 (2H, t), 3.34-3.52 (1H, m), 3.52-3.95 (6H, m), 8.64 (2H, s)。 步驟6. 2-(3-((5-溴-2-硝基苯基)氨基)-3-甲基氮雜環庚-1-基)乙醇(INT-A13-6)的製備。向2-(3-氨基-3-甲基氮雜環庚-1-基)乙醇(460 mg,2.67 mmol,1當量)和4-溴-2-氟-1-硝基苯(1174 mg,5.34 mmol,2當量)於乙腈(5 mL)中的攪拌混合物中分批添加K 2CO 3(738 mg,5.34 mmol,2當量)。將混合物在80℃下攪拌過夜,然後冷卻到室溫。將反應用水(200 mL)淬滅,並且將混合物用乙酸乙酯(3×200 mL)萃取。將合併的有機層用鹽水(3×200 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法使用石油醚/乙酸乙酯(1/10)洗脫來進行純化,以得到呈黃色油的530 mg 2-{3-[(5-溴-2-硝基苯基)氨基]-3-甲基氮雜環庚-1-基}乙醇(INT-A13-6)(53%)。 1H NMR (400 MHz, DMSO-d 6) δ 1.31-1.63 (7H, m), 1.64-1.77 (1H, m), 2.21 (1H, d), 2.58-2.86 (6H, m), 3.60 (2H, d), 4.44 (1H, t), 6.81 (1H, d), 7.41 (1H, d), 8.01 (1H, d), 9.07 (1H, s)。 步驟7. 5-[5-(2-{3-[(5-溴-2-硝基苯基)氨基]-3-甲基氮雜環庚-1-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A13-7)的製備。在氮氣氣氛下在0℃下向5-(5-羥基-1-甲基吡唑-4-基)-1-甲基-6氧代吡啶-3-甲酸甲酯(M2,405 mg,1.53 mmol,1當量)和三苯基膦(1210 mg,4.614 mmol,3當量)於四氫呋喃(5 mL)中的攪拌混合物中添加偶氮二甲酸二異丙酯(933 mg,4.61 mmol,3當量)和2-{3-[(5-溴-2-硝基苯基)氨基]-3-甲基氮雜環庚-1-基}乙醇(629 mg,1.69 mmol,1.1當量)。將混合物在室溫下攪拌2 h。將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用CH 2Cl 2/甲醇(10/1)洗脫進行純化,以得到呈黃色油的550 mg 5-[5-(2-{3-[(5-溴-2-硝基苯基)氨基]-3-甲基氮雜環庚-1-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A13-7)(57%)。 1H NMR (400 MHz, DMSO-d 6) δ 1.37 (3H, s), 1.44-1.50 (1H, m), 1.65-1.75 (1H, m), 2.17-2.25 (1H, m), 2.63-2.77 (2H, m), 2.83 (1H, s), 2.91 (1H, d), 3.10 (1H, d), 3.10-3.20 (1H, m), 3.33 (2H, s), 3.71 (6H, d), 4.06-4.22 (2H, m), 5.76 (1H, s), 6.80 (1H, d), 7.20-7.30 (2H, m), 7.33-7.50 (5H, m), 7.94 (1H, s), 7.98 (1H, d), 8.07 (1H, d), 8.40 (1H, d), 9.01 (1H, s)。 步驟8. 5-(5-(2-(3-((2-氨基-5-溴苯基)氨基)-3-甲基氮雜環庚-1-基)乙氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-甲酸甲酯(INT-A13-8)的製備。將5-[5-(2-{3-[(5-溴-2-硝基苯基)氨基]-3-甲基氮雜環庚-1-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(540 mg,0.87 mmol,1當量)、雷尼鎳(74 mg,0.87 mmol,1當量)和NH 2NH 2 .H 2O(44 mg,0.88 mmol,1當量)於甲醇(2 mL)中的攪拌混合物在氮氣氣氛下在0℃下攪拌1 h。將所得混合物過濾。將濾餅用甲醇(50 mL)洗滌。將所得溶液在減壓下濃縮,以得到呈棕色油的300 mg 5-[5-(2-{3-[(2-氨基-5-溴苯基)氨基]-3-甲基氮雜環庚-1-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸酯(INT-A13-8)。此材料在不經純化的情況下用於下一反應中。LCMS: m/z (ESI), [M+H]+=589.20。 步驟9. 5-(5-{2-[3-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-甲基氮雜環庚-1-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A13-9)的製備。將5-[5-(2-{3-[(2-氨基-5-溴苯基)氨基]-3-甲基氮雜環庚-1-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(280 mg,0.47 mmol,1當量)和BrCN(50 mg,0.47 mmol,1當量)於EtOH(4 mL)中的混合物在氮氣氣氛下在60 0℃下攪拌2 h。將混合物冷卻到室溫,並且在減壓下濃縮。將殘餘物通過Prep-TLC使用CH 2Cl 2/甲醇(10/1)洗脫進行純化,以得到呈棕色油的210 mg 5-(5-{2-[3-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-甲基氮雜環庚-1-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A13-9)(71%)。 1H NMR (400 MHz, DMSO-d 6) δ 1.19-1.23 (1H, m), 1.43-1.49 (1H, m), 1.60-1.64 (2H, m), 1.73-1.90 (5H, m), 2.65-2.81 (1H, m), 2.83-2.90 (1H, m), 2.90-3.05 (2H, m), 3.17 (3H, d), 3.55 (3H, s), 3.66 (3H, s), 3.74 (3H, s), 4.00-4.18 (3H, m), 6.95-7.11 (2H, m), 7.54 (1H, d), 7.70 (2H, s), 7.94 (1H, s), 8.06 (1H, d), 8.37 (1H, d)。 步驟10. 5-(5-{2-[3-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-甲基氮雜環庚-1-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A13-10)的製備。將5-(5-{2-[3-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-甲基氮雜環庚-1-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(200 mg,0.32 mmol,1當量)和LiOH .H 2O(27 mg,0.65 mmol,2當量)於四氫呋喃/H 2O(4 ml/1 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將混合物在減壓下濃縮。將殘餘物通過C18FLASH使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈棕色固體的150 mg 5-(5-{2-[3-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-甲基氮雜環庚-1-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A13-10)(76%)。 1H NMR (400 MHz, DMSO-d 6) δ 1.43-1.68 (2H, m), 1.81 (3H, d), 2.70-2.74 (1H, m), 2.82-3.02 (2H, m), 3.06-3.12 (1H, m), 3.38-3.44 (4H, m), 3.62 (2H, s), 3.97-4.10 (1H, m), 6.94-7.08 (1H, m), 7.54 (1H, d), 7.82 (1H, s), 7.88 (1H, s), 8.02 (1H, s), 8.16 (1H, d)。 步驟11. 5-溴-1,15,21-三甲基-23-氧雜-2,9,11,15,20, 21,26-七氮雜六環[24.4.1.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]三十二-3,5,7,9,13,17(32),18(22),19-八烯-12,16-二酮(INT-A13-11)的製備。在氮氣氣氛下向5-(5-{2-[3-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-甲基氮雜環庚-1-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(140 mg,0.23 mmol,1當量)和N,N,N,N-四甲基-O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(133 mg,0.35 mmol,1.5當量)於二噁烷(4 mL)中的攪拌混合物中添加 N,N-二異丙基乙胺(90 mg,0.70 mmol,3.0當量)。將所得混合物在60℃下攪拌2 h,冷卻到室溫,並且在減壓下濃縮。將殘餘物用水(30 mL)處理,並且將混合物用二氯甲烷(3×30 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用CH 2Cl 2/甲醇(12/1)洗脫進行純化,以得到呈棕色固體的100 mg 5-溴-1,15,21-三甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜六環[24.4.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十二-3,5,7,9,13,17(32),18(22),19-八烯-12,16-二酮(INT-A13-11)(73%)。 1H NMR (400 MHz, DMSO-d 6) δ 1.23-1.29 (7H, m), 1.68-1.72 (2H, m), 1.78-1.86 (5H, m), 1.87-1.91 (1H, m), 2.02-2.13 (1H, m), 2.69 (3H, s), 3.10-3.21 (3H, m), 3.23 (1H, t), 3.41 (1H, d), 3.60-3.66 (4H, m), 3.72 (3H, s), 4.30-4.47 (3H, m), 7.35-7.44 (1H, m), 7.57 (1H, d), 7.69 (1H, d), 8.10 (1H, s), 8.30 (1H, d), 8.62 (1H, d), 13.06 (1H, s)。 步驟12. 1,15,21-三甲基-5-(嗎啉-4-基)-23-氧雜-2,9, 11,15,20,21,26七氮雜六環[24.4.1.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]三十二-3,5,7,9,13,17(32),18(22),19-八烯-12,16-二酮(實施例A13)的製備。向5-溴-1,15,21-三甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜六環[24.4.1.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十二-3,5,7,9,13,17(32), 18(22),19-八烯-12,16-二酮(90 mg,0.15 mmol,1當量)和BrettPhos Pd G 3(42 mg,0.046 mmol,0.3當量)於1,4-二噁烷(3 mL)中的攪拌混合物中添加嗎啉(54 mg,0.62 mmol,4當量)和LiHMDS(0.93 mL,0.93 mmol,6當量)。將混合物在60℃下攪拌1 h,然後冷卻到室溫。將反應用水(30 mL)淬滅,並且將混合物用乙酸乙酯(3×30 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用CH 2Cl 2/甲醇(25/1)洗脫進行純化,以得到呈白色固體的25.8 mg 1,15,21-三甲基-5-(嗎啉-4-基)-23-氧雜-2,9,11,15,20,21,26七氮雜六環[24.4.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十二-3,5,7,9,13,17(32),18(22),19-八烯-12,16-二酮(A13)(28%)。LCMS: m/z (ESI), [M+H] +=587.40。 1H NMR (400 MHz, DMSO-d 6) δ 1.60-1.75 (3H, m) 1.80-1.88 (4H, s), 1.96-2.06 (1H, m), 2.57-2.67 (1H, m), 2.85-2.93 (1H, m), 2.95-3.09 (3H, m), 3.06-3.15 (2H, m), 3.21-3.29 (2H, m), 3.61 (3H, s), 3.70-3.80 (8H, m), 4.32 (1H, d), 4.39 (1H, t), 4.53 (1H, d), 6.94 (1H, d), 7.04 (1H, d), 7.49 (1H, d), 8.09 (1H, s), 8.26 (1H, d), 8.59 (1H, d), 12.85 (1H, s)。 實施例A14 (11 R)-26-乙基-5,11-二甲基-16-(4-甲基哌嗪-1-基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮 步驟1. 5-溴-1-乙基-6-氧代-1,6-二氫吡啶-3-甲酸甲酯(INT-A14-1)的製備。在0℃下向5-溴-6-羥基吡啶-3-甲酸甲酯(10 g,43.09 mmol,1當量)於 N,N-二甲基甲醯胺(200 mL)中的攪拌混合物中添加NaH(3.45 g,86.19 mmol,2當量,60%)。在添加乙基碘(10.08 g, 64.64 mmol,1.5當量)之前,將反應混合物在60℃下攪拌30分鐘。將混合物在室溫下另外攪拌2 h,用水(1 L)處理,並且用乙酸乙酯(3×1 L)萃取。將合併的有機層用鹽水(3×1 L)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法使用石油醚/乙酸乙酯(3/1)洗脫進行純化,以得到呈黃色固體的5 g 5-溴-1-乙基-6-氧代吡啶-3-甲酸甲酯(INT-A14-1)(37%)。LCMS: m/z (ESI), [M+H] +=261.90。 1H NMR (400 MHz, DMSO-d 6) δ 1.25 (3H, t), 3.81 (3H, s), 4.08 (2H, q), 8.17 (1H, d), 8.61 (1H, d) 步驟2. 甲基-1-乙基-5-(2-甲基-3-氧代-1-((2-(三甲基甲矽烷基)乙氧基)甲基)-2,3-二氫-1H-吡唑-4-基)-6-氧代-1,6-二氫吡啶-3-甲酸酯(INT-A14-2)的製備。將5-溴-1-乙基-6-氧代吡啶-3-甲酸甲酯(INT-A14-1,2.0 g,7.69 mmol,1當量)、4,4,5,5-四甲基-2-(四甲基-1,3,2-二氧雜環戊硼烷-2-基)-1,3,2-二氧雜環戊硼烷(2.93 g,11.53 mmol,1.5當量)、Pd(dppf)Cl 2(0.56 g,0.77 mmol,0.1當量)和KOAc(2.26 g,23.07 mmol,3.0當量)於二噁烷(25 mL)中的混合物在氮氣氣氛下在60℃下攪拌1 h。將混合物冷卻到室溫,隨後添加4-碘-2-甲基-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-3-酮(2.72 g,7.69 mmol,1.0當量)、Pd(dppf)Cl 2(1.13 g,1.54 mmol,0.2當量)、K 2CO 3(3.19 g,23.07 mmol,3.0當量)和H 2O(8 mL)。將反應混合物在80℃下在氮氣氣氛下攪拌2 h。在冷卻到室溫後,將反應用水(300 mL)淬滅,並且將混合物用乙酸乙酯(3×300 mL)萃取。將合併的有機層用鹽水(3×300 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法使用二氯甲烷/甲醇(12/1)洗脫進行純化,以得到呈棕色固體的1 g 1-乙基-5-(2-甲基-3-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-4-基)-6-氧代吡啶-3-甲酸甲酯(INT-A14-2)(31%)。LCMS: m/z (ESI), [M+H] +=408.15。 1H NMR (400 MHz, DMSO-d 6) δ 0.05 (9H, s), 0.78-0.86 (2H, m), 1.27 (3H, t), 3.37 (3H, s), 3.43-3.50 (2H, m), 3.82 (3H, s), 4.08-4.12 (2H, m), 5.32 (2H, s), 8.41 (1H, d), 8.87 (1H, s), 9.20 (1H, d)。 步驟3. 1-乙基-5-(5-羥基-1-甲基-1H-吡唑-4-基)-6-氧代-1,6-二氫吡啶-3-甲酸甲酯(INT-A14-3)的製備。將1-乙基-5-(2-甲基-3-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-4-基)-6-氧代吡啶-3-甲酸甲酯(1.0 g,2.45 mmol,1當量)和HCl(10 mL,1,4-二噁烷中4 M)的混合物在室溫下攪拌過夜。將混合物在真空下濃縮。將殘餘物用CH 2Cl 2(20 mL)和飽和NaHCO 3(水溶液)(50 mL)處理。用二氯甲烷(3×100 mL)萃取所得混合物。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過反向快速色譜法使用0%至100%乙腈/水洗脫進行純化,以得到呈黃色固體的520 mg 1-乙基-5-(5-羥基-1-甲基吡唑-4-基)-6-氧代吡啶-3-甲酸甲酯(INT-A14-3) (76%)。LCMS: m/z (ESI), [M+H] +=278.05。 1H NMR (400 MHz, DMSO-d 6) δ 1.29 (3H, t), 3.33 (3H, s), 3.84 (3H, s), 4.13-4.17 (2H, m), 7.96 (1H, s), 8.45 (2H, d), 12.72-13.41 (1H, br s)。 步驟4. 甲基-( R)-5-(5-((5-((5-溴-2-硝基苯基)氨基)-4-甲基戊基)氧基)-1-甲基-1H-吡唑-4-基)-1-乙基-6-氧代-1,6-二氫吡啶-3-甲酸酯(INT-A14-4)的製備。在氮氣氣氛下在0℃下向1-乙基-5-(5-羥基-1-甲基吡唑-4-基)-6-氧代吡啶-3-甲酸甲酯(500 mg,1.80 mmol,1當量)和三苯基膦(1.41 g,5.41 mmol,3當量)於四氫呋喃(8 mL)中的攪拌混合物中添加偶氮二甲酸二異丙酯(1.09 g,5.41 mmol,3當量)和(4 R)-5-[(5-溴-2-硝基苯基)氨基]-4-甲基戊-1-醇(M3-7,1.14 g,3.60 mmol,2.0當量)。將所得混合物在0℃下攪拌2 h。將反應用水(100 mL)淬滅,並且將混合物用二氯甲烷(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過反向快速色譜法使用0%至100%乙腈/水洗脫進行純化,以得到呈黃色固體的370 mg 甲基-( R)-5-(5-((5-((5-溴-2-硝基苯基)氨基)-4-甲基戊基)氧基)-1-甲基-1H-吡唑-4-基)-1-乙基-6-氧代-1,6-二氫吡啶-3-甲酸酯(INT-A14-4)(35%)。LCMS: m/z (ESI), [M+H] +=577.90。 1H NMR (DMSO-d 6, 400 MHz) δ 0.96 (3H, d), 1.27 (3H, t), 1.29-1.45 (1H, m), 1.58-1.64 (1H, m), 1.69-1.96 (3H, m), 3.16-3.27 (1H, m), 3.27-3.38 (1H, m), 3.68 (3H, s), 3.78 (3H, s), 3.96 (2H, t), 4.08 (2H, q), 6.80-6.86 (1H, m), 7.26 (1H, d), 7.95-8.02 (2H, m), 8.13 (1H, d), 8.21 (1H, t), 8.43 (1H, d) 步驟5. ( R)-5-(5-((5-((2-氨基-5-溴苯基)氨基)-4-甲基戊基)氧基)-1-甲基-1H-吡唑-4-基)-1-乙基-6-氧代-1,6-二氫吡啶-3-甲酸甲酯(INT-A14-5)的製備。在氮氣氣氛下在0℃下向雷尼鎳(53 mg,0.62 mmol,1當量)和甲基-( R)-5-(5-((5-((5-溴-2-硝基苯基)氨基)-4-甲基戊基)氧基)-1-甲基-1H-吡唑-4-基)-1-乙基-6-氧代-1,6-二氫吡啶-3-甲酸酯(INT-A14-4,360 mg,0.62 mmol,1當量)於甲醇(5 mL)中的攪拌混合物中添加NH 2NH 2 .H 2O(62 mg,1.25 mmol,2當量)。在60℃下攪拌1 h後,將所得混合物過濾,並且將有機溶液在減壓下濃縮,以得到呈棕色固體的350 mg ( R)-5-(5-((5-((2-氨基-5-溴苯基)氨基)-4-甲基戊基)氧基)-1-甲基-1H-吡唑-4-基)-1-乙基-6-氧代-1,6-二氫吡啶-3-甲酸甲酯(INT-A14-5)。此材料在不經純化的情況下用於直接下一反應中。 步驟6. 甲基-( R)-5-(5-((5-(2-氨基-6-溴-1H-苯並[d]咪唑-1-基)-4-甲基戊基)氧基)-1-甲基-1H-吡唑-4-基)-1-乙基-6-氧代-1,6-二氫吡啶-3-甲酸甲酯(INT-A14-6)的製備。在氮氣氣氛下在室溫下向新鮮製備的( R)-5-(5-((5-((2-氨基-5-溴苯基)氨基)-4-甲基戊基)氧基)-1-甲基-1H-吡唑-4-基)-1-乙基-6-氧代-1,6-二氫吡啶-3-甲酸酯(INT-A14-5,350 mg)於醇(5 mL)中的溶液中添加BrCN(83 mg,0.79 mmol,1.5當量)。在氮氣氣氛下在60℃下攪拌1 h後,將混合物在真空下濃縮。將殘餘物通過prep-TLC使用石油醚/乙酸乙酯(5/1)洗脫進行純化,以得到呈棕色固體的260 mg 甲基-( R)-5-(5-((5-(2-氨基-6-溴-1H-苯並[d]咪唑-1-基)-4-甲基戊基)氧基)-1-甲基-1H-吡唑-4-基)-1-乙基-6-氧代-1,6-二氫吡啶-3-甲酸酯(INT-A14-6)(69%)。LCMS: m/z (ESI), [M+H] +=573.25。 11H NMR (DMSO-d 6, 400 MHz) δ 0.83 (3H, d), 1.26 (3H, t), 1.28-1.40 (1H, m), 1.49-1.53 (1H, m), 1.67-1.71 (1H, m), 1.78-1.94 (1H, m), 1.96-2.02 (1H, m), 3.61 (3H, s), 3.79 (3H, s), 3.80-3.97 (4H, m), 4.08 (2H, q), 6.56 (2H, s), 7.04 (2H, d), 7.36 (1H, d), 7.99 (1H, s), 8.13 (1H, d), 8.45 (1H, d)。 步驟7. 5-(5-{[(4 R)-4-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]戊基]氧基}-1-甲基吡唑-4-基)-1-乙基-6-氧代吡啶-3-羧酸(INT-A14-7)的製備。在室溫下向5-(5-{[(4 R)-4-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]戊基]氧基}-1-甲基吡唑-4-基)-1-乙基-6-氧代吡啶-3-甲酸甲酯(260 mg,0.45 mmol,1當量)於THF(2 mL)中的攪拌混合物中添加含LiOH .H 2O(38 mg,0.91 mmol,2.0當量)的H 2O(0.5 mL)。在氮氣氣氛下在60℃下攪拌30分鐘後。將混合物冷卻到室溫,並且在真空下濃縮。將殘餘物通過反向快速色譜法使用0%至20%乙腈/水洗脫進行純化,以得到呈白色固體的160 mg 5-(5-{[(4 R)-4-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]戊基]氧基}-1-甲基吡唑-4-基)-1-乙基-6-氧代吡啶-3-羧酸(INT-A14-7)(63%)。LCMS: m/z (ESI), [M+H] += 559.15。 步驟8. (11 R)-16-溴-26-乙基-5,11-二甲基-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(INT-A14-8)的製備在氮氣氣氛下在室溫下向5-(5-{[(4 R)-4-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]戊基]氧基}-1-甲基吡唑-4-基)-1-乙基-6-氧代吡啶-3-羧酸(160 mg,0.28 mmol,1當量)和 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(163 mg,0.43 mmol,1.5當量)於二噁烷(2 mL)中的攪拌混合物中添加 N,N-d二異丙基乙胺(111 mg,0.86 mmol,3.0當量)。將反應混合物在室溫下攪拌1 h。將反應用水(50 mL)淬滅,並且將混合物用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-TLC使用二氯甲烷/甲醇(12/1)洗脫進行純化,以得到呈棕色固體的110 mg (11 R)-16-溴-26-乙基-5,11-二甲基-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(INT-A14-8)(71%)。LCMS: m/z (ESI), [M+H] += 541.05。 步驟9. (11 R)-26-乙基-5,11-二甲基-16-(4-甲基哌嗪-1-基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(實施例A14)的製備。在氮氣氣氛下向(11 R)-16-溴-26-乙基-5,11-二甲基-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(120 mg,0.22 mmol,1當量)、1-甲基哌嗪(89 mg,0.89 mmol,4當量)和BrettPhos Pd G 3(60 mg,0.07 mmol,0.3當量)於二噁烷(5 mL)中的攪拌混合物中逐滴添加LiHMDS(223 mg,1.33 mmol,6當量)。將反應混合物在60℃下攪拌30分鐘。在冷卻到室溫後,將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-TLC使用二氯甲烷/甲醇(15/1)洗脫進行純化,以得到呈白色固體的50 mg (11 R)-26-乙基-5,11-二甲基-16-(4-甲基哌嗪-1-基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3, 14,16,18,20,24-八烯-23,27-二酮(實施例A14)(39%)。LCMS: m/z (ESI), [M+H] +=559.35。 1H NMR (400 MHz, DMSO-d 6) δ 0.80-0.86 (4H, m), 1.13-1.34 (4H, m), 1.40-1.46 (2H, m), 1.91 (2H, d), 2.26-2.34 (5H, m), 2.80 (1H, s), 3.16 (4H, t), 3.72 (3H, s), 3.94 (2H, m), 4.05-4.18 (3H, m), 4.35 (1H, t), 6.82-6.88 (1H, m), 7.13 (1H, d), 7.36 (1H, d), 8.26 (1H, d), 8.35 (1H, s), 8.80 (1H, d), 12.35 (1H, s)。 實施例A15 (11 R)-26-環丙基-5,11-二甲基-16-(4-甲基哌嗪-1-基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮 步驟1. 5-溴-1-環丙基-6-氧代吡啶-3-甲酸甲酯(INT-A15-1)的製備。將5-溴-6-羥基吡啶-3-甲酸甲酯(5 g,21.55 mmol,1當量)、環丙基硼酸(4.07 g,47.41 mmol,2.2當量)、Cu(OAc) 2(4.31 g,23.70 mmol,1.1當量)和三甲胺(4.36 g,43.10 mmol,2當量)於乙腈(100 mL)和乙醇(5 mL)中的混合物在氮氣氣氛下在80℃下攪拌12 h。將混合物冷卻到室溫並且過濾。將濾餅用乙酸乙酯(3×400 mL)洗滌,並且將所得溶液在減壓下濃縮。將殘餘物在矽膠柱上使用石油醚/乙酸乙酯(1/1)洗脫進行純化,以得到呈黃色固體的350 mg 5-溴-1-環丙基-6-氧代吡啶-3-甲酸甲酯(INT-A15-1)(30%)。LCMS: m/z (ESI), [M+H] +=271.90。 1H NMR (DMSO-d 6, 400 MHz) δ 0.90-0.97 (2H, m), 1.01-1.09 (2H, m), 3.38-3.42 (1H, m), 3.80 (3H, s), 8.17 (1H, d), 8.22 (1H, d)。 步驟2. 1-環丙基-5-(2-甲基-3-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-4-基)-6-氧代吡啶-3-甲酸甲酯(INT-A15-2)的製備。將5-溴-1-環丙基-6-氧代吡啶-3-甲酸甲酯(400 mg,1.47 mmol,1當量)和4,4,5,5-四甲基-2-(四甲基-1,3,2-二氧雜環戊硼烷-2-基)-1,3,2-二氧雜環戊硼烷(560 mg,2.20 mmol,1.5當量)、Pd(dppf)Cl 2CH 2Cl 2(239 mg,0.29 mmol,0.2當量)和KOAc(432 mg,4.41 mmol,3當量)於二噁烷(8 mL)中的混合物在氮氣氣氛下在80℃下攪拌1 h,然後冷卻到室溫。向上述混合物中添加4-碘-2-甲基-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-3-酮(520.79 mg,1.470 mmol,1當量)、K 2CO 3(406 mg,2.94 mmol,2當量)和水(2 mL)。將所得混合物在80℃下另外攪拌2 h。冷卻到室溫後,將反應用水(100 mL)淬滅,並且將混合物用二氯甲烷(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-TLC使用二氯甲烷/甲醇(1/1)洗脫進行純化,以得到呈黃色固體的210 mg 1-甲基-5-(2-環丙基-3-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-4-基)-6-氧代吡啶-3-甲酸甲酯(INT-A15-2)(34%)。LCMS: m/z (ESI), [M+H] +=420.15。 1H NMR (DMSO-d 6, 400 MHz) δ 0.06 (9H, s), 0.83 (2H, t), 0.89-0.95 (2H, m), 1.04-1.10 (2H, m), 3.24-3.26 (1H, m), 3.37 (3H, s), 3.45-3.50 (2H, m), 3.81(3H, d), 5.32 (2H, s), 8.06 (1H, d), 8.89 (1H, s), 9.18(1H, d)。 步驟3. 1-環丙基-5-(5-羥基-1-甲基吡唑-4-基)-6-氧代吡啶-3-甲酸甲酯(INT-A15-3)的製備。將1-環丙基-5-(2-甲基-3-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-4-基)-6-氧代吡啶-3-甲酸甲酯(210 mg,0.50 mmol,1當量)和HCl(5 mL,1,4-二噁烷中4 M)的溶液在室溫下攪拌6 h。將所得混合物在減壓下濃縮。將殘餘物用CH 2Cl 2(20 mL)和飽和NaHCO 3(水溶液)(50 mL)處理。將混合物用二氯甲烷(3×100 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥,並且在減壓下濃縮,以得到190 mg 1-環丙基-5-(5-羥基-1-甲基吡唑-4-基)-6-氧代吡啶-3-甲酸甲酯(INT-A15-3),其在不進行進一步純化的情況下直接用於下一步驟。LCMS: m/z (ESI), [M+H] +=290.00。 1H NMR (DMSO-d 6, 400 MHz) δ 0.95-1.00 (2H, m), 1.08-1.12 (2H, m), 3.45-3.49 (1H, m), 3.51 (3H, s), 3.83 (3H, s), 7.46-7.69 (1H, m), 8.03 (1H, s), 8.08 (1H, d), 8.38 (1H, d)。 步驟4. 5-(5-{[(4 R)-5-[(5-溴-2-硝基苯基)氨基]-4-甲基戊基]氧基}-1-甲基吡唑-4-基)-1-環丙基-6-氧代吡啶-3-甲酸甲酯(INT-A15-4)的製備。在氮氣氣氛下在0℃下向1-環丙基-5-(5-羥基-1-甲基吡唑-4-基)-6-氧代吡啶-3-甲酸甲酯(粗製物,190 mg,0.66 mmol,1當量)和三苯基膦(516 mg,1.97 mmol,3當量)於四氫呋喃(10 mL)中的溶液中添加偶氮二甲酸二異丙酯(398 mg,1.97 mmol,3當量)和(4 R)-5-[(5-溴-2-硝基苯基)氨基]-4-甲基戊-1-醇(208 mg,0.66 mmol,1當量)。將所得混合物在0℃下攪拌3小時。將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用乙酸乙酯洗脫進行純化,以得到呈黃色固體的135 mg 5-(5-{[(4 R)-5-[(5-溴-2-硝基苯基)氨基]-4-甲基戊基]氧基}-1-甲基吡唑-4-基)-1-環丙基-6-氧代吡啶-3-甲酸甲酯(INT-A15-4)(33%)。LCMS: m/z (ESI), [M+H] +=589.90。 1H NMR (DMSO-d 6, 400 MHz) δ 0.85-1.00 (5H, m), 1.02-1.12 (2H, m), 1.15-1.30 (2H, m), 1.30-1.52 (1H, m), 1.53-1.96 (3H, m), 3.19-3.23 (1H, m), 3.38-3.42 (1H, m), 3.68 (3H, s), 3.77 (3H, s), 3.95 (2H, t), 6.80-6.86 (1H, m), 7.26 (1H, d), 7.97-8.01 (2H, m), 8.06 (1H, d), 8.11 (1H, d), 8.20 (1H, t) 步驟5. 5-(5-{[(4 R)-5-[(2-氨基-5-溴苯基)氨基]-4-甲基戊基]氧基}-1-甲基吡唑-4-基)-1-環丙基-6-氧代吡啶-3-甲酸甲酯(INT-A15-5)的製備。在氮氣氣氛下在0℃下向雷尼鎳(60 mg,0.70 mmol,3當量)和5-(5-{[(4 R)-5-[(5-溴-2-硝基苯基)氨基]-4-甲基戊基]氧基}-1-甲基吡唑-4-基)-1-環丙基-6-氧代吡啶-3-甲酸甲酯(135 mg,0.2 mmol,1當量)於甲醇(3 mL)中的混合物中添加水合肼(23 mg,0.46 mmol,2當量)。將所得混合物在室溫下攪拌2 h。將所得混合物過濾。將濾餅用甲醇(3×50 mL)洗滌。將所得溶液在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(16/1)洗脫進行純化,以得到呈棕色固體的85 mg 5-(5-{[(4 R)-5-[(2-氨基-5-溴苯基)氨基]-4-甲基戊基]氧基}-1-甲基吡唑-4-基)-1-環丙基-6-氧代吡啶-3-甲酸甲酯(INT-A15-5)(65%)。LCMS: m/z (ESI), [M+H] +=558.05。 1H NMR (DMSO-d 6, 400 MHz) δ 0.80-1.12 (7H, m), 1.20-1.40 (2H, m), 1.52-1.95 (4H, m), 2.70-3.02 (2H, m), 3.68 (3H, s), 3.79 (3H, s), 3.96 (2H, t), 4.59-4.75 (3H, m), 6.41-6.54 (3H, m), 8.01 (1H, s), 8.09 (1H, d), 8.14 (1H, d) 步驟6. 5-(5-{[(4 R)-4-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]戊基]氧基}-1-甲基吡唑-4-基)-1-環丙基-6-氧代吡啶-3-甲酸甲酯(INT-A15-6)的製備。將5-(5-{[(4 R)-5-[(2-氨基-5-溴苯基)氨基]-4-甲基戊基]氧基}-1-甲基吡唑-4-基)-1-環丙基-6-氧代吡啶-3-甲酸甲酯(85 mg,0.15 mmol,1當量)和溴化氰(24 mg,0.23 mmol,1.5當量)於乙醇(18 mL)中的混合物在室溫下攪拌3 h。將所得混合物在減壓下濃縮,並且將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(12/1)洗脫進行純化,以得到呈棕色固體的60 mg 5-(5-{[(4 R)-4-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]戊基]氧基}-1-甲基吡唑-4-基)-1-環丙基-6-氧代吡啶-3-甲酸甲酯(INT-A15-6)(65%)。LCMS: m/z (ESI), [M+H] +=585.05。 步驟7. 5-(5-{[(4 R)-4-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]戊基]氧基}-1-甲基吡唑-4-基)-1-環丙基-6-氧代吡啶-3-羧酸(INT-A15-7)的製備。向5-(5-{[(4 R)-4-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]戊基]氧基}-1-甲基吡唑-4-基)-1-環丙基-6-氧代吡啶-3-甲酸甲酯(70 mg,0.12 mmol,1當量)於四氫呋喃(4 mL)中的混合物中添加含LiOH(11 mg,0.48 mmol,4當量)的水(1 mL)。在室溫下攪拌2 h後,將混合物在減壓下濃縮。將殘餘物通過反向快速色譜法使用10%至50%乙腈/水洗脫進行純化,以得到呈灰白色固體的50 mg 5-(5-{[(4R)-4-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]戊基]氧基}-1-甲基吡唑-4-基)-1-環丙基-6-氧代吡啶-3-羧酸(INT-A15-7)(73%)。LCMS: m/z (ESI), [M+H] +=569.25。 步驟8. (11 R)-16-溴-26-環丙基-5,11-二甲基-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(INT-A15-8)的製備。將5-(5-{[(4 R)-4-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]戊基]氧基}-1-甲基吡唑-4-基)-1-環丙基-6-氧代吡啶-3-羧酸(60 mg,0.10 mmol,1當量)、 N, N-二異丙基乙胺(27 mg,0.21 mmol,2當量)和 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(80 mg,0.21 mmol,2當量)於1,4-二噁烷(1 mL,11.80 mmol,112當量)中的混合物在氮氣氣氛下在室溫下攪拌2小時。將反應用水(50 mL)淬滅,並且將混合物用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(25/1)洗脫進行純化,以得到呈黃色固體的27 mg (11 R)-16-溴-26-環丙基-5,11-二甲基-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(INT-A15-8)(46%)。LCMS: m/z (ESI), [M+H] +=553.10。 步驟9. (11 R)-26-環丙基-5,11-二甲基-16-(4-甲基哌嗪-1-基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(實施例A15)的製備。在氮氣氣氛下向(11 R)-16-溴-26-環丙基-5,11-二甲基-7-氧雜-4,5,13, 20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(20 mg,0.04 mmol,1當量)、1-甲基哌嗪(14.53 mg,0.144 mmol,4當量)和BrettPhos Pd G 3(9.86 mg,0.01 mmol,0.3當量)於二噁烷(1 mL)中的攪拌混合物中逐滴添加LiHMDS(0.14 mL,0.144 mmol,4當量)。將反應混合物在60℃下攪拌30分鐘。在冷卻到室溫後,將反應用水(50 mL)淬滅,並且將混合物用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過反向快速色譜法使用10%至50%乙腈/水洗脫進行純化,以得到呈白色固體的8.6 mg (11 R)-26-環丙基-5,11-二甲基-16-(4-甲基哌嗪-1-基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(實施例A15)(41%)。LCMS: m/z (ESI), [M+H] += 571.35。 1H NMR (DMSO-d 6, 400 MHz) δ 0.80 (3H, d), 0.87-0.89 (2H, m), 1.09-1.13 (2H, m), 1.42-1.48 (1H, m), 1.90-1.92 (2H, m), 2.18-2.28 (4H, m), 2.50 (4H, t), 2.78-2.82 (1H, m), 3.16 (4H, t), 3.42-3.46 (1H, m), 3.72 (3H, s), 3.87-3.94 (2H, m), 4.09 (1H, d), 4.33 (1H, t), 6.84-6.88 (1H, m), 7.14 (1H, d), 7.31-7.37 (1H, m), 8.04 (1H, d), 8.35 (1H, s), 8.77 (1H, d), 12.38 (1H, s)。 實施例A16A和實施例A16B 11-乙基-5,26-二甲基-16-(4-甲基哌嗪-1-基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮,異構體1(實施例A16A)和異構體2(實施例A16B) 步驟1. N, N-二苄基丁醯胺(INT-A16-1)的製備。在氮氣氣氛下在室溫下向丁酸(10 g,113.50 mmol,1當量)和二苄胺(22.39 g,113.50 mmol,1當量)於1,4-二噁烷(100 mL)中的攪拌混合物中添加 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(64.73 g,170.25 mmol,1.5當量)和 N,N-二異丙基乙胺(44.01 g,340.50 mmol,3當量)。將所得混合物在氮氣氣氛下在室溫下攪拌過夜。將反應用水(300 mL)淬滅,並且將混合物用CH 2Cl 2(3×300 mL)萃取。將合併的有機層用鹽水(3×200 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過C18柱使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的25 g N, N-二苄基丁醯胺(INT-A16-1)(82%)。LCMS: m/z (ESI), [M+H] +=268.00。 步驟2. N, N-二苄基-2-乙基戊-4-烯醯胺(INT-A16-2)的製備。在氮氣氣氛下在-78℃下向 N, N-二苄基丁醯胺(10 g,37.40 mmol,1當量)於四氫呋喃(100 mL)中的攪拌混合物中添加LiHMDS(41.14 mL,41.14 mmol,1.1當量)和烯丙基溴(6.79 g,56.10 mmol,1.5當量)。將所得混合物在氮氣氣氛下在-60℃下攪拌2 h,並且升溫到室溫。將反應用水(200 mL)淬滅,並且將混合物用乙酸乙酯(3×200 mL)萃取。將合併的有機層用鹽水(3×200 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過C18柱使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈無色油的8 g N, N-二苄基-2-乙基戊-4-烯醯胺(INT-A16-2)(69%)。LCMS: m/z (ESI), [M+H] += 308.15。 1H NMR (DMSO-d 6, 400 MHz) δ 0.75 (3H, t), 1.30-1.46 (1H, m), 1.46-1.63 (1H, m), 2.02-2.17 (1H, m), 2.21-2.31 (1H, m), 2.63-2.79 (1H, m), 4.49-4.61 (4H, m), 4.86-5.07 (2H, m), 5.59-5.83 (1H, m), 7.12-7.49 (10H, m)。 步驟3. N, N-二苄基-2-乙基-5-羥基戊醯胺(INT-A16-3)的製備。將 N, N-二苄基-2-乙基戊-4-烯醯胺(6.0 g,19.51 mmol,1當量)和9-硼雜雙環[3.3.1]壬烷(5.95 g,48.79 mmol,2.5當量)於THF(15 mL)中的混合物在氮氣氣氛下在室溫下攪拌1 h。然後在0℃下添加含NaOH(1.17 g,29.27 mmol,1.5當量)的H 2O(1.50 mL,83.33 mmol,4.27當量)和H 2O 2(12.12 mL,156.13 mmol,8.0當量,30%)。將所得混合物在室溫下攪拌2 h。將反應用飽和Na 2S 2O 3溶液(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過C18柱使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈無色油的5.0 g N, N-二苄基-2-乙基-5-羥基戊醯胺(INT-A16-3)(79%)。LCMS: m/z (ESI), [M+H] +=326.15。 1H NMR (DMSO-d 6, 400 MHz) δ 0.75 (3H, t), 1.30-1.56 (6H, m), 2.60 (1H, t), 3.23-3.31 (2H, m), 4.56 (4H, d), 7.05-7.54 (10H, m)。 步驟4. 4-[(二苄基氨基)甲基]己-1-醇(INT-A16-4)的製備。在氮氣氣氛下在0℃下向 N, N-二苄基-2-乙基-5-羥基戊醯胺(5.0 g,15.36 mmol,1當量)於四氫呋喃(50 mL)中的混合物中添加LiAlH 4(1.16 g,30.73 mmol,2.0當量)。將所得混合物在0℃下攪拌2 h。將反應在室溫下用水(1.2 mL)和含NaOH(387.12 mg,9.679 mmol,0.63當量)的H 2O(800 mg)淬滅。將所得混合物過濾。將濾餅用四氫呋喃(2×50 mL)洗滌。將有機溶液在減壓下濃縮。將殘餘物通過C18柱使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈無色油的3.0 g 4-[(二苄基氨基)甲基]己-1-醇(INT-A16-4)(63%)。LCMS: m/z (ESI), [M+H] +=312.15。 步驟5. 甲基4-(氨基甲基)己-1-醇(INT-A16-5)的製備。將4-[(二苄基氨基)甲基]己-1-醇(3.0 g,9.63 mmol,1當量)和Pd/C(0.31 g,2.89 mmol,0.3當量)於MeOH(50 mL)中的混合物在氫氣氣氛下在20℃下攪拌2 h。將混合物過濾。將濾餅用甲醇(300 mL)洗滌。將溶液在減壓下濃縮,以得到呈無色油的1.2 g 4-(氨基甲基)己-1-醇(INT-A16-5)。此材料在不經進一步純化的情況下用於下一反應中。 1H NMR (DMSO-d 6, 400 MHz) δ 0.82 (3H, t), 1.0-1.44 (7H, m), 2.18-2.48 (2H, m), 3.37 (2H, t)。 步驟6. 4-{[(5-溴-2-硝基苯基)氨基]甲基}己-1-醇(INT-A16-6)的製備。將4-(氨基甲基)己-1-醇(140 mg,1.07 mmol,1當量)、4-溴-2-氟-1-硝基苯(234.72 mg,1.07 mmol,1.0當量)和K 2CO 3(294.91 mg,2.13 mmol,2.0當量)於乙腈(5.0 mL)中的混合物在氮氣氣氛下在60℃下攪拌過夜。將混合物冷卻到室溫。將反應用水(50 mL)淬滅,並且將混合物用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過C18柱使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃色固體的300 mg 4-{[(5-溴-2-硝基苯基)氨基]甲基}己-1-醇(INT-A16-6)(85%)。LCMS: m/z (ESI), [M+H] +=330.90。 1H NMR (DMSO-d 6, 400 MHz) δ 0.90 (3H, t), 1.34-1.49 (6H, m), 1.60-1.70 (1H, m), 3.28 (2H, t), 3.39 (2H, d), 4.32-4.43 (1H, m), 6.84 (1H, d), 7.26 (1H, d), 7.99 (1H, d), 8.15 (1H, t) 步驟7. 5-{5-[(4-{[(5-溴-2-硝基苯基)氨基]甲基}己基)氧基]-1-甲基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A16-7)的製備。在氮氣氣氛下在0℃下向5-(5-羥基-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M2,262.29 mg,0.99 mmol,1.00當量)和三苯基膦(784.00 mg,2.99 mmol,3當量)於四氫呋喃(5 mL)中的攪拌混合物中添加偶氮二甲酸二異丙酯(604.41 mg,2.99 mmol,3當量)和4-{[(5-溴-2-硝基苯基)氨基]甲基}己-1-醇(330 mg,0.99 mmol,1.00當量)。將所得混合物在氮氣氣氛下在0℃下攪拌2 h。將反應用水(50 mL)淬滅,並且將混合物用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過C18柱使用水和乙腈作為流動相進行純化,以得到呈橙色油的500 mg 5-{5-[(4-{[(5-溴-2-硝基苯基)氨基]甲基}己基)氧基]-1-甲基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A16-7)(87%)。LCMS: m/z (ESI), [M+H] +=577.85。 步驟8. 5-{5-[(4-{[(2-氨基-5-溴苯基)氨基]甲基}己基)氧基]-1-甲基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A16-8)的製備。將5-{5-[(4-{[(5-溴-2-硝基苯基)氨基]甲基}己基)氧基]-1-甲基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸甲酯(400 mg,0.69 mmol,1當量)、雷尼鎳(297.25 mg,3.47 mmol,5當量)和水合肼(98%)(52.11 mg,1.04 mmol,1.5當量)於甲醇(5 mL)中的混合物在氮氣氣氛下在0℃下攪拌2 h。將所得混合物過濾。將濾餅用甲醇(3×30 mL)洗滌。將溶液在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(10/1)洗脫進行純化,以得到呈棕色固體的300 mg 5-{5-[(4-{[(2-氨基-5-溴苯基)氨基]甲基}己基)氧基]-1-甲基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸酯(INT-A16-8)(79%)。LCMS: m/z (ESI), [M+H] +=546.00。 步驟9. 5-[5-({4-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]己基}氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A16-9)的製備。將5-{5-[(4-{[(2-氨基-5-溴苯基)氨基]甲基}己基)氧基]-1-甲基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸甲酯(300 mg,0.55 mmol,1當量)和BrCN(69.78 mg,0.66 mmol,1.2當量)於CH 2Cl 2(5 mL)中的混合物在氮氣氣氛下在室溫下攪拌過夜,並且在減壓下濃縮。將殘餘物通過Prep-TLC使用石油醚/乙酸乙酯(1/3)洗脫進行純化,以得到呈棕色固體的220 mg 5-[5-({4-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]己基}氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A16-9)(90%)。LCMS: m/z (ESI), [M+H] +=572.95。 步驟10. 5-[5-({4-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]己基}氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-羧酸(INT-A16-10)的製備。將5-[5-({4-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]己基}氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(170 mg,0.23 mmol,1當量)和LiOH .H 2O(24.96 mg,0.55 mmol,2.5當量)於四氫呋喃(10 mL)和H 2O(2.5 mL)中的混合物在氮氣氣氛下在20℃下攪拌2 h。將所得混合物在減壓下濃縮。將殘餘物通過C18柱使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈橙色油的150 mg 5-[5-({4-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]己基}氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-羧酸(INT-A16-10)(87%)。LCMS: m/z (ESI), [M+H] +=559.00。 步驟11. 甲基16-溴-11-乙基-5,26-二甲基-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(INT-A16-11)的製備。將5-[5-({4-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]己基}氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-羧酸(150 mg,0.27 mmol,1當量)、N,N,N,N-四甲基-O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(204.63 mg,0.54 mmol,2.0當量)和 N,N-二異丙基乙胺(104.33 mg,0.81 mmol,3.0當量)於二噁烷(5 mL)中的混合物在氮氣氣氛下在60℃下攪拌2 h。將混合物冷卻到室溫。將反應用水(30 mL)淬滅,並且將混合物用乙酸乙酯(3×30 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(5/1)洗脫進行純化,以得到呈淡黃色固體的100 mg 16-溴-11-乙基-5,26-二甲基-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(INT-A16-11)(69%)。LCMS: m/z (ESI), [M+H] += 540.85。 步驟12. 5-{5-[(4-{[(2-氨基-5-溴苯基)氨基]甲基}己基)氧基]-1-甲基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸甲酯(A16)的製備。將1-甲基哌嗪(83.56 mg,0.85 mmol,5.0當量)、16-溴-11-乙基-5,26-二甲基-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(90 mg,0.17 mmol,1.00當量)、BrettPhos Pd G 3(45.37 mg,0.05 mmol,0.3當量)和LiHMDS(1.00 mL,1.00 mmol,6.0當量)於二噁烷(5.0 mL)中的混合物在氮氣氣氛下在60℃下攪拌2 h。將混合物冷卻到室溫。將反應通過添加飽和NH 4Cl溶液(30 mL)淬滅,並且將混合物用乙酸乙酯(3×30 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(8/1)洗脫進行純化,以得到呈淡黃色固體的2.0 mg 11-乙基-5,26-二甲基-16-(4-甲基哌嗪-1-基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1. 0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16, 18,20,24-八烯-23,27-二酮(A16)(2%)。LCMS: m/z (ESI), [M+H] +=559.50。 1H NMR (CD 3OD, 400 MHz) δ 0.82 (3H, t), 1.17-1.47 (6H, m), 1.74 (2H, s), 2.08 (1H, s), 2.28 (1H, s), 2.71 (1H, d), 2.88 (3H, s), 3.43 (4H, s), 3.68 (3H, s), 3.77 (3H, s), 3.80-3.92 (2H, m), 4.04 (1H, s), 4.30 (1H, d), 4.42 (1H, d), 6.99 (1H, s), 7.01 (1H, s), 7.39 (1H, d), 8.24 (1H, s), 8.40 (1H, s), 8.87 (1H, s) 步驟13. 11-乙基-5,26-二甲基-16-(4-甲基哌嗪-1-基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮,異構體1(實施例A16A)和異構體2(實施例A16B)的製備。將11-乙基-5,26-二甲基-16-(4-甲基哌嗪-1-基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(A16,25 mg,0.04 mmol)的外消旋混合物通過Prep-手性-HPLC使用CHIRALPAK IG柱(2*25 cm,5 μm)使用含0.1%二乙胺的叔丁基甲基醚作為流動相A和EtOH/CH 2Cl 2(1/1)作為流動相B分離,以得到呈白色固體的1.8 mg異構體1(實施例A16A,7%)和2.2 mg異構體2(實施例A16B,9%)。 異構體1:LCMS: m/z (ESI), [M+H] +=559.45。 1H NMR (CD 3OD, 400 MHz) δ 0.72-0.91 (3H, m), 1.16-1.45 (2H, m), 1.65-1.80 (2H, m), 2.03-2.12 (1H, m), 2.20-2.36 (1H, m), 2.60-2.74 (1H, m), 2.80 (4H, s), 3.15-3.26 (4H, m), 3.38 (4H, t), 3.56-3.71 (4H, m), 3.80 (3H, d), 4.00-4.06 (1H, m), 4.25-4.31 (1H, m), 4.39-4.45 (1H, m), 6.91 (1H, s), 7.02 (1H, d), 7.37 (1H, s), 8.12 (1H, s), 8.41 (1H, d), 8.83 (1H, s)。手性-HPLC,Rt=4.108分鐘。 異構體2:LCMS: m/z (ESI), [M+H] +=559.35。 1H NMR (CD 3OD, 400 MHz) δ 0.72-0.91 (3H, m), 1.16-1.45 (2H, m), 1.65-1.80 (2H, m), 2.03-2.12 (1H, m), 2.20-2.36 (1H, m), 2.60-2.74 (1H, m), 2.80 (4H, s), 3.15-3.26 (4H, m), 3.38 (4H, t), 3.56-3.71 (4H, m), 3.80 (3H, d), 4.00-4.06 (1H, m), 4.25-4.31 (1H, m), 4.39-4.45 (1H, m), 6.83 (1H, s), 6.98 (1H, d), 7.35 (1H, s), 8.05 (1H, s), 8.41 (1H, d), 8.77 (1H, s)。手性-HPLC,Rt=5.613分鐘。 實施例A18 5',26'-二甲基-16'-(4-甲基哌嗪-1-基)-7'-氧雜-4',5',13',20',22',26'-六氮雜螺[環丙烷-1,11'-五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八]-1'(28'), 2'(6'),3',14',16',18',20',24'-八烯-23',27'-二酮 步驟1. 3-(1-(((5-溴-2-硝基苯基)氨基)甲基)環丙基)丙-1-醇(INT-A18-1)的製備。將3-[1-(氨基甲基)環丙基]丙-1-醇(500 mg,3.87mmol,1當量)、K 2CO 3(1069 mg,7.74 mmol,2當量)和4-溴-2-氟-1-硝基苯(936 mg,4.25 mmol,1.1當量)於乙腈(5 mL)中的混合物在氮氣氣氛下在60℃下攪拌2 h。將混合物冷卻到室溫。將所得混合物在真空下濃縮。將殘餘物負載於矽膠柱上並且使用石油醚/乙酸乙酯(5:1)洗脫,以得到呈無色油的1.0 g 3-(1-{[(5-溴-2-硝基苯基)氨基]甲基}環丙基)丙-1-醇(INT-A18-1)(78%)。 1H NMR (DMSO-d 6, 400 MHz) δ 0.37-0.55 (4H, m), 1.31-1.44 (2H, m), 1.45-1.60 (2H, m), 3.25 (2H, d), 3.36-3.39 (2H, m), 4.45 (1H, s), 6.78-6.89 (1H, m), 7.26 (1H, d), 8.00 (1H, d), 8.08 (1H, t) 步驟2. 5-(5-(3-(1-(((5-溴-2-硝基苯基)氨基)甲基)環丙基)丙氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-甲酸甲酯(INT-A18-2)的製備。在氮氣氣氛下在0℃下向5-(5-羥基-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M2,700 mg,2.66 mmol,1當量)和三苯基膦(2092 mg,7.97 mmol,3當量)於四氫呋喃(10 mL)中的攪拌混合物中添加偶氮二甲酸二異丙酯(1613 mg,7.97 mmol,3當量)和3-(1-{[(5-溴-2-硝基苯基)氨基]甲基}環丙基)丙-1-醇(875 mg,2.66 mmol,1當量)。將混合物在室溫下攪拌2 h。將反應用水(80 mL)淬滅,並且將混合物用乙酸乙酯(3×60 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物負載於矽膠柱上並且使用石油醚/乙酸乙酯(1/10)洗脫進行純化,以得到呈黃色油的700 mg 5-{5-[3-(1-{[(5-溴-2-硝基苯基)氨基]甲基}環丙基)丙氧基]-1-甲基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A18-2)(45%)。 1H NMR (DMSO-d 6, 400 MHz) δ 0.42-0.47 (m, 2H), 0.52 (2H, d), 1.50-1.58 (m, 2H), 1.80-1.88 (2H, m), 3.20-3.29 (2H, m), 3.57 (3H, s), 3.66 (3H, s), 3.77 (3H, s), 3.88-3.96 (2H, m), 6.79-6.84 (1H, m), 7.25 (1H, d), 7.55-7.66 (1H, m), 7.98 (1H, d), 8.07 (1H, d), 8.10 (1H, d), 8.42 (1H, d)。 步驟3. 5-(5-(3-(1-(((2-氨基-5-溴苯基)氨基)甲基)環丙基)丙氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-甲酸甲酯(INT-A18-3)的製備。在氮氣氣氛下在0℃下向5-{5-[3-(1-{[(5-溴-2-硝基苯基)氨基]甲基}環丙基)丙氧基]-1-甲基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A18-2,700 mg,1.22 mmol,1當量)和雷尼鎳(50 mg)於甲醇(10 mL)中的攪拌混合物中分批添加NH 2NH 2 .H 2O(122 mg,2.43 mmol,2當量)。將混合物在0℃下攪拌1 h。將所得混合物過濾。將濾餅用甲醇(3×30 mL)洗滌,並且將溶液在真空下濃縮。將殘餘物直接用於下一步驟。 步驟4. 5-(5-(3-(1-((2-氨基-6-溴-1H-苯並[d]咪唑-1-基)甲基)環丙基)丙氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-甲酸甲酯(INT-A18-4)的製備。將5-{5-[3-(1-{[(2-氨基-5-溴苯基)氨基]甲基}環丙基)丙氧基]-1-甲基吡唑-4-基}-1-甲基-6-氧代吡啶-3-甲酸甲酯(580 mg,1.06 mmol,1當量)和BrCN(135 mg,1.27 mmol,1.2當量)於乙醇(6 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將混合物在真空下濃縮。將殘餘物負載於矽膠柱上並且使用亞甲基氯/甲醇(12/1)洗脫進行純化,以得到呈棕色固體的600 mg 5-[5-(3-{1-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]環丙基}丙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A18-4)(98%)。 1H NMR (DMSO-d 6, 400 MHz) δ 0.36 (2H, t), 0.45-0.54 (2H, m), 1.40-1.50 (2H, m), 1.78-1.88 (2H, m), 3.58 (3H, s), 3.65 (3H, s), 3.74 (3H, s), 3.88 (2H, t), 4.08 (2H, s), 6.65 (2H, s), 7.06 (2H, s), 7.35 (1H, s), 7.97 (1H, s), 8.08 (1H, d), 8.43 (1H, d) 步驟5. 5-(5-(3-(1-((2-氨基-6-溴-1H-苯並[d]咪唑-1-基)甲基)環丙基)丙氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-羧酸(INT-A18-5)的製備。將5-[5-(3-{1-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]環丙基}丙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(580 mg,1.02 mmol,1當量)和LiOH .H 2O(85 mg,2.03 mmol,2當量)於四氫呋喃/H 2O(0.8 mL/0.2 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將混合物在真空下濃縮。將殘餘物通過C18柱使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈棕色固體的400 mg 5-[5-(3-{1-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]環丙基}丙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-羧酸(INT-A18-5)(70%)。 1H NMR (DMSO-d 6, 400 MHz) δ 0.24-0.35 (2H, m), 0.42 (2H, t), 1.40-1.59 (2H, m), 1.89-1.96 (2H, m), 3.51 (3H, s), 3.63 (3H, s), 3.90 (2H, t), 4.17 (2H, s), 6.94 (2H, s), 7.02 (2H, d), 7.33-7.37 (1H, m),7.94(1H, s), 8.07 (1H, s), 8.38 (1H, d)。 步驟6. 16'-溴-5',26'-二甲基-7'-氧雜-4',5',13',20',22', 26'-六氮雜螺[環丙烷-1,11'-五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八]-1'(28'),2'(6'),3',14',16',18',20',24'-八烯-23',27'-二酮(INT-A18-6)的製備。在氮氣氣氛下在室溫下向5-[5-(3-{1-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]環丙基}丙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-羧酸(330 mg,0.59 mmol,1當量)和 N,N,N,N-四甲基-O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(338 mg,0.89 mmol,1.5當量)於二噁烷(5 mL)中的混合物中添加 N,N-二異丙基乙胺(230 mg,1.78 mmol,3當量)。將混合物在所述溫度下攪拌3小時。將反應用水(80 mL)淬滅,並且將混合物用乙酸乙酯(3×60 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC進行純化並且使用亞甲基二氯/甲醇(12/1)洗脫,以得到呈棕色固體的300 mg 16'-溴-5',26'-二甲基-7'-氧雜-4',5',13',20',22',26'-六氮雜螺[環丙烷-1,11'-五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八]-1'(28'), 2'(6'),3',14',16',18',20',24'-八烯-23',27'-二酮(INT-A18-6)(93%)。 1H NMR (DMSO-d 6, 400 MHz) δ 0.39 (2H, t), 0.63 (2H, t), 1.78 (2H, s), 2.20-2.28 (2H, m), 3.10-3.18 (2H, m), 3.65 (3H, d), 3.73 (3H, s), 4.15-4.28 (2H, m), 7.34-7.51 (2H, m), 7.75 (1H, d), 8.24-8.36 (2H, m), 8.71 (1H, d), 12.82 (1H, s) 步驟7. 5',26'-二甲基-16'-(4-甲基哌嗪-1-基)-7'-氧雜-4',5',13',20',22',26'-六氮雜螺[環丙烷-1,11'-五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八]-1'(28'), 2'(6'),3',14',16',18',20',24'-八烯-23',27'-二酮(實施例A18)的製備。在氮氣氣氛下在室溫下向16'-溴-5',26'-二甲基-7'-氧雜-4',5',13',20',22',26'-六氮雜螺[環丙烷-1,11'-五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八]-1'(28'), 2'(6'),3',14',16',18',20',24'-八烯-23',27'-二酮(100 mg,0.18 mmol,1當量)、1-甲基哌嗪(74 mg,0.74 mmol,4.0當量)和BrettPhos Pd G 3(50 mg,0.05 mmol,0.3當量)於1,4-二噁烷(4 mL)中的攪拌混合物中添加LiHMDS(1.12 mL,1.12 mmol,6當量)。將混合物在60℃下攪拌1 h,然後冷卻到室溫。將反應用飽和NH 4Cl(水溶液)溶液(20 mL)淬滅,並且將混合物用乙酸乙酯(3×20 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC進行純化並且使用亞甲基二氯/甲醇(12/1)洗脫,以得到呈白色固體的18 mg 5',26'-二甲基-16'-(4-甲基哌嗪-1-基)-7'-氧雜-4',5',13',20',22',26'-六氮雜螺[環丙烷-1,11'-五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八]-1'(28'),2'(6'),3',14',16',18',20',24'-八烯-23',27'-二酮(實施例A18)(17%)。LCMS: m/z (ESI), [M+H] +=557.35。 1H NMR (DMSO-d 6, 400 MHz) δ 0.35 (2H, t), 0.62 (2H, t), 1.73-1.81 (2H, m), 2.24 (5H, s), 2.45-2.49 (4H, m), 3.10-3.17 (4H, m), 3.62 (3H, s), 3.72 (3H, s), 4.18-4.29 (4H, m), 6.75-6.89 (1H, m), 6.95 (1H, d), 7.36 (1H, d), 8.29 (2H, d), 8.72 (1H, d), 12.51 (1H, s)。 實施例A19 5,12,26-三甲基-16-(4-甲基哌嗪-1-基)-7-氧雜-4,5, 13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮 步驟1. 5-氨基己-1-醇(INT-A19-1)的製備。將6-羥基-己-2-酮(1.0 g,8.60 mmol,1當量)、NH 4OAc(3.32 g,43.04 mmol,5.0當量)和NaBH 3CN(1.35 g,21.52 mmol,2.5當量)於甲醇(10 mL)中的混合物在氮氣氣氛下在80℃下攪拌2 h。將混合物冷卻到室溫。將反應用水(200 mL)淬滅,並且將混合物用乙酸乙酯(3×200 mL)萃取。將合併的有機層用鹽水(3×200 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過C18FLASH,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3 .H 2O和乙腈的水作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈無色油的2.5 g 5-氨基己-1-醇(INT-A19-1)(74%)。LCMS: m/z (ESI), [M+H] +=118.15。 步驟2. 5-((5-溴-2-硝基苯基)氨基)己-1-醇(INT-A19-2)的製備。將5-氨基己-1-醇(2.5 g,21.33 mmol,1當量)、K 2CO 3(8.84 g,63.99 mmol,3當量)和4-溴-2-氟-1-硝基苯(14.08 g,63.99 mmol,3當量)於乙腈(80 mL)中的混合物在氮氣氣氛下在60℃下攪拌過夜。將混合物冷卻到室溫。將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上使用石油醚/乙酸乙酯(5/1)洗脫進行純化,以得到呈黃色油的3.1 g 5-[(5-溴-2-硝基苯基)氨基]己-1-醇(INT-A19-2) (42%)。LCMS: m/z (ESI), [M+H] +=319.05。 1H NMR (DMSO-d 6, 400 MHz) δ 1.18-1.26 (3H, m), 1.29-1.49 (4H, m), 1.50-1.68 (2H, m), 3.36-3.43 (2H, m), 3.81-3.90 (1H, m), 4.37 (1H, t), 6.78-6.86 (1H, m), 7.31 (1H, d), 7.94 (1H, d), 7.99 (1H, d)。 步驟3. 5-(5-((5-((5-溴-2-硝基苯基)氨基)己基)氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-甲酸酯(INT-A19-3)的製備。在氮氣氣氛下在0℃下向5-(5-羥基-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M2,500 mg,1.89 mmol,1.00當量)和三苯基膦(1494 mg,5.69 mmol,3當量)於四氫呋喃(20 mL)中的攪拌混合物中添加含偶氮二甲酸二異丙酯(1152 mg,5.69 mmol,3當量)和5-[(5-溴-2-硝基苯基)氨基]己-1-醇(662 mg,2.08 mmol,1.1當量)的四氫呋喃(10 mL)。將混合物在0℃下攪拌2 h。將反應用水(50 mL)淬滅,並且將混合物用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上使用石油醚/乙酸乙酯(1/1)洗脫進行純化,以得到呈黃色固體的510 mg 5-[5-({5-[(5-溴-2-硝基苯基)氨基]己基}氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A19-3)(47%)。LCMS: m/z (ESI), [M+H] +=564.05。 步驟4. 5-(5-((5-((2-氨基-5-溴苯基)氨基)己基)氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-甲酸甲酯(INT-A19-4)的製備。在0℃下向5-[5-({5-[(5-溴-2-硝基苯基)氨基]己基}氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(490 mg,0.87 mmol,1當量)和雷尼鎳(599 mg,6.96 mmol,8當量)於甲醇(5 mL)中的攪拌混合物中添加肼(87.23 mg,1.742 mmol,2.0當量)。將混合物在氮氣氣氛下在0℃下攪拌2 h。將混合物過濾。將濾餅用甲醇(3×50 mL)洗滌,並且將所得溶液在減壓下濃縮,以得到呈棕色固體的400 mg 5-[5-({5-[(2-氨基-5-溴苯基)氨基]己基}氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A19-4)。此材料在不經進一步純化的情況下用於下一反應中。LCMS: m/z (ESI), [M+H] +=534.20。 步驟5. 5-(5-((5-(2-氨基-6-溴-1H-苯並[d]咪唑-1-基)己基)氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-甲酸甲酯(INT-A19-5)的製備。將5-[5-({5-[(2-氨基-5-溴苯基)氨基]己基}氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(400 mg,0.751 mmol,1當量)和BrCN(95.49 mg,0.901 mmol,1.2當量)於乙醇(5 mL)中的混合物在氮氣氣氛下在室溫下攪拌過夜。將反應用水(20 mL)淬滅,並且將混合物用二氯甲烷(3×30 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在Prep-TLC上使用二氯甲烷/甲醇(12/1)洗脫進行純化,以得到呈棕色固體的320 mg 5-(5-{[5-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)己基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A19-5)(76%)。LCMS: m/z (ESI), [M+H] +=557.20。 步驟6. 5-(5-((5-(2-氨基-6-溴-1H-苯並[d]咪唑-1-基)己基)氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-羧酸(INT-A19-6)的製備。將5-(5-{[5-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)己基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(300 mg,0.54 mmol,1當量)和LiOH.H 2O(67 mg,1.61 mmol,3.0當量)於四氫呋喃(4 mL)和水(1 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將混合物在減壓下濃縮。將殘餘物通過C18FLASH,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3.H 2O和乙腈的水作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈棕色固體的260 mg 5-(5-{[5-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)己基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A19-6)(88%)。LCMS: m/z (ESI), [M+H] +=545.20。 1H NMR (DMSO-d 6, 400 MHz) δ 1.37-1.39 (3H, m), 1.60-1.84 (1H, m), 1.90-1.97 (1H, m), 3.51 (3H, s), 3.61 (3H, s), 3.80-3.83 (1H, m), 3.89-3.92 (1H, m), 4.69-4.73 (1H, m), 7.02-7.04 (2H, m), 7.11 (2H, s), 7.37 (1H, t), 8.01 (2H, d), 8.34 (1H, d) 步驟7. 16-溴-5,12,26-三甲基-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(INT-A19-7)的製備。將5-(5-{[5-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)己基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(250 mg,0.46 mmol,1當量)、 N, N, N, N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(262 mg,0.69 mmol,1.5當量)和 N, N-二異丙基乙胺(178 mg,1.38 mmol,3.0當量)於1,4-二噁烷(5 mL)中的混合物在氮氣氣氛下在室溫下攪拌過夜。將反應用水(20 mL)淬滅,並且將混合物用二氯甲烷(3×30 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在Prep-TLC上使用二氯甲烷/甲醇(12/1)洗脫進行純化,以得到呈棕色固體的170 mg 16-溴-5,12,26-三甲基-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(INT-A19-7)(70%)。LCMS: m/z (ESI), [M+H] +=525.15。 步驟8. 5,12,26-三甲基-16-(4-甲基哌嗪-1-基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(實施例A19)的製備。在氮氣氣氛下在室溫下向16-溴-5,12,26-三甲基-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3, 14,16,18,20,24-八烯-23,27-二酮(100 mg,0.19 mmol,1當量)、1-甲基哌嗪(76 mg,0.76 mmol,4當量)和BrettPhos Pd G3(51 mg,0.06 mmol,0.3當量)於1,4-二噁烷(4 mL)中的攪拌混合物中添加LiHMDS(4 mL,1.14 mmol,6當量)。將混合物在60℃下攪拌1 h,並且冷卻到室溫。將反應用飽和NH 4Cl(水溶液)溶液(10 mL)淬滅,並且將混合物用CH 2Cl 2(3×30 mL)萃取。將合併的有機層經無水Na 2SO 4乾燥,並且在減壓下濃縮。將殘餘物通過prep-HPLC用XBridge Shield RP18 OBD柱,使用水10 mmol/L NH 4HCO 3和0.1% NH 3.H 2O和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的11 mg 5,12,26-三甲基-16-(4-甲基哌嗪-1-基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(實施例A19)(10%)。LCMS: m/z (ESI), [M+H] += 545.25。 1H NMR (DMSO-d 6, 400 MHz) δ 1.54-1.62 (4H, m), 1.63-1.70 (1H, m), 1.73-1.79 (1H, m), 1.82-1.92 (1H, m), 2.12-2.20 (2H, m), 2.22-2.31 (1H, m), 2.38-2.45 (4H, m), 3.12-3.27 (4H, m), 3.31 (3H, s), 3.61 (3H, s), 3.71 (3H, s), 3.90-4.08 (1H, m), 4.23-4.27 (1H, m), 4.82-4.98 (1H, m), 6.87-6.86 (1H, m), 7.09 (1H, s), 7.35-7.32 (1H, m), 8.23-8.31 (2H, m), 8.89-8.86 (1H, m), 12.29 (1H, s)。 實施例A20 (11 R)-11,26-二甲基-16-(4-甲基哌嗪-1-基)-5-(2,2,2-三氟乙基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮 步驟1. 4-(3-氯-4-硝基苯基)哌嗪-1-甲酸叔丁酯(INT-A20-1)的製備。將2-氯-4-氟-1-硝基苯(10 g,56.967 mmol,1當量)、K 2CO 3(15.75 g,113.934 mmol,2當量)和哌嗪-1-甲酸叔丁酯(11.67 g,62.664 mmol,1.1當量)於乙腈(200 mL)中的混合物在氮氣氣氛下在60℃下攪拌過夜。將混合物冷卻到室溫。將反應用水(800 mL)淬滅,並且將混合物用乙酸乙酯(3×600 mL)萃取。將合併的有機層用鹽水(3×500 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法使用石油醚/乙酸乙酯(1/1)洗脫進行純化,以得到呈黃色固體的5.4 g 4-(3-氯-4-硝基苯基)哌嗪-1-甲酸叔丁酯(INT-A20-1)(27%)。LCMS: m/z (ESI), [M+H] +=342.10。 步驟2. ( R)-4-(3-((5-羥基-2-甲基戊基)氨基)-4-硝基苯基)哌嗪-1-甲酸叔丁酯(INT-A20-2)的製備。將4-(3-氯-4-硝基苯基)哌嗪-1-甲酸叔丁酯(1.4 g,4.096 mmol,1當量)、K 2CO 3(1132.20 mg,8.192 mmol,2當量)和(4 R)-5-氨基-4-甲基戊-1-醇(M3-6,480.03 mg,4.096 mmol,1當量)於 N,N-二甲基乙醯胺(15 mL)中的混合物在氮氣氣氛下在100℃下攪拌過夜。將混合物冷卻到室溫。將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用石油醚/乙酸乙酯(1/1)洗脫來進行純化,以得到呈黃色固體的730 mg 4-(3-{[(2R)-5-羥基-2-甲基戊基]氨基}-4-硝基苯基)哌嗪-1-甲酸叔丁酯(INT-A20-2)(42%)。LCMS: m/z (ESI), [M+H] +=423.25。 步驟3. 2-(2,2,2-三氟乙基)吡唑-3-醇(INT-A20-3)的製備。將(2E)-3-甲氧基丙-2-烯酸甲酯(5 g,43.06 mmol,1當量)和(2,2,2-三氟乙基)肼HCl鹽(7.37 g,64.59 mmol,1.5當量)於甲醇(20 mL)中的混合物在氮氣氣氛下在80℃下攪拌2 h。將混合物冷卻到室溫。將反應用水(200 mL)淬滅,並且將混合物用二氯甲烷(3×200 mL)萃取。將合併的有機層用鹽水(3×200 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠柱使用二氯甲烷/甲醇(20/1)洗脫來進行純化,以得到呈黃色固體的6 g 2-(2,2,2-三氟乙基)吡唑-3-醇(INT-A20-3)(83%)。 1H NMR (DMSO, 400 MHz) δ 4.71 (2H, q), 5.38 (1H, d), 7.25 (1H, s), 11.40 (1H, s) 步驟4. 2-(2,2,2-三氟乙基)-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-3-酮(INT-A20-4)的製備。在0℃下向2-(2,2,2-三氟乙基)吡唑-3-醇(6 g,36.12 mmol,1當量)和K 2CO 3(9.98 g,72.24 mmol,2當量)於乙腈(100 mL)中的攪拌混合物中添加SEM-Cl(12.04 g,72.24 mmol,2當量)。將混合物在氮氣氣氛下在0℃下攪拌2 h。將反應用水(200 mL)淬滅,並且將混合物用二氯甲烷(3×200 mL)萃取。將合併的有機層用鹽水(3×200 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠柱使用石油醚/乙酸乙酯(1/3)洗脫進行純化,以得到呈棕色固體的900 mg 2-(2,2,2-三氟乙基)-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-3-酮(INT-A20-4)(8%)。 1H NMR (DMSO, 400 MHz) δ 0.81 (2H, d), 1.06 (1H, t), 1.24 (1H, s), 3.18 (1H, d), 3.45 (2H, m), 4.64 (2H, q), 5.09 (2H, s), 5.44 (1H, d), 8.08 (1H, d) 步驟5. 4-碘-2-(2,2,2-三氟乙基)-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-3-酮(INT-A20-5)的製備。向2-(2,2,2-三氟乙基)-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-3-酮(250 mg,0.84 mmol,1當量)於乙腈(5 mL)中的混合物中添加NIS(227 mg,1.01 mmol,1.2當量)。將所得混合物在氮氣氣氛下在0℃下攪拌2 h。將反應用飽和硫代硫酸鈉溶液(水溶液)(30 mL)淬滅,並且將混合物用二氯甲烷(3×20 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠柱使用石油醚/乙酸乙酯(1/3)洗脫進行純化,以得到呈棕色油的200 mg 4-碘-2-(2,2,2-三氟乙基)-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-3-酮(INT-A20-5)(56%)。 1H NMR (DMSO, 400 MHz) δ -0.05 (9H, s), 0.74-0.87 (2H, m), 3.42-3.50 (2H, m), 4.72 (2H, q), 5.10 (2H, s), 8.35 (1H, s)。 步驟6. 1-甲基-6-氧代-5-[3-氧代-2-(2,2,2-三氟乙基)-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-4-基]吡啶-3-甲酸甲酯(INT-A20-6)的製備。將5-溴-1-甲基-6-氧代吡啶-3-甲酸甲酯(561 mg,2.28 mmol mmol,1當量)、Pd(dppf)Cl 2 .CH 2Cl 2(167 mg,0.23 mmol,0.1當量)、雙(頻哪醇)二硼(868 mg,3.42 mmol,1.5當量)和KOAc(671 mg,6.84 mmol,3當量)於1,4-二噁烷(6 mL)中的混合物在氮氣氣氛下在80℃下攪拌過夜。將混合物冷卻到室溫,並且隨後添加4-碘-2-(2,2,2-三氟乙基)-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-3-酮(966 mg,2.28 mmol,1當量)、K 2CO 3(948 mg,6.86 mmol,3當量)、Pd(dppf)Cl 2(167 mg,0.23 mmol,0.1當量)、1,4-二噁烷(10 mL)和H 2O(5 mL)。將反應混合物在氮氣氣氛下在80℃下攪拌2 h。將混合物冷卻到室溫。將反應用水(50 mL)淬滅,並且將混合物用二氯甲烷(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用石油醚/乙酸乙酯(1/5)洗脫進行純化,以得到呈棕色固體的1 g 1-甲基-6-氧代-5-[3-氧代-2-(2,2,2-三氟乙基)-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-4-基]吡啶-3-甲酸甲酯(INT-A20-6)(94%)。LCMS: m/z (ESI), [M+H] +=462.00。 步驟7. 5-[5-羥基-1-(2,2,2-三氟乙基)吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A20-7)的製備。將1-甲基-6-氧代-5-[3-氧代-2-(2,2,2-三氟乙基)-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-4-基]吡啶-3-甲酸甲酯(1 g,2.17 mmol,1當量)和HCl於1,4-二噁烷(20 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將所得混合物在減壓下濃縮。將殘餘物用飽和NaHCO 3(水溶液)溶液(100 mL)處理,並且用二氯甲烷(3×100 mL)萃取。將合併的有機層用鹽水(3×200 mL)洗滌,經無水Na 2SO 4乾燥,並且在減壓下濃縮,以得到呈棕色固體的700 mg 5-[5-羥基-1-(2,2,2-三氟乙基)吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A20-7)(97%)。 1H NMR (DMSO, 400 MHz) δ 3.67 (3H, s), 3.84 (3H, s), 4.78 (2H, q), 8.11 (1H, s), 8.33 (1H, s), 8.50 (1H, d)。 步驟8. 4-(3-{[(2 R)-5-({4-[5-(甲氧基羰基)-1-甲基-2-氧代吡啶-3-基]-2-(2,2,2-三氟乙基)吡唑-3-基}氧基)-2-甲基戊基]氨基}-4-硝基苯基)哌嗪-1-甲酸叔丁酯(INT-A20-8)的製備。在氮氣氣氛下在0℃下向5-[5-羥基-1-(2,2,2-三氟乙基)吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(970 mg,2.93 mmol,1.00當量)和三苯基膦(2.304 g,8.78 mmol,3當量)於四氫呋喃(15 mL)中的攪拌溶液中添加4-(3-{[(2R)-5-羥基-2-甲基戊基]氨基}-4-硝基苯基)哌嗪-1-甲酸叔丁酯(INT-A20-2,1.237 g,2.93 mmol,1.00當量)和偶氮二甲酸二異丙酯(1.776 g,8.78 mmol,3當量)。將所得混合物在氮氣氣氛下在0℃下攪拌2 h。將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用石油醚/乙酸乙酯洗脫來進行純化,以得到呈棕色油的1.2 g 4-(3-{[(2 R)-5-({4-[5-(甲氧基羰基)-1-甲基-2-氧代吡啶-3-基]-2-(2,2,2-三氟乙基)吡唑-3-基}氧基)-2-甲基戊基]氨基}-4-硝基苯基)哌嗪-1-甲酸叔丁酯(INT-A20-8)(55%)。LCMS: m/z (ESI), [M+H] +=736.25。 步驟9. 4-(4-氨基-3-{[(2 R)-5-({4-[5-(甲氧基羰基)-1-甲基-2-氧代吡啶-3-基]-2-(2,2,2-三氟乙基)吡唑-3-基}氧基)-2-甲基戊基]氨基}苯基)哌嗪-1-甲酸叔丁酯(INT-A20-9)的製備。在氮氣氣氛下在0℃下向4-(3-{[(2 R)-5-({4-[5-(甲氧基羰基)-1-甲基-2-氧代吡啶-3-基]-2-(2,2,2-三氟乙基)吡唑-3-基}氧基)-2-甲基戊基]氨基}-4-硝基苯基)哌嗪-1-甲酸叔丁酯(600 mg,0.81 mmol,1當量)和雷尼鎳(699 mg,8.15 mmol,10當量)於甲醇(5 mL)中的攪拌混合物中添加水合肼(61 mg,1.22 mmol,1.5當量)。將所得混合物在室溫下攪拌2 h。將所得混合物過濾。將濾餅用甲醇(3×30 mL)洗滌,並且將溶液在減壓下濃縮,以得到呈黃色的575 mg 4-(4-氨基-3-{[(2 R)-5-({4-[5-(甲氧基羰基)-1-甲基-2-氧代吡啶-3-基]-2-(2,2,2-三氟乙基)吡唑-3-基}氧基)-2-甲基戊基]氨基}苯基)哌嗪-1-甲酸酯(INT-A20-9)。此材料在不經進一步純化的情況下用於下一反應中。LCMS: m/z (ESI), [M+H] +=706.45。 步驟10. 4-{2-氨基-3-[(2 R)-5-({4-[5-(甲氧基羰基)-1-甲基-2-氧代吡啶-3-基]-2-(2,2,2-三氟乙基)吡唑-3-基}氧基)-2-甲基戊基]-1,3-苯並二氮唑-5-基}哌嗪-1-甲酸叔丁酯(INT-A20-10)的製備。將4-(4-氨基-3-{[(2R)-5-({4-[5-(甲氧基羰基)-1-甲基-2-氧代吡啶-3-基]-2-(2,2,2-三氟乙基)吡唑-3-基}氧基)-2-甲基戊基]氨基}苯基)哌嗪-1-甲酸叔丁酯(600 mg,0.85 mmol,1當量)和BrCN(108 mg,1.02 mmol,1.2當量)於二氯甲烷(8 mL)中的溶液在氮氣氣氛下在室溫下攪拌過夜。將所得混合物在真空下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(10/1)洗脫來進行純化,以得到呈紫色固體的180 mg 4-{2-氨基-3-[(2 R)-5-({4-[5-(甲氧基羰基)-1-甲基-2-氧代吡啶-3-基]-2-(2,2,2-三氟乙基)吡唑-3-基}氧基)-2-甲基戊基]-1,3-苯並二氮唑-5-基}哌嗪-1-甲酸叔丁酯(INT-A20-10)(29%)。 1H NMR (DMSO, 400 MHz) δ 0.81 (4H, d), 1.15-1.34 (4H, m), 1.36-1.48 (11H, m), 1.70-1.76 (1H, m), 1.85-1.91 (1H, m), 1.97-2.05 (1H, m), 2.93-2.99 (4H, m), 3.30-3.37 (3H, m), 3.55-3.63 (3H, m), 3.75-3.83 (3H, m), 3.82-3.98 (5H, m), 4.86-4.96 (2H, m), 5.77 (6H, s), 6.67-6.77 (1H, m), 6.90 (1H, s), 6.97-7.16 (3H, m), 8.03 (1H, s), 8.05 (1H, d), 8.52 (1H, d)。 步驟11. 5-(5-{[(4 R)-4-({2-氨基-6-[4-(叔丁氧基羰基)哌嗪-1-基]-1,3-苯並二氮唑-1-基}甲基)戊基]氧基}-1-(2,2,2-三氟乙基)吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A20-11)的製備。將4-{2-氨基-3-[(2 R)-5-({4-[5-(甲氧基羰基)-1-甲基-2-氧代吡啶-3-基]-2-(2,2,2-三氟乙基)吡唑-3-基}氧基)-2-甲基戊基]-1,3-苯並二氮唑-5-基}哌嗪-1-甲酸叔丁酯(150 mg,0.20 mmol,1當量)和LiOH.H 2O(13 mg, 0.31 mmol, 1.5當量)於四氫呋喃(4 mL)和H 2O(1 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將所得混合物在減壓下濃縮。將殘餘物通過反向快速色譜法在C18矽膠柱上使用乙腈/水(0%至100%梯度)洗脫進行純化,以得到呈紫色固體的70 mg 5-(5-{[(4 R)-4-({2-氨基-6-[4-(叔丁氧基羰基)哌嗪-1-基]-1,3-苯並二氮唑-1-基}甲基)戊基]氧基}-1-(2,2,2-三氟乙基)吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A20-11)(47%)。 1H NMR (DMSO, 400 MHz) δ 0.76 (3H, d), 1.25 (1H, s), 1.42 (9H, s), 1.54-1.61 (1H, m), 1.70-1.76 (1H, m), 1.85-1.91 (1H, m), 1.97-2.05 (1H, s), 2.93-2.99 (4H, m), 3.30-3.37 (3H, m), 3.55-3.63 (3H, m), 3.75-3.83 (3H, m), 3.82-3.98 (5H, m), 4.88 (2H, m), 6.54 (2H, s), 6.60 (1H, q), 6.79 (1H, d), 6.96 (1H, d), 8.02 (1H, s), 8.04 (1H, d), 8.25 (1H, d)。 步驟12. 4-[(11R)-11,26-二甲基-23,27-二氧代-5-(2,2,2-三氟乙基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3, 14,16,18,20,24-辛-16-基]哌嗪-1-甲酸叔丁酯(INT-A20-12)的製備。將 N,N-二異丙基乙胺(27 mg,0.21 mmol,3當量)、5-(5-{[(4R)-4-({2-氨基-6-[4-(叔丁氧基羰基)哌嗪-1-基]-1,3-苯並二氮唑-1-基}甲基)戊基]氧基}-1-(2,2,2-三氟乙基)吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(50 mg,0.07 mmol,1當量)和 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(40 mg,0.10 mmol,1.5當量)於1,4-二噁烷(2 mL)中的混合物在氮氣氣氛下在60℃下攪拌過夜。將混合物冷卻到室溫。將反應用水(30 mL)淬滅,並且將混合物用乙酸乙酯(3×20 mL)萃取。將合併的有機層用鹽水(3×20 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(10/1)洗脫進行純化,以得到呈黃色固體的48 mg 4-[(11R)-11,26-二甲基-23,27-二氧代-5-(2,2,2-三氟乙基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28), 2(6),3,14,16,18,20,24-辛-16-基]哌嗪-1-甲酸叔丁酯(INT-A20-12)(98%)。LCMS: m/z (ESI), [M+H] +=699.45。 步驟13. (11 R)-11,26-二甲基-16-(哌嗪-1-基)-5-(2,2,2-三氟乙基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3, 14,16,18,20,24-八烯-23,27-二酮(INT-A20-13)的製備。將4-[(11R)-11,26-二甲基-23,27-二氧代-5-(2,2,2-三氟乙基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-辛-16-基]哌嗪-1-甲酸叔丁酯(INT-A20-13 40 mg,0.06 mmol,1當量)和三氟乙酸(0.5 mL)於二氯甲烷(2 mL)中的混合物在氮氣氣氛下在室溫下攪拌1 h。將所得混合物在減壓下濃縮。將殘餘物用飽和NaHCO 3(水溶液)溶液(100 mL)處理,並且用二氯甲烷(3×100 mL)萃取。將合併的有機層用鹽水(3×200 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將粗產物在不進行進一步純化的情況下直接用於下一步驟。LCMS: m/z (ESI), [M+H] +=599.15。 步驟14. (11 R)-11,26-二甲基-16-(4-甲基哌嗪-1-基)-5-(2,2,2-三氟乙基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3, 14,16,18,20,24-八烯-23,27-二酮(實施例A20)的製備。在室溫下向(11 R)-11,26-二甲基-16-(哌嗪-1-基)-5-(2,2,2-三氟乙基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3, 14,16,18,20,24-八烯-23,27-二酮(40 mg,0.07 mmol,1當量)、三甲胺(20 mg,0.20 mmol,3當量)和多聚甲醛(6 mg,0.06 mmol,1當量)於二氯甲烷(2 mL)中的攪拌混合物中添加乙醯氧基硼氫化鈉(42 mg,0.20 mmol,3當量)。將所得混合物在氮氣氣氛下在室溫下攪拌2 h,並且在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(5/1)洗脫進行純化,以得到呈黃綠色固體的6.7 mg (11 R)-11,26-二甲基-16-(4-甲基哌嗪-1-基)-5-(2,2,2-三氟乙基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(實施例A20)(16%)。LCMS: m/z (ESI), [M+H] +=613.30。 1H NMR (DMSO-d 6, 400 MHz) δ 0.77-0.89 (5H, m), 1.11-1.29 (9H, m), 1.40-1.50 (2H, m), 1.85-1.92 (2H, m), 2.15-2.21 (2H, m), 2.28 (3H, s), 2.75-2.80 (1H, m), 3.14-3.20 (3H, m), 3.63 (3H, s), 3.90-3.96 (2H, m), 4.07-4.11 (1H, m), 4.43-4.48 (1H, m), 4.93-5.00 (1H, m), 6.87 (1H, d), 7.14 (1H, s), 7.36 (1H, d), 8.31 (1H, d), 8.46 (1H, s), 8.73 (1H, d), 12.37 (1H, s) 實施例A21 (11 R)-5-乙基-11,26-二甲基-16-(4-甲基哌嗪-1-基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮 步驟1. 2-乙基吡唑-3-醇(INT-A21-1)的製備。將乙肼二鹽酸鹽(5.7 g,43.06 mmol,1當量)和(2E)-3-甲氧基丙-2-烯酸甲酯(5 g,43.06 mmol,1當量)於甲醇(50 mL)中的混合物在氮氣氣氛下在80℃下攪拌過夜。冷卻到室溫後,將反應混合物在減壓下濃縮,並且將殘餘物通過矽膠色譜法使用石油醚/乙酸乙酯(1/1)洗脫來進行純化,以得到呈白色固體的4 g 2-乙基吡唑-3-醇(INT-A21-1)(82%)。 1H NMR (DMSO-d 6, 400 MHz) δ 1.26 (3H, t), 3.94-3.98 (2H, m), 5.82 (1H, m), 7.48 (1H, d), 10.99 (1H, s)。 步驟2. 2-乙基-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-3-酮(INT-A21-2)的製備。將2-乙基吡唑-3-醇(5 g,44.59 mmol,1當量)、2-(三甲基甲矽烷基)乙氧基甲基氯(11.15 g,66.88 mmol,1.5當量)和K 2CO 3(15.41 g,111.47 mmol,2.5當量)於乙腈(50 mL)中的混合物在氮氣氣氛下在室溫下攪拌0.5 h。將反應用水(500 mL)淬滅,並且將混合物用乙酸乙酯(3×500 mL)萃取。將合併的有機層用鹽水(3×500 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法使用石油醚/乙酸乙酯(1/1)洗脫進行純化,以得到呈黃色油的1.5 g 2-乙基-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-3-酮(INT-A21-2)(13%)。 1H NMR (DMSO-d 6, 400 MHz) δ 0.00 (9H, s), 0.79-0.84 (2H, m), 1.04 (3H, t), 3.38-3.47 (2H, m), 3.77 (2H, q), 5.08 (2H, s), 5.27 (1H, d), 7.89 (1H, d) 步驟3. 2-乙基-4-碘-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-3-酮(INT-A21-3)的製備。將2-乙基-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-3-酮(1.15 g,4.74 mmol,1當量)和 N-碘代琥珀醯亞胺(1.28 g,5.69 mmol,1.2當量於乙腈(15 mL)中的混合物在氮氣氣氛下在0℃下攪拌3 h。將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法使用石油醚/乙酸乙酯(1/1)洗脫進行純化,以得到呈黃色固體的1.9 g 2-乙基-4-碘-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-3-酮(INT-A21-3)(97%)。 1H NMR (DMSO-d 6, 400 MHz) δ 0.03 (9H, s), 0.81 (2H, t), 1.05 (3H, t), 3.43 (2H, t), 3.83 (2H, q), 5.09 (2H, s), 8.17 (1H, s)。 步驟4. 5-(2-乙基-3-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A21-4)的製備。將5-溴-1-甲基-6-氧代吡啶-3-甲酸甲酯(1.4 g,5.69 mmol,1當量)、雙(頻哪醇)二硼(1.73 g,6.82 mmol,1.2當量)、KOAc(1.4 g,14.22 mmol,2.5當量)和Pd(dppf)Cl 2二氯甲烷(0.46 g,0.57 mmol,0.1當量)於二噁烷(16 mL)中的混合物在氮氣氣氛下在80℃下攪拌2 h。在添加2-乙基-4-碘-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-3-酮(1.7 g,4.61 mmol,0.9當量)、Pd(dppf)Cl 2二氯甲烷(0.28 g,0.51 mmol,0.1當量)、K 2CO 3(1.91 g,13.82 mmol,2.7當量)和H 2O(4 mL)之前,將反應混合物冷卻到室溫。將反應在氮氣氣氛下在80℃下另外攪拌2 h。在冷卻到室溫後,將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法使用二氯甲烷/甲醇(10/1)洗脫進行純化,以得到得到呈黃色固體的1.6 g 5-(2-乙基-3-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸乙酯(INT-A21-4)(76%)。LCMS: m/z (ESI), [M+H] +=408.10。 1H NMR (DMSO-d 6, 400 MHz) δ 0.05 (9H, s), 0.82 (2H, t), 1.10-1.13 (3H, m), 3.47 (2H, t), 3.60 (3H, s), 3.81 (3H, s), 3.89 (2H, q), 5.29 (2H, s), 8.43 (1H, d), 8.88 (1H, s), 9.21 (1H, d)。 步驟5. 5-(1-乙基-5-羥基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A21-5)的製備。將5-(2-乙基-3-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(1.6 g,3.92 mmol,1當量)和HCl(20 mL,1,4-二噁烷中4 M)的溶液在室溫下攪拌2 h。將殘餘物用二氯甲烷(30 mL)處理,並且用K 2CO 3處理。將混合物過濾。將濾餅用二氯甲烷(3×20 mL)洗滌。將所得溶液在減壓下濃縮,以得到呈黃色固體的640 mg 5-(1-乙基-5-羥基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A21-5)。此材料在不經進一步純化的情況下用於下一反應中。LCMS: m/z (ESI), [M+H] +=278.00。 步驟6. 5-(5-{[(4 R)-5-[(5-溴-2-硝基苯基)氨基]-4-甲基戊基]氧基}-1-乙基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A21-6)的製備。將5-(1-乙基-5-羥基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(670 mg,2.41 mmol,1當量)和三苯基膦(1.58 g,6.04 mmol,2.5當量)於四氫呋喃(10 mL)中的混合物在氮氣氣氛下在0℃下用偶氮二甲酸二異丙酯(0.98 g,4.83 mmol,2當量)處理1 h,隨後在0℃下添加(4 R)-5-[(5-溴-2-硝基苯基)氨基]-4-甲基戊-1-醇(M3-7,766 mg,2.41 mmol,1當量)。在氮氣氣氛下在0℃下另外攪拌4 h後,將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(10/1)洗脫進行純化,以得到呈黃色固體的720 mg 5-(5-{[(4 R)-5-[(5-溴-2-硝基苯基)氨基]-4-甲基戊基]氧基}-1-乙基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A21-6)(51%)。LCMS: m/z (ESI), [M+H] +=578.00。 步驟7. 5-(5-{[(4 R)-5-[(2-氨基-5-溴苯基)氨基]-4-甲基戊基]氧基}-1-乙基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A21-7)的製備。將5-(5-{[(4 R)-5-[(5-溴-2-硝基苯基)氨基]-4-甲基戊基]氧基}-1-乙基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(720 mg,1.25 mmol,1當量)、雷尼鎳(54 mg,0.62 mmol,0.5當量)和肼(120 mg,3.74 mmol,3當量)於甲醇(15 mL)中的混合物在室溫下攪拌2 h。將所得混合物過濾,並且將濾餅用甲醇(3×20 mL)洗滌。將所得溶液在減壓下濃縮,以得到呈黃色油的600 mg 5-(5-{[(4 R)-5-[(2-氨基-5-溴苯基)氨基]-4-甲基戊基]氧基}-1-乙基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A21-7)。此材料在不經進一步純化的情況下用於下一反應中。LCMS: m/z (ESI), [M+H] +=546.20。 步驟8. 5-(5-{[(4 R)-4-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]戊基]氧基}-1-乙基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A21-8)的製備。將5-(5-{[(4 R)-5-[(2-氨基-5-溴苯基)氨基]-4-甲基戊基]氧基}-1-乙基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(600 mg,1.10 mmol,1當量)和溴化氰(232 mg,2.20 mmol,2當量)於乙醇(15 mL)中的混合物在室溫下攪拌2 h並濃縮。將殘餘物通過矽膠色譜法使用二氯甲烷/甲醇(10/1)洗脫進行純化,以得到呈黃色油的400 mg 5-(5-{[(4 R)-4-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]戊基]氧基}-1-乙基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A21-8)(63%)。LCMS: m/z (ESI), [M+H] +=573.30。 步驟9. 5-(5-{[(4 R)-4-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]戊基]氧基}-1-乙基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A21-9)的製備。向5-(5-{[(4 R)-4-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]戊基]氧基}-1-乙基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(700 mg,1.22 mmol,1當量)於四氫呋喃(15 mL)中的攪拌混合物中添加含LiOH(58 mg,2.45 mmol,2當量)的H 2O(3 mL)。將所得混合物在室溫下攪拌2 h,並且在減壓下濃縮。將殘餘物通過反向快速色譜法使用10%至50%乙腈/水洗脫進行純化,以得到呈白色固體的600 mg 5-(5-{[(4 R)-4-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]戊基]氧基}-1-乙基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A21-9)(87%)。LCMS: m/z (ESI), [M+H] +=559.30。 步驟10. (11 R)-16-溴-5-乙基-11,26-二甲基-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(INT-A21-10)的製備。將5-(5-{[(4 R)-4-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]戊基]氧基}-1-乙基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(460 mg,0.82 mmol,1當量)、 N,N,N,N-四甲基-O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(627 mg,1.65 mmol,2當量)和 N, N-二異丙基乙胺(213 mg,1.65 mmol,2當量)於二噁烷(10 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(15/1)洗脫進行純化,以得到呈白色固體的430 mg (11 R)-16-溴-5-乙基-11,26-二甲基-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3, 14,16,18,20,24-八烯-23,27-二酮(INT-A21-10)(96%)。LCMS: m/z (ESI), [M+H] +=541.00。 步驟11. (11 R)-5-乙基-11,26-二甲基-16-(4-甲基哌嗪-1-基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(實施例A21)的製備。在氮氣氣氛下在室溫下向(11 R)-16-溴-5-乙基-11,26-二甲基-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(100 mg,0.18 mmol,1當量)、BrettPhos Pd G 3(84.02 mg,0.09 mmol,0.5當量)和1-甲基哌嗪(74 mg,0.74 mmol,4當量)於1,4-二噁烷(5 mL)中的混合物中逐滴添加LiHMDS(1.1 mL,1.11 mmol,6當量)。將反應混合物在60℃下攪拌30分鐘。在冷卻到室溫後,將反應用飽和NH 4Cl溶液(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(18/1)洗脫進行純化,以得到呈白色固體的29 mg (11 R)-5-乙基-11,26-二甲基-16-(4-甲基哌嗪-1-基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(實施例A21)(28%)。LCMS: m/z (ESI), [M+H] +=559.35。 1H NMR (DMSO-d 6, 400 MHz) δ 0.80 (3H, d), 1.37 (3H, t), 1.45 (1H, t), 1.90 (2H, d), 2.17-2.25 (4H, m), 2.51 (4H, t), 2.79 (1H, s), 3.15 (4H, t), 3.61 (3H, s), 3.86-4.13 (5H, m), 4.36-4.40 (1H, m), 6.86 (1H, d), 7.12 (1H, d), 7.35 (1H, d), 8.26 (1H, d), 8.37 (1H, s), 8.78 (1H, d), 12.35 (1H, s)。 實施例A22 (11 R)-5-環丙基-11,26-二甲基-16-(4-甲基哌嗪-1-基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮 步驟1. 5-(5-{[(4 R)-5-[(5-溴-2-硝基苯基)氨基]-4-甲基戊基]氧基}-1-環丙基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A22-1)的製備。在0℃下向5-(1-環丙基-5-羥基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-2-7,750 mg,2.59 mmol,1當量)和三苯基膦(1360 mg,5.19 mmol,2當量)於四氫呋喃(10 mL)中的攪拌混合物中添加偶氮二甲酸二異丙酯(1048 mg,5.19 mmol,2當量)和(4 R)-5-[(5-溴-2-硝基苯基)氨基]-4-甲基戊-1-醇(M3-7,905 mg,2.85 mmol,1.1當量)。將所得混合物在氮氣氣氛下在0℃下攪拌2 h。將反應用水(30 mL)淬滅,並且將混合物用二氯甲烷(3×30 mL)萃取。將合併的有機層經無水Na 2SO 4乾燥,並且在減壓下濃縮。將殘餘物在矽膠柱上使用石油醚/乙酸乙酯(1/1)洗脫進行純化,以得到呈黃色油的700 mg 5-(5-{[(4 R)-5-[(5-溴-2-硝基苯基)氨基]-4-甲基戊基]氧基}-1-環丙基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A22-1)(45%)。LCMS: m/z (ESI), [M+H] +=590.10 步驟2. 5-(5-{[(4 R)-5-[(2-氨基-5-溴苯基)氨基]-4-甲基戊基]氧基}-1-環丙基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A22-2)的製備。在0℃下向5-(5-{[(4 R)-5-[(5-溴-2-硝基苯基)氨基]-4-甲基戊基]氧基}-1-環丙基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(700 mg,1.19 mmol,1當量)和雷尼鎳(51 mg,0.60 mmol,0.5當量)於甲醇(15 mL)中的攪拌混合物中添加NH 2NH 2.H 2O(90 mg,1.79 mmol,1.5當量)。將混合物在氮氣氣氛下在0℃下攪拌1 h。將混合物過濾。將濾餅用甲醇(2×20 mL)洗滌。將所得溶液在減壓下濃縮。將殘餘物在矽膠柱上使用二氯甲烷/甲醇(10/1)洗脫進行純化,以得到呈黃色固體的570 mg 5-(5-{[(4 R)-5-[(2-氨基-5-溴苯基)氨基]-4-甲基戊基]氧基}-1-環丙基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A22-2)(85%)。LCMS: m/z (ESI), [M+H] +=560.40 步驟3. 5-(5-{[(4 R)-4-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]戊基]氧基}-1-環丙基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A22-3)的製備。將5-(5-{[(4 R)-5-[(2-氨基-5-溴苯基)氨基]-4-甲基戊基]氧基}-1-環丙基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(570 mg,1.02 mmol,1當量)和BrCN(162 mg,1.53 mmol,1.5當量)於乙醇(5 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將混合物在減壓下濃縮,並且將殘餘物在矽膠柱上使用二氯甲烷/甲醇(10/1)洗脫進行純化,以得到呈棕色固體的550 mg 5-(5-{[(4 R)-4-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]戊基]氧基}-1-環丙基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A22-3)(92%)。LCMS: m/z (ESI), [M+H] +=585.15 步驟4. 5-(5-{[(4 R)-4-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]戊基]氧基}-1-環丙基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A22-4)的製備。在室溫下向5-(5-{[(4 R)-4-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]戊基]氧基}-1-環丙基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(480 mg,0.823 mmol,1當量)於四氫呋喃(20 mL)中的攪拌混合物中添加LiOH(78.81 mg,3.292 mmol,4當量)和H 2O(5 mL)。將所得混合物在60℃下攪拌30分鐘。將混合物冷卻到室溫並在減壓下濃縮。將殘餘物通過反向快速色譜法使用10%至40%乙腈/水洗脫進行純化,以得到呈灰白色固體的380 mg 5-(5-{[(4 R)-4-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]戊基]氧基}-1-環丙基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A22-4)(81%)。LCMS: m/z (ESI), [M+H] += 571.00。 步驟5. (11 R)-16-溴-5-環丙基-11,26-二甲基-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(INT-A22-5)的製備。將5-(5-{[(4 R)-4-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]戊基]氧基}-1-環丙基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(375 mg,0.66 mmol,1當量)、 N, N-二異丙基乙胺(255 mg,1.98 mmol,3當量)和 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(375 mg,0.98 mmol,1.5當量)於1,4-二噁烷(10 mL)中的混合物在氮氣氣氛下在60℃下攪拌2 h。將反應混合物冷卻到室溫,用飽和NH 4Cl(水溶液)(20 mL)處理,並且用二氯甲烷(3×20 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上使用二氯甲烷/甲醇(10/1)洗脫進行純化,以得到呈紅棕色固體的330 mg (11 R)-16-溴-5-環丙基-11,26-二甲基-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(INT-A22-5)(90%)。LCMS: m/z (ESI), [M+H] += 553.10。 步驟6. (11 R)-5-環丙基-11,26-二甲基-16-(4-甲基哌嗪-1-基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(實施例A22)的製備。在氮氣氣氛下在室溫下向(11 R)-16-溴-5-環丙基-11,26-二甲基-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(90 mg,0.16 mmol,1當量)、BrettPhos Pd G 3(44 mg,0.05 mmol,0.3當量)和1-甲基哌嗪(49 mg,0.49 mmol,3當量)於1,4-二噁烷(9 mL)中的攪拌混合物中添加LiHMDS(2.7 mL,2.7 mmol,16.9當量)。將混合物在氮氣氣氛下在60℃下攪拌30分鐘,並且冷卻到室溫。將反應用飽和NH 4Cl(水溶液)溶液(30 mL)淬滅,並且將混合物用二氯甲烷(3×20 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上使用二氯甲烷/甲醇(15/1)洗脫來進行純化,以得到呈灰白色固體的24.7 mg標題化合物(實施例A22)(26%)。LCMS: m/z (ESI), [M+H] +=571.40。 1H NMR (DMSO-d 6, 400 MHz) δ 0.81 (3H, d), 0.971.06 (4H, m), 1.37-1.54 (1H, m), 1.83-2.03 (2H, m), 2.17-2.23 (1H, m), 2.24 (3H, s), 2.50 (4H, t), 2.82 (1H, br s), 3.15 (4H, br s), 3.53-3.60 (1H, m), 3.61 (3H, s), 3.85-4.20 (3H, m), 4.30-4.47 (1H, m), 6.86 (1H, dd), 7.13 (1H, d), 7.35 (1H, d), 8.26 (1H, d), 8.30 (1H, s), 8.84 (1H, d), 12.35 (1H, s)。 實施例A23 (11 R)-15-氯-5,11,26-三甲基-16-(4-甲基哌嗪-1-基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮 步驟1. (4 R)-5-[(2-氯-3-氟-6-硝基苯基)氨基]-4-甲基戊-1-醇(INT-A23-1)的製備。在室溫下向2-氯-1,3-二氟-4-硝基苯(1.73 g,8.96 mmol,1.05當量)和(4 R)-5-氨基-4-甲基戊-1-醇(M3-6,1 g,8.53 mmol,1當量)於乙腈(25 mL)中的攪拌混合物中分批添加K 2CO 3(2.36 g,17.06 mmol,2當量)。將混合物在氮氣氣氛下在60℃下攪拌2 h,並且冷卻到室溫。將反應用水(100 mL)淬滅,並且將混合物用CH 2Cl 2(3×100 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法使用石油醚/乙酸乙酯(3/1)洗脫來進行純化,以得到呈黃色油的2 g (4 R)-5-[(2-氯-3-氟-6-硝基苯基)氨基]-4-甲基戊-1-醇(INT-A23-1)(80%)。LCMS: m/z (ESI), [M+H] +=290.95。 1H NMR (DMSO-d6, 400 MHz) δ 0.82 (3H, d), 1.01 - 1.30 (2H, m), 1.31-1.72 (3H, m), 2.78-2.89 (1H, m), 2.94-3.09 (1H, m), 3.33 (2H, d), 4.37 (1H, t), 6.59 (1H, t), 7.22 (1H, d), 7.76 (1H, d)。 步驟2. 4-(2-氯-3-{[(2 R)-5-羥基-2-甲基戊基]氨基}-4-硝基苯基)哌嗪-1-甲酸叔丁酯(INT-A23-2)的製備。在室溫下向(4 R)-5-[(2-氯-3-氟-6-硝基苯基)氨基]-4-甲基戊-1-醇(2 g,6.87 mmol,1當量)和哌嗪-1-甲酸叔丁酯(2.56 g,13.75 mmol,2當量)於二甲基亞碸(30 mL)中的攪拌混合物中逐滴添加 N,N-二異丙基乙胺(1.78 g,13.75 mmol,2當量)。將混合物在氮氣氣氛下在120℃下攪拌4 h,並且冷卻到室溫。將反應用水(50 mL)淬滅,並且將混合物用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法使用石油醚/乙酸乙酯(10/1)洗脫來進行純化,以得到呈黃色油的3.1 g 4-(2-氯-3-{[(2 R)-5-羥基-2-甲基戊基]氨基}-4-硝基苯基)哌嗪-1-甲酸叔丁酯(INT-A23-2)(98%)。LCMS: m/z (ESI), [M+H] +=457.15。 步驟3. 4-(2-氯-3-{[(2 R)-5-({4-[5-(甲氧基羰基)-1-甲基-2-氧代吡啶-3-基]-2-甲基吡唑-3-基}氧基)-2-甲基戊基]氨基}-4-硝基苯基)哌嗪-1-甲酸叔丁酯(INT-A23-3)的製備。在0℃下向5-(5-羥基-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M2,576.04 mg,1.09 mmol,1當量)和三苯基膦(1.7 g,3.28 mmol,3當量)於四氫呋喃(10 mL)中的攪拌混合物中添加偶氮二甲酸二異丙酯(1.3 g,3.28 mmol,3當量)和4-(2-氯-3-{[(2 R)-5-羥基-2-甲基戊基]氨基}-4-硝基苯基)哌嗪-1-甲酸叔丁酯(1 g,1.09 mmol,1當量)。將混合物在氮氣氣氛下在0℃下攪拌1 h。將反應用水(50 mL)淬滅,並且將混合物用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC進行純化並且使用二氯甲烷/甲醇(25/1)洗脫,以得到呈黃色油的1.1 g 4-(2-氯-3-{[(2 R)-5-({4-[5-(甲氧基羰基)-1-甲基-2-氧代吡啶-3-基]-2-甲基吡唑-3-基}氧基)-2-甲基戊基]氨基}-4-硝基苯基)哌嗪-1-甲酸叔丁酯(INT-A23-3)(66%)。LCMS: m/z (ESI), [M+H] +=702.15。 步驟4. 4-(4-氨基-2-氯-3-{[(2 R)-5-({4-[5-(甲氧基羰基)-1-甲基-2-氧代吡啶-3-基]-2-甲基吡唑-3-基}氧基)-2-甲基戊基]氨基}苯基)哌嗪-1-甲酸叔丁酯(INT-A23-4)的製備。在氮氣氣氛下在0℃下向雷尼鎳(500 mg)和4-(2-氯-3-{[(2 R)-5-({4-[5-(甲氧基羰基)-1-甲基-2-氧代吡啶-3-基]-2-甲基吡唑-3-基}氧基)-2-甲基戊基]氨基}-4-硝基苯基)哌嗪-1-甲酸叔丁酯(1.1 g,1.56 mmol,1當量)於甲醇(10 mL)中的攪拌混合物中添加NH 2NH 2 .H 2O(235.26 mg,4.69 mmol,3當量)。將混合物在0℃下攪拌1小時,並且過濾。將濾餅用甲醇(2×20 mL)洗滌。將所得溶液在減壓下濃縮。將殘餘物通過Prep-TLC進行純化並且使用二氯甲烷/甲醇(10/1)洗脫,以得到呈淡棕色油的610 mg 4-(4-氨基-2-氯-3-{[(2 R)-5-({4-[5-(甲氧基羰基)-1-甲基-2-氧代吡啶-3-基]-2-甲基吡唑-3-基}氧基)-2-甲基戊基]氨基}苯基)哌嗪-1-甲酸叔丁酯(INT-A23-4)(52%)。LCMS: m/z (ESI), [M+H] +=672.20。 1H NMR (DMSO- d 6, 400 MHz) δ 0.95 (3H, d), 1.95-1.25 (1H, m), 1.36-1.45 (11H, m), 1.59-1.75 (2H, m), 2.65-2.78 (5H, m), 2.81 (1H, d), 3.57 (4H, s), 3.62-3.76 (7H, m), 3.81 (1H, d), 3.92 (2H, t), 5.76 (3H, s), 6.49-6.59 (2H, m), 7.58-7.69 (1H, m), 8.00 (1H, s), 8.14 (1H, d), 8.45 (1H, d)。 步驟5. 4-{2-氨基-4-氯-3-[(2 R)-5-({4-[5-(甲氧基羰基)-1-甲基-2-氧代吡啶-3-基]-2-甲基吡唑-3-基}氧基)-2-甲基戊基]-1,3-苯並二氮唑-5-基}哌嗪-1-甲酸叔丁酯(INT-A23-5)的製備。將4-(4-氨基-2-氯-3-{[(2 R)-5-({4-[5-(甲氧基羰基)-1-甲基-2-氧代吡啶-3-基]-2-甲基吡唑-3-基}氧基)-2-甲基戊基]氨基}苯基)哌嗪-1-甲酸叔丁酯(600 mg,0.89 mmol,1當量)和BrCN(94.54 mg,0.89 mmol,1當量)於乙醇(8 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h並且在真空下濃縮。將殘餘物通過Prep-TLC進行純化並且使用二氯甲烷/甲醇(12/1)洗脫,以得到呈淡棕色固體的480 mg 4-{2-氨基-4-氯-3-[(2 R)-5-({4-[5-(甲氧基羰基)-1-甲基-2-氧代吡啶-3-基]-2-甲基吡唑-3-基}氧基)-2-甲基戊基]-1,3-苯並二氮唑-5-基}哌嗪-1-甲酸叔丁酯(INT-A23-5)(76%)。LCMS: m/z (ESI), [M+H] +=697.20。 1H NMR (DMSO- d 6, 400 MHz) δ 0.01 (5H, s), 1.24 (2H, s), 1.78-1.92 (8H, m), 2.60-2.79 (2H, m), 2.81-2.95 (4H, m), 3.17 (3H, d), 3.57 (3H, s), 3.71 (6H, d), 4.01-4.22 (2H, m), 5.95 (2H, s), 7.04 (2H, s), 7.47 (1H, s), 7.96 (1H, s), 8.12 (1H, d), 8.39 (1H, d)。 步驟6和步驟7. 4-[(11 R)-15-氯-5,11,26-三甲基-23,27-二氧代-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3, 14,16,18,20,24-辛-16-基]哌嗪-1-甲酸叔丁酯(INT-A23-7)的製備。將4-{2-氨基-4-氯-3-[(2 R)-5-({4-[5-(甲氧基羰基)-1-甲基-2-氧代吡啶-3-基]-2-甲基吡唑-3-基}氧基)-2-甲基戊基]-1,3-苯並二氮唑-5-基}哌嗪-1-甲酸叔丁酯(INT-A23-5,450 mg,0.76 mmol,1當量)和LiOH(18 mg,0.71 mmol,1.2當量)於四氫呋喃/H 2O(2 mL/1 mL)中的混合物在氮氣氣氛下在80℃下攪拌2 h。將混合物冷卻到室溫,並且在真空下濃縮。向所述殘餘物添加 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(166.96 mg,0.44 mmol,1.5當量 ) N,N-二異丙基乙胺(113.51 mg,0.87 mmol,3當量)和1,4-二噁烷(6 mL)。將混合物在氮氣氣氛下攪拌3 h。將混合物用水(10 mL)處理,用乙酸乙酯(3×10 mL)萃取,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC進行純化並且使用亞甲基二氯/甲醇(12/1)洗脫,以得到呈淡黃色固體的160 mg 4-[(11 R)-15-氯-5,11,26-三甲基-23,27-二氧代-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-辛-16-基]哌嗪-1-甲酸叔丁酯(INT-A23-7)(78%)。LCMS: m/z (ESI), [M+H] +=665.15。 步驟8. (11 R)-15-氯-5,11,26-三甲基-16-(哌嗪-1-基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(實施例A23)的製備。將4-[(11 R)-15-氯-5,11,26-三甲基-23,27-二氧代-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-辛-16-基]哌嗪-1-甲酸叔丁酯(150 mg,0.22 mmol,1當量)於三氟乙酸(1 mL)和CH 2Cl 2(4 mL)中的混合物在氮氣氣氛下在室溫下攪拌1 h。將混合物在減壓下濃縮。將殘餘物用飽和NaHCO 3溶液處理,並且用CH 2Cl 2(3×10 mL)萃取。將合併的有機層用鹽水(3×10 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC進行純化並且使用亞甲基二氯/甲醇(12/1)洗脫,以得到呈白色固體的13.5 mg (11 R)-15-氯-5,11,26-三甲基-16-(哌嗪-1-基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3, 14,16,18,20,24-八烯-23,27-二酮(實施例A23)(56%)。LCMS: m/z (ESI), [M+H] +=565.35。 1H NMR (DMSO-d 6, 400 MHz) δ1.23-1.32 (3H, m), 1.52-1.59 (2H, m), 1.73 (1H, s), 1.95 (1H, s), 2.12-2.19 (1H, m), 2.89-2.97 (9H, m), 3.62-3.69 (3H, m), 3.71-3.76 (3H, m), 3.90 (1H, s), 4.19 (1H, m), 4.32-4.39 (1H, m), 4.70-4.79 (1H, m), 7.09-7.27 (1H, m), 7.50 (1H, d), 8.28-8.45 (2H, m), 8.65-8.76 (1H, m)。 實施例A25 (11 R)-5,11,26-三甲基-16-(4-甲基哌嗪-1-基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮 步驟1. 1-(3-氯-4-硝基苯基)-4-甲基哌嗪(INT-A25-1)的製備。將2-氯-4-氟-1-硝基苯(4 g,22.79 mmol,1當量)和1-甲基哌嗪-(2.28 g,22.79 mmol,1當量)以及K 2CO 3(6.3 g,45.57 mmol,2當量)於乙腈(60 mL)中的混合物在氮氣氣氛下在60℃下攪拌2 h。將混合物冷卻到室溫,用水(100 mL)處理,並且用二氯甲烷(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用石油醚/乙酸乙酯(1/1)洗脫來進行純化,以得到呈黃色固體的5.6 g 1-(3-氯-4-硝基苯基)-4-甲基哌嗪(INT-A25-1)(96%)。LCMS: m/z (ESI), [M+H] +=256.05。 1H NMR (DMSO-d 6, 400 MHz) δ 2.22 (3H, s), 2.41 (4H, t), 3.45 (4H, t), 7.00-7.03 (1H, m), 7.10 (1H, d), 8.02 (1H, d)。 步驟2. (4 R)-4-甲基-5-{[5-(4-甲基哌嗪-1-基)-2-硝基苯基]氨基}戊-1-醇(INT-A25-1)的製備。將1-(3-氯-4-硝基苯基)-4-甲基哌嗪(1.5 g,5.87 mmol,1當量)、K 2CO 3(1.62 g,11.73 mmol,2當量)和(4 R)-5-氨基-4-甲基戊-1-醇(M3-6,0.82 g,7.04 mmol,1.2當量)於 N,N-二甲基甲醯胺(10 mL)中的混合物在氮氣氣氛下在120℃下攪拌16 h。將反應混合物冷卻到室溫,用水(80 mL)處理,並且用二氯甲烷(3×70 mL)萃取。將合併的有機層用鹽水(3×500 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-TLC使用石油醚/乙酸乙酯(1/1)洗脫來進行純化,以得到呈黃色油的1.5 g (4 R)-4-甲基-5-{[5-(4-甲基哌嗪-1-基)-2-硝基苯基]氨基}戊-1-醇(INT-A25-2)(76%)。LCMS: m/z (ESI), [M+H] +=337.15。 步驟3. 1-甲基-5-(1-甲基-5-{[(4 R)-4-甲基-5-{[5-(4-甲基哌嗪-1-基)-2-硝基苯基]氨基}戊基]氧基}吡唑-4-基)-6-氧代吡啶-3-甲酸甲酯(INT-A25-3)的製備。在氮氣氣氛下在0℃下向三苯基膦(1.57 g,5.97 mmol,3當量)和5-(5-羥基-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M2,498.05 mg,1.89 mmol,0.95當量)於四氫呋喃(20 mL)中的攪拌混合物中添加(4 R)-4-甲基-5-{[5-(4-甲基哌嗪-1-基)-2-硝基苯基]氨基}戊-1-醇(670 mg,1.99 mmol,1當量)和偶氮二甲酸二異丙酯(1.21 g,5.97 mmol,3當量)。將混合物在室溫下攪拌2 h,用水(80 mL)處理,並且用乙酸乙酯(3×80 mL)萃取。將合併的有機層用水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上使用二氯甲烷/甲醇(35/1)洗脫進行純化,以得到呈淡黃色固體的500 mg 1-甲基-5-(1-甲基-5-{[(4R)-4-甲基-5-{[5-(4-甲基哌嗪-1-基)-2-硝基苯基]氨基}戊基]氧基}吡唑-4-基)-6-氧代吡啶-3-甲酸甲酯(INT-A25-3)(43%)。LCMS: m/z (ESI), [M+H] +=582.20。 步驟4. 5-(5-{[(4 R)-5-{[2-氨基-5-(4-甲基哌嗪-1-基)苯基]氨基}-4-甲基戊基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A25-4)的製備。在氮氣氣氛下在0℃下向1-甲基-5-(1-甲基-5-{[(4 R)-4-甲基-5-{[5-(4-甲基哌嗪-1-基)-2-硝基苯基]氨基}戊基]氧基}吡唑-4-基)-6-氧代吡啶-3-甲酸甲酯(700 mg,1.20 mmol,1當量)於甲醇(6 mL)中的攪拌混合物中添加雷尼鎳(349.52 mg,4.08 mmol,3.4當量)和NH 2NH 2 .H 2O(90.37 mg,1.80 mmol,1.5當量)。將所得混合物在室溫下攪拌1 h,用水(20 mL)處理,並且用二氯甲烷(3×20 mL)萃取。將合併的有機層用水(3×10 mL)洗滌,經無水Na 2SO 4乾燥,並且在減壓下濃縮,以得到呈淡黃色固體的510 mg 5-(5-{[(4 R)-5-{[2-氨基-5-(4-甲基哌嗪-1-基)苯基]氨基}-4-甲基戊基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A25-4)(77%)。此材料在不經進一步純化的情況下用於下一反應中。LCMS: m/z (ESI), [M+H] +=552.25。 步驟5. 5-(5-{[(4 R)-4-{[2-氨基-6-(4-甲基哌嗪-1-基)-1,3-苯並二氮唑-1-基]甲基}戊基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A25-5)的製備。在氮氣氣氛下在室溫下向5-(5-{[(4 R)-5-{[2-氨基-5-(4-甲基哌嗪-1-基)苯基]氨基}-4-甲基戊基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(480 mg,0.87 mmol,1當量)於二氯甲烷(8 mL)中的攪拌混合物中添加溴化氰(138.24 mg,1.30 mmol,1.5當量)。將所得混合物在氮氣氣氛下在室溫下攪拌2 h,用水(20 mL)處理,並且用二氯甲烷(3×20 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法使用二氯甲烷/甲醇(15/1)洗脫進行純化,以得到呈紫色固體的170 mg 5-(5-{[(4 R)-4-{[2-氨基-6-(4-甲基哌嗪-1-基)-1,3-苯並二氮唑-1-基]甲基}戊基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A25-5)(34%)。LCMS: m/z (ESI), [M+H] +=577.40。 步驟6. 5-(5-{[(4 R)-4-{[2-氨基-6-(4-甲基哌嗪-1-基)-1,3-苯並二氮唑-1-基]甲基}戊基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A25-6)的製備。在室溫下向5-(5-{[(4 R)-4-{[2-氨基-6-(4-甲基哌嗪-1-基)-1,3-苯並二氮唑-1-基]甲基}戊基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(140 mg,0.24 mmol,1當量)於四氫呋喃(4 mL)中的攪拌混合物中添加含LiOH(11.63 mg,0.49 mmol,2當量)的水(0.3 mL)。將所得混合物在室溫下攪拌1 h,並且在減壓下濃縮。將殘餘物通過反向快速色譜法使用乙腈/水(0%至100%)洗脫進行純化,以得到呈白色固體的37 mg 5-(5-{[(4 R)-4-{[2-氨基-6-(4-甲基哌嗪-1-基)-1,3-苯並二氮唑-1-基]甲基}戊基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A25-6)(27%)。LCMS: m/z (ESI), [M+H] +=563.35。 步驟7. (11 R)-5,11,26-三甲基-16-(4-甲基哌嗪-1-基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(實施例A25)的製備。在室溫下向5-(5-{[(4 R)-4-{[2-氨基-6-(4-甲基哌嗪-1-基)-1,3-苯並二氮唑-1-基]甲基}戊基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(34 mg,0.06 mmol,1當量)於二噁烷(3 mL)中的攪拌混合物中添加 N,N-二異丙基乙胺(23 mg,0.18 mmol,3當量 ) N,N,N,N-四甲基-O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(34 mg,0.09 mmol,1.5當量)。將所得混合物在氮氣氣氛下在60℃下攪拌1 h,冷卻到室溫,並且在減壓下濃縮。將殘餘物通過Prep-HPLC用XBridge Shield RP18 OBD柱,使用含有10 mmol/L NH 4HCO 3的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的14.4 mg (11 R)-5,11,26-三甲基-16-(4-甲基哌嗪-1-基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3, 14,16,18,20,24-八烯-23,27-二酮(實施例A25)(43%)。LCMS: m/z (ESI), [M+H] +=545.35。 1H NMR (DMSO-d 6, 400 MHz) δ 0.81 (3H, d), 1.44 (1H, q), 1.92 (2H, d), 2.12-2.25 (1H, m), 2.25 (3H, s), 2.50 (4H, t), 2.80 (1H, br s), 3.16 (4H, t), 3.62 (3H, s), 3.72 (3H, s), 3.88-3.99 (2H, m), 4.10 (1H, d), 4.33-4.38 (1H, m), 6.86 (1H, dd), 7.13 (1H, d), 7.36 (1H, d), 8.27 (1H, d), 8.36 (1H, s), 8.81 (1H, d), 12.35 (1H, s)。 實施例A26 (11 R)5,11,26-三甲基-16-(嗎啉-4-基)-7-氧雜-4,5,13,20,22, 26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮 步驟1. 4-(3-氯-4-硝基苯基)嗎啉(INT-A26-1)的製備。將2-氯-4-氟-1-硝基苯(3 g,17.09 mmol,1當量)、K 2CO 3(4.72 g,34.20 mmol,2當量)和嗎啉(1.94 g,22.20 mmol,1.3當量)於乙腈(30 mL)中的混合物在氮氣氣氛下在80℃下攪拌4 h。將混合物冷卻到室溫,用水(100 mL)處理,並且用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在用石油醚/乙酸乙酯(5/1)洗脫的矽膠柱上進行純化,以得到呈黃色固體的2.7 g 4-(3-氯-4-硝基苯基)嗎啉(INT-A26-1)(65%)。 1H NMR (DMSO-d 6, 400 MHz) δ 3.38-3.49 (4H, m), 3.63-3.75 (4H, m,), 7.00-7.04 (1H, m), 7.12 (1H, d,), 8.04 (1H, d,)。 步驟2. ( R)-4-甲基-5-((5-嗎啉代-2-硝基苯基)氨基)戊-1-醇(INT-A26-2)的製備。將4-(3-氯-4-硝基苯基)嗎啉(1 g,4.12 mmol,1當量)、Cs 2CO 3(2.69 g,8.24 mmol,2當量)和(4 R)-5-氨基-4-甲基戊-1-醇(0.58 g,4.94 mmol,1.2當量)於二甲基乙醯胺(15 mL)中的混合物在氮氣氣氛下在120℃下攪拌4 h,並且冷卻到室溫。將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過反向快速色譜法使用C18矽膠柱使用乙腈/水(0%至100%梯度)作為流動相進行純化,以得到呈棕色固體的730 mg (4 R)-4-甲基-5-{[5-(嗎啉-4-基)-2-硝基苯基]氨基}戊-1-醇(INT-A26-2)(54%)。LCMS: m/z (ESI), [M+H] +=324.20。 1HNMR (DMSO-d 6, 400 MHz) δ 0.97 (3H, d), 1.21-1.29 (1H, m), 1.40-1.47 (3H, m), 1.78-1.87 (1H, m), 3.09-3.19 (1H, m), 3.21-3.28 (1H, m), 3.45-3.50 (4H, m) 3.69-3.73 (6H, m), 4.40 (1H, s), 6.40-6.43 (1H, m), 7.00-7.10 (1H, m), 7.92 (1H, d), 8.02-8.12 (1H, m)。 步驟3. ( R)-1-甲基-5-(1-甲基-5-((4-甲基-5-((5-嗎啉代-2-硝基苯基)氨基)戊基)氧基)-1H-吡唑-4-基)-6-氧代-1,6-二氫吡啶-3-甲酸甲酯(INT-A26-3)的製備。在0℃下向5-(5-羥基-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M2,260 mg,0.98 mmol,1當量)和三苯基膦(777 mg,2.964 mmol,3.0當量)於四氫呋喃(3 mL)中的攪拌混合物中添加偶氮二甲酸二異丙酯(599 mg,2.96 mmol,3當量)和(4 R)-4-甲基-5-{[5-(嗎啉-4-基)-2-硝基苯基]氨基}戊-1-醇(638 mg,1.97 mmol,2當量)。將混合物在氮氣氣氛下在0℃下攪拌2 h,用水(100 mL)處理,並且用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC進行純化並且使用石油醚/乙酸乙酯(1/1)洗脫,以得到呈棕色油的130 mg 1-甲基-5-(1-甲基-5-{[(4 R)-4-甲基-5-{[5-(嗎啉-4-基)-2-硝基苯基]氨基}戊基]氧基}吡唑-4-基)-6-氧代吡啶-3-甲酸甲酯(INT-A26-3)(23%)。LCMS: m/z (ESI), [M+H] +=569.30。 1H NMR (DMSO-d 6, 400 MHz) δ 0.98 (3H, d), 1.31-1.45 (1H, m), 1.60 (1H, m), 1.74-1.92 (3H, m), 3.16 (1H, m), 3.23-3.30 (1H, m), 3.37-3.43 (4H, m), 3.57 (3H, s), 3.65-3.73 (7H, m), 3.77 (3H, s), 3.96 (2H, t), 6.01 (1H, d), 6.43-6.48 (1H, m), 7.92 (1H, d), 7.99 (1H, s), 8.14 (1H, d), 8.41-8.46 (2H, m)。 步驟4. ( R)-5-(5-((5-((2-氨基-5-嗎啉代苯基)氨基)-4-甲基戊基)氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-甲酸甲酯(INT-A26-4)的製備。在氮氣氣氛下在0℃下向雷尼鎳(40 mg,0.47 mmol,1當量)和1-甲基-5-(1-甲基-5-{[(4 R)-4-甲基-5-{[5-(嗎啉-4-基)-2-硝基苯基]氨基}戊基]氧基}吡唑-4-基)-6-氧代吡啶-3-甲酸甲酯(INT-A26-3,270 mg,0.47 mmol,1當量)於甲醇(4 mL)中的攪拌混合物中添加NH 2NH 2 .H 2O(47 mg,0.95 mmol,2當量)。將混合物在0℃下攪拌1小時,並且過濾。將濾餅用甲醇(2×50 mL)洗滌,並且將所得溶液在減壓下濃縮。將粗產物在不進行進一步純化的情況下直接用於下一步驟。LCMS: m/z (ESI), [M+H] +=539.30。 步驟5. 甲基-( R)-5-(5-((5-(2-氨基-6-嗎啉代-1H-苯並[d]咪唑-1-基)-4-甲基戊基)氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-甲酸甲酯(INT-A26-5)的製備。將5-(5-{[(4 R)-5-{[2-氨基-5-(嗎啉-4-基)苯基]氨基}-4-甲基戊基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(190 mg,0.35 mmol,1當量)和BrCN(41 mg,0.38 mmol,1.1當量)於乙醇(3 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將混合物在真空下濃縮。將殘餘物通過Prep-TLC進行純化並且使用二氯甲烷/甲醇(12/1)洗脫,以得到呈粉紅色固體的100 mg 5-(5-{[(4 R)-4-{[2-氨基-6-(嗎啉-4-基)-1,3-苯並二氮唑-1-基]甲基}戊基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A26-5)(50%)。 1H NMR (DMSO-d 6, 400 MHz) δ 0.84 (d, 3H), 1.27-1.40 (1H, m), 1.44-1.60 (1H, m), 1.67-1.71 (1H, m), 1.80-1.94 (1H, m), 2.04 (1H, s), 2.94-3.03 (3H, m), 3.17 (1H, d), 3.55-3.61 (5H, m), 3.69-3.76 (4H, m), 3.78 (3H, s), 3.88-3.91 (2H, m), 6.21 (1H, s), 6.59-6.63 (1H, m), 6.76 (2H, d), 7.00 (1H, d), 7.99 (1H, s), 8.13 (1H, d), 8.45 (1H, d)。 步驟6 ( R)-5-(5-((5-(2-氨基-6-嗎啉代-1H-苯並[d]咪唑-1-基)-4-甲基戊基)氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-羧酸(INT-A26-6)的製備。將5-(5-{[(4 R)-4-{[2-氨基-6-(嗎啉-4-基)-1,3-苯並二氮唑-1-基]甲基}戊基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(100 mg,0.17 mmol,1當量)和LiOH(8.5 mg,0.35 mmol,2當量)於四氫呋喃/H 2O(4 mL/1 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h,並且在真空下濃縮。將殘餘物通過反向快速色譜法使用C18矽膠柱和乙腈/水(0%至100%梯度)作為流動相進行純化,以得到呈棕色固體的85 mg 5-(5-{[(4 R)-4-{[2-氨基-6-(嗎啉-4-基)-1,3-苯並二氮唑-1-基]甲基}戊基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A26-6)(87%)。 1H NMR (DMSO-d 6, 400 MHz) δ 0.78 (3H, d), 1.43 (1H, s), 1.54-1.75 (2H, m), 1.83 (1H, d), 2.03 (1H, s), 2.89-3.04 (4H, m), 3.50 (3H, s), 3.60 (3H, s), 3.70-3.75 (4H, m), 3.79-3.82 (1H, m), 3.94 (3H, m), 6.56-6.62 (2H, m), 6.76 (1H, d), 6.96 (1H, d), 8.00 (2H, d), 8.01-8.03 (1H, s), 8.31 (1H, d)。 步驟7 (11 R)5,11,26-三甲基-16-(嗎啉-4-基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(實施例A26)的製備。將5-(5-{[(4 R)-4-{[2-氨基-6-(嗎啉-4-基)-1,3-苯並二氮唑-1-基]甲基}戊基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(50 mg,0.09 mmol,1當量)、 N,N-二異丙基乙胺(35 mg,0.27 mmol,3當量)和 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(51 mg,0.13 mmol,1.5當量)於1,4-二噁烷(3 mL)中的混合物在氮氣氣氛下在60℃下攪拌1 h。將混合物冷卻到室溫,並且在真空下濃縮。將殘餘物通過Prep-HPLC用XBridge Prep OBD C18柱,使用含有10 mmol/L NH 4HCO 3的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的16 mg (11 R)5,11,26-三甲基-16-(嗎啉-4-基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3, 14,16,18,20,24-八烯-23,27-二酮(實施例A26)(34%)。LCMS: m/z (ESI), [M+H] +=532.30。 1H NMR (DMSO-d 6, 400 MHz) δ 0.81 (3H, d), 1.43 (1H, q), 1.92 (2H, d), 2.19 (1H, br s), 2.80 (1H, br s), 3.14 (4H, t), 3.62 (3H, s), 3.72 (3H, s), 3.78 (4H, t), 3.89-3.97 (2H, m), 4.06-4.14 (1H, m), 4.31-4.41 (1H, m), 6.87 (1H, dd), 7.15 (1H, d), 7.38 (1H, d), 8.27 (1H, d), 8.35 (1H, s), 8.81 (1H, d), 12.37 (1H, s)。 實施例A27 (11 R)-16-[3-(二甲基氨基)吡咯烷-1-基]-5,11,26-三甲基-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮 步驟1. (11 R)-16-[3-(二甲基氨基)吡咯烷-1-基]-5,11,26-三甲基-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3, 14,16,18,20,24-八烯-23,27-二酮(實施例A27)的製備。在氮氣氣氛下在室溫下向(11R)-16-溴-5,11,26-三甲基-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(M3,40 mg,0.07 mmol,1當量)、 N,N-二甲基吡咯烷-3-胺(34 mg,0.30 mmol,4當量)和BrettPhos Pd G 3(20 mg,0.02 mmol,0.3當量)於1,4-二噁烷(5 mL)中的攪拌混合物中逐滴添加LiHMDS(0.22 mL,0.22 mmol,3當量)。將反應混合物在60℃下攪拌30分鐘。在冷卻到室溫後,將反應用飽和NH 4Cl溶液(30 mL)淬滅,並且將混合物用二氯甲烷(3×20 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-TLC使用二氯甲烷/甲醇(15/1)洗脫進行純化,並且將產物通過Prep-HPLC使用XBridge Prep OBD C18柱使用含有10 mmol/L NH 4HCO 3的水和乙腈作為移動相進行進一步純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的5 mg (11 R)-16-[3-(二甲基氨基)吡咯烷-1-基]-5,11,26-三甲基-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3, 14,16,18,20,24-八烯-23,27-二酮(實施例A27)(13%)。LCMS: m/z (ESI), [M+H] +=559.50。 1H NMR (DMSO-d 6, 400 MHz) δ 0.81 (3H, d), 1.40-1.50 (1H, m), 1.88-1.95 (3H, m), 2.12-2.31 (8H, m), 2.79-2.84 (2H, m), 3.06-3.17 (1H, m), 3.23-3.49 (3H, m), 3.62 (3H, s), 3.72 (3H, s), 3.84-3.97 (2H, m), 4.06-4.15 (1H, m), 4.32-4.41 (1H, m), 6.47 (1H, d), 6.66 (1H, s), 7.33 (1H, d), 8.26 (1H, d), 8.36 (1H, s), 8.81 (1H, d), 12.28 (1H, s)。 使用上文針對實施例A27的合成描述的類似條件,由M3製備以下實施例: 實施例A28、實施例A29、實施例A30、實施例A31、實施例A32、實施例A33、實施例A34、實施例A35、實施例A36、實施例A37、實施例A38、實施例A29、實施例A40、實施例A41、實施例A42、實施例A43、實施例A44、實施例A45、實施例A46、實施例A47、實施例A48、實施例A49、實施例A50、實施例A51、實施例A52、實施例A53、實施例A54、實施例A55、實施例A56、實施例A57和實施例A58。 結構以及m/z和 1H NMR的資料列於表1中。 實施例A59、實施例A60A和實施例A60B 6-溴-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮(實施例A59), 15,21-二甲基-6-(4-甲基哌嗪-1-基)-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33), 18(22),19-八烯-12,16-二酮,異構體1(實施例A60A)和異構體2(實施例A60B) 步驟1. 1-[(4-溴-2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛烷-3-甲酸叔丁酯(INT-A60-1)的製備。將1-氨基-3-氮雜雙環[3.2.1]辛烷-3-甲酸叔丁酯(2.0 g,8.80 mmol,1當量)、K 2CO 3(2.44 g,17.6 mmol,2當量)和4-溴-1-氟-2-硝基苯(3.88 g,17.6 mmol,2當量)於DMSO(40 mL)中的混合物在氮氣氣氛下在120℃下攪拌2 h。將混合物冷卻到室溫。將反應用水(200 mL)淬滅,並且將混合物用乙酸乙酯(3×200 mL)萃取。將合併的有機層用鹽水(3×200 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過C18柱使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃色油的800 mg 1-[(4-溴-2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛烷-3-甲酸叔丁酯(INT-A60-1)(21%)。LCMS: m/z (ESI), [M+H] +=372.05。 步驟2. N-(4-溴-2-硝基苯基)-3-氮雜雙環[3.2.1]辛-1-胺(INT-A60-2)的製備。將1-[(4-溴-2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛烷-3-甲酸叔丁酯(550 mg,1.29 mmol,1當量)和三氟乙酸(2 mL)於二氯甲烷(6 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h,用飽和NaHCO 3(水溶液)溶液(50 mL)處理,並且用CH 2Cl 2(3×50 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥,並且在減壓下濃縮,以得到呈黃色固體的435 mg N-(4-溴-2-硝基苯基)-3-氮雜雙環[3.2.1]辛-1-胺(INT-A60-2)(100%)。LCMS: m/z (ESI), [M+H] +=328.00。 步驟3. N-(4-溴-2-硝基苯基)-3-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-3-氮雜雙環[3.2.1]辛-1-胺(INT-A60-3)的製備。將 N-(4-溴-2-硝基苯基)-3-氮雜雙環[3.2.1]辛-1-胺(435 mg,1.34 mmol,1當量)、(2-溴乙氧基)(叔丁基)二甲基矽烷(652 mg,2.72 mmol,2當量)、NaI(408 mg,2.72 mmol, 2當量)和K 2CO 3(377 mg,2.72 mmol,2當量)於DMF(5 mL)中的混合物在氮氣氣氛下在60℃下攪拌4 h。將混合物冷卻到室溫,用水(50 mL)處理,並且用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過C18柱使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃色固體的360 mg N-(4-溴-2-硝基苯基)-3-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-3-氮雜雙環[3.2.1]辛-1-胺(INT-A60-3)(54%)。LCMS: m/z (ESI), [M+H] +=485.90。 步驟4. 2-{1-[(4-溴-2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙醇(INT-A60-4)的製備。將 N-(4-溴-2-硝基苯基)-3-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-3-氮雜雙環[3.2.1]辛-1-胺(360 mg,0.74 mmol,1當量)和四丁基氟化銨(388 mg,1.48 mmol,2當量)於四氫呋喃(5 mL)中的混合物在氮氣氣氛下在室溫下攪拌過夜。將混合物在減壓下濃縮。將殘餘物負載於矽膠柱上並且使用二氯甲烷/甲醇(10/1)洗脫,以得到呈黃色固體的260 mg 2-{1-[(4-溴-2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙醇(INT-A60-4)(94%)。LCMS: m/z (ESI), [M+H] +=370.10。 1H NMR (DMSO-d 6, 400 MHz) δ 1.61-1.71 (3H, m), 1.79-1.83 (1H, m), 1.91-1.97 (1H, m), 2.11-2.17 (1H, m), 2.18-2.24 (1H, m), 2.30-2.49 (5H, m), 3.11-3.18 (1H, m), 3.45-3.55 (2H, m), 4.34 (1H, t), 7.17(1H, d), 7.57-7.67 (1H, m), 8.18 (1H, d), 8.29 (1H, s)。 步驟5. 5-[5-(2-{1-[(4-溴-2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A60-5)的製備。在氮氣氣氛下在0℃下向5-(5-羥基-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M2,260 mg,1.25 mmol,1.97當量)和三苯基膦(782 mg,2.98 mmol,4.70當量)於四氫呋喃(5 mL)中的攪拌混合物中添加2-{1-[(4-溴-2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙醇(235 mg,0.63 mmol,1當量)和偶氮二甲酸二異丙酯(451 mg,2.23 mmol,3.5當量)。將混合物在0℃下攪拌2 h,用水(50 mL)處理,並且用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過C18柱使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發,以得到呈橙色油的260 mg 5-[5-(2-{1-[(4-溴-2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A60-5)(66%)。LCMS: m/z (ESI), [M+H] +=615.15。 1H NMR (DMSO-d 6, 400 MHz) δ 1.47-1.63 (2H, m), 1.66-1.70 (1H, m), 1.72-1.85 (1H, m), 1.88-1.94 (1H, m), 2.04-2.23 (4H, m), 2.66-2.76 (2H, m), 2.80 -2.89 (1H, m), 3.07-3.11 (1H, m), 3.58 (3H, s),3.73 (3H, s), 3.78 (3H, s), 4.10 (1H, s), 4.11 (1H, s), 6.94 (1H, d), 7.56-7.66 (1H, m), 8.02 (1H, s), 8.12 (1H, d), 8.17 (1H, d), 8.26 (1H, s), 8.43 (1H, d)。 步驟6. 5-(5-(2-(1-((2-氨基-4-溴苯基)氨基)-3-氮雜雙環[3.2.1]辛-3-基)乙氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-甲酸甲酯(INT-A60-6)的製備。將5-[5-(2-{1-[(4-溴-2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(240 mg,0.39 mmol,1.00當量)、NH 2NH 2 .H 2O(39 mg,0.78 mmol,2當量)和雷尼鎳(33 mg,0.39 mmol,1當量)於甲醇(2 mL)中的混合物在氮氣氣氛下在0℃下攪拌2 h。將混合物過濾。將濾餅用甲醇(2×50 mL)洗滌。將所得溶液在減壓下濃縮。將殘餘物負載於矽膠柱上並且使用二氯甲烷/甲醇(10/1)洗脫,以得到呈黃色固體的200 mg 5-(5-(2-(1-((2-氨基-4-溴苯基)氨基)-3-氮雜雙環[3.2.1]辛-3-基)乙氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-甲酸甲酯(INT-A60-6)(84%)。LCMS: m/z (ESI), [M+H] +=585.10。 1H NMR (DMSO-d 6, 400 MHz) δ 1.43-1.51 (4H, m), 1.63 -1.75 (1H, m), 1.77-1.81 (1H, m), 1.87-1.91 (1H, m), 1.95-1.99 (1H, m), 2.01-2.05 (1H, m), 2.09-2.13 (1H, m), 2,67-2.77 (3H, m), 3.03-3.09 (1H, m), 3.58 (3H, s), 3.71(3H, s), 3.79 (3H, s), 3.97-4.14 (2H, m), 4.87 (2H, s), 6.38 (1H, d), 6.46-6.56 (1H, m), 6.66 (1H, d), 8.00 (1H, s), 8.13 (1H, d), 8.45 (1H, d)。 步驟7. 5-(5-{2-[1-(2-氨基-5-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A60-7)的製備。將5-(5-{2-[1-(2-氨基-5-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(200 mg,0.32 mmol,1當量)和BrCN(38 mg,0.36 mmol,1.1當量)於乙醇(2 mL)中的混合物在氮氣氣氛下在60℃下攪拌2 h。將混合物冷卻到室溫,用水(50 mL)處理,並且用二氯甲烷(2×50 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用(二氯甲烷/甲醇12/1)洗脫進行純化,以得到呈棕色固體的150 mg 5-(5-{2-[1-(2-氨基-5-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A60-7)(75%)。LCMS: m/z (ESI), [M+H] +=611.90。 1H NMR (DMSO-d 6, 400 MHz) δ 1.04-1.07 (1H, m), 1.22-1.26 (1H, m), 1.48-1.53 (1H, m), 1.71-1.84 (2H, m), 2.20-2.24 (2H, m), 2.27-2.33 (2H, m), 2.62 -2.76 (2H, m), 2.76 -2.84 (1H, m), 3.02 -3.08 (1H, m), 3.58 (3H, s), 3.73 (3H, s), 3.77 (3H, s), 4.09-4.13 (2H, m), 6.08 (2H, s), 6.93-6.97 (1H, m), 7.19 (1H, d), 7.25 (1H, d), 8.01 (1H, s), 8.11 (1H, d), 8.42 (1H, d)。 步驟8. 5-(5-{2-[1-(2-氨基-5-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A60-8)的製備。將5-(5-{2-[1-(2-氨基-5-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯甲酯(140 mg,0.22 mmol,1當量)和LiOH .H 2O(48 mg,1.14 mmol,5當量)於四氫呋喃/H 2O(2 mL/0.5 mL)中的混合物在氮氣氣氛下在60℃下攪拌2 h。將混合物冷卻到室溫並濃縮。將殘餘物通過C18柱使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的100 mg 5-(5-{2-[1-(2-氨基-5-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A60-8)(73%)。LCMS: m/z (ESI), [M+H] +=595.95。 1H NMR (DMSO-d 6, 400 MHz) δ 1.54 (1H, m), 1.73-1.79 (2H, m), 2.15-2.21 (1H, m), 2.28-2.34 (2H, m), 2.63-2.69 (3H, m), 2.72-2.78 (1H, m), 2.85-2.91 (2H, m), 3.05-3.11 (1H, m), 3.49 (3H, s), 3.68 (3H, s), 4.01-4.19 (2H, m), 6.29 (2H, s), 6.90-6.99 (1H, m), 7.22 (1H, d), 7.27 (1H, d), 7.95 (2H, d), 8.24 (1H, d)。 步驟9. 6-溴-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33), 18(22),19-八烯-12,16-二酮(實施例A59)的製備。將5-(5-{2-[1-(2-氨基-5-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(90 mg,0.15 mmol,1當量)、 N,N-二異丙基乙胺(58 mg,0.45 mmol,3當量)和 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(114 mg,0.30 mmol,2當量)與1,4-二噁烷(2 mL)中的混合物在氮氣氣氛下在60℃下攪拌30分鐘。將混合物冷卻到室溫。將反應用水(30 mL)淬滅,並且將混合物用二氯甲烷(2×30 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(12/1)洗脫進行純化,以得到呈白色固體的80 mg 6-溴-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮(實施例A59)(91%)。LCMS: m/z (ESI), [M+H] +=580.20。 1H NMR (DMSO-d 6, 400 MHz) δ δ 1.53-1.77 (3H, m), 2.00 (1H, d), 2.39-2.63 (3H, m), 2.64-2.77 (2H, m), 3.16-3.28 (1H, m), 3.29-3.39 (2H, m), 3.62 (3H, s), 3.63-3.69 (1H, m), 3.72 (3H, s), 4.45-4.70 (2H, m), 7.28 (1H, d), 7.72 (1H, d), 8.02 (1H, s), 8.39-8.43 (2H, m), 8.78 (1H, s), 12.81 (1H, s)。 步驟10. 15,21-二甲基-6-(4-甲基哌嗪-1-基)-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33), 18(22),19-八烯-12,16-二酮(A60)的製備。在室溫下向6-溴-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮(75 mg,0.13 mmol,1當量)、1-甲基哌嗪(38 mg,0.39 mmol,3當量)和BrettPhos Pd G 3(35 mg,0.03 mmol,0.3當量)與1,4-二噁烷(3 mL)中的攪拌混合物中添加LiHMDS(1.3 mL,1.30 mmol,10當量)。將所得混合物在氮氣氣氛下在60℃下攪拌2 h,並且冷卻到室溫。將反應用飽和NH 4Cl(水溶液)溶液(50 mL)淬滅,並且將混合物用二氯甲烷(2×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(10/1)洗脫進行純化,以得到呈黃色固體的22.3 mg 15,21-二甲基-6-(4-甲基哌嗪-1-基)-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}. 1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13, 17(33),18(22),19-八烯-12,16-二酮(A60)(28%)。LCMS: m/z (ESI), [M+H] +=598.35。 1H NMR (DMSO-d 6, 400 MHz) δ 1.50-1.80 (3H, m), 1.97 (1H, d), 2.26-2.58 (10H, m), 2.55-2.80 (3H, m), 3.03-3.43 (6H, m), 3.63 (3H, s), 3.64-3.70 (1H, m), 3.72 (3H, s), 4.57 (1H, t), 4.69 (1H, d), 6.86 (1H, dd), 7.17 (1H, s), 7.57 (1H, d), 8.29 (1H, d), 8.42 (1H, s), 8.83 (1H, d), 12.56 (1H, s)。 步驟11. 15,21-二甲基-6-(4-甲基哌嗪-1-基)-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33), 18(22),19-八烯-12,16-二酮,異構體1(實施例A60A)和異構體2(實施例A60B)的製備。將15,21-二甲基-6-(4-甲基哌嗪-1-基)-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮(16.9 mg,0.02 mmol)的外消旋混合物通過Pre-SFC使用手性ART纖維素-SB柱(2*25 cm,5 μm)使用含有0.1%三乙胺的叔丁基甲基醚作為流動相A和乙醇/二氯甲烷(1: 1)作為流動相B來分離,以得到呈黃色固體的1 mg異構體1(5%)和1 mg異構體2(5%)。 異構體1:LCMS: m/z (ESI), [M+H] +=598.40。 1H NMR (DMSO-d 6, 400 MHz) δ 1.50-1.80 (3H, m), 1.97 (1H, d), 2.26-2.58 (10H, m), 2.55-2.80 (3H, m), 3.03-3.43 (6H, m), 3.63 (3H, s), 3.64-3.70 (1H, m), 3.72 (3H, s), 4.55 (1H, t), 4.69 (1H, d), 6.84 (1H, dd), 7.16 (1H, s), 7.55 (1H, d), 8.28 (1H, d), 8.41 (1H, s), 8.83 (1H, d), 12.54 (1H, s)。手性-HPLC,Rt=3.374分鐘 異構體2:LCMS: m/z (ESI), [M+H] +=598.35。 1H NMR (DMSO-d 6, 400 MHz) δ 1.50-1.80 (3H, m), 1.97 (1H, d), 2.26-2.58 (10H, m), 2.55-2.80 (3H, m), 3.03-3.43 (6H, m), 3.63 (3H, s), 3.64-3.70 (1H, m), 3.72 (3H, s), 4.55 (1H, t), 4.69 (1H, d), 6.85 (1H, dd), 7.16 (1H, s), 7.56 (1H, d), 8.28 (1H, d), 8.41 (1H, s), 8.83 (1H, d), 12.55 (1H, s)。手性-HPLC,Rt=3.923分鐘 實施例A61A和實施例A61B 15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮,異構體1(實施例A61A)和異構體2(實施例A61B) 步驟1. 1-[(2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛烷-3-甲酸叔丁酯(INT-A61-1)的製備。將1-氨基-3-氮雜雙環[3.2.1]辛烷-3-甲酸叔丁酯(500 mg,2.21 mmol,1當量)、鄰氟硝基苯(312 mg,2.20 mmol,1當量)和K 2CO 3(610.66 mg,4.42 mmol,2當量)於二甲亞碸(10 mL)中的混合物在氮氣氣氛下在120℃下攪拌過夜,並且冷卻到室溫。將反應用水(50 mL)淬滅,並且將混合物用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用石油醚/乙酸乙酯(5/1)洗脫來進行純化,以得到呈橙色油的600 mg 1-[(2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛烷-3-甲酸叔丁酯(INT-A61-1)(78%)。LCMS: m/z (ESI), [M+H] += 291.95。 步驟2. N-(2-硝基苯基)-3-氮雜雙環[3.2.1]辛-1-胺(INT-A61-2)的製備。將1-[(2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛烷-3-甲酸叔丁酯(550 mg,1.58 mmol,1當量)於三氟乙酸(2 mL)和二氯甲烷(6 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將混合物用水(50 mL)和飽和NaHCO 3(水溶液)處理。將混合物用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥,並且在減壓下濃縮,以得到呈橙色油的380 mg N-(2-硝基苯基)-3-氮雜雙環[3.2.1]辛-1-胺(INT-A61-2)。此材料在不經進一步純化的情況下用於下一反應中。LCMS: m/z (ESI), [M+H] +=248.00。 步驟3. 3-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}- N-(2-硝基苯基)-3氮雜雙環[3.2.1]辛-1-胺(INT-A61-3)的製備。將 N-(2-硝基苯基)-3-氮雜雙環[3.2.1]辛-1-胺(380 mg,1.54 mmol,1當量)、(2-溴乙氧基)(叔丁基)二甲基矽烷(735 mg,3.07 mmol,2當量)和K 2CO 3(425 mg,3.07 mmol,2當量)於 N,N-二甲基甲醯胺(5 mL)中的混合物在氮氣氣氛下在60℃下攪拌2 h,並且冷卻到室溫。將反應用水(50 mL)淬滅,並且將混合物用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用石油醚/乙酸乙酯(5/1)洗脫來進行純化,以得到呈橙色油的450 mg 3-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}- N-(2-硝基苯基)-3-氮雜雙環[3.2.1]辛-1-胺(INT-A61-3)(72%)。LCMS: m/z (ESI), [M+H] +=406.15。 步驟4. 2-{1-[(2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙醇(INT-A61-4)的製備。將3-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}- N-(2-硝基苯基)-3-氮雜雙環[3.2.1]辛-1-胺(400 mg,1 mmol,1當量)和四丁基氟化銨(516 mg,2 mmol,2當量)於四氫呋喃(10 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將反應用飽和NaHCO 3(水溶液)(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥,並且在減壓下濃縮,以得到呈橙色油的280 mg 2-{1-[(2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙醇(INT-A61-4)。此材料在不經進一步純化的情況下用於下一反應中。LCMS: m/z (ESI), [M+H] +=292.00。 步驟5. 1-甲基-5-[1-甲基-5-(2-{1-[(2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙氧基)吡唑-4-基]-6-氧代吡啶-3-甲酸甲酯(INT-A61-5)的製備。在氮氣氣氛下在0℃下向2-{1-[(2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙醇(330 mg,1.13 mmol,1當量)、偶氮二甲酸二異丙酯(687 mg,3.40 mmol,3當量)和三苯基膦(891 mg,3.40 mmol,3當量)於四氫呋喃(10 mL)中的攪拌混合物中添加5-(5-羥基-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M2,298 mg,1.13 mmol,1當量)。在氮氣氣氛下在0℃下攪拌2 h後,將反應用水(100 mL)淬滅,並且將混合物用二氯甲烷(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用石油醚/乙酸乙酯(1/1)洗脫進行純化,以得到呈橙色固體的400 mg 1-甲基-5-[1-甲基-5-(2-{1-[(2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙氧基)吡唑-4-基]-6-氧代吡啶-3-甲酸甲酯(INT-A61-5)(66%)。 步驟6. 5-[5-(2-{1-[(2-氨基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A61-6)的製備。在0℃下向1-甲基-5-[1-甲基-5-(2-{1-[(2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙氧基)吡唑-4-基]-6-氧代吡啶-3-甲酸甲酯(340 mg,0.634 mmol,1當量)和雷尼鎳(54.29 mg,0.634 mmol,1.0當量)於甲醇(20 mL)中的攪拌混合物中添加NH 2NH 2 .H 2O(63.44 mg,1.268 mmol,2.0當量)。將混合物在氮氣氣氛下在室溫下攪拌1 h並且過濾。將濾餅用甲醇(3×10 mL)洗滌。將所得溶液在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(12/1)洗脫進行純化,以得到呈黃色固體的240 mg 5-[5-(2-{1-[(2-氨基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A61-6)(63%)。LCMS: m/z (ESI), [M+H] +=507.25。 步驟7. 5-(5-{2-[1-(2-氨基-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A61-7)的製備。將5-[5-(2-{1-[(2-氨基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(300 mg,0.59 mmol,1當量)和BrCN(94 mg,0.89 mmol,1.5當量)於乙醇(5 mL)中的混合物在氮氣氣氛下在室溫下攪拌過夜。將混合物在減壓下濃縮,並且將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(12/1)洗脫進行純化,以得到呈棕黃色固體的200 mg 5-(5-{2-[1-(2-氨基-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A61-7)(64%)。LCMS: m/z (ESI), [M+H] +=532.25。 步驟8. 5-(5-(2-(-1-(2-氨基-1H-苯並[d]咪唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基)乙氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-甲酸甲酯,異構體1(INT-A61-7A)和異構體2(INT-A61-7B)的製備。將5-(5-{2-[1-(2-氨基-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(200 mg)的外消旋混合物使用SFC DZ-CHIRALPAK IC-3柱(4.6*50 mm,3.0 μm)、含有0.2%二乙胺的叔丁基甲醚作為流動相A和乙醇/二氯甲烷(1/1)作為流動相B來分離,以得到呈黃色固體(35%)的70 mg異構體1(35%)和70 mg異構體2(35%)。 步驟9. 5-(5-{2-[1-(2-氨基-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A61-8A)的製備。將5-(5-{2-[1-(2-氨基-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A61-7A,70 mg,0.13 mmol,1當量)和LiOH .H 2O(22 mg,0.53 mmol,4當量)於水(1 mL)和四氫呋喃(4 mL)中的溶液在氮氣氣氛下在室溫下攪拌過夜。將混合物在減壓下濃縮。將殘餘物通過prep-HPLC在XBridge Shield RP18 OBD柱上,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3 .H 2O的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃色固體的45 mg 5-(5-{2-[1-(2-氨基-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A61-8A)(70%)。LCMS: m/z (ESI), [M+H] +=518.10。 步驟10. 5,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮,異構體1(實施例A61A)的製備。將5-(5-{2-[1-(2-氨基-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(45 mg,0.087 mmol,1當量)、 N,N-二異丙基乙胺(33.9,0.261 mmol,3當量)和 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(66 mg,0.17 mmol,2當量)於二噁烷(2 mL)中的混合物在氮氣氣氛下在室溫下攪拌過夜。在將混合物在減壓下濃縮後,將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(12/1)洗脫進行純化,以得到呈灰白色固體的26.8 mg 15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮,異構體1(實施例A61A)(61%)。LCMS: m/z (ESI), [M+H] += 500.15。 1H NMR (DMSO- d 6, 400 MHz) δ 1.53-1.77 (3H, m), 2.00 (1H, d), 2.39-2.63 (3H, m), 2.64-2.77 (2H, m), 3.16-3.28 (1H, m), 3.29-3.39 (2H, m), 3.62 (3H, s), 3.63-3.69 (1H, m), 3.72 (3H, s), 4.55 (1H, t), 4.68 (1H, d), 7.12-7.23 (2H, m), 7.55-7.60 (1H, m), 7.69-7.75 (1H, m), 8.29 (1H, d), 8.41 (1H, d), 8.83 (1H, d), 12.76 (1H, s)。 步驟11. 5-(5-{2-[1-(2-氨基-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A61-8B)的製備。將5-(5-{2-[1-(2-氨基-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A61-7B,70 mg,0.13 mmol,1當量)和LiOH .H 2O(22 mg,0.53 mmol,4當量)於水(1 mL)和四氫呋喃(4 mL)中的混合物在氮氣氣氛下在室溫下攪拌過夜。將混合物在減壓下濃縮後,將殘餘物通過prep-HPLC用XBridge Shield RP18 OBD柱,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3.H 2O的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈灰白色固體的45 mg 5-(5-{2-[-1-(2-氨基-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A61-8B)(70%)。LCMS: m/z (ESI), [M+H] += 518.15。 步驟12. 15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮、(異構體2 實施例A61B)的製備。將5-(5-{2-[1-(2-氨基-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(45 mg,0.087 mmol,1當量)、 N,N-二異丙基乙胺(33.9,0.261 mmol,3當量)和 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(66 mg,0.17 mmol,2當量)於二噁烷(2 mL)中的混合物在氮氣氣氛下在室溫下攪拌過夜。在將混合物在減壓下濃縮後,將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(12/1)洗脫進行純化,以得到呈灰白色固體的25.8 mg 15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮,異構體2(實施例A61B)(57%)。LCMS: m/z (ESI), [M+H] += 500.15。 1H NMR (DMSO- d 6, 400 MHz) δ 1.53-1.77 (3H, m), 2.00 (1H, d), 2.39-2.63 (3H, m), 2.64-2.77 (2H, m), 3.16-3.28 (1H, m), 3.29-3.39 (2H, m), 3.62 (3H, s), 3.63-3.69 (1H, m), 3.72 (3H, s), 4.55 (1H, t), 4.68 (1H, d), 7.12-7.23 (2H, m), 7.55-7.60 (1H, m), 7.69-7.75 (1H, m), 8.29 (1H, d), 8.41 (1H, d), 8.83 (1H, d), 12.76 (1H, s)。 實施例A62A 15,21-二甲基-5-(嗎啉-4-基)-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮,異構體1(實施例A62A) 步驟1. 15,21-二甲基-5-(嗎啉-4-基)-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33), 18(22),19-八烯-12,16-二酮,異構體1(實施例A62A)的製備。在氮氣氣氛下在室溫下向5-溴-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33), 18(22),19-八烯-12,16-二酮(M4A,50 mg,0.09 mmol,1當量)、嗎啉(30 mg,0.34 mmol,4當量)和BrettPhos Pd G 3(23 mg,0.03 mmol,0.3當量)於二噁烷(10 mL)中的攪拌混合物中逐滴添加LiHMDS(0.34 mL,0.34 mmol,4當量)。將反應混合物在60℃下攪拌30分鐘。在冷卻到室溫後,將反應用飽和NH 4Cl溶液(30 mL)淬滅,並且將混合物用二氯甲烷(3×20 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(20/1)洗脫進行純化,以得到呈白色固體的22 mg 15,21-二甲基-5-(嗎啉-4-基)-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}. 1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13, 17(33),18(22),19-八烯-12,16-二酮,異構體1(實施例A62A)(43%)。LCMS: m/z (ESI), [M+H] +=585.35。 1H NMR (DMSO-d 6, 400 MHz) δ 1.53-1.82 (3H, m), 2.02 (1H, d), 2.39-2.61 (3H, m), 2.65-2.77 (3H, m), 3.10 (4H, q), 3.17-3.43 (2H, m), 3.62 (3H, s), 3.63-3.69 (1H, m), 3.72 (3H, s), 3.77 (4H, t), 4.54 (1H, t), 4.69 (1H, d), 6.91 (1H, dd), 7.20 (1H, s), 7.44 (1H, d), 8.28 (1H, d), 8.42 (1H, s), 8.83 (1H, d), 12.57 (1H, s)。 使用上文針對實施例A62A的合成描述的類似反應條件,由中間體M4A製備以下實施例: 實施例A63A、實施例A64A、實施例A65A、實施例A66A、實施例A66C、實施例A67A、實施例A68A、實施例A69A、實施例A70A、實施例A71A、實施例A72A、實施例A73A、實施例A76A、實施例A77A、實施例A78A、實施例A79A、實施例A81A、實施例A82A、實施例A83A、實施例A85A、實施例A86A、實施例A87A、實施例A88A、實施例A89A、實施例A90A、實施例A91A、實施例A92A、實施例A95A、實施例A97A、實施例A98A、實施例A99A、實施例A104A、實施例A105A、實施例A106A、實施例A107A、實施例A108A。實施例A109A. 結構以及m/z和 1H NMR的資料列於表1中。 實施例A62B 15,21-二甲基-5-(嗎啉-4-基)-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮,異構體2(實施例A62B) 步驟1. 15,21-二甲基-5-(嗎啉-4-基)-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33), 18(22),19-八烯-12,16-二酮,異構體2(實施例A62B)的製備。在氮氣氣氛下在室溫下向5-溴-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33), 18(22),19-八烯-12,16-二酮(M4B,50 mg,0.09 mmol,1當量)、嗎啉(30 mg,0.34 mmol,4當量)和BrettPhos Pd G 3(23 mg,0.03 mmol,0.3當量)於二噁烷(5 mL)中的攪拌混合物中逐滴添加LiHMDS(0.34 mL,0.34 mmol,4當量)。將反應混合物在60℃下攪拌30分鐘。在冷卻到室溫後,將反應用飽和NH 4Cl溶液(30 mL)淬滅,並且將混合物用二氯甲烷(3×20 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(20/1)洗脫進行純化,以得到呈白色固體的6.4 mg 15,21-二甲基-5-(嗎啉-4-基)-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}. 1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13, 17(33),18(22),19-八烯-12,16-二酮,異構體2(實施例A62B)(12%)。LCMS: m/z (ESI), [M+H] +=585.35。 1H NMR (DMSO-d 6, 400 MHz) δ 1.53-1.82 (3H, m), 2.02 (1H, d), 2.39-2.61 (3H, m), 2.65-2.77 (3H, m), 3.10 (4H, q), 3.17-3.43 (2H, m), 3.62 (3H, s), 3.63-3.69 (1H, m), 3.72 (3H, s), 3.77 (4H, t), 4.54 (1H, t), 4.69 (1H, d), 6.91 (1H, dd), 7.20 (1H, s), 7.44 (1H, d), 8.28 (1H, d), 8.42 (1H, s), 8.83 (1H, d), 12.57 (1H, s)。 使用上文針對實施例A62B的合成描述的類似反應條件,由中間體M4B製備以下實施例: 實施例A63B、實施例A64B、實施例A65B、實施例A66B、實施例A66D、實施例A67B、實施例A68B、實施例A69B、實施例A70B、實施例A71B、實施例A72B、實施例A73B、實施例A76B、實施例A77B、實施例A78B、實施例A79B、實施例A81B、實施例A82B、實施例A83B、實施例A85B、實施例A86B、實施例A87B、實施例A88B、實施例A89B、實施例A90B、實施例A91B、實施例A92B、實施例A95B、實施例A97B、實施例A98B、實施例A99B、實施例A104B、實施例A105B、實施例A106B、實施例A107B、實施例A108B。實施例A109B. 結構以及m/z和 1H NMR的資料列於表1中。 實施例A74 15,21-二甲基-5-(嗎啉-4-基)-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]三十四-3,5,7,9,13,17(34),18(22),19-八烯-12,16-二酮 步驟1. N-(5-溴-2-硝基苯基)-3-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-3-氮雜雙環[3.2.2]壬-1-胺(INT-A74-1)的製備。將3-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-3-氮雜雙環[3.2.2]壬-1-胺(1 g,3.35 mmol,1當量)、4-溴-2-氟-1-硝基苯(1.84 g,8.37 mmol,2.5當量)和K 2CO 3(1.39 g,10.05 mmol,3當量)於二甲基亞碸(10 mL)中的混合物在氮氣氣氛下在120℃下攪拌4 h。將混合物冷卻到室溫。將反應用水(50 mL)淬滅,並且將混合物用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法使用石油醚/乙酸乙酯(5/1)洗脫來進行純化,以得到呈黃色油的1.5 g N-(5-溴-2-硝基苯基)-3-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-3-氮雜雙環[3.2.2]壬-1-胺(INT-A74-1) (85%)。LCMS: m/z (ESI), [M+H]+=498.15。 步驟2. 2-{1-[(5-溴-2-硝基苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙醇(INT-A74-2)的製備。將 N-(5-溴-2-硝基苯基)-3-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-3-氮雜雙環[3.2.2]壬-1-胺(1.5 g,3.00 mmol,1當量)和四丁基氟化銨(1.57 g,6.01 mmol,2當量)於四氫呋喃(15 mL)中的溶液在氮氣氣氛下在室溫下攪拌2 h。將反應用飽和NaHCO 3(水溶液)(100 mL)淬滅,並且將混合物用二氯甲烷(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上使用二氯甲烷/甲醇(10/1)洗脫來進行純化,以得到呈黃色油的1.2 g 2-{1-[(5-溴-2-硝基苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙醇(INT-A74-2)(93%)。LCMS: m/z (ESI), [M+H] +=384.00。 步驟3. 5-[5-(2-{1-[(5-溴-2-硝基苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A74-3)的製備。在0℃下向5-(5-羥基-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M2,0.66 g,2.49 mmol,0.8當量)和三苯基膦(2.46 g,9.369 mmol,3當量)於四氫呋喃(10 mL)中的攪拌混合物中添加偶氮二甲酸二異丙酯(1.89 g,9.36 mmol,3當量)和2-{1-[(5-溴-2-硝基苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙醇(1.20 g,3.11 mmol,1當量)。將混合物在氮氣氣氛下在0℃下攪拌2 h。將反應用水(50 mL)淬滅,並且將混合物用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC進行純化並且使用石油醚/乙酸乙酯(1/1)洗脫,以得到呈黃色油的1.2 g 5-[5-(2-{1-[(5-溴-2-硝基苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A74-3)(57%)。LCMS: m/z (ESI), [M+H] +=629.25。 1H NMR (DMSO-d 6, 400 MHz) δ 1.58-1.76 (9H, m), 2.56-2.79 (2H, m), 2.90 (4H, q), 3.56 (3H, s), 3.72 (6H, d), 4.13 (2H, t), 6.70-6.85 (1H, m), 7.01 (1H, d), 7.95-8.12 (4H, m), 8.28 (1H, d)。 步驟4. 5-[5-(2-{1-[(2-氨基-5-溴苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A74-4)的製備。在氮氣氣氛下在0℃下向雷尼鎳(0.90 g)和5-[5-(2-{1-[(5-溴-2-硝基苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(1.1 g,1.74 mmol,1當量)於甲醇(8 mL)中的攪拌混合物中添加NH 2NH 2.H 2O(47 mg,0.95 mmol,2當量)。將混合物在0℃下攪拌1 h。將混合物過濾,並且將濾餅用甲醇(2×20 mL)洗滌。將所得溶液在減壓下濃縮。將殘餘物通過Prep-TLC進行純化並且使用二氯甲烷/甲醇(10/1)洗脫,以得到呈淡黃色固體的910 mg 5-[5-(2-{1-[(2-氨基-5-溴苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A74-4)(76%)。LCMS: m/z (ESI), [M+H] += 599.20。 步驟5. 5-(5-{2-[1-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.2]壬-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A74-5)的製備。將5-[5-(2-{1-[(2-氨基-5-溴苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(900 mg,1.50 mmol,1當量)和BrCN(174 mg,1.65 mmol,1.1當量)於乙醇(10 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將所得混合物在真空下濃縮。將殘餘物通過Prep-TLC進行純化並且使用二氯甲烷/甲醇(12/1)洗脫,以得到呈淡黃色固體的640 mg 5-(5-{2-[1-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.2]壬-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A74-5)(68%)。LCMS: m/z (ESI), [M+H] +=624.10。 1H NMR (DMSO-d 6, 400 MHz) δ 1.24 (1H, s), 1.78-1.95 (8H, m), 2.60-2.74 (2H, m), 2.85-2.95 (4H, m), 3.57 (3H, s), 3.71 (6H, d), 4.08-4.17 (2H, m), 5.95 (2H, s), 7.04 (2H, s), 7.47 (1H, s), 7.96 (1H, s), 8.12 (1H, ), 8.39 (1H, d)。 步驟6. 5-(5-{2-[1-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.2]壬-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A74-6).將5-(5-{2-[1-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.2]壬-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(620 mg,0.99 mmol,1當量)和LiOH(28 mg,1.19 mmol,1.2當量)於四氫呋喃/H 2O(4 mL/1 mL)中的混合物在氮氣氣氛下在80℃下攪拌2 h。將混合物冷卻到室溫,並且在真空下濃縮,以得到呈白色固體的550 mg 5-(5-{2-[1-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.2]壬-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A74-6)(90%)。此材料在不經純化的情況下用於下一反應中。 步驟7. 5-溴-15,21-二甲基-23-氧雜-2,9,11,15,20,21, 26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]三十四-3,5,7,9,13,17(34),18(22),19-八烯-12,16-二酮(INT-A74-7)的製備。將5-(5-{2-[1-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.2]壬-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(550 mg,0.90 mmol,1當量)、 N,N,N,N-四甲基-O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(1027 mg,2.70 mmol,3當量)和 N,N-二異丙基乙胺(174 mg,1.35 mmol,1.5當量)於二噁烷(6 mL)中的混合物在氮氣氣氛下在60℃下攪拌2 h。將混合物冷卻到室溫,並且在真空下濃縮。將殘餘物通過prep-HPLC用XBridge Prep OBD C18柱,使用含有10 mmol/L NH 4HCO 3的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃色固體的500 mg 5-溴-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十四-3,5,7,9,13,17(34),18(22),19-八烯-12,16-二酮(INT-A74-7)(88%)。LCMS: m/z (ESI), [M+H] +=592.15。 步驟8. 15,21-二甲基-5-(嗎啉-4-基)-23-氧雜-2,9,11, 15,20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十四-3,5,7,9,13,17(34), 18(22),19-八烯-12,16-二酮(實施例A74)的製備。在氮氣氣氛下在室溫下向5-溴-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十四-3,5,7,9,13,17(34), 18(22),19-八烯-12,16-二酮(130 mg,0.21 mmol,1當量)、嗎啉(57 mg,0.65 mmol,3當量)和BrettPhos Pd G 3(59 mg,0.06 mmol,0.3當量)與1,4-二噁烷(12 mL)中的攪拌混合物中添加LiHMDS(5.2 mL,5.20 mmol,6當量)。將混合物在60℃下攪拌1 h,並且冷卻到室溫。將反應用飽和NH 4Cl(水溶液)(15 mL)淬滅,並且將混合物用CH 2Cl 2(3×30 mL)萃取。將合併的有機層經無水Na 2SO 4乾燥,並且在減壓下濃縮。將殘餘物通過Prep-TLC進行純化並且使用亞甲基二氯/甲醇(12/1)洗脫,以得到呈白色固體的38.3 mg 15,21-二甲基-5-(嗎啉-4-基)-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]三十四-3,5,7,9,13,17(34),18(22),19-八烯-12,16-二酮(實施例A74)(28%)。LCMS: m/z (ESI), [M+H] += 599.25。 1H NMR (DMSO-d 6, 400 MHz) δ 1.82-2.02 (4H, m), 2.02-2.12 (3H, m), 2.80 (2H, d), 2.87 (2H, t), 3.08 (4H, t), 3.38-3.52 (4H, m), 3.62 (3H, s), 3.75 (3H, s), 3.77-3.85 (4H, m), 4.28 (2H, t), 6.93 (1H, dd), 7.34 (1H, d), 7.49 (1H, d), 8.31 (2H, d), 9.12 (1H, d), 13.20 (1H, s)。 實施例A75 15,21-二甲基-5-(4-甲基哌嗪-1-基)-23-氧雜-2,9,11,15, 20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]三十四-3,5,7,9,13,17(34),18(22),19-八烯-12,16-二酮 步驟1. 15,21-二甲基-5-(4-甲基哌嗪-1-基)-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十四-3,5,7,9,13,17(34), 18(22),19-八烯-12,16-二酮(實施例A75)的製備。在氮氣氣氛下在室溫下向5-溴-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十四-3,5,7,9,13,17(34), 18(22),19-八烯-12,16-二酮(130 mg,0.21 mmol,1當量)、1-甲基哌嗪(65.93 mg,0.65 mmol,3當量)和BrettPhos Pd G 3(59.67 mg,0.06 mmol,0.3當量)與1,4-二噁烷(13 mL)中的攪拌混合物中添加LiHMDS(5.2 mL,5.2 mmol,6當量)。將混合物在60℃下攪拌1 h,並且冷卻到室溫。將混合物用飽和NH 4Cl(水溶液)(30 mL)處理,並且將混合物用二氯甲烷(3×20 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC進行純化並且使用亞甲基二氯/甲醇(12/1)洗脫,以得到呈淡黃色固體的31 mg 15,21-二甲基-5-(4-甲基哌嗪-1-基)-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十四-3,5,7,9,13,17(34),18(22),19-八烯-12,16-二酮(實施例A75)(22%)。LCMS: m/z (ESI), [M+H] +=612.30。 1H NMR (DMSO-d 6, 400 MHz) δ 1.85-1.99 (4H, m), 2.05-2.09 (3H, m), 2.24 (3H, t), 2.46-2.50 (4H, m), 2.80 (2H, d), 2.87 (2H, t), 3.09 (4H, t), 3.52-3.59 (4H, m), 3.62 (3H, s), 3.75 (3H, s), 4.28 (2H, t), 6.92 (1H, dd), 7.33 (1H, d), 7.47 (1H, d), 8.31 (2H, d), 9.12 (1H, d), 13.19 (1H, s)。 實施例A80A、實施例A80A1、實施例A80A2 15,21-二甲基-5-[3-(4-甲基哌嗪-1-基)吡咯烷-1-基]-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}. 1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13, 17(33),18(22),19-八烯-12,16-二酮,異構體A(實施例A80A)、異構體A1(實施例A80A1)和異構體A2(實施例A80A2) 步驟1. 15,21-二甲基-5-[3-(4-甲基哌嗪-1-基)吡咯烷-1-基]-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮,異構體A(實施例A80A)的製備。在氮氣氣氛下在0℃下向5-溴-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮(M4A,90 mg,0.15 mmol,1當量)、1-甲基-4-(吡咯烷-3-基)哌嗪(79.01 mg,0.46 mmol,3當量)和BrettPhos Pd G 3(42.31 mg,0.04 mmol,0.3當量)中的攪拌混合物中逐滴添加LiHMDS(3.6 mL,3.6 mmol,24當量)。將混合物在60℃下攪拌1 h,然後冷卻到室溫。將反應用飽和NH 4Cl溶液(水溶液)(10 mL)淬滅,並且將混合物用二氯甲烷(3×20 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-HPLC用XBridge Shield RP18 OBD柱,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3.H 2O的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃色固體的2.6 mg 15,21-二甲基-5-[3-(4-甲基哌嗪-1-基)吡咯烷-1-基]-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮(異構體A,實施例A80A)(2%)。LCMS: m/z (ESI), [M+H]+= 667.40。 1H NMR (DMSO-d 6, 400 MHz) δ 1.57-1.87 (4H, m), 2.00 (1H, s), 2.16 (3H, s), 2.23-2.63 (12H, m), 2.66-2.76 (2H, m), 2.91 (1H, t), 3.07 (1H, q), 3.20-3.59 (6H, m), 3.62 (3H, s), 3.63-3.69 (1H, m), 3.72 (3H, s), 4.57 (1H, t), 4.69 (1H, d), 6.49 (1H, dd), 6.75 (1H, s), 7.38 (1H, d), 8.26 (1H, d), 8.41 (1H, s), 8.81 (1H, d), 12.51 (1H, s)。SFC-HPLC,Rt=1.029分鐘。 步驟2. 15,21-二甲基-5-[3-(4-甲基哌嗪-1-基)吡咯烷-1-基]-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮,異構體A1(實施例A80A1)和異構體A2(實施例A80A2)的製備。將15,21-二甲基-5-[3-(4-甲基哌嗪-1-基)吡咯烷-1-基]-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}. 1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13, 17(33),18(22),19-八烯-12,16-二酮(實施例A80A,20 mg,0.17 mmol)的混合物通過Prep-手性-HPLC使用XBridge Shield RP18 OBD柱(30*150 mm,5μm)使用含有10 mmol/L NH 4HCO 3和0.1% NH 3.H 2O的水和乙腈作為移動相來分離,以得到呈黃色固體的8.5 mg異構體A1(實施例A80A1,18%)和3.2 mg異構體A2(實施例A80A2,6%)。 異構體A1:LCMS: m/z (ESI), [M+H] +=667.45。 1H NMR (DMSO-d 6, 400 MHz) δ 1.57-1.87 (4H, m), 2.00 (1H, s), 2.16 (3H, s), 2.23-2.63 (12H, m), 2.66-2.76 (2H, m), 2.91 (1H, t), 3.07 (1H, q), 3.20-3.59 (6H, m), 3.62 (3H, s), 3.63-3.69 (1H, m), 3.72 (3H, s), 4.57 (1H, t), 4.69 (1H, d), 6.49 (1H, dd), 6.76 (1H, s), 7.38 (1H, d), 8.26 (1H, d), 8.42 (1H, s), 8.82 (1H, d), 12.53 (1H, s)。手性-HPLC,Rt=5.235分鐘。 異構體A2:LCMS: m/z (ESI), [M+H]+=667.45。 1H NMR (DMSO-d 6, 400 MHz) δ 1.57-1.87 (4H, m), 2.00 (1H, s), 2.16 (3H, s), 2.23-2.63 (12H, m), 2.66-2.76 (2H, m), 2.91 (1H, t), 3.07 (1H, q), 3.20-3.59 (6H, m), 3.62 (3H, s), 3.63-3.69 (1H, m), 3.72 (3H, s), 4.57 (1H, t), 4.69 (1H, d), 6.47 (1H, dd), 6.76 (1H, s), 7.37 (1H, d), 8.26 (1H, d), 8.42 (1H, s), 8.82 (1H, d), 12.49 (1H, s)。手性-HPLC,Rt=6.080分鐘 實施例A80B、實施例A80B1、實施例A80B2 15,21-二甲基-5-[3-(4-甲基哌嗪-1-基)吡咯烷-1-基]-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}. 1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13, 17(33),18(22),19-八烯-12,16-二酮,異構體B(實施例A80B)、異構體B1(實施例A80B1)和異構體B2(實施例A80B2) 步驟1. 15,21-二甲基-5-[3-(4-甲基哌嗪-1-基)吡咯烷-1-基]-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮,異構體B(實施例A80B)的製備。在氮氣氣氛下在0℃下向5-溴-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮(M4B,90 mg,0.15 mmol,1當量)、1-甲基-4-(吡咯烷-3-基)哌嗪(79.01 mg,0.46 mmol,3當量)和BrettPhos Pd G 3(42.31 mg,0.04 mmol,0.3當量)中的攪拌混合物中逐滴添加LiHMDS(3.6 mL,3.60 mmol,24當量)。將混合物在60℃下攪拌1 h,然後冷卻到室溫。將反應用飽和NH 4Cl溶液(水溶液)(10 mL)淬滅,並且將混合物用二氯甲烷(3×20 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-HPLC使用XBridge Shield RP18 OBD柱,含有10 mmol/L NH 4HCO 3和0.1% NH 3.H 2O的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃色固體的1.6 mg 15,21-二甲基-5-[3-(4-甲基哌嗪-1-基)吡咯烷-1-基]-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}. 1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13, 17(33),18(22),19-八烯-12,16-二酮,異構體B(實施例A80B)(1%)。LCMS: m/z (ESI), [M+H] +=667.40 1H NMR (DMSO-d 6, 400 MHz) δ 1.57-1.87 (4H, m), 2.00 (1H, s), 2.16 (3H, s), 2.23-2.63 (12H, m), 2.66-2.76 (2H, m), 2.91 (1H, t), 3.07 (1H, q), 3.20-3.59 (6H, m), 3.62 (3H, s), 3.63-3.69 (1H, m), 3.72 (3H, s), 4.57 (1H, t), 4.69 (1H, d), 6.49 (1H, d), 6.75 (1H, s), 7.38 (1H, d), 8.26 (1H, d), 8.41 (1H, s), 8.81 (1H, d), 12.51 (1H, s)。SFC-HPLC,Rt=0.869, 步驟2. 15,21-二甲基-5-[3-(4-甲基哌嗪-1-基)吡咯烷-1-基]-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮,異構體B1(實施例A80B1)和異構體B2(實施例A80B2)的製備。將15,21-二甲基-5-[3-(4-甲基哌嗪-1-基)吡咯烷-1-基]-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}. 1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13, 17(33),18(22),19-八烯-12,16-二酮(實施例A80B,18 mg,0.17 mmol)的混合物通過Prep-手性-HPLC使用XBridge Prep OBD C18柱(30*150 mm,5μm)使用含有10 mmol/L NH4HCO3的水和乙腈作為移動相來分離,以得到呈黃色固體的2.4 mg異構體B1(實施例A80B1,5%)和1.2 mg異構體B2(實施例A80B2,2%)。 異構體B1:LCMS: m/z (ESI), [M+H] +=667。 1H NMR (DMSO-d 6, 400 MHz) δ 1.57-1.87 (4H, m), 2.00 (1H, s), 2.16 (3H, s), 2.23-2.63 (12H, m), 2.66-2.76 (2H, m), 2.91 (1H, t), 3.07 (1H, q), 3.20-3.59 (6H, m), 3.62 (3H, s), 3.63-3.69 (1H, m), 3.72 (3H, s), 4.57 (1H, t), 4.69 (1H, d), 6.49 (1H, d), 6.75 (1H, s), 7.38 (1H, d), 8.26 (1H, d), 8.41 (1H, s), 8.81 (1H, d), 12.51 (1H, s)。手性HPLC,Rt=3.906分鐘。 異構體B2:LCMS: m/z (ESI), [M+H] +=667.45。 1H NMR (DMSO-d 6, 400 MHz) δ 1.57-1.87 (4H, m), 2.00 (1H, s), 2.16 (3H, s), 2.23-2.63 (12H, m), 2.66-2.76 (2H, m), 2.91 (1H, t), 3.07 (1H, q), 3.20-3.59 (6H, m), 3.62 (3H, s), 3.63-3.69 (1H, m), 3.72 (3H, s), 4.57 (1H, t), 4.69 (1H, d), 6.49 (1H, d), 6.75 (1H, s), 7.38 (1H, d), 8.26 (1H, d), 8.41 (1H, s), 8.81 (1H, d), 12.51 (1H, s)。手性HPLC,Rt=5.026分鐘。 實施例A84A和實施例A84B 15,21-二甲基-5-[4-氟-3-( N, N-二甲基氨基)吡咯烷基]-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}. 1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13, 17(33),18(22),19-八烯-12,16-二酮,異構體1(實施例A84A)和異構體2(實施例A84B) 步驟1. 3-(二甲基氨基)-4-氟吡咯烷-1-甲酸叔丁酯(INT-A84-1)的製備。在氮氣氣氛下在0℃下向3-氨基-4-氟吡咯烷-1-甲酸叔丁酯(500 mg,2.44 mmol,1當量)、聚甲醛(4410.29 mg,48.96 mmol,20當量)和NaBH(OAc) 3(2075.35 mg,9.79 mmol,4當量)於Cl 2Cl 2(20 mL)中的攪拌混合物中逐滴添加三甲胺(990.90 mg,9.79 mmol,4當量)。將混合物在0℃下攪拌2 h。將反應用水(10 mL)淬滅,並且將混合物用CH 2Cl 2(3×20 mL)萃取。將合併的有機層用鹽水(3×20 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上使用石油CH 2Cl 2/甲醇(10/1)洗脫進行純化,以得到呈棕色油的500 mg 3-(二甲基氨基)-4-氟吡咯烷-1-甲酸叔丁酯(INT-A84-1)(79%)。 1H NMR (DMSO- d 6, 400 MHz) δ 1.40 (9H, s), 2.10-2.23 (5H, m), 2.85-2.95 (1H, m), 3.25 (1H, d), 3.47-3.64 (4H, m), 5.11-5.23 (1H, m)。 步驟2. 4-氟- N, N-二甲基吡咯烷-3-胺(INT-A84-2)的製備。將3-(二甲基氨基)-4-氟吡咯烷-1-甲酸叔丁酯(500 mg,2.152 mmol,1當量)和HCl於1,4-二噁烷(4 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。向此反應溶液中添加固體K 2CO 3。過濾後,將濾液在減壓下濃縮,並且得到呈無色油的300 mg 4-氟- N, N-二甲基吡咯烷-3-胺(INT-A84-2)。此材料在不經進一步純化的情況下用於下一反應中。 1H NMR (DMSO- d 6, 400 MHz) δ 2.05-2.25 (6H, m), 2.40-2.65 (2H, m), 2.76-3.48 (4H, m), 4.94-5.08 (1H, m) 步驟3. 15,21-二甲基-5-[4-氟-3-( N, N-二甲基氨基)吡咯烷基]-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮,異構體1(實施例A84A)和異構體2(實施例A84B)的製備。在氮氣氣氛下在0℃下向5-溴-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33), 18(22),19-八烯-12,16-二酮(M4A,150 mg,0.25 mmol,1.00當量)、BrettPhos Pd G 3(70.52 mg,0.07 mmol,0.3當量)和4-氟- N, N-二甲基吡咯烷-3-胺(102.83 mg,0.77 mmol,3當量)於1,4-二噁烷(15 mL)中的攪拌混合物中逐滴添加LiHMDS(6 mL,6.00 mmol,24當量)。將混合物在60℃下攪拌1 h,然後冷卻到室溫。將反應用飽和NH 4Cl(水溶液)溶液(15 mL)淬滅,並且將混合物用二氯甲烷(3×20 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-HPLC用XBridge Shield RP18 OBD柱,使用含有0.1 %NH 3.H 2O的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃色固體的9.8 mg 15,21-二甲基-5-[4-氟-3-( N, N-二甲基氨基)吡咯烷基]-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}. 1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13, 17(33),18(22),19-八烯-12,16-二酮,異構體1(實施例A84A)(6%)和呈黃色固體的10.0 mg異構體2(實施例A84B)(6%)。 異構體1:LCMS: m/z (ESI), [M+H] +=630.40。 1H NMR (DMSO-d 6, 400 MHz) δ 1.48-1.68 (4H, m), 2.01-2.08 (1H, m), 2.30 (6H, s), 2.41-2.58 (3H, m), 2.55-2.79 (4H, m), 3.04-3.10 (2H, m), 3.22-3.32 (1H, m), 3.54-3.69 (3H, m), 3.62 (3H, s), 3.72 (3H, s), 4.55 (1H, t), 4.62-4.73 (1H, m), 5.25-5.35 (1H, m), 6.50 (1H, dd), 6.76 (1H, s), 7.40 (1H, d), 8.26 (1H, d), 8.42 (1H, s), 8.82 (1H, d), 12.52 (1H, s)。SFC-HPLC,Rt=4.025分鐘。 異構體2:LCMS: m/z (ESI), [M+H] +=630.40。 1H NMR (DMSO-d 6, 400 MHz) δ 1.48-1.68 (4H, m), 2.01-2.08 (1H, m), 2.30 (6H, s), 2.41-2.58 (3H, m), 2.55-2.79 (4H, m), 3.04-3.10 (2H, m), 3.22-3.32 (1H, m), 3.54-3.69 (3H, m), 3.62 (3H, s), 3.72 (3H, s), 4.55 (1H, t), 4.62-4.73 (1H, m), 5.25-5.35 (1H, m), 6.59 (1H, dd), 6.86 (1H, s), 7.41 (1H, d), 8.27 (1H, d), 8.42 (1H, s), 8.82 (1H, d), 12.51 (1H, s)。SFC-HPLC,Rt=3.757分鐘。 實施例A84C和實施例A84D 15,21-二甲基-5-[4-氟-3-( N, N-二甲基氨基)吡咯烷基]-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}. 1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13, 17(33),18(22),19-八烯-12,16-二酮,異構體3(實施例A84C)和異構體4(實施例A84B) 步驟1. 15,21-二甲基-5-[4-氟-3-( N, N-二甲基氨基)吡咯烷基]-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮,異構體3(實施例A84C)和異構體4(實施例A84D)的製備。在氮氣氣氛下在0℃下向5-溴-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33), 18(22),19-八烯-12,16-二酮(M4B,150 mg,0.25 mmol,1當量)、4-氟- N, N-二甲基吡咯烷-3-胺(102.83 mg,0.77 mmol,3當量)和BrettPhos Pd G 3(70.52 mg,0.07 mmol,0.3當量)中的攪拌混合物中逐滴添加LiHMDS(6 mL,6.00 mmol,24當量)。將混合物在60℃下攪拌1 h,然後冷卻到室溫。將反應用飽和NH 4Cl(水溶液)溶液(10 mL)淬滅,並且將混合物用二氯甲烷(3×20 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-HPLC用XBridge Shield RP18 OBD柱,使用含有0.1% NH 3.H 2O的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃色固體的11.8 mg 15,21-二甲基-5-[4-氟-3-( N, N-二甲基氨基)吡咯烷基]-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮,異構體3(實施例A84C)(7%)和呈黃色固體的6.0 mg異構體4(實施例A84D)(3%)。 異構體3:LCMS: m/z (ESI), [M+H] +=630.40。 1H NMR (DMSO-d 6, 400 MHz) δ 1.48-1.68 (4H, m), 2.01-2.08 (1H, m), 2.30 (6H, s), 2.41-2.58 (3H, m), 2.55-2.79 (4H, m), 3.04-3.10 (2H, m), 3.22-3.32 (1H, m), 3.54-3.69 (3H, m), 3.62 (3H, s), 3.72 (3H, s), 4.55 (1H, t), 4.62-4.73 (1H, m), 5.25-5.35 (1H, m), 6.50 (1H, dd), 6.76 (1H, s), 7.40 (1H, d), 8.26 (1H, d), 8.42 (1H, s), 8.82 (1H, d), 12.53 (1H, s)。SFC-HPLC,Rt=3.983分鐘 異構體4:LCMS: m/z (ESI), [M+H] +=630.40。 1H NMR (DMSO-d 6, 400 MHz) δ 1.48-1.68 (4H, m), 2.01-2.08 (1H, m), 2.30 (6H, s), 2.41-2.58 (3H, m), 2.55-2.79 (4H, m), 3.04-3.10 (2H, m), 3.22-3.32 (1H, m), 3.54-3.69 (3H, m), 3.62 (3H, s), 3.72 (3H, s), 4.55 (1H, t), 4.62-4.73 (1H, m), 5.25-5.35 (1H, m), 6.59 (1H, dd), 6.86 (1H, s), 7.41 (1H, d), 8.27 (1H, d), 8.42 (1H, s), 8.82 (1H, d), 12.55 (1H, s)。SFC-HPLC,Rt=3.724分鐘。 實施例A94A、實施例94A1和實施例A94A2 5-[3-(3-氟氮雜環丁烷-1-基)吡咯烷-1-基]-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}. 1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13, 17(33),18(22),19-八烯-12,16-二酮,異構體A(實施例A94A)、異構體A1(實施例A94A1)和異構體A2(實施例A94A2) 步驟1. 3-(3-氟氮雜環丁烷-1-基)吡咯烷-1-甲酸叔丁酯(INT-A94A-1)的製備。將3-氧代吡咯烷-1-甲酸叔丁酯(1500 mg,8.10 mmol,1當量)和3-氟氮雜環丁烷鹽酸鹽(903 mg,8.10 mmol,1當量)於CH 2Cl 2(50 mL)中的混合物在空氣氣氛下在室溫下攪拌1 h。在0℃下在2分鐘內向上述混合物中分批添加NaBH(OAc) 3(8582 mg,40.49 mmol,5當量)。將所得混合物攪拌另外2 h。將反應用飽和NH 4Cl(水溶液)(50 mL)淬滅,並且將混合物用CH 2Cl 2(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上使用二氯甲烷/甲醇(12/1)洗脫來進行純化,以得到呈黃色油的1.5 g 3-(3-氟氮雜環丁烷-1-基)吡咯烷-1-甲酸叔丁酯(INT-A94A-1)(75%)。 1H NMR (CDCl 3, 400 MHz) δ 1.46 (9H, d), 1.68-1.83 (1H, m), 1.85-1.95 (1H, m), 3.05-3.15 (1H, m), 3.18-3.48 (6H, m), 3.72-3.87 (2H, m) 步驟2. 3-(3-氟氮雜環丁烷-1-基)吡咯烷(INT-A94A-2)的製備。將3-(3-氟氮雜環丁烷-1-基)吡咯烷-1-甲酸叔丁酯(500 mg,2.05 mmol,1當量)和三氟乙酸(3 mL)於CH 2Cl 2(9 mL)中的溶液在室溫下攪拌2 h。將混合物在減壓下濃縮,用二氯甲烷/甲醇(10/1)(30 mL)處理,並且用K 2CO 3調節到pH 7。將混合物過濾,並且將濾餅用甲醇(2×10 mL)洗滌。將所得溶液在減壓下濃縮,以得到呈黃色油的150 mg 3-(3-氟氮雜環丁烷-1-基)吡咯烷(INT-A94A-2)。此材料在不經進一步純化的情況下用於下一反應中。 1H NMR (DMSO-d 6, 400 MHz) δ 1.75-1.84 (2H, m), 2.70-2.81 (2H, m), 2.82-2.95 (2H, m), 3.05-3.21 (3H, m), 3.59-3.62 (2H, m), 4.98-5.18 (2H, m) 步驟3. 5-[3-(3-氟氮雜環丁烷-1-基)吡咯烷-1-基]-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮(異構體A,實施例A94A)的製備。向3-(3-氟氮雜環丁烷-1-基)吡咯烷(125 mg,0.86 mmol,5當量)、BrettPhos Pd G 3(47 mg,0.05 mmol,0.3當量)和5-溴-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33), 18(22),19-八烯-12,16-二酮(M4A,100 mg,0.173 mmol,1當量)於1,4-二噁烷(10 mL)中的攪拌混合物中添加LiHMDS(2.1 mL,2.07 mmol,12當量)。將所得混合物在氮氣氣氛下在60℃下攪拌2 h。將混合物冷卻到室溫。將反應用飽和NH 4Cl(水溶液)(15 mL)淬滅,並且將混合物用CH 2Cl 2(3×50 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(10:1)洗脫進行純化。將產物通過Prep-HPLC用XBridge Prep苯基OBD柱,使用含有0.1% NH 4HCO 3的水和乙腈作為流動相進行進一步純化。將含有期望化合物的級分蒸發到乾燥,以得到呈灰白色固體的13 mg 5-[3-(3-氟氮雜環丁烷-1-基)吡咯烷-1-基]-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮,異構體A(實施例A94A)(12%)。LCMS: m/z (ESI), [M+H] += 642.40。 1H NMR (DMSO-d 6, 400 MHz) δ 1.48-1.85 (4H, m), 1.89-2.05 (2H, m), 2.35-2.56 (4H, m), 2.68-2.76 (2H, m), 2.96 (1H, t), 3.07-3.20 (4H, m), 3.20-3.43 (4H, m), 3.45-3.70 (3H, m), 3.62 (3H, s), 3.72 (3H, s), 4.55 (1H, t), 4.63-4.73 (1H, m), 4.95-5.31 (1H, m), 6.44 (1H, d), 6.73 (1H, s), 7.37 (1H, d), 8.25 (1H, d), 8.42 (1H, s), 8.82 (1H, d), 12.50 (1H, s)。 步驟4. 5-[3-(3-氟氮雜環丁烷-1-基)吡咯烷-1-基]-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮,異構體A1(實施例A94A1)和異構體A2(實施例A94A2)的製備。將5-[3-(3-氟氮雜環丁烷-1-基)吡咯烷-1-基]-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}. 1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13, 17(33),18(22),19-八烯-12,16-二酮(實施例A94A,10.5 mg)的混合物通過Prep-手性-HPLC使用CHIRALPAK IG柱(2×25 cm,5 μm)使用含有0.2%二乙胺和乙醇/CH 2Cl 2(1/1)的己烷作為流動相來分離,以得到呈黃色固體的2.3 mg 5-[3-(3-氟氮雜環丁烷-1-基)吡咯烷-1-基]-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33), 18(22),19-八烯-12,16-二酮,異構體A1(實施例A94A1) (21%)和呈黃色固體的3.4 mg異構體A2(實施例A94A2) (32%)。 異構體A1:LCMS: m/z (ESI), [M+H] +=642.35 1H NMR (DMSO-d 6, 400 MHz) δ 1.48-1.85 (4H, m), 1.89-2.05 (2H, m), 2.35-2.56 (4H, m), 2.68-2.76 (2H, m), 2.96 (1H, t), 3.07-3.20 (4H, m), 3.20-3.43 (4H, m), 3.45-3.70 (3H, m), 3.62 (3H, s), 3.72 (3H, s), 4.55 (1H, t), 4.63-4.73 (1H, m), 4.95-5.31 (1H, m), 6.44 (1H, d), 6.73 (1H, s), 7.37 (1H, d), 8.25 (1H, d), 8.42 (1H, s), 8.82 (1H, d), 12.50 (1H, s)。手性HPLC,Rt=2.381分鐘。 異構體A2:LCMS: m/z (ESI), [M+H] +=642.55。 1H NMR (DMSO-d 6, 400 MHz) δ 1.48-1.85 (4H, m), 1.89-2.05 (2H, m), 2.35-2.56 (4H, m), 2.68-2.76 (2H, m), 2.96 (1H, t), 3.07-3.20 (4H, m), 3.20-3.43 (4H, m), 3.45-3.70 (3H, m), 3.62 (3H, s), 3.72 (3H, s), 4.55 (1H, t), 4.63-4.73 (1H, m), 4.95-5.31 (1H, m), 6.44 (1H, d), 6.73 (1H, s), 7.37 (1H, d), 8.25 (1H, d), 8.42 (1H, s), 8.82 (1H, d), 12.50 (1H, s)。手性HPLC,Rt=2.959分鐘。 實施例A94B、實施例94B1和實施例A94B2 5-[3-(3-氟氮雜環丁烷-1-基)吡咯烷-1-基]-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮,異構體B(實施例A94B)、異構體B1(實施例A94B1)和異構體B2(實施例A94B2) 步驟1. 5-[3-(3-氟氮雜環丁烷-1-基)吡咯烷-1-基]-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮(異構體B,實施例A94B)的製備。向3-(3-氟氮雜環丁烷-1-基)吡咯烷(INT-A94A-2,125 mg,0.86 mmol,5當量)、BrettPhos Pd G 3(47 mg,0.05 mmol,0.3當量)和5-溴-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}. 1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13, 17(33),18(22),19-八烯-12,16-二酮(M4B,100 mg,0.173 mmol,1當量)於1,4-二噁烷(10 mL)中的攪拌混合物中添加LiHMDS(2.1 mL,2.07 mmol,12當量)。將所得混合物在氮氣氣氛下在60℃下攪拌2 h。將混合物冷卻到室溫。將反應用飽和NH 4Cl(水溶液)(15 mL)淬滅,並且將混合物用CH 2Cl 2(3×50 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(10/1)洗脫進行純化。將產物通過Prep-HPLC用XBridge Prep OBD C18柱,使用含有0.1% NH 4HCO 3的水和乙腈作為流動相進行進一步純化。將含有期望化合物的級分蒸發到乾燥,以得到呈灰白色固體的15.3 mg 5-[3-(3-氟氮雜環丁烷-1-基)吡咯烷-1-基]-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮,異構體B(實施例A94B)(13%)。LCMS: m/z (ESI), [M+H] += 642.40。 1H NMR (DMSO-d 6, 400 MHz) δ 1.48-1.85 (4H, m), 1.89-2.05 (2H, m), 2.35-2.56 (4H, m), 2.68-2.76 (2H, m), 2.96 (1H, t), 3.07-3.20 (4H, m), 3.20-3.43 (4H, m), 3.45-3.70 (3H, m), 3.62 (3H, s), 3.72 (3H, S), 4.55 (1H, t), 4.63-4.73 (1H, m), 4.95-5.31 (1H, m), 6.44 (1H, d), 6.73 (1H, s), 7.37 (1H, d), 8.25 (1H, d), 8.42 (1H, s), 8.82 (1H, d), 12.50 (1H, s)。 步驟2. 5-[3-(3-氟氮雜環丁烷-1-基)吡咯烷-1-基]-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮,異構體B1(實施例A94B1)和異構體B2(實施例A94B2)的製備。將5-[3-(3-氟氮雜環丁烷-1-基)吡咯烷-1-基]-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}. 1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13, 17(33),18(22),19-八烯-12,16-二酮(實施例A94B,11 mg)的混合物通過Prep-手性-HPLC使用CHIRALPAK IG柱(2×25 cm,5 μm)使用含有0.2%二乙胺作為移動相A和乙醇/CH 2Cl 2(1/1)作為流動相B的己烷來分離,以得到呈黃色固體的3.7 mg 5-[3-(3-氟氮雜環丁烷-1-基)吡咯烷-1-基]-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮,異構體B1(實施例A94B1)(28%)和呈黃色固體的4.5 mg異構體B2(實施例A94B2)(34%)。 異構體B1:LCMS: m/z (ESI), [M+H] +=642.30。 1H NMR (DMSO-d 6, 400 MHz) δ 1.48-1.85 (4H, m), 1.89-2.05 (2H, m), 2.35-2.56 (4H, m), 2.68-2.76 (2H, m), 2.96 (1H, t), 3.07-3.20 (4H, m), 3.20-3.43 (4H, m), 3.45-3.70 (3H, m), 3.62 (3H, s), 3.72 (3H, S), 4.55 (1H, t), 4.63-4.73 (1H, m), 4.95-5.31 (1H, m), 6.44 (1H, d), 6.73 (1H, s), 7.37 (1H, d), 8.25 (1H, d), 8.42 (1H, s), 8.82 (1H, d), 12.50 (1H, s)。手性HPLC,Rt=1.608分鐘。 異構體B2:LCMS: m/z (ESI), [M+H] +=642.55。 1H NMR (DMSO-d 6, 400 MHz) δ 1.48-1.85 (4H, m), 1.89-2.05 (2H, m), 2.35-2.56 (4H, m), 2.68-2.76 (2H, m), 2.96 (1H, t), 3.07-3.20 (4H, m), 3.20-3.43 (4H, m), 3.45-3.70 (3H, m), 3.62 (3H, s), 3.72 (3H, S), 4.55 (1H, t), 4.63-4.73 (1H, m), 4.95-5.31 (1H, m), 6.48 (1H, d), 6.76 (1H, s), 7.38 (1H, d), 8.26 (1H, d), 8.42 (1H, s), 8.82 (1H, d), 12.52 (1H, s)。手性HPLC,Rt=2.068分鐘。 實施例A100 15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十四-3,5,7,9,13,17(34),18(22),19-八烯-12,16-二酮 步驟1. 2-{1-[(2-硝基苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙醇(INT-A100-1)的製備。將3-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-3-氮雜雙環[3.2.2]壬-1-胺(250 mg,0.83 mmol,1當量)、K 2CO 3(347 mg,2.51 mmol,3當量)和鄰氟硝基苯(295 mg,2.09 mmol,2.5當量)於乙腈(8 mL)中的混合物在氮氣氣氛下在100℃下攪拌8 h。將混合物冷卻到室溫。將反應用水(20 mL)淬滅,並且將混合物用乙酸乙酯(3×30 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上使用石油醚/乙酸乙酯(5/1)洗脫來進行純化,以得到呈黃色油的110 mg 2-{1-[(2-硝基苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙醇(INT-A100-1)(38%)。LCMS: m/z (ESI), [M+H] +=306.15。 步驟2. 1-甲基-5-[1-甲基-5-(2-{1-[(2-硝基苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙氧基)吡唑-4-基]-6-氧代吡啶-3-甲酸甲酯(INT-A100-2)的製備。在氮氣氣氛下在0℃下向2-{1-[(2-硝基苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙醇(100 mg,0.32 mmol,1當量)和三苯基膦(257 mg,0.98 mmol,3當量)於四氫呋喃(20 mL)中的攪拌混合物中逐滴添加含偶氮二甲酸二異丙酯(198 mg,0.98 mmol,3當量)和5-(5-羥基-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M2,68 mg,0.26 mmol,0.8當量)的四氫呋喃(5 mL)。將混合物在0℃下攪拌2 h。將反應用水(20 mL)淬滅,並且將混合物用乙酸乙酯(3×30 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上使用石油醚/乙酸乙酯(1/1)洗脫進行純化,以得到呈黃色油的150 mg 1-甲基-5-[1-甲基-5-(2-{1-[(2-硝基苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙氧基)吡唑-4-基]-6-氧代吡啶-3-甲酸甲酯(INT-A100-2)(74%)。LCMS: m/z (ESI), [M+H] +=551.20。 步驟3. 5-[5-(2-{1-[(2-氨基苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A100-3)的製備。在0℃下向1-甲基-5-[1-甲基-5-(2-{1-[(2-硝基苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙氧基)吡唑-4-基]-6-氧代吡啶-3-甲酸甲酯(140 mg,0.25 mmol,1當量)和雷尼鎳(69 mg)於甲醇(5 mL)中的攪拌混合物中添加水合肼(51 mg,1.01 mmol,4.00當量)。將混合物在氮氣氣氛下在0℃下攪拌2 h。將所得混合物過濾,並且將濾餅用甲醇(3×10 mL)洗滌。將所得溶液在減壓下濃縮,以得到呈淡黃色固體的90 mg 5-[5-(2-{1-[(2-氨基苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A100-3)(58%)。LCMS: m/z (ESI), [M+H] +=521.30。 步驟4. 5-(5-{2-[1-(2-氨基-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.2]壬-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A100-4)的製備。將5-[5-(2-{1-[(2-氨基苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(90 mg,0.17 mmol,1當量)和BrCN(21 mg,0.20 mmol,1.20當量)於乙醇(5 mL)中的混合物在氮氣氣氛下在室溫下攪拌過夜。將反應用水(20 mL)淬滅,並且將混合物用乙酸乙酯(3×30 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用石油醚/乙酸乙酯(1/1)洗脫進行純化,以得到呈黃色固體的60 mg 5-(5-{2-[1-(2-氨基-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.2]壬-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A100-4)(57%)。LCMS: m/z (ESI), [M+H] +=546.35。 步驟5. 5-(5-{2-[1-(2-氨基-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.2]壬-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A100-5)的製備。將5-(5-{2-[1-(2-氨基-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.2]壬-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(50 mg,0.09 mmol,1當量)和LiOH.H 2O(7 mg,0.27 mmol,3當量)於四氫呋喃(4 mL)和水(1 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將混合物在減壓下濃縮。將殘餘物通過C18FLASH,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3.H 2O和乙腈的水作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃色固體的25 mg 5-(5-{2-[1-(2-氨基-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.2]壬-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A100-5)(50%)。LCMS: m/z (ESI), [M+H] +=532.25。 步驟6. 15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]三十四-3,5,7,9,13,17(34),18(22),19-八烯-12,16-二酮(實施例A100)的製備。將5-(5-{2-[1-(2-氨基-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.2]壬-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(30 mg,0.05 mmol,1當量)、 N, N, N, N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(32 mg,0.08 mmol,1.5當量)和 N, N-二異丙基乙胺(21 mg,0.16 mmol,3當量)於1,4-二噁烷(5 mL)中的混合物在氮氣氣氛下在60℃下攪拌2 h。將混合物冷卻到室溫。將反應用水(10 mL)淬滅,並且將混合物用乙酸乙酯(3×20 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-HPLC用XBridge Shield RP18 OBD柱,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3 .H 2O的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的26.8 mg 15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]三十四-3,5,7,9,13,17(34),18(22),19-八烯-12,16-二酮(實施例A100)(92%)。LCMS: m/z (ESI) , [M+H] += 514.20。 1H NMR (DMSO-d 6 , 400 MHz) δ 1.85-1.98 (4H, m), 2.02-2.18 (3H, m), 2.78-2.88 (2H, m), 2.88-2.95 (2H, m), 3.40-3.52 (4H, m), 3.63 (3H, s), 3.75 (3H, s), 4.29 (2H, s), 7.12-7.21 (2H, m), 7.63 (1H, d), 7.89 (1H, d), 8.38-8.24 (2H, m), 9.12 (1H, d), 13.30 (1H, s)。 實施例A101 (11R)-15-氯-5,11,26-三甲基-16-(4-甲基哌嗪-1-基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮 步驟1. (11 R)-15-氯-5,11,26-三甲基-16-(4-甲基哌嗪-1-基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(實施例A101)的製備。在氮氣氣氛下在0℃下向(11 R)-15-氯-5,11,26-三甲基-16-(哌嗪-1-基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(實施例A23,80 mg,0.14 mmol,1當量)、多聚甲醛(38 mg,0.42 mmol,3當量)和NaBH(OAc) 3(60 mg,0.28 mmol,2當量)於CH 2Cl 2(20 mL)中的攪拌混合物中逐滴添加 N, N-二異丙基乙胺(36 mg,0.28 mmol,2當量)。將混合物在0℃下攪拌2 h。將反應用水(10 mL)淬滅,並且將混合物用CH 2Cl 2(3×10 mL)萃取。將合併的有機層用鹽水(3×10 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-HPLC用XBridge Shield RP18 OBD柱,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3.H 2O的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的32.4 mg (11 R)-15-氯-5,11,26-三甲基-16-(4-甲基哌嗪-1-基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3, 14,16,18,20,24-八烯-23,27-二酮(實施例A101)(39%)。LCMS: m/z (ESI), [M+H] +=579.20。 1H NMR (DMSO- d 6, 400 MHz) δ 0.86 (3H, d), 1.51-1.59 (1H, m), 1.86-2.08 (2H, m), 2.21-2.29 (5H, m), 2.51-2.64 (4H, m), 2.80-2.94 (4H, m), 3.62 (3H, s), 3.71 (3H, s), 3.91-4.03 (1H, m), 4.18 (1H, dd), 4.32-4.49 (1H, m), 4.75-4.97 (1H, m), 7.15 (1H, d), 7.58 (1H, d), 8.31-8.47 (2H, m), 8.85 (1H, d), 12.73 (1H, s)。 實施例A102和實施例A103 (11 R)-5,11,30-三甲基-7-氧雜-4,5,13,18,24,26,30-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]三十二-1(32),2(6),3,14,20,22,24,28-八烯-27,31-二酮(實施例A102)和(11 R)-5,11,18,30-四甲基-7-氧雜-4,5,13,18,24,26, 30-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}. 0^{15,20}]三十二-1(32),2(6),3,14,20,22,24,28-八烯-27,31-二酮(實施例A103)。 步驟1. 2,2,2-三氟- N-(2-氟苯乙基)乙醯胺(INT-A102-1)的製備。將2-(2-氟苯基)乙胺(50 g,359 mmol,1當量)、三甲胺(73 g,359 mmol,2當量)和三氟乙酸酐(75 g,1.1當量)於二氯甲烷(800 mL)中的混合物在氮氣氣氛下在0℃下攪拌3 h。將反應用水(200 mL)淬滅,並且將混合物用二氯甲烷(3×200 mL)萃取。將合併的有機層用鹽水(3×200 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法使用石油醚/乙酸乙酯(5/1)洗脫進行純化,以得到呈黃色油的76 g 2,2,2-三氟- N-[2-(2-氟苯基)乙基]乙醯胺(INT-A102-1)(90%)。LCMS: m/z (ESI), [M+H] +=236.05。 步驟2. 2,2,2-三氟-1-(5-氟-3,4-二氫異喹啉-2(1H)-yl)乙-1-酮(INT-A102-2)的製備。將2,2,2-三氟- N-[2-(2-氟苯基)乙基]乙醯胺(76 g,323.15 mmol,1當量)、(HCHO) n(43 g,969.5 mmol,3當量)和H 2SO 4(224 mL,4201 mmol,13當量)和AcOH(296 mL)的混合物在氮氣氣氛下在0℃下攪拌3 h。將反應用水(800 mL)淬滅,並且用NaOH中和到pH 7。將混合物用二氯甲烷(3×500 mL)萃取。將合併的有機層用鹽水(3×500 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法使用石油醚/乙酸乙酯(1/1)洗脫來進行純化,以得到呈棕黃色油的10 g 2,2,2-三氟-1-(5-氟-3,4-二氫-1H-異喹啉-2-基)乙酮(INT-A102-2)(13%)。LCMS: m/z (ESI), [M+H] +=248.10。 步驟3. 2,2,2-三氟-1-(5-氟-6-硝基-3,4-二氫異喹啉-2(1H)-yl)乙-1-酮(INT-A102-3)的製備。將2,2,2-三氟-1-(5-氟-3,4-二氫-1H-異喹啉-2-基)乙酮(10 g,40.45 mmol,1當量)和KNO 3(4 g,40.45 mmol,1當量)於H 2SO 4(100 mL)中的混合物在0℃下攪拌過夜。將反應用水(500 mL)淬滅,並且用NaOH調節到pH 9。將混合物用二氯甲烷(3×200 mL)萃取。將合併的有機層用鹽水(3×200 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法使用石油醚/乙酸乙酯(3/1)洗脫來進行純化,以得到呈黃色油的1.4 g 2,2,2-三氟-1-(5-氟-6-硝基-3,4-二氫-1H-異喹啉-2-基)乙酮(INT-A102-3)(12%)。 1H NMR (DMSO, 400 MHz) δ 2.91-2.99 (2H, m), 3.85-3.92 (2H, m), 4.91 (2H, d), 7.45 (1H, d), 8.12 (1H, d)。 步驟4. 2,2,2-三氟-1-(5-{[(2 R)-5-羥基-2-甲基戊基]氨基}-6-硝基-3,4-二氫-1H-異喹啉-2-基)乙酮(INT-A102-4)的製備。將2,2,2-三氟-1-(5-氟-6-硝基-3,4-二氫-1H-異喹啉-2-基)乙酮(INT-A102-3,15 g,51.34 mmol,1當量)、 N,N-二異丙基乙胺(19.9 g,154.01 mmol,3當量)和(4 R)-5-氨基-4-甲基戊-1-醇(M3-6,6016 mg,51.34 mmol,1當量)於乙腈(150 mL)中的混合物在80℃下攪拌2 h。將混合物冷卻到室溫。將反應用水(500 mL)淬滅,並且將混合物用二氯甲烷(3×500 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠柱使用石油醚/乙酸乙酯(1/1)洗脫來進行純化,以得到呈黃色固體的10 g 2,2,2-三氟-1-(5-{[(2 R)-5-羥基-2-甲基戊基]氨基}-6-硝基-3,4-二氫-1H-異喹啉-2-基)乙酮(INT-A102-3)(50%)。LCMS: m/z (ESI), [M+H] +=390.10。 步驟5. (4 R)-4-甲基-5-[(6-硝基-1,2,3,4-四氫異喹啉-5-基)氨基]戊-1-醇(INT-A102-5)的製備。將2,2,2-三氟-1-(5-{[(2 R)-5-羥基-2-甲基戊基]氨基}-6-硝基-3,4-二氫-1H-異喹啉-2-基)乙酮(10 g,25.68 mmol,1當量)和K 2CO 3(10.6g,77.05 mmol,3當量)於甲醇(80 mL)和H 2O(40 mL)中的混合物在室溫下攪拌5 h。將反應用水(500 mL)處理,並且將混合物用二氯甲烷(3×500 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠柱使用石油醚/乙酸乙酯(1/1)洗脫來進行純化,以得到呈黃色固體的7 g (4 R)-4-甲基-5-[(6-硝基-1,2,3,4-四氫異喹啉-5-基)氨基]戊-1-醇(INT-A102-5)(93%)。LCMS: m/z (ESI), [M+H] +=294.15。 1H NMR (DMSO-d 6,400 MHz) δ 0.83-0.88 (3 H, m), 1.07-1.11 (1 H, m), 1.22-1.51 (3 H, m), 1.57-1.64 (1 H, m), 2.59 (2 H, t), 2.88-2.92 (3 H, m), 3.10-3.20 (1 H, m), 3.30-3.33 (2 H, m), 3.87 (2H, s), 4.37 (1 H, s), 6.56 (1 H, d), 6.69 (1 H, t), 7.26-7.37 (1 H, m), 7.70 (1 H, d)。 步驟6. 5-{[(2 R)-5-羥基-2-甲基戊基]氨基}-6-硝基-3,4-二氫-1H-異喹啉-2-甲酸叔丁酯(INT-A102-6)的製備。將(4 R)-4-甲基-5-[(6-硝基-1,2,3,4-四氫異喹啉-5-基)氨基]戊-1-醇(8 g,27.27 mmol,1當量)、三甲胺(5.5 g,54.54 mmol,2當量)和二碳酸二叔丁酯(7.1 g,32.72 mmol,1.2當量)於二氯甲烷(100 mL)中的混合物在室溫下攪拌2 h。將反應用水(300 mL)淬滅,並且將混合物用二氯甲烷(3×300 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠柱使用石油醚/乙酸乙酯(1/1)洗脫來進行純化,以得到呈黃色油的6.5 g 5-{[(2 R)-5-羥基-2-甲基戊基]氨基}-6-硝基-3,4-二氫-1H-異喹啉-2-甲酸叔丁酯(INT-A102-6)(60%)。LCMS: m/z (ESI), [M+H] +=394.20。 1H NMR (DMSO-d 6400 MHz) δ 0.84 (3H, d), 1.03-1.17 (1H, m), 1.22-1.45 (2H, m), 1.45 (9H, s), 1.61 (1H, q), 2.75 (2H, t), 2.93-2.97 (1H, m), 3.08-3.12 (1H, m), 3.30-3.33 (2H, m), 3.51 (2H, t), 4.36 (1H, t), 4.52 (2H, s), 6.73 (1H, d), 6.83 (1H, s), 7.80 (1H, d) 步驟7. 5-{[(2 R)-5-({4-[5-(甲氧基羰基)-1-甲基-2-氧代吡啶-3-基]-2-甲基吡唑-3-基}氧基)-2-甲基戊基]氨基}-6-硝基-3,4-二氫-1H-異喹啉-2-甲酸叔丁酯(INT-A102-7)的製備。在氮氣氣氛下在室溫下向5-{[(2 R)-5-羥基-2-甲基戊基]氨基}-6-硝基-3,4-二氫-1H-異喹啉-2-甲酸叔丁酯(INT-A102-6,800 mg,2.03 mmol,1當量)和5-(5-羥基-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M2,535 mg,2.03 mmol,1.0當量)於甲苯(30 mL)中的攪拌混合物中添加2-(三丁基-l^[5]-膦亞基)乙腈(2 g,10.16 mmol,5當量)。將所得混合物在100℃下攪拌2 h,然後冷卻到室溫。將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過C18柱使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發,以得到呈黃色油的700 mg 5-{[(2 R)-5-({4-[5-(甲氧基羰基)-1-甲基-2-氧代吡啶-3-基]-2-甲基吡唑-3-基}氧基)-2-甲基戊基]氨基}-6-硝基-3,4-二氫-1H-異喹啉-2-甲酸叔丁酯(INT-A102-7)(52%)。LCMS: m/z (ESI), [M+H] += 639.30。 1H NMR (DMSO- d 6, 400 MHz) δ 0.87 (3H, d), 0.92-0.96 (1H, m), 1.27 (1H, d), 1.44 (11H, s), 2.72-2.77 (2H, m), 2.94-3.03 (1H, m), 3.09-3.16 (1H, m), 3.18 (1H, d), 3.50 (2H, s), 3.58 (3H, s), 3.66 (3H, s), 3.78 (3H, s), 3.88-3.92 (2H, t), 4.50 (2H, s), 6.72 (1H, d), 6.83 (1H, s), 7.78 (1H, d), 8.00 (1H, s), 8.11 (1H, d), 8.45 (1H, d)。 步驟8. 6-氨基-5-{[(2 R)-5-({4-[5-(甲氧基羰基)-1-甲基-2-氧代吡啶-3-基]-2-甲基吡唑-3-基}氧基)-2-甲基戊基]氨基}-3,4-二氫-1H-異喹啉-2-甲酸叔丁酯(INT-A102-8)的製備。向5-{[(2R)-5-({4-[5-(甲氧基羰基)-1-甲基-2-氧代吡啶-3-基]-2-甲基吡唑-3-基}氧基)-2-甲基戊基]氨基}-6-硝基-3,4-二氫-1H-異喹啉-2-甲酸叔丁酯(700 mg,1.09 mmol,1當量)、雷尼鎳(187 mg,2.19 mmol,2當量)於甲醇(8 mL)中的混合物中添加NH 2NH 2 .H 2O(109 mg,2.19 mmol,2當量)。將混合物在氮氣氣氛下在0℃下攪拌30分鐘。將混合物過濾,並且將濾餅用甲醇(3×40 mL)洗滌。將所得溶液在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(10/1)洗脫來進行純化,以得到呈棕色固體的400 mg 6-氨基-5-{[(2 R)-5-({4-[5-(甲氧基羰基)-1-甲基-2-氧代吡啶-3-基]-2-甲基吡唑-3-基}氧基)-2-甲基戊基]氨基}-3,4-二氫-1H-異喹啉-2-甲酸叔丁酯(INT-A102-8)(59%)。LCMS: m/z (ESI), [M+H] +=609.30。 步驟9. 5-(5-{[(4 R)-4-{[2-氨基-7-(叔丁氧基羰基)-6H,8H,9H-咪唑並[4,5-f]異喹啉-1-基]甲基}戊基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A102-9)的製備。將6-氨基-5-{[(2 R)-5-({4-[5-(甲氧基羰基)-1-甲基-2-氧代吡啶-3-基]-2-甲基吡唑-3-基}氧基)-2-甲基戊基]氨基}-3,4-二氫-1H-異喹啉-2-甲酸叔丁酯(400 mg,0.65 mmol,1當量)和BrCN(104 mg,0.98 mmol,1.5當量)於乙醇(5 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將反應用水(50 mL)淬滅,並且將混合物用二氯甲烷(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(12/1)洗脫進行純化,以得到呈棕色固體的300 mg 5-(5-{[(4 R)-4-{[2-氨基-7-(叔丁氧基羰基)-6H,8H,9H-咪唑並[4,5-f]異喹啉-1-基]甲基}戊基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A102-9)(72%)。LCMS: m/z (ESI), [M+H] +=634.30。 步驟10. 5-(5-{[(4 R)-4-{[2-氨基-7-(叔丁氧基羰基)-6H,8H,9H-咪唑並[4,5-f]異喹啉-1-基]甲基}戊基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A102-10)的製備。將5-(5-{[(4 R)-4-{[2-氨基-7-(叔丁氧基羰基)-6H,8H,9H-咪唑並[4,5-f]異喹啉-1-基]甲基}戊基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(300 mg,0.47 mmol,1當量)和LiOH .H 2O(99 mg,2.36 mmol,5當量)於四氫呋喃/H 2O(2 mL/0.5 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將所得混合物在減壓下濃縮。將殘餘物通過C18柱使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的250 mg 5-(5-{[(4R)-4-{[2-氨基-7-(叔丁氧基羰基)-6H,8H,9H-咪唑並[4,5-f]異喹啉-1-基]甲基}戊基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A102-10)(85%)。LCMS: m/z (ESI), [M+H] +=620.40。 步驟11. (11 R)-5,11,30-三甲基-27,31-二氧代-7-氧雜-4,5,13,18,24,26,30-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}. 0^{14,23}.0^{15,20}]三十二-1(32),2(6),3,14,20,22,24,28-八烯-18-甲酸叔丁酯(INT-A102-11)的製備。將5-(5-{[(4 R)-4-{[2-氨基-7-(叔丁氧基羰基)-6H,8H,9H-咪唑並[4,5-f]異喹啉-1-基]甲基}戊基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(200 mg,0.32 mmol,1當量)、 N,N,N,N-四甲基-O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(245 mg,0.64 mmol,2當量)和 N,N-二異丙基乙胺(125 mg,0.96 mmol,3當量)於1,4-二噁烷(5 mL)中的混合物在氮氣氣氛下在60℃下攪拌30分鐘。將混合物冷卻到室溫。將反應用水(20 mL)淬滅,並且將混合物用二氯甲烷(2×20 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用(二氯甲烷/甲醇12/1)洗脫進行純化,以得到呈白色固體的180 mg (11 R)-5,11,30-三甲基-27,31-二氧代-7-氧雜-4,5,13,18,24,26,30-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}. 0^{14,23}.0^{15,20}]三十二-1(32),2(6),3,14,20,22,24,28-八烯-18-甲酸叔丁酯(INT-A102-11)(92%)。LCMS: m/z (ESI), [M+H] +=602.30。 步驟12. (11 R)-5,11,30-三甲基-7-氧雜-4,5,13,18,24, 26,30-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}. 0^{15,20}]三十二-1(32),2(6),3,14,20,22,24,28-八烯-27,31-二酮(實施例A102)的製備。將(11R)-5,11,30-三甲基-27,31-二氧代-7-氧雜-4,5,13,18,24,26,30-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]三十二-1(32),2(6),3,14,20,22,24,28-八烯-18-甲酸叔丁酯(200 mg,0.33 mmol,1當量)於二氯甲烷/三氟乙酸(3 mL/1 mL)中的混合物在氮氣氣氛下在室溫下攪拌30分鐘。將反應混合物在減壓下濃縮。將殘餘物用飽和NaHCO 3(水溶液)(50 mL)處理,並且用二氯甲烷(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(10/1)洗脫進行純化,以得到呈白色固體的110 mg (11 R)-5,11,30-三甲基-7-氧雜-4,5,13,18,24,26,30-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]三十二-1(32),2(6),3,14,20,22,24,28-八烯-27,31-二酮(實施例A102)(65%)。LCMS: m/z (ESI), [M+H] +=502.45。 1H NMR (DMSO- d 6, 400 MHz) δ 0.81 (3H, d), 1.21-1.27 (1H, m), 1.41-1.45 (1H, m), 1.76-2.00 (4H, m), 2.35 (3H, s), 2.60-2.76 (2H, m), 3.52-3.58 (2H, m), 3.62 (3H, s), 3.71 (3H, s), 3.85-3.93 (1H, m), 4.11-4.27 (1H, m), 4.27-4.37 (1H, m), 6.91 (1H, d), 7.38 (1H, d), 8.28 (1H, d), 8.36 (1H, s), 8.77 (1H, d), 12.62 (1H, s)。 步驟13. (11R)-5,11,18,30-四甲基-7-氧雜-4,5,13,18, 24,26,30-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}. 0^{15,20}]三十二-1(32),2(6),3,14,20,22,24,28-八烯-27,31-二酮(實施例A103)的製備。在氮氣氣氛下在室溫下向(11 R)-5,11,30-三甲基-7-氧雜-4,5,13,18,24,26,30-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]三十二-1(32),2(6),3,14,20,22,24,28-八烯-27,31-二酮(實施例A102,50 mg,0.10 mmol,1當量)、多聚甲醛(26 mg,0.30 mmol,3當量)和 N, N-二異丙基乙胺(25 mg,0.20 mmol,2當量)於二氯甲烷(2 mL)中的攪拌混合物中添加NaBH(OAc) 3(42 mg,0.20 mmol,2當量)。將反應用水(50 mL)淬滅,並且將混合物用二氯甲烷(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-HPLC用XBridge Prep OBD C18柱,使用含有10 mmol/L NH 4HCO 3的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃色固體的26 mg (11 R)-5,11,18,30-四甲基-7-氧雜-4,5,13,18,24,26,30-七氮雜六環[26.3.1.0^{2,6}. 0^{13,25}.0^{14,23}.0^{15,20}]三十二-1(32),2(6),3,14,20, 22,24,28-八烯-27,31-二酮(實施例A103)(50%)。LCMS: m/z (ESI), [M+H] +=516.30 1H NMR (DMSO- d 6, 400 MHz) δ 0.81 (3H, d), 1.21-1.27 (1H, m), 1.41-1.45 (1H, m), 1.76-2.00 (3H, m), 2.35 (3H, s), 2.60-2.76 (2H, m), 3.21 (3H, s), 3.52-3.58 (2H, m), 3.62 (3H, s), 3.71 (3H, s), 3.85-3.93 (1H, m), 4.11-4.27 (1H, m), 4.27-4.37 (1H, m), 6.91 (1H, d), 7.38 (1H, d), 8.28 (1H, d), 8.36 (1H, s), 8.77 (1H, d), 12.62 (1H, s)。 實施例A109 (12 R)-12,27-二甲基-17-(4-甲基哌嗪-1-基)-8-氧雜-14,21, 23,27-四氮雜五環[23.3.1.0^{2,7}.0^{14,22}.0^{15,20}]二十九-1(29),2(7),3,5,15,17,19,21,25-壬烯-24,28-二酮 步驟1. 5-(2-羥基苯基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A109-1)的製備。將5-溴-1-甲基-6-氧代吡啶-3-甲酸甲酯(8 g,32.51 mmol,1當量)、2-(4,4,5,5-四甲基-1,3,2-二氧雜環戊硼烷-2-基)苯酚(7.87 g,35.76 mmol,1.1當量)、Pd(dppf)Cl 2(1.43 g,1.95 mmol,0.06當量)和K 2CO 3(8.99 g,65.02 mmol,2當量)於1,4-二噁烷(240 mL)和H 2O(60 mL)中的混合物在氮氣氣氛下在80℃下攪拌1.5 h。將混合物冷卻到室溫。將反應用水(500 mL)淬滅,並且將混合物用CH 2Cl 2(3×500 mL)萃取。將合併的有機層用鹽水(3×500 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-TLC使用石油醚/乙酸乙酯(3/1)洗脫進行純化,以得到呈棕色固體的5.6 g 5-(2-羥基苯基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A109-1)(66%)。LCMS: m/z (ESI), [M+H] +=260.05。 1H NMR (DMSO-d 6, 400 MHz) δ 3.61 (3H, s), 3.82 (3H, s), 6.81-6.93 (2H, m), 7.18-7.22 (1H, m), 7.24-7.28 (1H, m), 7.86 (1H, d), 8.60 (1H, d), 9.39 (1H, s)。 步驟2. 5-(2-{[(4 R)-5-[(5-溴-2-硝基苯基)氨基]-4-甲基戊基]氧基}苯基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A109-2)的製備。將5-(2-羥基苯基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(1 g,3.86 mmol,1當量)、2-(三丁基-l^[5]-膦亞基)乙腈(4.65 g,19.29 mmol,5當量)和(4 R)-5-[(5-溴-2-硝基苯基)氨基]-4-甲基戊-1-醇(M3-7,1.22 g,3.86 mmol,1當量)於甲苯(40 mL)中的混合物在氮氣氣氛下在130℃下攪拌1.5 h。將混合物冷卻到室溫。將反應用水(200 mL)淬滅,並且將混合物用乙酸乙酯(3×200 mL)萃取。將合併的有機層用鹽水(3×200 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-TLC使用石油醚/乙酸乙酯(3/1)洗脫進行純化,以得到呈紅棕色固體的1.7 g 5-(2-{[(4 R)-5-[(5-溴-2-硝基苯基)氨基]-4-甲基戊基]氧基}苯基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A109-2)(79%)。LCMS: m/z (ESI), [M+H] +=558.10。 步驟3. 5-(2-{[(4 R)-5-[(2-氨基-5-溴苯基)氨基]-4-甲基戊基]氧基}苯基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A109-3)的製備。 在0℃下向5-(2-{[(4 R)-5-[(5-溴-2-硝基苯基)氨基]-4-甲基戊基]氧基}苯基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(800 mg,1.43 mmol,1當量)和雷尼鎳(80 mg,0.93 mmol,0.65當量)於甲醇(35 mL)中的攪拌混合物中添加NH 2NH 2 .H 2O(143 mg,2.87 mmol,2當量)。將所得混合物在室溫下攪拌2 h。將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×80 mL)萃取。將合併的有機層用鹽水(3×80 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用石油醚/乙酸乙酯(1/5)洗脫進行純化,以得到呈紅棕色固體的690 mg 5-(2-{[(4 R)-5-[(2-氨基-5-溴苯基)氨基]-4-甲基戊基]氧基}苯基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A109-3)(91%)。LCMS: m/z (ESI), [M+H] +=528.00。 步驟4. 5-(2-{[(4 R)-4-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]戊基]氧基}苯基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A109-4)的製備。將5-(2-{[(4 R)-5-[(2-氨基-5-溴苯基)氨基]-4-甲基戊基]氧基}苯基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(1.3 g,2.46 mmol,1當量)和BrCN(521 mg,4.92 mmol,2當量)於二氯甲烷(30 mL)中的混合物在室溫下攪拌4 h。將反應用水(200 mL)淬滅,並且將混合物用二氯甲烷(3×200 mL)萃取。將合併的有機層用鹽水(3×200 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(20/1)洗脫進行純化,以得到呈紅棕色固體的1.32 g 5-(2-{[(4 R)-4-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]戊基]氧基}苯基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A109-4)(97%)。LCMS: m/z (ESI), [M+H] +=554.95。 步驟5. 5-(2-{[(4 R)-4-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]戊基]氧基}苯基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A109-5)的製備。在室溫下向5-(2-{[(4 R)-4-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]戊基]氧基}苯基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(1.3 g,2.35 mmol,1當量)於四氫呋喃(10 mL)中的攪拌混合物中添加LiOH(225.02 mg,9.39 mmol,4當量)和H 2O(4 mL)。將所得混合物在室溫下攪拌1 h並在真空下濃縮。將殘餘物通過C18 FLASH色譜法使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈紅色固體的900 mg 5-(2-{[(4 R)-4-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]戊基]氧基}苯基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A109-5)(71%)。LCMS: m/z (ESI), [M+H] +=540.95。 步驟6. (12 R)-17-溴-12,27-二甲基-8-氧雜-14,21,23,27-四氮雜五環[23.3.1.0^{2,7}.0^{14,22}.0^{15,20}]二十九-1(29),2(7),3,5,15,17,19,21,25-壬烯-24,28-二酮(INT-A109-6)的製備。在室溫下向5-(2-{[(4 R)-4-[(2-氨基-6-溴-1,3-苯並二氮唑-1-基)甲基]戊基]氧基}苯基)-1-甲基-6-氧代吡啶-3-羧酸(450 mg,0.83 mmol,1當量)於二噁烷(10 mL)中的攪拌混合物中添加 N,N-二異丙基乙胺(323 mg,2.50 mmol,3當量)和 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(476 mg,1.25 mmol,1.5當量)。將所得混合物在氮氣氣氛下在60℃下攪拌1 h,並且冷卻到室溫。將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-TLC使用亞甲基二氯甲烷/甲醇(30/1)洗脫進行純化,以得到呈紅棕色固體的350 mg (12 R)-17-溴-12,27-二甲基-8-氧雜-14,21,23,27-四氮雜五環[23.3.1.0^{2,7}.0^{14,22}. 0^{15,20}]二十九-1(29),2(7),3,5,15,17,19,21,25-壬烯-24,28-二酮(INT-A109-6)(80%)。LCMS: m/z (ESI), [M+H] +=523.05。 步驟7. (12 R)-12,27-二甲基-17-(4-甲基哌嗪-1-基)-8-氧雜-14,21,23,27-四氮雜五環[23.3.1.0^{2,7}.0^{14,22}. 0^{15,20}]二十九-1(29),2(7),3,5,15,17,19,21,25-壬烯-24,28-二酮(實施例A109)的製備。在氮氣氣氛下在室溫下向(12 R)-17-溴-12,27-二甲基-8-氧雜-14,21,23,27-四氮雜五環[23.3.1.0^{2,7}.0^{14,22}.0^{15,20}]二十九-1(29),2(7), 3,5,15,17,19,21,25-壬烯-24,28-二酮(200 mg,0.38 mmol,1當量)、1-甲基哌嗪(127 mg,1.27 mmol,3.31當量)和BrettPhos Pd G3(90 mg,0.10 mmol,0.26當量)於1,4-二噁烷(10 mL)中的攪拌混合物中逐滴添加LiHMDS(3.8 mL,3.8 mmol,10當量)。將所得混合物在60℃下攪拌50分鐘,並且冷卻到室溫。將反應用飽和NH 4Cl(水溶液)(100 mL)淬滅,並且將混合物用二氯甲烷(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-TLC使用二氯甲烷/甲醇(30/1)洗脫進行純化,並且將產物通過prep-HPLC使用XBridge Prep OBD C18柱使用含有10 mmol/L NH 4HCO 3的水和乙腈作為移動相進行進一步純化。將含有期望化合物的級分蒸發到乾燥,以得到呈灰白色固體的13.7 mg (12 R)-12,27-二甲基-17-(4-甲基哌嗪-1-基)-8-氧雜-14,21,23,27-四氮雜五環[23.3.1.0^{2,7}.0^{14,22}.0^{15,20}]二十九-1(29),2(7),3,5,15,17,19,21,25-壬烯-24,28-二酮(實施例A109)(7%)。LCMS: m/z (ESI), [M+H] +=541.35。 1H NMR (DMSO-d 6, 400 MHz) δ 0.92 (3H, d), 1.29-1.33 (1H, m), 1.60-1.66 (1H, m), 2.02-2.06 (2H, m), 2.24 (3H, s), 2.26-2.30 (1H, m), 2.49-2.51 (4H, m), 3.14 (4H, t), 3.60 (3H, s), 3.93-4.08 (3H, m), 4.20 (1H, t), 6.83-6.89 (1H, m), 6.86 (1H, dd), 6.92-6.96 (1H, m), 7.12 (1H, d), 7.25-7.29 (1H, m), 7.34 (1H, d), 7.69 (1H, dd), 8.33 (1H, d), 8.85 (1H, d), 12.27 (1H, s)。 實施例A110 (12 R)-4-氟-12,27-二甲基-17-(4-甲基哌嗪-1-基)-8-氧雜-14,21,23,27-四氮雜五環[23.3.1.0^{2,7}.0^{14,22}. 0^{15,20}]二十九-1(29),2(7),3,5,15,17,19,21,25-壬烯-24,28-二酮 步驟1. 5-(5-氟-2-羥基苯基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A110-1)的製備。將5-溴-1-甲基-6-氧代吡啶-3-甲酸甲酯(2 g,8.128 mmol,1當量)、4-氟-2-(4,4,5,5-四甲基-1,3,2-二氧雜環戊硼烷-2-基)苯酚(2.32 g,9.754 mmol,1.2當量)、Pd(dppf)Cl 2(0.48 g,0.656 mmol,0.08當量)和K 2CO 3(2.81 g,20.320 mmol,2.5當量)於二噁烷(40 mL)和H 2O(10 mL)中的混合物在氮氣氣氛下在80℃下攪拌2 h。將混合物冷卻到室溫。將反應用水(150 mL)淬滅,並且將混合物用二氯甲烷(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法使用二氯甲烷/甲醇(15/1)洗脫進行純化,以得到呈黃色固體的1 g 5-(5-氟-2-羥基苯基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A110-1) (44%)。 1H NMR (DMSO-d 6, 400 MHz) 3.60 (3H, s), 3.81 (3H, s), 6.84-6.95 (1H,m), 6.96-7.08 (1H, m), 7.14 (1H, d), 7.94 (1H, d), 8.61 (1H, d), 9.41 (1H, s) 步驟2. 5-(5-氟-2-{[(4R)-4-甲基-5-{[5-(4-甲基哌嗪-1-基)-2-硝基苯基]氨基}戊基]氧基}苯基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A110-2).將5-(5-氟-2-羥基苯基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(526 mg,1.90 mmol,1當量)、(4 R)-4-甲基-5-{[5-(4-甲基哌嗪-1-基)-2-硝基苯基]氨基}戊-1-醇(639.07 mg,1.90 mmol,1當量)和2-(三丁基-l^[5]-膦亞基)乙腈(2.30 g,9.50 mmol,5當量)於甲苯(5 mL)中的混合物在氮氣氣氛下在130℃下攪拌過夜。將混合物冷卻到室溫。將反應用水(100 mL)淬滅,並且將混合物用二氯甲烷(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用石油醚/乙酸乙酯(1/1)洗脫進行純化,以得到呈白色固體的500 mg 5-(5-氟-2-{[(4R)-4-甲基-5-{[5-(4-甲基哌嗪-1-基)-2-硝基苯基]氨基}戊基]氧基}苯基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A110-2)(44%)。 1H NMR (DMSO-d 6 , 400 MHz) 0.90 (3H, s), 1.31-1.39 (4H, m), 2.20 (3H, s), 2.32-2.41 (4H, m), 2.44 (1H, t), 2.98-3.28 (2H, m), 3.40 (4H, t), 3.56 (3H, s), 3.76 (3H, s), 3.95 (2H, t), 5.77 (1H, s), 5.96 (1H, d), 6.43 (1H, d), 6.97-7.29 (4H, m), 7.83-7.95 (2H, m), 8.05 (1H, d), 8.39 (1H, t), 8.53 (1H, d)。 步驟3. 5-(2-{[(4 R)-5-{[2-氨基-5-(4-甲基哌嗪-1-基)苯基]氨基}-4-甲基戊基]氧基}-5-氟苯基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A110-3)的製備。在0℃下向5-(5-氟-2-{[(4 R)-4-甲基-5-{[5-(4-甲基哌嗪-1-基)-2-硝基苯基]氨基}戊基]氧基}苯基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(500 mg,0.83 mmol,1當量)和雷尼鎳(3.60 mg,0.042 mmol,0.05當量)於甲醇(20 mL)中的攪拌混合物中添加肼(126.06 mg,2.51 mmol,3當量)。將混合物在氮氣氣氛下在0℃下攪拌30分鐘。將所得混合物過濾,並且將濾餅用甲醇(3×30 mL)洗滌。將所得溶液在減壓下濃縮。將殘餘物通過C18 FLASH色譜法使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃色固體的400 mg 5-(2-{[(4R)-5-{[2-氨基-5-(4-甲基哌嗪-1-基)苯基]氨基}-4-甲基戊基]氧基}-5-氟苯基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A110-3)(84%)。LCMS: m/z (ESI), [M+H] += 566.15。 步驟4. 5-(2-{[(4 R)-4-{[2-氨基-6-(4-甲基哌嗪-1-基)-1,3-苯並二氮唑-1-基]甲基}戊基]氧基}-5-氟苯基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A110-4)的製備。將5-(2-{[(4 R)-5-{[2-氨基-5-(4-甲基哌嗪-1-基)苯基]氨基}-4-甲基戊基]氧基}-5-氟苯基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(240 mg,0.42 mmol,1當量)和BrCN(51 mg,0.46 mmol,1.1當量)於二氯甲烷(5 mL)中的混合物在氮氣氣氛下在室溫下攪拌4 h,並且在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(1/1)洗脫進行純化,以得到呈黃色固體的140 mg 5-(2-{[(4 R)-4-{[2-氨基-6-(4-甲基哌嗪-1-基)-1,3-苯並二氮唑-1-基]甲基}戊基]氧基}-5-氟苯基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A110-4)(55%)。LCMS: m/z (ESI), [M+H] +=591.15。 步驟5. 5-(2-{[(4 R)-4-{[2-氨基-6-(4-甲基哌嗪-1-基)-1,3-苯並二氮唑-1-基]甲基}戊基]氧基}-5-氟苯基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A110-5)的製備。將5-(2-{[(4 R)-4-{[2-氨基-6-(4-甲基哌嗪-1-基)-1,3-苯並二氮唑-1-基]甲基}戊基]氧基}-5-氟苯基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(75 mg,0.12 mmol,1當量)和LiOH .H 2O(10 mg,0.25 mmol,2當量)於四氫呋喃(2 mL)和水(0.5 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將反應混合物在減壓下濃縮。將殘餘物通過C18 FLASH色譜法使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃色油的40 mg 5-(2-{[(4 R)-4-{[2-氨基-6-(4-甲基哌嗪-1-基)-1,3-苯並二氮唑-1-基]甲基}戊基]氧基}-5-氟苯基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A110-5)(54%)。LCMS: m/z (ESI), [M+H] +=577.25。 步驟6. (12 R)-4-氟-12,27-二甲基-17-(4-甲基哌嗪-1-基)-8-氧雜-14,21,23,27-四氮雜五環[23.3.1.0^{2,7}. 0^{14,22}.0^{15,20}]二十九-1(29),2(7),3,5,15,17,19,21, 25-壬烯-24,28-二酮(實施例A110)的製備。將5-(2-{[(4 R)-4-{[2-氨基-6-(4-甲基哌嗪-1-基)-1,3-苯並二氮唑-1-基]甲基}戊基]氧基}-5-氟苯基)-1-甲基-6-氧代吡啶-3-羧酸(35 mg,0.06 mmol,1當量)、 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(34 mg,0.09 mmol,1.5當量)和 N,N-二異丙基乙胺(23 mg,0.18 mmol,3當量)於1,4-二噁烷(2 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將反應用水(50 mL)淬滅,並且將混合物用二氯甲烷(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-HPLC用XBridge Shield RP18 OBD柱,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3.H 2O的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃綠色固體的3.6 mg (12 R)-4-氟-12,27-二甲基-17-(4-甲基哌嗪-1-基)-8-氧雜-14,21,23,27-四氮雜五環[23.3.1.0^{2,7}. 0^{14,22}.0^{15,20}]二十九-1(29),2(7),3,5,15,17,19,21, 25-壬烯-24,28-二酮(實施例A110)(10%)。LCMS: m/z (ESI), [M+H] +=559.40。 1H NMR (DMSO-d 6 , 400 MHz) δ 0.94 (3H, d), 1.31-1.40 (1H, m), 1.64-1.70 (1H, m), 2.01-2.14 (2H, m), 2.21-2.30 (1H, m), 2.41 (3H, s), 2.66-2.73 (4H, m), 3.22 (4H, t), 3.60 (3H, s), 3.76-3.85 (1H, m), 3.99-4.09 (2H, m), 4.13 (1H, s), 6.87-6.97 (2H, m), 6.98-7.09 (2H, m), 7.24 (1H, d), 7.47-7.55 (1H, m), 8.23 (1H, s), 8.91 (1H, s)。 實施例A111 (12 R)-4,5-二氟-12,27-二甲基-17-(4-甲基哌嗪-1-基)-8-氧雜-14,21,23,27-四氮雜五環[23.3.1.0^{2,7}.0^{14,22}. 0^{15,20}]二十九-1(29),2(7),3,5,15,17,19,21,25-壬烯-24,28-二酮 步驟1. 5-(4,5-二氟-2-羥基苯基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A111-1)的製備。在氮氣氣氛下在室溫下向5-溴-1-甲基-6-氧代吡啶-3-甲酸甲酯(2.00 g,8.12 mmol,1當量)和4,5-二氟-2-羥苯基硼酸(2.12 g,12.19 mmol,1.5當量)於1,4-二噁烷(32 mL)和水(8 mL)中的攪拌混合物中分批添加1,1'-雙(二苯基膦基)二茂鐵-鈀(II)(1.19 g,1.62 mmol,0.2當量)和K 2CO 3(2.81 g,20.32 mmol,2.5當量)。將所得混合物在80℃下攪拌2 h。將混合物冷卻到室溫。將反應用水(100 mL)淬滅,並且將混合物用二氯甲烷(3×80 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過C18 FLASH色譜法,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3 .H 2O和乙腈的水作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的1.38 g 5-(4,5-二氟-2-羥基苯基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A111-1)(57%)。 1H NMR (DMSO-d 6, 400 MHz) δ 3.59 (3H, s), 3.81 (3H, s), 6.81-6.91 (1H, m), 7.33-7.41 (1H, d), 7.92 (1H, d), 8.61 (1H, d), 9.90 (1H, s)。 步驟2. 5-(4,5-二氟-2-{[(4 R)-4-甲基-5-{[5-(4-甲基哌嗪-1-基)-2-硝基苯基]氨基}戊基]氧基}苯基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A111-2)的製備。在氮氣氣氛下在室溫下向5-(4,5-二氟-2-羥基苯基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(500 mg,1.69 mmol,1當量)和(4 R)-4-甲基-5-{[5-(4-甲基哌嗪-1-基)-2-硝基苯基]氨基}戊-1-醇(INT-A25-3,569 mg,1.69 mmol,1當量)於甲苯(7 mL)中的攪拌混合物中分批添加2-(三丁基-λ5-膦亞基)乙腈(2.04 g,8.47 mmol,5當量)。將所得混合物在80℃下攪拌2 h。將混合物冷卻到室溫。將反應用水(200 mL)淬滅,並且將混合物用二氯甲烷(3×300 mL)萃取。將合併的有機層用鹽水(3×200 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過C18 FLASH,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3 .H 2O和乙腈的水作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃色油的620 mg 5-(4,5-二氟-2-{[(4R)-4-甲基-5-{[5-(4-甲基哌嗪-1-基)-2-硝基苯基]氨基}戊基]氧基}苯基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A111-2)(59%)。LCMS: m/z (ESI), [M+H] += 614.25。 1H NMR (DMSO-d 6, 400 MHz) δ 0.91 (4H, d), 1.20-1.32 (1H, m), 1.59-1.72 (3H, m), 1.75-1.87 (1H, m), 2.32 (3H, d), 3.04-3.13 (1H, m), 3.19-3.25 (1H, m), 3.320-3.36 (8H, m), 3.56 (3H, s), 3.76 (3H, s), 3.97 (3H, t), 5.76 (1H, s), 5.98 (1H, s), 6.40-6.46 (1H, m), 7.18-7.28 (1H, m), 7.40-7.50 (1H, m), 7.84-7.95 (2H, m), 8.37 (1H, t), 8.53 (1H, d)。 步驟3. 5-(2-{[(4 R)-5-{[2-氨基-5-(4-甲基哌嗪-1-基)苯基]氨基}-4-甲基戊基]氧基}-4,5-二氟苯基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A111-3)的製備。在氮氣氣氛下在室溫下向5-(4,5-二氟-2-{[(4 R)-4-甲基-5-{[5-(4-甲基哌嗪-1-基)-2-硝基苯基]氨基}戊基]氧基}苯基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(600 mg,0.97 mmol,1當量)和雷尼鎳(251 mg,2.93 mmol,3當量)於甲醇(5 mL)中的攪拌混合物中添加水合肼(98%)(2.45 mg,0.04 mmol,3當量)。將所得混合物在氮氣氣氛下在0℃下攪拌30分鐘。將混合物過濾,並且將濾餅用甲醇(3×10 mL)洗滌。將所得溶液在減壓下濃縮,以得到呈白色固體的310 mg 5-(2-{[(4 R)-5-{[2-氨基-5-(4-甲基哌嗪-1-基)苯基]氨基}-4-甲基戊基]氧基}-4,5-二氟苯基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A111-3)。此材料在不經進一步純化的情況下用於下一反應中。LCMS: m/z (ESI), [M+H] +=582.25。 步驟4. 5-(2-{[(4 R)-4-{[2-氨基-6-(4-甲基哌嗪-1-基)-1,3-苯並二氮唑-1-基]甲基}戊基]氧基}-4,5-二氟苯基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A111-4)的製備。將5-(2-{[(4 R)-5-{[2-氨基-5-(4-甲基哌嗪-1-基)苯基]氨基}-4-甲基戊基]氧基}-4,5-二氟苯基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(700 mg,1.20 mmol,1當量)和溴化氰(190 mg,1.80 mmol,1.5當量)於二氯甲烷(10 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將反應用水(10 mL)淬滅,並且將混合物用二氯甲烷(3×60 mL)萃取。將合併的有機層用鹽水(3×20 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過C18FLASH,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3 .H 2O和乙腈的水作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的120 mg 5-(2-{[(4 R)-4-{[2-氨基-6-(4-甲基哌嗪-1-基)-1,3-苯並二氮唑-1-基]甲基}戊基]氧基}-4,5-二氟苯基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A111-4)(16%)。LCMS: m/z (ESI), [M+H] +=609.15。 步驟5. 5-(2-{[(4R)-4-{[2-氨基-6-(4-甲基哌嗪-1-基)-1,3-苯並二氮唑-1-基]甲基}戊基]氧基}-4,5-二氟苯基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A111-5)的製備。將5-(2-{[(4 R)-4-{[2-氨基-6-(4-甲基哌嗪-1-基)-1,3-苯並二氮唑-1-基]甲基}戊基]氧基}-4,5-二氟苯基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(100 mg,0.16 mmol,1當量)和氫氧化鋰一水合物(10 mg,0.24 mmol,1.5當量)於四氫呋喃(2 mL)和水(0.5 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將反應用水(5 mL)淬滅,並且將混合物用二氯甲烷(3×10 mL)萃取。將合併的有機層用鹽水(3×5 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(10/1)洗脫進行純化,以得到呈白色固體的50 mg 5-(2-{[(4 R)-4-{[2-氨基-6-(4-甲基哌嗪-1-基)-1,3-苯並二氮唑-1-基]甲基}戊基]氧基}-4,5-二氟苯基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A111-5)(51%)。LCMS: m/z (ESI), [M+H] +=595.20。 步驟6. (12 R)-4,5-二氟-12,27-二甲基-17-(4-甲基哌嗪-1-基)-8-氧雜-14,21,23,27-四氮雜五環[23.3.1.0^{2,7}. 0^{14,22}.0^{15,20}]二十九-1(29),2(7),3,5,15,17,19,21, 25-壬烯-24,28-二酮(實施例A111)的製備。將5-(2-{[(4 R)-4-{[2-氨基-6-(4-甲基哌嗪-1-基)-1,3-苯並二氮唑-1-基]甲基}戊基]氧基}-4,5-二氟苯基)-1-甲基-6-氧代吡啶-3-羧酸(40 mg,0.06 mmol,1當量)、 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(38 mg,0.10 mmol,1.5當量)和 N,N-二異丙基乙胺(26 mg,0.20 mmol,3當量)於1,4-二噁烷(4 mL)中的混合物在氮氣氣氛下在室溫下攪拌3 h。將反應用水(10 mL)淬滅,並且將混合物用二氯甲烷(3×15 mL)萃取。將合併的有機層用鹽水(3×10 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(5/1)洗脫進行純化,以得到呈白色固體的9 mg (12 R)-4,5-二氟-12,27-二甲基-17-(4-甲基哌嗪-1-基)-8-氧雜-14,21,23,27-四氮雜五環[23.3.1.0^{2,7}.0^{14,22}. 0^{15,20}]二十九-1(29),2(7),3,5,15,17,19,21,25-壬烯-24,28-二酮(實施例A111)(22%)。LCMS: m/z (ESI), [M+H] +=577.30。 1H NMR (DMSO-d 6, 400 MHz) δ 0.92 (3H, d), 1.15-1.65 (3H, m), 2.00-2.10 (2H, m), 2.35-2.45 (4H, m), 2.50 (3H, s), 3.10-3.30 (4H, m), 3.32 (3H, s), 3.91-3.97 (2H, m), 4.05 (1H, d), 4.19 (1H, t), 6.88 (1H, dd), 7.11 (1H, s), 7.26 (1H, dd), 7.35 (1H, d), 7.76 (1H, dd), 8.38 (1H, d), 8.89 (1H, d), 12.31 (1H, s) 實施例A112 5,12,12,26-四甲基-16-(4-甲基哌嗪-1-基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮 步驟1. N-[(3E)-6-(苄氧基)-2-甲基己-3-烯-2-基]氨基甲酸叔丁酯(INT-A112-1)的製備。在氮氣氣氛下在0℃下向(3-苄氧基丙基)三苯基溴化膦(14.5 g,30 mmol,1.1當量)於四氫呋喃(100 mL)中的攪拌混合物中逐滴添加KHMDS(5.9 g,30 mmol,1.1當量)。將所得混合物在氮氣氣氛下在0℃下攪拌1 h。在0℃下在10分鐘內向上述混合物中添加 N-(2-甲基-1-氧代丙-2-基)氨基甲酸叔丁酯(5 g,26.7 mmol,1當量)。將所得混合物在室溫下攪拌過夜。將反應用水(50 mL)淬滅,並且將混合物用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用石油醚/乙酸乙酯(5/1)洗脫來進行純化,以得到呈無色油的6 g N-[(3E)-6-(苄氧基)-2-甲基己-3-烯-2-基]氨基甲酸叔丁酯(INT-A112-1)(70%)。 1H NMR (CDCl 3, 400 MHz) δ 1.44 (9H, s), 1.46 (6H, s), 2.50-2.62 (2H, m), 3.45-3.52 (2H, m), 4.56 (2H, s), 5.32-5.42 (1H, m), 5.56-6.02 (1H, m), 7.29-7.39 (5H, m)。 步驟2. N-(6-羥基-2-甲基己-2-基)氨基甲酸叔丁酯(INT-A112-2)的製備。將 N-[(3E)-6-(苄氧基)-2-甲基己-3-烯-2-基]氨基甲酸叔丁酯(2 g,6.26 mmol,1當量)和Pd/C(3.3 g,31 mmol,5當量)於甲醇(40 mL)中的混合物在氫氣氣氛下在室溫下攪拌16 h。將混合物過濾,並且將濾餅用甲醇(3×40 mL)洗滌。將所得溶液在減壓下濃縮,以得到呈無色油的1.4 g N-(6-羥基-2-甲基己-2-基)氨基甲酸叔丁酯(INT-A112-2)(97%)。 1H NMR (CDCl 3, 400 MHz) δ1.26 (6H, s), 1.32-1.41 (2H, m), 1.45 (9H, s), 1.60 (2H, d), 1.65-1.72 (2H, m), 3.62-3.73 (2H, m)。 步驟3. 5-氨基-5-甲基己-1-醇(INT-A112-3)的製備。將 N-(6-羥基-2-甲基己-2-基)氨基甲酸叔丁酯(1.3 g,5.6 mmol,1當量)於三氟乙酸(4 mL)和二氯甲烷(16 mL)中的溶液在氮氣氣氛下在室溫下攪拌2 h。將混合物在減壓下濃縮。將殘餘物用飽和NaHCO 3(水溶液)(30 mL)處理,並且用二氯甲烷(3×30 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(10/1)洗脫來進行純化,以得到呈無色油的680 mg 5-氨基-5-甲基己-1-醇(INT-A112-3)(92%)。 1H NMR (CD 3OD, 400 MHz) δ 1.35 (6H, d), 1.48-1.55 (2H, m), 1.65-1.72 (2H, m), 1.80-1.86 (2H, m), 4.40-4.49 (2H, m)。 步驟4. 5-((5-溴-2-硝基苯基)氨基)-5-甲基己-1-醇(INT-A112-4)的製備。將5-氨基-5-甲基己-1-醇(680 mg,5.18 mmol,1當量)、K 2CO 3(2150 mg,15.52 mmol,3當量)和4-溴-2-氟-1-硝基苯(1140 mg,5.18 mmol,1當量)於乙腈中的混合物在60℃下攪拌16 h,並且冷卻到室溫。將所得混合物過濾,並且將濾餅用二氯甲烷(3×20 mL)洗滌。將所得溶液在減壓下濃縮。將殘餘物通過Prep-TLC使用石油醚/乙酸乙酯(1/1)洗脫來進行純化,以得到呈黃色油的1.3 g 5-((5-溴-2-硝基苯基)氨基)-5-甲基己-1-醇(INT-A112-4)(76%)。LCMS: m/z (ES+), [M+H ] +=331.00。 步驟5. 5-(5-((5-((5-溴-2-硝基苯基)氨基)-5-甲基己基)氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-甲酸酯(INT-A112-5)的製備。在氮氣氣氛下在0℃下向5-(5-羥基-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M2,509 mg,1.93 mmol,1當量)和PPh 3(1520 mg,5.80 mmol,3當量)於四氫呋喃(20 mL)中的攪拌混合物中添加5-((5-溴-2-硝基苯基)氨基)-5-甲基己-1-醇(638 mg,1.93 mmol,1當量)和偶氮二甲酸二異丙酯(1172 mg,5.80 mmol,3當量)。將所得混合物在室溫下攪拌1 h。將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×60 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(20/1)洗脫進行純化,以得到呈黃色固體的610 mg 5-[5-({5-[(5-溴-2-硝基苯基)氨基]-5-甲基己基}氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A112-5)(55%)。LCMS: m/z (ESI), [M+H] +=578.30。 步驟6. 5-(5-((5-((2-氨基-5-溴苯基)氨基)-5-甲基己基)氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-甲酸甲酯(INT-A112-6)的製備。在氮氣氣氛下在室溫下向5-[5-({5-[(5-溴-2-硝基苯基)氨基]-5-甲基己基}氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(600 mg,1.04 mmol,1當量)和NH 2NH 2 .H 2O(104 mg,2.08 mmol,2當量)於甲醇(20 mL)中的攪拌混合物中添加雷尼鎳(445.88 mg,5.21 mmol,5當量)。將所得混合物在室溫下攪拌2 h。將所得混合物過濾,並且將濾餅用甲醇(3×20 mL)洗滌。將所得溶液在減壓下濃縮,以得到呈黃色固體的450 mg 5-(5-((5-((2-氨基-5-溴苯基)氨基)-5-甲基己基)氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-甲酸甲酯(INT-A112-6)。此材料在不經進一步純化的情況下用於下一反應中。LCMS: m/z (ESI), [M+H] +=546.05。 步驟7. 5-(5-((5-(2-氨基-6-溴-1H-苯並[d]咪唑-1-基)-5-甲基己基)氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-甲酸甲酯(INT-A112-7)的製備。將5-[5-({5-[(2-氨基-5-溴苯基)氨基]-5-甲基己基}氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(450 mg,0.82 mmol,1當量)和BrCN(174 mg,1.65mmol,2當量)於乙醇(15 mL)中的混合物在室溫下攪拌2 h。將反應用水(20 mL)淬滅,並且將混合物用二氯甲烷(3×20 mL)萃取。將合併的有機層用鹽水(3×20 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(10/1)洗脫進行純化,以得到呈黃色固體的300 mg 5-(5-{[5-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-5-甲基己基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A112-7)(64%)。 LCMS: m/z (ESI), [M+H] +=573.25。 1H NMR (DMSO, 400 MHz) δ 1.24 (2H, s), 1.66-1.72 (2H, m), 1.77 (6H, s), 1.99-2.07 (2H, m), 3.58 (6H, s), 3.80 (3H, s), 3.85-3.90 (2H, m), 6.31 (2H, s), 7.10 (2H, d), 7.61 (1H, s), 7.97 (1H, s), 8.09 (1H, d), 8.45 (1H, d)。 步驟8. 5-(5-((5-(2-氨基-6-溴-1,3-苯並[d]咪唑-1-基)-5-甲基己基)氧基)-1-甲基-1H-吡唑-4-基)-1-甲基-6-氧代-1,6-二氫吡啶-3-羧酸(INT-A112-8)的製備。在室溫下向5-[5-({5-[(氨基甲基)(3-溴苯基)氨基]-5-甲基己基}氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(280 mg,0.5 mmol,1當量)於THF(6 mL)和H 2O(1.5 mL)中的攪拌混合物中添加LiOH .H 2O(63 mg,1.5 mmol,3當量)。將所得混合物在氮氣氣氛下在室溫下攪拌16 h,並且在減壓下濃縮。將殘餘物通過C18 FLASH柱,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3 .H 2O和乙腈的水作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的150 mg 5-(5-{[5-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-5-甲基己基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A112-8)(54%)。LCMS: m/z (ESI), [M+H] +=557.25。 步驟9. 16-溴-5,12,12,26-四甲基-7-氧雜-4,5,13,20,22, 26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(INT-A112-9)的製備。將5-(5-{[5-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-5-甲基己基]氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(150 mg,0.27 mmol,1當量)、 N,N-二異丙基乙胺(104 mg,0.81 mmol,3當量)和 N,N,N,N-四甲基-O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(205 mg,0.54 mmol,2當量)於二噁烷(5 mL)中的混合物在氮氣氣氛下在60℃下攪拌2 h。將混合物冷卻到室溫。將反應用水(20 mL)淬滅,並且將混合物用二氯甲烷(3×20 mL)萃取。將合併的有機層用鹽水(3×20 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法使用二氯甲烷/甲醇(10/1)洗脫進行純化,以得到呈白色固體的140 mg 16-溴-5,12,12,26-四甲基-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3, 14,16,18,20,24-八烯-23,27-二酮(INT-A112-9)(96%)。LCMS: m/z (ESI), [M+H] +=539.20 步驟10. 5,12,12,26-四甲基-16-(4-甲基哌嗪-1-基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(28),2(6),3,14,16,18,20,24-八烯-23,27-二酮(實施例A112)的製備。在氮氣氣氛下在室溫下向16-溴-5,12,12,26-四甲基-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3, 14,16,18,20,24-八烯-23,27-二酮(70 mg,0.13 mmol,1當量)、BrettPhos(7 mg,0.013 mmol,0.1當量)和1-甲基-哌嗪(65 mg,0.65 mmol,5當量)於二噁烷(5 mL)中的攪拌混合物中逐滴添加LiHMDS(1.3 mL,1.3 mmol,10當量)。將混合物在氮氣氣氛下在60℃下攪拌2 h,並且冷卻到室溫。將反應用飽和NH 4Cl(水溶液)(10 mL)淬滅,並且將混合物用二氯甲烷(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法使用二氯甲烷/甲醇(12/1)洗脫進行純化,以得到呈灰白色固體的24.7 mg 5,12,12,26-四甲基-16-(4-甲基哌嗪-1-基)-7-氧雜-4,5,13,20,22,26-六氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(28),2(6),3, 14,16,18,20,24-八烯-23,27-二酮(實施例112)(33%)。LCMS: m/z (ESI), [M+H] +=559.30。 1H NMR (DMSO-d 6, 400 MHz) δ 1.20-1.26 (2H, m), 1.68-1.76 (4H, m), 1.92 (6H, s), 2.28 (3H, s), 2.40-2.60 (4H, m), 3.11 (4H, br s), 3.63 (3H, s), 3.70 (3H, s), 4.08 (2H, t), 6.92 (1H, dd), 7.27 (1H, d), 7.44 (1H, d), 8.27 (1H, d), 8.32 (1H, s), 8.81 (1H, d), 12.71 (1H, s)。 實施例A113 6-氟-15,21-二甲基-5-(嗎啉-4-基)-23-氧雜-2,9,11,15,20,21, 26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]三十四-3,5,7,9,13,17(34),18(22),19-八烯-12,16-二酮 步驟1. 2-{1-[(5-溴-4-氟-2-硝基苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙醇(INT-A113-1)的製備。在室溫下向3-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-3-氮雜雙環[3.2.2]壬-1-胺(M5-9,700 mg,2.35 mmol,1當量)於DMSO(10 mL)中的攪拌混合物中添加三甲胺(712 mg,7.04 mmol,3當量)和1-溴-2,5-二氟-4-硝基苯(1.12 g,4.69 mmol,2當量)。將所得混合物在氮氣氣氛下在100℃下攪拌2 h,並且冷卻到室溫。將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過C18 FLASH使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃色固體的800 mg 2-{1-[(5-溴-4-氟-2-硝基苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙醇(INT-A113-1)(85%)。LCMS: m/z (ESI), [M+H] +=404.00。 1H NMR (DMSO-d 6, 400 MHz) δ 1.49-1.61 (2H, m), 1.74-1.83 (4H, m), 1.90 (1H, s), 2.35-2.46 (2H, m), 2.55 (2H, t), 2.64 (2H, d), 2.86 (2H, s), 3.58 (2H, q), 4.43 (1H, t), 7.25 (1H, d), 7.99 (1H, s), 8.07 (1H, d)。 步驟2. 5-[5-(2-{1-[(5-溴-4-氟-2-硝基苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A113-2)的製備。在氮氣氣氛下在室溫下向2-{1-[(5-溴-4-氟-2-硝基苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙醇(785 mg,1.95 mmol,1當量)和5-(5-羥基-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M2,667 mg,2.54 mmol,1.3當量)於甲苯(20 mL)中的攪拌混合物中添加2-(三丁基-λ5-膦亞基)乙腈(2826 mg,11.71 mmol,6當量)。將所得混合物在100℃下攪拌3 h。將混合物冷卻到室溫。將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC進行純化並且使用石油醚/乙酸乙酯(1/2)洗脫,以得到呈黃色固體的670 mg 5-[5-(2-{1-[(5-溴-4-氟-2-硝基苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A113-2)(53%)。LCMS: m/z (ESI), [M+H] +=647.15。 步驟3. 5-[5-(2-{1-[(2-氨基-5-溴-4-氟苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A113-3)的製備。在室溫下向5-[5-(2-{1-[(5-溴-4-氟-2-硝基苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(660 mg,1.02 mmol,1當量)和雷尼鎳(175 mg,2.04 mmol,2當量)於甲醇(10 mL)中的攪拌混合物中添加NH 2NH 2.H 2O(153 mg,3.06 mmol,3當量)。將所得混合物在室溫下攪拌1 h。將反應用水(80 mL)淬滅,並且將混合物用二氯甲烷(3×80 mL)萃取。將合併的有機層用鹽水(3×80 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC進行純化並且使用石油醚/乙酸乙酯(1/32)洗脫,以得到呈棕色固體的490 mg 5-[5-(2-{1-[(2-氨基-5-溴-4-氟苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A113-3)(78%)。LCMS: m/z (ESI), [M+H] += 617.10。 步驟4. 5-(5-{2-[1-(2-氨基-6-溴-5-氟-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.2]壬-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A113-4)的製備。在室溫下向5-[5-(2-{1-[(2-氨基-5-溴-4-氟苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(480 mg,0.78 mmol,1當量)於乙醇(5 mL)中的攪拌混合物中添加BrCN(165 mg,1.55 mmol,2當量)。將所得混合物在60℃下攪拌1 h,並且冷卻到室溫。將反應用飽和硫代硫酸鈉(水溶液)(60 mL)淬滅,並且將混合物用二氯甲烷(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC進行純化並且使用二氯甲烷/甲醇(20/1)洗脫,以得到呈紅棕色固體的250 mg 5-(5-{2-[1-(2-氨基-6-溴-5-氟-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.2]壬-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A113-4)(50%)。LCMS: m/z (ESI), [M+H] +=642.15。 1H NMR (DMSO-d 6, 400 MHz) δ 1.04-1.30 (2H, m), 1.81 (1H, s), 1.80-1.98 (5H, m), 2.69 (2H, s), 2.81-2.94 (4H, m), 3.56 (3H, s), 3.65-3.77 (6H, m), 4.12 (2H, t), 6.11 (2H, s), 7.05 (1H, d), 7.48 (1H, d), 7.97 (1H, s), 8.10 (1H, d), 8.38 (1H, d)。 步驟5. 5-(5-{2-[1-(2-氨基-6-溴-5-氟-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.2]壬-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A113-5)的製備。在室溫下向5-(5-{2-[1-(2-氨基-6-溴-5-氟-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.2]壬-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(220 mg,0.34 mmol,1當量)於四氫呋喃(4 mL)中的攪拌溶液中添加含LiOH(21 mg,0.86 mmol,2.5當量)的H 2O(1 mL)。將所得混合物在60℃下攪拌1 h,並且冷卻到室溫。將混合物在減壓下濃縮。將殘餘物通過C18 FLASH色譜法使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的190 mg 5-(5-{2-[1-(2-氨基-6-溴-5-氟-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.2]壬-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A113-5)(88%)。LCMS: m/z (ESI), [M+H] +=630.10。 步驟6. 5-溴-6-氟-15,21-二甲基-23-氧雜-2,9,11,15,20, 21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]三十四-3,5,7,9,13,17(34),18(22),19-八烯-12,16-二酮(INT-A113-6)的製備。在室溫下向5-(5-{2-[1-(2-氨基-6-溴-5-氟-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.2]壬-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(190 mg,0.30 mmol,1當量)於1,4-二噁烷(5 mL)中的攪拌溶液中添加 N,N-二異丙基乙胺(117 mg,0.91 mmol,3當量)和 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(230 mg,0.60 mmol,2當量)。將所得混合物在氮氣氣氛下在60℃下攪拌1 h。將混合物冷卻到室溫。將反應用水(30 mL)淬滅,並且將混合物用二氯甲烷(3×30 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC進行純化並且使用二氯甲烷/甲醇(22/1)洗脫,以得到呈淡黃色固體的150 mg 5-溴-6-氟-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十四-3,5,7,9,13,17(34), 18(22),19-八烯-12,16-二酮(INT-A113-6)(81%)。LCMS: m/z (ESI), [M+H] +=610.30。 步驟7. 6-氟-15,21-二甲基-5-(嗎啉-4-基)-23-氧雜-2,9, 11,15,20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十四-3,5,7,9,13,17(34), 18(22),19-八烯-12,16-二酮(實施例A113)的製備。在氮氣氣氛下在室溫下向5-溴-6-氟-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十四-3,5,7,9,13,17(34), 18(22),19-八烯-12,16-二酮(50 mg,0.08 mmol,1當量)、BrettPhos Pd G 3(23 mg,0.03 mmol,0.3當量)和嗎啉(22 mg,0.25 mmol,3當量)與1,4-二噁烷(4 mL)中的攪拌混合物中添加LiHMDS(0.82 mL,0.82 mmol,10當量)。將所得混合物在氮氣氣氛下在60℃下攪拌30分鐘,並且冷卻到室溫。將反應用飽和NH 4Cl(水溶液)(80 mL)淬滅,並且將混合物用二氯甲烷(3×80 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC進行純化並且使用二氯甲烷/甲醇(25/1)洗脫。將產物通過prep-HPLC用XBridge Shield RP18 OBD柱,使用含有10 mmol/L NH 4HCO 3的水和乙腈作為流動相進行進一步純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的1.2 mg 6-氟-15,21-二甲基-5-(嗎啉-4-基)-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十四-3,5,7,9,13,17(34),18(22),19-八烯-12,16-二酮(實施例A113)(2%)。LCMS: m/z (ESI), [M+H] +=617.35。 1H NMR (CDCl 3, 400 MHz) δ 2.00-2.20 (9H, m), 2.84 (2H, d), 2.95 (2H, t), 3.06 (4H, t), 3.40-3.50 (2H, m), 3.71 (3H, s), 3.80 (3H, s), 3.91 (4H, t), 4.23 (2H, t), 7.01 (1H, d), 7.39 (1H, d), 8.23 (1H, d), 8.50 (1H, s), 9.19 (1H, d), 13.50 (1H, s)。 實施例A115 6-氟-15,21-二甲基-5-(4-甲基哌嗪-1-基)-23-氧雜-2,9, 11,15,20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十四-3,5,7,9,13,17(34), 18(22),19-八烯-12,16-二酮 在氮氣氣氛下在室溫下向5-溴-6-氟-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[26.2.2.1^{1,26}. 1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十四-3,5,7,9,13, 17(34),18(22),19-八烯-12,16-二酮(INT-A113-6,50 mg,0.08 mmol,1當量)、BrettPhos Pd G 3(23 mg,0.03 mmol,0.3當量)和1-甲基哌嗪(25 mg,0.25 mmol,3當量)與1,4-二噁烷(4 mL)中的攪拌混合物中緩慢添加LiHMDS(0.82 mL,0.82 mmol,10當量)。將所得混合物在60℃下攪拌30分鐘,然後冷卻到室溫。將反應用飽和NH 4Cl(水溶液)(80 mL)淬滅,並且將混合物用二氯甲烷(3×80 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-TLC進行純化並且使用二氯甲烷/甲醇(12/1)洗脫。將產物通過prep-HPLC用XBridge Prep OBD C18柱,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3.H 2O的水和乙腈作為流動相進行進一步純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的0.8 mg 6-氟-15,21-二甲基-5-(4-甲基哌嗪-1-基)-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十四-3,5,7,9,13,17(34), 18(22),19-八烯-12,16-二酮(實施例A115)(2%)。LCMS: m/z (ESI), [M+H] +=630.40。 1H NMR (CD 3OD, 400 MHz) δ 2.02-2.07 (4H, m), 2.08-2.19 (4H, m), 2.40 (3H, s), 2.62-2.78 (4H, m), 2.85-2.97 (4H, m), 3.03-3.13 (3H, m), 3.43-3.55 (4H, m), 3.69 (3H, s), 3.79 (3H, s), 4.31 (2H, t), 7.23 (1H, s), 7.51 (1H, d), 8.33 (1H, s), 8.34 (1H, s), 9.27 (1H, s)。 實施例A116 6-氟-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十四-3,5,7,9,13,17(34),18(22),19-八烯-12,16-二酮 在氮氣氣氛下在室溫下向5-溴-6-氟-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[26.2.2.1^{1,26}. 1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十四-3,5,7,9,13, 17(34),18(22),19-八烯-12,16-二酮(INT-A113-6,20 mg,0.03 mmol,1當量)和BrettPhos Pd G 3(9 mg,0.01 mmol,0.3當量)與1,4-二噁烷(2 mL)中的攪拌混合物中緩慢添加LiHMDS(0.33 mL,0.33 mmol,10當量)。將所得混合物在60℃下攪拌30分鐘,並且冷卻到室溫。將反應用飽和NH 4Cl(水溶液)(50 mL)淬滅。將混合物用二氯甲烷(3×50 mL)萃取。將合併的有機層用鹽水(3×20 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-TLC進行純化並且使用二氯甲烷/甲醇(22/1)洗脫。將產物通過prep-HPLC用XBridge Shield RP18 OBD柱,使用含有10 mmol/L NH 4HCO 3的水和乙腈作為流動相進行進一步純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的0.9 mg 6-氟-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18, 22}]三十四-3,5,7,9,13,17(34),18(22),19-八烯-12,16-二酮(實施例A116)(5%)。LCMS: m/z (ESI), [M+H] +=532.15。 1H NMR (DMSO-d 6, 400 MHz) δ 1.88-1.94 (4H, m), 1.96-2.13 (3H, m), 2.79 (2H, d), 2.86 (2H, t), 3.39-3.42 (4H, m), 3.62 (3H, s), 3.74 (3H, s), 4.28 (2H, t), 6.91-7.01 (1H, m), 7.45 (1H, dd), 7.87 (1H, dd), 8.27-8.35 (2H, m), 9.10 (1H, d), 13.34 (1H, s)。 實施例A117A、實施例A117B、實施例A118A和實施例A118B 6-氟-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮,異構體1(實施例A117A)和異構體2(實施例A117B);6-氟-15,21-二甲基-5-(嗎啉-4-基)-23-氧雜-2,9,11,15,20,21, 26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]三十三-3,5,7,9,13,17(33), 18(22),19-八烯-12,16-二酮,異構體1(實施例A118A)和異構體2(實施例A118B)。 步驟1. 3-(2-((叔丁基二甲基甲矽烷基)氧基)乙基)-3-氮雜雙環[3.2.1]辛-1-胺(INT-A117-1)的製備。將 N-(3-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-3-氮雜雙環[3.2.1]辛-1-基)氨基甲酸叔丁酯(M4-9,10 g,26 mmol,1當量)於三氟乙酸(35 mL)和二氯甲烷(100 mL)中的混合物在室溫下攪拌2 h。將所得混合物在減壓下濃縮。將殘餘物用飽和NaHCO 3(水溶液)(100 mL)處理,並且用二氯甲烷(3×100 mL)萃取。將合併的有機層用鹽水(3×200 mL)洗滌,經無水Na 2SO 4乾燥,並且在減壓下濃縮,以得到呈黃色固體的7.3 g 3-(2-((叔丁基二甲基甲矽烷基)氧基)乙基)-3-氮雜雙環[3.2.1]辛-1-胺(INT-A117-1)(98%)。使用粗產物,無需純化。LCMS: m/z (ESI), [M+H] +=285.15。 步驟2. 2-{1-[(5-溴-4-氟-2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙醇(INT-A117-2)的製備。將3-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-3-氮雜雙環[3.2.1]辛-1-胺(2 g,7.03 mmol,1當量)、三乙胺(2.84 g,28.11 mmol,4當量)和1-溴-2,5-二氟-4-硝基苯(5.01 g,21.08 mmol,3當量)於DMSO(30 mL)中的混合物在100℃下攪拌3 h。將混合物冷卻到室溫。將反應用水(50 mL)淬滅,並且將混合物用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上進行純化並且使用乙酸乙酯/石油醚(1/1)洗脫,以得到呈橙色油的1.5 g 2-{1-[(5-溴-4-氟-2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙醇(INT-A117-2)(55%)。LCMS: m/z (ESI), [M+H] +=387.95。 步驟3. 5-[5-(2-{1-[(5-溴-4-氟-2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A117-3)的製備。將2-{1-[(5-溴-4-氟-2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙醇(700 mg,1.80 mmol,1當量)、5-(5-羥基-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(M2,712 mg,2.70 mmol,1.5當量)和2-(三丁基膦烯)乙腈(2.61 g,10.82 mmol,6當量)於甲苯(20 mL)中的混合物在氮氣氣氛下在100℃下攪拌2 h。將混合物冷卻到室溫,並且在減壓下濃縮。將殘餘物用水(50 mL)處理,並且將混合物用CH 2Cl 2(3×50 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-TLC使用CH 2Cl 2/甲醇(25/1)洗脫進行純化,以得到呈黃色固體的700 mg 5-[5-(2-{1-[(5-溴-4-氟-2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A117-3)(61%)。LCMS: m/z (ESI), [M+H] +=633.35。 1H NMR (DMSO-d 6, 400 MHz) δ 1.53-1.74 (4H, m), 1.75-1.87 (1H, m), 1.93 (1H, s), 2.16 (1H, d), 2.21 (2H, d), 2.70 (1H, d), 2.76-2.86 (2H, m), 3.16 (1H, d), 3.57 (3H, s), 3.76 (6H, d), 4.09 (2H, t), 7.26 (1H, d), 7.97 (1H, s), 8.04-8.11 (2H, m), 8.17 (1H, s), 8.41 (1H, d)。 步驟4. 5-[5-(2-{1-[(2-氨基-5-溴-4-氟苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A117-4)的製備。在室溫下向5-[5-(2-{1-[(5-溴-4-氟-2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(680 mg,1.07 mmol,1當量)和雷尼鎳(184 mg,2.15 mmol,2當量)於甲醇(10 mL)中的攪拌混合物中添加水合肼(107 mg,2.15 mmol,2當量)。將所得混合物在室溫下攪拌1 h,並且過濾。將濾餅用甲醇(3×10 mL)洗滌。將濾液在減壓下濃縮,以得到呈白色固體的600 mg 5-[5-(2-{1-[(2-氨基-5-溴-4-氟苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A117-4)(99%)。將粗產物在不純化的情況下用於下一反應。LCMS: m/z (ESI), [M+H] += 603.10。 步驟5. 5-(5-{2-[1-(2-氨基-6-溴-5-氟-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A117-5)的製備。將5-[5-(2-{1-[(2-氨基-5-溴-4-氟苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙氧基)-1-甲基吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(550 mg,0.91 mmol,1當量)和BrCN(193 mg,1.82 mmol,2當量)於乙醇(10 mL)中的混合物在氮氣氣氛下在室溫下攪拌過夜。將反應混合物在真空下濃縮。將殘餘物通過與 N, N-二甲基甲醯胺(10 mL)一起研磨進行純化,以得到呈黃色固體的280 mg 5-(5-{2-[1-(2-氨基-6-溴-5-氟-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A117-5)(48%)。LCMS: m/z (ESI), [M+H] +=628.10。 步驟6. 5-(5-{2-[1-(2-氨基-6-溴-5-氟-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(異構體1,INT-A117A-6和異構體2,INT-A117B-6)的製備。將5-(5-{2-[1-(2-氨基-6-溴-5-氟-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(200 mg)的外消旋混合物通過prep-手性-HPLC使用CHIRALPAK IG的柱(2×25 cm,5 μm)用己烷(含有0.1%二乙胺)的流動相A和乙醇/CH 2Cl 2(1/1)的流動相B洗脫來分離,以得到呈灰白色固體的5-(5-{2-[1-(2-氨基-6-溴-5-氟-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯、90 mg異構體1(INT-A117A-6,45%)和90 mg異構體2(INT-A117B-6,45%)。 INT-A117A-6:手性-HPLC,Rt=3.619分鐘。 INT-A117B-6:手性-HPLC,Rt=4.508分鐘。 步驟7. 5-(5-{2-[1-(2-氨基-6-溴-5-氟-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A117A-7)的製備。將5-(5-{2-[1-(2-氨基-6-溴-5-氟-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A117A-6,80 mg,0.13 mmol,1當量)和LiOH(6 mg,0.25 mmol,2當量)於THF(2 mL)和H 2O(0.5 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將混合物在減壓下濃縮,以得到呈黃色固體的65 mg 5-(5-{2-[1-(2-氨基-6-溴-5-氟-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A117A-7)(83%)。此材料在不經進一步純化的情況下用於下一步驟中。LCMS: m/z (ESI), [M+H] +=614.20。 步驟8. 5-溴-6-氟-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33), 18(22),19-八烯-12,16-二酮(INT-A117A-8)的製備。將5-(5-{2-[1-(2-氨基-6-溴-5-氟-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A117A-7,60 mg,0.10 mmol,1當量)、 N, N, N, N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(74 mg,0.20 mmol,2當量)和 N,N-二異丙基乙胺(38 mg,0.30 mmol,3當量)於1,4-二噁烷(6 mL)中的混合物在氮氣氣氛下在60℃下攪拌2 h。將混合物冷卻到室溫。將反應用水(20 mL)淬滅,並且將混合物用CH 2Cl 2(3×30 mL)萃取。將合併的有機層經無水Na 2SO 4乾燥,並且在減壓下濃縮。將殘餘物通過prep-TLC進行純化並且使用二氯甲烷/甲醇(12/1)洗脫,以得到呈淡黃色固體的50 mg 5-溴-6-氟-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮(INT-A117A-8)(85%)。LCMS: m/z (ESI), [M+H] +=594.05。 步驟9. 6-氟-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33), 18(22),19-八烯-12,16-二酮(異構體1,實施例A117A)和6-氟-15,21-二甲基-5-(嗎啉-4-基)-23-氧雜-2,9,11,15,20,21, 26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮(異構體1,實施例A118A)的製備。 將5-溴-6-氟-15,21-二甲基-23-氧雜-2,9,11,15,20,21, 26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮(INT-A117A-8,50 mg,0.08 mmol,1當量)、嗎啉(146 mg,1.68 mmol,20當量)、BrettPhos Pd G 3(22 mg,0.03 mmol,0.3當量)和t-BuOK(113 mg,1.01 mmol,12當量)於1,4-二噁烷(4 mL)中的混合物在氮氣氣氛下在80℃下攪拌2 h。將混合物冷卻到室溫。將反應用水(20 mL)淬滅,並且將混合物用CH 2Cl 2(3×30 mL)萃取。將合併的有機層經無水Na 2SO 4乾燥,並且在減壓下濃縮。將殘餘物通過prep-TLC進行純化並且使用二氯甲烷/甲醇(12/1)洗脫。將產物通過Prep-HPLC用XBridge Shield RP18 OBD柱,使用含有0.1% NH 4HCO 3的水和乙腈作為流動相進行進一步純化。將含有期望化合物的級分蒸發到乾燥,以得到呈淡黃色固體的5.5 mg 6-氟-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33), 18(22),19-八烯-12,16-二酮(異構體1,實施例A117A,8%)和8.1 mg 6-氟-15,21-二甲基-5-(嗎啉-4-基)-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33), 18(22),19-八烯-12,16-二酮(異構體1,實施例A118A,11%)。 實施例117A:LCMS: m/z (ESI), [M+H] +=518.25。 1H NMR (DMSO-d 6, 400 MHz) δ 1.52-1.83 (3H, m), 2.02 (1H, d), 2.38-2.43 (2H, m), 2.51-2.65 (2H, m), 2.72-2.78 (1H, m), 3.21-3.27 (2H, m), 3.37-3.46 (1H, m), 3.63 (3H, s), 3.64-3.70 (1H, m), 3.72 (3H, s), 4.57 (1H, t), 4.71 (1H, d), 6.98-7.03 (1H, m), 7.37 (1H, dd), 7.72 (1H, dd), 8.29 (1H, d), 8.41 (1H, d), 8.83 (1H, s), 12.82 (1H, s)。 實施例118A:LCMS: m/z (ESI), [M+H] +=603.20。 1H NMR (DMSO-d 6, 400 MHz) δ 1.48-1.82 (3H, m), 2.02 (1H, d), 2.38-2.49 (2H, m), 2.51-2.56 (2H, m), 2.62-2.77 (2H, m), 2.85-3.07 (4H, m), 3.21-3.27 (1H, m), 3.37-3.46 (1H, m), 3.62 (3H, s), 3.63-3.70 (1H, m), 3.72 (3H, s), 3.77 (4H, t), 4.57 (1H, t), 4.71 (1H, d), 7.21-7.43 (2H, m), 8.28 (1H, d), 8.41 (1H, d), 8.81 (1H, s), 12.71 (1H, s)。 使用與實施例A117A的合成中所述的相同反應順序由呈淡黃色固體的INT-A117B-6製備實施例A117B,6-氟-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮(異構體2)。LCMS: m/z (ESI), [M+H] +=518.30。 1H NMR (DMSO-d 6, 400 MHz) δ 1.56-1.82 (3H, m), 2.02 (1H, d), 2.38-2.45 (2H, m), 2.51-2.55 (1H, m), 2.62-2.78 (2H, m), 3.15-3.25 (2H, m), 3.37-3.46 (1H, m), 3.63 (3H, s), 3.64-3.70 (1H, m), 3.72 (3H, s), 4.53 (1H, t), 4.65 (1H, d), 6.98-7.04 (1H, m), 7.37 (1H, dd), 7.72 (1H, dd), 8.29 (1H, d), 8.41 (1H, d), 8.83 (1H, s), 12.82 (1H, s)。 使用與實施例A118A的合成中所述的相同反應順序由呈淡黃色固體的INT-A117B-6製備實施例A118B,6-氟-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮(異構體2)。LCMS: m/z (ESI), [M+H] +=603.30。 1H NMR (DMSO-d 6, 400 MHz) δ 1.52-1.82 (3H, m), 2.03 (1H, d), 2.38-2.45 (2H, m), 2.51-2.62 (3H, m), 2.65-3.12 (6H, m), 3.21-3.27 (1H, m), 3.37-3.46 (1H, m), 3.62 (3H, s), 3.63-3.70 (1H, m), 3.72 (3H, s), 3.77 (4H, t), 4.55 (1H, t), 4.67 (1H, d), 7.29 (1H, d), 7.37 (1H, d), 8.27 (1H, s), 8.41 (1H, s), 8.81 (1H, s), 12.71 (1H, s)。 實施例A119 5-[4-(二甲基氨基)哌啶-1-基]-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十四-3,5,7,9,13,17(34), 18(22),19-八烯-12,16-二酮 在氮氣氣氛下在室溫下向5-溴-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十四-3,5,7,9,13,17(34), 18(22),19-八烯-12,16-二酮(M5,100 mg,0.16 mmol,1當量)、 N,N-二甲基哌啶-4-胺(64.92 mg,0.50 mmol,3當量)和BrettPhos Pd G 3(45.90 mg,0.05 mmol,0.3當量)與1,4-二噁烷(10 mL)中的攪拌混合物中逐滴添加LiHMDS(1 mL,1.014 mmol,6當量)。將混合物在60℃下攪拌1 h,然後冷卻到室溫。將反應用飽和NH 4Cl(水溶液)(15 mL)淬滅,並且將混合物用二氯甲烷(3×20 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-HPLC用XBridge Shield RP18 OBD柱,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3.H 2O的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈淡黃色固體的50.1 mg 5-[4-(二甲基氨基)哌啶-1-基]-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十四-3,5,7,9,13,17(34), 18(22),19-八烯-12,16-二酮(實施例A119)(46%)。LCMS: m/z (ESI), [M+H] +=640.40。 1H NMR (DMSO-d 6, 400 MHz) δ 1.47-1.59 (2H, m), 1.81-1.99 (6H, m), 2.00-2.13 (3H, m), 2.22 (6H, s), 2.57-2.70 (2H, m), 2.75-2.82 (2H, m), 2.86 (2H, t), 3.38-3.50 (4H, m), 3.50-3.59 (3H, m), 3.62 (3H, s), 3.74 (3H, s), 4.27 (2H, t), 6.91 (1H, d), 7.34 (1H, s), 7.45 (1H, d), 8.30 (2H, d), 9.11 (1H, d), 13.18 (1H, s)。 實施例A120-實施例A128(表2)使用上文針對由M5合成實施例A119所描述的相同的反應順序來製備。 實施例A129B 21-甲基-5-(嗎啉-4-基)-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18, 22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮,異構體2(實施例A129B) 步驟1. 1-苄基-2-甲基吡唑-3-酮(INT-A129-1)的製備。將2-甲基-1H-吡唑-3-酮(10 g,101.93 mmol,1當量)、K 2CO 3(42.26 g,305.79 mmol,3當量)和BnBr(18.31 g,107.02 mmol,1.05當量)於乙腈(100 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將反應用水(300 mL)淬滅,並且將混合物用乙酸乙酯(3×200 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過C18 FLASH使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈棕色固體的5.5 g 1-苄基-2-甲基吡唑-3-酮(INT-A129-1) (28%)。LCMS: m/z (ESI), [M+H] +=189.10。 1H NMR (DMSO-d 6 , 400 MHz) δ 3.14 (3H, s), 4.97 (2H, s), 5.23 (1H, d), 7.14-7.22 (2H, m), 7.26-7.38 (3H, m), 7.92 (1H, d)。 步驟2. 1-苄基-4-碘-2-甲基吡唑-3-酮(INT-A129-2)的製備。向1-苄基-2-甲基吡唑-3-酮(6.35 g,33.73 mmol,1當量)於乙腈(200 mL)中的攪拌混合物中分批添加NIS(8.35 g,37.10 mmol,1.1當量),並且在氮氣氣氛下在0℃下攪拌2 h。將反應用飽和硫代硫酸鈉溶液(200 mL)淬滅,並且將混合物用乙酸乙酯(3×300 mL)萃取。將合併的有機層用鹽水(3×300 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過C18 FLASH使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈紅棕-棕色半固體的9.5 g 1-苄基-4-碘-2-甲基吡唑-3-酮(INT-A129-2)(89%)。LCMS: m/z (ESI), [M+H] +=315.05。 1H NMR (DMSO-d 6 , 400 MHz) δ 3.22 (3H, s), 5.00 (2H, s), 7.16-7.41 (5H, m), 8.18 (1H, s)。 步驟3. 5-溴-6-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡啶-3-甲酸甲酯(INT-A129-3)的製備。在氮氣氣氛下在0℃下向5-溴-6-羥基吡啶-3-甲酸甲酯(10 g,43.09 mmol,1當量)和K 2CO 3(11.91 g,86.194 mmol,2當量)於乙腈(200 mL)中的攪拌混合物中逐滴添加2-(三甲基甲矽烷基)乙氧基甲基氯(10.78 g,64.64 mmol,1.5當量)。將所得混合物在室溫下攪拌3 h。將反應用水(200 mL)淬滅,並且將混合物用乙酸乙酯(3×200 mL)萃取。將合併的有機層用鹽水(3×200 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物從石油醚/乙酸乙酯(20/1)中重結晶,以得到呈白色固體的8 g 5-溴-6-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡啶-3-甲酸甲酯(INT-A129-3)(51%)。LCMS: m/z (ESI), [M+H] +=364.10。 1H NMR (DMSO-d 6 , 400 MHz) δ 0.00 (9H, s), 0.89-0.94 (2H, m), 3.60-3.68 (2H, m), 3.84 (3H, s), 5.44 (2H, s), 8.24 (1H, d), 8.58 (1H, d)。 步驟4. 6-氧代-5-(4,4,5,5-四甲基-1,3,2-二氧雜環戊硼烷-2-基)-1-((2-(三甲基甲矽烷基)乙氧基)甲基)-1,6-二氫吡啶-3-甲酸甲酯(INT-A129-4)的製備。將5-溴-6-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡啶-3-甲酸甲酯(6.89 g,19.01 mmol,1當量)、KOAc(5.60 g,57.054 mmol,3當量)、Pd(dppf)Cl 2CH 2Cl 2(1.55 g,1.90 mmol,0.1當量)和雙(頻哪醇)二硼(7.24 g,28.52 mmol,1.5當量)於1,4-二噁烷(100 mL)中的混合物在氮氣氣氛下在80℃下攪拌2 h,然後冷卻到室溫。將此混合物在不進行進一步後處理的情況下直接用於下一步驟。LCMS: m/z (ESI), [M+H] +=328.05。 步驟5. 5-(1-苄基-2-甲基-3-氧代吡唑-4-基)-6-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡啶-3-甲酸甲酯(INT-A129-5)的製備。將新鮮製備的6-氧代-5-(4,4,5,5-四甲基-1,3,2-二氧雜環戊硼烷-2-基)-1-((2-(三甲基甲矽烷基)乙氧基)甲基)-1,6-二氫吡啶-3-甲酸甲酯(INT-A129-4)、K 2CO 3(5.28 g,38.20 mmol,3當量)、Pd(dppf)Cl 2CH 2Cl 2(1.04 g,1.27 mmol,0.1當量)和1-苄基-4-碘-2-甲基吡唑-3-酮(4 g,12.73 mmol,1.00當量)於二噁烷(100 mL)和水(25 mL)中的混合物在氮氣氣氛下在80℃下攪拌2 h。將反應混合物冷卻到室溫,用水(100 mL)處理,並且用二氯甲烷(3×100 mL)萃取。將合併的有機層用鹽水(3×300 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過反向C18 FLASH色譜法使用乙腈和水(0%至100%梯度)作為移動相進行純化,以得到呈棕色固體的2.5 g 5-(1-苄基-2-甲基-3-氧代吡唑-4-基)-6-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡啶-3-甲酸甲酯(INT-A129-5)(41%)。LCMS: m/z (ESI), [M+H] +=470.30。 步驟6. 5-(5-羥基-1-甲基吡唑-4-基)-6-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡啶-3-甲酸甲酯(INT-A129-6)的製備。將5-(1-苄基-2-甲基-3-氧代吡唑-4-基)-6-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡啶-3-甲酸甲酯(2.5 g,5.32 mmol,1.00當量)和Pd/C(2.83 g,26.62 mmol,5當量)於甲醇(50 mL)中的混合物在氫氣氣氛下在室溫下攪拌過夜。將所得混合物過濾。將濾餅用甲醇(3×100 mL)洗滌。將濾液在減壓下濃縮,以得到呈棕色固體的1.5 g 5-(5-羥基-1-甲基吡唑-4-基)-6-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡啶-3-甲酸甲酯(INT-A129-6)(74%)。LCMS: m/z (ESI), [M+H] +=380.00。 步驟7. 2-{1-[(5-氟-2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙醇(INT-A129-7)的製備。將2-{1-氨基-3-氮雜雙環[3.2.1]辛-3-基}二鹽酸乙醇(4.8 g,19.75 mmol,1當量)、三甲胺(9.99 g,98.77 mmol,5當量)和2,4-二氟-1-硝基苯(4.71 g,29.63 mmol,1.5當量)於乙腈(100 mL)中的混合物氮氣氣氛下在60℃下攪拌2 h,並且冷卻到室溫。將反應用水(300 mL)淬滅,並且將混合物用乙酸乙酯(3×200 mL)萃取。將合併的有機層用鹽水(3×300 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上使用石油醚/乙酸乙酯(2/1)洗脫來進行純化,以得到呈橙色油的5.5 g 2-{1-[(5-氟-2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙醇(INT-A129-7)(90 %)。LCMS: m/z (ESI), [M+H] +=310.10。 1H NMR (DMSO-d 6 , 400 MHz) δ 1.58-1.73 (3H, m), 1.76-1.98 (2H, m), 2.08-2.25 (4H, m), 2.36-2.51 (2H, m), 2.68 (1H, dd), 3.12-3.20 (1H, m), 3.44-3.58 (2H, m), 4.35 (1H, t), 6.53-6.62 (1H, m), 6.83 (1H, dd), 8.19 (1H, dd), 8.43-8.48 (1H, m)。 步驟8. 2-{1-[(5-氟-2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙醇(異構體2,INT-A129-7B)的製備。將2-{1-[(5-氟-2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙醇(15 g)的外消旋混合物通過Prep_SFC(CHIRALPAK IG,3*25 cm,5 μm),用CO 2的流動相A和甲醇的流動相B洗脫來分離,以得到呈橙色油的6.2 g(41%)異構體1(INT-A129-7A)和呈淡黃色油的6.5 g異構體2(INT-A129-7B) (43%)。 異構體1:LCMS: m/z (ESI), [M+H] +=310.20。Rt= 1.429分鐘。 異構體2:LCMS: m/z (ESI), [M+H] +=310.20。 1H NMR (DMSO-d 6 , 400 MHz) δ 1.59-1.73 (3H, m), 1.82 (1H, d), 1.94 (1H, s), 2.17 (2H, dd), 2.44 (2H, d), 2.68 (1H, d), 3.17 (2H, d), 3.51 (2H, s), 4.11 (1H, q), 4.35 (1H, d), 6.58 (1H, t), 6.83 (1H, d), 8.19 (1H, dd), 8.45 (1H, s)。Rt=1.987分鐘。 步驟9. 2-(1-{[5-(嗎啉-4-基)-2-硝基苯基]氨基}-3-氮雜雙環[3.2.1]辛-3-基)乙醇(INT-A129-9B)的製備。將2-{1-[(5-氟-2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙醇(INT-A129-7B,6.5 g,21.01 mmol,1當量)、嗎啉(6.4 g,73.54 mmol,3.5當量)和三甲胺(6.38 g,63.03 mmol,3當量)於二甲亞碸(100 mL)中的混合物在氮氣氣氛下在120℃下攪拌2 h。將混合物冷卻到室溫。將反應用水(800 mL)淬滅,並且將混合物用乙酸乙酯(3×500 mL)萃取。將合併的有機層用鹽水(3×500 mL)洗滌,經無水Na 2SO 4乾燥,並且在減壓下濃縮,以得到呈橙色固體的7.4 g 2-(1-{[5-(嗎啉-4-基)-2-硝基苯基]氨基}-3-氮雜雙環[3.2.1]辛-3-基)乙醇(INT-A129-9B)(93%)。LCMS: m/z (ESI), [M+H] +=377.25。 1H NMR (DMSO-d 6 , 400 MHz) δ 1.63-1.71 (3H, m), 1.83-1.87 (1H, m), 1.92-1.96 (1H, m), 2.05 (1H, d), 2.14-2.22 (2H, m), 2.32-2.51 (3H, m), 2.51-2.57 (1H, m), 2.65 (1H, dd), 3.31-3.37 (4H, m), 3.45-3.56 (2H, m), 3.73 (4H, t), 4.35 (1H, t), 6.09 (1H, d), 6.46 (1H, dd), 7.93 (1H, d), 8.67 (1H, s)。 步驟10. 5-(1-甲基-5-{2-[1-{[5-(嗎啉-4-基)-2-硝基苯基]氨基}-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}吡唑-4-基)-6-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡啶-3-甲酸甲酯(INT-A129-10B)的製備。在氮氣氣氛下在0℃下向5-(5-羥基-1-甲基吡唑-4-基)-6-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡啶-3-甲酸甲酯(INT-A129-6,756 mg,1.99 mmol,1.5當量)、2-[1-{[5-(嗎啉-4-基)-2-硝基苯基]氨基}-3-氮雜雙環[3.2.1]辛-3-基]乙醇(INT-A129-9B,500 mg,1.32 mmol,1.00當量)和三苯基膦(1045 mg,3.98 mmol,3當量)於四氫呋喃(10 mL)中的攪拌混合物中逐滴添加偶氮二甲酸二異丙酯(805 mg,3.98 mmol,3當量)。將所得混合物在氮氣氣氛下在0℃下攪拌2 h。將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在Prep-TLC上使用石油醚/乙酸乙酯(1/3)洗脫進行純化,以得到呈橙色固體的400 mg 5-(1-甲基-5-{2-[1-{[5-(嗎啉-4-基)-2-硝基苯基]氨基}-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}吡唑-4-基)-6-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡啶-3-甲酸甲酯(INT-A129-10B)(40%)。LCMS: m/z (ESI), [M+H] += 738.60。 步驟11. 5-(5-{2-[1-{[2-氨基-5-(嗎啉-4-基)苯基]氨基}-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-6-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡啶-3-甲酸甲酯(INT-A129-11B)的製備。向5-(1-甲基-5-{2-[1-{[5-(嗎啉-4-基)-2-硝基苯基]氨基}-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}吡唑-4-基)-6-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡啶-3-甲酸甲酯(INT-A129-10B,380 mg,0.51 mmol,1當量)和雷尼鎳(220.59 mg,2.57 mmol,5當量)於甲醇(10 mL)中的攪拌混合物中逐滴添加水合肼(98%) (38.67 mg,0.77 mmol,1.5當量),並且在氮氣氣氛下在0℃下攪拌2 h。將所得混合物過濾。將濾餅用甲醇(3×50 mL)洗滌。將濾液在減壓下濃縮。將粗產物在不進行進一步純化的情況下直接用於下一步驟。LCMS: m/z (ESI), [M+H] +=708.70。 步驟12. 5-(5-{2-[1-[2-氨基-6-(嗎啉-4-基)-1,3-苯並二氮唑-1-基]-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-6-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡啶-3-甲酸甲酯(INT-A129-12B)的製備。將5-(5-{2-[1-{[2-氨基-5-(嗎啉-4-基)苯基]氨基}-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-6-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡啶-3-甲酸甲酯(INT-A129-11B)和BrCN(62.54 mg,0.59 mmol,1.1當量)於乙醇(10 mL)中的混合物在氮氣氣氛下在室溫下攪拌過夜。將反應用飽和NaHCO 3溶液(50 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(15/1)洗脫進行純化,以得到呈棕色固體的250 mg 5-(5-{2-[1-[2-氨基-6-(嗎啉-4-基)-1,3-苯並二氮唑-1-基]-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-6-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡啶-3-甲酸甲酯(INT-A129-12B)(63%)。LCMS: m/z (ESI), [M+H] +=733.65。 步驟13. 5-(5-{2-[1-[2-氨基-6-(嗎啉-4-基)-1,3-苯並二氮唑-1-基]-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-6-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡啶-3-羧酸(INT-A129-13B)的製備。將5-(5-{2-[1-[2-氨基-6-(嗎啉-4-基)-1,3-苯並二氮唑-1-基]-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-6-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡啶-3-甲酸甲酯(INT-A129-12B,80 mg,0.10 mmol,1當量)和LiOH(4 mg,0.17 mmol,1.5當量)於四氫呋喃(2 mL)和水(0.5 mL)中的混合物在氮氣氣氛下在室溫下攪拌過夜。將所得混合物在減壓下濃縮。將所得混合物在不進行進一步純化的情況下直接用於下一步驟。LCMS: m/z (ESI), [M+H] +=719.55。 步驟14. 21-甲基-5-(嗎啉-4-基)-15-{[2-(三甲基甲矽烷基)乙氧基]甲基}-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮(INT-A129-14B)的製備。將5-(5-{2-[1-[2-氨基-6-(嗎啉-4-基)-1,3-苯並二氮唑-1-基]-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-6-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡啶-3-羧酸(INT-A129-13B,80 mg,0.10 mmol,1當量)、 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(63 mg,0.16 mmol,1.5當量)和 N,N-二異丙基乙胺(43 mg,0.33 mmol,3當量)於二噁烷(5 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將反應用水(50 mL)淬滅,並且將混合物用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(20/1)洗脫進行純化,以得到呈白色固體的50 mg 21-甲基-5-(嗎啉-4-基)-15-{[2-(三甲基甲矽烷基)乙氧基]甲基}-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33), 18(22),19-八烯-12,16-二酮(INT-A129-14B)(64%)。LCMS: m/z (ESI), [M+H] +=701.60。 步驟15. 21-甲基-5-(嗎啉-4-基)-23-氧雜-2,9,11,15,20, 21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮(實施例A129B)的製備。將21-甲基-5-(嗎啉-4-基)-15-{[2-(三甲基甲矽烷基)乙氧基]甲基}-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33), 18(22),19-八烯-12,16-二酮(INT-A129-14B,45 mg,0.06 mmol,1當量)於1,4-二噁烷(2 mL)的HCl溶液中的混合物在氮氣氣氛下在室溫下攪拌2 h。將混合物用飽和NaHCO 3溶液中和到pH 7。將混合物用二氯甲烷(3×20 mL)萃取,並且將合併的有機層在減壓下濃縮。將殘餘物通過Prep-HPLC(XBridge Prep OBD C18柱,30*150 mm,5μm)使用水(含有10 mmol/L NH 4HCO 3和0.05% NH 3H 2O)的流動相A和CAN的流動相B進行純化,以得到呈白色固體的14.3 mg 21-甲基-5-(嗎啉-4-基)-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18, 22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮(39%)。LCMS: m/z (ESI), [M+H] +=571.45。 1H NMR (DMSO-d 6 , 400 MHz) δ 1.50-1.83 (4H, m), 2.00 (1H, d), 2.71 (2H, s), 3.05-3.15 (5H, m), 3.17-3.23 (3H, m), 3.62-3.92 (9H, m), 4.56 (1H, t), 4.65-4.72 (1H, m), 6.90 (1H, d), 7.19 (1H, s), 7.41 (1H, d), 7.87 (1H, s), 8.40 (1H, s), 8.82 (1H, d), 11.96 (1H, s), 12.62 (1H, s)。 實施例A130B 15-甲基-5-(嗎啉-4-基)-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18, 22}]三十三-3,5,7,9,13,17(33),18,21-八烯-12,16-二酮,異構體2(實施例A130B) 步驟1. 3-羥基-4-碘吡唑-1-甲酸叔丁酯(INT-A130-1)的製備。在氮氣氣氛下在0℃下向3-羥基吡唑-1-甲酸叔丁酯(4 g,21.71 mmol,1當量)於乙腈(80 mL)中的攪拌混合物中分批添加NIS(5.86 g,26.05 mmol,1.2當量)。將所得混合物在氮氣氣氛下在0℃下攪拌2 h。將反應用飽和硫代硫酸鈉溶液淬滅,並且將混合物用乙酸乙酯(3×200 mL)萃取。將合併的有機層用鹽水(3×200 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過C18 FLASH使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈棕色固體的2.5 g 3-羥基-4-碘吡唑-1-甲酸叔丁酯(40%)。LCMS: m/z (ESI), [M+H] += 311.00。 1H NMR (DMSO-d 6 , 400 MHz) δ 1.53 (9H, s), 8.18 (1H, s), 11.63 (1H, s)。 步驟2. 4-碘-3-{2-[1-{[5-(嗎啉-4-基)-2-硝基苯基]氨基}-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}吡唑-1-甲酸叔丁酯(INT-A130-2B)的製備。在氮氣氣氛下在0℃下向2-[1-{[5-(嗎啉-4-基)-2-硝基苯基]氨基}-3-氮雜雙環[3.2.1]辛-3-基]乙醇(INT-A129-9B,500 mg,1.32 mmol,1.00當量)和三苯基膦(1045 mg,3.98 mmol,3當量)於四氫呋喃(10 mL)中的攪拌混合物中添加偶氮二甲酸二異丙酯(805 mg,3.98 mmol,3當量)和3-羥基-4-碘吡唑-1-甲酸叔丁酯(617 mg,1.99 mmol,1.5當量)。將所得混合物在氮氣氣氛下在0℃下攪拌2 h。將反應用水(100 mL)淬滅,並且將混合物用二氯甲烷(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用石油醚/乙酸乙酯(1/1)洗脫來進行純化,以得到呈黃色固體的550 mg 4-碘-3-{2-[1-{[5-(嗎啉-4-基)-2-硝基苯基]氨基}-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}吡唑-1-甲酸叔丁酯(61%)。LCMS: m/z (ESI), [M+H] += 669.40。 步驟3. 5-[1-(叔丁氧基羰基)-3-{2-[1-{[5-(嗎啉-4-基)-2-硝基苯基]氨基}-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(INT-A130-3B)的製備。將4-碘-3-{2-[1-{[5-(嗎啉-4-基)-2-硝基苯基]氨基}-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}吡唑-1-甲酸叔丁酯(INT-A130-3B,550 mg,0.823 mmol,1當量)、K 2CO 3(341 mg,2.46 mmol,3當量)、Pd(dppf)Cl 2CH 2Cl 2(67 mg,0.08 mmol,0.1當量)和1-甲基-6-氧代-5-(4,4,5,5-四甲基-1,3,2-二氧雜環戊硼烷-2-基)吡啶-3-甲酸甲酯(361 mg,1.23 mmol,1.5當量)於二噁烷(20 mL)和水(5 mL)中的混合物在氮氣氣氛下在80℃下攪拌2 h。將混合物冷卻到室溫,用水(100 mL)處理,並且用二氯甲烷(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過C18 FLASH,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3 .H 2O的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃色固體的130 mg 5-[1-(叔丁氧基羰基)-3-{2-[1-{[5-(嗎啉-4-基)-2-硝基苯基]氨基}-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(22%)。LCMS: m/z (ESI), [M+H] +=708.55。 步驟4. 5-(3-{2-[1-[2-氨基-6-(嗎啉-4-基)-1,3-苯並二氮唑-1-基]-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1H-吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A130-4B)的製備。在氮氣氣氛下在0℃下向雷尼鎳(6.05 mg,0.07 mmol,1當量)和5-[1-(叔丁氧基羰基)-3-{2-[1-{[5-(嗎啉-4-基)-2-硝基苯基]氨基}-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}吡唑-4-基]-1-甲基-6-氧代吡啶-3-甲酸甲酯(50 mg,0.07 mmol,1當量)於甲醇(1 mL)中的攪拌混合物中添加水合肼(10.61 mg,0.21 mmol,3當量)。將所得混合物在室溫下攪拌2 h。將所得混合物過濾,並且將濾餅用甲醇(3×5 mL)洗滌。將濾液在減壓下濃縮。向此殘餘物中添加乙醇(1 mL)和BrCN(8.23 mg,0.07 mmol,1.1當量)。將所得混合物在室溫下攪拌另外2 h,並且用LiOH.H 2O(5.93 mg,0.14 mmol,2當量)於H 2O(0.2 mL)中溶液處理。將所得混合物在40℃下攪拌2 h,並且在減壓下濃縮。將殘餘物通過反向快速色譜法使用10%至50%乙腈/水洗脫進行純化,以得到呈白色固體的15 mg 5-(3-{2-[(1S,5S)-1-[2-氨基-6-(嗎啉-4-基)-1,3-苯並二氮唑-1-基]-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1H-吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(36%)。LCMS: m/z (ESI), [M+H] +=589.40。 步驟5. 15-甲基-5-(嗎啉-4-基)-23-氧雜-2,9,11,15,20, 21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33),18,21-八烯-12,16-二酮(實施例A130B)的製備。將5-(3-{2-[1-[2-氨基-6-(嗎啉-4-基)-1,3-苯並二氮唑-1-基]-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1H-吡唑-4-基)-1-甲基-6-氧代吡啶-3-羧酸(INT-A130-4B,20 mg,0.03 mmol,1當量)、 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(19.38 mg,0.05 mmol,1.5當量)和 N,N-二異丙基乙胺(13.17 mg,0.10 mmol,3當量)於1,4-二噁烷(2 mL)中的混合物在氮氣氣氛下在40℃下攪拌2 h。將反應用水(30 mL)淬滅,並且將混合物用乙酸乙酯(3×30 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上使用二氯甲烷/甲醇(25/1)洗脫進行純化,以得到呈白色固體的4.9 mg 15-甲基-5-(嗎啉-4-基)-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}. 1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13, 17(33),18,21-八烯-12,16-二酮(25%)。LCMS: m/z (ESI), [M+H] +=571.20。 1H NMR (DMSO-d 6, 400 MHz) δ 1.51-1.61 (2H, m), 1.68 (1H, d), 2.45-2.67 (6H, m), 3.09 (4H, q), 3.37-3.55 (3H, m), 3.62 (3H, s), 3.76 (4H, t), 4.14 (2H, d), 4.32 (1H, d), 6.87-6.93 (1H, m), 7.18 (1H, d), 7.42 (1H, d), 8.20 (1H, d), 8.44 (1H, d), 9.19 (1H, d), 11.95 (1H, s), 12.53 (1H, s)。 實施例A132B 5-羥基-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18, 22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮,異構體2(實施例A132B) 將5-溴-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮(M4B,150 mg,0.26 mmol,1當量)、Pd 2(dba) 3(47.49 mg,0.05 mmol,0.2當量)、t-BuXPhos(22.02 mg,0.052 mmol,0.2當量)、KOH(58.19 mg,1.036 mmol,4當量)於1,4-二噁烷(3 mL)和H 2O(2 mL)中的混合物在氮氣氣氛下在100℃下攪拌2 h。將反應用水(50 mL)淬滅,並且將混合物用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過C18 FLASH使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的4.8 mg 5-羥基-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮(異構體2)(3%)。LCMS: m/z (ESI), [M+H] +=516.25。 1H NMR (DMSO-d 6, 400 MHz) δ 1.52-1.63 (1H, m), 1.63-1.79 (2H, m), 1.99 (1H, d), 2.42-2.72 (8H, m), 3.62 (3H, s), 3.63-3.69 (1H, m), 3.71 (3H, s), 4.54 (1H, t), 4.67 (1H, d), 6.63 (1H, dd), 7.16 (1H, d), 7.35 (1H, d), 8.26 (1H, d), 8.41 (1H, s), 8.81 (1H, d), 9.32 (1H, s), 12.55 (1H, s)。 實施例A133B 5-[(2-羥乙基)氨基]-15,21-二甲基-23-氧雜-2,9,11,15,20, 21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33), 18(22),19-八烯-12,16-二酮,異構體2 步驟1. 5-({2-[(叔丁基二甲基甲矽烷基)氧基]乙基}氨基)-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮(INT-A133-1B).將5-溴-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33), 18(22),19-八烯-12,16-二酮(120 mg,0.20 mmol,1當量)、(2-氨基乙氧基)(叔丁基)二甲基矽烷(M4B,109.12 mg,0.62 mmol,3當量)、BrettPhos Pd G3(56.41 mg,0.06 mmol,0.3當量)和LiHMDS(347.12 mg,2.07 mmol,10當量)於二噁烷(5 mL)中的混合物在氮氣氣氛下在60℃下攪拌2 h。將反應用NH 4Cl(水溶液)(50 mL)淬滅,並且將混合物用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠柱色譜法使用二氯甲烷/甲醇(20:1)洗脫進行純化,以得到呈黃色固體的65 mg 5-({2-[(叔丁基二甲基甲矽烷基)氧基]乙基}氨基)-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33), 18(22),19-八烯-12,16-二酮(46%)。LCMS: m/z (ESI), [M+H] +=673.30。 步驟2. 5-[(2-羥乙基)氨基]-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,17(33), 18(22),19-八烯-12,16-二酮(實施例A133B).將5-({2-[(叔丁基二甲基甲矽烷基)氧基]乙基}氨基)-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}. 1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13, 17(33),18(22),19-八烯-12,16-二酮(INT-A133-1B,60 mg,0.09 mmol,1當量)於HCl於1,4-二噁烷(2 mL)中的溶液中的溶液在氮氣氣氛下在15℃下攪拌2 h。將反應用NaHCO 3溶液(50 mL)淬滅,並且將混合物用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過C18 FLASH使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的32.3 mg 5-[(2-羥乙基)氨基]-15,21-二甲基-23-氧雜-2,9,11,15,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18, 22}]三十三-3,5,7,9,13,17(33),18(22),19-八烯-12,16-二酮(60%)。LCMS: m/z (ESI), [M+H] +=559.20。 1H NMR (DMSO-d 6, 400 MHz) δ 1.48-1.82 (3H, m), 1.98 (1H, d), 2.42 (3H, d), 2.56 (1H, s), 2.73 (1H, d), 3.10 (2H, s), 3.18-3.25 (1H, m), 3.31 (1H, s), 3.55-3.63 (6H, m), 3.66 (1H, d), 3.72 (3H, s), 4.55 (1H, t), 4.64-4.74 (2H, m), 5.47 (1H, s), 6.51 (1H, dd), 6.96 (1H, s), 7.28 (1H, d), 8.25 (1H, d), 8.41 (1H, s), 8.82 (1H, d), 12.47 (1H, s)。 實施例B1 15,21-二甲基-23-氧雜-2,9,11,16,20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十四-3,5,7,9,13,15,17(34),18(22), 19-壬-12-酮 步驟1. 2-甲基-6-[1-甲基-5-(2-{1-[(2-硝基苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙氧基)吡唑-4-基]吡啶-4-甲酸甲酯(INT-B1-1)的製備。在氮氣氣氛下向2-{1-[(2-硝基苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙醇(250 mg,0.81 mmol,1當量)和2-(5-羥基-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(M1,202 mg,0.81 mmol,1當量)於甲苯(100 mL)中的攪拌混合物中添加2-(三丁基-λ5-膦亞基)乙腈(1580 mg,6.55 mmol,8當量)。將混合物在100℃下攪拌4 h,冷卻到室溫,並且在減壓下濃縮。將反應用水(80 mL)淬滅,並且將混合物用乙酸乙酯(3×150 mL)萃取。將合併的有機層用鹽水(3×150 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(10/1)洗脫進行純化,以得到呈黃色油的420 mg 2-甲基-6-[1-甲基-5-(2-{1-[(2-硝基苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙氧基)吡唑-4-基]吡啶-4-甲酸甲酯(INT-B1-1) (95%)。LCMS: m/z (ESI), [M+H] +=535.30。 步驟2. 2-[5-(2-{1-[(2-氨基苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙氧基)-1-甲基吡唑-4-基]-6-甲基吡啶-4-甲酸甲酯(INT-B1-2)的製備。在氮氣氣氛下在室溫下向2-甲基-6-[1-甲基-5-(2-{1-[(2-硝基苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙氧基)吡唑-4-基]吡啶-4-甲酸甲酯(400 mg,0.75 mmol,1當量)和雷尼鎳(512 mg,5.98 mmol,8當量)於MeOH(30 mL)中的攪拌混合物中添加NH 2NH 2.H 2O(74 mg,1.50 mmol,2當量)。將所得混合物在室溫下攪拌2 h,並且過濾。將濾餅用甲醇(3×20 mL)洗滌。將所得溶液在減壓下濃縮,以得到呈黃色固體的360 mg 2-[5-(2-{1-[(2-氨基苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙氧基)-1-甲基吡唑-4-基]-6-甲基吡啶-4-甲酸甲酯(INT-B1-2)。此材料在不經進一步純化的情況下用於下一反應中。LCMS: m/z (ESI) , [M+H] +=505.30。 步驟3. 2-(5-{2-[1-(2-氨基-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.2]壬-3-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(INT-B26-3)的製備。在氮氣氣氛下在室溫下向2-[5-(2-{1-[(2-氨基苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙氧基)-1-甲基吡唑-4-基]-6-甲基吡啶-4-甲酸甲酯(360 mg,0.71 mmol,1當量)和BrCN(113 mg,1.06 mmol,1.5當量)於乙醇(30 mL)中的攪拌混合物中。將所得混合物在室溫下攪拌1 h並在真空下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(15/1)洗脫進行純化,以得到呈粉色固體的256 mg 2-(5-{2-[1-(2-氨基-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.2]壬-3-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(INT-B26-3)(67%)。LCMS: m/z (ESI) , [M+H]+=530.35。 步驟4. 2-(5-{2-[1-(2-氨基-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.2]壬-3-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-羧酸(INT-B1-4)的製備。將2-(5-{2-[1-(2-氨基-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.2]壬-3-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(250 mg,0.47 mmol,1當量)和LiOH.H 2O(39 mg,0.94 mmol,2當量)於四氫呋喃(20 mL)和H 2O(4 mL)中的混合物在室溫下攪拌過夜,並且在真空中濃縮。將殘餘物通過C18 FLASH色譜法,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3 .H 2O和乙腈的水作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃色固體的170 mg 2-(5-{2-[1-(2-氨基-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.2]壬-3-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-羧酸(INT-B1-4)(69%)。LCMS: m/z (ESI), [M+H] +=516.15。 1H NMR (CD 3OD, 400 MHz) δ 1.78-1.98 (7H, m), 2.43 (3H, s), 2.64 (2H, d), 2.82 (2H, t), 2.90 (2H, t), 3.16 (2H, s), 3.68 (3H, s), 4.26 (2H, t), 3.74-6.79 (1H, m), 6.89 (1H, t), 7.07-7.10 (1H, m), 7.35 (1H, d), 7.38 (1H, d), 7.73 (1H, s), 7.76 (1H, s)。 步驟5. 15,21-二甲基-23-氧雜-2,9,11,16,20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]三十四-3,5,7,9,13,15,17(34),18(22), 19-壬-12-酮(實施例B1)的製備。在氮氣氣氛下在室溫下向2-(5-{2-[1-(2-氨基-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.2]壬-3-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-羧酸(160 mg,0.31 mmol,1當量)和 N, N, N, N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(353 mg,0.93 mmol,3當量)於1,4-二噁烷(10 mL)中的攪拌混合物中添加 N, N-二異丙基乙胺(160 mg,1.24 mmol,4當量)。將混合物在空氣氣氛下在60℃下攪拌3 h。將混合物冷卻到室溫。在室溫下,將反應用水淬滅。用二氯甲烷(3×200 mL)萃取所得混合物。將合併的有機層用鹽水(3×150 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-HPLC用XBridge Shield RP18 OBD柱,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3.H 2O的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的63.1 mg 15,21-二甲基-23-氧雜-2,9,11,16,20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十四-3,5,7,9,13,15,17(34),18(22), 19-壬-12-酮(實施例B1) (40%)。LCMS: m/z (ESI), [M+H] +=498.20。 1H NMR (DMSO-d 6, 400 MHz) δ 1.82-2.04 (4H, m), 2.04-2.15 (3H, m), 2.55 (3H, s), 2.82 (2H, s), 2.87 (2H, t), 3.42-3.55 (4H, m), 3.77 (3H, s), 4.38 (2H, t), 7.18 (2H, dt), 7.62 (1H, d), 7.65 (1H, d), 7.89 (1H, s), 7.91 (1H, d), 8.77 (1H, s), 13.32 (1H, s)。 實施例B2 15,21-二甲基-5-(嗎啉-4-基)-23-氧雜-2,9,11,16,20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]三十四-3,5,7,9,13,15,17(34),18(22), 19-壬-12-酮 步驟1. 2-[5-(2-{1-[(5-溴-2-硝基苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙氧基)-1-甲基吡唑-4-基]-6-甲基吡啶-4-甲酸甲酯(INT-B2-1)的製備。將2-{1-[(5-溴-2-硝基苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙醇(380 mg,0.98 mmol,1當量)、2-(三丁基-λ5-膦亞基)乙腈(1193 mg,4.94 mmol,5當量)和2-(5-羥基-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(M1,366 mg,1.48 mmol,1.5當量)於甲苯(5 mL)中的混合物在氮氣氣氛下在100℃下攪拌2 h。將混合物冷卻到室溫。將反應用水(100 mL)淬滅,並且將混合物用二氯甲烷(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在Prep-TLC上使用石油醚/乙酸乙酯(3/1)洗脫進行純化,以得到呈橙色油的500 mg 2-[5-(2-{1-[(5-溴-2-硝基苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙氧基)-1-甲基吡唑-4-基]-6-甲基吡啶-4-甲酸甲酯(INT-B2-1)(82 %)。LCMS: m/z (ESI), [M+H] +=613.15。 1H NMR (DMSO-d 6 , 400 MHz) δ 1.44-1.47 (2H, m), 1.74-1.81 (6H, m), 1.89-1.96 (1H, m), 2.48 (3H, s), 2.66-2.72 (2H, m), 2.89-2.97 (4H, m), 3.74 (3H, s), 3.84 (3H, s), 4.39 (2H, t), 6.72-6.77 (1H, m), 7.01 (1H, d), 7.33 (1H, d), 7.77 (1H, d), 7.86 (1H, s), 7.95 (1H, d), 8.06 (1H, s)。 步驟2. 2-[5-(2-{1-[(2-氨基-5-溴苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙氧基)-1-甲基吡唑-4-基]-6-甲基吡啶-4-甲酸甲酯(INT-B2-2)的製備。在氮氣氣氛下在0℃下向2-[5-(2-{1-[(5-溴-2-硝基苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙氧基)-1-甲基吡唑-4-基]-6-甲基吡啶-4-甲酸甲酯(480 mg,0.78 mmol,1當量)和雷尼鎳(335 mg,3.91 mmol,5當量)於甲醇(5 mL)中的攪拌混合物中添加水合肼(98%)(61 mg,1.22 mmol,1.5當量)。將混合物在氮氣氣氛下在0℃下攪拌2 h。將反應用水(100 mL)淬滅,並且將混合物用二氯甲烷(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用石油醚/乙酸乙酯(1/1)洗脫進行純化,以得到呈棕色固體的370 mg 2-[5-(2-{1-[(2-氨基-5-溴苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙氧基)-1-甲基吡唑-4-基]-6-甲基吡啶-4-甲酸甲酯(INT-B2-2)(81 %)。LCMS: m/z (ESI), [M+H] +=583.20。 1H NMR (DMSO-d 6 , 400 MHz) δ 1.38-1.49 (2H, m), 1.69-1.76 (4H, m), 1.85-1.88 (1H, m), 2.25-2.32 (2H, m), 2.52 (3H, s), 2.63-2.68 (2H, m), 2.78-2.83 (2H, m), 2.84-2.91 (2H, m), 3.72 (3H, s), 3.86 (3H, s), 4.03 (1H, s), 4.27-4.37(2H, m), 4.68 (2H, s), 6.46 (2H, d), 6.58 (1H, d), 7.43 (1H, d), 7.83 (1H, d), 7.89 (1H, s)。 步驟3. 2-(5-{2-[1-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.2]壬-3-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(INT-B2-3)的製備。將2-[5-(2-{1-[(2-氨基-5-溴苯基)氨基]-3-氮雜雙環[3.2.2]壬-3-基}乙氧基)-1-甲基吡唑-4-基]-6-甲基吡啶-4-甲酸甲酯(350 mg,0.60 mmol,1當量)和BrCN(95 mg,0.90 mmol,1.5當量)於乙醇(2 mL)中的混合物在氮氣氣氛下在室溫下攪拌過夜。將所得混合物在減壓下濃縮。將殘餘物通過Prep-TLC使用石油醚/乙酸乙酯(1/3)洗脫進行純化,以得到呈棕色固體的360 mg 2-(5-{2-[1-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.2]壬-3-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(INT-B2-3)(98%)。LCMS: m/z (ESI), [M+H] +=610.15。 1H NMR (DMSO-d 6 , 400 MHz) δ 1.79-1.97 (7H, m), 2.51 (3H, s), 2.70-2.74 (2H, m), 2.79-2.87 (2H, m), 2.92-2.99 (2H, m), 3.18 (2H, s), 3.74 (3H, s), 3.84 (3H, s), 4.37 (2H, t), 6.05 (2H, s), 6.99-7.08 (2H, m), 7.41 (1H, d), 7.46 (1H, d), 7.81 (1H, d), 7.89 (1H, s)。 步驟4. 鋰代2-(5-{2-[1-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.2]壬-3-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸酯(INT-B2-4)的製備。將2-(5-{2-[1-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.2]壬-3-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(340 mg,0.55 mmol,1當量)和LiOH(16 mg,0.67 mmol,1.2當量)於乙醇(2 mL)中的混合物在氮氣氣氛下在80℃下攪拌2 h。將混合物冷卻到室溫,並且在減壓下濃縮,以得到呈白色固體的2-(5-{2-[1-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.2]壬-3-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-羧酸(INT-B2-4)。將粗產物在不進行進一步純化的情況下直接用於下一反應。LCMS: m/z (ESI), [M+H] +=594.45。 步驟5. 5-溴-15,21-二甲基-23-氧雜-2,9,11,16,20,21, 26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3, 8}.0^{18,22}]三十四-3,5,7,9,13,15,17(34), 18(22),19-壬-12-酮(INT-B2-5)的製備。將2-(5-{2-[1-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.2]壬-3-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-羧酸(330 mg粗製物,約0.5 mmol,約1當量)、 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(417 mg,1.10 mmol,2當量)和 N,N-二異丙基乙胺(213 mg,1.65 mmol,3當量)於1,4-二噁烷(5 mL)中的混合物在氮氣氣氛下在60℃下攪拌2 h。將混合物冷卻到室溫。將反應用水(50 mL)淬滅,並且將混合物用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×150 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過與石油醚(10 mL)一起研磨進行純化,以得到呈黃色固體的280 mg 5-溴-15,21-二甲基-23-氧雜-2,9,11,16,20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]三十四-3,5,7,9,13,15,17(34), 18(22),19-壬-12-酮(INT-B2-5)(88%)。LCMS: m/z (ESI), [M+H] +=578.05。 步驟6. 15,21-二甲基-5-(嗎啉-4-基)-23-氧雜-2,9,11, 16,20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}.0^{2, 10}.0^{3,8}.0^{18,22}]三十四-3,5,7,9,13,15,17(34), 18(22),19-壬-12-酮(實施例B2)的製備。在氮氣氣氛下在室溫下向5-溴-15,21-二甲基-23-氧雜-2,9,11,16,20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]三十四-3,5,7,9,13,15,17(34),18(22),19-壬-12-酮(100 mg,0.17 mmol,1當量)、Brettphos Pd G 3(47 mg,0.05 mmol,0.3當量)和嗎啉(52.89 mg,0.605 mmol,3.5當量)與1,4-二噁烷(10 mL)中的攪拌混合物中逐滴添加LiHMDS(2 mL,2.00 mmol,11.53當量)。將所得混合物在氮氣氣氛下在60℃下攪拌1 h,並且冷卻到室溫。將反應用飽和NH 4Cl(水溶液)(50 mL)淬滅,並且將混合物用CH 2Cl 2(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-HPLC用XBridge Prep OBD C18柱,使用含有10 mmol/L NH 4HCO 3的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃色固體的14.4 mg 15,21-二甲基-5-(嗎啉-4-基)-23-氧雜-2,9,11,16,20, 21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]三十四-3,5,7,9,13,15,17(34),18(22),19-壬-12-酮(實施例B2)(14%)。LCMS: m/z (ESI), [M+H] += 583.40。 1H NMR (DMSO-d 6 , 400 MHz) δ 1.88-1.99 (4H, m), 2.03-2.11 (3H, m), 2.54 (3H, s), 2.78-2.90 (4H, m), 3.04-3.10 (4H, m), 3.42-3.50 (4H, m), 3.75-3.80 (7H, m), 4.36-4.40 (2H, m), 6.95 (1H, dd), 7.35 (1H, s), 7.51 (1H, d), 7.61 (1H, d), 7.89 (1H, s), 8.76 (1H, s), 13.21 (1H, s)。 實施例B3 15,21-二甲基-5-(4-甲基哌嗪-1-基)-23-氧雜-2,9,11,16, 20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]三十四-3,5,7,9,13,15,17(34),18(22), 19-壬-12-酮 步驟1. 15,21-二甲基-5-(4-甲基哌嗪-1-基)-23-氧雜-2, 9,11,16,20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十四-3,5,7,9,13,15,17(34), 18(22),19-壬-12-酮(實施例B3)的製備。在氮氣氣氛下在室溫下向5-溴-15,21-二甲基-23-氧雜-2,9,11,16,20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]三十四-3,5,7,9,13,15,17(34),18(22),19-壬-12-酮(100 mg,0.17 mmol,1當量)、BrettPhos Pd G 3(550 mg,0.60 mmol,3.5當量)和1-甲基哌嗪(60.81 mg,0.605 mmol,3.5當量)與1,4-二噁烷(10 mL)中的攪拌混合物中逐滴添加LiHMDS(2 mL,2.000 mmol,11.53當量)。將所得混合物在氮氣氣氛下在60℃下攪拌1 h,並且冷卻到室溫。將反應用飽和NH 4Cl(水溶液)(50 mL)淬滅,並且將混合物用CH 2Cl 2(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-HPLC用XBridge Shield RP18 OBD柱,使用含有10 mmol/L NH 4HCO 3的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃色固體的13.7 mg 15,21-二甲基-5-(4-甲基哌嗪-1-基)-23-氧雜-2,9,11,16,20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十四-3,5,7,9,13,15,17(34), 18(22),19-壬-12-酮(實施例B3)(13%)。LCMS: m/z (ESI), [M+H] +=596.40。 1H NMR (DMSO-d 6 , 400 MHz) δ 1.84-2.01 (4H, m), 2.03-2.10 (3H, m), 2.24 (3H, s), 2.54 (4H, s), 2.79-2.84 (2H, m), 2.85-2.90 (2H, m), 3.10 (4H, t), 3.28-3.36 (3H, m), 3.42-3.50 (4H, m), 3.76 (3H, s), 4.37 (2H, d), 6.94 (1H, dd), 7.34 (1H, s), 7.49 (1H, d), 7.61 (1H, d), 7.89 (1H, s), 8.76 (1H, s), 13.20 (1H, s)。 實施例B4 15,21-二甲基-23-氧雜-2,9,11,16,20,21,26-七氮雜六環[24.4.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十二-3,5,7,9,13(32),14,16,18(22),19-壬-12-酮 步驟1. 3-[(2-硝基苯基)氨基]氮雜環庚烷-1-甲酸叔丁酯(INT-B4-1)的製備。將鄰氟硝基苯(5.00 g,35.44 mmol,1當量)、K 2CO 3(9.79 g,70.87 mmol,2當量)和3-氨基氮雜環庚烷-1-甲酸叔丁酯(7.59 g,35.44 mmol,1當量)於乙腈(80 mL)中的混合物在氮氣氣氛下在80℃下攪拌3 h。將混合物冷卻到室溫。將反應用水(200 mL)淬滅,並且將混合物用乙酸乙酯(3×200 mL)萃取。將合併的有機層用鹽水(3×200 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法使用石油醚/乙酸乙酯(21/1)洗脫進行純化,以得到呈紅棕色油的9 g 3-[(2-硝基苯基)氨基]氮雜環庚烷-1-甲酸叔丁酯(INT-B4-1)(75%)。LCMS: m/z (ESI), [M+H] +=336.15。 1H NMR (400 MHz, DMSO-d 6) δ 1.38 (9H, s), 1.49-1.65 (1H, m), 1.62-1.75 (3H, m), 1.75-1.82 (1H, m), 1.96-2.01 (1H, m), 3.37-3.50 (2H, m), 3.61-3.69 (1H, m), 3.83-4.03 (2H, m), 6.66-6.76 (1H, m), 7.21-7.25 (1H, m), 7.51-7.61 (1H, m), 8.01-8.13 (1H, m), 8.20 (1H, d)。 步驟2. N-(2-硝基苯基)氮雜環庚-3-胺(INT-B4-2)的製備。在室溫下向3-[(2-硝基苯基)氨基]氮雜環庚烷-1-甲酸叔丁酯(4.00 g,11.93 mmol,1當量)於二氯甲烷(12 mL)中的攪拌混合物中緩慢添加含HCl的1,4-二噁烷(60 mL)。將所得混合物在室溫下攪拌1.5 h。將混合物在減壓下濃縮。將殘餘物用飽和NaHCO 3(水溶液)(100 mL)處理,並且用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥,並且在減壓下濃縮,以得到呈黃色油的2.8 g N-(2-硝基苯基)氮雜環庚烷-3-胺(INT-B4-2)。此材料在不經進一步純化的情況下用於下一反應中。LCMS: m/z (ESI), [M+H] +=236.10。 步驟3. 1-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}- N-(2-硝基苯基)氮雜環庚-3-胺(INT-B4-3)的製備。將新鮮製備的 N-(2-硝基苯基)氮雜環庚烷-3-胺(2.8 g,約1當量)、(2-溴乙氧基)(叔丁基)二甲基矽烷(6.39 g,26.69 mmol,2當量)、K 2CO 3(3.69 g,26.69 mmol,2當量)和KI(4.43 g,26.69 mmol,2當量)於DMF(80 mL)中的混合物在氮氣氣氛下在60℃下攪拌2 h。將混合物冷卻到室溫。將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-TLC使用石油醚/乙酸乙酯(19/1)洗脫來進行純化,以得到呈黃色油的5.2 g 1-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}- N-(2-硝基苯基)氮雜環庚烷-3-胺(INT-B4-3)(99%)。LCMS: m/z (ESI), [M+H] +=394.20。 1H NMR (400 MHz, DMSO-d 6) δ 0.03 (6H, s), 0.81 (9H, s), 1.45-1.63 (3H, m), 1.74-1.81 (3H, m), 2.57-2.77 (3H, m), 2.78-2.95 (3H, m), 3.29 (1H, t), 3.63-3.84 (3H, m), 6.65-6.71 (1H, m), 7.05 (1H, d), 7.51-7.58 (1H, m), 8.07-8.13 (1H, m), 8.74 (1H, d)。 步驟4. 2-{3-[(2-硝基苯基)氨基]氮雜環庚-1-基}乙醇(INT-B4-4)的製備。在氮氣氣氛下在室溫下向1-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-N-(2-硝基苯基)氮雜環庚-3-胺(3 g,7.62 mmol,1當量)於四氫呋喃(50 mL)中的攪拌混合物中添加含四丁基氟化銨的四氫呋喃(12 mL)。將所得混合物在室溫下攪拌1 h。將反應用水(100 mL)淬滅,並且將混合物用二氯甲烷(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用石油醚/乙酸乙酯(1/1)洗脫進行純化,以得到呈黃色油的1.5 g 2-{3-[(2-硝基苯基)氨基]氮雜環庚-1-基}乙醇(INT-B4-4)(70%)。LCMS: m/z (ESI), [M+H] +=280.10。 步驟5. 2-甲基-6-[1-甲基-5-(2-{3-[(2-硝基苯基)氨基]氮雜環庚-1-基}乙氧基)吡唑-4-基]吡啶-4-甲酸甲酯(INT-B4-5)的製備。在氮氣氣氛下在0℃下向2-{3-[(2-硝基苯基)氨基]氮雜環庚-1-基}乙醇(1.5 g,5.370 mmol,1當量)和三苯基膦(4.30 g,16.38 mmol,3.05當量)於四氫呋喃(30 mL)中的攪拌混合物中添加2-(5-羥基-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(M1,1.46 g,5.91 mmol,1.1當量)和偶氮二甲酸二異丙酯(2.71 g,13.43 mmol,2.5當量)。將所得混合物在室溫下攪拌2 h。將反應用水(300 mL)淬滅,並且將混合物用二氯甲烷(3×300 mL)萃取。將合併的有機層用鹽水(3×300 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過反向快速色譜法使用10%至50%乙腈/水洗脫進行純化,以得到呈黃色油的1.6 g 2-甲基-6-[1-甲基-5-(2-{3-[(2-硝基苯基)氨基]氮雜環庚-1-基}乙氧基)吡唑-4-基]吡啶-4-甲酸甲酯(INT-B4-5)(59%)。LCMS: m/z (ESI), [M+H] +=509.25。 1H NMR (400 MHz, DMSO-d 6) δ 1.44-1.80 (6H, m), 2.48-2.54 (2H, m), 2.71-2.81 (1H, m), 2.85-2.92 (2H, m), 2.94-3.14 (3H, m), 3.70 (3H, s), 3.86 (3H, s), 3.92 (1H, s), 4.28-4.35 (2H, m), 6.63-6.68 (1H, m), 6.96-7.03 (1H, m), 7.38 (1H, d), 7.46-7.51 (1H, m), 7.80-7.83 (1H, m), 7.88 (1H, s), 8.00-8.05 (1H, m), 8.62 (1H, d)。 步驟6. 2-[5-(2-{3-[(2-氨基苯基)氨基]氮雜環庚-1-基}乙氧基)-1-甲基吡唑-4-基]-6-甲基吡啶-4-甲酸甲酯(INT-B4-6)的製備。在氮氣氣氛下在0℃下向2-甲基-6-[1-甲基-5-(2-{3-[(2-硝基苯基)氨基]氮雜環庚-1-基}乙氧基)吡唑-4-基]吡啶-4-甲酸甲酯(1.00 g,1.97 mmol,1當量)和雷尼鎳(340 mg,3.93 mmol,2當量)於MeOH(30 mL)中的攪拌混合物中添加NH 2NH 2.H 2O(295 mg,5.90 mmol,3當量)。將所得混合物在室溫下攪拌1 h。將混合物過濾,並且將濾餅用二氯甲烷(3×30 mL)洗滌。將所得溶液在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(30/1)洗脫進行純化,以得到呈紅棕色油的500 mg 2-[5-(2-{3-[(2-氨基苯基)氨基]氮雜環庚-1-基}乙氧基)-1-甲基吡唑-4-基]-6-甲基吡啶-4-甲酸甲酯(INT-B4-6)(53%)。LCMS: m/z (ESI), [M+H] +=479.25。 步驟7. 2-(5-{2-[3-(2-氨基-1,3-苯並二氮唑-1-基)氮雜環庚-1-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(INT-B4-7)的製備。將2-[5-(2-{3-[(2-氨基苯基)氨基]氮雜環庚-1-基}乙氧基)-1-甲基吡唑-4-基]-6-甲基吡啶-4-甲酸甲酯(490 mg,1.02 mmol,1當量)和溴化氰(108 mg,1.02 mmol,1當量)於二氯甲烷(5 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將混合物在減壓下濃縮,並且將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(30/1)洗脫進行純化,以得到呈黃色油的490 mg 2-(5-{2-[3-(2-氨基-1,3-苯並二氮唑-1-基)氮雜環庚-1-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(INT-B4-7)(95%)。LCMS: m/z (ESI), [M+H] +=504.25。 1H NMR (400 MHz, DMSO-d 6) δ 1.50-1.56 (1H, m), 1.61-1.70 (1H, m), 1.73-1.99 (2H, m), 2.52 (5H, s), 2.93-3.03 (1H, m), 3.17 (1H, s), 3.26-3.32 (3H, m), 3.70 (3H, s), 3.84 (3H, s), 4.28 (2H, s), 4.62 (1H, s), 6.96 (1H, s), 7.00 (1H, d), 7.20 (1H, d), 7.39 (1H, s), 7.44 (1H, d), 7.83-7.88 (1H, m), 7.91 (1H, s)。 步驟8. 2-(5-{2-[3-(2-氨基-1,3-苯並二氮唑-1-基)氮雜環庚-1-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-羧酸(INT-B4-8)的製備。將2-(5-{2-[3-(2-氨基-1,3-苯並二氮唑-1-基)氮雜環庚-1-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(480 mg,0.95 mmol,1當量)和LiOH(45 mg,1.91 mmol,2當量)於四氫呋喃(6 mL)和H 2O(1.5 mL)中的混合物在室溫下攪拌1 h。將混合物在減壓下濃縮,並且將殘餘物通過反向快速色譜法使用10%至30%乙腈/水洗脫進行純化,以得到呈白色固體的125 mg 2-(5-{2-[3-(2-氨基-1,3-苯並二氮唑-1-基)氮雜環庚-1-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-羧酸(INT-B4-8)(27%)。LCMS: m/z (ESI), [M+H] +=490.15。 步驟9. 15,21-二甲基-23-氧雜-2,9,11,16,20,21,26-七氮雜六環[24.4.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十二-3,5,7,9,13(32),14,16,18(22),19-壬-12-酮(實施例B4)的製備。將2-(5-{2-[3-(2-氨基-1,3-苯並二氮唑-1-基)氮雜環庚-1-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-羧酸(120 mg,0.25 mmol,1當量)、 N,N-二異丙基乙胺(95 mg,0.74 mmol,3當量)和 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(140 mg,0.37 mmol,1.5當量)於1,4-二噁烷(6 mL)中的混合物在氮氣氣氛下在室溫下攪拌1 h。將混合物在減壓下濃縮。將殘餘物通過Prep-HPLC使用含有10 mmol/L NH 4HCO 3的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發,以得到呈白色固體的60.9 mg 15,21-二甲基-23-氧雜-2,9,11,16,20,21,26-七氮雜六環[24.4.1.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]三十二-3,5,7,9,13(32),14,16,18(22),19-壬-12-酮(實施例B4) (52%)。LCMS: m/z (ESI), [M+H] +=472.25。 1H NMR (DMSO-d 6, 400 MHz) δ 1.63-2.02 (6H, m), 2.57 (3H, s), 2.62-2.76 (1H, m), 2.79-2.85 (1H, m), 2.88-3.01 (3H, m), 3.10-3.17 (1H, m), 3.76 (3H, s), 4.04 (1H, br s), 4.38 (1H, br s), 5.15 (1H, br s), 7.19-7.30 (2H, m), 7.52-7.59 (2H, m), 7.68 (1H, d), 7.91 (1H, s), 8.62 (1H, s), 12.72 (1H, s)。 實施例B6 11-環丙基-26-甲基-8-(2,2,2-三氟乙基)-11H,51H-11-氧雜-4,8-二氮雜-5(2,1)-苯並[d]咪唑-2(2,4)-吡啶-1(4,5)-吡唑環十一烷-3-酮 步驟1. 2-氯-6-(2-環丙基-3-氧代-1-((2-(三甲基甲矽烷基)乙氧基)甲基)-2,3-二氫-1H-吡唑-4-基)異煙酸甲酯(INT-B6-1)的製備。在氮氣氣氛下在室溫下向2-環丙基-4-碘-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-3-酮(2.3 g,6.04 mmol,1當量)和新鮮製備的(6-氯-4-(甲氧基羰基)吡啶-2-基)硼酸(M1-3,9.0 g粗製材料,約30 mmol,約5當量)於甲苯(20 mL)和水(5 mL)中的攪拌混合物中添加Pd 2(dba) 3(1.1 g,1.21 mmol,0.2當量)、二(1-金剛烷基)-正丁基膦(0.1 g,1.21 mmol,0.2當量)和Cs 2CO 3(4.9 g,15.12 mmol,2.5當量)。將所得混合物在80℃下攪拌2 h,並且冷卻到室溫。將反應用水(300 mL)淬滅,並且將混合物用乙酸乙酯(3×200 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠柱色譜法使用石油醚/乙酸乙酯(1/1)洗脫進行純化,以得到呈黃色油的1.8 g 2-氯-6-(2-環丙基-3-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-4-基)吡啶-4-甲酸甲酯(INT-B6-1)(70%)。 1H NMR (DMSO-d 6, 400 MHz) δ -0.05 (9H, s), 0.78-0.83 (2H, m), 0.87-0.90 (4H, m), 3.30 (2H, d), 3.94-3.98 (3H, m), 2.05-2.10 (1H, m), 5.41 (2H, s), 7.55-7.51 (1H, m), 8.66-8.70 (2H, m)。 步驟2. 2-(2-環丙基-3-氧代-1-((2-(三甲基甲矽烷基)乙氧基)甲基)-2,3-二氫-1H-吡唑-4-基)-6-甲基異煙酸甲酯(INT-B6-2)的製備。在氮氣氣氛下在室溫下向2-氯-6-(2-環丙基-3-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-4-基)吡啶-4-甲酸甲酯(1.8 g,4.24 mmol,1當量)和三甲基-1,3,5,2,4,6-三氧雜三硼環己烷(3.2 g,25.47 mmol,6.0當量)於1,4-二噁烷(25 mL)中的攪拌混合物中添加PCy 3.HBF 4(0.5 g,1.27 mmol,0.3當量)、Pd(dppf)Cl 2CH 2Cl 2(0.6 g,0.84 mmol,0.2當量)和K 2CO 3(1.1 g,8.49 mmol,2.0當量)。將所得混合物在100℃下攪拌過夜並且冷卻到室溫。將反應用水(50 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×200 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上使用石油醚/乙酸乙酯(1/1)洗脫進行純化,以得到呈棕色固體的1.3 g 2-(2-環丙基-3-氧代-1-((2-(三甲基甲矽烷基)乙氧基)甲基)-2,3-二氫-1H-吡唑-4-基)-6-甲基異煙酸甲酯(INT-B6-2)(75%)。 1H NMR (DMSO-d 6, 400 MHz) δ -0.05 (9H, s), 0.80-0.84 (2H, m), 1.00-1.05 (4H, m), 2.53 (3H, s), 2.74-2.80 (1H, m), 3.45-3.50 (2H, m), 3.89 (3H, s), 5.38 (2H, s), 7.45 (1H, d), 8.55 (1H, d), 8.59 (1H, s)。 步驟3. 2-(1-環丙基-5-羥基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(INT-B6-3)的製備。將2-(2-環丙基-3-氧代-1-{[2-(三甲基甲矽烷基)乙氧基]甲基}吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(1.3 g,3.22 mmol,1當量)和HCl於1,4-二噁烷(10 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h,並且在真空下濃縮。將殘餘物用CH 2Cl 2(20 mL)和飽和NaHCO 3(水溶液)(50 mL)處理。用二氯甲烷(3×100 mL)萃取所得混合物。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物負載於矽膠柱上並且使用二氯甲烷/甲醇(10/1)洗脫,以得到呈棕色固體的844 mg 2-(1-環丙基-5-羥基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(INT-B6-3)(95%)。 1H NMR (DMSO-d 6, 400 MHz) δ 0.88-1.00 (4H, m), 1.24 (1H, s), 2.57 (3H, s), 3.91 (3H, s), 7.33 (1H, s), 7.91 (2H, s)。 步驟4. 2-(1-環丙基-5-(2-((2-((2-硝基苯基)氨基)乙基)(2,2,2-三氟乙基)氨基)乙氧基)-1H-吡唑-4-基)-6-甲基異煙酸甲酯(INT-B6-4)的製備。在氮氣氣氛下在0℃下向2-(1-環丙基-5-羥基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(142 mg,0.52 mmol,1.0當量)和三苯基膦(409 mg,1.56 mmol,3.0當量)於四氫呋喃(10 mL)中的攪拌混合物中添加偶氮二甲酸二異丙酯(315 mg,1.56 mmol,3.0當量)和2-({2-[(2-硝基苯基)氨基]乙基}(2,2,2-三氟乙基)氨基)乙醇(INT-A1-2,400 mg,1.30 mmol,2.5當量)。將所得混合物攪拌2 h。將反應用水(50 mL)淬滅,並且將混合物用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠柱使用CH 2Cl 2/甲醇(10/1)洗脫進行純化,以得到呈黃色油的540 mg 2-(1-環丙基-5-(2-((2-((2-硝基苯基)氨基)乙基)(2,2,2-三氟乙基)氨基)乙氧基)-1H-吡唑-4-基)-6-甲基異煙酸甲酯(INT-B6-4)(57%)。 1H NMR (DMSO-d 6, 400 MHz) δ 0.91-0.99 (2H, m), 1.02-1.11 (2H, m), 2.53 (3H, s), 3.01-3.07 (2H, m), 3.39-3.43 (4H, m), 3.51-3.67 (3H, m), 3.88 (3H, s), 4.32 (2H, t), 6.61-6.66 (1H, m), 7.02-7.08 (1H. m), 7.38-7.54 (2H, m), 7.83-7.78 (2H, m), 8.03-8.07 (1H, m), 8.22 (1H, t)。 步驟5. 2-{5-[2-({2-[(2-氨基苯基)氨基]乙基}(2,2,2-三氟乙基)氨基)乙氧基]-1-環丙基吡唑-4-基}-6-甲基吡啶-4-甲酸甲酯(INT-B6-5)的製備。在氮氣氣氛下在室溫下向2-{1-環丙基-5-[2-({2-[(2-硝基苯基)氨基]乙基}(2,2,2-三氟乙基)氨基)乙氧基]吡唑-4-基}-6-甲基吡啶-4-甲酸甲酯(500 mg,0.88 mmol,1當量)和Zn(464 mg,7.11 mmol,8當量)於四氫呋喃(10 mL)和H 2O(2.5 mL)中的攪拌混合物中添加NH 4Cl(475 mg,8.89 mmol,10當量)。將混合物在所述溫度下攪拌2小時。將反應用水(50 mL)淬滅,並且將混合物用二氯甲烷(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠柱使用CH 2Cl 2/甲醇(5/1)洗脫進行純化,以得到呈黃色油的190 mg 2-{5-[2-({2-[(2-氨基苯基)氨基]乙基}(2,2,2-三氟乙基)氨基)乙氧基]-1-環丙基吡唑-4-基}-6-甲基吡啶-4-甲酸甲酯(INT-B6-5)(40%)。 1H NMR (DMSO-d 6, 400 MHz) δ 0.92-1.01 (2H, m), 1.03-1.07 (2H, m), 2.55 (3H, s), 3.00 (2H, t), 3.14 (2H, s), 3.48-3.52 (2H, m), 3.58-3.63 (1H, m), 3.89 (3H, s), 3.98-4.04 (1H, m), 4.29-4.31 (2H, m), 4.40 (2H, s), 6.37-6.49 (3H, m), 6.48-6.54 (1H, m), 7.49 (1H, d), 7.81-7.91 (2H, m)。 步驟6. 2-[5-(2-{[2-(2-氨基-1,3-苯並二氮唑-1-基)乙基](2,2,2-三氟乙基)氨基}乙氧基)-1-環丙基吡唑-4-基]-6-甲基吡啶-4-甲酸甲酯(INT-B6-6)的製備。將2-{5-[2-({2-[(2-氨基苯基)氨基]乙基}(2,2,2-三氟乙基)氨基)乙氧基]-1-環丙基吡唑-4-基}-6-甲基吡啶-4-甲酸甲酯(170 mg,0.31 mmol,1當量)和BrCN(37 mg,0.35 mmol,1.1當量)於乙醇(5 mL)中的混合物在氮氣氣氛下在室溫下攪拌4 h。將混合物在真空中濃縮。將殘餘物通過Prep-TLC使用CH 2Cl 2/甲醇(10/1)洗脫進行純化,以得到呈棕色油的125 mg 2-[5-(2-{[2-(2-氨基-1,3-苯並二氮唑-1-基)乙基](2,2,2-三氟乙基)氨基}乙氧基)-1-環丙基吡唑-4-基]-6-甲基吡啶-4-甲酸甲酯(INT-B6-6)(70%)。 1H NMR (DMSO-d 6, 400 MHz) δ 0.92-0.98 (2H, m), 1.03-1.07 (2H, m), 2.54 (3H, s), 2.97-3.07 (2H, m), 3.14-3.24 (2H, m), 3.50-3.64 (3H, m), 3.86 (3H, s), 4.08 (2H, t), 4.25 (2H, t), 6.47 (2H, s), 6.78-6.80 (1H, m), 6.87-6.91 (1H, m), 7.03-7.17 (2H, m), 7.47 (1H, d), 7.85 (2H, d)。 步驟7. 2-[5-(2-{[2-(2-氨基-1,3-苯並二氮唑-1-基)乙基](2,2,2-三氟乙基)氨基}乙氧基)-1-環丙基吡唑-4-基]-6-甲基吡啶-4-羧酸(INT-B6-7)的製備。將2-[5-(2-{[2-(2-氨基-1,3-苯並二氮唑-1-基)乙基](2,2,2-三氟乙基)氨基}乙氧基)-1-環丙基吡唑-4-基]-6-甲基吡啶-4-甲酸甲酯(120 mg,0.22 mmol,1當量)和LiOH(10 mg,0.43 mmol,2當量)於四氫呋喃(4 mL)和H 2O(1 mL)中的混合物在氮氣氣氛下在室溫下攪拌1 h。將混合物在真空中濃縮。將殘餘物通過反向快速色譜法使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈棕色固體的90 mg 2-[5-(2-{[2-(2-氨基-1,3-苯並二氮唑-1-基)乙基](2,2,2-三氟乙基)氨基}乙氧基)-1-環丙基吡唑-4-基]-6-甲基吡啶-4-羧酸(INT-B6-7)(76%)。LCMS: m/z (ESI), [M+H] += 544.30。 步驟8. 5-環丙基-26-甲基-10-(2,2,2-三氟乙基)-7-氧雜-4,5,10,13,20,22,27-七氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(27),2(6),3,14,16,18,20,24(28),25-壬-23-酮(實施例B6)的製備。在氮氣氣氛下在室溫下向2-[5-(2-{[2-(2-氨基-1,3-苯並二氮唑-1-基)乙基](2,2,2-三氟乙基)氨基}乙氧基)-1-環丙基吡唑-4-基]-6-甲基吡啶-4-羧酸(80 mg,0.14 mmol,1當量)和 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(83 mg,0.22 mmol,1.5當量)和1,4-二噁烷(3 mL)中的攪拌混合物中添加 N,N-二異丙基乙胺(57 mg,0.44 mmol,3.0當量)。將反應混合物在所述溫度下攪拌2 h,並在真空下濃縮。將殘餘物通過Prep-HPLC使用含有10 mmol/L NH 4HCO 3和0.1% NH 4OH的水和乙腈作為移動相進行純化,以得到呈白色固體的40 mg 5-環丙基-26-甲基-10-(2,2,2-三氟乙基)-7-氧雜-4,5,10,13,20, 22,27-七氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(27),2(6),3,14,16,18,20,24(28),25-壬-23-酮(實施例B6)(52%)。LCMS: m/z (ESI), [M+H] +=526.20。 1H NMR (DMSO-d 6, 400 MHz) δ 0.98-1.08 (2H, m), 1.04-1.15 (2H, m), 2.48-2.54 (1H, m), 2.57 (3H, s), 3.14-3.23 (2H, m), 3.33-3.38 (1H, m), 3.56-3.73 (3H, m), 4.31 (2H, t), 4.34-4.42 (2H, m), 7.20-7.35 (2H, m), 7.51-7.59 (2H, m), 7.72-7.79 (1H, m), 7.85 (1H, s), 8.55 (1H, d), 12.73 (1H, s)。 實施例B7A和實施例B7B 15,21-二甲基-23,29-二氧雜-2,9,11,16,20,21,26-七氮雜六環[24.4.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十二-3,5,7,9,13(32),14,16,18(22),19-壬-12-酮,異構體1(實施例B7A)和異構體2(實施例B7B) 步驟1. 6-[(2-硝基苯基)氨基]-1,4-氧氮雜環庚烷-4-甲酸叔丁酯(INT-B7-1)的製備。將鄰氟硝基苯(1 g,7.09 mmol,1當量)、K 2CO 3(1.96 g,14.17 mmol,2當量)和6-氨基-1,4-氧氮雜環庚烷-4-甲酸叔丁酯(1.69 g,7.79 mmol,1.1當量)於乙腈(20 mL)中的混合物在氮氣氣氛下在100℃下攪拌12 h。將混合物冷卻到室溫。將反應用水(200 mL)淬滅,並且將混合物用乙酸乙酯(3×200 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上使用石油醚/乙酸乙酯(3/1)洗脫進行純化,以得到呈黃色油的1.9 g 6-[(2-硝基苯基)氨基]-1,4-氧氮雜環庚烷-4-甲酸叔丁酯(INT-B7-1)(79%)。LCMS: m/z (ESI), [M+H] +=338.10。 1H NMR (DMSO-d 6, 400 MHz) δ 1.03 (6H, s), 1.33 (3H, s), 3.29 (2H, d), 3.58-3.79 (2H, m), 3.81-3.96 (2H, m), 4.02-4.13 (2H, m), 6.72 (1H, q), 7.17 (1H, d), 7.55 (1H, q), 8.05-8.13 (1H, m), 8.36-8.46 (1H, m)。 步驟2. N-(2-硝基苯基)-1,4-氧氮雜環庚-6-胺(INT-B7-2)的製備。將6-[(2-硝基苯基)氨基]-1,4-氧氮雜環庚烷-4-甲酸叔丁酯(2 g,5.928 mmol,1當量)於三氟乙酸(10 mL)和二氯甲烷(40 mL)中的混合物在氮氣氣氛下在室溫下攪拌過夜。將混合物在減壓下濃縮。將殘餘物用飽和NaHCO 3(水溶液)(100 mL)處理,並且用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×200 mL)洗滌,經無水Na 2SO 4乾燥,並且在減壓下濃縮,以得到呈黃色油的1.4 g N-(2-硝基苯基)-1,4-氧氮雜環庚-6-胺(INT-B7-2)(99%)。此材料在不經進一步純化的情況下用於下一反應中。LCMS: m/z (ESI), [M+H] +=238.10。 步驟3. 4-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}- N-(2-硝基苯基)-1,4-氧氮雜環庚-6-胺(INT-B7-3)的製備。將 N-(2-硝基苯基)-1,4-氧氮雜環庚-6-胺(1.4 g,5.90 mmol,1當量)、(2-溴乙氧基)(叔丁基)二甲基矽烷(2.12 g,8.85 mmol,1.5當量)和K 2CO 3(2.45 g,17.70 mmol,3當量)於乙腈(20 mL)中的混合物在氮氣氣氛下在60℃下攪拌過夜,並且冷卻到室溫。將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×200 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上使用石油醚/乙酸乙酯(1/1)洗脫來進行純化,以得到呈橙色油的1.8 g 4-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}- N-(2-硝基苯基)-1,4-氧氮雜環庚-6-胺(INT-B7-3)(77%)。LCMS: m/z (ESI), [M+H] +=396.15。 步驟4. 2-{6-[(2-硝基苯基)氨基]-1,4-氧氮雜環庚-4-基}乙醇(INT-B7-4)的製備。將4-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}- N-(2-硝基苯基)-1,4-氧氮雜環庚-6-胺(1.7 g,4.29 mmol,1當量)和四丁基氟化銨(2.25 g,8.59 mmol,2當量)於四氫呋喃(20 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×200 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過C18 FLASH,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3 .H 2O和乙腈的水作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈橙色油的1 g 2-{6-[(2-硝基苯基)氨基]-1,4-氧氮雜環庚-4-基}乙醇(INT-B7-4) (82%)。LCMS: m/z (ESI), [M+H] +=282.10。 1H NMR (DMSO-d 6, 400 MHz) δ 2.64-2.71 (3H, m), 2.81-2.94 (2H, m), 3.03-3.18 (1H, m), 3.45-3.59 (2H, m), 3.59-3.75 (3H, m), 3.95-3.99 (1H, m), 4.04-4.13 (1H, m), 4.41 (1H, t), 6.66-7.00 (1H, m), 7.01-7.02 (1H, m), 7.49-7.53 (1H, m), 8.07-8.10 (1H, m), 8.79 (1H, d) 步驟5. 2-甲基-6-[1-甲基-5-(2-{6-[(2-硝基苯基)氨基]-1,4-氧氮雜環庚-4-基}乙氧基)吡唑-4-基]吡啶-4-甲酸甲酯(INT-B7-5)的製備。在氮氣氣氛下在0℃下向2-{6-[(2-硝基苯基)氨基]-1,4-氧氮雜環庚-4-基}乙醇(1.00 g,3.55 mmol,1.2當量)和三苯基膦(2.33 g,8.89 mmol,3當量)於四氫呋喃(20 mL)中的攪拌混合物中添加含偶氮二甲酸二異丙酯(1.80 g,8.89 mmol,3當量)和2-(5-羥基-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(0.73 g,2.96 mmol,1.00當量)的THF(10 mL)。將混合物在0℃下攪拌2 h。將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×200 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上使用石油醚/乙酸乙酯(1/1)洗脫進行純化,以得到呈橙色油的0.9 g 2-甲基-6-[1-甲基-5-(2-{6-[(2-硝基苯基)氨基]-1,4-氧氮雜環庚-4-基}乙氧基)吡唑-4-基]吡啶-4-甲酸甲酯(INT-B7-5)(59%)。LCMS: m/z (ESI), [M+H] +=511.20。 1H NMR (DMSO-d 6, 400 MHz) δ 2.52 (3H, s), 2.91-2.95 (2H, m), 3.05 (1H, d), 3.10 (1H, q), 3.17 (2H, d), 3.67 (2H, d), 3.70 (3H, s), 3.87 (3H, s), 3.90-3.95 (1H, m), 4.05-4.12 (2H, m), 4.20-4.34 (2H, m), 6.64-6.68 (1H, m), 6.97 (1H, d), 7.42 (1H, d), 7.45-7.50 (1H, m), 7.81 (1H, d), 7.88 (1H, s), 8.02-8.05 (1H, m), 8.67 (1H, d)。 步驟6. 2-[5-(2-{6-[(2-氨基苯基)氨基]-1,4-氧氮雜環庚-4-基}乙氧基)-1-甲基吡唑-4-基]-6-甲基吡啶-4-甲酸甲酯(INT-B7-6)的製備。在0℃下向2-甲基-6-[1-甲基-5-(2-{6-[(2-硝基苯基)氨基]-1,4-氧氮雜環庚-4-基}乙氧基)吡唑-4-基]吡啶-4-甲酸甲酯(540 mg,1.06 mmol,1當量)和雷尼鎳(725 mg,8.46 mmol,8當量)於甲醇(5 mL)中的攪拌混合物中添加水合肼(79 mg,1.59 mmol,1.5當量)。將混合物在氮氣氣氛下在0℃下攪拌2 h。將所得混合物過濾。將濾餅用甲醇(3×50 mL)洗滌,並且將所得溶液在減壓下濃縮,以得到呈棕色固體的480 mg 2-[5-(2-{6-[(2-氨基苯基)氨基]-1,4-氧氮雜環庚-4-基}乙氧基)-1-甲基吡唑-4-基]-6-甲基吡啶-4-甲酸甲酯(INT-B7-6)。此材料在不經進一步純化的情況下用於下一反應中。LCMS: m/z (ESI), [M+H] += 481.25。 步驟7. 2-(5-{2-[6-(2-氨基-1,3-苯並二氮唑-1-基)-1,4-氧氮雜環庚-4-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(INT-B7-7)的製備。將2-[5-(2-{6-[(2-氨基苯基)氨基]-1,4-氧氮雜環庚-4-基}乙氧基)-1-甲基吡唑-4-基]-6-甲基吡啶-4-甲酸甲酯(500 mg,1.04 mmol,1當量)和BrCN(132 mg,1.25 mmol,1.2當量)於二氯甲烷(5 mL)中的混合物在氮氣氣氛下在室溫下攪拌過夜。將反應用水(80 mL)淬滅,並且將混合物用乙酸乙酯(3×60 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用石油醚/乙酸乙酯(1/1)洗脫進行純化,以得到呈橙色油的400 mg 2-(5-{2-[6-(2-氨基-1,3-苯並二氮唑-1-基)-1,4-氧氮雜環庚-4-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(INT-B7-7)(76%)。LCMS: m/z (ESI), [M+H] +=506.30。 步驟8. 2-(5-{2-[6-(2-氨基-1,3-苯並二氮唑-1-基)-1,4-氧氮雜環庚-4-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-羧酸(INT-B7-8)的製備。將2-(5-{2-[6-(2-氨基-1,3-苯並二氮唑-1-基)-1,4-氧氮雜環庚-4-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(200 mg,0.39 mmol,1當量)和LiOH.H 2O(25 mg,0.59 mmol,1.5當量)於四氫呋喃(4 mL)和水(1 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h,並且在減壓下濃縮。將殘餘物通過C18 FLASH,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3 .H 2O和乙腈的水作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈粉色固體的180 mg 2-(5-{2-[6-(2-氨基-1,3-苯並二氮唑-1-基)-1,4-氧氮雜環庚-4-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-羧酸(INT-B7-8)(92%)。LCMS: m/z (ESI), [M+H] +=492.30。 步驟9. 15,21-二甲基-23,29-二氧雜-2,9,11,16,20,21, 26-七氮雜六環[24.4.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18, 22}]三十二-3,5,7,9,13(3 2),14,16,18(22),19-壬-12-酮(實施例B7)的製備。將2-(5-{2-[6-(2-氨基-1,3-苯並二氮唑-1-基)-1,4-氧氮雜環庚-4-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-羧酸(160 mg,0.33 mmol,1當量)、HATU(186 mg,0.49 mmol,1.5當量)和 N,N-二異丙基乙胺(126 mg,0.98 mmol,3當量)於1,4-二噁烷(5 mL)中的混合物在氮氣氣氛下在室溫下攪拌過夜。將反應用水(30 mL)淬滅,並且將混合物用乙酸乙酯(3×20 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過與甲醇(50 mL)一起研磨進行純化,以得到呈白色固體的8.3 mg 15,21-二甲基-23,29-二氧雜-2,9,11,16,20,21,26-七氮雜六環[24.4.1.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]三十二-3,5,7,9,13(32),14,16,18(22),19-壬-12-酮(實施例B7)(5%)。LCMS: m/z (ESI), [M+H] += 474.15。 1H NMR (DMSO-d 6, 400 MHz) δ 2.57 (3H, s), 2.82-2.91 (3H, m), 2.93-3.01 (1H, m), 3.24-3.44 (2H, m), 3.75 (3H, s), 3.72-3.78 (1H, m), 3.96 (1H, br s), 4.03-4.26 (2H, m), 4.59 (2H, d), 5.45 (1H, br s), 7.20-7.33 (2H, m), 7.52-7.58 (2H, m), 7.69 (1H, d), 7.92 (1H, s), 8.61 (1H, s), 12.67 (1H, s)。 步驟10. 15,21-二甲基-23,29-二氧雜-2,9,11,16,20,21, 26-七氮雜六環[24.4.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18, 22}]三十二-3,5,7,9,13(32),14,16,18(22),19-壬-12-酮,異構體1(實施例B7A)和異構體2(實施例B7B)的製備。將15,21-二甲基-23,29-二氧雜-2,9,11,16,20,21,26-七氮雜六環[24.4.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十二-3,5,7,9,13(32),14,16,18(22),19-壬-12-酮(實施例B7,80 mg)的混合物通過Prep-手性-HPLC使用CHIRALPAK IG柱(2*25 cm,5 μm)使用含有0.2% DEA的己烷和EtOH/CH 2Cl 2(1/1)作為流動相來分離,以得到呈白色固體的32 mg異構體1(實施例B7A,40%)和34 mg異構體2(實施例B7B,42%)。 異構體1:LCMS: m/z (ESI), [M+H] +=474.15。 1H NMR (DMSO-d 6, 400 MHz) δ 2.57 (3H, s), 2.82-2.91 (3H, m), 2.93-3.01 (1H, m), 3.24-3.44 (2H, m), 3.75 (3H, s), 3.72-3.78 (1H, m), 3.96 (1H, br s), 4.03-4.26 (2H, m), 4.59 (2H, d), 5.45 (1H, br s), 7.20-7.33 (2H, m), 7.52-7.58 (2H, m), 7.69 (1H, d), 7.92 (1H, s), 8.61 (1H, s), 12.67 (1H, s)。手性-HPLC,Rt=2.016分鐘。 異構體2:LCMS: m/z (ESI), [M+H] +=474.15。 1H NMR (DMSO-d 6, 400 MHz) δ 2.57 (3H, s), 2.82-2.91 (3H, m), 2.93-3.01 (1H, m), 3.24-3.44 (2H, m), 3.75 (3H, s), 3.72-3.78 (1H, m), 3.96 (1H, br s), 4.03-4.26 (2H, m), 4.59 (2H, d), 5.45 (1H, br s), 7.20-7.33 (2H, m), 7.52-7.58 (2H, m), 7.69 (1H, d), 7.92 (1H, s), 8.61 (1H, s), 12.67 (1H, s)。手性-HPLC,Rt=2.626分鐘。 實施例B8A和實施例B8B 15,21-二甲基-23-氧雜-2,9,11,16,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,15,17(33),18(22), 19-壬-12-酮,異構體1(實施例B8A)和異構體2(實施例B8B) 步驟1. 2-甲基-6-[1-甲基-5-(2-{1-[(2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙氧基)吡唑-4-基]吡啶-4-甲酸甲酯(INT-B8-1)的製備。在氮氣氣氛下在0℃下向2-{1-[(2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙醇(INT-A61-4,380 mg,1.30 mmol,1當量)和三苯基膦(685 mg,2.61 mmol,2當量)於四氫呋喃(10 mL)中的攪拌混合物中添加2-(5-羥基-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(M1,323 mg,1.30 mmol,1當量)和偶氮二甲酸二異丙酯(528 mg,2.61 mmol,2當量)。將所得混合物在0℃下攪拌2 h。將反應用水(30 mL)淬滅,並且將混合物用CH 2Cl 2(3×50 mL)萃取。將合併的有機層用鹽水(3×75 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-TLC使用石油醚/乙酸乙酯(1/1)洗脫進行純化,以得到呈白色固體的460 mg 2-甲基-6-[1-甲基-5-(2-{1-[(2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙氧基)吡唑-4-基]吡啶-4-甲酸甲酯(INT-B8-1)(67%)。LCMS: m/z (ESI), [M+H] +=521.15。 步驟2. 2-[5-(2-{1-[(2-氨基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙氧基)-1-甲基吡唑-4-基]-6-甲基吡啶-4-甲酸甲酯(INT-B8-2)的製備。在氮氣氣氛下在室溫下向2-甲基-6-[1-甲基-5-(2-{1-[(2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙氧基)吡唑-4-基]吡啶-4-甲酸甲酯(500 mg,0.96 mmol,1當量)和雷尼鎳(80 mg)於甲醇(10 mL)中的攪拌混合物中添加水合肼(92 mg,2.88 mmol,3當量)。將所得混合物在室溫下攪拌1 h。將所得混合物過濾,並且將濾餅用甲醇(3×20 mL)洗滌。將所得溶液在減壓下濃縮。將混合物通過prep-TLC使用二氯甲烷/甲醇(20/1)洗脫進行純化,以得到呈黃色固體的400 mg 2-[5-(2-{1-[(2-氨基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙氧基)-1-甲基吡唑-4-基]-6-甲基吡啶-4-甲酸甲酯(INT-B8-2)(85%)。LCMS: m/z (ESI), [M+H] +=491.15。 步驟3. 2-(5-{2-[1-(2-氨基-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(INT-B8-3)的製備。在氮氣氣氛下在室溫下向2-[5-(2-{1-[(2-氨基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙氧基)-1-甲基吡唑-4-基]-6-甲基吡啶-4-甲酸甲酯(455 mg,0.93 mmol,1當量)於乙醇(20 mL)中的攪拌混合物中添加BrCN(334 mg,3.15 mmol,3.4當量)。將所得混合物在室溫下攪拌1 h,並且在減壓下濃縮。將殘餘物通過prep-TLC使用二氯甲烷/甲醇(20/1)洗脫進行純化,以得到呈棕色固體的350 mg 2-(5-{2-[1-(2-氨基-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(INT-B8-3)(73%)。LCMS: m/z (ESI), [M+H] +=516.15。 步驟4. 2-(5-{2-[1-(2-氨基-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯,異構體1(INT-B8-3A)和異構體2(INT-B8-3B)的製備。 將2-(5-{2-[1-(2-氨基-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(350 mg)的外消旋混合物通過Prep-手性-HPLC使用CHIRALPAK IG手性柱(2×25 cm,5 μm)使用含有0.1%二乙胺的甲基叔丁基醚和乙醇/CH 2Cl 2(1/1)作為流動相來分離,以得到呈白色固體的90 mg異構體1(INT-B8-3A,25%)和80 mg異構體2(INT-B8-3B,22%)。手性-HPLC:Rt(異構體1)=1.312分鐘,Rt(異構體2)=2.047分鐘。 步驟5. 2-(5-{2-[1-(2-氨基-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-羧酸(INT-B8-4A)的製備。在氮氣氣氛下在室溫下向2-(5-{2-[1-(2-氨基-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(INT-B8-3A,85 mg,0.16 mmol,1當量)於四氫呋喃(8 mL)中的攪拌混合物中添加含LiOH .H 2O(16 mg,0.66 mmol,4當量)的H 2O(2 mL)。將所得混合物在60℃下攪拌30分鐘,冷卻到室溫,並且在減壓下濃縮。將殘餘物通過反向快速色譜法使用C18矽膠柱使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃色固體的80 mg 2-(5-{2-[1-(2-氨基-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-羧酸(INT-B8-4A)(96%)。LCMS: m/z (ESI), [M+H] +=502.20。 步驟6. 15,21-二甲基-23-氧雜-2,9,11,16,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]三十三-3,5,7,9,13,15,17(33),18(22), 19-壬-12-酮,異構體1(實施例B8A)的製備。將 N, N-二異丙基乙胺(58 mg,0.45 mmol,3當量)、2-(5-{2-[1-(2-氨基-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-羧酸(INT-B8-4A,75 mg,0.15 mmol,1當量)和 N, N, N, N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(85 mg,0.22 mmol,1.5當量)於1,4-二噁烷(7 mL)中的混合物在氮氣氣氛下在60℃下攪拌1 h。將混合物冷卻到室溫。將反應用水(20 mL)淬滅,並且將混合物用CH 2Cl 2(3×30 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC進行純化並且使用二氯甲烷/甲醇(25/1)洗脫,以得到呈黃色固體的57.4 mg 15,21-二甲基-23-氧雜-2,9,11,16,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,15,17(33), 18(22),19-壬-12-酮(異構體1,實施例B8A)(79%)。LCMS: m/z (ESI), [M+H] +=484.30。 1H NMR (DMSO-d 6, 400 MHz) δ 1.59-1.80 (3H, m), 2.02 (1H, d), 2.38-2.50 (3H, m), 2.56 (3H, s), 2.63-2.82 (3H, m), 3.22-3.45 (2H, m), 3.74 (3H, s), 3.77-3.83 (1H, m), 4.56 (1H, t), 4.71 (1H, d), 7.16-7.25 (2H, m), 7.55-7.63 (2H, m), 7.73-7.84 (1H, m), 7.94 (1H, s), 8.44 (1H, s), 12.87 (1H, s)。SFC-HPLC,Rt=1.282分鐘。 步驟7. 2-(5-{2-[1-(2-氨基-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-羧酸(INT-B8-4B)的製備。在氮氣氣氛下在室溫下向2-(5-{2-[1-(2-氨基-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(INT-B8-3B,75 mg,0.15 mmol,1當量)於四氫呋喃(8 mL)中的攪拌混合物中添加含LiOH .H 2O(14 mg,0.58 mmol,4當量)的H 2O(2 mL)。將所得混合物在60℃下攪拌30分鐘。將混合物冷卻到室溫並在減壓下濃縮。將殘餘物通過反向快速色譜法使用C18矽膠柱使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈灰白色固體的70 mg 2-(5-{2-[1-(2-氨基-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-羧(INT-B8-4B)酸(96%)。LCMS: m/z (ESI), [M+H] +=502.20。 步驟8. 15,21-二甲基-23-氧雜-2,9,11,16,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]三十三-3,5,7,9,13,15,17(33),18(22), 19-壬-12-酮,異構體2(實施例B8B)的製備。將 N, N-二異丙基乙胺(50 mg,0.39 mmol,3當量)、2-(5-{2-[1-(2-氨基-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-羧酸(INT-B8-4B,65 mg,0.13 mmol,1當量)和 N, N, N, N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(74 mg,0.20 mmol,1.5當量)於1,4-二噁烷(6 mL)中的混合物在氮氣氣氛下在60℃下攪拌1 h。將混合物冷卻到室溫。將反應用水(20 mL)淬滅,並且將混合物用CH 2Cl 2(3×30 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC進行純化並且使用二氯甲烷/甲醇(25/1)洗脫,以得到呈黃色固體的46.5 mg 15,21-二甲基-23-氧雜-2,9,11,16,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,15,17(33), 18(22),19-壬-12-酮,異構體2(實施例B8B)(74%)。LCMS: m/z (ESI), [M+H] +=484.25。 1H NMR (DMSO-d 6, 400 MHz) δ 1.59-1.80 (3H, m), 2.02 (1H, d), 2.38-2.50 (3H, m), 2.56 (3H, s), 2.63-2.82 (3H, m), 3.22-3.45 (2H, m), 3.74 (3H, s), 3.77-3.83 (1H, m), 4.56 (1H, t), 4.71 (1H, d), 7.16-7.25 (2H, m), 7.55-7.63 (2H, m), 7.73-7.84 (1H, m), 7.94 (1H, s), 8.44 (1H, s), 12.87 (1H, s)。SFC-HPLC,Rt=1.060分鐘。 實施例B9A和實施例B9B 15,21-二甲基-5-(4-甲基哌嗪-1-基)-23-氧雜-2,9,11,16, 20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,15,17(33),18(22), 19-壬-12-酮,異構體1(實施例B9A)和異構體2(實施例B9B)。 步驟1. 2-[5-(2-{1-[(5-溴-2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙氧基)-1-甲基吡唑-4-基]-6-甲基吡啶-4-甲酸甲酯(INT-B9-1)的製備。在氮氣氣氛下在0℃下向2-(5-羥基-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(M1,1.40 g,5.67 mmol,1.05當量)和三苯基膦(3.97 g,15.12 mmol,2.8當量)於四氫呋喃(30 mL)中的攪拌混合物中添加偶氮二甲酸二異丙酯(2.73 g,13.50 mmol,2.5當量),隨後添加2-{1-[(5-溴-2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙醇(M4-11,2 g,5.40 mmol,1當量)。在0℃下攪拌3 h後,將反應用水(400 mL)淬滅,並且將混合物用乙酸乙酯(3×300 mL)萃取。將合併的有機層用鹽水(3×300 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠柱使用二氯甲烷/甲醇(30/1)洗脫進行純化,以得到呈黃色油的2 g 2-[5-(2-{1-[(5-溴-2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙氧基)-1-甲基吡唑-4-基]-6-甲基吡啶-4-甲酸甲酯(INT-B9-1)(61%)。LCMS: m/z (ESI), [M+H] +=601.00。 步驟2. 2-[5-(2-{1-[(2-氨基-5-溴苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙氧基)-1-甲基吡唑-4-基]-6-甲基吡啶-4-甲酸甲酯(INT-B9-2)的製備。在室溫下向2-[5-(2-{1-[(5-溴-2-硝基苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙氧基)-1-甲基吡唑-4-基]-6-甲基吡啶-4-甲酸甲酯(2 g,3.33 mmol,1當量)於甲醇(60 mL)中的攪拌混合物中添加雷尼鎳(457 mg,5.33 mmol,1.6當量)和NH 2NH 2 .H 2O(835 mg,16.68 mmol,5當量)。將所得混合物在室溫下攪拌1 h。將反應用水(300 mL)淬滅,並且將混合物用乙酸乙酯(3×200 mL)萃取。將合併的有機層用鹽水(3×200 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法使用二氯甲烷/甲醇(20/1)洗脫進行純化,以得到呈紅棕色固體的1.34 g 2-[5-(2-{1-[(2-氨基-5-溴苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙氧基)-1-甲基吡唑-4-基]-6-甲基吡啶-4-甲酸甲酯(INT-B9-2)(70%)。LCMS: m/z (ESI), [M+H] += 571.05。 步驟3. 2-(5-{2-[1-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(INT-B9-3)的製備。在室溫下向2-[5-(2-{1-[(2-氨基-5-溴苯基)氨基]-3-氮雜雙環[3.2.1]辛-3-基}乙氧基)-1-甲基吡唑-4-基]-6-甲基吡啶-4-甲酸甲酯(500 mg,0.87 mmol,1當量)於乙醇(8 mL)中的攪拌混合物中添加BrCN(139 mg,1.31 mmol,1.5當量)。將所得混合物在室溫下攪拌2小時。將反應用飽和NaHCO 3(100 mL)處理,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層經無水Na 2SO 4乾燥,並且在減壓下濃縮。將殘餘物通過矽膠色譜法使用二氯甲烷/甲醇(15/1)洗脫進行純化,以得到呈紅棕色油的400 mg 2-(5-{2-[1-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(INT-B9-3)(76.63%)。LCMS: m/z (ESI), [M+H] +=596.10。 步驟4. 2-(5-{2-[1-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯,異構體1(INT-B9-3A)和異構體2(INT-B9-3B)的製備。將2-(5-{2-[1-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(400 mg)的外消旋混合物通過Prep-手性-HPLC使用CHIRAL ART纖維素-SB柱(5*25 cm,5 μm)使用CO 2和CH 3OH/CH 2Cl 2(2/1)作為移動相來分離,以得到呈白色固體的150 mg異構體1(INT-B9-3A,42%)和150 mg異構體2(INT-B9-3B,42%)。SFC-HPLC,Rt(異構體1)=1.760分鐘,Rt(異構體2)=1.889分鐘。 步驟5. 2-(5-{2-1-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-羧酸(異構體1,INT-B9-4A)的製備。在室溫下向2-(5-{2-[1-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(490 mg,0.82 mmol,1當量)於THF(8 mL)中的攪拌溶液中添加含LiOH(78 mg,3.29 mmol,4當量)的H 2O(2 mL)。將所得混合物在室溫下攪拌3 h。將混合物在減壓下濃縮,並且將殘餘物通過反向快速色譜法使用水和乙腈作為移動相洗脫進行純化,以得到呈白色固體的410 mg 2-(5-{2-[1-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-羧酸(異構體1,INT-B9-4A)(85%)。LCMS: m/z (ESI), [M+H] +=580.05。 步驟6. 5-溴-15,21-二甲基-23-氧雜-2,9,11,16,20,21, 26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3, 8}.0^{18,22}]三十三-3,5,7,9,13,15,17(33),18(22), 19-壬-12-酮(異構體1,INT-B9-5A)的製備。在室溫下向2-(5-{2-[1-(2-氨基-6-溴-1,3-苯並二氮唑-1-基)-3-氮雜雙環[3.2.1]辛-3-基]乙氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-羧酸(390 mg,0.67 mmol,1當量)於二噁烷(10 mL)中的攪拌溶液中添加 N,N-二異丙基乙胺(260 mg,2.01 mmol,3當量)和 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(383 mg,1.01 mmol,1.5當量)。將反應混合物在氮氣氣氛下在50℃下攪拌2 h。在冷卻到室溫後,將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將粗產物通過prep-TLC使用二氯甲烷/甲醇(25/1)洗脫進行純化,以得到呈淡黃色固體的350 mg 5-溴-15,21-二甲基-23-氧雜-2,9,11,16,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,15,17(33),18(22), 19-壬-12-酮(異構體1,INT-B9-5A)(92%)。LCMS: m/z (ESI), [M+H] +=564.05。 步驟7. 15,21-二甲基-5-(4-甲基哌嗪-1-基)-23-氧雜-2,9,11,16,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,15,17(33), 18(22), 19-壬-12-酮(異構體1,實施例B9A)的製備。在氮氣氣氛下在室溫下向5-溴-15,21-二甲基-23-氧雜-2,9,11,16,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,15,17(33), 18(22),19-壬-12-酮(130 mg,0.23 mmol,1當量)、1-甲基哌嗪(34.7 mg,0.35 mmol,1.5當量)和BrettPhos Pd G 3(62.8 mg,0.07 mmol,0.3當量)於二噁烷(5 mL)中的攪拌混合物中逐滴添加LiHMDS(2.3 mL,2.31 mmol,10當量)。將反應混合物在氮氣氣氛下在60℃下攪拌1 h。在冷卻到室溫後,將反應用飽和NH 4Cl(30 mL)淬滅,並且將混合物用二氯甲烷(3×20 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-TLC使用二氯甲烷/甲醇(18/1)洗脫進行純化,以得到呈白色固體的41.6 mg 15,21-二甲基-5-(4-甲基哌嗪-1-基)-23-氧雜-2,9,11,16,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,15,17(33),18(22),19-壬-12-酮,異構體1(實施例B9A)(30%)。LCMS: m/z (ESI), [M+H] +=582.40。 1H NMR (DMSO-d 6, 400 MHz) δ 1.52-1.82 (3H, m), 2.03 (1H, d), 2.24 (3H, s), 2.40-2.60 (8H, m), 2.55 (3H, s), 2.64-2.79 (2H, m), 3.12 (4H, q), 3.20-3.30 (1H, m), 3.37-3.45 (1H, m), 3.74 (3H, s), 3.77 (1H, br s), 4.56 (1H, t), 4.70 (1H, d), 6.93 (1H, dd), 7.19 (1H, d), 7.42 (1H, d), 7.57 (1H, d), 7.94 (1H, s), 8.42 (1H, s), 12.69 (1H, s)。SFC-HPLC,Rt=3.968分鐘。 使用INT-B9-3B作為起始材料,上文針對實施例B9A的合成描述的相同反應順序得到呈白色固體的48.4 mg 15,21-二甲基-5-(4-甲基哌嗪-1-基)-23-氧雜-2,9,11,16,20, 21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,15,17(33), 18(22),19-壬-12-酮,異構體2(實施例B9B)。LCMS: m/z (ESI), [M+H] +=582.40。 1H NMR (DMSO-d 6, 400 MHz) δ 1.52-1.82 (3H, m), 2.03 (1H, d), 2.24 (3H, s), 2.40-2.60 (8H, m), 2.55 (3H, s), 2.64-2.79 (2H, m), 3.12 (4H, q), 3.20-3.30 (1H, m), 3.37-3.45 (1H, m), 3.74 (3H, s), 3.77 (1H, br s), 4.56 (1H, t), 4.70 (1H, d), 6.93 (1H, dd), 7.19 (1H, d), 7.42 (1H, d), 7.57 (1H, d), 7.94 (1H, s), 8.42 (1H, s), 12.69 (1H, s)。SFC-HPLC,Rt=3.745分鐘。 實施例B10A和實施例B10B 15,21-二甲基-5-(嗎啉-4-基)-23-氧雜-2,9,11,16,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]三十三-3,5,7,9,13,15,17(33),18(22),19-壬-12-酮,異構體1(實施例B10A)和異構體2(實施例B10B) 標題化合物使用針對實施例B9A和實施例B9B的製備所描述的相同的反應順序來製備。 實施例B10A:LCMS: m/z (ESI), [M+H]+=569.40。 1H NMR (DMSO-d 6, 400 MHz) δ 1.56-1.81 (3H, m), 2.03 (1H, d), 2.37-2.48 (3H, m), 2.55 (3H, s), 2.57-2.79 (3H, m), 3.04-3.18 (4H, m), 3.19-3.29 (2H, m), 3.37-3.47 (1H, m), 3.74 (3H, s), 3.75-3.83 (4H, m), 4.56 (1H, t), 4.71 (1H, d), 6.94 (1H, dd), 7.21 (1H, d), 7.45 (1H, d), 7.57 (1H, d), 7.94 (1H, s), 8.43 (1H, s), 12.71 (1H, s)。SFC-HPLC,Rt=1.549分鐘 實施例B10B:LCMS: m/z (ESI), [M+H]+=569.25。 1H NMR (DMSO-d 6, 400 MHz) δ 1.56-1.81 (3H, m), 2.03 (1H, d), 2.37-2.48 (3H, m), 2.55 (3H, s), 2.57-2.79 (3H, m), 3.04-3.18 (4H, m), 3.19-3.29 (2H, m), 3.37-3.47 (1H, m), 3.74 (3H, s), 3.75-3.83 (4H, m), 4.56 (1H, t), 4.71 (1H, d), 6.94 (1H, dd), 7.21 (1H, d), 7.45 (1H, d), 7.57 (1H, d), 7.94 (1H, s), 8.43 (1H, s), 12.71 (1H, s)。SFC-HPLC,Rt=1.639分鐘 實施例B11 (12 R)-5,12,27-三甲基-8-氧雜-5,14,21,23,28-五氮雜五環[23.3.1.0^{2,7}.0^{14,22}.0^{15,20}]二十九-1(29),2,6,15, 17,19,21,25,27-壬烯-4,24-二酮 步驟1. (4 R)-4-甲基-5-[(2-硝基苯基)氨基]戊-1-醇(INT-B11-1)的製備。將鄰氟硝基苯(1.32 g,9.386 mmol,1.1當量)、(4 R)-5-氨基-4-甲基戊-1-醇(1 g,8.533 mmol,1.00當量)和K 2CO 3(3.54 g,25.599 mmol,3當量)於乙腈(20 mL)中的混合物在氮氣氣氛下在60℃下攪拌2 h。將混合物冷卻到室溫。將反應用水(800 mL)淬滅,並且將混合物用二氯甲烷(3×600 mL)萃取。將合併的有機層用鹽水(3×500 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法使用石油醚/乙酸乙酯(1/1)洗脫進行純化,以得到呈橙色油的1.6 g (4 R)-4-甲基-5-[(2-硝基苯基)氨基]戊-1-醇(INT-B11-1)(78%)。LCMS: m/z (ESI), [M+H] +=239.05。 步驟2. 2-(苄氧基)-5-[(2-甲氧基乙氧基)甲氧基]吡啶(INT-B11-2)的製備。在0℃下向6-(苄氧基)吡啶-3-醇(5 g,24.84 mmol,1當量)於 N,N-二甲基甲醯胺中的攪拌混合物中添加氫化鈉(油中60%,4 g,12.42 mmol,2當量)。在添加1-(氯甲氧基)-2-甲氧基乙烷(3.1 g,24.84 mmol,1當量)之前,將混合物攪拌15分鐘。將混合物在0℃下攪拌2 h。將反應用水(50 mL)淬滅,並且將混合物用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在用石油醚/乙酸乙酯(3/1)洗脫的矽膠柱上進行純化,以得到呈淡黃色油的6 g 2-(苄氧基)-5-[(2-甲氧基乙氧基)甲氧基]吡啶(INT-B11-2)(75%)。LCMS: m/z (ESI), [M+H] +=290.10。 1H NMR (DMSO- d 6, 400 MHz) δ 3.22 (3H, s), 3.36-3.51 (2H, m), 3.65-3.80 (2H, m), 5.21 (2H, s), 5.29 (2H, s), 6.85 (1H, d), 7.28-7.42 (3H, m), 7.35-7.50 (2H, m), 7.50 (1H, d), 7.93 (1H, d)。 步驟3. 2-(苄氧基)-4-碘-5-[(2-甲氧基乙氧基)甲氧基]吡啶(INT-B11-3)的製備。在氮氣氣氛下在-78℃下向2-(苄氧基)-5-[(2-甲氧基乙氧基)甲氧基]吡啶(5.8 g,20.04 mmol,1當量)於四氫呋喃(5 mL)中的攪拌混合物中添加正丁基鋰溶液(四氫呋喃中2.5 M,17 mL,2 mmol)。將混合物在-78℃下攪拌30分鐘。然後逐滴添加I 2(8.1 g,32.07 mmol,1.60當量)於10 mL四氫呋喃中的溶液,並且將混合物在氮氣氣氛下在-78℃下攪拌1 h。將反應用飽和NH 4Cl(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在用石油醚/乙酸乙酯(3/1)洗脫的矽膠柱上進行純化,以得到呈黃色油的5.5 g 2-(苄氧基)-4-碘-5-[(2-甲氧基乙氧基)甲氧基]吡啶(INT-B11-3)(59%)。LCMS: m/z (ESI), [M+H] +=415.95。 1H NMR (CDCl 3, 400 MHz) δ 3.42 (3H, s), 3.52-3.70 (2H, m), 3.85-4.02 (2H, m), 5.27 (2H, s), 5.33 (2H, s), 7.35-7.46 (6H, m), 7.94 (1H, s) 步驟4. 2'-(苄氧基)-6-氯-5'-[(2-甲氧基乙氧基)甲氧基]-[2,4'-雙吡啶]-4-甲酸甲酯(INT-B11-4)的製備。將2-氯-6-(4,4,5,5-四甲基-1,3,2-二氧雜環戊硼烷-2-基)吡啶-4-甲酸甲酯(M1-1,4 g粗產物)、2-(苄氧基)-4-碘-5-[(2-甲氧基乙氧基)甲氧基]吡啶(4.1 g,10.08 mmol,1當量)、Pd 2(dba) 3(0.92 g,1.00 mmol,0.1當量)、雙(金剛烷-1-基)(丁基)膦(0.36 g,1.00 mmol,0.1當量)和Cs 2CO 3(9.8 g,30.24 mmol,3.00當量)於甲苯/H 2O(40 mL/10 mL)中的混合物在氮氣氣氛下在80℃下攪拌16 h。將混合物冷卻到室溫。將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上使用石油醚/乙酸乙酯(1/1)洗脫進行純化,以得到呈棕色固體的4.8 g 2'-(苄氧基)-6-氯-5'-[(2-甲氧基乙氧基)甲氧基]-[2,4'-雙吡啶]-4-甲酸甲酯(INT-B11-4)(93%)。LCMS: m/z (ESI), [M+H] +=459.05。 步驟5. 2'-(苄氧基)-5'-[(2-甲氧基乙氧基)甲氧基]-6-甲基-[2,4'-雙吡啶]-4-甲酸甲酯(INT-B11-5)的製備。將2'-(苄氧基)-6-氯-5'-[(2-甲氧基乙氧基)甲氧基]-[2,4'-雙吡啶]-4-甲酸甲酯(2.8 g,6.10 mmol,1當量)、Pd(dppf)Cl 2(0.89 g,1.22 mmol,0.2當量)、K 2CO 3(1.6 g,12.20 mmol,2當量)、PCy 3.HBF 4(0.67 g,1.83 mmol,0.3當量)和三甲基-1,3,5,2,4,6-三氧雜三硼環己烷(2.07 g,16.47 mmol,2.7當量)於1,4-二噁烷(28 mL)中的混合物在氮氣氣氛下在100℃下攪拌3 h。將混合物冷卻到室溫。將反應用水(200 mL)淬滅,並且將混合物用乙酸乙酯(3×200 mL)萃取。將合併的有機層用鹽水(3×200 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上使用石油醚/乙酸乙酯(1/1)洗脫進行純化,以得到呈黃色固體的1.8 g 2'-(苄氧基)-5'-[(2-甲氧基乙氧基)甲氧基]-6-甲基-[2,4'-雙吡啶]-4-甲酸甲酯(INT-B11-5)(60%)。LCMS: m/z (ESI), [M+H] += 439.15。 步驟6. 2'-羥基-5'-[(2-甲氧基乙氧基)甲氧基]-6-甲基-[2,4'-雙吡啶]-4-甲酸甲酯(INT-B11-6)的製備。將2'-(苄氧基)-5'-[(2-甲氧基乙氧基)甲氧基]-6-甲基-[2,4'-雙吡啶]-4-甲酸甲酯(1.8 g,4.105 mmol,1當量)和Pd/C(10%,500 mg)於甲醇(10 mL)中的混合物在氫氣氣氛下攪拌1 h。將所得混合物過濾,並且將濾餅用甲醇(150 mL)洗滌。將溶液在減壓下濃縮。將殘餘物在矽膠柱上使用二氯甲烷/甲醇(10/1)洗脫進行純化,以得到呈黃色固體的1.3 g 2'-羥基-5'-[(2-甲氧基乙氧基)甲氧基]-6-甲基-[2,4'-雙吡啶]-4-甲酸甲酯(INT-B11-6)(82%)。LCMS: m/z (ESI), [M+H] += 349.05。 步驟7. 5'-[(2-甲氧基乙氧基)甲氧基]-1',6-二甲基-2'-氧代-[2,4'-雙吡啶]-4-甲酸甲酯(INT-B11-7)的製備 將methyl 2'-羥基-5'-[(2-甲氧基乙氧基)甲氧基]-6-甲基-[2,4'-雙吡啶]-4-甲酸甲酯(1.1 g,3.15 mmol,1當量)、K 2CO 3(1.3 g,9.47 mmol,3當量)和CH 3I(672 mg,4.73 mmol,1.5當量)於丙酮(10 mL)中的混合物在氮氣氣氛下在60℃下攪拌12 h。將混合物冷卻到室溫。將反應用水(50 mL)淬滅,並且將混合物用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過C18 FLASH,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3 .H 2O和乙腈的水作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈淡黃色油的1 g 5'-[(2-甲氧基乙氧基)甲氧基]-1',6-二甲基-2'-氧代-[2,4'-雙吡啶]-4-甲酸甲酯(INT-B11-7)(83%)。LCMS: m/z (ESI), [M+H] +=363.10。 1H NMR (CDCl 3, 400 MHz) δ 2.66 (3H, s), 3.41 (3H, s), 3.58 (5H, d), 3.71-3.96 (2H, m), 3.96 (3H, s), 5.02 (2H, s), 6.92 (1H, s), 7.45 (1H, s), 7.73 (1H, d), 7.99 (1H, d) 步驟8. 5'-羥基-1',6-二甲基-2'-氧代-[2,4'-雙吡啶]-4-甲酸甲酯(INT-B11-8)的製備。將5'-[(2-甲氧基乙氧基)甲氧基]-1',6-二甲基-2'-氧代-[2,4'-雙吡啶]-4-甲酸甲酯(370 mg,1.02 mmol,1當量)和HCl(10 mL,MeOH中4 M)的溶液在氮氣氣氛下在室溫下攪拌1.5 h。將混合物在減壓下濃縮。將殘餘物用二氯甲烷(30 mL)和NaHCO 3溶液處理並過濾。將濾液在減壓下濃縮,以得到呈黃色固體的260 mg 5'-羥基-1',6-二甲基-2'-氧代-[2,4'-雙吡啶]-4-甲酸甲酯(INT-B11-8)。此材料在不經進一步純化的情況下用於下一反應中。LCMS: m/z (ESI), [M+H] +=275.05。 步驟9. 1',6-二甲基-5'-{[(4 R)-4-甲基-5-[(2-硝基苯基)氨基]戊基]氧基}-2'-氧代-[2,4'-雙吡啶]-4-甲酸甲酯(INT-B11-9)的製備。將5'-羥基-1',6-二甲基-2'-氧代-[2,4'-雙吡啶]-4-甲酸甲酯(282 mg,1.03 mmol,1當量)、(4 R)-4-甲基-5-[(2-硝基苯基)氨基]戊-1-醇(247 mg,1.03 mmol,1當量)和2-(三丁基-l^[5]-膦亞基)乙腈(1.00 g,4.14 mmol,4.00當量)於甲苯(10 mL)中的混合物在氮氣氣氛下在100℃下攪拌2 h。將混合物冷卻到室溫。將反應用水(100 mL)淬滅,並且將混合物用二氯甲烷(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上使用石油醚/乙酸乙酯(1/1)洗脫進行純化,以得到呈黃色油的400 mg 1',6-二甲基-5'-{[(4R)-4-甲基-5-[(2-硝基苯基)氨基]戊基]氧基}-2'-氧代-[2,4'-雙吡啶]-4-甲酸甲酯(INT-B11-9)(77%)。LCMS: m/z (ESI), [M+H] +=495.10。 步驟10. 5'-{[(4R)-5-[(2-氨基苯基)氨基]-4-甲基戊基]氧基}-1',6-二甲基-2'-氧代-[2,4'-雙吡啶]-4-甲酸甲酯(INT-B11-10)的製備。在0℃下向1',6-二甲基-5'-{[(4R)-4-甲基-5-[(2-硝基苯基)氨基]戊基]氧基}-2'-氧代-[2,4'-雙吡啶]-4-甲酸甲酯(400 mg,0.80 mmol,1當量)和雷尼鎳(34 mg,0.40 mmol,0.50當量)於甲醇(10 mL)中的攪拌混合物中添加水合肼(77 mg,2.42 mmol,3.00當量)。將混合物在氮氣氣氛下在0℃下攪拌2 h。將混合物過濾,並且將濾餅用甲醇(3×10 mL)洗滌。將所得溶液在減壓下濃縮。將殘餘物在prep-TLC上使用石油醚/乙酸乙酯(1/1)洗脫進行純化,以得到呈黃色固體的350 mg 5'-{[(4 R)-5-[(2-氨基苯基)氨基]-4-甲基戊基]氧基}-1',6-二甲基-2'-氧代-[2,4'-雙吡啶]-4-甲酸甲酯(INT-B11-10)(93%)。LCMS: m/z (ESI), [M+H] +=465.20。 步驟11. 5'-{[(4 R)-4-[(2-氨基-1,3-苯並二氮唑-1-基)甲基]戊基]氧基}-1',6-二甲基-2'-氧代-[2,4'-雙吡啶]-4-甲酸甲酯(INT-B11-11)的製備。將5'-{[(4 R)-5-[(2-氨基苯基)氨基]-4-甲基戊基]氧基}-1',6-二甲基-2'-氧代-[2,4'-雙吡啶]-4-甲酸甲酯(400 mg,0.86 mmol,1當量)和BrCN(182 mg,1.722 mmol,2.00當量)於乙醇(10 mL)中的混合物在氮氣氣氛下在室溫下攪拌過夜。將反應用水(50 mL)淬滅,並且將混合物用乙酸乙酯(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在prep-TLC上使用二氯甲烷/甲醇(20/1)洗脫進行純化,以得到呈黃色油的350 mg 5'-{[(4 R)-4-[(2-氨基-1,3-苯並二氮唑-1-基)甲基]戊基]氧基}-1',6-二甲基-2'-氧代-[2,4'-雙吡啶]-4-甲酸甲酯(INT-B11-11)(83%)。LCMS: m/z (ESI), [M+H] +=490.10。 步驟12. 5'-{[(4 R)-4-[(2-氨基-1,3-苯並二氮唑-1-基)甲基]戊基]氧基}-1',6-二甲基-2'-氧代-[2,4'-雙吡啶]-4-羧酸(INT-B11-12)的製備。將5'-{[(4 R)-4-[(2-氨基-1,3-苯並二氮唑-1-基)甲基]戊基]氧基}-1',6-二甲基-2'-氧代-[2,4'-雙吡啶]-4-甲酸甲酯(300 mg,0.61 mmol,1當量)和LiOH .H 2O(44 mg,1.83 mmol,3當量)於四氫呋喃/水(4 mL/1 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將所得混合物在減壓下濃縮。將殘餘物通過C18 FLASH色譜法,使用含有0.1% NH 3 .H 2O和乙腈的水作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃色固體的200 mg 5'-{[(4 R)-4-[(2-氨基-1,3-苯並二氮唑-1-基)甲基]戊基]氧基}-1',6-二甲基-2'-氧代-[2,4'-雙吡啶]-4-羧酸(INT-B11-12)(68%)。LCMS: m/z (ESI), [M+H] += 476.10。 步驟13. (12 R)-5,12,27-三甲基-8-氧雜-5,14,21,23,28-五氮雜五環[23.3.1.0^{2,7}.0^{14,22}.0^{15,20}]二十九-1(29),2,6,15,17,19,21,25,27-壬烯-4,24-二酮(實施例B11)的製備。將5'-{[(4 R)-4-[(2-氨基-1,3-苯並二氮唑-1-基)甲基]戊基]氧基}-1',6-二甲基-2'-氧代-[2,4'-雙吡啶]-4-羧酸(400 mg,0.84 mmol,1當量)、 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(479 mg,1.26 mmol,1.5當量)和 N, N-二異丙基乙胺(217 mg,1.68 mmol,2當量)於1,4-二噁烷(10 mL)中的混合物在氮氣氣氛下在室溫下攪拌過夜。將反應用水(100 mL)淬滅,並且將混合物用二氯甲烷(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-HPLC用XBridge Shield RP18 OBD柱,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3 .H 2O的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的186 mg (12 R)-5,12,27-三甲基-8-氧雜-5,14,21,23,28-五氮雜五環[23.3.1.0^{2,7}.0^{14,22}.0^{15,20}]二十九-1(29),2,6,15,17,19,21,25,27-壬烯-4,24-二酮(實施例B11) (48%)。LCMS: m/z (ESI), [M+H] +=458.25。 1H NMR (DMSO-d 6, 400 MHz) δ 0.93 (3H, d), 1.47-1.60 (1H, m), 1.66-1.80 (1H, m), 1.96-2.20 (2H, m), 2.25-2.37 (1H, m), 2.63 (3H, s), 3.48 (3H, s), 3.93-4.08 (3H, m), 4.18 (1H, dd), 7.12 (1H, s), 7.19-7.34 (2H, m), 7.53-7.61 (3H, m), 7.83 (1H, d), 8.92 (1H, d), 12.80 (1H, s)。 實施例B12 (11 R)-18-[2-(二甲基氨基)乙基]-5,11,30-三甲基-7-氧雜-4,5,13,18,24,26,31-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}. 0^{14,23}.0^{15,20}]三十二-1(31),2(6),3,14,20,22,24, 28(32),29-壬-27-酮 步驟1. 2,2,2-三氟-1-(5-{[(2 R)-5-羥基-2-甲基戊基]氨基}-6-硝基-3,4-二氫-1H-異喹啉-2-基)乙酮(INT-B12-1)的製備。將2,2,2-三氟-1-(5-氟-6-硝基-3,4-二氫-1H-異喹啉-2-基)乙酮(INT-A102-3,15 g,51.34 mmol,1當量)、 N,N-二異丙基乙胺(19.9 g,154.01 mmol,3當量)和(4 R)-5-氨基-4-甲基戊-1-醇(M3-6,6016 mg,51.34 mmol,1當量)於乙腈(150 mL)中的混合物在80℃下攪拌2 h。將混合物冷卻到室溫。將反應用水(500 mL)淬滅,並且將混合物用二氯甲烷(3×500 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠柱使用石油醚/乙酸乙酯(1/1)洗脫來進行純化,以得到呈黃色固體的10 g 2,2,2-三氟-1-(5-{[(2R)-5-羥基-2-甲基戊基]氨基}-6-硝基-3,4-二氫-1H-異喹啉-2-基)乙酮(INT-B12-1)(50%)。LCMS: m/z (ESI), [M+H] +=390.10。 步驟2. (4 R)-4-甲基-5-[(6-硝基-1,2,3,4-四氫異喹啉-5-基)氨基]戊-1-醇(INT-B12-2)的製備。將2,2,2-三氟-1-(5-{[(2 R)-5-羥基-2-甲基戊基]氨基}-6-硝基-3,4-二氫-1H-異喹啉-2-基)乙酮(10 g,25.68 mmol,1當量)和K 2CO 3(10.6g,77.05 mmol,3當量)於甲醇(80 mL)和H 2O(40 mL)中的混合物在室溫下攪拌5 h。將反應用水(500 mL)處理,並且將混合物用二氯甲烷(3×500 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上使用石油醚/乙酸乙酯(1/1)洗脫來進行純化,以得到呈黃色固體的7 g (4 R)-4-甲基-5-[(6-硝基-1,2,3,4-四氫異喹啉-5-基)氨基]戊-1-醇(INT-B12-2)(93%)。LCMS: m/z (ESI), [M+H] +=294.15。 1H NMR (DMSO-d 6,400 MHz) δ 0.83-0.88 (3 H, m), 1.07-1.11 (1 H, m), 1.22-1.51 (3 H, m), 1.57-1.64 (1 H, m), 2.59 (2 H, t), 2.88-2.92 (3 H, m), 3.10-3.20 (1 H, m), 3.30-3.33 (2 H, m), 3.87 (2H, s), 4.37 (1 H, s), 6.56 (1 H, d), 6.69 (1 H, t), 7.26-7.37 (1 H, m), 7.70 (1 H, d)。 步驟3. 5-{[(2 R)-5-羥基-2-甲基戊基]氨基}-6-硝基-3,4-二氫-1H-異喹啉-2-甲酸叔丁酯(INT-B12-3)的製備。將(4 R)-4-甲基-5-[(6-硝基-1,2,3,4-四氫異喹啉-5-基)氨基]戊-1-醇(8 g,27.27 mmol,1當量)、三甲胺(5.5 g,54.54 mmol,2當量)和二碳酸二叔丁酯(7.1 g,32.72 mmol,1.2當量)於二氯甲烷(100 mL)中的混合物在室溫下攪拌2 h。將反應用水(300 mL)淬滅,並且將混合物用二氯甲烷(3×300 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上使用石油醚/乙酸乙酯(1/1)洗脫來進行純化,以得到呈黃色油的6.5 g 5-{[(2 R)-5-羥基-2-甲基戊基]氨基}-6-硝基-3,4-二氫-1H-異喹啉-2-甲酸叔丁酯(INT-B12-3)(60%)。LCMS: m/z (ESI), [M+H] +=394.20。 1H NMR (DMSO-d 6400 MHz) δ 0.84 (3H, d), 1.03-1.17 (1H, m), 1.22-1.45 (2H, m), 1.45 (9H, s), 1.61 (1H, q), 2.75 (2H, t), 2.93-2.97 (1H, m), 3.08-3.12 (1H, m), 3.30-3.33 (2H, m), 3.51 (2H, t), 4.36 (1H, t), 4.52 (2H, s), 6.73 (1H, d), 6.83 (1H, s), 7.80 (1H, d) 步驟4. 5-{[(2 R)-5-({4-[4-(甲氧基羰基)-6-甲基吡啶-2-基]-2-甲基吡唑-3-基}氧基)-2-甲基戊基]氨基}-6-硝基-3,4-二氫-1H-異喹啉-2-甲酸叔丁酯(INT-B12-4)的製備。在氮氣氣氛下在0℃下向5-{[(2 R)-5-羥基-2-甲基戊基]氨基}-6-硝基-3,4-二氫-1H-異喹啉-2-甲酸叔丁酯(1 g,2.54 mmol,1當量)和三苯基膦(2 g,7.62 mmol,3當量)於四氫呋喃(20 mL)中的攪拌混合物中添加2-(5-羥基-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(M1,691 mg,2.79 mmol,1.1當量)和偶氮二甲酸二異丙酯(1.5 g,7.62 mmol,3當量)。將混合物在室溫下攪拌2 h。將反應用水(100 mL)淬滅,並且將混合物用二氯甲烷(3×80 mL)萃取。將合併的有機層用鹽水(3×80 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上使用石油醚/乙酸乙酯(2/3)洗脫來進行純化,以得到呈淡黃色固體的900 mg 5-{[(2 R)-5-({4-[4-(甲氧基羰基)-6-甲基吡啶-2-基]-2-甲基吡唑-3-基}氧基)-2-甲基戊基]氨基}-6-硝基-3,4-二氫-1H-異喹啉-2-甲酸叔丁酯(INT-B12-4)(57%)。LCMS: m/z (ESI), [M+H] +=623.30。 步驟5. 6-氨基-5-{[(2 R)-5-({4-[4-(甲氧基羰基)-6-甲基吡啶-2-基]-2-甲基吡唑-3-基}氧基)-2-甲基戊基]氨基}-3,4-二氫-1H-異喹啉-2-甲酸叔丁酯(INT-B12-5)的製備。將5-{[(2 R)-5-({4-[4-(甲氧基羰基)-6-甲基吡啶-2-基]-2-甲基吡唑-3-基}氧基)-2-甲基戊基]氨基}-6-硝基-3,4-二氫-1H-異喹啉-2-甲酸叔丁酯(500 mg,0.80 mmol,1當量)和Pd/C (10%,30 mg)於甲醇(10 mL)中的混合物在氫氣氣氛下攪拌1 h。將所得混合物過濾,並且將濾餅用甲醇(3×50 mL)洗滌。將所得溶液在減壓下濃縮。將殘餘物通過prep-TLC使用二氯甲烷/甲醇(10/1)洗脫來進行純化,以得到呈淡黃色固體的360 mg 6-氨基-5-{[(2 R)-5-({4-[4-(甲氧基羰基)-6-甲基吡啶-2-基]-2-甲基吡唑-3-基}氧基)-2-甲基戊基]氨基}-3,4-二氫-1H-異喹啉-2-甲酸叔丁酯(INT-B12-5) (76%)。LCMS: m/z (ESI), [M+H] +=593.40。 步驟6. 2-(5-{[(4 R)-4-{[2-氨基-7-(叔丁氧基羰基)-6H,8H,9H-咪唑並[4,5-f]異喹啉-1-基]甲基}戊基]氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(INT-B12-6)的製備。將6-氨基-5-{[(2 R)-5-({4-[4-(甲氧基羰基)-6-甲基吡啶-2-基]-2-甲基吡唑-3-基}氧基)-2-甲基戊基]氨基}-3,4-二氫-1H-異喹啉-2-甲酸叔丁酯(350 mg,0.59 mmol,1當量)和BrCN(75 mg,0.71 mmol,1.2當量)於二氯甲烷(4 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將反應用水(60 mL)淬滅,並且將混合物用二氯甲烷(3×60 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-TLC使用二氯甲烷/甲醇(11/1)洗脫進行純化,以得到呈黃色油的300 mg 2-(5-{[(4 R)-4-{[2-氨基-7-(叔丁氧基羰基)-6H,8H,9H-咪唑並[4,5-f]異喹啉-1-基]甲基}戊基]氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(INT-B12-6)(82%)。LCMS: m/z (ESI), [M+H] +=618.35。 步驟7. 2-(5-{[(4 R)-4-{[2-氨基-7-(叔丁氧基羰基)-6H,8H,9H-咪唑並[4,5-f]異喹啉-1-基]甲基}戊基]氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-羧酸(INT-B12-7)的製備。將2-(5-{[(4 R)-4-{[2-氨基-7-(叔丁氧基羰基)-6H,8H,9H-咪唑並[4,5-f]異喹啉-1-基]甲基}戊基]氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(290 mg,0.47 mmol,1當量)和LiOH(30 mg,0.70 mmol,1.5當量)於四氫呋喃/H 2O(4 mL/1 mL)中的混合物在室溫下攪拌2 h。將混合物在真空中濃縮。將殘餘物通過C18快速色譜法使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈紅色固體的900 mg 2-(5-{[(4 R)-4-{[2-氨基-7-(叔丁氧基羰基)-6H,8H,9H-咪唑並[4,5-f]異喹啉-1-基]甲基}戊基]氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-羧酸(INT-B12-7)(71%)。LCMS: m/z (ESI), [M+H] +=604.30。 步驟8. (11 R)-5,11,30-三甲基-27-氧代-7-氧雜-4,5,13, 18,24,26,31-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}.0^{14, 23}.0^{15,20}]三十二-1(31),2(6),3,14,20,22,24,28(32),29-壬烯-18-甲酸叔丁酯(INT-B12-8)的製備。將2-(5-{[(4 R)-4-{[2-氨基-7-(叔丁氧基羰基)-6H,8H,9H-咪唑並[4,5-f]異喹啉-1-基]甲基}戊基]氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-羧酸(200 mg,0.33 mmol,1當量)、 N,N-二異丙基乙胺(128 mg,0.99 mmol,3當量)和 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(252 mg,0.66 mmol,2當量)於二噁烷(5 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將反應用水(60 mL)淬滅,並且將混合物用二氯甲烷(3×60 mL)萃取。將合併的有機層用鹽水(3×40 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-TLC使用二氯甲烷/甲醇(15/1)洗脫進行純化,以得到呈黃色固體的160 mg (11 R)-5,11,30-三甲基-27-二氧代-7-二氧雜-4,5,13, 18,24,26,31-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}.0^{14, 23}.0^{15,20}]三十二-1(31),2(6),3,14,20,22,24,28(32),29-壬烯-18-甲酸叔丁酯(INT-B12-8)(82%)。LCMS: m/z (ESI), [M+H] +=586.30。 步驟9. (11R)-5,11,30-三甲基-7-氧雜-4,5,13,18,24,26, 31-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15, 20}]三十二-1(31),2(6),3,14,20,22,24,28(32),29-壬-27-酮(INT-B12-9)的製備。在室溫下向(11 R)-5,11,30-三甲基-27-氧代-7-氧雜-4,5,13,18,24,26,31-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]三十二-1(31),2(6),3,14,20,22,24,28(32),29-壬烯-18-甲酸叔丁酯(120 mg,0.21 mmol,1當量)於1,4-二噁烷(0.5 mL)中的攪拌混合物中添加HCl溶液(3 mL,1,4-二噁烷中4 M)。將混合物在室溫下攪拌2 h。將所得混合物用飽和NaHCO 3(水溶液)(100 mL)處理,並且用二氯甲烷(3×100 mL)萃取。將合併的有機層用鹽水(3×70 mL)洗滌,經無水Na 2SO 4乾燥,並且在減壓下濃縮,以得到呈黃色固體的99 mg (11 R)-5,11,30-三甲基-7-氧雜-4,5,13,18,24,26,31-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]三十二-1(31),2(6),3,14,20,22,24,28(32),29-壬-27-酮(INT-B12-9)(99%)。此材料在不經純化的情況下用於下一步驟中。LCMS: m/z (ESI), [M+H] +=486.30。 步驟10. (11 R)-18-[2-(二甲基氨基)乙基]-5,11,30-三甲基-7-氧雜-4,5,13,18,24,26,31-七氮雜六環[26.3.1.0^{2,6}. 0^{13,25}.0^{14,23}.0^{15,20}]三十二-1(31),2(6),3,14, 20,22,24,28(32),29-壬-27-酮(實施例B12).在室溫下向(11 R)-5,11,30-三甲基-7-氧雜-4,5,13,18,24,26,31-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]三十二-1(31),2(6),3,14,20,22,24,28(32),29-壬-27-酮(99 mg,0.20 mmol,1當量)和三甲胺(62 mg,0.61 mmol,3當量)於 N,N-二甲基甲醯胺(5 mL)中的攪拌混合物中添加(2-溴乙基)二甲胺(56 mg,0.37 mmol,1.8當量)和KI(10 mg,0.06 mmol,0.3當量)。將所得混合物在氮氣氣氛下在80℃下攪拌2 h。將混合物冷卻到室溫。將反應用水(50 mL)淬滅,並且將混合物用二氯甲烷(3×60 mL)萃取。將合併的有機層用鹽水(3×40 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-TLC使用二氯甲烷/甲醇(20/1)洗脫進行純化。將產物通過prep-HPLC用XBridge Prep OBD C18柱,使用含有10 mmol/L NH 4HCO 3的水和乙腈作為流動相進行進一步純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃色固體的9.3 mg (11 R)-18-[2-(二甲基氨基)乙基]-5,11,30-三甲基-7-氧雜-4,5,13,18,24,26,31-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]三十二-1(31),2(6),3,14,20,22,24,28(32),29-壬-27-酮(實施例B12) (8%)。LCMS: m/z (ESI), [M+H] +=557.30。 1H NMR (DMSO- d 6, 400 MHz) δ 0.85 (3H, d), 1.41-1.53 (1H, m), 1.77-1.92 (1H, m), 1.95-2.08 (1H, m), 2.19 (6H, s), 2.21-2.29 (1H, m), 2.49-2.61 (5H, m), 2.72-2.86 (2H, m), 2.88-3.02 (1H, m), 3.10-3.27 (2H, m), 3.60-3.70 (2H, m), 3.73 (3H, s), 3.96-4.02 (1H, m), 4.22-4.28 (4H, m), 6.95 (1H, d), 7.39 (1H, d), 7.57 (1H, s), 7.93 (1H, s), 8.36 (1H, d), 12.75 (1H, s)。 實施例B13 ( R)-11,26,7-三甲基-57-(氧雜環丁-3-基)-56,57,58,59-四氫-11H,51H-11-氧雜-4-氮雜-5(2,1)-咪唑並[4,5-f]異喹啉-2(2,4)-吡啶-1(4,5)-吡唑環十一烷-3-酮 步驟1. (11 R)-11,26,7-三甲基-57-(氧雜環丁-3-基)-56, 57,58,59-四氫-11H,51H-11-氧雜-4-氮雜-5(2,1)-咪唑並[4,5-f]異喹啉-2(2,4)-吡啶-1(4,5)-吡唑環十一烷-3-酮(實施例B13)的製備。將(11 R)-5,11,30-三甲基-7-氧雜-4,5,13,18, 24,26,31-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}. 0^{15,20}]三十二-1(31),2(6),3,14,20,22,24,28(32),29-壬-27-酮(INT-B12-9,40 mg,0.08 mmol,1當量)於二氯甲烷(2 mL)中的混合物用3-氧雜環丁烷酮(12 mg,0.16 mmol,2當量)和三甲胺(17 mg,0.16 mmol,2當量)在氮氣氣氛下在室溫下處理0.5 h,隨後在0℃下添加NaBH(AcO) 3(52 mg,0.25 mmol,3當量)。將所得混合物在氮氣氣氛下在室溫下攪拌過夜。將反應用水(20 mL)淬滅,並且將混合物用二氯甲烷(3×20 mL)萃取。將合併的有機層用鹽水(3×20 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-HPLC用XBridge Shield RP18 OBD柱,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3 .H 2O的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的10.5 mg (11 R)-5,11,30-三甲基-18-(氧雜環丁-3-基)-7-氧雜-4,5,13,18,24,26,31-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]三十二-1(31),2(6),3,14,20,22,24,28(32),29-壬-27-酮(實施例B13) (22%)。LCMS: m/z (ESI), [M+H] +=542.20。 1H NMR (DMSO- d 6, 400 MHz) δ 0.86 (3H, d), 1.38-1.53 (1H, m), 1.79-1.93 (1H, m), 1.96-2.06 (1H, m), 2.16-2.28 (1H, m), 2.58 (3H, s), 2.58-2.73 (3H, m), 2.90-3.02 (1H, m), 3.15-3.26 (1H, m), 3.45-3.64 (3H, m), 3.73 (3H, s), 3.97-4.01 (1H, m), 4.14-4.38 (3H, m), 4.56 (2H, t), 4.65 (2H, t), 6.96 (1H, d), 7.41 (1H, d), 7.58 (1H, s), 7.93 (1H, s), 8.39 (1H, s), 12.78 (1H, s)。 實施例B14 30-甲基-11-氧雜-8,9,17,24,26,31-六氮雜六環[26.3.1.0^{2,10}.0^{3,8}.0^{17,25}.0^{18,23}]三十二-1(31),2,4,6,9,18,20,22,24,28(32),29-十一烷-27-酮 步驟1. 1-(5-溴戊基)-1,3-苯並二氮唑-2-胺(INT-B14-1)的製備。將2-氨基苯並咪唑(4 g,30.04 mmol,1當量)、K 2CO 3(12.46 g,90.12 mmol,3當量)和1,5-二溴戊烷(27.63 g,120.16 mmol,4當量)於乙腈(80 mL)中的混合物在60℃下攪拌5 h。將混合物冷卻到室溫。將反應用水(500 mL)淬滅,並且將混合物用乙酸乙酯(3×500 mL)萃取。將合併的有機層用鹽水(3×500 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上使用二氯甲烷/甲醇(10/1)洗脫來進行純化,以得到呈棕色固體的4 g 1-(5-溴戊基)-1,3-苯並二氮唑-2-胺(INT-B14-1)(47%)。LCMS: m/z (ESI), [M+H] +=283.95。 步驟2. 1-[5-({3-溴吡唑並[1,5-a]吡啶-2-基}氧基)戊基]-1,3-苯並二氮唑-2-胺(INT-B14-2)的製備。將1-(5-溴戊基)-1,3-苯並二氮唑-2-胺(3.97 g,14.08 mmol,1.5當量)、3-溴吡唑並[1,5-a]吡啶-2-醇(2 g,9.38 mmol,1.00當量)和K 2CO 3(2.59 g,18.77 mmol,2當量)於乙腈(80 mL)中的混合物在60℃下攪拌2 h。將混合物冷卻到室溫。將反應用水(500 mL)淬滅,並且將混合物用乙酸乙酯(3×500 mL)萃取。將合併的有機層用鹽水(3×500 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過C18 FLASH色譜法使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈棕色固體的2.3 g 1-[5-({3-溴吡唑並[1,5-a]吡啶-2-基}氧基)戊基]-1,3-苯並二氮唑-2-胺(INT-B14-2)(39%)。LCMS: m/z (ESI), [M+H] +=414.20。 1H NMR (DMSO-d 6 , 400 MHz) δ 1.40-1.52 (2H, m), 1.63.175 (2H, m), 1.76-1.88 (2H, m), 3.92-4.03 (2H, m), 4.30 (2H, t), 6.37 (2H, s), 6.79-6.87 (2H, m), 6.88-6.94 (1H, m), 7.12 (2H, t), 7.26-7.39 (2H, m),8.54 (1H, d)。 步驟3. 6-氯-4-(甲氧基羰基)吡啶-2-基硼酸)(M1-1)的製備。將雙(頻哪醇)二硼(1631.71 mg,6.426 mmol,1.5當量)於甲基叔丁基醚(20 mL)中的混合物溫熱到80℃,並且在氮氣氣氛下攪拌0.5 h,然後冷卻到室溫。向此混合物中添加雙(1,5-環辛二烯)二-μ-甲氧基二銥(I)(283.95 mg,0.428 mmol,0.1當量)和4-叔丁基-2-(4-叔丁基吡啶-2-基)吡啶(229.95 mg,0.857 mmol,0.2當量)。將混合物在25℃下攪拌0.5 h,然後添加2-氯吡啶-4-甲酸甲酯(735 mg,4.284 mmol,1當量)。將反應混合物在80℃下繼續攪拌16 h。將混合物冷卻到室溫,並且在減壓下濃縮,以得到呈棕色油的850 mg(粗製物)6-氯-4-(甲氧基羰基)吡啶-2-基硼酸(M1-1)。此材料在不經進一步純化的情況下用於下一反應中。LCMS: m/z (ESI), [M+H] +=216.05。 步驟4. 2-(2-{[5-(2-氨基-1,3-苯並二氮唑-1-基)戊基]氧基}吡唑並[1,5-a]吡啶-3-基)-6-氯吡啶-4-甲酸甲酯(INT-B14-3).將新鮮製備的6-氯-4-(甲氧基羰基)吡啶-2-基硼酸(850 mg粗製物,約2.5當量)、1-[5-({3-溴吡唑並[1,5-a]吡啶-2-基}氧基)戊基]-1,3-苯並二氮唑-2-胺(654 mg,1.58 mmol,1.00當量)、Cs 2CO 3(1543 mg,4.73 mmol,3當量)、Pd 2(dba) 3(144 mg,0.15 mmol,0.1當量)和雙(金剛烷-1-基)(丁基)膦(57 mg,0.16 mmol,0.1當量)於H 2O(4 mL)和甲苯(20 mL)中的混合物在氮氣氣氛下在80℃下攪拌2 h。將混合物冷卻到室溫。將反應用水(200 mL)淬滅,並且將混合物用乙酸乙酯(3×200 mL)萃取。將合併的有機層用鹽水(3×200 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上使用石油醚/乙酸乙酯(1/3)洗脫進行純化,以得到呈棕色固體的250 mg 2-(2-{[5-(2-氨基-1,3-苯並二氮唑-1-基)戊基]氧基}吡唑並[1,5-a]吡啶-3-基)-6-氯吡啶-4-甲酸甲酯(INT-B14-3)(31%)。LCMS: m/z (ESI), [M+H] +=505.15 步驟5. 2-(2-{[5-(2-氨基-1,3-苯並二氮唑-1-基)戊基]氧基}吡唑並[1,5-a]吡啶-3-基)-6-甲基吡啶-4-甲酸甲酯(INT-B14-4)的製備。將2-(2-{[5-(2-氨基-1,3-苯並二氮唑-1-基)戊基]氧基}吡唑並[1,5-a]吡啶-3-基)-6-氯吡啶-4-甲酸甲酯(140 mg,0.27 mmol,1當量)、三甲基-1,3,5,2,4,6-三氧雜三硼環己烷(69 mg,0.55 mmol,2當量)、Pd(dppf)Cl 2 .CH 2Cl 2(3 mg,0.01 mmol,0.2當量)、K 2CO 3(76 mg,0.55 mmol,2當量)和PCy 3HBF 4(30 mg,0.08 mmol,0.3當量)於1,4-二噁烷(10 mL)中的混合物在氮氣氣氛下在100℃下攪拌2 h。將混合物冷卻到室溫。將反應用水(200 mL)淬滅,並且將混合物用乙酸乙酯(3×200 mL)萃取。將合併的有機層用鹽水(3×200 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在Prep-TLC上使用二氯甲烷/甲醇(20/1)洗脫進行純化,以得到呈棕色固體的80 mg 2-(2-{[5-(2-氨基-1,3-苯並二氮唑-1-基)戊基]氧基}吡唑並[1,5-a]吡啶-3-基)-6-甲基吡啶-4-甲酸甲酯(INT-B14-4)(59%)。LCMS: m/z (ESI), [M+H] +=485.25。 步驟6. 2-(2-{[5-(2-氨基-1,3-苯並二氮唑-1-基)戊基]氧基}吡唑並[1,5-a]吡啶-3-基)-6-甲基吡啶-4-羧酸(INT-B14-5)的製備。將2-(2-{[5-(2-氨基-1,3-苯並二氮唑-1-基)戊基]氧基}吡唑並[1,5-a]吡啶-3-基)-6-甲基吡啶-4-甲酸甲酯(70 mg,0.14 mmol,1當量)和LiOH .H 2O(18 mg,0.43 mmol,3當量)於四氫呋喃(5 mL)和水(1.25 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將所得混合物在減壓下濃縮。將殘餘物通過C18 FLASH使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的30 mg 2-(2-{[5-(2-氨基-1,3-苯並二氮唑-1-基)戊基]氧基}吡唑並[1,5-a]吡啶-3-基)-6-甲基吡啶-4-羧酸(INT-B14-5)(44%)。LCMS: m/z (ESI), [M+H] +=471.30。 步驟7. 30-甲基-11-氧雜-8,9,17,24,26,31-六氮雜六環[26.3.1.0^{2,10}.0^{3,8}.0^{17,25}.0^{18,23}]三十二-1(31),2,4,6,9,18,20,22,24,28(32),29-十一烷-27-酮(實施例B14)的製備。將2-(2-{[5-(2-氨基-1,3-苯並二氮唑-1-基)戊基]氧基}吡唑並[1,5-a]吡啶-3-基)-6-甲基吡啶-4-羧酸(25 mg,0.05 mmol,1當量)、 N,N-二異丙基乙胺(20 mg,0.15 mmol,3當量)和 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(30 mg,0.08 mmol,1.5當量)於1,4-二噁烷(10 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將所得混合物在減壓下濃縮。將殘餘物通過prep-HPLC用XBridge Prep OBD C18柱,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3 .H 2O的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的9.4 mg 30-甲基-11-氧雜-8,9,17,24,26,31-六氮雜六環[26.3.1.0^{2,10}.0^{3,8}.0^{17,25}.0^{18,23}]三十二-1(31),2,4,6,9,18,20,22,24,28(32),29-十一烷-27-酮(實施例B14)(39%)。LCMS: m/z (ESI), [M+H] +=453.20。 1H NMR (DMSO-d 6 , 400 MHz) δ 1.91-2.12 (6H, m), 2.64 (3H, s), 4.23 (2H, t), 4.48 (2H, t), 6.91 (1H, t), 7.20-7.33 (2H, m), 7.42 (1H, t), 7.49 (1H, s), 7.55 (2H, d), 8.57-8.67 (2H, m), 9.04 (1H, s), 12.66 (1H, s)。 實施例B15 30-氯-11-氧雜-8,9,17,24,26,31-六氮雜六環[26.3.1.0^{2,10}.0^{3,8}.0^{17,25}. 0^{18,23}]三十二-1(31),2,4,6,9,18,20,22,24,28(32),29-十一烷-27-酮 步驟1. 2-(2-{[5-(2-氨基-1,3-苯並二氮唑-1-基)戊基]氧基}吡唑並[1,5-a]吡啶-3-基)-6-氯吡啶-4-羧酸(INT-B15-1)的製備。將2-(2-{[5-(2-氨基-1,3-苯並二氮唑-1-基)戊基]氧基}吡唑並[1,5-a]吡啶-3-基)-6-氯吡啶-4-甲酸甲酯(330 mg,0.65 mmol,1當量)和LiOH .H2O(41 mg,0.98 mmol,1.5當量)於四氫呋喃(10 mL)和H 2O(2.5 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將所得混合物在減壓下濃縮。將殘餘物通過prep-HPLC用XBridge Shield RP18 OBD柱,使用含有10 mmol/L NH 4HCO 3的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈棕色固體的134 mg 2-(2-{[5-(2-氨基-1,3-苯並二氮唑-1-基)戊基]氧基}吡唑並[1,5-a]吡啶-3-基)-6-氯吡啶-4-羧酸(INT-B15-1)(41%)。LCMS: m/z (ESI), [M+H] +=491.15。 步驟2. 30-氯-11-氧雜-8,9,17,24,26,31-六氮雜六環[26.3.1.0^{2,10}.0^{3,8}.0^{17,25}. 0^{18,23}]三十二-1(31),2,4,6,9,18,20,22,24,28(32),29-十一烷-27-酮(實施例B15)的製備。將2-(2-{[5-(2-氨基-1,3-苯並二氮唑-1-基)戊基]氧基}吡唑並[1,5-a]吡啶-3-基)-6-氯吡啶-4-羧酸(120 mg,0.24 mmol,1當量)、 N,N-二異丙基乙胺(94 mg,0.73 mmol,3當量)和 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(139 mg,0.36 mmol,1.5當量)於1,4-二噁烷(10 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將反應用水淬滅,並且將混合物用乙酸乙酯萃取。將合併的有機層在減壓下乾燥和濃縮。將殘餘物通過prep-HPLC用XBridge Prep OBD C18柱,使用含有10 mmol/L NH 4HCO 3的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃色固體的3.9 mg 30-氯-11-氧雜-8,9,17,24,26,31-六氮雜六環[26.3.1.0^{2,10}.0^{3,8}. 0^{17,25}.0^{18,23}]三十二-1(31),2,4,6,9,18,20,22,25, 28(32),29-十一烷-27-酮(實施例B15)(3%)。LCMS: m/z (ESI), [M+H] +=473.10。 1H NMR (DMSO-d 6 , 400 MHz) δ 1.93-2.13 (6H, m), 4.24 (2H, t), 4.49 (2H, t), 6.96-7.02 (1H, m), 7.22-7.35 (2H, m), 7.48-7.61 (4H, m), 8.44 (1H, d), 8.67 (1H, d), 9.17 (1H, d), 12.76 (1H, s)。 實施例B16 5,30-二甲基-7-氧雜-4,5,13,18,24,26,31-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]三十二-1(31),2(6),3,14,20,22,24,28(32),29-壬-27-酮 步驟1. 5-((5-((4-(4-(甲氧基羰基)-6-甲基吡啶-2-基)-1-甲基-1H-吡唑-5-基)氧基)戊基)氨基)-6-硝基-3,4-二氫異喹啉-2(1H)-甲酸叔丁酯(INT-B16-1)的製備。在氮氣氣氛下在0℃下向5-[(5-羥戊基)氨基]-6-硝基-3,4-二氫-1H-異喹啉-2-甲酸叔丁酯(1.6 g,0.79 mmol,1當量)、三苯基膦(3 g,2.37 mmol,3當量)和2-(5-羥基-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(1 g,0.79 mmol,1當量)於四氫呋喃(50 mL)中的攪拌混合物中添加偶氮二甲酸二異丙酯(2.5 g,1.19 mmol,3當量)。將所得混合物在氮氣氣氛下在室溫下攪拌3 h。將反應用水(50 mL)淬滅,並且將混合物用二氯甲烷(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法使用石油醚/乙酸乙酯(1/1)洗脫來進行純化,以得到呈黃色油的900 mg 5-{[5-({4-[4-(甲氧基羰基)-6-甲基吡啶-2-基]-2-甲基吡唑-3-基}氧基)戊基]氨基}-6-硝基-3,4-二氫-1H-異喹啉-2-甲酸叔丁酯(INT-B16-1)(36%)。LCMS: m/z (ESI), [M+H] +=609.20。 步驟2. 6-氨基-5-((5-((4-(4-(甲氧基羰基)-6-甲基吡啶-2-基)-1-甲基-1H-吡唑-5-基)氧基)戊基)氨基)-3,4-二氫異喹啉-2(1H)-甲酸叔丁酯(INT-B16-2)的製備。將5-{[5-({4-[4-(甲氧基羰基)-6-甲基吡啶-2-基]-2-甲基吡唑-3-基}氧基)戊基]氨基}-6-硝基-3,4-二氫-1H-異喹啉-2-甲酸叔丁酯(900 mg,1.48 mmol,1當量)和Pd/C(15.73 mg,0.148 mmol,0.1當量)於甲醇(10 mL)中的混合物在氫氣氣氛下在室溫下攪拌1 h。將所得混合物過濾,並且將濾餅用甲醇(3×10 mL)洗滌。將所得溶液在減壓下濃縮,以得到呈黃色油的570 mg 6-氨基-5-((5-((4-(4-(甲氧基羰基)-6-甲基吡啶-2-基)-1-甲基-1H-吡唑-5-基)氧基)戊基)氨基)-3,4-二氫異喹啉-2(1H)-甲酸叔丁酯(INT-B16-2)(67%)。將粗產物混合物在不進行進一步純化的情況下直接用於下一步驟。LCMS: m/z (ESI), [M+H] +=579.25。 步驟3. 2-氨基-1-(5-((4-(4-(甲氧基羰基)-6-甲基吡啶-2-基)-1-甲基-1H-吡唑-5-基)氧基)戊基)-1,6,8,9-四氫-7H-咪唑並[4,5-f]異喹啉-7-甲酸叔丁酯(INT-B16-3)的製備。將6-氨基-5-{[5-({4-[4-(甲氧基羰基)-6-甲基吡啶-2-基]-2-甲基吡唑-3-基}氧基)戊基]氨基}-3,4-二氫-1H-異喹啉-2-甲酸叔丁酯(570 mg,0.99 mmol,1當量)和BrCN(115 mg,1.08 mmol,1.1當量)於甲醇(20 mL)中的混合物在氮氣氣氛下在室溫下攪拌1 h。將反應用水(20 mL)淬滅,並且將混合物用二氯甲烷(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法使用二氯甲烷/甲醇(20/1)洗脫進行純化,以得到呈灰白色固體的500 mg 2-[5-({5-[2-氨基-7-(叔丁氧基羰基)-6H,8H,9H-咪唑並[4,5-f]異喹啉-1-基]戊基}氧基)-1-甲基吡唑-4-基]-6-甲基吡啶-4-甲酸甲酯(INT-B16-3)(84%)。LCMS: m/z (ESI), [M+H] +=604.20。 步驟4. 2-(5-((5-(2-氨基-7-(叔丁氧基羰基)-6,7,8,9-四氫-1H-咪唑並[4,5-f]異喹啉-1-基)戊基)氧基)-1-甲基-1H-吡唑-4-基)-6-甲基異煙酸(INT-B16-4)的製備。將2-[5-({5-[2-氨基-7-(叔丁氧基羰基)-6H,8H,9H-咪唑並[4,5-f]異喹啉-1-基]戊基}氧基)-1-甲基吡唑-4-基]-6-甲基吡啶-4-甲酸甲酯(500 mg,0.828 mmol,1當量)和LiOH .H 2O(59.50 mg,2.484 mmol,3當量)於MeOH(10 mL)和H 2O(5 mL)中的混合物在氮氣氣氛下在室溫下攪拌1 h,並且在減壓下濃縮。將殘餘物通過prep-HPLC用XBridge Shield RP18 OBD柱,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3 .H 2O的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的150 mg 2-[5-({5-[2-氨基-7-(叔丁氧基羰基)-6H,8H,9H-咪唑並[4,5-f]異喹啉-1-基]戊基}氧基)-1-甲基吡唑-4-基]吡啶-4-羧酸(INT-B16-4)(31%)。LCMS: m/z (ESI), [M+H] +=590.25。 步驟5. 叔丁基5,30-二甲基-18-(叔丁氧基羰基)-7-氧雜-4,5,13,18,24,26,31-七氮雜六環[26.3.1.0^{2,6}. 0^{13,25}.0^{14,23}.0^{15,20}]三十二-1(31),2(6),3,14,20, 22,24,28(32),29-壬-27-酮(INT-B16-5)的製備。將2-[5-({5-[2-氨基-7-(叔丁氧基羰基)-6H,8H,9H-咪唑並[4,5-f]異喹啉-1-基]戊基}氧基)-1-甲基吡唑-4-基]-6-甲基吡啶-4-羧酸(500 mg,0.85 mmol,1當量)、 N,N-二異丙基乙胺(219 mg,1.7 mmol,2當量)和 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(644.8 mg,1.7 mmol,2當量)於二噁烷(20 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將反應用水(10 mL)淬滅,並且將混合物用二氯甲烷(3×10 mL)萃取。將合併的有機層用鹽水(3×10 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠柱色譜法使用二氯甲烷/甲醇(10/1)洗脫進行純化,以得到呈棕色固體的200 mg 5,30-二甲基-18-(叔丁氧基羰基)-7-氧雜-4,5,13,18,24,26,31-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}. 0^{14,23}.0^{15,20}]三十二-1(31),2(6),3,14,20,22,24, 28(32),29-壬-27-酮(INT-B16-5)(41%)。LCMS: m/z (ESI), [M+H] +=572.30。 1H NMR (CDCl 3, 400 MHz) δ 1.64 (17H, s), 1.95 (2H, d), 2.10-2.15 (2H, m), 2.68 (3H, s), 3.24 (2H, s), 3.80 (5H, d), 4.20-4.29 (2H, m), 4.38-4.47 (2H, m), 4.72 (2H, s), 7.03 (1H, d), 7.22 (1H, d), 7.69 (1H, s), 8.21 (1H, s), 8.49 (1H, s), 12.00 (1H, s)。 步驟6. 5,30-二甲基-7-氧雜-4,5,13,18,24,26,31-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]三十二-1(31),2(6),3,14,20,22,24,28(32),29-壬-27-酮(實施例B16)的製備。將1 1,2 6-二甲基-3-氧代-5 6,5 7,5 8,5 9-四氫-1 1H,5 1H-11-氧雜-4-氮雜-5(2,1)-咪唑並[4,5-f]異喹啉-2(2,4)-吡啶-1(4,5)-吡唑環十一烷-5 7-甲酸叔丁酯(30 mg,0.090 mmol,1當量)和HCl於1,4-二噁烷(5 mL)中的混合物在氮氣氣氛下在室溫下攪拌1 h。將混合物用飽和NaHCO 3(水溶液)調節到pH 7,並且將混合物用乙酸乙酯(3×10 mL)萃取。將合併的有機層用鹽水(3×10 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-HPLC用XBridge Shield RP18 OBD柱,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3 .H 2O的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的1.5 mg 5,30-二甲基-7-氧雜-4,5,13,18,24,26,31-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]三十二-1(31),2(6),3,14,20,22,24,28(32),29-壬-27-酮(實施例B16)(0.37%)。LCMS: m/z (ESI), [M+H] +=472.35。 1H NMR (DMSO, 400 MHz) δ 1.74-1.82 (2H, m), 1.96-2.10 (4H, m), 2.55 (3H, s), 3.02-3.16 (4H, m), 3.72 (3H, s), 3.95 (2H, s), 4.21 (2H, t), 4.35 (1H, d), 6.91 (1H, d), 7.33 (1H, d), 7.56 (1H, s), 7.93 (1H, s), 8.40 (1H, s)。 實施例B17 5,30-二甲基-18-(氧雜環丁-3-基)-7-氧雜-4,5,13,18,24,26, 31-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15, 20}]三十二-1(31),2(6),3,14,20,22,24,28(32),29-壬-27-酮 步驟1. 5,30-二甲基-18-(氧雜環丁-3-基)-7-氧雜-4,5,13,18,24,26,31-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}. 0^{14,23}.0^{15,20}]三十二-1(31),2(6),3,14,20,22,24, 28(32),29-壬-27-酮(實施例B17)的製備。將(25E)-5,30-二甲基-7-氧雜-4,5,13,18,24,26,31-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]三十二-1(31),2(6),3,14,20,22,25,28(32),29-壬-27-酮(實施例B16,30 mg,0.06 mmol,1當量)、3-氧雜環丁烷酮(9.17 mg,0.12 mmol,2當量)和三甲胺(12.88 mg,0.12 mmol,2當量)於二氯甲烷(5 mL)中的攪拌混合物在氮氣氣氛下在室溫下攪拌30分鐘。在室溫下向上述混合物中添加NaBH(AcO) 3(40.45 mg,0.18 mmol,3當量)。將所得混合物在氮氣氣氛下在室溫下攪拌過夜。將反應用水(10 mL)淬滅,並且將混合物用二氯甲烷(3×10 mL)萃取。將合併的有機層用鹽水(3×10 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-HPLC用XBridge Shield RP18 OBD柱,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3.H 2O的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃色固體的5.8 mg 5,30-二甲基-18-(氧雜環丁-3-基)-7-氧雜-4,5,13,18,24,26,31-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}.0 ^{14,23}.0^{15,20}]三十二-1(31),2(6),3,14,20,22,24,28(32),29-壬-27-酮(實施例B17)(17%)。LCMS: m/z (ESI), [M+H] +=528.20。 1H NMR (DMSO- d 6, 400 MHz) δ 1.71-1.85 (2H, m), 1.96-2.13 (4H, m), 2.56 (3H, s), 2.59-2.70 (2H, m), 3.22-3.30 (2H, m), 3.54 (2H, s), 3.57-3.65 (1H, m), 3.73 (3H, s), 4.21 (1H, t), 4.33 (2H, t), 4.55 (2H, t), 4.65 (2H, t), 6.96 (1H, d), 7.46 (1H, d), 7.57 (1H, s), 7.76 (1H, d), 7.98 (1H, s), 8.40 (1H, s)。 實施例B18 5,30-二甲基-17-(氧雜環丁-3-基)-7-氧雜-4,5,13,17,24,26, 31-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}. 0^{15,20}]三十二-1(31),2(6),3,14,20,22,24,28(32),29-壬-27-酮 步驟1. 2-(2-溴-5-氟苯基)乙胺(INT-B18-1)的製備。將2-(2-溴-5-氟苯基)乙腈(10 g,46.72 mmol,1當量)和BH 3-Me 2S(10.65 g,140.16 mmol,3當量)於四氫呋喃(100 mL)中的混合物在氮氣氣氛下在80℃下攪拌2 h。將混合物冷卻到室溫。將反應在0℃下用水(800 mL)淬滅,並且將混合物用二氯甲烷(3×200 mL)萃取。將合併的有機層用鹽水(3×200 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法使用二氯甲烷/甲醇(10/1)洗脫來進行純化,以得到呈灰白色固體的8 g 2-(2-溴-5-氟苯基)乙胺(實施例INT-B18-1)(79%)。LCMS: m/z (ESI), [M+H] += 220.10。 步驟2. N-[2-(2-溴-5-氟苯基)乙基]-2,2,2-三氟乙醯胺(INT-B18-2)的製備。將2-(2-溴-5-氟苯基)乙胺(5 g,23 mmol,1當量)、三氟乙酸酐(5.3 g,25 mmol,1.1當量)和三甲胺(4.64 g,45.86 mmol,2當量)於二氯甲烷(100 mL)中的混合物在氮氣氣氛下在0℃下攪拌2 h。將反應用水(200 mL)淬滅,並且將混合物用二氯甲烷(3×200 mL)萃取。將合併的有機層用鹽水(3×200 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠柱色譜法使用石油醚/乙酸乙酯(10/1)洗脫進行純化,以得到呈白色固體的4 g N-[2-(2-溴-5-氟苯基)乙基]-2,2,2-三氟乙醯胺(INT-B18-2)(56%)。 1H NMR (DMSO-d 6, 400 MHz) δ 2.90-2.96 (2H, m), 3.45-3.50 (2H, m), 7.04-7.26 (2H, m), 7.65 (1H, d), 9.55 (1H, s)。 步驟3. 1-(5-溴-8-氟-3,4-二氫-1H-異喹啉-2-基)-2,2,2-三氟乙酮(INT-B18-3)的製備。將 N-[2-(2-溴-5-氟苯基)乙基]-2,2,2-三氟乙醯胺(6.8 g,21.65 mmol,1當量)和多聚甲醛(28.6 g,64.95 mmol,3當量)於H 2SO 4(25 ml)和AcOH(30 ml)的混合物在氮氣氣氛下在0℃下攪拌過夜。將反應在0℃下用水(200 mL)淬滅,並且用NaOH調節到pH 7。將混合物用二氯甲烷(3×200 mL)萃取。將合併的有機層用鹽水(3×200 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠色譜法使用石油醚/乙酸乙酯(1/1)洗脫來進行純化,以得到呈灰白色固體的5 g 1-(5-溴-8-氟-3,4-二氫-1H-異喹啉-2-基)-2,2,2-三氟乙酮(71%)。 1H NMR (DMSO-d 6, 400 MHz) δ 2.85-2.92 (2H, m), 3.85-3.95 (2H, m), 4.77 (2H, d), 7.17 (1H, d), 7.63 (1H, d)。 步驟4. 1-(5-溴-8-氟-7-硝基-3,4-二氫-1H-異喹啉-2-基)-2,2,2-三氟乙酮(INT-B18-4)的製備。將1-(5-溴-8-氟-3,4-二氫-1H-異喹啉-2-基)-2,2,2-三氟乙酮(6.46 g,19.81 mmol,1當量)和KNO 3(2 g,19.81 mmol,1當量)於H 2SO 4(20 mL)中的混合物在氮氣氣氛下在0℃下攪拌0.5 h。將反應用水(200 mL)淬滅,並且用NaOH調節到pH 9。將混合物用二氯甲烷(3×200 mL)萃取。將合併的有機層用鹽水(3×200 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠柱色譜法使用石油醚/乙酸乙酯(3/1)洗脫來進行純化,以得到呈棕黃色固體的775 mg 1-(5-溴-8-氟-7-硝基-3,4-二氫-1H-異喹啉-2-基)-2,2,2-三氟乙酮(INT-B18-4)(12%)。 1H NMR (DMSO-d 6, 400 MHz) δ 2.95-3.02 (2H, m), 3.81-3.96 (2H, m), 4.75-4.90 (2H, m), 8.07 (1H, t)。 步驟5. 5-[(5-溴-7-硝基-1,2,3,4-四氫異喹啉-8-基)氨基]戊-1-醇(INT-B18-5)的製備。將1-(5-溴-8-氟-7-硝基-3,4-二氫-1H-異喹啉-2-基)-2,2,2-三氟乙酮(4.6 g,12.4 mmol,1當量)、K 2CO 3(5.14 g,37.2 mmol,3當量)和5-氨基戊醇(1.53 g,14.88 mmol,1.2當量)於乙腈(50 mL)中的混合物在氮氣氣氛下在60℃下攪拌2 h。將混合物冷卻到室溫。將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠柱色譜法使用二氯甲烷/甲醇(10/1)洗脫來進行純化,以得到呈黃色油的600 mg 5-[(5-溴-7-硝基-1,2,3,4-四氫異喹啉-8-基)氨基]戊-1-醇(INT-B18-5)(14%)。LCMS: m/z (ESI), [M+H] +=360.00。 步驟6. 5-溴-8-((5-羥戊基)氨基)-7-硝基-3,4-二氫異喹啉-2(1H)-甲酸叔丁酯(INT-B18-6)的製備。將5-[(5-溴-7-硝基-1,2,3,4-四氫異喹啉-8-基)氨基]戊-1-醇(650 mg,1.81 mmol,1當量)、二碳酸二叔丁酯(475 mg,2.18 mmol,1.2當量)和三甲胺(367 mg,3.63 mmol,2當量)於二氯甲烷(20 mL)中的混合物在氮氣氣氛下在室溫下攪拌過夜。將反應混合物在減壓下濃縮。將殘餘物通過C18 FLASH使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃色油的450 mg 5-溴-8-((5-羥戊基)氨基)-7-硝基-3,4-二氫異喹啉-2(1H)-甲酸叔丁酯(INT-B18-6)(54%)。LCMS: m/z (ESI), [M+H] +=460.25。 步驟7. 5-溴-8-{[5-({4-[4-(甲氧基羰基)-6-甲基吡啶-2-基]-2-甲基吡唑-3-基}氧基)戊基]氨基}-7-硝基-3,4-二氫-1H-異喹啉-2-甲酸叔丁酯(INT-B18-7)的製備。在氮氣氣氛下在0℃下向5-溴-8-((5-羥戊基)氨基)-7-硝基-3,4-二氫異喹啉-2(1H)-甲酸叔丁酯(450 mg,0.98 mmol,1當量)、三苯基膦(772 mg,2.95 mmol,3當量)於四氫呋喃(20 mL)中的攪拌混合物中添加2-(5-羥基-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(267 mg,1.08 mmol,1.1當量)和偶氮二甲酸二異丙酯(595 mg,2.95 mmol,3當量)。將反應混合物在氮氣氣氛下在60℃下攪拌3 h,並且冷卻到室溫。將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過C18 FLASH使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃色固體的350 mg 5-溴-8-{[5-({4-[4-(甲氧基羰基)-6-甲基吡啶-2-基]-2-甲基吡唑-3-基}氧基)戊基]氨基}-7-硝基-3,4-二氫-1H-異喹啉-2-甲酸叔丁酯(INT-B18-7)(52%)。LCMS: m/z (ESI), [M+H] +=689.05。 步驟8. 7-氨基-8-((5-((4-(4-(甲氧基羰基)-6-甲基吡啶-2-基)-1-甲基-1H-吡唑-5-基)氧基)戊基)氨基)-3,4-二氫異喹啉-2(1H)-甲酸叔丁酯(INT-B18-8)的製備。將5-溴-8-{[5-({4-[4-(甲氧基羰基)-6-甲基吡啶-2-基]-2-甲基吡唑-3-基}氧基)戊基]氨基}-7-硝基-3,4-二氫-1H-異喹啉-2-甲酸叔丁酯(330 mg,0.48 mmol,1當量)和Pd/C(5.1 mg,0.048 mmol,0.1當量)於甲醇(20 mL)中的混合物在氫氣氣氛下在室溫下攪拌1 h。將所得混合物過濾,並且將濾餅用甲醇(3×30 mL)洗滌。將所得溶液在減壓下濃縮,以得到呈黃色油的172 mg 7-氨基-8-((5-((4-(4-(甲氧基羰基)-6-甲基吡啶-2-基)-1-甲基-1H-吡唑-5-基)氧基)戊基)氨基)-3,4-二氫異喹啉-2(1H)-甲酸叔丁酯(INT-B18-8)。此材料在不經進一步純化的情況下用於下一反應中。LCMS: m/z (ESI), [M+H] +=579.25。 步驟9. 2-[5-({5-[2-氨基-5-溴-8-(叔丁氧基羰基)-6H,7H,9H-咪唑並[4,5-h]異喹啉-1-基]戊基}氧基)-1-甲基吡唑-4-基]-6-甲基吡啶-4-甲酸甲酯(INT-B18-9)的製備。將7-氨基-8-((5-((4-(4-(甲氧基羰基)-6-甲基吡啶-2-基)-1-甲基-1H-吡唑-5-基)氧基)戊基)氨基)-3,4-二氫異喹啉-2(1H)-甲酸叔丁酯(135 mg,約0.21 mmol,約1當量)和BrCN(33 mg,0.31 mmol,1.5當量)於二氯甲烷(5 mL)中的混合物在氮氣氣氛下在室溫下攪拌過夜。將反應用水(20 mL)淬滅,並且將混合物用二氯甲烷(3×50 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠柱色譜法進行純化並且使用二氯甲烷/甲醇(10/1)洗脫,以得到呈淡黃色油的90 mg 2-[5-({5-[2-氨基-5-溴-8-(叔丁氧基羰基)-6H,7H,9H-咪唑並[4,5-h]異喹啉-1-基]戊基}氧基)-1-甲基吡唑-4-基]-6-甲基吡啶-4-甲酸甲酯(INT-B18-9)(64%)。LCMS: m/z (ESI), [M+H] +=604.25。 步驟10. 2-[5-({5-[2-氨基-8-(叔丁氧基羰基)-6H,7H,9H-咪唑並[4,5-h]異喹啉-1-基]戊基}氧基)-1-甲基吡唑-4-基]-6-甲基吡啶-4-羧酸(INT-B18-10)的製備。將2-[5-({5-[2-氨基-5-溴-8-(叔丁氧基羰基)-6H,7H,9H-咪唑並[4,5-h]異喹啉-1-基]戊基}氧基)-1-甲基吡唑-4-基]-6-甲基吡啶-4-甲酸酯(90 mg,0.15 mmol,1當量)和LiOH .H 2O(19 mg,0.45 mmol,3當量)於MeOH(2 ml)和H 2O(0.5 ml)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將反應混合物在減壓下濃縮。將殘餘物通過C18 FLASH使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的50 mg 2-[5-({5-[2-氨基-8-(叔丁氧基羰基)-6H,7H,9H-咪唑並[4,5-h]異喹啉-1-基]戊基}氧基)-1-甲基吡唑-4-基]-6-甲基吡啶-4-羧酸(INT-B18-10)(57%)。LCMS: m/z (ESI), [M+H] +=590.50。 步驟11. 5,30-二甲基-27-氧代-7-氧雜-4,5,13,17,24,26, 31-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15, 20}]三十二-1(31),2(6),3,14,20,22,24,28(32),29-壬烯-17-甲酸叔丁酯(INT-B18-11)的製備。將2-[5-({5-[2-氨基-8-(叔丁氧基羰基)-6H,7H,9H-咪唑並[4,5-h]異喹啉-1-基]戊基}氧基)-1-甲基吡唑-4-基]-6-甲基吡啶-4-羧酸(35 mg,0.05 mmol,1當量)、 N,N-二異丙基乙胺(19.26 mg,0.15 mmol,3當量)和 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(58.03 mg,0.15 mmol,3當量)於二噁烷(2 mL)中的混合物在氮氣氣氛下在室溫下攪拌2 h。將反應用水(10 mL)淬滅,並且將混合物用二氯甲烷(3×10 mL)萃取。將合併的有機層用鹽水(3×10 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠柱色譜法進行純化並且使用二氯甲烷/甲醇(10/1)洗脫,以得到呈白色固體的30 mg 5,30-二甲基-27-氧代-7-氧雜-4,5,13,17,24, 26,31-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}. 0^{15,20}]三十二-1(31),2(6),3,14,20,22,24,28(32),29-壬烯-17-甲酸叔丁酯(INT-B18-11)(93%)。LCMS: m/z (ESI), [M+H] +=670.55。 步驟12. 5,30-二甲基-7-氧雜-4,5,13,17,24,26,31-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]三十二-1(31),2(6),3,14,20,22,24,28(32),29-壬-27-酮(INT-B18-12)的製備。將5,30-二甲基-27-氧代-7-氧雜-4,5,13, 17,24,26,31-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}.0^{14, 23}.0^{15,20}]三十二-1(31),2(6),3,14,20,22,24,28(32),29-壬烯-17-甲酸叔丁酯(30 mg,0.05 mmol,1當量)於三氟乙酸(1 mL)和二氯甲烷(2 mL)中的混合物在室溫下攪拌30分鐘。將所得混合物在減壓下濃縮。將殘餘物通過prep-HPLC用XBridge Shield RP18 OBD柱,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3 .H 2O的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的2 mg 5,30-二甲基-7-氧雜-4,5,13,17,24,26,31-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}. 0^{15,20}]三十二-1(31),2(6),3,14,20,22,24,28(32),29-壬-27-酮(INT-B18-12)(8%)。LCMS: m/z (ESI), [M+H] +=472.15。 1H NMR (DMSO-d 6, 400 MHz) δ 1.79 (2H, s), 2.04 (4H, s), 2.56 (3H, s), 2.80 (2H, d), 2.97 (2H, t), 3.73 (3H, s), 4.20-4.29 (4H, m), 4.32 (2H, s), 6.95 (1H, d), 7.34 (1H, d), 7.57 (1H, d), 7.92 (1H, s), 8.39 (1H, s)。 步驟13. 5,30-二甲基-17-(氧雜環丁-3-基)-7-氧雜-4,5, 13,17,24,26,31-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}. 0^{14,23}.0^{15,20}]三十二-1(31),2(6),3,14,20,22,24, 28(32),29-壬-27-酮(實施例B18)的製備。將5,30-二甲基-7-氧雜-4,5,13,17,24,26,31-七氮雜六環[26.3.1.0^{2,6}. 0^{13,25}.0^{14,23}.0^{15,20}]三十二-1(31),2(6),3,14,20, 22,24,28(32),29-壬-27-酮(7 mg,0.015 mmol,1當量)、3-氧雜環丁烷酮(1.28 mg,0.018 mmol,1.2當量)和三甲胺(3 mg,0.030 mmol,2當量)於二氯甲烷(2 mL)中的混合物在氮氣氣氛下在室溫下攪拌30分鐘。在室溫下向上述混合物中分批添加乙醯氧基硼氫化鈉(9.44 mg,0.045 mmol,3當量)。將所得混合物在氮氣氣氛下在室溫下攪拌過夜。將反應用水(10 mL)淬滅,並且將混合物用二氯甲烷(3×10 mL)萃取。將合併的有機層用鹽水(3×10 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將所得混合物在減壓下濃縮。將殘餘物通過prep-HPLC用XBridge Shield RP18 OBD柱,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3 .H 2O的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的1.8 mg 5,30-二甲基-17-(氧雜環丁-3-基)-7-氧雜-4,5,13,17,24,26,31-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]三十二-1(31),2(6),3,14,20,22,24,28(32),29-壬-27-酮(實施例B18)(23%)。LCMS: m/z (ESI), [M+H] +=528.20。 1H NMR (DMSO-d 6, 400 MHz) δ 1.66-1.80 (2H, m), 1.86-2.11 (4H, m), 2.56 (3H, s), 2.87-2.98 (4H, m), 3.17 (2H, d), 3.73 (3H, s), 3.75-3.81 (1H, m), 4.04 (4H, s), 4.16-4.28 (4H, m), 4.61 (2H, t), 4.70 (2H, t), 7.01 (1H, d), 7.38 (1H, d), 7.57 (1H, s), 7.93 (1H, s), 8.40 (1H, s)。 實施例B20 5,16,26-三甲基-7-氧雜-4,5,13,16,20,22,27-七氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(27),2(6),3, 14(19),17,20,24(28),25-八烯-15,23-二酮 步驟1. 4-甲氧基-1-甲基-3-硝基吡啶-2-酮(INT-B20-1)的製備。將4-羥基-3-硝基-1H-吡啶-2-酮(10 g,64.0 mmol,1當量)、碘甲烷(36.4 g,256.2 mmol,4當量)和K 2CO 3(35.4 g,256.2 mmol,4當量)於 N,N-二甲基甲醯胺(100 mL)中的混合物在氮氣氣氛下在60℃下攪拌過夜,並且冷卻到室溫。將反應用水(500 mL)淬滅,並且將混合物用二氯甲烷(3×500 mL)萃取。將合併的有機層用鹽水(3×500 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過C18 FLASH使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃色固體的1600 mg 4-甲氧基-1-甲基-3-硝基吡啶-2-酮(INT-B20-1)(13%)。LCMS: m/z (ESI), [M+H] +=185.15。 1H NMR (DMSO-d 6, 400 MHz) δ 3.48 (3H, s), 3.96 (3H, s), 6.50 (1H, d), 8.03 (1H, d)。 步驟2. 4-氨基-1-甲基-3-硝基吡啶-2-酮(INT-B20-2)的製備。將4-甲氧基-1-甲基-3-硝基吡啶-2-酮(800 mg,4.34 mmol,1當量)和NH 3於甲醇(20 mL)中的混合物在氮氣氣氛下在室溫下攪拌4 h。將沉澱的固體通過過濾收集,並且用甲醇(3×40 mL)洗滌,以得到呈黃色固體的630 mg 4-氨基-1-甲基-3-硝基吡啶-2-酮(INT-B20-2)(85%)。LCMS: m/z (ESI), [M+H] +=170.05。 1H NMR (DMSO-d 6, 400 MHz) δ 3.28 (3H, s), 5.87 (1H, d), 7.55 (1H, d), 8.03 (2H, s)。 步驟3. 3,4-二氨基-1-甲基吡啶-2-酮(INT-B20-3)的製備。將4-氨基-1-甲基-3-硝基吡啶-2-酮(1 g,5.91 mmol,1當量)和Pd/C(0.31 g,2.91 mmol,0.49當量)於甲醇(30 mL)中的混合物在氫氣氣氛下在室溫下攪拌4 h。將所得混合物過濾,並且將濾餅用甲醇(3×40 mL)洗滌。將有機溶液在減壓下濃縮,以得到呈黑色固體的900 mg 3,4-二氨基-1-甲基吡啶-2-酮(INT-B20-3)。此材料在不經進一步純化的情況下用於下一反應中。LCMS: m/z (ESI), [M+H] += 140.10。 步驟4. 2-氨基-5-甲基-3H-咪唑並[4,5-c]吡啶-4-酮(INT-B20-4)的製備。將3,4-二氨基-1-甲基吡啶-2-酮(INT-B20-3,800 mg,5.74 mmol,1當量)和BrCN(730.71 mg,6.89 mmol,1.20當量)於乙醇(15 mL)中的混合物在氮氣氣氛下在室溫下攪拌3 h。將所得混合物在真空下濃縮。將粗產物混合物在不進行進一步純化的情況下直接用於下一步驟。LCMS: m/z (ESI), [M+H] +=165.15。 步驟5. 2-氨基-3-(5-溴戊基)-5-甲基咪唑並[4,5-c]吡啶-4-酮(INT-B20-5)的製備。將2-氨基-5-甲基-3H-咪唑並[4,5-c]吡啶-4-酮(INT-B20-4,0.8 g,4.87 mmol,1當量)、1,5-二溴戊烷(5.6 g,24.36 mmol,5當量)和K 2CO 3(1.68 g,12.13 mmol,2.49當量)於乙腈(15 mL)中的混合物在氮氣氣氛下在60℃下攪拌2 h,並且冷卻到室溫。將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×60 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用乙酸乙酯洗脫進行純化,以得到呈白色固體的230 mg 2-氨基-3-(5-溴戊基)-5-甲基咪唑並[4,5-c]吡啶-4-酮(INT-B20-5)(15%)。LCMS: m/z (ESI), [M+H] +=313.10。 1H NMR (DMSO-d 6, 400 MHz) δ 1.38-1.52 (2H, m), 1.53-1.65 (2H, m), 1.74-1.85 (2H, m), 3.07-3.20 (2H, m), 3.28 (3H, s), 3.48-3.56 (2H, m), 5.76 (1H, d), 6.39 (2H, s), 7.34 (1H, d)。 步驟6. 2-{5-[(5-{2-氨基-5-甲基-4-氧代咪唑並[4,5-c]吡啶-3-基}戊基)氧基]-1-甲基吡唑-4-基}-6-甲基吡啶-4-甲酸甲酯(INT-B20-6)的製備。將2-氨基-3-(5-溴戊基)-5-甲基咪唑並[4,5-c]吡啶-4-酮(350 mg,1.11 mmol,1當量)、2-(5-羥基-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(M1,303 mg,1.23 mmol,1.10當量)和K 2CO 3(386 mg,2.79 mmol,2.5當量)於乙腈(5 mL)中的混合物在氮氣氣氛下在60℃下攪拌4 h,然後冷卻到室溫。將反應用水(100 mL)淬滅,並且將混合物用二氯甲烷(3×60 mL)萃取。將合併的有機層用鹽水(3×50 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過Prep-TLC使用二氯甲烷/甲醇(15/1)洗脫進行純化,以得到呈黃色固體的160 mg 2-{5-[(5-{2-氨基-5-甲基-4-氧代咪唑並[4,5-c]吡啶-3-基}戊基)氧基]-1-甲基吡唑-4-基}-6-甲基吡啶-4-甲酸甲酯(INT-B20-6)(29%)。LCMS: m/z (ESI), [M+H] +=480.15。 步驟7. 2-{5-[(5-{2-氨基-5-甲基-4-氧代咪唑並[4,5-c]吡啶-3-基}戊基)氧基]-1-甲基吡唑-4-基}-6-甲基吡啶-4-羧酸(INT-B20-7)的製備。將2-{5-[(5-{2-氨基-5-甲基-4-氧代咪唑並[4,5-c]吡啶-3-基}戊基)氧基]-1-甲基吡唑-4-基}-6-甲基吡啶-4-甲酸甲酯(160 mg,0.33 mmol,1當量)和LiOH .H 2O(28 mg,0.66 mmol,2當量)於THF(1 mL)和H 2O(0.25 mL)中的混合物在室溫下攪拌2 h。將所得混合物在減壓下濃縮。將殘餘物通過反向快速色譜法使用乙腈和水洗脫進行純化,以得到呈白色固體的130 mg 2-{5-[(5-{2-氨基-5-甲基-4-氧代咪唑並[4,5-c]吡啶-3-基}戊基)氧基]-1-甲基吡唑-4-基}-6-甲基吡啶-4-羧酸(INT-B20-7)(83%)。LCMS: m/z (ESI), [M+H] +=466.10。 步驟8. 5,16,26-三甲基-7-氧雜-4,5,13,16,20,22,27-七氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(27),2(6),3,14(19),17,20,24(28),25-八烯-15,23-二酮(實施例B20)的製備。將2-{5-[(5-{2-氨基-5-甲基-4-氧代咪唑並[4,5-c]吡啶-3-基}戊基)氧基]-1-甲基吡唑-4-基}-6-甲基吡啶-4-羧酸(105 mg,0.22 mmol,1當量)、 N,N,N,N-四甲基-O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(128 mg,0.33 mmol,1.5當量)和 N,N-二異丙基乙胺(87 mg,0.67 mmol,3當量)於1,4-二噁烷(3 mL)中的混合物在氮氣氣氛下在室溫下攪拌過夜。將反應用水(60 mL)淬滅,並且將混合物用二氯甲烷(3×40 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過與DMF/DMSO(5 mL)一起研磨進行純化,以得到呈黃色固體的15 mg 5,16,26-三甲基-7-氧雜-4,5,13,16,20,22,27-七氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(27),2(6),3,14(19),17,20,24(28),25-八烯-15,23-二酮(實施例B20)(13%)。LCMS: m/z (ESI), [M+H] +=448.10。 1H NMR (DMSO-d 6, 400 MHz) δ 1.66-1.80 (2H, m), 1.86-2.11 (4H, m), 2.55 (3H, s), 3.53 (3H, s), 3.73 (3H, s), 4.40 (2H, t), 4.49 (2H, t), 6.58 (1H, d), 7.55 (1H, s), 7.60 (1H, d), 7.92 (1H, s), 8.38 (1H, s), 12.87 (1H, s)。 實施例B21 5,16,26-三甲基-7-氧雜-4,5,13,16,17,20,22,27-八氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(27),2(6),3, 14(19),17,20,24(28),25-八烯-15,23-二酮 步驟1. 4-羥基-2-甲基-5-硝基噠嗪-3-酮(INT-B21-1)的製備。將4,5-二氯-2-甲基噠嗪-3-酮(4.0 g,22.34 mmol,1當量)和NaNO 2(6.1 g,89.38 mmol,4.0當量)於 N,N-二甲基甲醯胺(20 mL)中的混合物在氮氣氣氛下在90℃下攪拌24 h。將混合物冷卻到室溫,並且在減壓下濃縮。將殘餘物再溶解於溫熱的4 M HCl(20 mL)中並且冷卻到室溫。將固體過濾,並且與醚(100 mL)一起研磨,以得到呈黃色固體的1.0 g 4-羥基-2-甲基-5-硝基噠嗪-3-酮(INT-B21-1)(26%)。 1H NMR (DMSO-d 6, 400 MHz) δ 3.56 (3H, s), 8.20 (1H, s)。 步驟2. 4-氨基-2-甲基-5-硝基噠嗪-3-酮(INT-B21-2)的製備。將4-羥基-2-甲基-5-硝基噠嗪-3-酮(2.5 g,14.61 mmol,1當量)和NH 3於甲醇(60 mL)中的混合物在氮氣氣氛下在60℃下攪拌20 h。將混合物冷卻到室溫,並且在減壓下濃縮。將殘餘物通過C18 FLASH使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到以得到呈黃色固體的800 mg 4-氨基-2-甲基-5-硝基噠嗪-3-酮(INT-B21-2)(32%)。 1H NMR (400 MHz, DMSO-d 6) δ 3.60 (s, 3H), 8.26 (s, 1H), 8.83 (s, 2H)。 步驟3. 4,5-二氨基-2-甲基噠嗪-3-酮(INT-B21-3)的製備。將4-氨基-2-甲基-5-硝基噠嗪-3-酮(340 mg,2.00 mmol,1當量)和Pd/C(106 mg,1.00 mmol,0.5當量)於甲醇(15 mL)中的混合物在氫氣氣氛下在室溫下攪拌3 h。將所得混合物過濾,並且將濾餅用甲醇(50 mL)洗滌。將溶液在減壓下濃縮,以得到呈淡黃色固體的200 mg 4,5-二氨基-2-甲基噠嗪-3-酮(INT-B21-3)(71%)。此材料在不經純化的情況下用於下一反應中。LCMS: m/z (ESI), [M+H] += 141.00。 步驟4. 2-氨基-5-甲基-3H-咪唑並[4,5-d]噠嗪-4-酮(INT-B21-4)的製備。將4,5-二氨基-2-甲基噠嗪-3-酮(160 mg,1.14 mmol,1當量)和溴化氰(181 mg,1.71 mmol,1.5當量)於乙醇(5 mL)中的混合物在氮氣氣氛下在室溫下攪拌16 h。將混合物在減壓下濃縮,並且將殘餘物在Prep-TLC上使用二氯甲烷/甲醇(10/1)洗脫進行純化,以得到呈黃色固體的160 mg 2-氨基-5-甲基-3H-咪唑並[4,5-d]噠嗪-4-酮(INT-B21-4)(59%)。LCMS: m/z (ESI), [M+H] += 166.00。 步驟5. 2-氨基-3-(5-溴戊基)-5-甲基咪唑並[4,5-d]噠嗪-4-酮(INT-B21-5)的製備。將2-氨基-5-甲基-3H-咪唑並[4,5-d]噠嗪-4-酮(100 mg,0.60 mmol,1當量)、1,5-二溴戊烷(409 mg,1.78 mmol,3.0當量)和K 2CO 3(246.02 mg,1.78 mmol,3.0當量)於乙腈(10 mL)中的混合物在氮氣氣氛下在60℃下攪拌18 h,並且冷卻到室溫。將反應用水(30 mL)淬滅,並且將混合物用乙酸乙酯(3×30 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過C18FLASH使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃色固體的70 mg 2-氨基-3-(5-溴戊基)-5-甲基咪唑並[4,5-d]噠嗪-4-酮(INT-B21-5)(37%)。LCMS: m/z (ESI), [M+H] +=315.95。 步驟6. 2-{5-[(5-{2-氨基-6-甲基-7-氧代咪唑並[4,5-d]噠嗪-1-基}戊基)氧基]-1-甲基吡唑-4-基}-6-甲基吡啶-4-甲酸甲酯(INT-B21-6)的製備。將2-氨基-3-(5-溴戊基)-5-甲基咪唑並[4,5-d]噠嗪-4-酮(55 mg,0.17 mmol,1當量)、2-(5-羥基-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(M1,65 mg,0.26 mmol,1.5當量)和K 2CO 3(72 mg,0.52 mmol,3.0當量)於乙腈(2.0 mL)中的混合物在氮氣氣氛下在60℃下攪拌3 h,然後冷卻到室溫。將反應用水(30 mL)淬滅,並且將混合物用乙酸乙酯(3×30 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過C18 FLASH使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈淡黃色固體的25 mg 2-{5-[(5-{2-氨基-6-甲基-7-氧代咪唑並[4,5-d]噠嗪-1-基}戊基)氧基]-1-甲基吡唑-4-基}-6-甲基吡啶-4-甲酸甲酯(INT-B21-6)(29%)。LCMS: m/z (ESI), [M+H] +=481.20。 步驟7. 2-{5-[(5-{2-氨基-6-甲基-7-氧代咪唑並[4,5-d]噠嗪-1-基}戊基)氧基]-1-甲基吡唑-4-基}-6-甲基吡啶-4-羧酸(INT-B21-7)的製備。 將2-{5-[(5-{2-氨基-6-甲基-7-氧代咪唑並[4,5-d]噠嗪-1-基}戊基)氧基]-1-甲基吡唑-4-基}-6-甲基吡啶-4-甲酸甲酯(25 mg,0.05 mmol,1當量)和LiOH .H 2O(4.4 mg,0.10 mmol,2.0當量)於甲醇(0.5 mL)和H 2O(1.0 mL)中的混合物在氮氣氣氛下在20℃下攪拌2 h。將混合物在減壓下濃縮。將殘餘物通過C18 FLASH使用水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈淡黃色固體的15 mg 2-{5-[(5-{2-氨基-6-甲基-7-氧代咪唑並[4,5-d]噠嗪-1-基}戊基)氧基]-1-甲基吡唑-4-基}-6-甲基吡啶-4-羧酸(INT-B21-7)(61%)。LCMS: m/z (ESI), [M+H] +=467.15。 步驟8. 5,16,26-三甲基-7-氧雜-4,5,13,16,17,20,22,27-八氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(27),2(6),3,14(19),17,20,24(28),25-八烯-15,23-二酮(實施例B21)的製備。將2-{5-[(5-{2-氨基-6-甲基-7-氧代咪唑並[4,5-d]噠嗪-1-基}戊基)氧基]-1-甲基吡唑-4-基}-6-甲基吡啶-4-羧酸(15 mg,0.03 mmol,1當量)、 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(24 mg,0.06 mmol,2.0當量)和 N,N-二異丙基乙胺(12 mg,0.10 mmol,3.0當量)於1,4-二噁烷(1.0 mL)中的混合物在氮氣氣氛下在室溫下攪拌18 h。將混合物在減壓下濃縮。將殘餘物用二甲亞碸(2 mL)處理。將固體通過過濾收集,並且用甲醇(3×5 mL)洗滌,以得到呈白色固體的14 mg 5,16,26-三甲基-7-氧雜-4,5,13,16,17,20,22,27-八氮雜五環[22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]二十八-1(27),2(6),3,14(19),17,20, 24(28),25-八烯-15,23-二酮(實施例B21)(99%)。LCMS: m/z (ESI), [M+H] +=449.15。 1H NMR (CDCl 3, 400 MHz) δ 1.72-1.82 (2H, m), 2.00-2.14 (4H, m), 2.56 (3H, s), 3.73 (3H, s), 3.76 (3H, s), 4.20 (2H, t), 4.47 (2H, t), 7.56 (1H, s), 7.92 (1H, s), 8.22 (1H, s), 8.37 (1H, s)。 實施例B22 16-(2-羥乙基)-5,26-二甲基-7-氧雜-4,5,13,16,20,22,27-七氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(27),2(6),3,14(19),17,20,24(28),25-八烯-15,23-二酮 步驟1. 4-(2-{[(叔丁基二甲基甲矽烷基)氧基]氨基}乙氧基)-1-(2-{[(叔丁基二甲基甲矽烷基)氧基]氨基}乙基)-3-硝基吡啶-2-酮(INT-B22-1)的製備。將4-羥基-3-硝基-1H-吡啶-2-酮(7 g,44.84 mmol,1當量)、碳酸銫(43.8 g,134.53 mmol,3當量)和(2-溴乙氧基)(叔丁基)二甲基矽烷(42.9 g,179.38 mmol,4當量)於 N,N-二甲基甲醯胺(100 mL)中的混合物在60℃下攪拌12 h。冷卻到室溫後,將反應用水(600 mL)處理,並且將混合物用乙酸乙酯(3×500 mL)萃取。將合併的有機層用鹽水(3×500 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過矽膠柱色譜法使用石油醚/乙酸乙酯(2/1)洗脫來進行純化,以得到呈黃色固體的8.6 g 4-(2-{[(叔丁基二甲基甲矽烷基)氧基]氨基}乙氧基)-1-(2-{[(叔丁基二甲基甲矽烷基)氧基]氨基}乙基)-3-硝基吡啶-2-酮(INT-B22-1)(37%)。LCMS: m/z (ESI), [M+H] +=473.30。 1H NMR (400 MHz, DMSO-d 6) δ 0-0.19 (12H, m), 0.69-0.97 (18H, m), 3.75-3.96 (4H, m), 3.98-4.15 (2H, m), 4.20-4.46 (2H, m), 6.47-6.61 (1H, m), 7.81-7.96 (1H, m)。 步驟2. 4-氨基-1-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-3-硝基吡啶-2-酮(INT-B22-2)的製備。將4-{2-[(叔丁基二甲基甲矽烷基)氧基]乙氧基}-1-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-3-硝基吡啶-2-酮(8.6 g,18.01 mmol,1當量)和NH 3於甲醇(60 mL)中的混合物在60℃下攪拌12 h。冷卻到室溫後,將混合物在減壓下濃縮,以得到呈黃色固體的5.6 g 4-氨基-1-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-3-硝基吡啶-2-酮(INT-B22-2)(96%)。此材料在不經進一步純化的情況下用於下一反應中。LCMS: m/z (ESI), [M+H] +=314.10。 1H NMR (400 MHz, DMSO-d 6) δ 0.05(6H, s), 0.82 (9H, s), 3.72-3.78 (2H, m), 3.82-3.88 (2H, m), 5.84-5.89 (1H, m), 7.45 (1H, d), 8.04 (2H, s)。 步驟3. 3,4-二氨基-1-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}吡啶-2-酮(INT-B22-3)的製備。在氮氣氣氛下在0℃下向4-氨基-1-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-3-硝基吡啶-2-酮(4.4 g,13.89 mmol,1當量)和雷尼鎳(1.10 g,12.79 mmol,0.92當量)於甲醇(200 mL)中的攪拌混合物中添加水合肼(4.26 g,83.39 mmol,6當量)。將所得混合物在室溫下攪拌4 h,並且過濾。將濾餅用甲醇(3×100 mL)洗滌。將溶液在減壓下濃縮。將殘餘物通過矽膠柱色譜法使用石油二氯甲烷/甲醇(10/1)洗脫來進行純化,以得到呈灰色粉末的3.7 g 3,4-二氨基-1-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}吡啶-2-酮(INT-B22-3)(93%)。LCMS: m/z (ESI), [M+H] +=284.15。 1H NMR (400 MHz, DMSO-d 6) δ 0.07 (6H, s), 0.82 (9H, s), 3.74 (2H, t), 3.80-3.89 (4H, m), 5.11 (2H, s), 5.73 (1H, d), 6.79(1H,d)。 步驟4. 2-氨基-5-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-3H-咪唑並[4,5-c]吡啶-4-酮(INT-B22-4)的製備。在氮氣氣氛下在室溫下向3,4-二氨基-1-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}吡啶-2-酮(3.7 g,13.05 mmol,1當量)於乙醇(60 mL)中的攪拌混合物中添加溴化氰(2.1 g,19.58 mmol,1.5當量)。將所得混合物在室溫下攪拌4 h,並且在減壓下濃縮。將殘餘物通過矽膠柱色譜法進行純化並且使用二氯甲烷/甲醇(7/1)洗脫,以得到呈棕色固體的6.6 g 2-氨基-5-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-3H-咪唑並[4,5-c]吡啶-4-酮(INT-B22-4)(95%)。LCMS: m/z (ESI), [M+H] +=309.25。 1H NMR (400 MHz, DMSO-d 6) δ 0.14 (6H, s), 0.71 (9H, s), 3.95 (2H, t), 4.36(2H, t), 6.73 (1H, d), 7.88 (1H, d), 8.03(2H,s)。 步驟5. 2-氨基-3-(5-溴戊基)-5-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}咪唑並[4,5-c]吡啶-4-酮(INT-B22-5)的製備。在室溫下向2-氨基-5-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-3H-咪唑並[4,5-c]吡啶-4-酮(6.6 g,21.40 mmol,1當量)和K 2CO 3(5.9 g,42.79 mmol,2當量)於乙腈(100 mL)中的攪拌混合物中添加1,5-二溴-戊烷(24.6 g,106.99 mmol,5當量)。將所得混合物在60℃下攪拌4 h。冷卻到室溫後,將混合物過濾並且將濾液在減壓下濃縮。將殘餘物通過矽膠柱色譜法進行純化並且使用二氯甲烷/甲醇(10/1)洗脫,以得到呈棕色固體的2.2 g 2-氨基-3-(5-溴戊基)-5-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}咪唑並[4,5-c]吡啶-4-酮(INT-B22-5)(21%)。LCMS: m/z (ESI), [M+H] +=457.20。 1H NMR (400 MHz, DMSO-d 6) δ 0.12 (6H, s), 0.80 (9H, s), 1.31-1.43 (2H,m), 1.62-1.68 (2H, m), 1.78-1.84 (2H, m), 3.51 (2H, t), 3.80 (2H, t), 3.99 (2H,t), 4.19 (2H, t), 6.22 (1H, d), 6.50 (2H, s), 7.16 (1H, d)。 步驟6. 2-(5-{[5-(2-氨基-5-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-4-氧代咪唑並[4,5-c]吡啶-3-基)戊基]氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(INT-B22-6)的製備。在氮氣氣氛下在室溫下向2-氨基-3-(5-溴戊基)-5-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}咪唑並[4,5-c]吡啶-4-酮(2.2 g,4.81 mmol,1當量)和K 2CO 3(1.3 g,9.62 mmol,2當量)於乙腈(20 mL)中的攪拌混合物中添加含2-(5-羥基-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(1.19 g,4.81 mmol,1當量)的乙腈(5 mL)。將所得混合物在60℃下攪拌12 h。冷卻到室溫後,將反應用水(100 mL)淬滅,並且將混合物用乙酸乙酯(3×100 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物在矽膠柱上使用二氯甲烷/甲醇(10/1)洗脫進行純化,以得到呈棕色固體的2.5 g 2-(5-{[5-(2-氨基-5-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-4-氧代咪唑並[4,5-c]吡啶-3-基)戊基]氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(INT-B22-6)(80%)。LCMS: m/z (ESI), [M+H] +=610.30。 步驟7. 2-(5-{[5-(2-氨基-5-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-4-氧代咪唑並[4,5-c]吡啶-3-基)戊基]氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-羧酸(INT-B22-7)的製備。在氮氣氣氛下在室溫下向2-(5-{[5-(2-氨基-5-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-4-氧代咪唑並[4,5-c]吡啶-3-基)戊基]氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-甲酸甲酯(2.5 g,4.00 mmol,1當量)於四氫呋喃(20 mL)和H 2O(5 mL)中的攪拌混合物中添加氫氧化鋰(336 mg,8.01 mmol,2當量)。將反應混合物攪拌3 h。將所得混合物在減壓下濃縮,並且通過反向快速色譜法使用10%至50%乙腈/水洗脫進行純化,以得到呈棕色固體的2.0 g 2-(5-{[5-(2-氨基-5-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-4-氧代咪唑並[4,5-c]吡啶-3-基)戊基]氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-羧酸(INT-B22-7)(75%)。LCMS: m/z (ESI), [M+H] +=610.30。 步驟8. 16-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-5,26-二甲基-7-氧雜-4,5,13,16,20,22,27-七氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(27),2(6),3, 14(19),17,20,24(28),25-八烯-15,23-二酮(INT-B22-8)的製備。在氮氣氣氛下在室溫下向2-(5-{[5-(2-氨基-5-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-4-氧代咪唑並[4,5-c]吡啶-3-基)戊基]氧基}-1-甲基吡唑-4-基)-6-甲基吡啶-4-羧酸(20 mg,0.03 mmol,1當量)和 N,N,N,N-四甲基- O-(7-氮雜苯並三唑-1-基)六氟磷酸脲(18.71 mg,0.05 mmol,1.5當量)於二噁烷(4 mL)中的攪拌混合物中添加 N,N-二異丙基乙胺(12.72 mg,0.10 mmol,3當量)。在室溫下攪拌12 h後,將反應用水(100 mL)淬滅,並且將混合物用二氯甲烷(2×80 mL)萃取。將合併的有機層用鹽水(3×100 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-TLC使用二氯甲烷/甲醇(10/1)洗脫進行純化,以得到呈黃色固體的8 mg 16-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-5,26-二甲基-7-氧雜-4,5,13,16,20,22,27-七氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(27),2(6),3, 14(19),17,20,24(28),25-八烯-15,23-二酮(INT-B22-8)(39%)。LCMS: m/z (ESI), [M+H] +=592.35。 1H NMR (400 MHz, CD 3OD) δ 0.09 (6H, s), 0.83 (9H, s), 1.51-1.63 (2H, m), 1.76-1.91 (4H, m), 2.56 (3H, d), 3.72 (3H, s), 3.89-3.96 (2H, m), 4.09-4.16 (4H, m), 4.25-4.35 (2H, m), 6.39 (1H, d), 7.25 (1H, d),7.51 (1H, d), 7.84 (1H, s), 7.88-7.93 (1H, m)。 步驟9. 16-(2-羥乙基)-5,26-二甲基-7-氧雜-4,5,13, 16,20,22,27-七氮雜五環[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]二十八-1(27),2(6),3,14(19),17,20,24(28),25-八烯-15,23-二酮(實施例B22).將16-{2-[(叔丁基二甲基甲矽烷基)氧基]乙基}-5,26-二甲基-7-氧雜-4,5,13,16,20,22,27-七氮雜五環[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]二十八-1(27),2(6),3,14(19),17,20,24(28),25-八烯-15,23-二酮(40 mg,0.068 mmol,1當量)和四丁基氟化銨(35.35 mg,0.136 mmol,2當量)於四氫呋喃(2 mL)中的混合物在室溫下攪拌4 h。將所得混合物在減壓下濃縮。將殘餘物通過反向快速色譜法使用10%至100% MeOH/水洗脫進行純化,以得到呈黃色固體的7.7 mg 16-(2-羥乙基)-5,26-二甲基-7-氧雜-4,5,13,16,20,22,27-七氮雜五環[22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]二十八-1(27),2(6),3,14(19),17,20, 24(28),25-八烯-15,23-二酮(實施例B22)(21%)。LCMS: m/z (ESI), [M+H] +=478.15。 1H NMR (CD 3OD, 400 MHz) δ 1.85-1.93 (2H, m), 2.11-2.23 (4H, m), 2.89 (3H, s), 3.87 (3H, s), 3.88-3.94 (2H, m), 4.22 (2H, t), 4.41 (2H, t), 4.60-4.69 (2H, m), 6.69 (1H, d), 7.59 (1H, d), 8.10 (1H, s), 8.36 (1H, s), 8.95 (1H, s)。 實施例B23 18-[2-(二甲基氨基)乙基]-5,30-二甲基-7-氧雜-4,5,13, 18,24,26,31-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}.0^{14, 23}.0^{15,20}]三十二-1(31),2(6),3,14,20,22,24,28(32),29-壬-27-酮 步驟1. 18-[2-(二甲基氨基)乙基]-5,30-二甲基-7-氧雜-4,5,13,18,24,26,31-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}. 0^{14,23}.0^{15,20}]三十二-1(31),2(6),3,14,20,22,24, 28(32),29-壬-27-酮(實施例B23).將5,30-二甲基-7-氧雜-4,5,13,18,24,26,31-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}. 0^{14,23}.0^{15,20}]三十二-1(31),2(6),3,14,20,22,24, 28(32),29-壬-27-酮(實施例B16,50 mg,0.10 mmol,1當量)、(2-溴乙基)二甲胺(32 mg,0.21 mmol,2當量)、三甲胺(21 mg,0.21 mmol,2當量)和NaI(3.1 mg,0.02 mmol,0.2當量)於 N,N-二甲基甲醯胺(2 mL)中的混合物在60℃下攪拌6 h。將混合物冷卻到室溫。將反應用水(20 mL)淬滅,並且將混合物用乙酸乙酯(3×20 mL)萃取。將合併的有機層用鹽水(3×20 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-HPLC用XBridge Prep OBD C18柱,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3 .H 2O的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈白色固體的5.4 mg 18-[2-(二甲基氨基)乙基]-5,30-二甲基-7-氧雜-4,5,13,18,24,26,31-七氮雜六環[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]三十二-1(31),2(6),3,14,20,22,24,28(32),29-壬-27-酮(實施例B23) (8%)。LCMS: m/z (ESI), [M+H] +=543.40。 1H NMR (DMSO- d 6, 400 MHz) δ 1.79-1.83 (2H, m), 2.02-2.06 (4H, m), 2.21 (6H, s), 2.56 (3H, s), 2.54-2.64 (2H, m), 2.77-2.81 (2H, m), 3.15-3.44 (4H, m), 3.67 (2H, s), 3.73 (3H, s), 4.22 (2H, t), 4.35 (2H, t), 6.94 (1H, d), 7.35 (1H, d), 7.57 (1H, s), 7.93 (1H, s), 8.41 (1H, s), 12.66 (1H, s)。 實施例B27 5-[4-(二甲基氨基)哌啶-1-基]-15,21-二甲基-23-氧雜-2,9, 11,16,20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十四-3,5,7,9,13,15,17(34), 18(22),19-壬-12-酮 在氮氣氣氛下在室溫下向5-溴-15,21-二甲基-23-氧雜-2,9,11,16,20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十四-3,5,7,9,13,15,17(34), 18(22),19-壬-12-酮(INT-B2-5,70 mg,0.12 mmol,1當量)、 N,N-二甲基哌啶-4-胺(93 mg,0.73 mmol,6當量)和BrettPhos Pd G 3(33 mg,0.04 mmol,0.3當量)與1,4-二噁烷(6 mL)中的攪拌混合物中逐滴添加LiHMDS(1.5 mL,1.45 mmol,12當量)。將混合物在氮氣氣氛下在60℃下攪拌2 h,並且冷卻到室溫。將反應用飽和NH 4Cl(水溶液)(20 mL)在室溫下淬滅,並且將混合物用CH 2Cl 2(3×50 mL)萃取。將合併的有機層經無水Na 2SO 4乾燥,並且在減壓下濃縮。將殘餘物通過prep-TLC使用二氯甲烷/甲醇(10:1)洗脫進行純化。將產物通過prep-HPLC用XBridge Prep OBD C18柱使用含有0.1% NH 4HCO 3的水和乙腈進行進一步純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃色固體的20.4 mg 5-[4-(二甲基氨基)哌啶-1-基]-15,21-二甲基-23-氧雜-2,9,11,16,20,21,26-七氮雜七環[26.2.2.1^{1,26}.1^{13, 17}.0^{2,10}.0^{3,8}.0^{18,22}]三十四-3,5,7,9,13,15, 17(34),18(22), 19-壬-12-酮(實施例B27)(26%)。LCMS: m/z (ESI), [M+H] +=624.30 1H NMR (DMSO-d 6, 400 MHz) δ 1.47-1.60 (2H, m), 1.88-2.04 (6H, m), 2.06-2.15 (3H, m), 2.21-2.28 (7H, m), 2.54 (3H, s), 2.61-2.71 (2H, m), 2.81 (2H, d), 2.86 (2H, d), 3.46 (4H, d), 3.56 (2H, d), 3.76 (3H, s), 4.38 (2H, t), 6.94 (1H, dd), 7.36 (1H, d), 7.48 (1H, d), 7.61 (1H, d), 7.89 (1H, s), 8.76 (1H, s), 13.20 (1H, s)。 實施例B28-實施例B39(表3)使用上文針對由INT-B2-5合成實施例B27所描述的相同的反應條件來製備。 實施例B40A和實施例B40B 5-[4-(二甲基氨基)哌啶-1-基]-15,21-二甲基-23-氧雜-2,9, 11,16,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,15,17(33), 18(22),19-壬-12-酮(異構體1,實施例B40A) 在氮氣氣氛下在室溫下向5-溴-15,21-二甲基-23-氧雜-2,9,11,16,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,15,17(33), 18(22),19-壬-12-酮(異構體1,INT-B9-5A,80 mg,0.142 mmol,1當量)、 N,N-二甲基哌啶-4-胺(54.71 mg,0.426 mmol,3當量)和BrettPhos Pd G 3(38.68 mg,0.043 mmol,0.3當量)與1,4-二噁烷(8 mL)中的攪拌混合物中逐滴添加LiHMDS(1.420 mmol,10當量)。將混合物在60℃下攪拌1 h,然後冷卻到室溫。將反應用飽和NH 4Cl(水溶液)(10 mL)淬滅,並且將混合物用二氯甲烷(3×20 mL)萃取。將合併的有機層用鹽水(3×30 mL)洗滌,經無水Na 2SO 4乾燥並在減壓下濃縮。將殘餘物通過prep-HPLC用XBridge Shield RP18 OBD柱,使用含有10 mmol/L NH 4HCO 3和0.1% NH 3.H 2O的水和乙腈作為流動相進行純化。將含有期望化合物的級分蒸發到乾燥,以得到呈黃色固體的33.2 mg 5-[4-(二甲基氨基)哌啶-1-基]-15,21-二甲基-23-氧雜-2,9,11, 16,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2, 10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,15,17(33), 18(22),19-壬-12-酮(異構體1,實施例B40A)(37%)。LCMS: m/z (ESI), [M+H] +=610.35。 1H NMR (DMSO- d 6, 400 MHz) δ 1.65-1.78 (6H, m), 1.87-1.93 (2H, m), 1.96-2.12 (1H, d), 2.25 (6H, s), 2.45-2.49 (2H, m), 2.51-2.55 (4H, m), 2.65-2.69 (4H, m), 3.20-3.26 (2H, m), 3.35-3.45 (1H, m), 3.61-3.68 (2H, m), 3.74 (4H, s), 4.56 (1H, t), 4.70 (1H, d), 6.90-7.02 (1H, m), 7.21 (1H, d), 7.42 (1H, d), 7.56 (1H, s), 7.94 (1H, s), 8.42 (1H, s), 12.69 (1H, s)。SFC-HPLC,Rt=8.811分鐘。 5-[4-(二甲基氨基)哌啶-1-基]-15,21-二甲基-23-氧雜-2,9,11,16,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,15,17(33), 18(22), 19-壬-12-酮(異構體2,實施例B40B) 在氮氣氣氛下在室溫下向5-溴-15,21-二甲基-23-氧雜-2,9,11,16,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]三十三-3,5,7,9,13,15,17(33), 18(22),19-壬-12-酮(異構體2,INT-B9-5B,80 mg,0.14 mmol,1當量)和 N,N-二甲基哌啶-4-胺(109 mg,0.85 mmol,6當量)和BrettPhos Pd G 3(38 mg,0.04 mmol,0.3當量)與1,4-二噁烷中的攪拌混合物中逐滴添加LiHMDS(3 mL,3.00 mmol,21.0當量)。將混合物在氮氣氣氛下在60℃下攪拌1.5 h。將混合物冷卻到室溫,然後用飽和氯化銨(水溶液)(100 mL)在室溫下淬滅。將混合物用二氯甲烷(3×150 mL)萃取。將合併的有機層經無水硫酸鈉乾燥並在減壓下濃縮。將殘餘物通過prep-TLC使用二氯甲烷/甲醇(10/1)洗脫進行純化。將產物通過prep-HPLC使用XBridge Prep OBD C18的柱(30*150 mm,5μm)使用水(10 mmol/L NH 4HCO 3和0.05% NH 3H 2O)移動相A和乙腈的移動相B進一步進行純化,以得到呈黃色固體的29.7 mg 5-[4-(二甲基氨基)哌啶-1-基]-15,21-二甲基-23-氧雜-2,9,11,16,20,21,26-七氮雜七環[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]三十三-3,5,7,9,13,15,17(33),18(22),19-壬-12-酮(異構體2,實施例B40B)(34%)。LCMS: m/z (ESI), [M+H] +=610.30。 1H NMR (DMSO-d 6, 400 MHz) δ 1.51-1.75 (5H, m), 1.87-1.93 (2H, m), 2.03-2.12 (2H, m), 2.26 (6H, s), 2.45-2.49 (2H, m), 2.51-2.55 (4H, m), 2.72 (4H, d), 3.20-3.26 (2H, m), 3.35-3.45 (1H, m), 3.61-3.68 (2H, m), 3.74 (4H, s), 4.52-4.60 (1H, m), 4.70 (1H, d), 6.93 (1H, d), 7.21 (1H, s), 7.42 (1H, d), 7.57 (1H, s), 7.94 (1H, s), 8.42 (1H, s), 12.70 (1H, s)。SFC-HPLC,Rt=7.034,ee=100%。Rt1=3.894,Rt2=4.686。 實施例B41A、實施例B42A、實施例B43A、實施例B44A、實施例B44A1、實施例B44A2、實施例B45A、實施例B46A、實施例B47A、實施例B48A、實施例B49A、實施例B50A、實施例B51A、實施例B52A、實施例B53A、實施例B54A、實施例B55A、實施例B56A、實施例B57A、實施例B58A、實施例B59A(表4)使用上文針對由INT-9B-5A合成實施例B40A所描述的類似反應條件來製備。 實施例B41B、實施例B42B、實施例B43B、實施例B44B、實施例B44B1、實施例B44B2、實施例B45B、實施例B46B、實施例B47B、實施例B48B、實施例B49B、實施例B50B、實施例B51B、實施例B52B、實施例B53B、實施例B54B、實施例B55B、實施例B56B、實施例B57B、實施例B58B、實施例B59B(表4)使用上文針對由INT-9B-5B合成實施例B40B所描述的類似反應條件來製備。 生物實施例 本文所公開的示例性化合物已在以下生物測定中的一種或多種測定中表徵。 生物實施例1:EGFR WT和突變生化酶活性的抑制 重組EGFR野生型、L858R、L858R/T790M、Exon19Del、Exon19Del/T790M, EGFR[T790M/C797S/ L858R]和EGFR[T790M/C797S/d746-750]購自卡納生物公司(Carna Bio)。EGFR(d746-750 C797S)和EGFR(C797S L858R)購自新格諾康公司(signalchem)。 使用均相時間分辨螢光方法評估化合物對這些酶的抑制效力(HTRF,CisBio公司(CisBio.)目錄號62TK0PEJ)。 通過在存在1 mM 5'-三磷酸腺苷(ATP)和不同濃度的測試化合物的情況下測量EGFR酶磷酸化1 μM TK底物-生物素的酶活性來確定化合物的抑制作用。酶反應緩衝液含有5 mM MgCl2、1 mM MnCl2、0.01% CHAPS、1 mM二硫蘇糖醇(DTT)、0.5%二甲亞碸(DMSO)和1X補充酶緩衝液。將WT和突變EGFR酶(新格諾康公司)添加到含有抑制劑的板中,隨後通過添加ATP和肽底物啟動激酶反應。在室溫下溫育1 h後,通過添加含有EDTA的檢測試劑混合物終止反應。分別在615 nm和665 nm處測量螢光,其中激發波長在320 nm處。計算出的665 nm/615 nm信號比與激酶活性成正比。使用XL-FIT的四參數邏輯擬合計算對相應激酶產生50%抑制的化合物濃度(IC 50)。表5列出了通過這些測定獲得的資料。 生物實施例2:細胞增殖測定 將表達EGFR突變的Ba/F3細胞以5000個細胞/孔接種在384孔板中含有10% FBS的RPMI1640培養基中。PC-9以500個細胞/孔接種到384孔板中具有10% FBS的RPMI1640培養基中。溫育過夜後,向測定板給予一系列濃度的化合物。除了對測定板進行給予外,還使用CellTiter-Glo®發光細胞活力測定(普洛麥格公司(Promega))處理第0天的板,以測量活細胞(G0)的數量。將測定板進一步溫育72小時,並且測量活細胞(G3)的數量。增殖百分比計算如下:增殖%=100×(樣品孔的G3值- G0值)/(DMSO對照的G3值- G0值)。使用XLFit軟體在最佳擬合曲線中進一步計算產生50%增殖抑制(GI 50)的化合物濃度。表6列出了通過這些測定獲得的資料。N/A =不可用。 前述說明書被認為僅是對本公開的原理的說明。進一步地,由於許多修改和變化對於本領域技術人員來說將是顯而易見的,所以並不期望將本發明限於如上文所描述的所示確切構造和過程。因此,所有合適的修改和等效物可以被視為落入由所附權利要求書限定的本發明的範圍內。 Reference will now be made in detail to certain embodiments of the disclosure, embodiments of which are shown in the attached structures and formulas. Although the disclosure will be described in conjunction with the enumerated embodiments, it should be understood that the embodiments are not intended to limit the disclosure to those embodiments. On the contrary, the disclosure is intended to cover all alternatives, modifications and equivalents, which may be included within the scope of the disclosure as defined in the claims. Those skilled in the art will recognize many methods and materials that are similar or equivalent to the methods and materials described herein and that can be used in practicing the disclosure. The disclosure is in no way limited to the methods and materials described. In the event that one or more of the incorporated references and similar materials (including but not limited to defined terms, term usage, described techniques, etc.) are different from or contradictory to the present application, the disclosure shall prevail. All references, patents, and patent applications cited in this disclosure are hereby incorporated by reference in their entirety. It should be understood that certain features of the disclosure described in the context of separate embodiments for the sake of clarity may also be provided in combination in a single embodiment. Conversely, various features of the disclosure described in the context of a single embodiment for the sake of brevity may also be provided individually or in any suitable subcombination. It must be noted that, as used in the specification and the appended claims, the singular forms "a or an" and "the" include their plural forms unless the context clearly indicates otherwise. Thus, for example, a reference to "a compound" includes a plurality of compounds. Definitions Definitions of specific functional groups and chemical terms are described in more detail below. For purposes of this disclosure, chemical elements are identified according to the Periodic Table of the Elements, CAS version, Handbook of Chemistry and Physics, 75th ed., inside cover, and specific functional groups are generally defined as described herein. In addition, general principles of organic chemistry as well as specific functional moieties and reactivities are described in the following references: Organic Chemistry, Thomas Sorrell, University Science Books, Sausalito, 1999; Smith and March, March's Advanced Organic Chemistry, 5th edition, John Wiley & Sons, Inc., New York, 2001; LaEGFR, Comprehensive Organic Transformations, VCH Publishers, Inc., New York, 1989; Carruthers, Some Modern Methods of Organic Synthesis, 3rd edition, Cambridge University Press, Cambridge, 1996. 1987; each of which is incorporated herein by reference in its entirety. Throughout this disclosure, linking substituents are described. Where a structure clearly requires a linking group, the Markush variable listed for that group is to be understood as the linking group. For example, if a structure requires a linking group and the Markush group definition for the variable lists "alkyl", it is to be understood that "alkyl" means a linking alkylene group. In any variable (e.g., R i) occurs more than once in any constituent or formula of a compound, its definition at each occurrence is independent of its definition at every other occurrence. Thus, for example, if the indicated group is represented by 0 to 2 R iPartially substituted, the group may be optionally replaced by up to two R iPartial replacement, and R iSelected independently from R at each occurrencei. Furthermore, combinations of substituents and/or variables are permitted, but only if such combinations result in stable compounds. As used herein, for convenience, a dash "-" is used in front of or at the end of a chemical group to indicate the point of attachment of a substituent. For example, -OH is attached through an oxygen atom; a chemical group may be depicted with one or more dashes or without one or more dashes without losing its general meaning. A wavy line drawn through a line in a structure indicates the point of attachment of a group. Unless chemically or structurally required, directionality does not indicate or imply the order in which the chemical groups are written or named. As used herein, a solid line from the center of a ring indicates that the point of attachment of a substituent on the ring can be at any ring atom. When substituents are listed without specifying the atom via which such substituents are bonded to the rest of the compound of a given formula, such substituents may be bonded via any atom in the formula. Combinations of substituents and/or variables are permitted, but only if such combinations result in stable compounds. Unless otherwise indicated herein, the recitation of numerical ranges is merely intended to serve as a shorthand method of referring individually to each separate value falling within the range, and each separate value is incorporated into the specification as if it were individually recited herein. As used herein, ranges include both limits of the range unless otherwise indicated. For example, the expressions "n is an integer between 1 and 6" and "n is an integer from 1 to 6" both mean "n is 1, 2, 3, 4, 5, or 6." As used herein, the term "compounds provided herein", or "compounds disclosed herein" or "compounds disclosed herein" refers to compounds of formula (I), (II), (III) and (IV) as well as specific compounds disclosed herein. As used herein, the term "C ij" indicates a range of carbon atom numbers, where i and j are integers, and the range of carbon atom numbers includes the endpoints (i.e., i and j) and every integer point in between, and where j is greater than i. For example, C 1-6Indicates a range of one to six carbon atoms, including one carbon atom, two carbon atoms, three carbon atoms, four carbon atoms, five carbon atoms, and six carbon atoms. In some embodiments, the term "C 1-12” indicates 1 to 12 carbon atoms, especially 1 to 10 carbon atoms, especially 1 to 8 carbon atoms, especially 1 to 6 carbon atoms, especially 1 to 5 carbon atoms, especially 1 to 4 carbon atoms, especially 1 to 3 carbon atoms or especially 1 to 2 carbon atoms. As used herein, the term "alkyl", whether used as part of another term or independently, refers to a saturated straight or branched chain alkyl group that may be optionally substituted independently by one or more substituents described below. The term "C ij"Alkyl" refers to an alkyl group having i to j carbon atoms. In some embodiments, the alkyl group contains 1 to 10 carbon atoms. In some embodiments, the alkyl group contains 1 to 9 carbon atoms. In some embodiments, the alkyl group contains 1 to 8 carbon atoms, 1 to 7 carbon atoms, 1 to 6 carbon atoms, 1 to 5 carbon atoms, 1 to 4 carbon atoms, 1 to 3 carbon atoms, or 1 to 2 carbon atoms. "C 1-10Examples of "alkyl" include, but are not limited to, methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, and decyl. "C 1-6Examples of "alkyl" are methyl, ethyl, propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, 2-pentyl, 3-pentyl, 2-methyl-2-butyl, 3-methyl-2-butyl, 3-methyl-1-butyl, 2-methyl-1-butyl, 1-hexyl, 2-hexyl, 3-hexyl, 2-methyl-2-pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 3-methyl-3-pentyl, 2-methyl-3-pentyl, 2,3-dimethyl-2-butyl, 3,3-dimethyl-2-butyl, etc. As used herein, the term "alkenyl", whether used as part of another term or independently, refers to a straight or branched chain alkyl group having at least one carbon-carbon double bond that may be optionally substituted independently with one or more substituents described herein and includes groups having "cis" orientation and "trans" orientation or alternatively "E" orientation and "Z" orientation. In some embodiments, the alkenyl group contains 2 to 12 carbon atoms. In some embodiments, the alkenyl group contains 2 to 11 carbon atoms. In some embodiments, the alkenyl group contains 2 to 12 carbon atoms. In some embodiments, the alkenyl group contains 2 to 11 carbon atoms. In the formula, the alkenyl group contains 2 to 11 carbon atoms, 2 to 10 carbon atoms, 2 to 9 carbon atoms, 2 to 8 carbon atoms, 2 to 7 carbon atoms, 2 to 6 carbon atoms, 2 to 5 carbon atoms, 2 to 4 carbon atoms, 2 to 3 carbon atoms, and in some embodiments, the alkenyl group includes 2 carbon atoms. Examples of alkenyl groups include, but are not limited to, ethylenyl or vinyl, propenyl (allyl), butenyl, pentenyl, 1-methyl-2-butene-1-yl, 5-hexenyl, etc. As used herein, the term "alkynyl", whether used as part of another term or independently, refers to a straight or branched alkyl group having at least one carbon-carbon triple bond that may be optionally independently substituted with one or more substituents described herein. In some embodiments, the alkenyl group contains 2 to 12 carbon atoms. In some embodiments, the alkynyl group contains 2 to 11 carbon atoms. In some embodiments, the alkynyl group contains 2 to 11 carbon atoms, 2 to 10 carbon atoms, 2 to 9 carbon atoms, 2 to 8 carbon atoms, 2 to 7 carbon atoms, 2 to 6 carbon atoms, 2 to 5 carbon atoms, 2 to 4 carbon atoms, 2 to 3 carbon atoms, and in some embodiments, the alkynyl group includes 2 carbon atoms. Examples of alkynyl groups include, but are not limited to, ethynyl, 1-propynyl, 2-propynyl, and the like. As used herein, the term "amino" refers to -NH 2The amino group may also be substituted by one or more groups such as alkyl, alkenyl, alkynyl, aryl, carbonyl or other amino groups. As used herein, the term "aryl", whether used as part of another term or independently, refers to monocyclic and polycyclic systems having a total of 5 to 20 ring members, wherein at least one ring in the system is aromatic, and wherein each ring in the system includes 3 to 12 ring members. Examples of "aryl" include, but are not limited to, phenyl, biphenyl, naphthyl, anthracenyl, etc., which may carry one or more substituents. As used herein, the scope of the term "aryl" also includes groups in which an aromatic ring is fused to one or more additional rings. In the case of a polycyclic system, only one ring needs to be aromatic (e.g., 2,3-dihydroindole), but all rings can be aromatic (e.g., quinoline). The second ring can also be fused or bridged. Examples of polycyclic aryl groups include, but are not limited to, benzofuranyl, indanyl, phthalimidyl, naphthalimidyl, phenanthridinyl, or tetrahydronaphthyl, etc. The aryl group can be substituted at one or more ring positions with substituents as described above. As used herein, the term "cyano" refers to -CN. As used herein, the term "cycloalkyl", whether used as part of another term or by itself, refers to a monovalent non-aromatic saturated or partially unsaturated monocyclic and polycyclic system in which all ring atoms are carbon and the system includes at least three ring-forming carbon atoms. In some embodiments, the cycloalkyl group may contain 3 to 12 ring-forming carbon atoms, 3 to 10 ring-forming carbon atoms, 3 to 9 ring-forming carbon atoms, 3 to 8 ring-forming carbon atoms, 3 to 7 ring-forming carbon atoms, 3 to 6 ring-forming carbon atoms, 3 to 5 ring-forming carbon atoms, 4 to 12 ring-forming carbon atoms, 4 to 10 ring-forming carbon atoms, 4 to 9 ring-forming carbon atoms, 4 to 8 ring-forming carbon atoms, 4 to 7 ring-forming carbon atoms, 4 to 6 ring-forming carbon atoms, 4 to 5 ring-forming carbon atoms. The cycloalkyl group may be saturated or partially unsaturated. The cycloalkyl group may be substituted. In some embodiments, the cycloalkyl group may be a saturated cyclic alkyl group. In some embodiments, the cycloalkyl group may be a partially unsaturated cyclic alkyl group including at least one double or triple bond in its ring system. In some embodiments, the cycloalkyl group may be monocyclic or polycyclic. In the case of polycyclic systems, fused, spiro and bridged ring systems are included within the scope of this definition. Examples of monocyclic cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, 1-cyclopent-1-enyl, 1-cyclopent-2-enyl, 1-cyclopent-3-enyl, cyclohexyl, 1-cyclohex-1-enyl, 1-cyclohex-2-enyl, 1-cyclohex-3-enyl, cyclohexadienyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, cycloundecyl, and cyclododecyl. Examples of polycyclic cycloalkyl groups include, but are not limited to, adamantyl, norbornyl, fluorenyl, spiro-pentadienyl, spiro[3.6]-decyl, bicyclo[1,1,1]pentenyl, bicyclo[2,2,1]heptenyl, etc. As used herein, the term "halogen" refers to an atom selected from fluorine (fluorine or fluoro), chlorine (chlorine or chloro), bromine (bromine or bromo) and iodine (iodine or iodo). As used herein, the term "heteroatom" refers to nitrogen, oxygen, sulfur, phosphorus, and includes any oxidized form of nitrogen, sulfur or phosphorus and any quaternized form of basic nitrogen (including N-oxide). As used herein, the term "heteroalkyl" refers to an alkyl group in which at least one of its carbon atoms is replaced by a heteroatom selected from N, O or S. The heteroalkyl group can be a carbon group or a heteroatom group (i.e., the heteroatom can appear in the middle or at the end of the group) and can be optionally substituted independently by one or more substituents described herein. The term "heteroalkyl" covers alkoxy and heteroalkoxy groups. As used herein, the term "heteroalkenyl" refers to an alkenyl group in which at least one of its carbon atoms is replaced by a heteroatom selected from N, O or S. The heteroalkenyl group may be a carbon group or a heteroatom group (i.e., the heteroatom may be present in the middle or at a terminal of the group), and may be optionally and independently substituted with one or more substituents described herein. As used herein, the term "heteroalkynyl" refers to an alkynyl group in which at least one of its carbon atoms is replaced by a heteroatom selected from N, O or S. The heteroalkynyl group may be a carbon group or a heteroatom group (i.e., the heteroatom may be present in the middle or at a terminal of the group), and may be optionally and independently substituted with one or more substituents described herein. As used herein, the term "heteroaryl", whether used as part of another term or independently, refers to an aryl group having one or more heteroatoms in addition to carbon atoms. Heteroaryl can be monocyclic. Examples of monocyclic heteroaryl include, but are not limited to, thienyl, furanyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, thiadiazolyl, pyridyl, oxazinyl, pyrimidinyl, pyrazinyl, indolizinyl, purinyl, naphthyridinyl, benzofuranyl, and pteridinyl. Heteroaryl also includes polycyclic groups in which a heteroaromatic ring is fused to one or more aryl, cycloalkyl, or heterocyclic rings, wherein the connecting group or point of attachment is located on the heteroaromatic ring. Examples of polycyclic heteroaryl groups include, but are not limited to, indolyl, isoindolyl, benzothienyl, benzofuranyl, benzo[1,3]dioxolyl, dibenzofuranyl, indazolyl, benzimidazolyl, benzothiazolyl, quinolyl, isoquinolyl, dihydroquinolyl, dihydroisoquinolyl, tetrahydroquinolyl, tetrahydroisoquinolyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, 4H-quinolizinyl, oxazolyl, acridinyl, phenazinyl, phenothiazinyl, phenoxazinyl, tetrahydroquinolyl, tetrahydroisoquinolyl, etc. As used herein, the term "heterocyclic group" refers to a saturated or partially unsaturated carbocyclic group, wherein one or more ring atoms are heteroatoms independently selected from oxygen, sulfur, nitrogen, phosphorus, etc., and the remaining ring atoms are carbon, wherein one or more ring atoms can be optionally independently substituted by one or more substituents. In some embodiments, the heterocyclic group is a saturated heterocyclic group. In some embodiments, the heterocyclic group is a partially unsaturated heterocyclic group having one or more double bonds in its ring system. In some embodiments, the heterocyclic group can include any oxidized form of carbon, nitrogen, sulfur or phosphorus and any quaternized form of basic nitrogen. "Heterocyclic" also includes groups in which the heterocyclic group is fused to a saturated, partially unsaturated, or fully unsaturated (i.e., aromatic) carbocyclic or heterocyclic group. Where possible, the heterocyclic group can be carbon-linked or nitrogen-linked. In some embodiments, the heterocyclic group is carbon-linked. In some embodiments, the heterocyclic group is nitrogen-linked. For example, a group derived from pyrrole can be pyrrol-1-yl (nitrogen-linked) or pyrrol-3-yl (carbon-linked). Further, a group derived from imidazole can be imidazol-1-yl (nitrogen-linked) or imidazol-3-yl (carbon-linked). In some embodiments, the term "3- to 12-membered heterocyclic group" refers to a 3- to 12-membered saturated or partially unsaturated monocyclic or polycyclic heterocyclic system having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, sulfur or phosphorus. In the case of a polycyclic system, fused, spiro and bridged ring systems are also included within the scope of this definition. Examples of monocyclic heterocyclic groups include, but are not limited to, oxacyclobutanyl, 1,1-dioxothiacyclobutanyl, pyrrolidinyl, tetrahydrofuranyl, tetrahydrothienyl, pyrrolyl, furanyl, thienyl, pyrazolyl, imidazolyl, triazolyl, oxazolyl, thiazolyl, piperidinyl, piperazinyl, piperidinyl, morpholinyl, pyridinyl, pyrazinyl, pyrimidinyl, oxazinyl, triazinyl, pyridonyl, pyrimidonyl, pyrazinonyl, pyrimidonyl, oxazinonyl, pyrrolidinyl, triazinonyl, and the like. Examples of fused heterocyclic radicals include, but are not limited to, quinolinyl, isoquinolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, quinoxalinyl, quinolizinyl, quinazolinyl, azaindolizinyl, pteridinyl, chromenyl, isochromenyl, indolyl, isoindolyl, indolizinyl, indazolyl, purinyl, benzofuranyl, isobenzofuranyl, benzimidazolyl, benzothiophenyl, benzothiophene, benzothiazolyl ... oxazolyl, oxazolyl, phenazinyl, phenothiazinyl, phenanthridinyl, hexahydro-1H-pyrrolizinyl, imidazo[1,2-a]pyridinyl, [1,2,4]triazol[4,3-a]pyridinyl, [1,2,3]triazol[4,3-a]pyridinyl, octahydropyrrolo[3,4-b]pyrrolyl, octahydropyrrolo[3,4-c]pyrrolyl, etc. Examples of spiroheterocyclic groups include, but are not limited to, spiropyranyl, spirooxazinyl, 2,6-diazaspiro[3.3]heptanyl, 2,5-diazaspiro[3.4]octanyl, 2,6-diazaspiro[3.4]octanyl, 2,7-diazaspiro[3.5]nonanyl, 5-oxa-2,8-diazaspiro[3.5]nonanyl, 2,7-diazaspiro[4.4]nonanyl, 1,7-diazaspiro[3.5]nonanyl, 2,8-diazaspiro[4.5]decanyl, 2,8-diazaspiro[4.5]decanyl, and the like. Examples of bridged heterocyclic groups include, but are not limited to, morpholinyl, hexamethylenetetramine, 3-aza-bicyclo[3.1.0]hexane, 3,6-diazabicyclo[3.1.1]heptane, 2,5-diazabicyclo[2.2.1]heptane, 8-aza-bicyclo[3.2.1]octane, 3-azabicyclo[3.2.1]octane, 1-aza-bicyclo[2.2.2]octane, 1,4-diazabicyclo[2.2.2]octane, 3-azabicyclo[3.2.2]nonane, etc. As used herein, the term "hydroxyl" or "hydroxy" refers to -OH. As used herein, the term "partially unsaturated" refers to a group that includes at least one double or triple bond. The term "partially unsaturated" is intended to cover rings with multiple sites of unsaturation, but is not intended to include aromatic (i.e., fully unsaturated) moieties. As used herein, the term "optionally" or "optionally" means that the subsequently described event or circumstance may or may not occur, and the description includes instances where the event or circumstance occurs and instances where the event or circumstance does not occur. As used herein, the term "substituted", whether preceded by the term "optionally" or not, means that one or more hydrogens of the designated moiety are replaced with a suitable substituent. It should be understood that "substituted" or "substituted by" includes the implicit proviso that such substitution is consistent with the allowed valencies of the substituted atom and that the substitution results in a stable or chemically feasible compound, e.g., a compound that does not spontaneously undergo transformations such as rearrangement, cyclization, elimination, etc. Unless otherwise specified, an "optionally substituted" group may have a suitable substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent may be the same or different at each position. Substitutions may include, but are not limited to, alkyl, alkenyl, alkynyl, alkoxy, acyl, amino, amido, amidino, aryl, azido, carbamoyl, carboxyl, carboxyl ester, cyano, guanidino, halogen, halogenated alkyl, heteroalkyl, heteroaryl, heterocyclic, hydroxyl, hydrazine, imino, oxo, nitro, alkylsulfinyl, sulfonic acid, alkylsulfonyl, thiocyanate, thiol, thione, or combinations thereof. It will be understood by those skilled in the art that the substituents themselves may be substituted, if appropriate. Unless specifically stated as "unsubstituted," references to chemical moieties herein are to be understood to include substituted variants. For example, reference to an "aryl" group or moiety implicitly includes both substituted and unsubstituted variants. As used herein, the term "substituted", whether preceded by the term "optionally", means that one or more hydrogens of the designated moiety are replaced with a suitable substituent. It should be understood that "substituted" or "substituted by..." includes the implicit precondition that such substitution is consistent with the allowed valence of the substituted atom and that the substitution produces a stable or chemically feasible compound, for example, a compound that does not spontaneously undergo transformations such as rearrangement, cyclization, elimination, etc. Unless otherwise specified, an "optionally substituted" group may have a suitable substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent may be the same or different at each position. It will be understood by those skilled in the art that the substituent itself may be substituted if appropriate. Unless specifically stated as "unsubstituted," references to chemical moieties herein are to be understood to include substituted variants. For example, reference to an "aryl" group or moiety implicitly includes both substituted and unsubstituted variants. The symbols "R" and "S" represent the configuration of substituents around a chiral carbon atom. The isomer descriptors "R" and "S" are used as described herein to indicate the configuration of atoms relative to the core molecule and are intended to be used as defined in the literature (IUPAC Recommendations 1996, Pure and Applied Chemistry, 68: 2193-2222 (1996)). Compounds In one aspect, the present disclosure provides compounds of formula (I): Formula (I) or a pharmaceutically acceptable salt thereof, wherein, Ring A is selected from the group consisting of: cycloalkyl, heterocyclic, aryl and heteroaryl; Ring B is selected from the group consisting of: cycloalkyl, heterocyclic, aryl and heteroaryl; L 1Selected from the group consisting of: a bond, an alkyl group, an alkenyl group, an alkynyl group, a heteroalkyl group, a heteroalkenyl group and a heteroalkynyl group, wherein the alkyl group, the alkenyl group, the alkynyl group, the heteroalkyl group, the heteroalkenyl group and the heteroalkynyl group are optionally substituted with one or more groups independently selected from the group consisting of: a hydroxyl group, a halogen group, a cyano group and an amino group; L 2Select from the group consisting of: key, N(R A), alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted by one or more R BReplace; Where R ASelected from the group consisting of: hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from: hydroxyl, halogen, cyano, amino, alkyl and halogenated alkyl; wherein each R BIndependently selected from the group consisting of hydroxyl, halogen, cyano, amino, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, cycloalkyl, heterocyclo, aryl and heteroaryl; L 3Selected from the group consisting of: a bond, an alkyl group, an alkenyl group, an alkynyl group, a heteroalkyl group, a heteroalkenyl group and a heteroalkynyl group, wherein the alkyl group, the alkenyl group, the alkynyl group, the heteroalkyl group, the heteroalkenyl group and the heteroalkynyl group are optionally substituted with one or more groups independently selected from the group consisting of: a hydroxyl group, a halogen group, a cyano group and an amino group; L 4Choose from O, S or N(R C); Where R CSelected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, halogenated alkyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl; Each R 1Independently selected from the group consisting of: hydroxyl, halogen, cyano, amino, -N(R D) 2, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted by one or more R EReplace; where each R DIndependently selected from the group consisting of: hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the following: hydroxyl, halogen, cyano,  -N(R F) 2OR G; where each R EIndependently selected from the group consisting of: hydrogen, hydroxyl, halogen, cyano, amino, -N(R F) 2、-alkyl-N(R F) 2、-C(O)OR G, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the following: hydroxyl, halogen, cyano, amino and alkyl; wherein R Fand R GEach of R is independently selected from the group consisting of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl; Each R 2Independently selected from the group consisting of: hydrogen, hydroxyl, halogen, cyano, amino, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from: hydroxyl, halogen, cyano, amino, alkyl and halogenated alkyl; R 3Selected from the group consisting of: hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl and heterocyclo, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl and heterocyclo are optionally substituted with one or more groups independently selected from: hydroxyl, halogen, cyano, amino, alkyl and halogenated alkyl; m is an integer from 0 to 5; and n is an integer from 0 to 4. In some embodiments, Ring A is aryl. In certain embodiments, Ring A is C 6-12Aryl, C 6-11Aryl, C 6-10Aryl, C 6-9Aryl or C 6-8Aryl. In some embodiments, Ring A is phenyl. In some embodiments, Ring A is heteroaryl. In some embodiments, Ring A is 5-12 membered heteroaryl, 5-11 membered heteroaryl, 5-10 membered heteroaryl, 5-9 membered heteroaryl, 5-8 membered heteroaryl, 5-7 membered heteroaryl or 5-6 membered heteroaryl. In some embodiments, Ring A is selected from the group consisting of furanyl, phenylthio, pyrrolyl, pyridyl, pyranyl, pyrimidinyl, oxazinyl, pyrazinyl and tetrahydroisoquinolinyl. In some embodiments, Ring A is selected from the group consisting of: and . In some embodiments, Ring B is aryl. In some embodiments, Ring B is C 6-12Aryl, C 6-11Aryl, C 6-10Aryl, C 6-9Aryl or C 6-8In some embodiments, ring B is phenyl. In some embodiments, ring B is heteroaryl. In some embodiments, ring B is 5-12 membered heteroaryl, 5-11 membered heteroaryl, 5-10 membered heteroaryl, 5-9 membered heteroaryl, 5-8 membered heteroaryl, 5-7 membered heteroaryl or 5-6 membered heteroaryl. In some embodiments, ring B is pyridyl or pyrazolyl. In some embodiments, ring B is selected from the group consisting of: and . In some implementations, L 1is a key. In some implementations, L 1is an alkyl group. In certain embodiments, L 1It is C 1-6Alkyl, C 1-5Alkyl, C 1-4Alkyl, C 1-3Alkyl or C 1-2Alkyl. In certain embodiments, L 1Choose from a group consisting of: and , where L 1*end and L 2connection. In some embodiments, L 2is a key. In some implementations, L 2It is N(R A), and R ASelected from alkyl or heterocyclic groups, wherein the alkyl or heterocyclic group is optionally substituted by one or more halogens or alkyl groups. In certain embodiments, L 2It is N(R A), and R Ais ethyl, difluoroethyl, trifluoroethyl or cyclohexane. In some embodiments, L 2is optionally represented by one or more R BSubstituted cycloalkyl. In certain embodiments, L 2It is C 3-10Cycloalkyl, C 3-9Cycloalkyl, C 3-8Cycloalkyl, C 3-7Cycloalkyl, C 3-6Cycloalkyl or C 3-5Cycloalkyl, each of which is optionally substituted by one or more R BReplace. In some embodiments, L 2is optionally represented by one or more R Breplaced . In some implementations, L 2is optionally represented by one or more R BSubstituted heterocyclic group. In certain embodiments, L 2is a heterocyclic group containing one or more heteroatoms selected from N, O or S. In certain embodiments, L 2is a 3-12-membered heterocyclic group, a 3-11-membered heterocyclic group, a 3-10-membered heterocyclic group or a 3-9-membered heterocyclic group, each of which is optionally substituted by one or more R BReplace. In some embodiments, L 2is a heterocyclic group selected from the group consisting of: and , each of which is optionally represented by one or more R BReplacement, where L 2* end and L 3connection. In some embodiments, L 2is one or more R BSubstituted cycloalkyl or heterocyclic group, and R Bis an alkyl group. In certain embodiments, R BIt is C 1-6Alkyl, C 1-5Alkyl, C 1-4Alkyl, C 1-3Alkyl or C 1-2Alkyl. In certain embodiments, R Bis methyl. In some embodiments, L 2Choose from a group consisting of: and , where L 2*end and L 3connection. In some embodiments, L 3is an alkyl group. In some embodiments, L 3It is C 1-6Alkyl, C 1-5Alkyl, C 1-4Alkyl, C 1-3Alkyl or C 1-2Alkyl. In certain embodiments, L 3is ethyl. In some embodiments, L 4is O or NH. In some embodiments, L 1is the key, and L 2is a key or optionally replaced by one or more R BSubstituted heterocyclic group. In some embodiments, L 1is an alkyl group, and L 2Yes key, N(R A) or optionally one or more R BSubstituted cycloalkyl. In some embodiments, -L 1-L 2-L 3-L 4- is -alkyl-O-. In certain embodiments, -L 1-L 2-L 3-L 4-Yes-(C 3-8Alkyl)-O-. In certain embodiments, -L 1-L 2-L 3-L 4-Yes-CH 2CH(CH 3)(CH 2) 3-O- or    -CH 2CH(CH 2CH 3)(CH 2) 3-O-. In some implementations, -L 1-L 2-L 3-L 4-is-heterocyclic-alkyl-O-, wherein the heterocyclic group is optionally replaced by one or more R BReplace. In some embodiments, -L 1-L 2-L 3-L 4- is -(5-10 membered heterocyclic group)-(C 1-6Alkyl)-O-, wherein the 5-10 membered heterocyclic group is optionally replaced by one or more R BReplace. In some embodiments, -L 1-L 2-L 3-L 4-Choose from a group consisting of: as well as . In some implementations, -L 1-L 2-L 3-L 4- is - alkyl-N(R A)-alkyl-O-, where R ASelected from alkyl or heterocyclic groups, wherein the alkyl or heterocyclic group is optionally substituted by one or more halogens or alkyl groups. In certain embodiments, -L 1-L 2-L 3-L 4-Yes-(C 1-6Alkyl)-N(R A)-(C 1-6Alkyl)-O-, where R ASelected from C 1-6Alkyl or 3- to 6-membered heterocyclic group, wherein the alkyl or heterocyclic group is optionally substituted by one or more halogens or C1-6Alkyl substitution. In certain embodiments, -L 1-L 2-L 3-L 4-yes or . In some implementations, -L 1-L 2-L 3-L 4-is-alkyl-cycloalkyl-alkyl-O-, wherein the cycloalkyl is optionally replaced by one or more RBReplace. In some embodiments, -L 1-L 2-L 3-L 4-Yes-(C 1-6Alkyl)-(C 3-6Cycloalkyl)-(C 1-6Alkyl)-O-, wherein the C 3-6The cycloalkyl group is optionally replaced by one or more RBReplace. In some embodiments, -L 1-L 2-L 3-L 4-yes . In some implementations, R 1is hydroxyl and m is 1. In some embodiments, R 1is a halogen and m is 1. In certain embodiments, R 1is bromine or fluorine, and m is 1. In some embodiments, R 1Yes-N(R D) 2, and m is 1. In some embodiments, R 1Yes-N(R D) 2, and each R Dindependently hydrogen or optionally one or more independently selected from -N(R F) 2OR GAn alkyl group substituted with a group. In certain embodiments, R 1Yes-N(R D) 2, and each R Dis arbitrarily selected by one or more independently selected from -N(R F) 2or -OR GC 1-6Alkyl, C 1-5Alkyl, C 1-4Alkyl, C 1-3Alkyl or C 1-2Alkyl. In certain embodiments, R 1Yes-N(R D) 2, and each R Dare independently methyl, methoxyethyl, N,N-dimethylaminoethyl, hydroxyethyl or N,N-dimethylaminopropyl. In some embodiments, R 1is optionally represented by one or more R Esubstituted heterocyclic group, and m is 1. In certain embodiments, R 1is optionally represented by one or more R Esubstituted 3- to 10-membered heterocyclic group, and m is 1. In certain embodiments, R 1Choose from a group consisting of: and , each of which is optionally represented by one or more R EReplace. In some embodiments, R Eis a halogen. In certain embodiments, R Eis F. In some embodiments, each R EIndependently, it is a halogen, -N(R F) 2、-alkyl-N(R F) 2、-C(O)OR Gor an alkyl group optionally substituted with one or more halogens. In some embodiments, R Fand R GEach of R is independently an alkyl group. In certain embodiments, R Fand R GEach of them is independently C 1-6Alkyl, C 1-5Alkyl, C 1-4Alkyl, C 1-3Alkyl or C 1-2Alkyl. In certain embodiments, R Fand R GEach of them is independently C 1-3Alkyl. In certain embodiments, each R EIndependently selected from the group consisting of F, -N(CH 3) 2、-C 1-3Alkyl-N(CH 3) 2, -C(O)O(tert-butyl), methyl, ethyl or trifluoroethyl. In certain embodiments, R Eis a cycloalkyl or heterocyclic group, wherein the cycloalkyl and heterocyclic groups are optionally substituted with one or more alkyl and/or halogen groups. In certain embodiments, R ESelected from cyclopropyl, morpholine, piperazine, oxacyclobutenyl or azocyclobutane, each of which is optionally substituted with one or more alkyl and/or halogen. In certain embodiments, R ESelected from cyclopropyl, morpholinyl, piperazinyl, oxacyclobutenyl or azocyclobutanyl, each of which is optionally substituted with one or more alkyl and/or halogen. In some embodiments, m is 2, R 1One of them is a halogen, and the other R 1is optionally represented by one or more R ESubstituted heterocyclic group. In certain embodiments, m is 2, R 1One of them is a halogen, and the other R 1is a heterocyclic group selected from the group consisting of: and , each of which is optionally represented by one or more R EReplace. In some embodiments, each R EIndependently selected from halogens,     -N(R F) 2、-alkyl-N(R F) 2、-C(O)OR Gor an alkyl group optionally substituted with one or more halogens. In some embodiments, R 1Selected from the group consisting of: OH, F, Br, and . In some implementations, R 2is a halogen, and n is 1 or 2. In certain embodiments, R 2is F. In some embodiments, R 2is an alkyl or cycloalkyl group, wherein the alkyl and cycloalkyl groups are optionally substituted with one or more or halogens. In certain embodiments, R2It is C 1-6Alkyl or C 3-6Cycloalkyl, wherein the C1-6Alkyl and C 3-6The cycloalkyl group is optionally substituted with one or more halogens. In certain embodiments, R2is methyl, ethyl, cyclopropyl or trifluoroethyl. In some embodiments, R 3is an alkyl group or a cycloalkyl group. In certain embodiments, R 3It is C 1-6Alkyl or C 3-6Cycloalkyl. In certain embodiments, R 3is methyl, ethyl or cyclopropyl. In some embodiments, m is 0, 1 or 2. In some embodiments, n is 0 or 1. Exemplary compounds of formula (I) are described below. and . On the one hand, the present disclosure provides a compound of formula (II): (II) or a pharmaceutically acceptable salt thereof, wherein: Ring A 1is a 7-12 membered cycloalkyl, a 7-12 membered heterocyclic group, a 7-12 membered aryl group or a 7-12 membered heteroaryl group; Ring B is selected from the group consisting of: cycloalkyl, heterocyclic group, aryl group and heteroaryl group; L 1Selected from the group consisting of: a bond, an alkyl group, an alkenyl group, an alkynyl group, a heteroalkyl group, a heteroalkenyl group and a heteroalkynyl group, wherein the alkyl group, the alkenyl group, the alkynyl group, the heteroalkyl group, the heteroalkenyl group and the heteroalkynyl group are optionally substituted with one or more groups independently selected from the group consisting of: a hydroxyl group, a halogen group, a cyano group and an amino group; L 2Select from the group consisting of: key, N(R A), alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted by one or more R BReplace; Where R ASelected from the group consisting of: hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the following: hydroxyl, halogen, cyano, amino, alkyl and halogenated alkyl; wherein each R BIndependently selected from the group consisting of hydroxyl, halogen, cyano, amino, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, cycloalkyl, heterocyclo, aryl and heteroaryl; L 3Selected from the group consisting of: a bond, an alkyl group, an alkenyl group, an alkynyl group, a heteroalkyl group, a heteroalkenyl group and a heteroalkynyl group, wherein the alkyl group, the alkenyl group, the alkynyl group, the heteroalkyl group, the heteroalkenyl group and the heteroalkynyl group are optionally substituted with one or more groups independently selected from the group consisting of: a hydroxyl group, a halogen group, a cyano group and an amino group; L 4Choose from O, S or N(R C); Where R CSelected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, halogenated alkyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl; Each R 1Independently selected from the group consisting of: hydroxyl, halogen, cyano, amino, -N(R D) 2, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted by one or more R EReplace; where each R DIndependently selected from the group consisting of: hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the following: hydroxyl, halogen, cyano,  -N(R F) 2OR G; where each R EIndependently selected from the group consisting of: hydrogen, hydroxyl, halogen, cyano, amino, -N(R F) 2、-alkyl-N(R F) 2、-C(O)OR G, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the following: hydroxyl, halogen, cyano, amino and alkyl; wherein R Fand R GEach of R is independently selected from the group consisting of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl; Each R 2Independently selected from the group consisting of: hydrogen, hydroxyl, halogen, cyano, amino, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from: hydroxyl, halogen, cyano, amino, alkyl and halogenated alkyl; Each R 3Selected from the group consisting of: hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl and heterocyclo, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl and heterocyclo are optionally substituted with one or more groups independently selected from: hydroxyl, halogen, cyano, amino, alkyl and halogenated alkyl; m is an integer from 0 to 5; n is an integer from 0 to 4; and p is an integer from 0 to 3. In some embodiments, Ring A 1is a 7-12 membered heteroaryl group. In certain embodiments, Ring A 1is tetrahydroisoquinolinyl. In certain embodiments, Ring A 1yes or . In some embodiments, Ring B is aryl. In some embodiments, Ring B is C 6-12Aryl, C 6-11Aryl, C 6-10Aryl, C 6-9Aryl or C 6-8Aryl. In some embodiments, Ring B is phenyl. In some embodiments, Ring B is heteroaryl. In some embodiments, Ring B is 5-12 membered heteroaryl, 5-11 membered heteroaryl, 5-10 membered heteroaryl, 5-9 membered heteroaryl, 5-8 membered heteroaryl, 5-7 membered heteroaryl or 5-12 membered heteroaryl. In some embodiments, Ring B is pyridyl or pyrazolyl. In some embodiments, Ring B is selected from the group consisting of: and . In some implementations, L 1is an alkyl group. In certain embodiments, L 1It is C 1-6Alkyl, C 1-5Alkyl, C 1-4Alkyl, C 1-3Alkyl or C 1-2Alkyl. In certain embodiments, wherein L 1yes or , where L 1*end and L 2connection. In some embodiments, L 2is a key. In some implementations, L 1is an alkyl group, and L 2is a key. In some implementations, L 3is an alkyl group. In certain embodiments, L 3It is C 1-6Alkyl, C 1-5Alkyl, C 1-4Alkyl, C 1-3Alkyl or C 1-2Alkyl. In certain embodiments, L 3is ethyl. In some embodiments, L 4is O or NH. In some embodiments, -L 1-L 2-L 3-L 4- is -alkyl-O-. In certain embodiments, -L 1-L 2-L 3-L 4-Yes-(C 3-8Alkyl)-O-. In certain embodiments, -L 1-L 2-L 3-L 4-Yes-(CH 2) 5-O-, -CH 2CH(CH 3)(CH 2) 3-O- or -CH 2CH(CH 2CH 3)(CH 2) 3-O-. In some embodiments, m is 0. In some embodiments, m is 1, and R 1is optionally represented by one or more R ESubstituted alkyl. In certain embodiments, m is 1, and R 1is optionally represented by one or more R ESubstituted C 1-6Alkyl, C 1-5Alkyl, C 1-4Alkyl, C 1-3Alkyl or C 1-2Alkyl. In certain embodiments, R 1is optionally represented by one or more R Esubstituted ethyl. In some embodiments, R EYes-N(R F) 2, and R Fis an alkyl group. In certain embodiments, R EYes-N(R F) 2, and R FIt is C 1-6Alkyl, C 1-5Alkyl, C 1-4Alkyl, C 1-3Alkyl or C 1-2Alkyl. In certain embodiments, R EYes-N(CH 3) 2. In some embodiments, m is 1, and R 1is optionally represented by one or more R ESubstituted heterocyclic group. In certain embodiments, m is 1, and R 1is a 3- to 10-membered heterocyclic group, a 3- to 9-membered heterocyclic group, a 3- to 8-membered heterocyclic group, a 3- to 7-membered heterocyclic group, a 3- to 6-membered heterocyclic group or a 3- to 5-membered heterocyclic group, each of which is optionally substituted by one or more R EReplace. In some embodiments, R 1is an oxacyclobutane. In certain embodiments, R 1yes . In some embodiments, n is 1. In some embodiments, R 2is an alkyl group. In certain embodiments, R 2It is C 1-6Alkyl, C 1-5Alkyl, C 1-4Alkyl, C 1-3Alkyl or C 1-2Alkyl. In certain embodiments, R 2is methyl. In some embodiments, n is 1, and R 2is an alkyl group. In certain embodiments, n is 1, and R 2It is C 1-6Alkyl, C 1-5Alkyl, C 1-4Alkyl, C 1-3Alkyl or C 1-2Alkyl. In certain embodiments, n is 1, and R 2is methyl. In some embodiments, yes . In some embodiments, p is 1. In some embodiments, R 3is an alkyl group. In certain embodiments, R 3It is C 1-6Alkyl, C 1-5Alkyl, C 1-4Alkyl, C 1-3Alkyl or C 1-2Alkyl. In certain embodiments, R 3is methyl. In some embodiments, p is 1, and R 3is an alkyl group. In certain embodiments, p is 1, and R 3It is C 1-6Alkyl, C 1-5Alkyl, C 1-4Alkyl, C 1-3Alkyl or C 1-2Alkyl. In certain embodiments, p is 1, and R 3is methyl. In some embodiments, yes . In some implementations, yes . In some embodiments, m is 0 or 1. In some embodiments, n is 1. In some embodiments, p is 1. On the other hand, the present disclosure provides a compound of formula (III): (III) or a pharmaceutically acceptable salt thereof, wherein: Ring A is selected from the group consisting of cycloalkyl, heterocyclic, aryl and heteroaryl; Ring B 1is a 7-12-membered cycloalkyl group, a 7-12-membered heterocyclic group, a 7-12-membered aryl group, or a 7-12-membered heteroaryl group; L 1Selected from the group consisting of: a bond, an alkyl group, an alkenyl group, an alkynyl group, a heteroalkyl group, a heteroalkenyl group and a heteroalkynyl group, wherein the alkyl group, the alkenyl group, the alkynyl group, the heteroalkyl group, the heteroalkenyl group and the heteroalkynyl group are optionally substituted with one or more groups independently selected from the group consisting of: a hydroxyl group, a halogen group, a cyano group and an amino group; L 2Select from the group consisting of: key, N(R A), alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted by one or more R BReplace; Where R ASelected from the group consisting of: hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from: hydroxyl, halogen, cyano, amino, alkyl and halogenated alkyl; wherein each R BIndependently selected from the group consisting of hydroxyl, halogen, cyano, amino, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, cycloalkyl, heterocyclo, aryl and heteroaryl; L 3Selected from the group consisting of: a bond, an alkyl group, an alkenyl group, an alkynyl group, a heteroalkyl group, a heteroalkenyl group and a heteroalkynyl group, wherein the alkyl group, the alkenyl group, the alkynyl group, the heteroalkyl group, the heteroalkenyl group and the heteroalkynyl group are optionally substituted with one or more groups independently selected from the group consisting of: a hydroxyl group, a halogen group, a cyano group and an amino group; L 4Choose from O, S or N(R C); Where R CSelected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, halogenated alkyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl; Each R 1Independently selected from the group consisting of: hydroxyl, halogen, cyano, amino, -N(R D) 2, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted by one or more R EReplace; where each R DIndependently selected from the group consisting of: hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the following: hydroxyl, halogen, cyano,  -N(R F) 2OR G; where each R EIndependently selected from the group consisting of: hydrogen, hydroxyl, halogen, cyano, amino, -N(R F) 2、-alkyl-N(R F) 2、-C(O)OR G, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the following: hydroxyl, halogen, cyano, amino and alkyl; wherein R Fand R GEach of R is independently selected from the group consisting of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl; Each R 2Independently selected from the group consisting of: hydrogen, hydroxyl, halogen, cyano, amino, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from: hydroxyl, halogen, cyano, amino, alkyl and halogenated alkyl; Each R 3Selected from the group consisting of: hydrogen, halogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl and heterocyclo, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl and heterocyclo are optionally substituted with one or more groups independently selected from: hydroxyl, halogen, cyano, amino, alkyl and halogenated alkyl; m is an integer from 0 to 5; n is an integer from 0 to 4; and p is an integer from 0 to 3. In some embodiments, Ring A is aryl. In certain embodiments, Ring A is C 6-12Aryl, C 6-11Aryl, C 6-10Aryl, C 6-9Aryl or C 6-8Aryl. In some embodiments, Ring A is phenyl. In some embodiments, Ring A is heteroaryl. In some embodiments, Ring A is 5-12 membered heteroaryl, 5-11 membered heteroaryl, 5-10 membered heteroaryl, 5-9 membered heteroaryl, 5-8 membered heteroaryl, 5-7 membered heteroaryl or 5-6 membered heteroaryl. In some embodiments, Ring A is selected from the group consisting of furanyl, phenylthio, pyrrolyl, pyridyl, pyranyl, pyrimidinyl, oxazinyl, pyrazinyl and tetrahydroisoquinolinyl. In some embodiments, Ring B 1is a 7-12 membered heteroaryl group. In certain embodiments, ring B 1is pyrazolopyridinyl. In certain embodiments, ring B 1yes . In some implementations, L 1is a key. In some implementations, L 1is an alkyl group. In certain embodiments, L 1It is C 1-6Alkyl, C 1-5Alkyl, C 1-4Alkyl, C 1-3Alkyl or C 1-2Alkyl. In certain embodiments, L 1Choose from a group consisting of: and , where L 1*end and L 2connection. In some embodiments, L 2is a key. In some implementations, L 2It is N(R A), and R ASelected from alkyl or heterocyclic groups, wherein the alkyl or heterocyclic group is optionally substituted by one or more halogens or alkyl groups. In certain embodiments, L 2It is N(R A), and R Ais ethyl, difluoroethyl, trifluoroethyl or cyclohexane. In some embodiments, L 2is optionally represented by one or more R BSubstituted cycloalkyl. In certain embodiments, L 2It is C 3-10Cycloalkyl, C 3-9Cycloalkyl, C 3-8Cycloalkyl, C 3-7Cycloalkyl, C 3-6Cycloalkyl or C 3-5Cycloalkyl, each of which is optionally substituted by one or more R BReplace. In some embodiments, L 2is optionally represented by one or more R Breplaced . In some implementations, L 2is optionally represented by one or more R BSubstituted heterocyclic group. In certain embodiments, L 2is a heterocyclic group containing one or more heteroatoms selected from N, O or S. In certain embodiments, L 2is a 3-12-membered heterocyclic group, a 3-11-membered heterocyclic group, a 3-10-membered heterocyclic group or a 3-9-membered heterocyclic group, each of which is optionally substituted by one or more R BReplace. In some embodiments, L 2is a heterocyclic group selected from the group consisting of: and , each of which is optionally represented by one or more R BReplacement, where L 2*end and L 3connection. In some embodiments, L 2is one or more R BSubstituted cycloalkyl or heterocyclic group, and R Bis an alkyl group. In certain embodiments, R BIt is C 1-6Alkyl, C 1-5Alkyl, C 1-4Alkyl, C 1-3Alkyl or C 1-2Alkyl. In certain embodiments, R Bis methyl. In some embodiments, L 2Choose from a group consisting of: and , where L 2* end and L 3connection. In some embodiments, L 3is an alkyl group. In some embodiments, L 3It is C 1-6Alkyl, C 1-5Alkyl, C 1-4Alkyl, C 1-3Alkyl or C 1-2Alkyl. In certain embodiments, L 3is ethyl. In some embodiments, L 4is O or NH. In some embodiments, L 1is the key, and L 2is a key or optionally replaced by one or more R BSubstituted heterocyclic group. In some embodiments, L 1is an alkyl group, and L 2Yes key, N(R A) or optionally one or more R BSubstituted cycloalkyl. In some embodiments, -L 1-L 2-L 3-L 4- is -alkyl-O-. In certain embodiments, -L 1-L 2-L 3-L 4-Yes-(C 3-8Alkyl)-O-. In certain embodiments, -L 1-L 2-L 3-L 4-Yes-(CH 2) 5-O-, -CH 2CH(CH 3)(CH 2) 3-O- or -CH 2CH(CH 2CH 3)(CH 2) 3-O-. In some implementations, -L 1-L 2-L 3-L 4-is-heterocyclic-alkyl   -O-, wherein the heterocyclic group is optionally replaced by one or more RBReplace. In some embodiments, -L 1-L 2-L 3-L 4- is -(5-10 membered heterocyclic group)-(C 1-6Alkyl)-O-, wherein the 5-10 membered heterocyclic group is optionally replaced by one or more R BReplace. In some embodiments, -L 1-L 2-L 3-L 4-Choose from a group consisting of: as well as . In some implementations, -L 1-L 2-L 3-L 4- is - alkyl-N(R A)-alkyl-O-, where R ASelected from alkyl or heterocyclic groups, wherein the alkyl or heterocyclic group is optionally substituted by one or more halogens or alkyl groups. In certain embodiments, -L 1-L 2-L 3-L 4-Yes-(C 1-6Alkyl)-N(R A)-(C 1-6Alkyl)-O-, where RA is selected from C 1-6Alkyl or 3- to 6-membered heterocyclic group, wherein the alkyl or heterocyclic group is optionally substituted by one or more halogens or C1-6Alkyl substitution. In certain embodiments, -L 1-L 2-L 3-L 4-yes or . In some implementations, -L 1-L 2-L 3-L 4-is-alkyl-cycloalkyl-alkyl-O-, wherein the cycloalkyl is optionally replaced by one or more RBReplace. In some embodiments, -L 1-L 2-L 3-L 4-Yes-(C 1-6Alkyl)-(C 3-6Cycloalkyl)-(C 1-6Alkyl)-O-, wherein the C 3-6The cycloalkyl group is optionally replaced by one or more RBReplace. In some embodiments, -L 1-L 2-L 3-L 4-yes . In some implementations, R 1is a halogen and m is 1. In some embodiments, R 1Yes-N(R D) 2, and m is 1. In some embodiments, R 1Yes-N(R D) 2, and each R DIndependently is optionally selected by one or more independently selected from -N(R F) 2OR GAn alkyl group substituted with a group. In certain embodiments, R 1Yes-N(R D) 2, and each R Dis arbitrarily selected by one or more independently selected from -N(R F) 2OR GC 1-6Alkyl, C 1-5Alkyl, C 1-4Alkyl, C 1-3Alkyl or C 1-2Alkyl. In certain embodiments, R 1Yes-N(R D) 2, and each R Dare independently methyl, methoxyethyl, N,N-dimethylaminoethyl or N,N-dimethylaminopropyl. In some embodiments, R 1is optionally represented by one or more R Esubstituted heterocyclic group, and m is 1. In certain embodiments, R 1is optionally represented by one or more R Esubstituted 3- to 10-membered heterocyclic group, and m is 1. In certain embodiments, R 1Choose from a group consisting of: and , each of which is optionally represented by one or more R EReplace. In some embodiments, R Eis a halogen. In certain embodiments, R Eis F. In some embodiments, R EYes-N(R F) 2、-alkyl-N(R F) 2、 -C(O)OR GOr an alkyl group optionally substituted with one or more halogens. In some embodiments, R Fand R GEach of R is independently an alkyl group. In certain embodiments, R Fand R GEach of them is independently C 1-6Alkyl, C 1-5Alkyl, C 1-4Alkyl, C 1-3Alkyl or C 1-2Alkyl. In certain embodiments, R Fand R GEach of them is independently C 1-3Alkyl. In certain embodiments, R ESelect from the group consisting of:     -N(CH 3) 2、-C 1-3Alkyl-N(CH 3) 2, -C(O)O(tert-butyl), methyl, ethyl and trifluoroethyl. In certain embodiments, R Eis a cycloalkyl or heterocyclic group, wherein the cycloalkyl and heterocyclic groups are optionally substituted with one or more alkyl and/or halogen groups. In certain embodiments, R ESelected from cyclopropyl, morpholinyl, piperazinyl, oxacyclobutane or azocyclobutane, each of which is optionally substituted with one or more alkyl and/or halogen. In certain embodiments, R ESelected from cyclopropyl, morpholinyl, piperazinyl, oxacyclobutane or azocyclobutane, each of which is optionally substituted with one or more alkyl and/or halogen. In certain embodiments, R 2is a halogen, and n is 1 or 2. In certain embodiments, R 2is F. In some embodiments, R 2is an alkyl or cycloalkyl group, wherein the alkyl and cycloalkyl groups are optionally substituted with one or more or halogens. In certain embodiments, R2It is C 1-6Alkyl or C 3-6Cycloalkyl, wherein the C1-6Alkyl and C 3-6The cycloalkyl group is optionally substituted with one or more halogens. In certain embodiments, R2is methyl, ethyl, cyclopropyl or trifluoroethyl. In some embodiments, yes . In some implementations, R 3is an alkyl group or a cycloalkyl group. In certain embodiments, R 3It is C 1-6Alkyl or C 3-6Cycloalkyl. In certain embodiments, R 3is methyl, ethyl or cyclopropyl. In some embodiments, p is 1, and R 3is an alkyl group. In certain embodiments, p is 1, and R 3It is C 1-6Alkyl, C 1-5Alkyl, C 1-4Alkyl, C 1-3Alkyl or C 1-2Alkyl. In certain embodiments, p is 1, and R 3is methyl. In some embodiments, yes . In some implementations, yes . In some embodiments, m is 0, 1 or 2. In some embodiments, n is 0 or 1. In some embodiments, p is 1. On the other hand, the present disclosure provides a compound of formula (IV): (IV) or a pharmaceutically acceptable salt thereof, wherein: Ring A is selected from the group consisting of cycloalkyl, heterocyclic, aryl and heteroaryl; Ring B is selected from the group consisting of cycloalkyl, heterocyclic, aryl and heteroaryl; L 1Selected from the group consisting of: a bond, an alkyl group, an alkenyl group, an alkynyl group, a heteroalkyl group, a heteroalkenyl group and a heteroalkynyl group, wherein the alkyl group, the alkenyl group, the alkynyl group, the heteroalkyl group, the heteroalkenyl group and the heteroalkynyl group are optionally substituted with one or more groups independently selected from the group consisting of: a hydroxyl group, a halogen group, a cyano group and an amino group; L twenty oneis a 7-12 membered cycloalkyl or a 7-12 membered heterocyclic group, wherein the cycloalkyl and heterocyclic groups are optionally substituted by one or more R BReplace; where each R BIndependently selected from the group consisting of hydroxyl, halogen, cyano, amino, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, cycloalkyl, heterocyclo, aryl and heteroaryl; L 3Selected from the group consisting of: a bond, an alkyl group, an alkenyl group, an alkynyl group, a heteroalkyl group, a heteroalkenyl group and a heteroalkynyl group, wherein the alkyl group, the alkenyl group, the alkynyl group, the heteroalkyl group, the heteroalkenyl group and the heteroalkynyl group are optionally substituted with one or more groups independently selected from the group consisting of: a hydroxyl group, a halogen group, a cyano group and an amino group; L 4Choose from O, S or N(R C); Where R CSelected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, halogenated alkyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl; Each R 1Independently selected from the group consisting of: hydroxyl, halogen, cyano, amino, -N(R D) 2, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted by one or more R EReplace; where each R DIndependently selected from the group consisting of: hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the following: hydroxyl, halogen, cyano,  -N(R F) 2OR G; where each R EIndependently selected from the group consisting of: hydrogen, hydroxyl, halogen, cyano, amino, -N(R F) 2、-alkyl-N(R F) 2、-C(O)OR G, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the following: hydroxyl, halogen, cyano, amino and alkyl; wherein R Fand R GEach of R is independently selected from the group consisting of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl; Each R 2Independently selected from the group consisting of: hydrogen, hydroxyl, halogen, cyano, amino, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from: hydroxyl, halogen, cyano, amino, alkyl and halogenated alkyl; R 3Selected from the group consisting of: hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl and heterocyclo, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl and heterocyclo are optionally substituted with one or more groups independently selected from: hydroxyl, halogen, cyano, amino, alkyl and halogenated alkyl; m is an integer from 0 to 5; n is an integer from 0 to 4; and p is an integer from 0 to 3. In some embodiments, Ring A is aryl. In certain embodiments, Ring A is C 6-12Aryl, C 6-11Aryl, C 6-10Aryl, C 6-9Aryl or C 6-8In some embodiments, Ring A is phenyl. In some embodiments, Ring A is heteroaryl. In some embodiments, Ring A is 5-12 membered heteroaryl, 5-11 membered heteroaryl, 5-10 membered heteroaryl, 5-9 membered heteroaryl, 5-8 membered heteroaryl, 5-7 membered heteroaryl or 5-6 membered heteroaryl. In some embodiments, Ring A is selected from the group consisting of furanyl, phenylthio, pyrrolyl, pyridyl, pyranyl, pyrimidinyl, oxazinyl, pyrazinyl and tetrahydroisoquinolinyl. In some embodiments, Ring B is aryl. In some embodiments, Ring B is C 6-12Aryl, C 6-11Aryl, C 6-10Aryl, C 6-9Aryl or C 6-8Aryl. In some embodiments, Ring B is phenyl. In some embodiments, Ring B is heteroaryl. In some embodiments, Ring B is 5-12 membered heteroaryl, 5-11 membered heteroaryl, 5-10 membered heteroaryl, 5-9 membered heteroaryl, 5-8 membered heteroaryl, 5-7 membered heteroaryl or 5-6 membered heteroaryl. In some embodiments, Ring B is pyridyl or pyrazolyl. In some embodiments, Ring B is selected from the group consisting of: and . In some implementations, L 1is a key. In some implementations, L twenty oneis a 7-10 membered heterocyclic group containing one or more heteroatoms selected from N or O. In certain embodiments, L twenty oneChoose from a group consisting of: and , where L twenty one*end and L 3connection. In some embodiments, L 3is an alkyl group. In certain embodiments, L 3It is C 1-6Alkyl, C 1-5Alkyl, C 1-4Alkyl, C 1-3Alkyl or C 1-2Alkyl. In certain embodiments, L 3is ethyl. In some embodiments, L 4is O or NH. In some embodiments, -L 1-L twenty one-L 3-L 4-is-heterocyclic-alkyl-O-, wherein the heterocyclic group is optionally replaced by one or more RBReplace. In some embodiments, -L 1-L twenty one-L 3-L 4- is -(5-10 membered heterocyclic group)-(C 1-6Alkyl)-O-, wherein the 5-10 membered heterocyclic group is optionally replaced by one or more R BReplace. In some implementations, -L 1-L twenty one-L 3-L 4-Choose from a group consisting of: and . In some implementations, R 1is a halogen and m is 1. In some embodiments, R 1Yes-N(R D) 2, and m is 1. In some embodiments, R 1Yes-N(R D) 2, and each R DIndependently is optionally selected by one or more independently selected from -N(R F) 2OR GAn alkyl group substituted with a group. In certain embodiments, R 1Yes-N(R D) 2, and each R Dis arbitrarily selected by one or more independently selected from -N(R F) 2OR GC 1-6Alkyl, C 1-5Alkyl, C 1-4Alkyl, C 1-3Alkyl or C 1-2Alkyl. In certain embodiments, R 1Yes-N(R D) 2, and each R Dare independently methyl, methoxyethyl or N,N-dimethylaminopropyl. In some embodiments, R 1is optionally represented by one or more R Esubstituted heterocyclic group, and m is 1. In certain embodiments, R 1is optionally represented by one or more R Esubstituted 3- to 10-membered heterocyclic group, and m is 1. In certain embodiments, R 1Choose from a group consisting of: and , each of which is optionally represented by one or more R EReplace. In some embodiments, R Eis a halogen. In certain embodiments, R Eis F. In some embodiments, R EYes-N(R F) 2、-alkyl-N(R F) 2、 -C(O)OR Gor an alkyl group optionally substituted with one or more halogens. In some embodiments, R Fand R GEach of R is independently an alkyl group. In certain embodiments, R Fand R GEach of them is independently C 1-6Alkyl, C 1-5Alkyl, C 1-4Alkyl, C 1-3Alkyl or C 1-2Alkyl. In certain embodiments, R Fand R GEach of them is independently C 1-3Alkyl. In certain embodiments, R ESelect from the group consisting of:     -N(CH 3) 2、-C 1-3Alkyl-N(CH 3) 2, -C(O)O(tert-butyl), methyl, ethyl and trifluoroethyl. In certain embodiments, R Eis a cycloalkyl or heterocyclic group, wherein the cycloalkyl and heterocyclic groups are optionally substituted with one or more alkyl and/or halogen groups. In certain embodiments, R ESelected from cyclopropyl, morpholinyl, piperazinyl, oxacyclobutane or azocyclobutane, each of which is optionally substituted with one or more alkyl and/or halogen. In certain embodiments, R ESelected from cyclopropyl, morpholinyl, piperazinyl, oxacyclobutenyl or azocyclobutanyl, each of which is optionally substituted with one or more alkyl and/or halogen. In certain embodiments, R 1Choose from a group consisting of: and . In some implementations, R 2is a halogen, and n is 1 or 2. In certain embodiments, R 2is F. In some embodiments, R 2is an alkyl or cycloalkyl group, wherein the alkyl and cycloalkyl groups are optionally substituted with one or more or halogens. In certain embodiments, R2It is C 1-6Alkyl or C 3-6Cycloalkyl, wherein the C1-6Alkyl and C 3-6The cycloalkyl group is optionally substituted with one or more halogens. In certain embodiments, R2is methyl, ethyl, cyclopropyl or trifluoroethyl. In certain embodiments, R 2is methyl. In some embodiments, R 3is an alkyl group. In certain embodiments, R 3Each of them is independently C 1-6Alkyl, C 1-5Alkyl, C 1-4Alkyl, C 1-3Alkyl or C 1-2Alkyl. In certain embodiments, R 3is methyl. In some embodiments, p is 1, and R 3is an alkyl group. In certain embodiments, p is 1, and R 3It is C 1-6Alkyl, C 1-5Alkyl, C 1-4Alkyl, C 1-3Alkyl or C 1-2Alkyl. In certain embodiments, p is 1, and R 3is methyl. In some embodiments, yes . In some implementations, yes . In some embodiments, m is 1. In some embodiments, n is 1. In some embodiments, p is 1. Exemplary compounds of the present disclosure are described below. , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , and . The compounds provided herein are described with reference to both general formulae and specific compounds. In addition, the compounds disclosed herein may exist in a variety of different forms or derivatives, including but not limited to prodrugs, active metabolic derivatives (active metabolites), solvates, pharmaceutically acceptable salts or isotope derivatives, all of which are within the scope of the present disclosure. As used herein, the term "prodrug" refers to a compound or a pharmaceutically acceptable salt thereof that produces the desired active compound when metabolized or converted by solvent decomposition under physiological conditions. Prodrugs include but are not limited to esters, amides, carbamates, carbonates, ureas, solvates or hydrates of active compounds. Typically, prodrugs are inactive or less active than active compounds, but can provide one or more advantageous handling, administration and/or metabolic properties. For example, some prodrugs are esters of active compounds; during metabolism, the ester group is cleaved to produce the active drug. In addition, some prodrugs are enzymatically activated to generate active compounds or compounds that generate active compounds after further chemical reactions. Prodrugs can be developed from prodrug forms to active forms in a single step, or can have one or more intermediate forms that may or may not be active themselves. The preparation and use of prodrugs are discussed in the following references: T. Higuchi and V. Stella, "Pro-drugs as Novel Delivery Systems", Volume 14 of the A.C.S. Symposium Series, Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987; Prodrugs: Challenges and Rewards, eds. V. Stella, R. Borchardt, M. Hageman, R. Oliyai, H. Maag, J. Tilley, Springer Verlag New York, 1991. 2007, all of which are hereby incorporated by reference in their entirety. As used herein, the term "metabolite", e.g., active metabolite, overlaps with prodrugs as described above. Thus, such metabolites are pharmacologically active compounds or compounds that are further metabolized to pharmacologically active compounds that are derivatives produced by metabolic processes in the subject. For example, such metabolites can be produced by oxidation, reduction, hydrolysis, amidation, deamidation, esterification, deesterification, enzymatic cleavage, etc. of the administered compound or salt or prodrug. Among them, the active metabolite is such a pharmacologically active derivative compound. For prodrugs, the prodrug compound is generally inactive or less active than the metabolite. For active metabolites, the parent compound can be an active compound or can be an inactive prodrug. Prodrugs and active metabolites can be identified using conventional techniques known in the art. See, e.g., Bertolini et al., 1997, J Med Chem 40:2011-2016; Shan et al., J Pharm Sci 86:756-757; Bagshawe, 1995, Drug Dev Res 34:220-230; Wermuth, supra. As used herein, the term "pharmaceutically acceptable" indicates that a substance or composition is chemically and/or toxicologically compatible with the other ingredients making up the formulation and/or the subject being treated. As used herein, unless otherwise indicated, the term "pharmaceutically acceptable salt" includes salts that retain the biological effectiveness of the free acid and base of the specified compound and are not biologically or otherwise undesirable. Pharmaceutically acceptable salt forms contemplated include, but are not limited to, monosalts, disalts, trisalts, tetrasalts, and the like. Pharmaceutically acceptable salts are nontoxic in the amounts and concentrations in which they are administered. The preparation of such salts can facilitate pharmacological use by altering the physical properties of the compound without interfering with its ability to exert its physiological effects. Useful changes in physical properties include lowering the melting point to facilitate transmucosal administration and increasing solubility to facilitate administration of higher concentrations of the drug. Pharmaceutically acceptable salts include acid addition salts, such as the following acid addition salts: sulfate, chloride, hydrochloride, fumarate, maleate, phosphate, sulfamate, acetate, citrate, lactate, tartrate, methanesulfonate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate, cyclohexylaminosulfonate and quinate. Pharmaceutically acceptable salts can be obtained from acids such as hydrochloric acid, maleic acid, sulfuric acid, phosphoric acid, sulfamate, acetic acid, citric acid, lactic acid, tartaric acid, malonic acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, cyclohexylaminosulfonate, fumaric acid and quinic acid. When an acidic functional group such as carboxylic acid or phenol is present, pharmaceutically acceptable salts also include base addition salts, such as base addition salts including benzathine, chloroprocaine, choline, diethanolamine, ethanolamine, tert-butylamine, ethylenediamine, meglumine, procaine, aluminum, calcium, lithium, magnesium, potassium, sodium, ammonium, alkylamines and zinc. See, e.g., Remington's Pharmaceutical Sciences, 19th ed., Mack Publishing Co., Easton, PA, Vol. 2, p. 1457, 1995; Handbook of Pharmaceutical Salts: Properties, Selection, and Use, Stahl and Wermuth, Wiley-VCH, Weinheim, Germany, 2002. Such salts may be prepared using the appropriate corresponding base. Pharmaceutically acceptable salts may be prepared by standard techniques. For example, the free base form of the compound may be dissolved in a suitable solvent (e.g., an aqueous or aqueous-alcoholic solution containing an appropriate acid) and then isolated by evaporation of the solution. Thus, if a particular compound is a base, the desired pharmaceutically acceptable salt may be prepared by any suitable method available in the art, for example, by treating the free base with an inorganic acid such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, or an organic acid such as acetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, or a pyranosyl acid such as glucuronic acid or galacturonic acid; an α-hydroxy acid such as citric acid or tartaric acid; an amino acid such as aspartic acid or glutamic acid; an aromatic acid such as benzoic acid or cinnamic acid; a sulfonic acid such as p-toluenesulfonic acid or ethanesulfonic acid; and the like. Similarly, if the particular compound is an acid, the desired pharmaceutically acceptable salt may be prepared by any suitable method, for example, by treating the free acid with an inorganic or organic base such as an amine (primary, secondary or tertiary), an alkali metal hydroxide or an alkali earth metal hydroxide, etc. Illustrative examples of suitable salts include organic salts derived from amino acids such as L-glycine, L-lysine and L-arginine; ammonia, primary, secondary and tertiary amines; and cyclic amines such as hydroxyethylpyrrolidine, piperidine, morpholine or piperazine; and inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum and lithium. It is also understood that the compounds of the present disclosure may exist in non-solvated forms, solvated forms (e.g., hydrated forms), and solid forms (e.g., crystalline forms or polycrystalline forms), and the present disclosure is intended to encompass all such forms. As used herein, the term "solvate" or "solvated form" refers to a solvent addition form that includes a stoichiometric or non-stoichiometric amount of solvent. Some compounds have a tendency to capture a fixed molar ratio of solvent molecules in the crystalline solid state, thereby forming a solvate. If the solvent is water, the solvate formed is a hydrate, and if the solvent is an alcohol, the solvate formed is an alcoholate. Hydrates are formed by the addition of one or more water molecules to a solid state in which the water retains its hydrogen bond as H 2O molecular state of a substance to form a molecular combination. Examples of solvents that form solvates include but are not limited to water, isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid and ethanolamine. The present disclosure is also intended to include all isotopic forms of the compounds provided herein. Isotopes of atoms include atoms with the same atomic number but different mass numbers. For example, unless otherwise stated, hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine, bromine or iodine in the compounds of the present disclosure is meant to also include its isotopes, such as but not limited to 1H. 2H. 3H. 11C. 12C. 13C. 14C. 14N. 15N. 16O. 17O. 18O. 31P. 32P. 32S. 33S. 34S. 36S. 17F. 18F. 19F. 35Cl, 37Cl, 79Br, 81Br, 124I. 127I and 131I. In some embodiments, hydrogen includes protium, deuterium, and tritium. In some embodiments, carbon includes 12C and 13C. The compounds provided herein or their pharmaceutically acceptable salts may contain one or more asymmetric centers, thus giving rise to enantiomers, diastereomers and other stereoisomeric forms, which may be defined by absolute stereochemistry as (R)- or (S)- or (D)- or (L)- of amino acids, or by relative configuration as rel-(R)- or rel-(S)-. The present disclosure includes all such possible isomers, as well as racemic and optically pure forms thereof. Optically active (+) and (-), (R)- and (S)- or (D)- and (L)-isomers may be prepared by chiral synthons or chiral reagents or resolved using conventional techniques, such as chromatography and fractional crystallization. Traditional techniques for preparing and separating individual enantiomers include chiral synthesis from suitable optically pure precursors or resolution of racemates (or racemates of salts or derivatives) using, for example, chiral high pressure liquid chromatography (HPLC). When a compound is represented in its chiral form, it should be understood that the embodiments include but are not limited to specific diastereomers or enantiomerically enriched forms. Where chirality is not specified but exists, it should be understood that the embodiments are intended to include specific diastereomers or enantiomerically enriched forms; or racemic or non-scalemic mixtures of such compounds. The term "stereoisomer" refers to compounds containing the same atoms connected by the same bonds but with different three-dimensional structures, which are not interchangeable. The present disclosure contemplates various stereoisomers and mixtures thereof and includes "enantiomers," which refers to two stereoisomers whose molecules are non-superimposable mirror images of each other. The term "enantiomers" refers to a pair of stereoisomers that are non-superimposable mirror images of each other. A 1:1 mixture of a pair of enantiomers is a "racemic" mixture. A mixture of enantiomers in a ratio other than 1:1 is a "non-racemic" mixture. The term "diastereomer" refers to stereoisomers that have at least two asymmetric atoms but are not mirror images of each other. The term "tautomer" or "tautomeric form" refers to structural isomers of different energies that can interconvert via low energy barriers. The presence and concentration of isomeric forms will depend on the environment in which the compound is located and may differ, for example, whether the compound is a solid or in an organic or aqueous solution. For example, proton tautomers (also called prototropic tautomers) include interconversions by proton migration, such as keto-enol, amide-imidic acid, lactam-lactimide, imine-enamine isomerizations, and cyclic forms in which protons can occupy two or more positions in heterocyclic systems. Valence tautomers include interconversions by reorganization of some of the bonding electrons. Tautomers may be in equilibrium or sterically locked into one form by appropriate substitution. Unless otherwise indicated, compounds disclosed herein that are identified by name or structure as a particular tautomeric form are intended to include other tautomeric forms. When the compounds provided herein contain olefinic double bonds or other centers of geometric asymmetry, and unless otherwise indicated, the compounds include both E and Z geometric isomers. Synthesis Methods The compounds provided herein can be prepared using any known organic synthesis technique and can be synthesized according to any of a number of possible synthetic routes. The reactions used to prepare the compounds disclosed herein can be carried out in suitable solvents that can be readily selected by those skilled in the art of organic synthesis. Suitable solvents can be substantially non-reactive with starting materials (reactants), intermediates or products at temperatures at which the reaction is carried out, e.g., temperatures that can range from the freezing temperature of the solvent to the boiling temperature of the solvent. A given reaction can be carried out in one solvent or a mixture of more than one solvent. Depending on the particular reaction step, a suitable solvent for a particular reaction step can be selected by a person skilled in the art. The preparation of the compounds disclosed herein can involve the protection and deprotection of various chemical groups. The need for protection and deprotection and the selection of appropriate protecting groups can be readily determined by a person skilled in the art. The chemistry of protecting groups can be found in, for example, T. W. Greene and P. G. M. Wuts, Protective Groups in Organic Synthesis, 3rd ed., John Wiley & Sons, New York, (1999); P. Kocienski, Protecting Groups, Georg Thieme Verlag, 2003; and Peter G. M. Wuts, Greene's Protective Groups in Organic Synthesis, 5th ed., Wiley, 2014, which are incorporated herein by reference in their entirety. The reaction can be monitored according to any suitable method known in the art. For example, the reaction can be monitored by methods such as nuclear magnetic resonance spectroscopy (e.g., 1H or 13 C), infrared spectroscopy, spectrophotometry (e.g., UV visible), mass spectrometry or by chromatographic methods such as high performance liquid chromatography (HPLC), liquid chromatography-mass spectrometry (LCMS) or thin layer chromatography (TLC) to monitor product formation. Those skilled in the art can purify the compound by various methods, including high performance liquid chromatography (HPLC) ("Preparative LC-MS Purification: Improved Compound Specific Method Optimization" Karl F. Blom, Brian Glass, Richard Sparks, Andrew P. Combs "Journal of Combi. Chem. (Journal of Combi. Chem.) 2004, 6 (6), 874-883, the reference is incorporated herein by reference in its entirety) and normal phase silica gel chromatography. Pharmaceutical Compositions In another aspect, a pharmaceutical composition is provided, which comprises one or more compounds disclosed herein or a pharmaceutically acceptable salt thereof. In some embodiments, the pharmaceutical composition disclosed herein comprises the first compound or its pharmaceutically acceptable salt provided herein and one or more other compounds of the same formula, but the first compound and the other compound are not the same molecule. On the other hand, a pharmaceutical composition is provided, which comprises one or more compounds disclosed herein or its pharmaceutically acceptable salt and at least one pharmaceutically acceptable excipient. In some embodiments, the pharmaceutical composition disclosed herein comprises a therapeutically effective amount of one or more compounds disclosed herein or its pharmaceutically acceptable salt and at least one pharmaceutically acceptable excipient. In some embodiments, the pharmaceutical composition disclosed herein comprises a therapeutically effective amount of one or more compounds disclosed herein or its pharmaceutically acceptable salt and at least one pharmaceutically acceptable excipient. As used herein, the term "therapeutically effective amount" refers to the amount of a molecule, compound, or composition comprising the molecule or compound that treats, ameliorates, or prevents an identified disease or condition, or shows a detectable therapeutic or inhibitory effect. The effect can be detected by any assay method known in the art. The exact effective amount for a subject will depend on the subject's weight, size, and health; the nature and extent of the condition; the rate of administration; the treatment or combination of treatments selected for administration; and the judgment of the prescribing physician. The therapeutically effective amount for a given situation can be determined by routine experiments within the skill and judgment of the clinician. As used herein, the term "drug composition" refers to a formulation comprising a molecule or compound disclosed herein in a form suitable for administration to a subject. The pharmaceutical composition comprises a composition suitable for oral administration, rectal administration, topical administration, parenteral administration (including subcutaneous, intramuscular and intravenous), sublingual administration, ocular administration, transdermal administration or nasal administration, but the most suitable route in any given case will depend on the nature and severity of the particular host and the condition to which the active ingredient is administered. The pharmaceutical composition can be conveniently presented in unit dosage form and can be prepared by any method well known in the pharmaceutical art. As used herein, the term "pharmaceutically acceptable excipient" means an excipient that can be used to prepare a generally safe, non-toxic and biologically and otherwise desirable pharmaceutical composition, and includes excipients that are acceptable for veterinary use as well as human pharmaceutical use. As used herein, "pharmaceutically acceptable excipients" include one and more than one such excipients. The term "pharmaceutically acceptable excipients" also encompasses "pharmaceutically acceptable carriers" and "pharmaceutically acceptable diluents". The specific excipient used will depend on the means and purpose for which the compounds disclosed herein are applied. Solvents are typically selected based on solvents that are considered safe by those skilled in the art for administration to mammals, including humans. Typically, safe solvents are non-toxic aqueous solvents, such as water and other non-toxic solvents that are soluble or miscible in water. Suitable aqueous solvents include water, ethanol, propylene glycol, polyethylene glycol (e.g., PEG 400, PEG 300), and the like, and mixtures thereof. In some embodiments, suitable excipients may include buffers such as phosphates, citrates, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzylammonium chloride; hexamethonium chloride; benzoyl chloride; benzethonium chloride; phenol, butyl alcohol, or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; o-catechin; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins such as serum albumin, gelatin, or immunoglobulin; immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine or lysine; monosaccharides, disaccharides and other carbohydrates including glucose, mannose or dextran; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counterions such as sodium; metal complexes (e.g., Zn protein complexes); and/or non-ionic surfactants such as TWEEN™, PLURONICS™ or polyethylene glycol (PEG). In some embodiments, suitable excipients may include one or more stabilizers, surfactants, wetting agents, lubricants, emulsifiers, suspending agents, preservatives, antioxidants, opacifiers, glidants, processing aids, coloring agents, sweeteners, aromas, flavoring agents and other known additives to provide the best presentation form of the drug (i.e., the compounds disclosed herein or their pharmaceutical compositions) or to assist in the manufacture of the drug product (i.e., the drug). The active pharmaceutical ingredient can also be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, such as hydroxymethylcellulose or gelatin microcapsules and poly-(methyl methacrylate) microcapsules, respectively, in colloidal drug delivery systems (e.g., liposomes, albumin microspheres, microemulsions, nanoparticles and nanocapsules) or in macroemulsions. Such techniques are disclosed in Remington's Pharmaceutical Sciences, 16th edition, Osol, A. ed. (1980). "Liposomes" are small vesicles comprising various types of lipids, phospholipids and/or surfactants that can be used to deliver drugs (such as the compounds disclosed herein and optionally chemotherapeutic agents) to mammals, including humans. The components of liposomes are usually arranged in a double layer, similar to the lipid arrangement of biological membranes. The pharmaceutical composition provided herein can be in a form that allows subjects, including but not limited to people, to use the composition and allows the composition to be formulated into any form compatible with the expected route of administration. A variety of approaches have been considered for the pharmaceutical composition provided herein, and therefore the pharmaceutical composition provided herein can be supplied in bulk or unit dosage form depending on the expected route of administration. For example, for oral, buccal and sublingual administration, powders, suspensions, granules, tablets, pills, capsules, soft capsules and caplets can be acceptable as solid dosage forms, and emulsions, syrups, elixirs, suspensions and solutions can be acceptable as liquid dosage forms. For injection, emulsions and suspensions may be acceptable as liquid dosage forms, and powders suitable for reconstitution with a suitable solution may be acceptable as solid dosage forms. For inhalation, solutions, sprays, dry powders and aerosols may be acceptable dosage forms. For topical (including buccal and sublingual) or transdermal administration, powders, sprays, ointments, pastes, creams, lotions, gels, solutions and patches may be acceptable dosage forms. For vaginal administration, vaginal suppositories, tampons, creams, gels, pastes, foams and sprays may be acceptable dosage forms. In some embodiments, the disclosed pharmaceutical composition may be in the form of a formulation for oral administration. In certain embodiments, the pharmaceutical composition of the present disclosure may be in the form of a tablet formulation. Pharmaceutically acceptable excipients suitable for tablet formulations include, for example, inert diluents such as lactose, sodium carbonate, calcium phosphate or calcium carbonate; granulating agents and disintegrants such as corn starch or alginic acid; binders such as starch; lubricants such as magnesium stearate, stearic acid or talc; preservatives such as ethyl or propyl p-hydroxybenzoate; and antioxidants such as ascorbic acid. Tablet formulations may be uncoated or coated to modulate disintegration and subsequent absorption of the active ingredient in the gastrointestinal tract, or to improve stability and/or appearance, in either case using conventional coating agents and procedures well known in the art. In certain embodiments, the pharmaceutical compositions disclosed herein may be in the form of hard gelatin capsules in which the active ingredient is mixed with an inert solid diluent, such as calcium carbonate, calcium phosphate or kaolin; or in the form of soft gelatin capsules in which the active ingredient is mixed with water or oil, such as peanut oil, liquid paraffin or olive oil. In certain embodiments, the pharmaceutical compositions of the present disclosure may be in the form of an aqueous suspension, which generally includes the active ingredient in the form of a fine powder and one or more suspending agents, such as sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethylcellulose, sodium alginate, polyvinylpyrrolidone, tragacanth gum and gum arabic; dispersants or wetting agents, such as lecithin or alkylene oxide and fatty acid. The aqueous suspension may also include one or more preservatives (e.g., ethyl or propyl paraben), antioxidants (e.g., ascorbic acid), coloring agents, flavoring agents, and/or sweeteners (e.g., sucrose, saccharin, or aspartame). In certain embodiments, the pharmaceutical compositions disclosed herein may be in the form of an oily suspension, which typically includes a suspended active ingredient in a vegetable oil (such as peanut oil, castor oil, olive oil, sesame oil, or coconut oil) or in a mineral oil (such as liquid paraffin). The oily suspension may also include a thickener, such as beeswax, hard wax, or cetyl alcohol. Sweeteners (such as those described above) and flavorings may be added to provide a palatable oral formulation. These compositions may be preserved by adding an antioxidant (such as ascorbic acid). In certain embodiments, the pharmaceutical compositions disclosed herein may be in the form of an oil-in-water emulsion. The oil phase may be a vegetable oil, such as olive oil or peanut oil; or a mineral oil, such as liquid paraffin; or a mixture of any of these oils. Suitable emulsifiers can be, for example, naturally occurring glues, such as gum arabic or tragacanth; naturally occurring phospholipids, such as soybeans, lecithin, esters or partial esters derived from fatty acids and hexitol anhydrides (e.g., sorbitan monooleate) and condensation products of the partial esters with ethylene oxide, such as polyoxyethylene sorbitan monooleate. Emulsions can also include sweeteners, flavorings and preservatives. In certain embodiments, the pharmaceutical compositions provided herein can be in the form of syrups and elixirs, which can include sweeteners, such as glycerol, propylene glycol, sorbitol, aspartame or sucrose; softeners; preservatives; flavorings and/or coloring agents. In certain embodiments, the pharmaceutical compositions disclosed herein can be in the form of formulations for injection. In certain embodiments, the pharmaceutical composition disclosed herein can be in the form of a sterile injectable preparation, such as a sterile injectable aqueous or oily suspension. Such a suspension can be prepared using those suitable dispersants or wetting agents and suspending agents mentioned above according to known techniques. The sterile injectable preparation can also be a sterile injectable solution or suspension in a non-toxic parenteral acceptable diluent or solvent, such as a solution in 1,3-butanediol or prepared as a lyophilized powder. Acceptable vehicles and solvents that can be used include water, Ringer's solution, and isotonic sodium chloride solution. In addition, sterile non-volatile oils can be used as solvents or suspension media conventionally. For this purpose, any mild fixed oil can be used, including synthetic monoglycerides or diglycerides. In addition, fatty acids such as oleic acid can be used to prepare injectables. In some embodiments, the disclosed pharmaceutical composition can be in the form of a formulation for inhalation administration. In certain embodiments, the disclosed pharmaceutical composition can be in the form of an aqueous and non-aqueous (e.g., in a fluorocarbon propellant) aerosol, and the aqueous and non-aqueous aerosols include any suitable solvents and optionally other compounds, such as but not limited to stabilizers, antimicrobial agents, antioxidants, pH regulators, surfactants, bioavailability regulators and combinations thereof. Carriers and stabilizers vary with the requirements of the specific compound, but generally include non-ionic surfactants (Tween, Pluronic or polyethylene glycol), harmless proteins (such as serum albumin), sorbitan esters, oleic acid, lecithin, amino acids such as glycine, buffers, salts, sugars or sugar alcohols. In some embodiments, the pharmaceutical compositions disclosed herein may be in the form of formulations for topical or transdermal administration. In certain embodiments, the pharmaceutical composition provided herein can be in the form of creams, ointments, gels and aqueous or oily solutions or suspensions, which can be prepared by mixing active ingredients with conventional local acceptable excipients, such as animal fat and vegetable fat, oil, wax, paraffin, starch, tragacanth, cellulose derivatives, polyethylene glycol, silicone, bentonite, silicic acid, talc and zinc oxide or their mixtures. In certain embodiments, the pharmaceutical composition provided herein can be prepared for ophthalmic administration. In certain embodiments, the pharmaceutical composition provided herein can be in the form of ophthalmic formulations, such as eye ointments, powders, solutions, etc. In certain embodiments, ophthalmic formulations are prepared at a comfortable pH and an appropriate buffer system. In addition to those representative dosage forms described above, pharmaceutically acceptable excipients and carriers are generally known to those skilled in the art and are therefore included in the present disclosure. Such excipients and carriers are described in, for example, the following references: Remingtons Pharmaceutical Sciences, Mack Pub. Co., New Jersey (1991); Remington: The Science and Practice of Pharmacy, ed., University of the Sciences in Philadelphia, 21st ed., LWW (2005), which are incorporated herein by reference. The dosage regimen of the compounds provided herein will vary according to known factors, such as the pharmacodynamic characteristics of a particular agent and its mode and route of administration; the species, age, sex, health condition, physical condition and weight of the recipient; the nature and extent of symptoms; the type of concurrent treatment; the frequency of treatment; the route of administration, the patient's renal and liver function, and the desired effect. A physician or veterinarian can determine and prescribe the effective amount of the drug required to prevent, counter or arrest the progress of the condition. In some embodiments, the pharmaceutical composition of the present disclosure can be formulated so that 0.001 mg/kg body weight/day to 1000 mg/kg body weight/day can be administered, for example, 0.01 mg/kg body weight/day to 800 mg/kg body weight/day, 0.01 mg/kg body weight/day to 700 mg/kg body weight/day, 0.01 mg/kg body weight/day to 600 mg/kg body weight/day, 0.01 mg/kg body weight/day to 500 mg/kg body weight/day, 0.01 mg/kg body weight/day to 400 mg/kg body weight/day, 0.01 mg/kg body weight/day to 300 mg/kg body weight/day, 0.1 mg/kg body weight/day to 200 mg/kg body weight/day, 0.1 mg/kg body weight/day to 150 mg/kg body weight/day, 0.1 mg/kg body weight/day to 100 [00136] The present invention relates to a compound or a pharmaceutically acceptable salt thereof provided herein in an amount of 1 mg/kg body weight/day, 0.5 mg/kg body weight/day to 100 mg/kg body weight/day, 0.5 mg/kg body weight/day to 80 mg/kg body weight/day, 0.5 mg/kg body weight/day to 60 mg/kg body weight/day, 0.5 mg/kg body weight/day to 50 mg/kg body weight/day, 1 mg/kg body weight/day to 50 mg/kg body weight/day, 1 mg/kg body weight/day to 45 mg/kg body weight/day, 1 mg/kg body weight/day to 40 mg/kg body weight/day, 1 mg/kg body weight/day to 35 mg/kg body weight/day, 1 mg/kg body weight/day to 30 mg/kg body weight/day, 1 mg/kg body weight/day to 25 mg/kg body weight/day. In some cases, dosage levels below the lower limit of the aforementioned range may be sufficient, while in other cases, larger doses may be used without causing any harmful side effects, provided that such larger doses are first divided into several small doses for all-day administration. For additional information on the route of administration and dosage regimen, see "Comprehensive Medicinal Chemistry" Volume 5, Chapter 25.3 (Corwin Hansch; Chairman of the Editorial Board), Pergamon Press 1990, which reference is particularly incorporated herein by reference. In some embodiments, the disclosed pharmaceutical composition can be formulated into a single dosage form. The amount of the compound provided herein in a single dosage form will vary according to the subject being treated and the specific mode of administration. In some embodiments, dosage forms suitable for administration may contain about 1 mg to about 1000 mg of active ingredient per dosage unit. In these pharmaceutical compositions, the active ingredient will generally be present in an amount of about 0.1-95% by weight, based on the gross weight of the composition. In some embodiments, the pharmaceutical compositions disclosed herein may be formulated into short-acting, rapid-release, long-acting, and sustained-release forms. Therefore, the pharmaceutical formulations disclosed herein may also be formulated for controlled release or slow release. In some embodiments, a certain dose of the compound provided herein or the pharmaceutical composition provided herein is administered to a subject every day, every other day, every two days, every three days, once a week, twice a week, three times a week, or once every two weeks. If necessary, the effective daily dose of the active compound can be used as a single dose at appropriate intervals in a day, optionally in two, three, four, five, six or more subdoses administered in a unit dosage form. In some embodiments, a certain dose of the compound provided herein or the pharmaceutical composition provided herein is administered for 2 days, 3 days, 5 days, 7 days, 14 days, 21 days, 1 month, 2 months, 2.5 months, 3 months, 4 months, 5 months, 6 months or longer. In other aspects, veterinary compositions are also provided, comprising one or more molecules or compounds disclosed herein or pharmaceutically acceptable salts thereof and veterinary carriers. Veterinary carriers are materials that can be used for the purpose of administering the composition, and can be solid, liquid or gaseous materials that are otherwise inert or acceptable in the veterinary field and compatible with the active ingredient. These veterinary compositions can be administered parenterally, orally or by any other desired route. Pharmaceutical compositions or veterinary compositions can be packaged in various ways depending on the method used to administer the drug. For example, a product for distribution can include a container in which a composition in an appropriate form is stored. Suitable containers are well known to those skilled in the art, and include materials such as bottles (plastic and glass), sachets, ampoules, plastic bags, metal cylinders, and the like. The container may also include an anti-disassembly assembly to prevent easy access to the contents of the package. In addition, the container has a label placed thereon describing the contents of the container. The label may also include appropriate warnings. The composition may also be packaged in unit dose or multi-dose containers, such as sealed ampoules and vials, and may be stored under freeze drying (lyophilization) conditions requiring only the addition of a sterile liquid carrier, such as water for injection, immediately prior to use. Extemporaneous injection solutions and suspensions are prepared from sterile powders, granules, and tablets of the types described previously. In some embodiments, the disclosed pharmaceutical composition comprising one or more compounds provided herein or pharmaceutically acceptable salts thereof further comprises one or more additional therapeutically active agents. The additional therapeutic agent has an activity that complements the compound provided herein, so that it does not adversely affect each other. Such agents are appropriately combined in an amount effective for the intended purpose. In certain embodiments, the additional therapeutic agent is selected from: EGFR TKI, EGFR antibody, MEK inhibitor, c-MET inhibitor, mitotic kinase inhibitor, immunotherapy, anti-angiogenic agent, apoptosis inducer, mTOR inhibitor, histone deacetylase inhibitor, IL6 inhibitor, JAK inhibitor. Examples of EGFR TKIs include, but are not limited to, for example, afatinib, erlotinib, gefitinib, lapatinib, dacomitinib, osimertinib, olmutinib, nazartinib, and AC0010. Examples of EGFR antibodies include, for example, cetuximab, panitumumab, and necitumumab. Examples of MEK inhibitors include trametinib, cobimetinib, binimetinib, selumetinib, and refametinib. Examples of c-MET inhibitors include, for example, savolitinib, cabozantinib, foretinib, and MET antibodies (such as emibetuzumab). Examples of mitotic kinase inhibitors include CDK4/6 inhibitors, such as palbociclib, ribociclib, and abemaciclib. Examples of immunotherapeutic agents include immune checkpoint inhibitors, such as anti-CTLA4 mAb, anti-PD1 mAb, anti-PD-L1 mAb, anti-PD-L2 mAb, anti-LAG3 mAb, anti-TM3 mAb, preferably anti-PD1 mAb, ipilimumab, nivolumab, pembrolizumab, atezolizumab, avelumab, durvalumab, pidilizumab, PDR-001, and immunomodulators, such as CD73 inhibitors or CD73 inhibitory antibodies. Examples of anti-angiogenic agents include, for example, bevacizumab, nintedanib. Examples of apoptosis inducers include Bcl-2 inhibitors (e.g., venetoclax, obatoclax, navitoclax), Mcl-1 inhibitors (e.g., AZD-5991, AMG-176, S-64315). Examples of mTOR inhibitors include, for example, rapamycin, temsirolimus, everolimus, ridaforolimus. Examples of histone deacetylase inhibitors include, for example, panobinostat, entinostat, romidepsin, and vorinostat. Examples of IL6 inhibitors include, for example, tocilizumab, siltuximab, olokizumab, elsilimomab, clazakizumab, sirukumab, levilimab, ARGX-109, FE301, FM101. Examples of JAK inhibitors include, for example, baricitinib, ruxolitinib, tofacitinib, oclacitinib, baricitinib, peficitinib, fedratinib, upadacitinib, filgotinib, delgocitinib, abrocitinib. One or more other therapeutic agents can be used simultaneously or sequentially with the compound provided herein. Sequential use includes use before or after the compound provided herein. In some embodiments, the one or more other therapeutic agents can be used in the same composition as the compound provided herein. In other embodiments, there can be a time interval between the use of the other therapeutic agent and the compound provided herein. In some embodiments, the use of the other therapeutic agent together with the compound provided herein can make it possible to reduce the dosage of other therapeutic agents and/or to use them at intervals with lower frequency. Methods for treatment The disclosed compound and the pharmaceutical composition comprising the compound can inhibit EGFR, and therefore can be used to inhibit the EGFR activity in a subject in need, and to prevent or treat EGFR-related diseases. In other aspects, the disclosure provides a method for treating EGFR related conditions, the method comprising administering an effective amount of a compound provided herein or a pharmaceutically acceptable salt thereof or a pharmaceutical composition to a subject in need thereof. As used herein, the term "treating/treatment" or "therapy" is intended to have its normal meaning, i.e., to treat a disease, to fully or partially alleviate one, some or all of its symptoms, or to correct or compensate for potential pathology, thereby achieving a useful or desired clinical outcome. For purposes of this disclosure, beneficial or desired clinical results include, but are not limited to, relief of symptoms, reduction in extent of disease, stabilization of the disease state (i.e., not worsening), delay or slowing of disease progression, improvement or alleviation of the disease state, and remission (whether partial or complete), whether detectable or undetectable. "Treatment" may also mean prolonging survival as compared to expected survival if not receiving treatment. Situations in which treatment is needed include those already suffering from the condition or disorder as well as those susceptible to developing the condition or disorder or those in which the condition or disorder is to be prevented. As used herein, the term "preventing, prevention or prophylaxis" is intended to have its normal meaning and includes primary prevention for preventing the development of a disease and secondary prevention for preventing a disease from developing and temporarily or permanently protecting a patient from aggravation or deterioration of the disease or from developing new symptoms associated with the disease. In some embodiments, the compounds provided herein or their pharmaceutically acceptable salts and compositions can be used to treat a variety of EGFR-related conditions, including cancer, autoimmune diseases, and the like. In certain embodiments, the compounds provided herein or their pharmaceutically acceptable salts and compositions can be used to treat cancer, including lung cancer (e.g., non-small cell lung cancer), brain cancer, colorectal cancer, bladder cancer, urothelial carcinoma, breast cancer, prostate cancer, ovarian cancer, head and neck cancer, pancreatic cancer, gastric cancer and mesothelioma, including metastasis (particularly brain metastasis), and the like. In some embodiments, the compound or its pharmaceutically acceptable salt and composition provided herein can be used to treat autoimmune diseases, including rheumatoid arthritis, graft-versus-host disease, systemic lupus erythematosus (SLE), scleroderma, multiple sclerosis, diabetes, organ rejection, inflammatory bowel disease, psoriasis and other pains. The concentration and the application path of the subject will be different due to the EGFR related conditions to be treated. In some embodiments, the application is carried out by the route selected from the group consisting of: enteral, intraperitoneal, intradermal, intracardiac, intraventricular, intracranial, intracerebrospinal, intrasynovial, intrathecal administration, intramuscular injection, intravitreal injection, intravenous injection, intraarterial injection, oral, oral, sublingual, transdermal, topical, intratracheal, rectal, subcutaneous and ocular administration. The embodiments of various aspects provided by the present disclosure are also described in any of the following paragraphs. Embodiment 1: A compound of formula (I):(I) or a pharmaceutically acceptable salt thereof, wherein: Ring A is selected from the group consisting of cycloalkyl, heterocyclic, aryl and heteroaryl; Ring B is selected from the group consisting of cycloalkyl, heterocyclic, aryl and heteroaryl; L 1Selected from the group consisting of: a bond, an alkyl group, an alkenyl group, an alkynyl group, a heteroalkyl group, a heteroalkenyl group and a heteroalkynyl group, wherein the alkyl group, the alkenyl group, the alkynyl group, the heteroalkyl group, the heteroalkenyl group and the heteroalkynyl group are optionally substituted with one or more groups independently selected from the group consisting of: a hydroxyl group, a halogen group, a cyano group and an amino group; L 2Select from the group consisting of: key, N(R A), alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted by one or more R BReplace; Where R ASelected from the group consisting of: hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from: hydroxyl, halogen, cyano, amino, alkyl and halogenated alkyl; wherein each R BIndependently selected from the group consisting of hydroxyl, halogen, cyano, amino, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, cycloalkyl, heterocyclo, aryl and heteroaryl; L 3Selected from the group consisting of: a bond, an alkyl group, an alkenyl group, an alkynyl group, a heteroalkyl group, a heteroalkenyl group and a heteroalkynyl group, wherein the alkyl group, the alkenyl group, the alkynyl group, the heteroalkyl group, the heteroalkenyl group and the heteroalkynyl group are optionally substituted with one or more groups independently selected from the group consisting of: a hydroxyl group, a halogen group, a cyano group and an amino group; L 4Choose from O, S or N(R C); Where R CSelected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, halogenated alkyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl; Each R 1Independently selected from the group consisting of: hydroxyl, halogen, cyano, amino, -N(R D) 2, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted by one or more R EReplace; where each R DIndependently selected from the group consisting of: hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the following: hydroxyl, halogen, cyano,  -N(R F) 2OR G; where each R EIndependently selected from the group consisting of: hydrogen, hydroxyl, halogen, cyano, amino, -N(R F) 2、-alkyl-N(R F) 2、-C(O)OR G, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the following: hydroxyl, halogen, cyano, amino and alkyl; wherein R Fand R GEach of R is independently selected from the group consisting of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl; Each R 2Independently selected from the group consisting of: hydrogen, hydroxyl, halogen, cyano, amino, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from: hydroxyl, halogen, cyano, amino, alkyl and halogenated alkyl; R 3Selected from the group consisting of: hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl and heterocyclo, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl and heterocyclo are optionally substituted by one or more groups independently selected from: hydroxyl, halogen, cyano, amino, alkyl and halogenated alkyl; m is an integer from 0 to 5; and n is an integer from 0 to 4. Embodiment 2. The compound or a pharmaceutically acceptable salt thereof according to Embodiment 1, wherein Ring A is an aryl or heteroaryl group. Embodiment 3. The compound or pharmaceutically acceptable salt thereof according to Embodiment 2, wherein Ring A is selected from the group consisting of furanyl, phenylthio, pyrrolyl, phenyl, pyridyl, pyranyl, pyrimidyl, oxazinyl, pyrazinyl and tetrahydroisoquinolinyl. Embodiment 4. The compound or pharmaceutically acceptable salt thereof according to Embodiment 3, wherein Ring A is selected from the group consisting of: or and . Embodiment 5. The compound or pharmaceutically acceptable salt thereof according to Embodiment 1, wherein Ring B is an aryl group or a heteroaryl group. Embodiment 6. The compound or pharmaceutically acceptable salt thereof according to Embodiment 5, wherein Ring B is a phenyl group or a pyrazolyl group. Embodiment 7. The compound or pharmaceutically acceptable salt thereof according to Embodiment 6, wherein Ring B is and . Implementation 8. The compound or a pharmaceutically acceptable salt thereof according to implementation 1, wherein L 1is a bond. Implementation 9. A compound or a pharmaceutically acceptable salt thereof according to Implementation 8, wherein L 1is an alkyl group. Embodiment 10. A compound according to Embodiment 9 or a pharmaceutically acceptable salt thereof, wherein L 1Choose from a group consisting of: and , where L 1*end and L 2Connected. Implementation 11. The compound or a pharmaceutically acceptable salt thereof according to Implementation 1, wherein L 2is a bond. Implementation 12. A compound or a pharmaceutically acceptable salt thereof according to Implementation 1, wherein L 2It is N(R A), and R ASelected from alkyl or heterocyclic groups, wherein the alkyl or heterocyclic group is optionally substituted by one or more halogens or alkyl groups. Embodiment 13. A compound or a pharmaceutically acceptable salt thereof according to Embodiment 12, wherein R Ais ethyl, difluoroethyl, trifluoroethyl or oxacyclobutane. Implementation 14. A compound according to Implementation 1 or a pharmaceutically acceptable salt thereof, wherein L 2is optionally represented by one or more R BSubstituted cycloalkyl. Embodiment 15. A compound or a pharmaceutically acceptable salt thereof according to Embodiment 14, wherein L 2yes , which is optionally replaced by one or more R BReplacement. Implementation 16. A compound or a pharmaceutically acceptable salt thereof according to implementation 1, wherein L 2is optionally represented by one or more R BSubstituted heterocyclic group. Embodiment 17. A compound or a pharmaceutically acceptable salt thereof according to Embodiment 16, wherein the heterocyclic group is selected from the group consisting of: and , each of which is optionally represented by one or more R BReplacement, where L 2*end and L 3connected. Implementation 18. A compound or a pharmaceutically acceptable salt thereof according to Implementation 16 or 17, wherein R Bis an alkyl group. Embodiment 19. A compound or a pharmaceutically acceptable salt thereof according to Embodiment 19, wherein R Bis methyl. Implementation 20. A compound or a pharmaceutically acceptable salt thereof according to implementation 1, wherein L 2Choose from a group consisting of: and , where L 2*end and L 3connected. Implementation 21. A compound or a pharmaceutically acceptable salt thereof according to implementation 1, wherein L 3is an alkyl group. Embodiment 22. A compound or a pharmaceutically acceptable salt thereof according to Embodiment 21, wherein L 3is ethyl. Implementation 23. A compound or a pharmaceutically acceptable salt thereof according to Implementation 1, wherein L 4is O or NH. Implementation 24. A compound or a pharmaceutically acceptable salt thereof according to implementation 1, wherein L 1is the key, and L 2is a key or optionally replaced by one or more R BSubstituted heterocyclic group. Embodiment 25. The compound or pharmaceutically acceptable salt thereof according to Embodiment 1, wherein L 1is an alkyl group, and L 2Yes key, N(R A) or optionally one or more R BSubstituted cycloalkyl. Embodiment 26. A compound or a pharmaceutically acceptable salt thereof according to Embodiment 1, wherein R 1is a halogen, and m is 1. Implementation 27. A compound or a pharmaceutically acceptable salt thereof according to Implementation 26, wherein R 1is bromine. Implementation 28. A compound or a pharmaceutically acceptable salt thereof according to Implementation 1, wherein R 1Yes-N(R D) 2, and m is 1. Implementation 29. A compound or a pharmaceutically acceptable salt thereof according to implementation 28, wherein each R DIndependently is optionally selected by one or more independently selected from -N(R F) 2OR Galkyl substituted with a group. Embodiment 30. A compound or a pharmaceutically acceptable salt thereof according to Embodiment 29, wherein each R Dare independently methyl, methoxyethyl or N,N-dimethylaminopropyl. Embodiment 31. A compound or a pharmaceutically acceptable salt thereof according to Embodiment 1, wherein R 1is optionally represented by one or more R Esubstituted heterocyclic group, and m is 1. Embodiment 32. A compound or a pharmaceutically acceptable salt thereof according to Embodiment 31, wherein the heterocyclic group is selected from the group consisting of: and , each of which is optionally represented by one or more R EReplacement. Implementation 33. A compound or a pharmaceutically acceptable salt thereof according to Implementation 31, wherein R Eis a halogen. Implementation 34. A compound or a pharmaceutically acceptable salt thereof according to Implementation 32, wherein R Eis F. Implementation 35. A compound or a pharmaceutically acceptable salt thereof according to implementation 31, wherein R EYes-N(R F) 2、-alkyl-N(R F) 2、    -C(O)OR GOr an alkyl group optionally substituted by one or more halogens. Embodiment 36. A compound according to Embodiment 35 or a pharmaceutically acceptable salt thereof, wherein R Fand R GEach of them is an alkyl group. Embodiment 37. A compound or a pharmaceutically acceptable salt thereof according to Embodiment 35, wherein R Fand R GEach of them is C 1-3Alkyl. Embodiment 38. A compound or a pharmaceutically acceptable salt thereof according to Embodiment 35, wherein R ESelect from the group consisting of: -N(CH 3) 2、-C 1-3Alkyl-N(CH 3) 2, -C(O)O(tert-butyl), methyl, ethyl or trifluoroethyl. Embodiment 39. A compound or a pharmaceutically acceptable salt thereof according to Embodiment 31, wherein R Eis a cycloalkyl or heterocyclic group, wherein the cycloalkyl and the heterocyclic group are optionally substituted by one or more alkyl or halogen groups. Embodiment 40. A compound according to Embodiment 39 or a pharmaceutically acceptable salt thereof, wherein R ESelected from cyclopropyl, morpholinyl, piperazinyl, oxadiazine or cyclobutane, each of which is optionally substituted by one or more alkyl or halogen groups. Embodiment 41. A compound according to Embodiment 1 or a pharmaceutically acceptable salt thereof, wherein R 1Choose from a group consisting of: and . Implementation 42. A compound or a pharmaceutically acceptable salt thereof according to implementation 1, wherein R 2is a halogen, and n is 1 or 2. Implementation 43. A compound or a pharmaceutically acceptable salt thereof according to implementation 42, wherein R 2is F. Implementation 44. A compound or a pharmaceutically acceptable salt thereof according to implementation 1, wherein R 2is an alkyl or cycloalkyl group, wherein the alkyl group and the cycloalkyl group are optionally substituted by one or more halogens. Embodiment 45. A compound according to Embodiment 44 or a pharmaceutically acceptable salt thereof, wherein R 2is methyl, ethyl, cyclopropyl or trifluoroethyl. Implementation 46. A compound or a pharmaceutically acceptable salt thereof according to Implementation 1, wherein R 3is an alkyl group or a cycloalkyl group. Embodiment 47. A compound or a pharmaceutically acceptable salt thereof according to Embodiment 46, wherein R 3is methyl, ethyl or cyclopropyl. Embodiment 48. The compound or a pharmaceutically acceptable salt thereof according to Embodiment 1, wherein m is 0, 1 or 2. Embodiment 49. The compound or a pharmaceutically acceptable salt thereof according to Embodiment 1, wherein n is 0 or 1. Embodiment 50. The compound or a pharmaceutically acceptable salt thereof according to Embodiment 1, wherein the compound is selected from the group consisting of: and . Embodiment 51. A pharmaceutical composition comprising a compound according to any one of Embodiments 1 to 50 or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier. Embodiment 52. A method for inhibiting EGFR activity in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound according to any one of Embodiments 1 to 50 or a pharmaceutically acceptable salt thereof or a pharmaceutical composition according to Embodiment 51. Embodiment 53. A method for treating an EGFR-related disorder in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound according to any one of Embodiments 1 to 50 or a pharmaceutically acceptable salt thereof or administering to the subject a pharmaceutical composition according to Embodiment 51. Embodiment 54. The method according to Embodiment 53, wherein the EGFR-related disorder is an autoimmune disease or cancer. Embodiment 55. The method according to embodiment 54, wherein the cancer is selected from the group consisting of lung cancer, brain cancer, colorectal cancer, bladder cancer, urothelial cancer, breast cancer, prostate cancer, ovarian cancer, head and neck cancer, pancreatic cancer, gastric cancer and mesothelioma, including metastasis (especially brain metastasis), etc. Embodiment 56. The method according to any one of embodiments 53 to 55, wherein the compound is administered simultaneously, separately or sequentially with one or more additional therapeutic agents. Embodiment 57. The method according to embodiment 56, wherein the one or more additional therapeutic agents are selected from the group consisting of: EGFR TKI, EGFR antibody, MEK inhibitor, c-MET inhibitor, mitotic kinase inhibitor, immunotherapy agent, anti-angiogenic agent, apoptosis inducer, mTOR inhibitor, histone deacetylase inhibitor, IL6 inhibitor and JAK inhibitor. In addition, the embodiments of various aspects provided by the present disclosure are also described in any of the following paragraphs. Item 1. A compound of formula (II) (II) or a pharmaceutically acceptable salt thereof, wherein: Ring A 1is a 7-12 membered cycloalkyl, a 7-12 membered heterocyclic group, a 7-12 membered aryl group or a 7-12 membered heteroaryl group; Ring B is selected from the group consisting of: cycloalkyl, heterocyclic group, aryl group and heteroaryl group; L 1Selected from the group consisting of: a bond, an alkyl group, an alkenyl group, an alkynyl group, a heteroalkyl group, a heteroalkenyl group and a heteroalkynyl group, wherein the alkyl group, the alkenyl group, the alkynyl group, the heteroalkyl group, the heteroalkenyl group and the heteroalkynyl group are optionally substituted with one or more groups independently selected from the group consisting of: a hydroxyl group, a halogen group, a cyano group and an amino group; L 2Select from the group consisting of: key, N(R A), alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted by one or more R BReplace; Where R ASelected from the group consisting of: hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the following: hydroxyl, halogen, cyano, amino, alkyl and halogenated alkyl; wherein each R BIndependently selected from the group consisting of hydroxyl, halogen, cyano, amino, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, cycloalkyl, heterocyclo, aryl and heteroaryl; L 3Selected from the group consisting of: a bond, an alkyl group, an alkenyl group, an alkynyl group, a heteroalkyl group, a heteroalkenyl group and a heteroalkynyl group, wherein the alkyl group, the alkenyl group, the alkynyl group, the heteroalkyl group, the heteroalkenyl group and the heteroalkynyl group are optionally substituted with one or more groups independently selected from the group consisting of: a hydroxyl group, a halogen group, a cyano group and an amino group; L 4Choose from O, S or N(R C); Where R CSelected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, halogenated alkyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl; Each R 1Independently selected from the group consisting of: hydroxyl, halogen, cyano, amino, -N(R D) 2, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted by one or more R EReplace; where each R DIndependently selected from the group consisting of: hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the following: hydroxyl, halogen, cyano,  -N(R F) 2OR G; where each R EIndependently selected from the group consisting of: hydrogen, hydroxyl, halogen, cyano, amino, -N(R F) 2、-alkyl-N(R F) 2、-C(O)OR G, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the following: hydroxyl, halogen, cyano, amino and alkyl; wherein R Fand R GEach of R is independently selected from the group consisting of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl; Each R 2Independently selected from the group consisting of: hydrogen, hydroxyl, halogen, cyano, amino, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from: hydroxyl, halogen, cyano, amino, alkyl and halogenated alkyl; Each R 3Selected from the group consisting of: hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl and heterocyclic, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl and heterocyclic are optionally substituted by one or more groups independently selected from the following: hydroxyl, halogen, cyano, amino, alkyl and halogenated alkyl; m is an integer from 0 to 5; n is an integer from 0 to 4; and p is an integer from 0 to 3. Item 2. A compound or a pharmaceutically acceptable salt thereof according to Project 1, wherein Ring A 1is a 7-12 membered heteroaryl group. Item 3. A compound or a pharmaceutically acceptable salt thereof according to Project 2, wherein Ring A 1is tetrahydroisoquinolinyl. Item 4. A compound or a pharmaceutically acceptable salt thereof according to Project 3, wherein Ring A 1yes or . Item 5. A compound or a pharmaceutically acceptable salt thereof according to any of the foregoing projects, wherein m is 0. Item 6. A compound or a pharmaceutically acceptable salt thereof according to Project 1, wherein m is 1. Item 7. A compound or a pharmaceutically acceptable salt thereof according to Project 6, wherein R 1is optionally replaced by one or more N(R F) 2Substituted alkyl. Item 8. A compound or a pharmaceutically acceptable salt thereof according to Item 7, wherein R 1is optionally replaced by one or more N(R F) 2Substituted ethyl. Item 9. A compound or a pharmaceutically acceptable salt thereof according to Project 7 or 8, wherein R Fis an alkyl group. Item 10. A compound or a pharmaceutically acceptable salt thereof according to Project 9, wherein R Fis a methyl group. Item 11. A compound or a pharmaceutically acceptable salt thereof according to Project 6, wherein R 1is a heterocyclic group. Item 12. A compound or a pharmaceutically acceptable salt thereof according to Project 11, wherein R 1is an oxacyclobutane. Item 13. A compound or a pharmaceutically acceptable salt thereof according to Item 12, wherein R 1yes . Item 14. A compound or a pharmaceutically acceptable salt thereof according to Project 1, wherein L 1is an alkyl group. Item 15. A compound or a pharmaceutically acceptable salt thereof according to Item 14, wherein L 1yes or , where L 1*end and L 2Connected. Item 16. A compound or a pharmaceutically acceptable salt thereof according to Project 1, wherein L 2is a bond. Item 17. A compound or a pharmaceutically acceptable salt thereof according to Project 1, wherein L 3is an alkyl group. Item 18. A compound or a pharmaceutically acceptable salt thereof according to Project 11, wherein L 3is ethyl. Item 19. A compound or a pharmaceutically acceptable salt thereof according to Project 1, wherein L 4is O or NH. Item 20. A compound or a pharmaceutically acceptable salt thereof according to Item 1, wherein Ring B is an aryl group or a heteroaryl group. Item 21. A compound or a pharmaceutically acceptable salt thereof according to Item 20, wherein Ring B is a phenyl group, a pyridyl group or a pyrazolyl group. Item 22. A compound or a pharmaceutically acceptable salt thereof according to Item 21, wherein Ring B is selected from the group consisting of: and . Item 23. A compound or a pharmaceutically acceptable salt thereof according to Item 1, wherein n is 1. Item 24. A compound or a pharmaceutically acceptable salt thereof according to Item 1, wherein R 2is an alkyl group. Item 25. A compound or a pharmaceutically acceptable salt thereof according to Item 24, wherein R 2is a methyl group. Item 26. A compound or a pharmaceutically acceptable salt thereof according to Project 1, wherein R 3is an alkyl group. Item 27. A compound or a pharmaceutically acceptable salt thereof according to Item 26, wherein R 3is methyl. Item 28. A compound according to Project 1 or a pharmaceutically acceptable salt thereof, wherein m is 0 or 1. Item 29. A compound according to Project 1 or a pharmaceutically acceptable salt thereof, wherein n is 1. Item 30. A compound according to Project 1 or a pharmaceutically acceptable salt thereof, wherein p is 1. Item 31. A compound of formula (III): (III) or a pharmaceutically acceptable salt thereof, wherein: Ring A is selected from the group consisting of cycloalkyl, heterocyclic, aryl and heteroaryl; Ring B 1is a 7-12-membered cycloalkyl group, a 7-12-membered heterocyclic group, a 7-12-membered aryl group, or a 7-12-membered heteroaryl group; L 1Selected from the group consisting of: a bond, an alkyl group, an alkenyl group, an alkynyl group, a heteroalkyl group, a heteroalkenyl group and a heteroalkynyl group, wherein the alkyl group, the alkenyl group, the alkynyl group, the heteroalkyl group, the heteroalkenyl group and the heteroalkynyl group are optionally substituted with one or more groups independently selected from the group consisting of: a hydroxyl group, a halogen group, a cyano group and an amino group; L 2Select from the group consisting of: key, N(R A), alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted by one or more R BReplace; Where R ASelected from the group consisting of: hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the following: hydroxyl, halogen, cyano, amino, alkyl and halogenated alkyl; wherein each R BIndependently selected from the group consisting of hydroxyl, halogen, cyano, amino, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, cycloalkyl, heterocyclo, aryl and heteroaryl; L 3Selected from the group consisting of: a bond, an alkyl group, an alkenyl group, an alkynyl group, a heteroalkyl group, a heteroalkenyl group and a heteroalkynyl group, wherein the alkyl group, the alkenyl group, the alkynyl group, the heteroalkyl group, the heteroalkenyl group and the heteroalkynyl group are optionally substituted with one or more groups independently selected from the group consisting of: a hydroxyl group, a halogen group, a cyano group and an amino group; L 4Choose from O, S or N(R C); Where R CSelected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, halogenated alkyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl; Each R 1Independently selected from the group consisting of: hydroxyl, halogen, cyano, amino, -N(R D) 2, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted by one or more R EReplace; where each R DIndependently selected from the group consisting of: hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the following: hydroxyl, halogen, cyano,  -N(R F) 2OR G; where each R EIndependently selected from the group consisting of: hydrogen, hydroxyl, halogen, cyano, amino, -N(R F) 2、-alkyl-N(R F) 2、-C(O)OR G, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the following: hydroxyl, halogen, cyano, amino and alkyl; wherein R Fand R GEach of R is independently selected from the group consisting of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl; Each R 2Independently selected from the group consisting of: hydrogen, hydroxyl, halogen, cyano, amino, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from: hydroxyl, halogen, cyano, amino, alkyl and halogenated alkyl; Each R 3Selected from the group consisting of: hydrogen, halogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl and heterocyclo, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl and heterocyclo are optionally substituted by one or more groups independently selected from: hydroxyl, halogen, cyano, amino, alkyl and halogenated alkyl; m is an integer from 0 to 5; n is an integer from 0 to 4; and p is an integer from 0 to 3. Item 32. A compound or a pharmaceutically acceptable salt thereof according to Project 31, wherein Ring A is an aryl or heteroaryl group. Item 33. A compound or a pharmaceutically acceptable salt thereof according to Item 32, wherein Ring A is selected from the group consisting of furanyl, phenylthio, pyrrolyl, phenyl, pyridyl, pyranyl, pyrimidyl, oxazinyl, pyrazinyl and tetrahydroisoquinolinyl. Item 34. A compound or a pharmaceutically acceptable salt thereof according to Item 31, wherein m is 0. Item 35. A compound or a pharmaceutically acceptable salt thereof according to Item 31, wherein R 1is a halogen, and m is 1. Item 36. A compound or a pharmaceutically acceptable salt thereof according to Item 35, wherein R 1is bromine. Item 37. A compound or a pharmaceutically acceptable salt thereof according to Item 31, wherein R 1Yes-N(R D) 2, and m is 1. Item 38. A compound or a pharmaceutically acceptable salt thereof according to Item 37, wherein each R DIndependently is optionally selected by one or more independently selected from -N(R F) 2OR G Item 39. A compound or a pharmaceutically acceptable salt thereof according to Item 38, wherein each R Dare independently methyl, methoxyethyl or N,N-dimethylaminopropyl. Item 40. A compound or a pharmaceutically acceptable salt thereof according to Project 31, wherein R 1is optionally represented by one or more R Esubstituted heterocyclic group, and m is 1. Item 41. A compound or a pharmaceutically acceptable salt thereof according to Item 40, wherein the heterocyclic group is selected from the group consisting of: and , each of which is optionally represented by one or more R ESubstituted. Item 42. A compound or a pharmaceutically acceptable salt thereof according to Item 40, wherein R Eis a halogen. Item 43. A compound or a pharmaceutically acceptable salt thereof according to Item 42, wherein R Eis F. Item 44. A compound or a pharmaceutically acceptable salt thereof according to Item 40, wherein R EYes-N(R F) 2、-alkyl-N(R F) 2、-C(O)OR GOr an alkyl group optionally substituted by one or more halogens. Item 45. A compound or a pharmaceutically acceptable salt thereof according to Item 44, wherein R Fand R GEach of them is an alkyl group. Item 46. A compound or a pharmaceutically acceptable salt thereof according to Item 45, wherein R Fand R GEach of them is C 1-3Alkyl. Item 47. A compound or a pharmaceutically acceptable salt thereof according to Item 44, wherein R ESelect from the group consisting of: -N(CH 3) 2、-C 1-3Alkyl-N(CH 3) 2, -C(O)O(tert-butyl), methyl, ethyl or trifluoroethyl. Item 48. A compound or a pharmaceutically acceptable salt thereof according to Project 40, wherein R Eis a cycloalkyl or heterocyclic group, wherein the cycloalkyl and the heterocyclic group are optionally substituted by one or more alkyl or halogen groups. Item 49. A compound or a pharmaceutically acceptable salt thereof according to Item 48, wherein R ESelected from cyclopropyl, morpholine, piperazine, oxacyclobutenyl or azocyclobutane, each of which is optionally substituted by one or more alkyl or halogen groups. Item 50. A compound or a pharmaceutically acceptable salt thereof according to Project 31, wherein R 1Choose from a group consisting of and . Item 51. A compound or a pharmaceutically acceptable salt thereof according to Project 31, wherein Ring B 1is a 7-12 membered heteroaryl group. Item 52. A compound or a pharmaceutically acceptable salt thereof according to Item 51, wherein Ring B 1yes . Item 53. A compound or a pharmaceutically acceptable salt thereof according to Project 31, wherein R 2is a halogen, and n is 1 or 2. Item 54. A compound or a pharmaceutically acceptable salt thereof according to Item 53, wherein R 2is F. Item 55. A compound or a pharmaceutically acceptable salt thereof according to Project 31, wherein R 2is an alkyl group or a cycloalkyl group, wherein the alkyl group and the cycloalkyl group are optionally substituted by one or more halogens. Item 56. A compound or a pharmaceutically acceptable salt thereof according to Item 55, wherein R 2is methyl, ethyl, cyclopropyl or trifluoroethyl. Item 57. A compound or a pharmaceutically acceptable salt thereof according to Project 31, wherein L 1is an alkyl group. Item 58. A compound or a pharmaceutically acceptable salt thereof according to Item 57, wherein L 1Is propyl. Item 59. A compound or a pharmaceutically acceptable salt thereof according to Project 31, wherein L 2is a bond. Item 60. A compound or a pharmaceutically acceptable salt thereof according to Project 31, wherein L 3is an alkyl group. Item 61. A compound or a pharmaceutically acceptable salt thereof according to Item 60, wherein L 3is ethyl. Item 62. A compound or a pharmaceutically acceptable salt thereof according to Project 31, wherein L 4is O or NH. Item 63. A compound or a pharmaceutically acceptable salt thereof according to Project 31, wherein R 3is an alkyl group. Item 64. A compound or a pharmaceutically acceptable salt thereof according to Item 63, wherein R 3is a methyl group. Item 65. A compound or a pharmaceutically acceptable salt thereof according to Project 31, wherein R 3is a halogen. Item 66. A compound or a pharmaceutically acceptable salt thereof according to Item 65, wherein R 3is Cl. Item 67. A compound according to Item 31 or a pharmaceutically acceptable salt thereof, wherein m is 0. Item 68. A compound according to Item 31 or a pharmaceutically acceptable salt thereof, wherein n is 0. Item 69. A compound according to Item 31 or a pharmaceutically acceptable salt thereof, wherein p is 1. Item 70. A compound of formula (IV): (IV) or a pharmaceutically acceptable salt thereof, wherein: Ring A is selected from the group consisting of cycloalkyl, heterocyclic, aryl and heteroaryl; Ring B is selected from the group consisting of cycloalkyl, heterocyclic, aryl and heteroaryl; L 1Selected from the group consisting of: a bond, an alkyl group, an alkenyl group, an alkynyl group, a heteroalkyl group, a heteroalkenyl group and a heteroalkynyl group, wherein the alkyl group, the alkenyl group, the alkynyl group, the heteroalkyl group, the heteroalkenyl group and the heteroalkynyl group are optionally substituted with one or more groups independently selected from the group consisting of: a hydroxyl group, a halogen group, a cyano group and an amino group; L twenty oneis a 7-12 membered cycloalkyl or a 7-12 membered heterocyclic group, wherein the cycloalkyl and heterocyclic groups are optionally substituted by one or more R BReplace; where each R BIndependently selected from the group consisting of hydroxyl, halogen, cyano, amino, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, cycloalkyl, heterocyclo, aryl and heteroaryl; L 3Selected from the group consisting of: a bond, an alkyl group, an alkenyl group, an alkynyl group, a heteroalkyl group, a heteroalkenyl group and a heteroalkynyl group, wherein the alkyl group, the alkenyl group, the alkynyl group, the heteroalkyl group, the heteroalkenyl group and the heteroalkynyl group are optionally substituted with one or more groups independently selected from the group consisting of: a hydroxyl group, a halogen group, a cyano group and an amino group; L 4Choose from O, S or N(R C); Where R CSelected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, halogenated alkyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl; Each R 1Independently selected from the group consisting of: hydroxyl, halogen, cyano, amino, -N(R D) 2, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted by one or more R EReplace; where each R DIndependently selected from the group consisting of: hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the following: hydroxyl, halogen, cyano,  -N(R F) 2OR G; where each R EIndependently selected from the group consisting of: hydrogen, hydroxyl, halogen, cyano, amino, -N(R F) 2、-alkyl-N(R F) 2、-C(O)OR G, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the following: hydroxyl, halogen, cyano, amino and alkyl; wherein R Fand R GEach of R is independently selected from the group consisting of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl; Each R 2Independently selected from the group consisting of: hydrogen, hydroxyl, halogen, cyano, amino, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from: hydroxyl, halogen, cyano, amino, alkyl and halogenated alkyl; R 3Selected from the group consisting of: hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl and heterocyclo, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl and heterocyclo are optionally substituted by one or more groups independently selected from: hydroxyl, halogen, cyano, amino, alkyl and halogenated alkyl; m is an integer from 0 to 5; n is an integer from 0 to 4; and p is an integer from 0 to 3. Item 71. A compound or a pharmaceutically acceptable salt thereof according to Item 70, wherein Ring A is an aryl group. Item 72. A compound or a pharmaceutically acceptable salt thereof according to Item 71, wherein Ring A is a phenyl group. Item 73. A compound or a pharmaceutically acceptable salt thereof according to Item 70, wherein R 1is a halogen, and m is 1. Item 74. A compound or a pharmaceutically acceptable salt thereof according to Item 73, wherein R 1is bromine. Item 75. A compound or a pharmaceutically acceptable salt thereof according to Item 70, wherein R 1Yes-N(R D) 2, and m is 1. Item 76. A compound or a pharmaceutically acceptable salt thereof according to Item 75, wherein each R DIndependently is optionally selected by one or more independently selected from -N(R F) 2OR G Item 77. A compound or a pharmaceutically acceptable salt thereof according to Item 76, wherein each R Dare independently methyl, methoxyethyl or N,N-dimethylaminopropyl. Item 78. A compound or a pharmaceutically acceptable salt thereof according to Item 70, wherein R 1is optionally represented by one or more R Esubstituted heterocyclic group, and m is 1. Item 79. A compound or a pharmaceutically acceptable salt thereof according to Item 78, wherein the heterocyclic group is selected from the group consisting of: and , each of which is optionally represented by one or more R ESubstituted. Item 80. A compound or a pharmaceutically acceptable salt thereof according to Project 78, wherein R Eis a halogen. Item 81. A compound or a pharmaceutically acceptable salt thereof according to Item 80, wherein R Eis F. Item 82. A compound or a pharmaceutically acceptable salt thereof according to Project 78, wherein R EYes-N(R F) 2、-alkyl-N(R F) 2、-C(O)OR GOr an alkyl group optionally substituted by one or more halogens. Item 83. A compound or a pharmaceutically acceptable salt thereof according to Item 82, wherein R Fand R GEach of which is an alkyl group. Item 84. A compound or a pharmaceutically acceptable salt thereof according to Item 83, wherein R Fand R GEach of them is C 1-3Alkyl. Item 85. A compound or a pharmaceutically acceptable salt thereof according to Item 82, wherein R ESelect from the group consisting of: -N(CH 3) 2、-C 1-3Alkyl-N(CH 3) 2, -C(O)O(tert-butyl), methyl, ethyl or trifluoroethyl. Item 86. A compound or a pharmaceutically acceptable salt thereof according to Project 78, wherein R Eis a cycloalkyl or heterocyclic group, wherein the cycloalkyl and the heterocyclic group are optionally substituted by one or more alkyl or halogen groups. Item 87. A compound or a pharmaceutically acceptable salt thereof according to Item 86, wherein R ESelected from cyclopropyl, morpholine, piperazine, oxacyclobutenyl or azocyclobutane, each of which is optionally substituted by one or more alkyl or halogen groups. Project 88 A compound or a pharmaceutically acceptable salt thereof according to Project 70, wherein R 1Choose from a group consisting of and . Item 89. A compound or a pharmaceutically acceptable salt thereof according to Item 70, wherein Ring B is an aryl group or a heteroaryl group. Item 90. A compound or a pharmaceutically acceptable salt thereof according to Item 89, wherein Ring B is a phenyl group, a pyridyl group or a pyrazolyl group. Item 91. A compound or a pharmaceutically acceptable salt thereof according to Item 90, wherein Ring B is selected from the group consisting of: and . Item 92. A compound or a pharmaceutically acceptable salt thereof according to Project 70, wherein L 1is a bond. Item 93. A compound or a pharmaceutically acceptable salt thereof according to Project 70, wherein L twenty oneis a 7-10 membered heterocyclic group containing one or more heteroatoms selected from N or O. Item 94. A compound or a pharmaceutically acceptable salt thereof according to Item 93, wherein L twenty oneChoose from a group consisting of and , where L twenty one*end and L 3connected. Item 95. A compound or a pharmaceutically acceptable salt thereof according to Project 70, wherein L 3is an alkyl group. Item 96. A compound or a pharmaceutically acceptable salt thereof according to Item 95, wherein L 3is ethyl. Item 97. A compound or a pharmaceutically acceptable salt thereof according to Project 70, wherein L 4is O or NH. Item 98. A compound or a pharmaceutically acceptable salt thereof according to Project 70, wherein R 1is optionally represented by one or more R Esubstituted heterocyclic group, and m is 1. Item 99. A compound or a pharmaceutically acceptable salt thereof according to Project 70, wherein R 3is an alkyl group. Item 100. A compound or a pharmaceutically acceptable salt thereof according to Project 99, wherein R 3is methyl. Item 101. A compound or a pharmaceutically acceptable salt thereof according to Item 70, wherein m is 0 or 1. Item 102. A compound or a pharmaceutically acceptable salt thereof according to Item 70, wherein n is 1. Item 103. A compound or a pharmaceutically acceptable salt thereof according to Item 70, wherein p is 1. Item 104. A compound or a pharmaceutically acceptable salt thereof, wherein the compound is selected from the group consisting of: , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , . Item 105. A pharmaceutical composition comprising a compound according to any one of Items 1 to 104 or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier. Item 106. A method for inhibiting EGFR activity in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound according to any one of Items 1 to 104 or a pharmaceutically acceptable salt thereof or a pharmaceutical composition according to Item 105. Item 107. A method for treating an EGFR-related disorder in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound according to any one of Items 1 to 104 or a pharmaceutically acceptable salt thereof or administering to the subject a pharmaceutical composition according to Item 105. Item 108. The method according to Item 107, wherein the EGFR-related disorder is an autoimmune disease or cancer. Item 109. The method according to Item 108, wherein the cancer is selected from the group consisting of lung cancer, brain cancer, colorectal cancer, bladder cancer, urothelial cancer, breast cancer, prostate cancer, ovarian cancer, head and neck cancer, pancreatic cancer, gastric cancer and mesothelioma, including metastasis (especially brain metastasis), etc. Item 110. The method according to any one of Items 107 to 109, wherein the compound is administered simultaneously, separately or sequentially with one or more additional therapeutic agents. Item 111. A method according to Item 110, wherein the one or more additional therapeutic agents are selected from the group consisting of: EGFR TKI, EGFR antibody, MEK inhibitor, c-MET inhibitor, mitotic kinase inhibitor, immunotherapy agent, anti-angiogenic agent, apoptosis inducer, mTOR inhibitor, histone deacetylase inhibitor, IL6 inhibitor, JAK inhibitor. Examples The general method of the present disclosure is further explained below. The compounds of the present disclosure can be prepared by methods known in the art. The detailed preparation methods of the preferred compounds of the present disclosure are described below. However, these in no way limit the preparation methods of the compounds of the present disclosure. Synthesis Examples The following examples are included for illustrative purposes. The examples provided herein describe the synthesis of the compounds disclosed herein and the intermediates used to prepare the compounds. However, it should be understood that these examples do not limit the present disclosure and are intended only to illustrate methods for practicing the present disclosure. Those skilled in the art will recognize that the described chemical reactions can be easily adapted to prepare a variety of other compounds of the present disclosure, and that alternative methods for preparing the compounds of the present disclosure are considered to be within the scope of the present disclosure. For example, non-exemplary compounds according to the present disclosure can be successfully synthesized by modifications that are obvious to those skilled in the art, such as by appropriately protecting interfering groups, by utilizing other suitable reagents and building blocks known in the art in addition to the described reagents and building blocks, and/or by routine modifications to reaction conditions. In addition, it will be understood by those skilled in the art that the various steps described herein or in separate batches of compounds can be combined. Alternatively, other reactions disclosed herein or known in the art will be considered applicable to the preparation of other compounds disclosed herein. Therefore, the following description is not intended to limit the scope of the disclosure, but is specified by the appended claims. Chemistry, Materials and General Methods Unless otherwise specified, all reagents and solvents are from commercial suppliers such as PharmaBlock, Bide Pharmatech, Shanghai Send Pharm, Aldrich, Sigma, etc., and can be used without further purification. Dry organic solvents (THF, DMF, 1,4-dioxane, etc.) were purchased from Anhui Senrise Technology, Beijing Innochem Science & Technology, and J & K Scientific and packaged in Sure/Seal bottles under nitrogen. All reactions involving air- or moisture-sensitive reagents were performed under nitrogen atmosphere. 1H NMR spectra were recorded on a Bruker AV III HD 400 MHz, Bruker AV NEO 400 MHz spectrometer at ambient temperature. Chemical shifts are reported in parts per million (ppm, delta units). The data are reported as follows: chemical shift, number of protons and multiplicity (s = singlet, d = doublet, dd = double of doublets, dt = double of triplets, t = triplet, q = quartet, br = broad, m = multiplet). The reactions were monitored using LCMS (Shimadzu 20AD) with UV detection at 254 nm and low resonance electrospray mode (ESI). Most of the final compounds were purified to > 95% purity as determined by LCMS (3 min). The LCMS (3 minutes) method used the following: Shimadzu 20AD spectrometer, Shim-pack Scepter C18-120 3.3 * 33 mm, 3.0 um, 30°C, with a flow rate of 1.5 ml/min; solvent A was water + 6.5 mM NH 4HCO 3+ Hydrogen hydroxide (pH = 10), solvent B is acetonitrile; 0.0-1.7 minutes, 30% B-70% B; 1.7-2.3 minutes, 70% B-95% B; 2.3-2.8 minutes, 95% B; 2.8-3.0 minutes, 10% B. The flow from the UV detector was split (1:3) to the MS detector, which was configured with ESI as the ionization source. The CHIRAL method used the following: column: CHIRALPAK IC-3, 0.46 * 5 cm, 3 μm; at 25 ° C, the flow rate was 1.0 mL/min; methyl tert-butyl ether (0.2% diethylamine)/(ethanol/dichloromethane = 1/1) = 20/80. The SFC method used the following: Column: (R, R) -WHELK-01-Kromasil, 5 * 25 cm, 5 μm; Mobile phase A: CO 2, mobile phase B: methanol/acetonitrile = 1/1. The abbreviations used in the synthesis of the compounds provided herein are listed below: Synthesis of intermediates Intermediate M1 2-(5-hydroxy-1-methyl-1H-pyrazol-4-yl)-6-methylisoxonitrile Step 1. Preparation of 2-methyl-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-3-one (M1-1). 2-Methyl-1H-pyrazol-3-one (100 g, 1019 mmol, 1 equiv) and K 2CO 3To a stirred solution of 2-(trimethylsilyl)ethoxymethyl chloride (352 g, 2548 mmol, 2.5 equiv) in acetonitrile (1000 mL) was added 2-(trimethylsilyl)ethoxymethyl chloride (254 g, 1528 mmol, 1.5 equiv) dropwise. The resulting mixture was stirred at room temperature for 3 h. The reaction was quenched with water (200 mL) at 0 °C, and the mixture was extracted with ethyl acetate (3×500 mL). The combined organic layers were washed with brine (3×500 mL) and purified by anhydrous Na 2SO 4Dry. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by trituration with petroleum ether (200 mL) to give 130 g of 2-methyl-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-3-one (M1-1) (55%) as a yellow solid. LCMS: m/z (ESI), [2M+H] +=457.25. 1H NMR (DMSO-d 6, 400 MHz) δ 0.00 (9H, s), 0.80-0.85 (2H, m), 3.28 (3H, s), 3.40-3.50 (2H, m), 5.15 (2H, s), 5.29 (1H, d), 7.89 (1H, d). Step 2. Preparation of 4-iodo-2-methyl-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-3-one (M1-2). To a stirred mixture of 2-methyl-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-3-one (47 g, 205.80 mmol, 1 equiv) in acetonitrile (400 mL) at 0 °C under nitrogen atmosphere was added N-iodosuccinimide (50.93 g, 226.39 mmol, 1.1 equiv). The resulting mixture was stirred at 0 °C for 1 h under nitrogen atmosphere. The reaction was quenched with aqueous sodium thiosulfate solution (400 mL) at 0 °C, and the mixture was extracted with dichloromethane (3×300 mL). The combined organic layers were washed with brine (400 mL) and purified by anhydrous Na 2SO 4Dry. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by trituration with petroleum ether (300 mL) to give 72 g of 4-iodo-2-methyl-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-3-one (M1-2) (97%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=354.95. 1H NMR (DMSO-d 6, 400 MHz) δ -0.04 (9H, s), 0.75-0.86 (2H, m), 3.32 (3H, s), 3.39-3.48 (2H, m), 5.13 (2H, s), 8.13 (1H, s). Step 3. Preparation of (6-chloro-4-(methoxycarbonyl)pyridin-2-yl)boronic acid (M1-3). A mixture of bis(pinacolato)diboron (11.5 g, 453.7 mmol, 1.3 equiv) in methyl tert-butyl ether (100 mL) was heated to 80°C and stirred for 0.5 h, then cooled to room temperature, and bis(1,5-cyclooctadiene)di-μ-methoxydiiridium(I) (1.16 g, 17 mmol, 0.05 equiv) and 4-tert-butyl-2-(4-tert-butylpyridin-2-yl)pyridine (1.4 g, 52 mmol, 0.15 equiv) were added. The mixture was stirred at 25°C for 0.5 h, then methyl 2-chloropyridine-4-carboxylate (60 g, 349 mmol, 1.00 equiv) was added, and stirred at 80°C for 16 h. The mixture was cooled to room temperature and concentrated under reduced pressure to give 104 g of methyl 2-chloro-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine-4-carboxylate (M1-3) (crude) as a brown oil. This crude material was used in the next step without purification. LCMS: m/z (ESI), [M+H] +=216.05. Step 4. Preparation of methyl 2-chloro-6-(2-methyl-3-oxo-1-((2-(trimethylsilyl)ethoxy)methyl)-2,3-dihydro-1H-pyrazol-4-yl)isonicotinate (M1-4). Freshly prepared 2-chloro-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine-4-carboxylic acid methyl ester (104 g crude, about 349 mmol, about 1 equiv), 4-iodo-2-methyl-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-3-one (M1-2, 124.30 g, 350.877 mmol, 1 equiv), bis(adamantan-1-yl)(butyl)phosphine (12.58 g, 35.088 mmol, 0.1 equiv), Cs 2CO 3(228.65 g, 701.754 mmol, 2 equiv) and Pd 2(dba) 3A mixture of (9.6 g, 10.4 mmol, 0.03 equiv) in toluene (600 mL) and water (150 mL) was stirred at 80 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (800 mL), and the mixture was extracted with ethyl acetate (3×600 mL). The combined organic layers were washed with brine (3×500 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column using petroleum ether/ethyl acetate (1/4) to give 38 g of methyl 2-chloro-6-(2-methyl-3-oxo-1-((2-(trimethylsilyl)ethoxy)methyl)-2,3-dihydro-1H-pyrazol-4-yl)isonicotinate (M1-4) (25%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=398.05. Step 5. Preparation of methyl 2-methyl-6-(2-methyl-3-oxo-1-((2-(trimethylsilyl)ethoxy)methyl)-2,3-dihydro-1H-pyrazol-4-yl)isonicotinate (M1-5). Under nitrogen atmosphere at room temperature, methyl 2-chloro-6-(2-methyl-3-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-4-yl)pyridine-4-carboxylate (38 g, 95 mmol, 1.00 equiv), Pd(dppf)Cl 2(3677 mg, 5.02 mmol, 0.05 equiv), PCy 3 .HBF 4(10.5 g, 28.64 mmol, 0.3 equiv) and K 2CO 3To a stirred mixture of 1,4-dioxane (26.4 g, 190 mmol, 2 equiv) was added trimethyl-1,3,5,2,4,6-trioxatriborinane (23.9 g, 190 mmol, 2 equiv). The mixture was stirred at 100 °C for 2 h and then cooled to room temperature. The reaction was quenched with water (300 mL) and the mixture was extracted with ethyl acetate (3×200 mL). The combined organic layers were washed with brine (3×300 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column using petroleum ether/ethyl acetate (1/3) to give 34 g of methyl 2-methyl-6-(2-methyl-3-oxo-1-((2-(trimethylsilyl)ethoxy)methyl)-2,3-dihydro-1H-pyrazol-4-yl)isonicotinate (M1-5) (92%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=378.15. 1H NMR (DMSO-d 6, 400 MHz) δ 0.05 (9H, s), 0.80-0.90 (2H, m), 2.53 (3H, s), 3.41 (3H, s), 3.50 (2H, t), 3.89 (3H, s), 5.37 (2H, s), 7.44 (1H, d), 8.52-8.60 (2H, m). Step 6. Preparation of methyl 2-(5-hydroxy-1-methyl-1H-pyrazol-4-yl)-6-methylisonicotinate (M1). A solution of methyl 2-methyl-6-(2-methyl-3-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-4-yl)pyridine-4-carboxylate (34 g, 90 mmol, 1.00 equiv) and HCl in 1,4-dioxane (340 mL) was stirred at room temperature for 2 h under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was treated with saturated NaHCO3(aqueous solution) solution (300 mL) and extracted with ethyl acetate (3×200 mL). The combined organic layers were washed with brine (3×500 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure to obtain 20 g of methyl 2-(5-hydroxy-1-methyl-1H-pyrazol-4-yl)-6-methylisonicotinate (M1) (80%) as a red solid. LCMS: m/z (ESI), [M+H] +=248.05. 1H NMR (DMSO-d 6, 400 MHz) δ 2.56 (3H, s), 3.48 (3H, s), 3.91 (3H, s), 7.31 (1H, s), 7.90-7.80 (2H, m). Intermediate M2 Methyl 5-(5-hydroxy-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate Step 1. Preparation of 5-bromo-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (M2-1). At 0°C, 5-bromo-6-hydroxypyridine-3-carboxylic acid methyl ester (20 g, 86.19 mmol, 1 equivalent) was added to N, N-To the mixture in dimethylformamide (100 mL), NaH (60%) (4.14 g, 172.39 mmol, 2 eq.) was added. The mixture was stirred at 0 °C under nitrogen atmosphere for 60 min. CH 3I (13.46 g, 94.81 mmol, 1.1 equiv) was added, and the mixture was stirred at 0 °C for 1 h. The reaction was quenched with water (1000 mL) at 0 °C, and the mixture was extracted with ethyl acetate (3×500 mL). The combined organic layers were washed with brine (500 mL) and purified by anhydrous Na 2SO 4Dry. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by trituration with petroleum ether (200 mL) to give 19 g of methyl 5-bromo-1-methyl-6-oxopyridine-3-carboxylate (M2-1) (84%) as a white solid. LCMS: m/z (ESI), [M+H] += 245.95. 1H NMR (DMSO-d 6, 400 MHz) δ 3.59 (3H, s), 3.81 (3H, s), 8.18 (1H, d), 8.62 (1H, d). Step 2. Preparation of (5-(methoxycarbonyl)-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)boronic acid (M2-2). 5-Bromo-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (30 g, 121.92 mmol, 1 equivalent), Pd(dppf)Cl 2 .CH 2Cl 2A mixture of (8.92 g, 12.2 mmol, 0.1 eq.), bis(pinacol)diboron (46.44 g, 182.88 mmol, 1.5 eq.) and KOAc (35.90 g, 365.76 mmol, 3 eq.) in 1,4-dioxane (600 mL) was stirred at 80 °C overnight under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction mixture was used directly in the next step without post-treatment. LCMS: m/z (ESI), [M+H] +=212.00. Step 3. Preparation of methyl 1-methyl-5-(2-methyl-3-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-4-yl)-6-oxopyridine-3-carboxylate (M2-3). To the mixture freshly prepared by the reaction described above, 4-iodo-2-methyl-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-3-one (43 g, 121.37 mmol, 1.00 equiv), Pd(dppf)Cl 2.CH 2Cl 2(7.91 g, 9.71 mmol, 0.08 equivalent), K 2CO 3(50.33 g, 364.14 mmol, 3 equiv.) and H 2O (150 mL). The mixture was stirred at 80 °C for 2 h under nitrogen atmosphere. The resulting mixture was cooled to room temperature, treated with water (200 mL), and extracted with dichloromethane (3×300 mL). The combined organic layers were washed with brine (500 mL) and purified by anhydrous Na 2SO 4Dry. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified on a silica gel column using petroleum ether/ethyl acetate (1/8) to obtain 30 g of 1-methyl-5-(2-methyl-3-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-4-yl)-6-oxopyridine-3-carboxylic acid methyl ester (M2-3) (62%) as a brown solid. LCMS: m/z (ESI), [M+H] += 394.10. 1H NMR (DMSO-d 6, 400 MHz) δ -0.06 (9H, s), 0.82 (2H, t), 3.37 (3H, s), 3.47 (2H, t), 3.60 (3H, s), 3.81 (3H, s), 5.33 (2H, s), 8.41 (1H, d), 8.85 (1H, s), 9.21 (1H, d). Step 4. Preparation of methyl 5-(5-hydroxy-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (M2). A mixture of methyl 1-methyl-5-(2-methyl-3-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-4-yl)-6-oxopyridine-3-carboxylate (30 g, 76.23 mmol, 1 eq.) and HCl in 1,4-dioxane (50 mL) was stirred at room temperature overnight. The resulting mixture was concentrated under reduced pressure. The residue was dissolved in dichloromethane (50 mL) and treated with K 2CO 3Alkalize to pH 9. The resulting mixture was filtered and the filter cake was washed with dichloromethane (5×50 mL). The resulting solution was concentrated under reduced pressure. The residue was purified on a silica gel column eluted with dichloromethane/methanol (10/1) to give 15 g of methyl 5-(5-hydroxy-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (M2) (74%) as a brown solid. LCMS: m/z (ESI), [M+H] +=264.10. 1H NMR (DMSO-d 6, 400 MHz) δ 3.52 (3H, s), 3.65 (3H, s), 3.83 (3H, s), 7.76 (1H, d), 8.09 (1H, s), 8.45 (1H, s), 13.34 (1H, s). Intermediate M3 (11 R)-16-bromo-5,11,26-trimethyl-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]twenty-eight-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione Step 1. (4 S)-4-benzyl-3-[(2 R)-2-methylpent-4-enyl]-1,3-oxazolidin-2-one (M3-1). In a nitrogen atmosphere at -78 ° C to (4 STo a stirred mixture of 4-benzyl-3-propionyl-1,3-oxazolidin-2-one (100 g, 428.69 mmol, 1 equiv) in tetrahydrofuran (640 mL) were added LiHMDS (471 mL, 471.56 mmol, 1.1 equiv) and allyl bromide (207.4 g, 1714.77 mmol, 4 equiv) dropwise in sequence. The resulting mixture was stirred at -50 °C under nitrogen atmosphere for 2 h and warmed to room temperature. The reaction mixture was treated with saturated NH4Cl(aq) solution (1000 mL) and treated with CH 2Cl 2(3×400 mL) extraction. The combined organic layers were washed with brine (3×500 mL) and purified by anhydrous Na 2SO 4Dry. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography using petroleum ether/ethyl acetate (40/1) to obtain 100 g (4 S)-4-benzyl-3-[(2 R)-2-methylpent-4-enyl]-1,3-oxazolidin-2-one (M3-1) (85%). LCMS: m/z (ESI), [M+H] +=274.00. 1H NMR (DMSO-d 6, 400 MHz) δ 1.00-1.12 (3H, t), 2.08-2.49 (2H, m), 2.84-3.06 (2H, m), 3.64-3.78 (1H, m), 4.11-4.26 (1H, m), 4.29-4.39 (1H, m), 4.59-4.75 (1H, m), 4.98-5.18 (2H, m), 5.70-5.87 (1H, m), 7.13-7.38 (5H, m). Step 2. (2 R)-2-methylpent-4-enoic acid (M3-2) preparation. At 0℃, (4 S)-4-benzyl-3-[(2 R)-2-methylpent-4-enyl]-1,3-oxazolidin-2-one (M3-1, 100 g, 365.85 mmol, 1 equivalent) in tetrahydrofuran (800 mL) and H 2Add LiOH to the stirred mixture in O (200 mL).H 2O (46.05 g, 1097.56 mmol, 3 equiv.) and H 2O 2(30%) (114.30 mL, 4906 mmol, 13.4 equiv.). The resulting mixture was stirred at 0 °C for 3 h under a nitrogen atmosphere. The reaction was heated to 40 °C with Na 2S 2O 3The solution (250 mL) was quenched, acidified to pH = 1 with HCl solution, and washed with CH2Cl 2(3×400 mL) extraction. The combined organic layers were washed with brine (3×500 mL) and purified by anhydrous Na 2SO 4Dry. After filtration, concentrate the filtrate under reduced pressure. The crude product is used directly in the next step without further purification. Step 3. (2 R)- N,N- Preparation of dibenzyl-2-methylpent-4-enamide (M3-3). In a nitrogen atmosphere at room temperature to (2 R)-2-methylpent-4-enoic acid (50 g, 219.02 mmol, 1 equivalent) and dibenzylamine (43.2 g, 219.022 mmol, 1 equivalent) were added to a stirred solution in dioxane (200 mL) N,N-diisopropylethylamine (56.6 g, 438.04 mmol, 2 equiv) and N,N,N,N-tetramethyl-O-(7-azabenzotriazol-1-yl)uronium hexafluorophosphate (99.94 g, 262.82 mmol, 1.2 equiv). The resulting mixture was stirred for 2 h. The reaction was quenched with water (1000 mL), and the mixture was washed with CH2Cl 2(3×300 mL) extraction. The combined organic layers were washed with brine (3×500 mL) and purified by anhydrous Na 2SO 4Dry. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified on a silica gel column using petroleum ether/ethyl acetate (40/1) to obtain 55.6 g (2 R)- N,N-Dibenzyl-2-methylpent-4-enamide (M3-3) (86%). LCMS: m/z (ESI), [M+H] +=294.15. 1H NMR (DMSO-d 6, 400 MHz) δ 1.01 (3H, d), 1.94-2.39 (2H, m), 2.76-2.90 (1H, m), 4.35-4.67 (4H, m), 4.89-5.11 (2H, m), 5.58-5.81 (1H, m), 7.08-7.43 (10H, m). Step 4. (2 R)- N,N- Preparation of dibenzyl-5-hydroxy-2-methylpentylamide (M3-4). At 0°C, (2 R)- N,N-dibenzyl-2-methylpent-4-enamide (50 g, 170.41 mmol, 1 eq.) and 9-boraherobicyclo[3.3.1]nonane (852.05 mL, 426.02 mmol, 2.5 eq.) in tetrahydrofuran (60 mL) were added dropwise NaOH (10 M) (220 mL) and H 2O 2(30%) (151 mL). The resulting mixture was stirred at room temperature for 2 h. The reaction was treated with Na 2S 2O 4(aqueous solution) solution (500 mL) was quenched. The mixture was washed with CH 2Cl 2(3×500 mL) extraction, and the combined organic layers were purified by anhydrous Na 2SO 4Dry. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified on a silica gel column using petroleum ether/ethyl acetate (1/1) to obtain 42.5 g (2 R)- N,N-Dibenzyl-5-hydroxy-2-methylpentanamide (M3-4) (68%). LCMS: m/z (ESI), [M+H] +=312.10. 1H NMR (DMSO-d 6, 400 MHz) δ 1.01 (3H, d), 1.24-1.43 (3H, m), 1.56-1.70 (1H, m), 2.60-2.80 (1H, m), 3.26-3.33 (2H, m), 4.30-4.42 (1H, m), 4.44-4.64 (4H, m), 7.15-7.23 (4H,m), 7.23-7.43 (6H, m). Step 5. (4 R)-5-(dibenzylamino)-4-methylpentan-1-ol (M3-5). At 0°C, LiAlH 4(6.73 g, 177.41 mmol, 1.3 equiv.) was added to a stirred mixture of tetrahydrofuran (600 mL) (2R)- N,N-dibenzyl-5-hydroxy-2-methylpentanamide (42.5 g, 136.46 mmol, 1 eq.). The resulting mixture was stirred at room temperature for 2 h. The reaction was quenched with water (7 mL) and 15 mL NaOH (w/w, 30%) at 0 °C and washed with anhydrous Na 2SO 4Dry. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified on a silica gel column using petroleum ether/ethyl acetate (10/1) to obtain 27.9 g (4 R)-5-(Dibenzylamino)-4-methylpentan-1-ol (M3-5) (68%). LCMS: m/z (ESI), [M+H] +=298.15. 1H NMR (DMSO-d 6, 400 MHz) δ 0.81 (3H, d), 0.88-1.00 (1H, m), 1.13-1.46 (3H, m), 1.64-1.78 (1H, m), 2.03-2.27 (2H, m), 3.27- 3.39 (2H, m), 3.44 (2H, d), 3.49-3.59 (2H, d), 4.32 (1H, t), 7.18-7.26 (2H, m), 7.30-7.35 (8H, m). Step 6. (4 R)-5-amino-4-methylpentan-1-ol (M3-6). (4 RA mixture of )-5-(dibenzylamino)-4-methylpentan-1-ol (18 g, 60.51 mmol, 1 eq.) and Pd/C (1.29 g, 12.10 mmol, 0.2 eq.) in methanol (200 mL) was stirred at room temperature under a hydrogen atmosphere for 6 h. The resulting mixture was filtered, and the filter cake was washed with methanol (3×70 mL). The resulting solution was concentrated under reduced pressure to give 6.5 g (4 R)-5-amino-4-methylpentan-1-ol (M3-6) (91%).1H NMR (DMSO-d 6, 400 MHz) δ 0.83 (3H, d), 0.94-1.09 (2H, m), 1.25-1.54 (3H, m),2.26-2.49 (2H, m), 3.17 (1H, s), 3.37 (2H, t). Step 7. (4 RPreparation of )-5-[(5-bromo-2-nitrophenyl)amino]-4-methylpentan-1-ol (M3-7). 4-Bromo-2-fluoro-1-nitrobenzene (10.00 g, 45.46 mmol, 1 equivalent), K 2CO 3(12.56 g, 90.91 mmol, 2 equiv.) and (4 RA mixture of 5-amino-4-methylpentan-1-ol (6.39 g, 54.55 mmol, 1.2 equiv.) in acetonitrile (60 mL) was stirred at 60 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature, treated with water (500 mL), and extracted with dichloromethane (3×500 mL). The combined organic layers were washed with brine (3×500 mL) and purified by anhydrous Na 2SO 4After filtration, the filtrate was concentrated under reduced pressure and purified by prep-TLC using petroleum ether/ethyl acetate (1/3) to obtain 11.7 g (4 R)-5-[(5-bromo-2-nitrophenyl)amino]-4-methylpentan-1-ol (M3-7) (81%). LCMS: m/z (ESI), [M+H] +=319.00. 1H NMR (400 MHz, DMSO-d 6 ) δ 0.95 (3H, d), 1.14-1.27 (1H, m), 1.37-1.59 (3H, m), 1.77-1.88 (1H, m), 3.18-3.22 (1H, m), 3.24-3.34 (1H , m), 3.35-3.44 (2H, m), 4.39 (1H, t), 6.83-6.87 (1H, m), 7.26 (1H, d), 7.99 (1H, d), 8.20 (1H, t).Step 8. 5-(5-{[(4 RPreparation of methyl 5-[(5-bromo-2-nitrophenyl)amino]-4-methylpentyl]oxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (M3-8). To a stirred mixture of methyl 5-(5-hydroxy-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (M2, 2.67 g, 10.09 mmol, 1 equiv) and triphenylphosphine (6.62 g, 25.22 mmol, 2.5 equiv) in tetrahydrofuran (60 mL) at 0°C under a nitrogen atmosphere were added diisopropyl azodicarboxylate (4.08 g, 20.18 mmol, 2 equiv) and (4 R)-5-[(5-bromo-2-nitrophenyl)amino]-4-methylpentan-1-ol (3.21 g, 10.09 mmol, 1 equiv). After stirring at room temperature for 5 h, the mixture was treated with water (500 mL) and extracted with dichloromethane (3×500 mL). The combined organic layers were washed with brine (3×500 mL) and purified by anhydrous Na 2SO 4Dry. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by prep-TLC using petroleum ether/ethyl acetate (1/4) to obtain 2.72 g of 5-(5-{[(4 R)-5-[(5-bromo-2-nitrophenyl)amino]-4-methylpentyl]oxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (M3-8) (48%). LCMS: m/z (ESI), [M+H] +=564.10. 1H NMR (400 MHz, DMSO-d 6 ) δ 0.97 (3H, d), 1.45-1.29 (1H, m), 1.60-1.64 (1H, m), 1.78-1.82 (1H, m), 1.86 (2H, s), 3.38-3.15 (2H, m ), 3.58 (3H, s), 3.68 (3H, s), 3.78 (3H, s), 3.96 (2H, t), 6.83-6.88 (1H, m), 7.26 (1H, d), 8.02-7.95 ( 2H, m), 8.14 (1H, d), 8.21 (1H, t), 8.43 (1H, d). Step 9. 5-(5-{[(4 RPreparation of methyl 5-[(2-amino-5-bromophenyl)amino]-4-methylpentyl]oxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (M3-9). In a nitrogen atmosphere at 0 ° C, 5-(5-{[(4 R)-5-[(5-bromo-2-nitrophenyl)amino]-4-methylpentyl]oxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (4.00 g, 7.11 mmol, 1 equiv) and ranyl nickel (200 mg, 2.33 mmol, 0.33 equiv) were added to a stirring mixture of methanol (40 mL) with NH 2NH 2 .H 2O (712 mg, 14.22 mmol, 2 eq.). After stirring at room temperature for 3 h, the mixture was filtered and the filter cake was washed with methanol (3×200 mL). The organic solution was concentrated under reduced pressure and the residue was purified by prep-TLC eluting with dichloromethane/methanol (30/1) to give 3.65 g of 5-(5-{[(4 R)-5-[(2-amino-5-bromophenyl)amino]-4-methylpentyl]oxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (M3-9) (96%). LCMS: m/z (ESI), [M+H] +=533.95. Step 10. 5-(5-{[(4 RPreparation of methyl 4-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]pentyl]oxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (M3-10). At room temperature, 5-(5-{[(4 RTo a stirred mixture of methyl 5-[(2-amino-5-bromophenyl)amino]-4-methylpentyl]oxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (3.00 g, 5.63 mmol, 1 equiv) in dichloromethane (40 mL) was added cyanogen bromide (895 mg, 8.45 mmol, 1.5 equiv). After stirring at room temperature for 2 h, the mixture was concentrated under reduced pressure. The residue was purified by prep-TLC eluting with dichloromethane/methanol (20/1) to obtain 2.6 g of 5-(5-{[(4 R)-4-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]pentyl]oxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (M3-10) (83%). LCMS: m/z (ESI), [M+H] +=559.05. Step 11. 5-(5-{[(4 R)-4-{[2-amino-6-(4-methylpiperazine-1-yl)-1,3-benzodiazol-1-yl]methyl}pentyl]oxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (M3-11). At room temperature, 5-(5-{[(4 RTo a stirred mixture of methyl 4-{[2-amino-6-(4-methylpiperazine-1-yl)-1,3-benzodiazol-1-yl]methyl}pentyl]oxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (3.71 g, 6.42 mmol, 1 eq.) in tetrahydrofuran (100 mL) was added LiOH (307 mg, 12.83 mmol, 2 eq.) in H 2O (20 mL). After stirring at room temperature for 1 h, the reaction mixture was concentrated under reduced pressure. The residue was purified by reverse-phase flash chromatography (10% to 40% acetonitrile/water) to obtain 3.5 g of 5-(5-{[(4 R)-4-{[2-amino-6-(4-methylpiperazine-1-yl)-1,3-benzodiazol-1-yl]methyl}pentyl]oxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (M3-11) (97%). LCMS: m/z (ESI), [M+H] +=545.15. Step 12. (11 R)-16-bromo-5,11,26-trimethyl-7-oxa-4,5,13, 20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octadecene-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (M3). At room temperature, 5-(5-{[(4 R)-4-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]pentyl]oxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (1.2 g, 2.20 mmol, 1 equivalent) was added to the stirred mixture in dioxane (18 mL) N, N-diisopropylethylamine (0.85 g, 6.62 mmol, 3 equiv) and N,N,N,N-tetramethyl-O-(7-azabenzotriazol-1-yl)uronium hexafluorophosphate (1.26 g, 3.31 mmol, 1.5 equiv). After stirring at 60 °C for 3 h under nitrogen atmosphere, the reaction mixture was cooled to room temperature, treated with water (300 mL), and washed with CH2Cl 2(2×200 mL) extraction. The combined organic layers were washed with brine (3×300 mL) and purified by anhydrous Na 2SO 4Dry. After filtration, concentrate the filtrate under reduced pressure. The residue is subjected to prep-TLC using CH 2Cl 2/methanol (40/1) for purification to obtain 1.14 g (11 R)-16-bromo-5,11,26-trimethyl-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]octadecene-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (M3) (98%). LCMS: m/z (ESI), [M+H] +=525.15. 1H NMR (DMSO-d 6, 400 MHz) δ 0.81 (3H, d), 1.43 (1H, q), 1.91 (2H, d), 2.06-2.26 (1H, m), 2.78 (1H, br s), 3.62 (3H, s), 3.72 (3H, s), 3.89-4.00 (2H, m), 4.07-4.15 (1H, m), 4.31-4.38 (1H, m), 7.36 (1H, dd), 7.46 (1H, d), 7.88 (1H, d), 8.30 (1H, d), 8.35 (1H, s), 8.79 (1H, d), 12.67 (1H, s). Intermediates M4A and M4B 5-Bromo-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontahedral-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione, isomer 1 (M4A) and isomer 2 (M4B) Step 1. Preparation of benzylbis(ethoxymethyl)amine (M4-1). Benzylamine (500 g, 4666 mmol, 1 equivalent), POM (840.62 g, 9332 mmol, 2 equivalents) and K 2CO 3(644.88 g, 4666. mmol, 1 eq.) in ethanol (1200 mL) was stirred at room temperature under nitrogen atmosphere for 16 h. The resulting mixture was filtered. The filter cake was washed with dichloromethane (3×200 mL), and the organic solution was concentrated under reduced pressure to give 1200 g of benzylbis(ethoxymethyl)amine (M4-1) (80%) as a yellow oil. This crude material was used in step 3 without further purification. 1H NMR (DMSO- d 6, 400 MHz) δ 1.01-1.19 (6H, m), 3.39-3.57 (3H, m), 3.90 (2H, t), 4.55-4.82 (4H, m), 7.27-7.39 (5H, m). Step 2. Preparation of 2-oxocyclopentane-1-carboxylic acid isopropyl ester (M4-2). 2-Oxocyclopentane-1-carboxylic acid ethyl ester (400 g, 2561 mmol, 1 equivalent) and DMAP (31.29 g, 256.11 mmol, 0.1 equivalent) were added to i-PrOH (500 mL) was stirred at 80 °C overnight under nitrogen atmosphere. The mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified on a silica gel column eluted with petroleum ether/ethyl acetate (10/1) to give 400 g of 2-oxocyclopentane-1-carboxylic acid isopropyl ester (M4-2) (92%) as a yellow oil. LCMS: m/z (ESI), [M - H] +=168.85. 1H NMR (DMSO- d 6, 400 MHz) δ 1.26 (6H, t), 1.80-1.92 (1H, m), 2.07-2.17 (1H, m), 2.25-2.35 (4H, m), 3.11 (1H, t), 5.00-5.10 (1H, m). Step 3. Preparation of 3-benzyl-8-oxo-3-azabicyclo[3.2.1]octane-1-carboxylic acid isopropyl ester (M4-3). A solution of benzylbis(methoxymethyl)amine (M4-1, 601.1 g, 3078.5 mmol, 2.62 equiv) in DMF (1.5 L) was treated with methyltrichlorosilane (400.4 g, 2679 mmol, 2.28 equiv) under nitrogen atmosphere at 0 °C for 10 min, followed by the slow addition of isopropyl 2-oxocyclopentane-1-carboxylate (M4-2, 200 g, 1175 mmol, 1 equiv) at 0 °C. The resulting mixture was stirred at room temperature overnight under nitrogen atmosphere. The reaction was quenched with water (3 L) and treated with saturated NaHCO 3(aqueous solution) solution (3 L) was alkalized to pH 7, and the mixture was extracted with ethyl acetate (3×2 L). The combined organic layers were washed with brine (3×2 L) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column eluting with petroleum ether:ethyl acetate (40/1) and concentrated under vacuum. The residue was further purified by Prep-HPLC by reverse phase flash chromatography using a C18 silica gel column. The fractions containing the desired compound were evaporated to dryness to give 110 g of 3-benzyl-8-oxo-3-azabicyclo[3.2.1]octane-1-carboxylic acid isopropyl ester (M4-3) (31%) as a yellow oil. LCMS: m/z (ESI), [M+H] +=470.15. 1H NMR (DMSO- d 6, 400 MHz) δ 1.04-1.26 (6H, m), 1.84-1.96 (2H, m), 2.16-2.37 (3H, m), 2.45 (1H, d), 2.62 (1H, d), 2.94 (1H, d), 3.04 (1H, d), 3.65 (2H, s), 4.75-5.02 (1H, m), 7.13-7.48 (5H, m) Step 4. Preparation of 3-benzyl-8-[(4-methylbenzenesulfonylamino)imino]-3-azabicyclo[3.2.1]octane-1-carboxylic acid isopropyl ester (M4-4). A solution of 3-benzyl-8-oxo-3-azabicyclo[3.2.1]octane-1-carboxylic acid isopropyl ester (100 g, 331.8 mmol, 1 equivalent) in methanol (1 L) was added with TsNHNH in portions at 0°C under nitrogen atmosphere.2(123.58 g, 663.5 mmol, 2.00 equiv.). The resulting mixture was stirred at room temperature overnight under a nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified on a silica gel column and eluted with petroleum ether/ethyl acetate (5/1) to give 80 g of 3-benzyl-8-[(4-methylbenzenesulfonylamide)imino]-3-azabicyclo[3.2.1]octane-1-carboxylic acid isopropyl ester (M4-4) (51%) as a white solid. 1H NMR (DMSO- d 6, 400 MHz) δ 0.99-1.15 (6H, m), 1.57-1.85 (2H, m), 1.87-2.03 (1H, ddd), 2.02-2.13 (2H, m), 2.25 (1H, d), 2.39 (3H, s), 2.62-2.73 (1H, m),2.84(1H, d) 3.14-3.19 (1H, m), 3.06-3.10 (1H, m), 3.50 (2H, s), 4.75-4.91 (1H, m), 7.26-7.43 (7H, m), 7.67 (2H, d), 10.38 (1H, s). Step 5. Preparation of 3-benzyl-3-azabicyclo[3.2.1]octane-1-carboxylate (M4-5). To a stirred solution of 3-benzyl-8-[(4-methylbenzenesulfonylamide)imino]-3-azabicyclo[3.2.1]octane-1-carboxylic acid isopropyl ester (100 g, 212.95 mmol, 1 equivalent) in methanol (2 L) was added NaBH under nitrogen atmosphere at 0 °C.4(120.8 g, 3194.20 mmol, 15 equiv.). The mixture was stirred at 80 °C overnight and then cooled to room temperature. The reaction was quenched with water (1 L), and the mixture was extracted with ethyl acetate (3×1 L). The combined organic layers were washed with brine (3×1 L), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by silica gel chromatography and eluted with petroleum ether/ethyl acetate (5/1) to give 33.0 g of 3-benzyl-3-azabicyclo[3.2.1]octane-1-carboxylic acid isopropyl ester (M4-5) (54%) as a colorless oil. LCMS: m/z (ESI), [M+H] +=288.05. 1H NMR (DMSO-d 6, 400 MHz) δ 1.14 (6H, d), 1.41 (1H, d), 1.62-1.69 (2H, m), 1.70-1.90 (3H, m), 1.9-2.04 (1H, m), 2.12 (1H, d), 2.18 (1H, d), 2.58 (1H, d), 2.82 (1H, d), 3.50 (2H, d), 4.81-4.90 (1H, m), 7.06-7.51 (5H, m). Step 6. Preparation of 3-benzyl-3-azabicyclo[3.2.1]octane-1-carboxylic acid (M4-6). A mixture of 3-benzyl-3-azabicyclo[3.2.1]octane-1-carboxylic acid isopropyl ester (33.0 g, 114.82 mmol, 1 eq.) and LiOH (3.0 g, 126.30 mmol, 1.1 eq.) in ethanol (400 mL) was stirred at 80 °C for 2 days under nitrogen atmosphere. The mixture was cooled to room temperature and concentrated under reduced pressure to give 30 g of 3-benzyl-3-azabicyclo[3.2.1]octane-1-carboxylic acid (M4-6) (96%) as a yellow oil. LCMS: m/z (ESI), [M+H] +=246.05. Step 7. NPreparation of tert-butyl-{3-benzyl-3-azabicyclo[3.2.1]octane-1-yl}carbamate (M4-7). A mixture of 3-benzyl-3-azabicyclo[3.2.1]octane-1-carboxylic acid (30 g, 110.06 mmol, 1 eq., 90%), DPPA (36.35 g, 132.07 mmol, 1.2 eq.) and triethylamine (33.41 g, 330.18 mmol, 3.0 eq.) in tetrahydrofuran (300 mL) was stirred at 80 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature and concentrated under reduced pressure. Toluene (500 mL) and t-BuOK (18.53 g, 165.09 mmol, 1.5 equiv). The mixture was stirred at 100 °C for 2 h under nitrogen atmosphere and then cooled to room temperature. The reaction was quenched with water (1 L), and the mixture was extracted with ethyl acetate (3×1 L). The combined organic layers were washed with brine (3×1 L), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue is purified by silica gel chromatography eluting with petroleum ether/ethyl acetate (5/1) to give 15 g of a white solid.N-tert-butyl {3-benzyl-3-azabicyclo[3.2.1]octan-1-yl}carbamate (M4-7) (43%). LCMS: m/z (ESI), [M+H] +=317.15. 1H NMR (DMSO-d 6, 400 MHz) δ 1.23 (9H, s), 1.38-1.60 (3H, m), 1.59-1.76 (2H, m), 1.75-2.25 (5H, m), 2.87 (1H, d), 3.38-3.56 (2H, m), 6.88 (1H, s), 7.17-7.37 (5H, m) Step 8. N- Preparation of tert-butyl {3-azabicyclo[3.2.1]oct-1-yl}carbamate (M4-8). NA mixture of tert-butyl-{3-benzyl-3-azabicyclo[3.2.1]octan-1-yl}carbamate (15 g, 47.40 mmol, 1 equiv) and Pd/C (7.57 g, 71.13 mmol, 1.50 equiv) in methanol (300 mL) was stirred at room temperature under hydrogen atmosphere for 2 h. The resulting mixture was filtered and the filter cake was washed with methanol (3×200 mL). The organic solution was concentrated under reduced pressure to obtain 10 g of a white solid.N-tert-Butyl {3-azabicyclo[3.2.1]octan-1-yl}carbamate (M4-8) (93%). This material was used in the next step without further purification. LCMS: m/z (ESI), [M+H] +=227.10. 1H NMR (DMSO-d 6, 400 MHz) δ 1.36 (9H, d), 1.45-1.60 (2H, m), 1.60-1.75 (1H, m), 1.73-1.91 (1H, m), 1.93-2.00 (2H, m), 2.45 ( 2H, d), 2.76-2.89 (1H, m), 3.14-3.20 (1H, m), 6.73 (1H, d). Step 9. NPreparation of tert-butyl-(3-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-3-azabicyclo[3.2.1]oct-1-yl)carbamate (M4-9). Freshly prepared N-tert-butyl {3-azabicyclo[3.2.1]oct-1-yl}carbamate (10 g, about 44 mmol, about 1 equivalent), (2-bromoethoxy)(tert-butyl)dimethylsilane (21.14 g, 88.37 mmol, 2 equivalents), NaI (13.25 g, 88.37 mmol, 2 equivalents) and K 2CO 3A mixture of (12.21 g, 88.37 mmol, 2 equiv) in DMF (160 mL) was stirred at 60 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (300 mL), and the mixture was extracted with ethyl acetate (3×200 mL). The combined organic layers were washed with brine (3×300 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was subjected to prep-HPLC using a HPLC-HPLC system containing 0.1% NH4HCO 3of water and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to obtain 12 g of a colorless oil.N-tert-Butyl (3-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-3-azabicyclo[3.2.1]oct-1-yl)carbamate (M4-9) (70%). LCMS: m/z (ESI), [M+H] += 385.25. 1H NMR (DMSO-d 6, 400 MHz) δ 0.00 (6H, s), 0.82 (9H, s), 0.85 (1H, s), 1.32 (9H, s), 1.37-1.48 (2H, m), 1.53-1.64 (2H, m), 1.91-2.05 (4H, m), 2.36-2.43 (2H, m), 2.54-2.57 (1H, m), 2.86-2.92 (1H, m), 3.61 (2H, t), 6.80 (1H, s). Step 10. Preparation of 2-(1-amino-3-azabicyclo[3.2.1]octan-3-yl)ethan-1-ol (M4-10). NA mixture of tert-butyl-(3-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-3-azabicyclo[3.2.1]octan-1-yl)carbamate (12 g, 31.19 mmol, 1 equiv) in trifluoroacetic acid (28 mL) and dichloromethane (84 mL) was stirred at room temperature under nitrogen atmosphere for 2 h. The resulting mixture was concentrated under reduced pressure. The crude product 2-(1-amino-3-azabicyclo[3.2.1]octan-3-yl)ethan-1-ol (M4-10) as an orange oil was used directly in the next step without further purification. LCMS: m/z (ESI), [M+H] += 285.15. Step 11. Preparation of 2-[1-[(5-bromo-2-nitrophenyl)amino]-3-azabicyclo[3.2.1]octan-3-yl]ethanol (M4-11). 2-(1-amino-3-azabicyclo[3.2.1]octan-3-yl)ethan-1-ol (crude, M4-10), K 2CO 3A mixture of (21.56 g, 156.25 mmol, 5 equiv.) and 4-bromo-2-fluoro-1-nitrobenzene (20.53 g, 93.73 mmol, 3 equiv.) in DMSO (160 mL) was stirred at 120 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (300 mL), and the mixture was extracted with ethyl acetate (3×200 mL). The combined organic layers were washed with brine (3×300 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The crude product was purified by prep-HPLC using HPLC containing 0.1% NH4HCO 3of water and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 12 g of 2-[1-[(5-bromo-2-nitrophenyl)amino]-3-azabicyclo[3.2.1]octan-3-yl]ethanol (M4-11) (76%) as an orange semisolid. LCMS: m/z (ESI), [M+H] += 371.95. 1H NMR (DMSO-d 6, 400 MHz) δ 1.68 (3H, d), 1.82-1.85 (1H, m), 1.94 (1H, s), 2.12 (1H, d), 2.17 (1H, d), 2.21 (1H, s), 2.36 (1H, s), 2.49 (2H, d), 2.65-2.73 (1H, m), 3.10-3.20 (1H, m), 3.47-3.57 (2H, m), 4.36 (1H, t), 6.86-3.89 (1H, m), 7.21 (1H, d), 8.01 (1H, d), 8.31 (1H, s). Step 12. Preparation of methyl 5-[5-(2-{1-[(5-bromo-2-nitrophenyl)amino]-3-azabicyclo[3.2.1]oct-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylate (M4-12). To a stirred mixture of 2-{1-[(5-bromo-2-nitrophenyl)amino]-3-azabicyclo[3.2.1]octan-3-yl}ethanol (8 g, 21.61 mmol, 1 eq) and triphenylphosphine (17.00 g, 64.82 mmol, 3 eq) in tetrahydrofuran (500 mL) was added diisopropyl azodicarboxylate (12.23 g, 60.50 mmol, 2.8 eq) and methyl 5-(5-hydroxy-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (5.69 g, 21.61 mmol, 1 eq) slowly under nitrogen atmosphere at 0° C. The resulting mixture was stirred at 0° C. for 2 h under nitrogen atmosphere. The reaction was quenched with water (300 mL), and the mixture was extracted with ethyl acetate (3×200 mL). The combined organic layers were washed with brine (3×300 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column using petroleum ether/ethyl acetate (1/3) to give 9.5 g of 5-[5-(2-{1-[(5-bromo-2-nitrophenyl)amino]-3-azabicyclo[3.2.1]octan-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (M4-12) (71%) as an orange oil. LCMS: m/z (ESI), [M+H] +=617.10. Step 13. Preparation of 5-[5-(2-{1-[(2-amino-5-bromophenyl)amino]-3-azabicyclo[3.2.1]oct-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (M4-13). To a mixture of methyl 5-[5-(2-{1-[(5-bromo-2-nitrophenyl)amino]-3-azabicyclo[3.2.1]octan-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylate (10.5 g, 17.06 mmol, 1 eq) and ranyl nickel (2 g) in methanol (150 mL) was added hydrazine hydrate (2.19 g, 68.24 mmol, 1.5 eq) and stirred at 0 °C for 2 h under nitrogen atmosphere. The resulting mixture was filtered and the filter cake was washed with dichloromethane (3 x 200 mL). The organic solution was concentrated under reduced pressure. The residue was purified on a silica gel column using dichloromethane/methanol (10/1) to give 9.5 g of 5-[5-(2-{1-[(2-amino-5-bromophenyl)amino]-3-azabicyclo[3.2.1]oct-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (M4-13) (91%) as a brown oil. LCMS: m/z (ESI), [M+H] +=585.20. 1H NMR (DMSO-d 6, 400 MHz) δ 1.45-1.60 (3H, m), 1.70-1.85 (2H, m), 1.95-2.17 (4H, m), 2.65-2.80 (3H, m), 3.09-3.15 (1H, m), 3.17 (2H, d), 3.57 (3H, s), 3.74 (3H, s), 3.79 (3H, s), 4.00-4.10 (3H, m), 4.52 (1H, s), 4.75 (2H, s), 6.44-6.50 (1H, m), 6.50-6.56 (1H, m), 6.64 (1H, d), 7.97 (1H, s), 8.10 (1H, d), 8.43 (1H, d). Step 14. Preparation of 5-(5-{2-[1-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (M4-14). A solution of methyl 5-[5-(2-{1-[(2-amino-5-bromophenyl)amino]-3-azabicyclo[3.2.1]octan-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylate (10 g, 17.08 mmol, 1 eq) and cyanogen bromide (2.17 g, 20.49 mmol, 1.20 eq) in ethanol (200 mL) was stirred at room temperature overnight under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure, and the residue was purified on a silica gel column using dichloromethane/methanol (20/1) for elution to obtain 9.5 g of 5-(5-{2-[1-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]octan-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (M4-14) (91%) as a brown solid. LCMS: m/z (ESI), [M+H] +=612.05. Step 15. Preparation of 5-(5-{2-[1-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester, isomer 1 (M4-14A) and isomer 2 (M4-14B). Chiral chromatography was performed using a (R, R)-WHELK-01-Kromasil column (5 * 25 cm, 5 μm), CO 2The racemic mixture of M4-14 (9 g, 14.74 mmol) was separated using methanol/acetonitrile (1/1) as mobile phase A and 3.6 g of isomer 1 (M4-14A, 41%) and 3.9 g of isomer 2 (M4-14B, 43%) as brown solids. SFC-HPLC, Rt (isomer 1) = 2.246 min, Rt (isomer 2) = 3.898 min. Step 16. Preparation of 5-(5-(2-(1-(2-amino-6-bromo-1H-benzo[d]imidazol-1-yl)-3-azabicyclo[3.2.1]octan-3-yl)ethoxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxo-1,6-dihydropyridine-3-carboxylic acid (Isomer 1, M4-15A). To a stirred solution of 5-(5-(2-(1-(2-amino-6-bromo-1H-benzo[d]imidazol-1-yl)-3-azabicyclo[3.2.1]octan-3-yl)ethoxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxo-1,6-dihydropyridine-3-carboxylic acid methyl ester (isomer 1, M4-14A, 4 g, 6.55 mmol, 1 eq) in tetrahydrofuran (8 mL) was added LiOH (0.31 g, 13.10 mmol, 2 eq) in H 2O (2 mL). The resulting mixture was stirred at room temperature for 2 h. The reaction was concentrated under reduced pressure, and the residue was purified by Prep-HPLC by reversed phase flash chromatography using a C18 silica gel column eluting with water and acetonitrile. The fractions containing the desired compound were evaporated to dryness to give 3.6 g of 5-(5-(2-(1-(2-amino-6-bromo-1H-benzo[d]imidazol-1-yl)-3-azabicyclo[3.2.1]octan-3-yl)ethoxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxo-1,6-dihydropyridine-3-carboxylic acid (Isomer 1, M4-15A) (92%) as a white solid. LCMS: m/z (ESI), [M+H] +=598.05. Step 17. Preparation of 5-bromo-15,21-dimethyl-23-oxo-2,9,11,15,20,21, 26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]tris-3,5,7,9,13,17(33), 18(22),19-octaene-12,16-dione (M4A). 5-(5-{2-[1-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (M4-15A, 3.6 g, 6.03 mmol, 1 equivalent), N,N,N,N-Tetramethyl- O-(7-Azabenzotriazol-1-yl)urea hexafluorophosphate (3.44 g, 9.05 mmol, 1.5 equivalents) and N,N- A mixture of diisopropylethylamine (2.34 g, 18.10 mmol, 3 equiv.) in 1,4-dioxane (80 mL) was stirred at room temperature under nitrogen atmosphere for 2 h. The reaction was quenched with water (300 mL), and the mixture was extracted with ethyl acetate (3×200 mL). The combined organic layers were washed with brine (3×300 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column using dichloromethane/methanol (20/1) to give 3.3 g of 5-bromo-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontahedral-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione (isomer 1, M4A) (94%) as a brown solid. LCMS: m/z (ESI), [M+H] += 580.00. SFC-HPLC, Rt=5.498, 1H NMR (DMSO-d 6, 400 MHz) δ 1.56-1.81 (3H, m), 2.01 (1H, d), 2.42 (2H, d), 2.54 (1H, d), 2.63 (1H, s), 2.69-2.75 (1H, m), 3.09-3.20 (2H, m), 3.19-3.30 (1H, m), 3.63 (3H, s), 3.72 (3H, s), 4.54 (1H, t), 4.63 (1H, d), 7.36 (1H, d), 7.51 (1H, d), 7.82 (1H, d), 8.16 (1H, s), 8.30 (1H, d), 8.41 (1H, s), 8.81 (1H, d), 12.84 (1H, s). Step 18. Preparation of 5-(5-(2-(1-(2-amino-6-bromo-1H-benzo[d]imidazol-1-yl)-3-azabicyclo[3.2.1]octan-3-yl)ethoxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxo-1,6-dihydropyridine-3-carboxylic acid (isomer 2, M4-15B). To a stirred solution of 5-(5-(2-(1-(2-amino-6-bromo-1H-benzo[d]imidazol-1-yl)-3-azabicyclo[3.2.1]octan-3-yl)ethoxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxo-1,6-dihydropyridine-3-carboxylic acid methyl ester (isomer 2, M4-14B, 4.2 g, 6.88 mmol, 1 eq.) in tetrahydrofuran (80 mL) was added LiOH (0.33 g, 13.76 mmol, 2 eq.) in H 2O (20 mL). The resulting mixture was stirred at room temperature for 2 h. The reaction was concentrated under reduced pressure, and the residue was further purified by Prep-HPLC by reversed phase flash chromatography using a C18 silica gel column eluting with water and acetonitrile. The fractions containing the desired compound were evaporated to dryness to give 3.8 g of 5-(5-(2-(1-(2-amino-6-bromo-1H-benzo[d]imidazol-1-yl)-3-azabicyclo[3.2.1]octan-3-yl)ethoxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxo-1,6-dihydropyridine-3-carboxylic acid (isomer 2, M4-15B) (92%) as a white solid. LCMS: m/z (ESI), [M+H] +=598.05. Step 19. Preparation of 5-bromo-15,21-dimethyl-23-oxa-2,9,11,15,20,21, 26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]tris-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione (isomer 2, M4B). 5-(5-{2-[1-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (M4-15B, 3.8 g, 6.37 mmol, 1 equivalent), N,N,N,N-Tetramethyl- O-(7-Azabenzotriazol-1-yl)urea hexafluorophosphate (3.63 g, 9.56 mmol, 1.5 equivalents) and N,N- A solution of diisopropylethylamine (2.47 g, 19.11 mmol, 3 equivalents) in 1,4-dioxane (80 mL) was stirred at room temperature under nitrogen atmosphere for 2 h. The reaction was quenched with water (300 mL), and the mixture was extracted with ethyl acetate (3×200 mL). The combined organic layers were washed with brine (3×300 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column using dichloromethane/methanol (20/1) to give 3.5 g of 5-bromo-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontahedral-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione (isomer 2, M4B) (94%) as a brown solid. LCMS: m/z (ESI), [M+H] += 580.00. SFC-HPLC, Rt=3.186, 1H NMR (DMSO-d 6, 400 MHz) δ 1.55-1.76 (2H, m), 2.01 (1H, d), 2.39-2.47 (2H, m), 2.51-2.57 (1H, m), 2.61-2.65 (1H, m), 2.70-2.77 (2H, m), 3.09-3.18 (2H, m), 3.19-3.29 (1H, m), 3.63 (3H, s), 3.72 (3H, s), 4.54 (1H, t), 4.63 (1H, d), 7.36 (1H, dd), 7.51 (1H, d), 7.82 (1H, d), 8.16 (1H, s), 8.30 (1H, d), 8.41 (1H, s), 8.81 (1H, d), 12.84 (1H, s) Intermediate M5 5-Bromo-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic [26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontria-3,5,7,9,13,17(34),18(22),19-octaene-12,16-dione Step 1. Preparation of ethyl 4-methylenecyclohexane-1-carboxylate (M5-1). To a mixture of methyltriphenylphosphonium bromide (314.81 g, 881.27 mmol, 1.5 equiv.) and potassium tert-butoxide (131.85 g, 1175.03 mmol, 2 equiv.) in tetrahydrofuran (500 mL) at 0°C was added ethyl 4-oxocyclohexane-1-carboxylate (100 g, 587.51 mmol, 1 equiv.). The mixture was stirred at room temperature under a nitrogen atmosphere for 2 h. The reaction was quenched with water (300 mL), and the mixture was extracted with ethyl acetate (3×200 mL). The combined organic layers were washed with brine (3×300 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by silica gel chromatography using petroleum ether/ethyl acetate (5/1) to give 80 g of ethyl 4-methylenecyclohexane-1-carboxylate (M5-1) (85%) as a yellow oil. 1H NMR (DMSO-d 6, 400 MHz) δ 1.18 (3H, t), 1.30-1.50 (2H, m), 1.80-1.95 (2H, m), 1.96-2.15 (2H, m), 2.15-2.35 (2H, m), 2.42-2.49 (1H, m), 4.04-4.08 (2H, m), 4.64 (2H, t). Step 2. Preparation of ethyl 1-(4-methylbenzenesulfonyl)-1-azaspiro[2.5]octane-6-carboxylate (M5-2). To a mixture of ethyl 4-methylenecyclohexane-1-carboxylate (80 g, 475.52 mmol, 1 eq.) and chloramine-T (216.50 g, 951.04 mmol, 2 eq.) in acetonitrile (800 mL) was added at 0°C.N,N,N-Trimethylphenylammonium dibromoethane (17.88 g, 47.55 mmol, 0.1 equiv). The mixture was stirred at room temperature overnight under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel chromatography using petroleum ether/ethyl acetate (5/1) to give 60 g of ethyl 1-(4-methylbenzenesulfonyl)-1-azaspiro[2.5]octane-6-carboxylate (M5-2) (33%) as a white solid. LCMS: m/z (ESI), [M+H] +=338.05. 1H NMR (DMSO-d 6, 400 MHz) δ 1.18 (3H, t), 1.54-1.58 (1H, m), 1.58-1.72 (1H, m), 1.72-1.93 (4H, m), 2.40 (3H, s), 2.50-2.53(2H, m), 4.05-4.09 (2H, m), 7.43 (2H, d), 7.73-7.79 (2H, m). Step 3. Preparation of ethyl 4-[(benzylamino)methyl]-4-(4-methylbenzenesulfonylamide)cyclohexane-1-carboxylate (M5-3). A solution of ethyl 1-(4-methylbenzenesulfonyl)-1-azaspiro[2.5]octane-6-carboxylate (60 g, 177.81 mmol, 1 eq) and benzylamine (28.58 g, 266.72 mmol, 1.5 eq) in tetrahydrofuran (600 mL) was stirred at 80 °C for 2 days under a nitrogen atmosphere. The mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by silica gel chromatography using petroleum ether/ethyl acetate (1/1) to give 60 g of ethyl 4-[(benzylamino)methyl]-4-(4-methylbenzenesulfonamido)cyclohexane-1-carboxylate (M5-3) (75%) as a white oil. LCMS: m/z (ESI), [M+H] +=445.20. 1H NMR (DMSO-d 6, 400 MHz) δ 1.15 (3H, t), 1.31-1.44 (2H, m), 1.48-1.76 (4H, m), 2.00 (1H, m), 2.21-2.30 (1H, m), 2.30 (2H, s), 2.33-2.37 (2H, m), 3.43 (2H, s), 4.02 (2H, m), 7.15-7.25 (3H, m), 7.25-7.36 (4H, m), 7.67-7.75 (2H, m). Step 4. Preparation of lithium 4-((benzylamino)methyl)-4-((4-methylphenyl)sulfonylamino)cyclohexane-1-carboxylate (M5-4). A mixture of ethyl 4-[(benzylamino)methyl]-4-(4-methylbenzenesulfonamido)cyclohexane-1-carboxylate (60 g, 134.95 mmol, 1 eq) and LiOH (6.46 g, 269.91 mmol, 2 eq) in ethanol (500 mL) was stirred at 80 °C for 16 h under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure to give 57 g of lithium 4-((benzylamino)methyl)-4-((4-methylphenyl)sulfonamido)cyclohexane-1-carboxylate (M5-4) as a white solid. This material was used in the next step without further purification. LCMS: m/z (ESI), [M+H] +=417.10. Step 5. N-{3-benzyl-4-oxo-3-azabicyclo[3.2.2]non-1-yl}-4-toluenesulfonamide (M5-5). N,N,N,N-Tetramethyl- O-(7-Azabenzotriazol-1-yl)urea hexafluorophosphate (77.03 g, 202.60 mmol, 1.5 equivalents), 4-((benzylamino)methyl)-4-((4-methylphenyl)sulfonylamino)cyclohexane-1-carboxylate lithium (57 g, about 135 mmol, about 1 equivalent) and N,N- A mixture of diisopropylethylamine (52.37 g, 405.21 mmol, 3 equivalents) in dichloromethane (150 mL) was stirred at room temperature overnight under a nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography using petroleum ether/ethyl acetate (1/1) to obtain 50 g of a white solid.N-{3-Benzyl-4-oxo-3-azabicyclo[3.2.2]non-1-yl}-4-toluenesulfonamide (M5-5) (93%). LCMS: m/z (ESI), [M+H] += 399.15. 1H NMR (DMSO-d 6, 400 MHz) δ 1.64-1.74 (8H, m), 2.38 (3H, s), 3.18 (2H, s), 4.37 (2H, s), 7.01-7.19 (2H, m), 7.21-7.41 (5H, m), 7.48-7.56 (2H, m), 7.73 (1H, s) Step 6. NPreparation of -{3-benzyl-3-azabicyclo[3.2.2]non-1-yl}-4-toluenesulfonylamide methyl (M5-6). Add lithium aluminum hydroxide (7.62 g, 200.74 mmol, 2 equivalents) in tetrahydrofuran (400 mL) at 0°C.N-{3-benzyl-4-oxo-3-azabicyclo[3.2.2]non-1-yl}-4-toluenesulfonamide (40 g, 100.37 mmol, 1 equiv) was added and the mixture was stirred under nitrogen atmosphere for 2 h. The reaction was quenched with water (7 mL) and 15 mL NaOH (w/w, 30%) at 0 °C. The mixture was washed with anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column using petroleum ether/ethyl acetate (1/1) to obtain 20 g of a white solid.N-{3-Benzyl-3-azabicyclo[3.2.2]non-1-yl}-4-toluenesulfonamide (M5-6) (46%). LCMS: m/z (ESI), [M+H] +=385.15. 1H NMR (DMSO-d 6, 400 MHz) δ 1.39-1.63 (6H, m), 1.71-1.77 (1H, m), 1.90-2.02 (2H, m), 2.36 (3H, s), 2.40 (2H, d), 2.64 (2H, s), 3.44 (2H, s), 7.18-7.31 (3H, m), 7.31-7.40 (4H, m), 7.39 (1H, s), 7.55-7.62 (2H, m). Step 7. N-{3-nitrobicyclo[3.2.2]non-1-yl}-4-toluenesulfonamide (M5-7). Pd/C (10%, 500 mg) and NA mixture of -{3-benzyl-3-azabicyclo[3.2.2]non-1-yl}-4-toluenesulfonamide (20 g, 52.01 mmol, 1 equiv) in methanol (200 mL) was stirred at room temperature under hydrogen pressure for 4 h. The resulting mixture was filtered. The filter cake was washed with methanol (3×70 mL), and the solution was concentrated under reduced pressure. The residue was purified on a silica gel column using dichloromethane/methanol (10/1) to give 15 g of white solid.N-{3-Azabicyclo[3.2.2]non-1-yl}-4-toluenesulfonamide (M5-7) (93%). LCMS: m/z (ESI), [M+H] +=295.00. 1H NMR (DMSO-d 6, 400 MHz) δ 1.35-1.60 (6H, m), 1.63-1.69 (1H, m), 1.79-1.83 (2H, m), 2.37 (3H, s), 2.59 (2H, d), 2.80 (2H, s), 3.17 (1H, d), 7.29-7.41 (3H, m), 7.51-7.79 (2H, m). Step 8. N-(3-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-3-azabicyclo[3.2.2]non-1-yl)-4-toluenesulfonamide (M5-8). N-{3-nitrobicyclo[3.2.2]non-1-yl}-4-toluenesulfonamide (15 g, 50.94 mmol, 1 equivalent), NaI (7.64 g, 50.94 mmol, 1 equivalent), K 2CO 3A mixture of (21.12 g, 152.84 mmol, 3 equiv.) and (2-bromoethoxy)(tert-butyl)dimethylsilane (14.63 g, 61.13 mmol, 1.2 equiv.) in dimethylformamide (150 mL) was stirred at 60 °C for 16 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (800 mL), and the mixture was extracted with ethyl acetate (3×600 mL). The combined organic layers were washed with brine (3×500 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column using dichloromethane/methanol (10/1) to give 20 g of yellow solid.N-(3-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-3-azabicyclo[3.2.2]non-1-yl)-4-toluenesulfonamide (M5-8) (78%). LCMS: m/z (ESI), [M+H] += 453.30. 1H NMR (DMSO-d 6, 400 MHz) δ 0.00 (6H, s), 0.83 (9H, s), 1.32-1.42 (2H, m), 1.44-1.54 (4H, m), 1.65-1.71 (1H, m), 1.80-1.90 (2H, m), 2.34 (3H, s), 2.38 (2H, t), 2.44 (2H, d), 2.65 (2H, s), 3.54 (2H, t), 7.32 (2H, d), 7.37 (1H, s), 7.59-7.76 (2H, m). Step 9. Preparation of 3-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-3-azabicyclo[3.2.2]nonan-1-amine (M5-9).NA mixture of -(3-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-3-azabicyclo[3.2.2]non-1-yl)-4-toluenesulfonamide (5 g, 11.04 mmol, 1 eq.) and Mg (5.37 g, 220.88 mmol, 20 eq.) in methanol (200 mL) was stirred at 70 °C overnight under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was terminated by adding saturated NH4Cl(aq) solution (300 mL) was quenched, and the mixture was extracted with ethyl acetate (3×300 mL). The combined organic layers were washed with brine (3×400 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column using dichloromethane/methanol (5/1) to give 2.3 g of 3-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-3-azabicyclo[3.2.2]nonan-1-amine (M5-9) (60%) as a yellow oil. 1H NMR (DMSO-d 6, 400 MHz) δ 0.00 (6H, s), 0.82 (9H, s), 1.54-1.64 (4H, m), 1.79-1.83 (1H, m), 1.85-1.91 (2H, m), 2.42-2.52 (3H, m), 2.54 (2H, d), 2.60 (2H, s), 3.63 (2H, t). Step 10. N-(5-bromo-2-nitrophenyl)-3-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-3-azabicyclo[3.2.2]nonan-1-amine (M5-10). 3-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-3-azabicyclo[3.2.2]nonan-1-amine (1 g, 3.35 mmol, 1 equivalent), 4-bromo-2-fluoro-1-nitrobenzene (1.84 g, 8.37 mmol, 2.5 equivalents) and K 2CO 3A mixture of (1.39 g, 10.05 mmol, 3 equiv.) in dimethyl sulfoxide (10 mL) was stirred at 120 °C for 4 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (50 mL), and the mixture was extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue is purified by silica gel chromatography eluting with petroleum ether/ethyl acetate (5/1) to give 1.5 g of yellow oil.N-(5-bromo-2-nitrophenyl)-3-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-3-azabicyclo[3.2.2]nonan-1-amine (M5-10) (85%). LCMS: m/z (ESI), [M+H]+=498.15. Step 11. Preparation of 2-{1-[(5-bromo-2-nitrophenyl)amino]-3-azabicyclo[3.2.2]nonan-3-yl}ethanol (M5-11). NA solution of -(5-bromo-2-nitrophenyl)-3-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-3-azabicyclo[3.2.2]nonan-1-amine (1.5 g, 3.00 mmol, 1 eq.) and tetrabutylammonium fluoride (1.57 g, 6.01 mmol, 2 eq.) in tetrahydrofuran (15 mL) was stirred at room temperature for 2 h under nitrogen atmosphere. The reaction was heated with saturated NaHCO3(aqueous solution) solution (100 mL) was quenched, and the mixture was extracted with dichloromethane (3×100 mL). The combined organic layers were washed with brine (3×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by silica gel chromatography using dichloromethane/methanol (10/1) to give 1.2 g of 2-{1-[(5-bromo-2-nitrophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethanol (M5-11) (93%) as a yellow oil. LCMS: m/z (ESI), [M+H] += 384.00. Step 12. Preparation of 5-[5-(2-{1-[(5-bromo-2-nitrophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (M5-12). To a stirred mixture of methyl 5-(5-hydroxy-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (M2, 0.66 g, 2.49 mmol, 0.8 eq.) and triphenylphosphine (2.46 g, 9.369 mmol, 3 eq.) in tetrahydrofuran (10 mL) at 0° C. were added diisopropyl azodicarboxylate (1.89 g, 9.36 mmol, 3 eq.) and methyl 5-(5-hydroxy-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (0.66 g, 2.49 mmol, 0.8 eq.). The mixture was stirred at 0° C. for 2 h under nitrogen atmosphere. The reaction was quenched with water (50 mL), and the mixture was extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×50 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC and eluted with petroleum ether/ethyl acetate (1/1) to give 1.2 g of 5-[5-(2-{1-[(5-bromo-2-nitrophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (M5-12) (57%) as a yellow oil. LCMS: m/z (ESI), [M+H] +=629.25. 1H NMR (DMSO-d 6, 400 MHz) δ 1.58-1.76 (9H, m), 2.56-2.79 (2H, m), 2.90 (4H, q), 3.56 (3H, s), 3.72 (6H, d), 4.13 (2H, t), 6.70-6.85 (1H, m), 7.01 (1H, d), 7.95-8.12 (4H, m), 8.28 (1H, d). Step 13. Preparation of 5-[5-(2-{1-[(2-amino-5-bromophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (M5-13). To a stirred mixture of ranyl nickel (0.90 g) and methyl 5-[5-(2-{1-[(5-bromo-2-nitrophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylate (1.1 g, 1.74 mmol, 1 equiv) in methanol (8 mL) at 0°C under nitrogen atmosphere was added NH2NH 2.H 2O (47 mg, 0.95 mmol, 2 eq.). The mixture was stirred at 0 °C for 1 h. The resulting mixture was filtered. The filter cake was washed with methanol (2×20 mL), and the solution was concentrated under reduced pressure. The residue was purified by Prep-TLC and eluted with dichloromethane/methanol (10/1) to give 910 mg of 5-[5-(2-{1-[(2-amino-5-bromophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (M5-13) (76%) as a light yellow solid. LCMS: m/z (ESI), [M+H] +=599.20. Step 14. Preparation of 5-(5-{2-[1-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.2]non-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (M5-14). A mixture of methyl 5-[5-(2-{1-[(2-amino-5-bromophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylate (900 mg, 1.50 mmol, 1 eq) and BrCN (174 mg, 1.65 mmol, 1.1 eq) in ethanol (10 mL) was stirred under nitrogen atmosphere at room temperature for 2 h. The mixture was concentrated under vacuum. The residue was purified by Prep-TLC and eluted with dichloromethane/methanol (12/1) to give 640 mg of 5-(5-{2-[1-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.2]non-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (M5-14) (68%) as a light yellow solid. LCMS: m/z (ESI), [M+H] +=624.10. 1H NMR (DMSO-d 6, 400 MHz) δ 1.24 (1H, s), 1.78-1.95 (8H, m), 2.60-2.74 (2H, m), 2.85-2.95 (4H, m), 3.57 (3H, s), 3.71 (6H, d), 4.08-4.17 (2H, m), 5.95 (2H, s), 7.04 (2H, s), 7.47 (1H, s), 7.96 (1H, s), 8.12 (1H, ), 8.39 (1H, d). Step 15. Preparation of 5-(5-{2-[1-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.2]non-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (M5-15). 5-(5-{2-[1-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.2]non-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (620 mg, 0.99 mmol, 1 eq.) and LiOH (28 mg, 1.19 mmol, 1.2 eq.) were dissolved in tetrahydrofuran/H 2O (4 mL/1 mL) was stirred at 80 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature and concentrated under vacuum to obtain 550 mg of 5-(5-{2-[1-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.2]non-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (M5-15) (90%) as a white solid. LCMS: m/z (ESI), [M+H] +=610.10. Step 16. Preparation of 5-bromo-15,21-dimethyl-23-oxo-2,9,11,15,20,21, 26-heptaazaheptacyclo[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]triacontriacontria-3,5,7,9,13,17(34),18(22),19-octaene-12,16-dione (M5). 5-(5-{2-[1-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.2]non-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (550 mg, 0.90 mmol, 1 equivalent), N,N,N,N-Tetramethyl-O-(7-azabenzotriazol-1-yl)urea hexafluorophosphate (1027 mg, 2.70 mmol, 3 equivalents) and N,N- A mixture of diisopropylethylamine (174 mg, 1.35 mmol, 1.5 equiv.) in dioxane (6 mL) was stirred at 60 °C for 2 h under a nitrogen atmosphere. The mixture was cooled to room temperature and concentrated under vacuum. The residue was purified by Prep-HPLC using an XBridge Prep OBD C18 column with 10 mmol/L NH4HCO 3of water and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 500 mg of 5-bromo-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontriacontria-3,5,7,9,13,17(34),18(22),19-octaene-12,16-dione (M5) as a yellow solid (88%). LCMS: m/z (ESI), [M+H] +=592.15. Synthesis Example Example A1 5,26-Dimethyl-10-(2,2,2-trifluoroethyl)-7-oxa-4,5,10,13,20,22, 26-heptaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]twenty-eight-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione Step 1. Preparation of 2-((2-((2-nitrophenyl)amino)ethyl)amino)ethan-1-ol (INT-A1-1). o-Fluoronitrobenzene (10 g, 70.871 mmol, 1 equivalent), K 2CO 3A mixture of (19.59 g, 141.742 mmol, 2 equiv) and aminoethylethanolamine (14.76 g, 141.742 mmol, 2 equiv) in ACN (300 mL) was stirred at 60 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (800 mL), and the mixture was extracted with ethyl acetate (3×600 mL). The combined organic layers were washed with brine (3×500 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by silica gel chromatography and eluted with PE/EA (1:1) to give 8.5 g of 2-({2-[(2-nitrophenyl)amino]ethyl}amino)ethanol (INT-A1-1) (53%) as a yellow solid. 1H NMR (DMSO-d 6, 400 MHz) δ 1.99 (1H, s), 2.62 (2H, t), 2.84 (2H, t), 3.36-3.40 (2H, m), 3.44-3.49 (2H, m), 4.50 (1H, t), 6.65-3.71 (1H, m), 7.04-7.08 (1H, m), 7.54 (1H, m), 8.34-8.38 (1H, t), 8.04-8.08 (1H, m). Step 2. Preparation of 2-({2-[(2-nitrophenyl)amino]ethyl}(2,2,2-trifluoroethyl)amino)ethanol (INT-A1-2). 2-({2-[(2-nitrophenyl)amino]ethyl}amino)ethanol (INT-A1-1, 2 g, 8.879 mmol, 1 equivalent), N,N-Diisopropylethylamine (2.30 g, 17.758 mmol, 2 equivalents) and 2,2,2-trifluoroethyl trifluoromethanesulfonate (2.47 g, 10.655 mmol, 1.2 equivalents) were added to N,N-The mixture in dimethylformamide (20 mL) was stirred at 60 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (200 mL), and the mixture was extracted with dichloromethane (3×200 mL). The combined organic layers were washed with brine (3×200 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by silica gel chromatography using petroleum ether/ethyl acetate (1/1) to give 2.19 g of 2-({2-[(2-nitrophenyl)amino]ethyl}(2,2,2-trifluoroethyl)amino)ethanol (INT-A1-2) (80%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=308.10. Step 3. Preparation of 1-methyl-5-{1-methyl-5-[2-({2-[(2-nitrophenyl)amino]ethyl}(2,2,2-trifluoroethyl)amino)ethoxy]pyrazol-4-yl}-6-oxopyridine-3-carboxylic acid methyl ester (INT-A1-3). To a stirred mixture of methyl 5-(5-hydroxy-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (M2, 0.56 g, 2.14 mmol, 1 eq) and triphenylphosphine (1.69 g, 6.44 mmol, 3 eq) in tetrahydrofuran (20 mL) at 0 °C under nitrogen atmosphere were added diisopropyl azodicarboxylate (1.30 g, 6.44 mmol, 3 eq) and 2-({2-[(2-nitrophenyl)amino]ethyl}(2,2,2-trifluoroethyl)amino)ethanol (0.66 g, 2.15 mmol, 1 eq). The mixture was stirred at 0 °C under nitrogen atmosphere for 2 h. The reaction was quenched with water (150 mL), and the mixture was extracted with dichloromethane (3×150 mL). The combined organic layers were washed with brine (3×150 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on Prep-TLC using petroleum ether/ethyl acetate (1/3) to give 1.15 g of 1-methyl-5-{1-methyl-5-[2-({2-[(2-nitrophenyl)amino]ethyl}(2,2,2-trifluoroethyl)amino)ethoxy]pyrazol-4-yl}-6-oxopyridine-3-carboxylic acid methyl ester (INT-A1-3) (96%) as an orange oil. LCMS: m/z (ESI), [M+H] +=553.25. 1H NMR (DMSO-d 6 , 400 MHz) δ 3.02 (2H, t), 3.12-3.19 (2H, m), 3.38-3.44 (2H, m), 3.50 (1H, s), 3.56 (3H, s), 3.67 (3H, s), 3.76 (3H, s), 4.03 (2H, t), 6.63-6.72 (1H, m), 6.97-7.05 (1H, m), 7.50-7.58 (1H, m), 7.62-7.68 (1H, m), 7.92 (1H, s), 8.01-8.06 (2H, m), 8.20 (1H, t), 8.37 (1H, d) Step 4. Preparation of methyl 5-{5-[2-({2-[(2-aminophenyl)amino]ethyl}(2,2,2-trifluoroethyl)amino)ethoxy]-1-methylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylate (INT-A1-4). To a stirred mixture of methyl 1-methyl-5-{1-methyl-5-[2-({2-[(2-nitrophenyl)amino]ethyl}(2,2,2-trifluoroethyl)amino)ethoxy]pyrazol-4-yl}-6-oxopyridine-3-carboxylate (1.1 g, 1.99 mmol, 1 eq) and ranyl nickel (0.85 g, 9.95 mmol, 5 eq) in methanol (30 mL) was added hydrazine hydrate (0.15 g, 2.98 mmol, 1.5 eq) at 0 °C. The mixture was stirred at 0°C for 30 minutes under nitrogen atmosphere. The resulting mixture was filtered. The filter cake was washed with methanol (3×20 mL), and the solution was concentrated under reduced pressure to give 1 g of 5-{5-[2-({2-[(2-aminophenyl)amino]ethyl}(2,2,2-trifluoroethyl)amino)ethoxy]-1-methylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A1-4) (96%) as a brown solid. This material was used in the next reaction without further purification. LCMS: m/z (ESI), [M+H] +=523.10. 1H NMR (DMSO-d 6 , 400 MHz) δ 2.95 (2H, t), 3.12 (2H, t), 3.17 (2H, d), 3.48 (2H, q), 3.57 (3H, s), 3.68 (3H, s), 3.79 (3H, s), 4.03 (2H, t), 4.36 (2H, s), 6.37-6.46 (2H, m), 7.51-7.59 (2H, m), 7.64-7.67 (1H, m), 7.94 (1H, s), 8.10 (1H, d), 8.45 (1H, d). Step 5. Preparation of 5-[5-(2-{[2-(2-amino-1,3-benzodiazol-1-yl)ethyl](2,2,2-trifluoroethyl)amino}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A1-5). 5-{5-[2-({2-[(2-aminophenyl)amino]ethyl}(2,2,2-trifluoroethyl)amino)ethoxy]-1-methylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (1 g, about 1.9 mmol, about 1 equivalent) and BrCN (0.24 g, 2.29 mmol, 1.2 equivalents) were mixed in CH2CI 2The mixture in 20 mL was stirred at room temperature under nitrogen atmosphere for 2 h. The mixture was concentrated under reduced pressure. The residue was separated by Prep-TLC using CH2Cl 2/methanol (40/1) for purification to obtain 1 g of 5-[5-(2-{[2-(2-amino-1,3-benzodiazol-1-yl)ethyl] (2,2,2-trifluoroethyl)amino}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A01-5) (95%) as a brown solid. LCMS: m/z (ESI), [M+H] +=548.25. 1H NMR (DMSO-d 6 , 400 MHz) δ 3.04 (2H, t), 3.16 (2H, t), 3.50 (2H, q), 3.58 (3H, s), 3.65 (3H, s), 3.77 (3H, s), 3.98 (2H, t), 4.18 (2H, t), 7.07-7.21 (2H, m), 7.32 (1H, d), 7.40 (1H, d), 7.93 (1H, s), 8.04 (1H, d), 8.11 (2H, s), 8.46 (1H, d). Step 6. Preparation of 5-[5-(2-{[2-(2-amino-1,3-benzodiazol-1-yl)ethyl](2,2,2-trifluoroethyl)amino}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A1-6). 5-[5-(2-{[2-(2-amino-1,3-benzodiazol-1-yl)ethyl](2,2,2-trifluoroethyl)amino}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (200 mg, 0.36 mmol, 1 equivalent) and LiOH.H 2O (23 mg, 0.55 mmol, 1.5 equiv.) in tetrahydrofuran (4 mL) and H 2The mixture in 400 mL was stirred at room temperature overnight under nitrogen atmosphere. The reaction mixture was concentrated under reduced pressure. The residue was purified by C18FLASH using water and acetonitrile as mobile phase. The fractions containing the desired compound were evaporated to dryness to give 180 mg of 5-[5-(2-{[2-(2-amino-1,3-benzodiazol-1-yl)ethyl](2,2,2-trifluoroethyl)amino}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A1-6) (92%) as a white solid. LCMS: m/z (ESI), [M+H] +=534.10. 1H NMR (DMSO-d 6 , 400 MHz) δ 2.99 (2H, t), 3.28 (2H, t), 3.52 (6H, s), 3.67 (3H, s), 3.99 (2H, t), 4.18 (2H, t), 6.80-7.01 (4H, m), 7.05-7.14 (2H, m), 8.02 (1H, s), 8.09 (1H, d), 8.37 (1H, d) Step 7. 5,26-Dimethyl-10-(2,2,2-trifluoroethyl)-7-oxa-4,5, 10,13,20,22,26-heptaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}. Preparation of 0^{14,19}]twenty-eight-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (Example A1). 5-[5-(2-{[2-(2-amino-1,3-benzodiazol-1-yl)ethyl](2,2,2-trifluoroethyl)amino}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid (150 mg, 0.28 mmol, 1 equivalent), N,N,N,N-tetramethyl-O-(7-azabenzotriazole-1-yl)urea hexafluorophosphate (160 mg, 0.42 mmol, 1.5 equivalents) and N,N- A mixture of diisopropylethylamine (109 mg, 0.84 mmol, 3 equiv.) in 1,4-dioxane (5 mL) was stirred at room temperature overnight under nitrogen atmosphere. The reaction was quenched with water (50 mL), and the mixture was extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×150 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was subjected to prep-HPLC using an XBridge Shield RP18 OBD column with 10 mmol/L NH4HCO 3and 0.1% NH 3.H 2O water and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 29.5 mg of 5,26-dimethyl-10-(2,2,2-trifluoroethyl)-7-oxa-4,5,10,13,20,22, 26-heptaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octacosa-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (Example A1) as a pink solid (20%). LCMS: m/z (ESI), [M+H] +=516.20. 1H NMR (DMSO, 400 MHz) δ 3.10 (2H, t), 3.27 (2H, t), 3.63 (3H, s), 3.59-3.69 (2H, m), 3.71 (3H, s), 4.19 (2H, t), 4.33 (2H, t), 7.18-7.31 (2H, m), 7.49-7.56 (1H, m), 7.70 (1H, d), 8.25 (1H, s), 8.29 (1H, d), 8.89 (1H, d), 12.55 (1H, s). 19F NMR (DMSO-d 6 , 376 MHz) δ 69.23 (s). Example A2 5-cyclopropyl-26-methyl-16-(4-methylpiperazine-1-yl)-10-(2,2,2-trifluoroethyl)-7-oxa-4,5,10,13,20,22,26-heptaazapentacyclic [22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]twenty-eight-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione Step 1. Preparation of 2-({2-[(5-bromo-2-nitrophenyl)amino]ethyl}amino)ethanol (INT-A2-1). 4-bromo-2-fluoro-1-nitrobenzene (2 g, 9.09 mmol, 1 equivalent), aminoethylethanolamine (1.42 g, 13.63 mmol, 1.5 equivalents) and K 2CO 3A mixture of (3 g, 27.27 mmol, 3 equiv.) in acetonitrile (20 mL) was stirred at 60 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (100 mL), and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (10/1) to give 2.1 g of 2-({2-[(5-bromo-2-nitrophenyl)amino]ethyl}amino)ethanol (INT-A2-1) (72%) as a yellow solid. LCMS: m/z (ESI), [M+H] += 306.00. 1H NMR (DMSO-d 6 , 400 MHz) δ 2.64 (2H, t), 2.83 (2H, t), 3.18 (1H, d), 3.37-3.44 (2H, m), 3.48 (2H, q), 4.53 (1H, t), 6.83 (1H, d), 7.28 (1H, d), 7.99 (1H, d), 8.42 (1H, t). Step 2. Preparation of 2-({2-[(5-bromo-2-nitrophenyl)amino]ethyl}(2,2,2-trifluoroethyl)amino)ethanol (INT-A2-2). 2-({2-[(5-bromo-2-nitrophenyl)amino]ethyl}amino)ethanol (INT-A2-1, 10 g, 32.87 mmol, 1 equivalent) and N,N-To a stirred mixture of diisopropylethylamine (12 g, 98.63 mmol, 3 equiv.) in dimethylformamide (100 mL) was added 2,2,2-trifluoroethyl trifluoromethanesulfonate (11 g, 49.31 mmol, 1.5 equiv.). The mixture was stirred at 60 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (100 mL), and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by prep-TLC using petroleum ether/ethyl acetate (5/1) to give 3 g of 2-({2-[(5-bromo-2-nitrophenyl)amino]ethyl}(2,2,2-trifluoroethyl)amino)ethanol (INT-A2-2) (21%) as a yellow oil. LCMS: m/z (ESI), [M+H] +=385.90 1H NMR (DMSO-d 6, 400 MHz) δ 2.76 (2H, t), 2.95 (2H, t), 3.40 (4H, d), 3.57-3.45 (2H, m), 4.55 (1H, t), 6.84 (1H, d), 7.27 (1H, d), 8.00 (1H, d), 8.30 (1H, t). Step 3. Preparation of 2-cyclopropylpyrazol-3-ol (INT-A2-3). A mixture of methyl (2E)-3-methoxyprop-2-enoate (1 g, 8.61 mmol, 1 eq.) and cyclopropylhydrazine (621 mg, 8.61 mmol, 1 eq.) in methanol (4 mL) was stirred at 80 °C under air atmosphere for 16 h. After cooling to room temperature, the reaction was heated with saturated NH4Cl solution (300 mL) was quenched, and the mixture was extracted with dichloromethane (3×200 mL). The combined organic layers were washed with brine (3×300 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by reverse phase flash chromatography using 0% to 15% acetonitrile/water to give 200 mg of 2-cyclopropylpyrazol-3-ol (INT-A2-3) (18%) as a yellow oil. LCMS: m/z (ESI), [M+H] +=125.15. 1H NMR (DMSO-d 6, 400 MHz) δ 1.08-0.93 (4H, m), 3.44-3.31 (1H, m), 5.64 (1H, d), 7.62 (1H, d). Step 4. Preparation of 2-cyclopropyl-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-3-one (INT-A2-4). 2-Cyclopropylpyrazol-3-ol (1.1 g, 8.86 mmol, 1 eq.) and 2-(trimethylsilyl)ethoxymethyl chloride (2.66 g, 15.95 mmol, 1.8 eq.) were added to 2-cyclopropylpyrazol-3-ol (1.1 g, 8.86 mmol, 1 eq.) at room temperature under a nitrogen atmosphere.N,N-Sodium hydride (0.85 g, 17.72 mmol, 2 eq., 60%) was added portionwise to the stirred mixture in dimethylformamide (10 mL). The mixture was stirred for 2 h. The reaction was heated with saturated NH4Cl solution (300 mL) was quenched, and the mixture was extracted with dichloromethane (3×200 mL). The combined organic layers were washed with brine (3×300 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column eluting with dichloromethane/methanol (10:1). The product was further purified by reverse phase flash chromatography eluting with 0% to 100% acetonitrile/water to give 330 mg of 2-cyclopropyl-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-3-one (INT-A2-4) (15%) as a brown solid. LCMS: m/z (ESI), [M+H] +=255.20 Step 5. Preparation of 2-cyclopropyl-4-iodo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-3-one (INT-A2-5). A mixture of 2-cyclopropyl-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-3-one (4.5 g, 17.69 mmol, 1 eq.) and NIS (5.97 g, 26.53 mmol, 1.5 eq.) in acetonitrile (50 mL) was stirred at 0 °C for 2 h under nitrogen atmosphere. The reaction was quenched by adding saturated sodium thiosulfate (aqueous solution) (100 mL), and the mixture was washed with CH 2Cl 2(3×50 mL) extraction. The combined organic layers were washed with brine (3×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure to obtain 5.5 g of 2-cyclopropyl-4-iodo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-3-one (INT-A2-5) (82%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=380.95 Step 6. Preparation of 5-(2-cyclopropyl-3-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A2-6). Freshly prepared 5-(methoxycarbonyl)-1-methyl-2-oxopyridin-3-ylboronic acid (M2-2, 800 mg, 3.8 mmol, 1 equivalent), K 2CO 3(1.31 g, 9.48 mmol, 2.5 equivalents), Pd(dppf)Cl 2(554 mg, 0.76 mmol, 0.2 equiv) and 2-cyclopropyl-4-iodo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-3-one (1.73 g, 4.55 mmol, 1.2 equiv) in 1,4-dioxane (8 mL) and H 2The mixture in 2% O (2 mL) was stirred at 80 °C for 2 h under nitrogen atmosphere. After cooling to room temperature, the reaction was quenched with water (300 mL), and the mixture was extracted with dichloromethane (3×200 mL). The combined organic layers were washed with brine (3×300 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column using petroleum ether/ethyl acetate (1/1) to give 1 g of methyl 5-(2-cyclopropyl-3-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (INT-A22-4) (92%) as a brown solid. LCMS: m/z (ESI), [M+H] +=420.05. Step 7. Preparation of 5-(1-cyclopropyl-5-hydroxypyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A2-7). A solution of 5-(2-cyclopropyl-3-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (750 mg, 1.79 mmol, 1 equivalent) in 1,4-dioxane (10 mL) containing HCl was stirred at 60 °C for 5 h under nitrogen atmosphere. The resulting mixture was cooled to room temperature and concentrated under reduced pressure. The residue was dissolved in methanol (10 mL) and treated with K 2CO 3Neutralize to pH 7. Filter the mixture. Wash the filter cake with methanol (2×5 mL). Concentrate the solution under reduced pressure to give 500 mg of methyl 5-(1-cyclopropyl-5-hydroxypyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (INT-A2-7) (96%) as a brown solid. This material was used in the next step without further purification. LCMS: m/z (ESI), [M+H] +=290.05 Step 8. Preparation of 5-{5-[2-({2-[(5-bromo-2-nitrophenyl)amino]ethyl}(2,2,2-trifluoroethyl)amino)ethoxy]-1-cyclopropylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A2-8). To a mixture of methyl 5-(1-cyclopropyl-5-hydroxypyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (0.7 g, 2.42 mmol, 1 eq.) and triphenylphosphine (1.9 g, 7.26 mmol, 3 eq.) in tetrahydrofuran (20 mL) was added diisopropyl azodicarboxylate (1.4 g, 7.26 mmol, 3 eq.) and 2-({2-[(5-bromo-2-nitrophenyl)amino]ethyl}(2,2,2-trifluoroethyl)amino)ethanol (INT-A2-2, 0.9 g, 2.42 mmol, 1 eq.) at 0 °C under nitrogen atmosphere. The mixture was stirred at 0 °C for 2 h. The reaction was quenched with water (100 mL), and the mixture was extracted with ethyl acetate (3 x 100 mL). The combined organic layers were washed with brine (3×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. Pass the residue through a C18 column using 10 mmol/L NH4HCO 3and 0.1% NH 3 .H 2O water and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 400 mg of 5-{5-[2-({2-[(5-bromo-2-nitrophenyl)amino]ethyl}(2,2,2-trifluoroethyl)amino)ethoxy]-1-cyclopropylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A2-8) (25%) as an orange solid. LCMS: m/z (ESI), [M+H] +=658.95. 1H NMR (DMSO-d 6, 400 MHz) δ 0.92-1.03 (2H, m), 0.99-1.10 (2H, m), 2.51 (3H, d), 3.04 (2H, t), 3.17 (2H, t), 3.44 (2H, q), 3.55-3.60 (3H, m), 3.77 (3H, s), 4.12-4.15 (2H, m), 6.80-6.83(1H, m), 7.23 (1H, d), 7.88 (1H, s), 7.94 (1H, d), 8.06 (1H, d), 8.25 (1H, t), 8.38 (1H, d). Step 9. Preparation of methyl 5-{5-[2-({2-[(2-amino-5-bromophenyl)amino]ethyl}(2,2,2-trifluoroethyl)amino)ethoxy]-1-cyclopropylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylate (INT-A2-9). To a mixture of methyl 5-{5-[2-({2-[(5-bromo-2-nitrophenyl)amino]ethyl}(2,2,2-trifluoroethyl)amino)ethoxy]-1-cyclopropylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylate (350 mg, 0.53 mmol, 1 eq) and ranyl nickel (365 mg, 4.26 mmol, 8 eq) in methanol (5 mL) was added hydrazine hydrate (98%) (40 mg, 0.80 mmol, 1.5 eq) under nitrogen atmosphere at 0 °C. The resulting mixture was stirred for 2 h and filtered. The filter cake was washed with methanol (3×50 mL), and the solution was concentrated under reduced pressure to give 300 mg of 5-{5-[2-({2-[(2-amino-5-bromophenyl)amino]ethyl}(2,2,2-trifluoroethyl)amino)ethoxy]-1-cyclopropylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A2-9) (about 90%) as a brown solid. LCMS: m/z (ESI), [M+H] +=627.15. 1H NMR (DMSO-d 6, 400 MHz) δ 0.91-1.03 (2H, m), 0.99-1.10 (2H, m), 2.98 (2H, t), 3.13-3.19 (4H, m), 3.51 (2H, q), 3.57 (3H, s), 3.79 (3H, s), 4.01-4.07 (3H, m), 4.12 (2H, t), 5.76 (1H, s), 6.38-6.58 (3H, m), 7.91 (1H, d), 8.13 (1H, d), 8.45 (1H, d). Step 10. Preparation of methyl 5-[5-(2-{[2-(2-amino-6-bromo-1,3-benzodiazol-1-yl)ethyl](2,2,2-trifluoroethyl)amino}ethoxy)-1-cyclopropylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylate (INT-A2-10). A mixture of methyl 5-{5-[2-({2-[(2-amino-5-bromophenyl)amino]ethyl}(2,2,2-trifluoroethyl)amino)ethoxy]-1-cyclopropylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylate (300 mg, 0.48 mmol, 1 eq) and BrCN (60 mg, 0.57 mmol, 1.2 eq) in dichloromethane (5 mL) was stirred under nitrogen atmosphere at room temperature for 2 h and concentrated under vacuum. The residue was purified on prep-TLC using dichloromethane/methanol (10/1) to give 300 mg of 5-[5-(2-{[2-(2-amino-6-bromo-1,3-benzodiazol-1-yl)ethyl](2,2,2-trifluoroethyl)amino}ethoxy)-1-cyclopropylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A2-10) (96%) as a brown solid. LCMS: m/z (ESI), [M+H] +=654.10. Step 11. Preparation of 5-[5-(2-{[2-(2-amino-6-bromo-1,3-benzodiazol-1-yl)ethyl](2,2,2-trifluoroethyl)amino}ethoxy)-1-cyclopropylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A2-11). 5-[5-(2-{[2-(2-amino-6-bromo-1,3-benzodiazol-1-yl)ethyl](2,2,2-trifluoroethyl)amino}ethoxy)-1-cyclopropylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (280 mg, 0.43 mmol, 1 equivalent) and LiOH .H 2O (27 mg, 0.64 mmol, 1.5 eq.) in THF (4 mL) and H 2The mixture in 4% O (1 mL) was stirred at room temperature for 2 h under nitrogen atmosphere and concentrated under reduced pressure. The residue was purified by prep-HPLC using an XBridge Shield RP18 OBD column with 10 mmol/L NH4HCO 3and 0.1% NH 3 .H 2O water and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 135 mg of 5-[5-(2-{[2-(2-amino-6-bromo-1,3-benzodiazol-1-yl)ethyl](2,2,2-trifluoroethyl)amino}ethoxy)-1-cyclopropylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A2-11) (49%) as a brown solid. LCMS: m/z (ESI), [M+H] +=638.10. 1H NMR (DMSO-d 6, 400 MHz) δ 0.90-1.03 (2H, m), 0.99-1.09 (2H, m), 3.01 (2H, t), 3.51 (3H, s), 3.53-3.63 (3H, m), 4.08 (2H, t), 4.23 (2H, t), 6.80-7.13 (3H, m), 7.19 (2H, s), 7.31 (1H, d), 7.97-8.05 (2H, m), 8.43 (1H, d) Step 12. Preparation of 16-bromo-5-cyclopropyl-26-methyl-10-(2,2,2-trifluoroethyl)-7-oxa-4,5,10,13,20,22,26-heptaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octacos-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (INT-A2-12). 5-[5-(2-{[2-(2-amino-6-bromo-1,3-benzodiazol-1-yl)ethyl](2,2,2-trifluoroethyl)amino}ethoxy)-1-cyclopropylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid (110 mg, 0.17 mmol, 1 equivalent), N,N-Diisopropylethylamine (67 mg, 0.52 mmol, 3 equivalents) and N,N,N,N-Tetramethyl- OA mixture of -(7-azabenzotriazol-1-yl)urea hexafluorophosphate (98 mg, 0.26 mmol, 1.5 equiv) in 1,4-dioxane (5 mL) was stirred at room temperature under nitrogen atmosphere for 2 h. The reaction was quenched with water (50 mL), and the mixture was extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was subjected to prep-HPLC using an XBridge Shield RP18 OBD column with 10 mmol/L NH4HCO 3and 0.1% NH 3 .H 2O water and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 60 mg of 16-bromo-5-cyclopropyl-26-methyl-10-(2,2,2-trifluoroethyl)-7-oxa-4,5,10,13,20,22,26-heptaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octacosa-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (INT-A2-12) (56%) as a brown solid. LCMS: m/z (ESI), [M+H] +=622.05. 1H NMR (DMSO-d 6, 400 MHz) δ 0.96-1.04 (2H, m), 1.08-1.35 (2H, m), 3.08-3.12 (2H, m), 3.28 (2H, q), 3.49-3.60 (1H, m), 3.62 (3H, s), 3.64-3.77 (2H, m), 4.21-4.33 (2H, m), 4.29 (2H, s), 7.36-7.38 (1H, m), 7.43 (1H, d), 8.09 (1H, d), 8.21 (1H, s), 8.30 (1H, d), 8.88 (1H, d), 12.63 (1H, s). Step 13. Preparation of 5-cyclopropyl-26-methyl-16-(4-methylpiperazin-1-yl)-10-(2,2,2-trifluoroethyl)-7-oxa-4,5,10,13,20,22,26-heptaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octacosa-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (Example A2). 16-Bromo-5-cyclopropyl-26-methyl-10-(2,2,2-trifluoroethyl)-7-oxa-4,5,10,13,20,22,26-heptaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octacos-1(28),2(6),3, 14,16,18, 20,24-octaene-23,27-dione (40 mg, 0.06 mmol, 1 eq.), 1-methylpiperazine (26 mg, 0.256 mmol, 4 eq.) and BrettPhos Pd G 3To a mixture of 1,4-dioxane (5 mL) and 1,4-dioxane (18 mg, 0.02 mmol, 0.3 equiv.) was added LiHMDS (65 mg, 0.38 mmol, 6 equiv.). The mixture was stirred at 60 °C for 2 h and then cooled to room temperature. The reaction mixture was treated with NH4Cl(aqueous) solution (50 mL) and extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×30 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was passed through prep-HPLC using a Shield RP18 OBD column with 10 mmol/L NH4HCO 3and 0.1% NH 3 .H 2O water and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 2.7 mg of 5-cyclopropyl-26-methyl-16-(4-methylpiperazin-1-yl)-10-(2,2,2-trifluoroethyl)-7-oxa-4,5,10,13,20,22,26-heptaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octacosa-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (Example A2) as a white solid (6%). LCMS: m/z (ESI), [M+H] +=640.35. 1H NMR (DMSO-d 6, 400 MHz) δ 0.96-1.04 (2H, m), 1.08-1.35 (2H, m), 2.25 (3H, s), 2.48-2.50 (4H, m), 3.04-3.15 (3H, m), 3.18 (4H, t), 3.57-3.64 (4H, m), 3.62 (3H, s), 4.24-4.29 (4H, m), 6.87 (1H, d), 7.23 (1H, s), 7.35 (1H, d), 8.21 (1H, s), 8.27 (1H, s), 8.89 (1H, s), 12.33 (1H, s). 19F NMR (DMSO-d 6, 376 MHz) δ 69.26 (s). Example A3 5,26-Dimethyl-16-(4-methylpiperazine-1-yl)-10-(2,2,2-trifluoroethyl)-7-oxo-4,5,10,13,20,22,26-heptaazapentacyclic [22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]twenty-eight-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione Step 1. Preparation of methyl-5-{5-[2-({2-[(5-bromo-2-nitrophenyl)amino]ethyl}(2,2,2-trifluoroethyl)amino)ethoxy]-1-methylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylate (INT-A3-1). To a stirred mixture of triphenylphosphine (1324 mg, 5.04 mmol, 3 eq.) and methyl 5-(5-hydroxy-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (M2, 354 mg, 1.34 mmol, 0.8 eq.) in tetrahydrofuran (9 mL) at 0°C were added 2-({2-[(5-bromo-2-nitrophenyl)amino]ethyl}(2,2,2-trifluoroethyl)amino)ethanol (650 mg, 1.68 mmol, 1 eq.) and diisopropyl azodicarboxylate (1021 mg, 5.04 mmol, 3 eq.). The mixture was stirred at 0°C under nitrogen atmosphere for 1 h. The reaction was quenched with water (50 mL), and the mixture was extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×50 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (10/1) to give 550 mg of 5-{5-[2-({2-[(5-bromo-2-nitrophenyl)amino]ethyl}(2,2,2-trifluoroethyl)amino)ethoxy]-1-methylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A3-1) (36%) as a yellow oil. LCMS: m/z (ESI), [M+H] +=630.85. 1H NMR (DMSO-d 6, 400 MHz) δ 3.01 (2H, t), 3.11 (2H, t), 3.31-3.45 (2H, m)3.49-3.54 (4H, m), 3.66 (3H, s), 3.76 (3H, s), 4.01 (2H, t), 5.73 (1H, s), 6.78-6.92 (1H, m), 7.20 (1H, d), 7.96-7.88 (2H, m), 8.00 (1H, d), 8.23 (1H, t), 8.33 (1H, d). Step 2. Preparation of 5-{5-[2-({2-[(2-amino-5-bromophenyl)amino]ethyl}(2,2,2-trifluoroethyl)amino)ethoxy]-1-methylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A3-2). To a stirred mixture of 5-{5-[2-({2-[(5-bromo-2-nitrophenyl)amino]ethyl}(2,2,2-trifluoroethyl)amino)ethoxy]-1-methylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (570 mg, 0.90 mmol, 1 eq.) and ranyl nickel (30 mg) in methanol (6 mL) was added NH 2NH 2 .H 2O (57 mg, 1.80 mmol, 2 eq.). The mixture was stirred at 0 °C for 30 min under nitrogen atmosphere. The mixture was filtered. The filter cake was washed with methanol (3×30 mL), and the solution was concentrated under reduced pressure to give 270 mg of 5-{5-[2-({2-[(2-amino-5-bromophenyl)amino]ethyl}(2,2,2-trifluoroethyl)amino)ethoxy]-1-methylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A3-2) (42%) as a yellow oil. LCMS: m/z (ESI), [M+H] +=601.05. Step 3. Preparation of 5-[5-(2-{[2-(2-amino-6-bromo-1,3-benzodiazol-1-yl)ethyl](2,2,2-trifluoroethyl)amino}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A3-3). A mixture of 5-{5-[2-({2-[(2-amino-5-bromophenyl)amino]ethyl}(2,2,2-trifluoroethyl)amino)ethoxy]-1-methylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (240 mg, 0.39 mmol, 1 eq.) and BrCN (42 mg, 0.39 mmol, 1 eq.) in ethanol (6 mL) was stirred at 0°C for 1 h under nitrogen atmosphere. The mixture was concentrated under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (10/1) to give 120 mg of 5-[5-(2-{[2-(2-amino-6-bromo-1,3-benzodiazol-1-yl)ethyl](2,2,2-trifluoroethyl)amino}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A3-3) (43%) as a brown solid. LCMS: m/z (ESI), [M+H] +=626.05 Step 4. Preparation of 5-[5-(2-{[2-(2-amino-6-bromo-1,3-benzodiazol-1-yl)ethyl](2,2,2-trifluoroethyl)amino}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A3-4). To a stirred mixture of 5-[5-(2-{[2-(2-amino-6-bromo-1,3-benzodiazol-1-yl)ethyl](2,2,2-trifluoroethyl)amino}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (100 mg, 0.16 mmol, 1 equivalent) in tetrahydrofuran (2 mL) was added LiOH .H 2O (7.6 mg, 0.32 mmol, 2 eq.) of H 2O (0.5 mL). The resulting mixture was stirred at room temperature for 2 h under a nitrogen atmosphere. The mixture was concentrated under reduced pressure. The residue was purified by C18 column using water and acetonitrile as mobile phase. The fractions containing the desired compound were evaporated to dryness to give 70 mg of 5-[5-(2-{[2-(2-amino-6-bromo-1,3-benzodiazol-1-yl)ethyl](2,2,2-trifluoroethyl)amino}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A3-4) (68%) as a white solid. LCMS: m/z (ESI), [M+H] +=612.00. 1H NMR (DMSO-d 6, 400 MHz) δ 2.98 (2H, t), 3.51 (3H, s), 3.57 (5H, t), 3.68 (3H, s), 3.99 (2H, t), 4.25 (2H, t), 7.06-6.97 (2H, m), 7.28 (2H, d), 7.33 (3H, s ), 7.98 (1H, d), 8.09 (1H, s), 8.47 (1H, d,). Step 5. Preparation of 16-bromo-5,26-dimethyl-10-(2,2,2-trifluoroethyl)-7-oxa-4,5,10,13,20,22,26-heptaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octacosa-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (INT-A3-5). 5-[5-(2-{[2-(2-amino-6-bromo-1,3-benzodiazol-1-yl)ethyl](2,2,2-trifluoroethyl)amino}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid (65 mg, 0.10 mmol, 1 equivalent) andN,N,N,N-Tetramethyl- O-(7-Azabenzotriazol-1-yl)urea hexafluorophosphate (60 mg, 0.15 mmol, 1.5 equivalents) was added to the stirred mixture in 1,4-dioxane (2 mL) N,N-diisopropylethylamine (41 mg, 0.31 mmol, 3 eq.) was added, and the mixture was stirred at 60 °C for 2 h under a nitrogen atmosphere. After cooling to room temperature, the mixture was concentrated under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (10/1) to give 45 mg of 16-bromo-5,26-dimethyl-10-(2,2,2-trifluoroethyl)-7-oxa-4,5,10,13,20,22,26-heptaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octacosa-1(28),2(6), 3,14,16,18,20,24-octaene-23,27-dione (INT-A3-5) (58%) as a brown solid. LCMS: m/z (ESI), [M+H] +=594.00. Step 6. Preparation of 5,26-dimethyl-16-(4-methylpiperazin-1-yl)-10-(2,2,2-trifluoroethyl)-7-oxa-4,5,10,13,20,22,26-heptaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octacosa-1(28),2(6),3, 14,16,18,20,24-octaene-23,27-dione (Example A3). 16-Bromo-5,26-dimethyl-10-(2,2,2-trifluoroethyl)-7-oxa-4,5,10,13,20,22,26-heptaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octadecano-1(28),2(6),3, 14,16,18,20,24-octaene-23,27-dione (50 mg, 0.08 mmol, 1 equivalent), BrettPhos Pd G 3To a stirred mixture of 1,4-dioxane (2 mL) and piperazine, 1-methyl- (22 mg, 0.02 mmol, 0.3 equiv.) was added LiHMDS (0.48 mL, 0.48 mmol, 6 equiv.) dropwise. The mixture was stirred at 60 °C for 40 min under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (30 mL), and the mixture was extracted with ethyl acetate (3×20 mL). The combined organic layers were washed with brine (3×30 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was subjected to Prep-HPLC using an XBridge Shield RP18 OBD column with 10 mmol/L NH4HCO 3and 0.1% NH 3 .H 2O water and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 3.1 mg of 5,26-dimethyl-16-(4-methylpiperazin-1-yl)-10-(2,2,2-trifluoroethyl)-7-oxa-4,5,10,13,20,22,26-heptaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octacosa-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (Example A3) as a yellow solid (5%). LCMS: m/z (ESI), [M+H] +=614.25. 1H NMR (DMSO-d 6, 400 MHz) δ 2.26 (3H, s), 3.04-3.10 (2H, m), 3.18 (4H, t), 3.15-3.27 (5H, m), 3.58-3.64 (4H, m), 3.64-3.78 (5H, m), 4.17 (2H, t), 4.29 (2H, t), 6.85-6.89 (1H, m), 7.23 (1H, d), 7.35 (1H, d), 8.26 (2H, d), 8.88 (1H, d), 12.34 (1H, s). 19F NMR (DMSO-d 6, 376 MHz) δ -69.31 (s). Example A4 10-(2,2-difluoroethyl)-16-{[2-(dimethylamino)ethyl](methyl)amino}-5,26-dimethyl-7-oxa-4,5,10,13,20,22,26-heptaazapentacyclic[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]twenty-eight-1(28),2(6),3, 14,16,18,20,24-octaene-23,27-dione Step 1. Preparation of 2-({2-[(5-bromo-2-nitrophenyl)amino]ethyl}(2,2,-difluoroethyl)amino)ethanol (INT-A4-1). 2-({2-[(5-bromo-2-nitrophenyl)amino]ethyl}amino)ethanol (2 g, 6.57 mmol, 1 equivalent), N,N- A mixture of diisopropylethylamine (2.55 g, 19.72 mmol, 3 equiv.) and 2,2-difluoroethyl trifluoromethanesulfonate (1.69 g, 7.89 mmol, 1.2 equiv.) in dimethylformamide (20 mL) was stirred at 60 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (80 mL), and the mixture was extracted with ethyl acetate (3×60 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (10/1) to give 1.1 g of 2-({2-[(5-bromo-2-nitrophenyl)amino]ethyl}(2,2-difluoroethyl)amino)ethanol (INT-A4-1) (43%) as a yellow oil. LCMS: m/z (ESI), [M+H] +=366.00. 1H NMR (DMSO-d 6 , 400 MHz) δ 2.71 (2H, t), 2.90 (2H, t), 2.99 (2H, t), 3.35-3.43 (2H, m), 3.46-3.64 (2H, m), 4.51 (1H, t), 5.87-6.18 (1H, m), 6.83-6.86 (1H, m), 7.27 (1H, d), 7.99 (1H, d), 8.33 (1H, t). Step 2. Preparation of 5-{5-[2-({2-[(5-bromo-2-nitrophenyl)amino]ethyl}(2,2-difluoroethyl)amino)ethoxy]-1-methylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A4-2). To a mixture of triphenylphosphine (1.38 g, 5.29 mmol, 3 eq.) and methyl 5-(5-hydroxy-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (M2, 836 mg, 3.17 mmol, 1.8 eq.) in tetrahydrofuran (10 mL) at 0°C were added 2-({2-[(5-bromo-2-nitrophenyl)amino]ethyl}(2,2-difluoroethyl)amino)ethanol (650 mg, 1.76 mmol, 1 eq.) and diisopropyl azodicarboxylate (1.07 g, 5.29 mmol, 3 eq.). The mixture was stirred at 0°C under nitrogen atmosphere for 1 h. The reaction was quenched with water (80 mL), and the mixture was extracted with ethyl acetate (3×60 mL). The combined organic layers were washed with brine (3×50 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using petroleum ether/ethyl acetate (1/3) to give 400 mg of 5-{5-[2-({2-[(5-bromo-2-nitrophenyl)amino]ethyl}(2,2-difluoroethyl)amino)ethoxy]-1-methylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A4-2) (33%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=613.00. Step 3. Preparation of 5-{5-[2-({2-[(2-amino-5-bromophenyl)amino]ethyl}(2,2-difluoroethyl)amino)ethoxy]-1-methylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A4-3). To a stirred mixture of 5-{5-[2-({2-[(5-bromo-2-nitrophenyl)amino]ethyl}(2,2-difluoroethyl)amino)ethoxy]-1-methylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (400 mg, 0.65 mmol, 1 eq.) and ranyl nickel (72 mg) in methanol (5 mL) was added hydrazine (41 mg, 1.30 mmol, 2 eq.) at 0°C. The mixture was stirred at 0°C for 30 minutes under nitrogen atmosphere. The resulting mixture was filtered. The filter cake was washed with methanol (3×10 mL), and the solution was concentrated under reduced pressure to give 300 mg of methyl-5-{5-[2-({2-[(2-amino-5-bromophenyl)amino]ethyl}(2,2-difluoroethyl)amino)ethoxy]-1-methylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylate (INT-A4-3) (55%) as a brown solid. LCMS: m/z (ESI) , [M+H]+=583.05. Step 4. Preparation of methyl 5-[5-(2-{[2-(2-amino-6-bromo-1,3-benzodiazol-1-yl)ethyl](2,2-difluoroethyl)amino}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylate (INT-A4-4). A mixture of methyl 5-{5-[2-({2-[(2-amino-5-bromophenyl)amino]ethyl}(2,2-difluoroethyl)amino)ethoxy]-1-methylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylate (300 mg, 0.51 mmol, 1 eq.) and BrCN (54 mg, 0.51 mmol, 1 eq.) in ethanol (5 mL) was stirred under nitrogen atmosphere at room temperature for 1 h. The mixture was concentrated under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (10/1) to give 250 mg of 5-[5-(2-{[2-(2-amino-6-bromo-1,3-benzodiazol-1-yl)ethyl](2,2-difluoroethyl)amino}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A4-4) (71%) as a brown solid. LCMS: m/z (ESI), [M+H]+=608.05. Step 5. Preparation of 5-[5-(2-{[2-(2-amino-6-bromo-1,3-benzodiazol-1-yl)ethyl](2,2-difluoroethyl)amino}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A4-5). To a stirred mixture of 5-[5-(2-{[2-(2-amino-6-bromo-1,3-benzodiazol-1-yl)ethyl](2,2-difluoroethyl)amino}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (200 mg, 0.32 mmol, 1 equiv.) in tetrahydrofuran (4 mL) was added LiOH .H 2O (19.6 mg, 0.82 mmol, 2.5 equiv) of H 2O (1 mL). The resulting mixture was stirred at room temperature for 2 h. The mixture was concentrated under reduced pressure. The residue was passed through C18FLASH using a 10 mmol/L NH4HCO 3and 0.1% NH 3 .H 2O water and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 175 mg of 5-[5-(2-{[2-(2-amino-6-bromo-1,3-benzodiazol-1-yl)ethyl](2,2-difluoroethyl)amino}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A4-5) (80%) as a brown solid. LCMS: m/z (ESI), [M+H] +=594.10. 1H NMR (DMSO-d 6 , 400 MHz) δ 2.90 (4H, t), 3.05 (4H, t), 3.27 (2H, d), 3.51 (6H, s), 3.68 (6H, s), 3.97 (4H, t), 4.24 (4H, t), 5.85-6.20 (1H, m), 7.06-6.97 (4H, m), 7.28 (4H, s), 7.32 (2H, d), 7.97 (2H, d), 8.08 (2H, s), 8.46 (1H, s), 8.47 (1H, s). Step 6. Preparation of 16-bromo-10-(2,2-difluoroethyl)-5,26-dimethyl-7-oxa-4,5,10,13,20,22,26-heptaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octacosa-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (INT-A4-6). 5-[5-(2-{[2-(2-amino-6-bromo-1,3-benzodiazol-1-yl)ethyl](2,2-difluoroethyl)amino}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid (160 mg, 0.26 mmol, 1 equivalent), N,N,N,N-tetramethyl-O-(7-azabenzotriazol-1-yl)uronium hexafluorophosphate (153 mg, 0.40 mmol, 1.5 equivalents) andN,N- A mixture of diisopropylethylamine (104 mg, 0.80 mmol, 3 equiv.) in 1,4-dioxane (6 mL) was stirred at 60 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (30 mL), and the mixture was extracted with ethyl acetate (3×20 mL). The combined organic layers were washed with brine (3×20 mL), purified by anhydrous Na 2SO 4The residue was purified by Prep-TLC eluting with dichloromethane/methanol (10/1) to give 130 mg of 16-bromo-10-(2,2-difluoroethyl)-5,26-dimethyl-7-oxa-4,5,10,13,20,22,26-heptaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octacosa-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (INT-A4-6) (79%) as a brown solid. LCMS (EB237475-165): m/z (ESI), [M+H]+=576.00; Step 7. Preparation of 10-(2,2-difluoroethyl)-16-{[2-(dimethylamino)ethyl](methyl)amino}-5,26-dimethyl-7-oxa-4,5,10,13,20,22,26-heptaazapentacyclic[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]twenty-eight-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (Example A4). 16-Bromo-10-(2,2-difluoroethyl)-5,26-dimethyl-7-oxa-4,5,10,13,20,22,26-heptaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octadecano-1(28),2(6),3, 14,16,18,20,24-octaene-23,27-dione (100 mg, 0.17 mmol, 1 equivalent), BrettPhos Pd G 3To a mixture of 2-(47 mg, 0.05 mmol, 0.3 eq.) and [2-(dimethylamino)ethyl](methyl)amine (70 mg, 0.69 mmol, 4 eq.) in 1,4-dioxane (3 mL) was added LiHMDS (0.7 mL, 1.02 mmol, 6 eq.) dropwise. The mixture was stirred at 60 °C under nitrogen atmosphere for 2 h and then cooled to room temperature. The reaction was heated with saturated NH4Cl(aq) solution (5 mL) was quenched, and the mixture was extracted with ethyl acetate (3×200 mL). The combined organic layers were washed with brine (3×300 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was subjected to prep-HPLC using an XBridge Shield RP18 OBD column with 10 mmol/L NH4HCO 3+0.1%NH 3 .H 2O water and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 1.2 mg of 10-(2,2-difluoroethyl)-16-{[2-(dimethylamino)ethyl](methyl)amino}-5,26-dimethyl-7-oxa-4,5,10,13,20,22,26-heptaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octacosa-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (Example A4) as a yellow solid (1%). LCMS: m/z (ESI), [M+H] +=598.25. 1H NMR (DMSO-d 6, 400 MHz) δ 2.19 (6H, s), 2.39 (2H, t), 2.96 (3H, s), 3.03 (2H, t), 3.11-3.52 (6H, m), 3.61 (3H, s), 3.73 (3H, s), 4.19 (2H, t), 4.28 (2H, t), 6.21 (1H, t), 6.64 (1H, d), 6.84 (1H, s), 7.31 (1H, d), 8.25 (2H, s), 8.87 (1H, s), 12.20 (1H, s). 19F NMR (DMSO-d 6, 376 MHz) δ 119.603 (s). Example A5 5,26-Dimethyl-10-(oxacyclobutane-3-yl)-7-oxa-4,5,10,13,20,22,26-heptaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]twenty-eight-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione Step 1. Preparation of 2-({2-[(2-nitrophenyl)amino]ethyl}(oxacyclobutan-3-yl)amino)ethanol (INT-A5-1). To a stirred mixture of 2-({2-[(2-nitrophenyl)amino]ethyl}amino)ethanol (1 g, 4.44 mmol, 1 eq), 3-oxacyclobutanone (383 mg, 5.33 mmol, 1.2 eq) and molecular sieves (20 mg) in dichloromethane (30 mL) was added sodium triacetoxyborohydride (1.88 g, 8.88 mmol, 2 eq) at room temperature. The mixture was stirred at room temperature for 12 h under nitrogen atmosphere. The reaction was quenched with water (80 mL), and the mixture was extracted with ethyl acetate (3×60 mL). The combined organic layers were washed with brine (3×50 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using petroleum ether/ethyl acetate (1/1) to give 1.2 g of 2-({2-[(2-nitrophenyl)amino]ethyl}(oxadiazine-3-yl)amino)ethanol (INT-A5-1) (95%) as a red oil. LCMS: m/z (ESI), [M+H] +=282.10. 1H NMR (DMSO-d 6, 400 MHz) δ 2.56-2.65(2H, m), 2.74-2.81(2H, m), 3.31-3.40(2H, m), 3.41-3.49(2H, m), 4.00-4.06(1H, m), 4.42-4.48(2H, m), 4.51-4.58(2H, m), 6.64-6.74(1H, m), 7.01-7.08(1H, m), 7.50-7.59(1H, m), 8.03-8.11(1H, m), 8.32(1H, t). Step 2. Preparation of methyl 1-methyl-5-{1-methyl-5-[2-({2-[(2-nitrophenyl)amino]ethyl}(oxacyclobutan-3-yl)amino)ethoxy]pyrazol-4-yl}-6-oxopyridine-3-carboxylate (INT-A5-2). To a stirred mixture of methyl 5-(5-hydroxy-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (308 mg, 1.17 mmol, 1.1 eq) and triphenylphosphine (839 mg, 3.20 mmol, 3 eq) in tetrahydrofuran (2 mL) at 0°C were added diisopropyl azodicarboxylate (647 mg, 3.20 mmol, 3 eq) and 2-({2-[(2-nitrophenyl)amino]ethyl}(oxacyclobutan-3-yl)amino)ethanol (300 mg, 1.07 mmol, 1 eq). The mixture was stirred at room temperature under nitrogen atmosphere for 12 h. The reaction was quenched with water (50 mL), and the mixture was extracted with ethyl acetate (3×30 mL). The combined organic layers were washed with brine (3×30 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using petroleum ether/ethyl acetate (1/1) to give 200 mg of 1-methyl-5-{1-methyl-5-[2-({2-[(2-nitrophenyl)amino]ethyl}(oxacyclobutan-3-yl)amino)ethoxy]pyrazol-4-yl}-6-oxopyridine-3-carboxylic acid methyl ester (INT-A5-2) (30%) as a yellow solid. LCMS: m/z (ESI), [M+H] += 527.20. 1H NMR (DMSO-d 6, 400 MHz) δ 2.79-2.87(2H, m), 2.94-3.00(2H, m), 3.29-3.40(5H, m), 3.78(3H, s), 3.94-4.03(2H, m), 4.04-4.16(1H, m), 4.42-4.51(2H, m), 4.52-4.60(2H, m), 6.64-6.73(1H, m), 6.94-7.01(1H, m), 7.45-7.53(1H, m), 7.91(1H, s), 8.01-8.08(2H, m), 8.25(1H, t), 8.39(1H, d). Step 3. Preparation of 5-{5-[2-({2-[(2-aminophenyl)amino]ethyl}(oxacyclobutan-3-yl)amino)ethoxy]-1-methylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A5-3). To a stirred mixture of 1-methyl-5-{1-methyl-5-[2-({2-[(2-nitrophenyl)amino]ethyl}(oxacyclobutan-3-yl)amino)ethoxy]pyrazol-4-yl}-6-oxopyridine-3-carboxylic acid methyl ester (150 mg, 0.29 mmol, 1 eq.) and ranyl nickel (30 mg, 0.35 mmol, 1.2 eq.) in methanol (30 mL) at 0°C was added NH 2NH 2.H 2O (90 mg). The mixture was stirred at 0 °C for 1 h under nitrogen atmosphere. The resulting mixture was filtered. The filter cake was washed with methanol (2×10 mL), and the solution was concentrated under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (10/1) to give 120 mg of 5-{5-[2-({2-[(2-aminophenyl)amino]ethyl}(oxacyclobutan-3-yl)amino)ethoxy]-1-methylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A5-3) (73%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=497.30. Step 4. Preparation of 5-[5-(2-{[2-(2-amino-1,3-benzodiazol-1-yl)ethyl](oxacyclobutan-3-yl)amino}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A5-4). A mixture of 5-{5-[2-({2-[(2-aminophenyl)amino]ethyl}(oxacyclobutan-3-yl)amino)ethoxy]-1-methylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (120 mg, 0.24 mmol, 1 equiv.) and cyanogen bromide (38 mg, 0.36 mmol, 1.5 equiv.) in ethanol (10 mL) was stirred at room temperature for 2 h under nitrogen atmosphere. The mixture was concentrated under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (5/1) to give 82 mg of 5-[5-(2-{[2-(2-amino-1,3-benzodiazol-1-yl)ethyl](oxacyclobutan-3-yl)amino}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A5-4) (65%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=522.25. 1H NMR (DMSO-d 6, 400 MHz) δ 2.77-2.86(2H, m), 2.99-3.07(2H, m), 3.57(3H, s), 3.68(3H, s), 3.77(3H, s), 3.91-3.98(2H, m), 3.98-4.11(3H, m), 4.29-4.36(2H, m), 4.43-4.50(2H, m), 6.39(2H, s), 6.78-6.85(1H, m), 6.87-6.95(1H, m), 7.04-7.14(1H, m), 7.93(1H, s), 8.08(1H, d), 8.45(1H, d). Step 5. Preparation of 5-[5-(2-{[2-(2-amino-1,3-benzodiazol-1-yl)ethyl](oxacyclobutan-3-yl)amino}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A5-5). A mixture of 5-[5-(2-{[2-(2-amino-1,3-benzodiazol-1-yl)ethyl](oxacyclobutan-3-yl)amino}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (82 mg, 0.16 mmol, 1 eq.) and lithium hydroxide (13 mg, 0.31 mmol, 2 eq.) in tetrahydrofuran (10 mL) and water (2.5 mL) was stirred at room temperature for 2 h under nitrogen atmosphere. The mixture was concentrated under reduced pressure. The residue was passed through C18FLASH using a 10 mmol/L NH4HCO 3and 0.1% NH 3 .H 2O water and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 20 mg of 5-[5-(2-{[2-(2-amino-1,3-benzodiazol-1-yl)ethyl](oxacyclobutan-3-yl)amino}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A5-5) (24%) as a red solid. LCMS: m/z (ESI), [M+H] +=508.25. Step 6. Preparation of 5,26-dimethyl-10-(oxacyclobutane-3-yl)-7-oxa-4,5,10,13,20,22,26-heptaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octaconic-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (Example A5). 5-[5-(2-{[2-(2-amino-1,3-benzodiazol-1-yl)ethyl](oxacyclobutan-3-yl)amino}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid (20 mg, 0.04 mmol, 1 equivalent), N, N-Diisopropylethylamine (15 mg, 0.12 mmol, 3 equivalents) and N,N,N,N-Tetramethyl- OA mixture of -(7-azabenzotriazol-1-yl)uronium hexafluorophosphate (22 mg, 0.06 mmol, 1.5 equiv.) in 1,4-dioxane (4 mL) was stirred at room temperature for 3 h under a nitrogen atmosphere. The mixture was concentrated under reduced pressure. The residue was purified by prep-HPLC using an XBridge Prep OBD C18 column containing 10 mmol/L NH4HCO 3and 0.1% NH 3 .H 2O water and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 8.8 mg of 5,26-dimethyl-10-(oxacyclobutane-3-yl)-7-oxa-4,5,10,13,20,22,26-heptaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octacosa-1(28),2(6),3, 14,16,18,20,24-octaene-23,27-dione (Example A5) as a white solid (45%). LCMS: m/z (ESI), [M+H] +=490.20. 1H NMR (DMSO-d 6, 400 MHz) δ 2.95-3.03(2H, m), 3.17-3.24(2H, m), 3.62(3H, s), 3.74(3H, s), 4.13-4.30(5H, m), 4.59-4.64(2H, m), 4.66-4.72(2H, m), 7.19-7.29(2H, m), 7.49-7.54(1H, m), 7.58-7.64(1H, m), 8.24(1H, s), 8.29(1H, d), 8.80(1H, d), 11.69(1H, s). Example A6 10-ethyl-5,12,26-trimethyl-7-oxa-4,5,10,13,20,22,26-heptaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octadecene-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione Step 1. NPreparation of tert-butyl-{1-[ethyl(2-hydroxyethyl)amino]propan-2-yl}carbamate (INT-A6-1). Ethylethanolamine (1 g, 11.21 mmol, 1 equivalent) and NTo a stirred mixture of tert-butyl-(1-bromopropan-2-yl)carbamate (3.2 g, 13.46 mmol, 1.2 equiv.) in acetonitrile (40 mL), add K 2CO 3(4.6 g, 33.65 mmol, 3 equiv.). The mixture was stirred at 60 °C overnight and cooled to room temperature. The reaction was quenched with water (50 mL), and the mixture was washed with CH2Cl 2(3×100 mL) extraction. The combined organic layers were washed with brine (3×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC eluting with dichloromethane/methanol (10/1) to give 1 g of N-tert-Butyl{1-[ethyl(2-hydroxyethyl)amino]propan-2-yl}carbamate (INT-A6-1) (36%). 1H NMR (DMSO-d 6, 400 MHz) δ 1.04 (3H, t), 1.14 (3H, d), 1.46 (9H, s), 2.32-2.47 (2H, m), 2.52-2.70 (4H, m), 3.50-3.63 (2H, m), 3.72-3.80 (1H, m), 4.45 (1H, brs). Step 2. Preparation of 2-[(2-aminopropyl)(ethyl)amino]ethanol (INT-A6-2). Add 2-[(2-aminopropyl)(ethyl)amino]ethanol to a 20 mL vial at room temperature.N-tert-butyl {1-[ethyl(2-hydroxyethyl)amino]propan-2-yl}carbamate (1 g, 4.05 mmol, 1 equiv) and 1,4-dioxane (10 mL) containing HCl. The mixture was stirred at room temperature for 10 h and concentrated under reduced pressure. The residue was treated with saturated NaHCO3(aqueous solution) solution (30 mL) and extracted with ethyl acetate (3×30 mL). The combined organic layers were washed with brine (3×40 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure to give 0.59 g of 2-[(2-aminopropyl)(ethyl)amino]ethanol (INT-A6-2) as a yellow solid. This material was used in the next step without further purification. LCMS: m/z (ESI), [M+H] +=146.15 Step 3. Preparation of 2-[ethyl({2-[(2-nitrophenyl)amino]propyl})amino]ethanol (INT-A6-3). To a stirred mixture of 2-[(2-aminopropyl)(ethyl)amino]ethanol (590 mg, about 4 mmol, about 1 equivalent) and o-fluoronitrobenzene (569 mg, 4.03 mmol, 1 equivalent) in acetonitrile (20 mL) was added K 2CO 3(1115 mg, 8.07 mmol, 2 eq.). The resulting mixture was stirred at 60 °C for 2 h and cooled to room temperature. The reaction was quenched with water (20 mL), and the mixture was extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (12/1) to give 640 mg of 2-[ethyl({2-[(2-nitrophenyl)amino]propyl})amino]ethanol (INT-A6-3) (59%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=268.10. 1H NMR (CDCl 3, 400 MHz) δ 1.16 (3H, t), 1.35 (3H, d), 2.34-3.01 (7H, m), 3.64-3.68 (2H, m), 6.68 (1H, t), 6.96 (1H, s), 7.47 (1H, t), 8.18-8.21 (2H, m). Step 4. Preparation of 5-(5-{2-[ethyl({2-[(2-nitrophenyl)amino]propyl})amino]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A6-4). To a stirred mixture of triphenylphosphine (1766 mg, 6.73 mmol, 3 eq.) and methyl 5-(5-hydroxy-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (M2, 590 mg, 2.24 mmol, 1 eq.) in tetrahydrofuran (20 mL) was added diisopropyl azodicarboxylate (1361 mg, 6.73 mmol, 3 eq.). The mixture was stirred at 0 °C for 0.5 h under nitrogen atmosphere. To the above mixture was added 2-[ethyl({2-[(2-nitrophenyl)amino]propyl})amino]ethanol (600 mg, 2.24 mmol, 1 eq.) in tetrahydrofuran (2 mL) at 0 °C over 2 min. The mixture was stirred at 0 °C for another 2 h. The reaction was quenched with water (20 mL), and the mixture was extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using petroleum ether/ethyl acetate (1/3) to give 400 mg of 5-(5-{2-[ethyl({2-[(2-nitrophenyl)amino]propyl})amino]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A6-4) (34%) as a yellow oil. LCMS: m/z (ESI), [M+H] +=513.20. 1H NMR (methanol-d 4, 400 MHz) δ 1.09 (3H, t), 1.23-1.31 (3H, m), 2.60-2.75 (3H, m), 2.84-2.95 (1H, m), 2.96-3.03 (1H, m), 3.66 (3H, s), 3.68 (3H, s), 3.80-3.92 (4H, m), 4.01 (2H, t), 6.42-6.62 (1H, m), 6.99 (1H, d), 7.39 (1H, d), 7.91 (1H, s), 8.05 (1H, d), 8.16 (1H, d), 8.35 (1H, d). Step 5. Preparation of methyl 5-{5-[2-({2-[(2-aminophenyl)amino]propyl}(ethyl)amino)ethoxy]-1-methylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylate (INT-A6-5). To a stirred mixture of methyl 5-(5-{2-[ethyl({2-[(2-nitrophenyl)amino]propyl})amino]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (450 mg, 0.87 mmol, 1 eq) and ranyl nickel (50 mg) in methanol (5 mL) was added hydrazine hydrate (140 mg, 4.39 mmol, 5 eq) at 0°C. The mixture was stirred at room temperature under nitrogen atmosphere for 1 h and filtered. The filter cake was washed with methanol (3×20 mL), and the solution was concentrated under reduced pressure to give 260 mg of methyl 5-{5-[2-({2-[(2-aminophenyl)amino]propyl}(ethyl)amino)ethoxy]-1-methylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylate (INT-A6-5) (61%) as a yellow oil. LCMS: m/z (ESI), [M+H] +=483.10. Step 6. Preparation of dimethyl 5-[5-(2-{[2-(2-amino-1,3-benzodiazol-1-yl)propyl](ethyl)amino}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylate (INT-A6-6). A mixture of methyl 5-{5-[2-({2-[(2-aminophenyl)amino]propyl}(ethyl)amino)ethoxy]-1-methylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylate (260 mg, 0.53 mmol, 1 eq.) and BrCN (114 mg, 1.07 mmol, 2 eq.) in ethanol (5 mL) was stirred at room temperature under a nitrogen atmosphere for 1 h and concentrated under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (20/1) to give 250 mg of 5-[5-(2-{[2-(2-amino-1,3-benzodiazol-1-yl)propyl](ethyl)amino}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid dimethyl ester (INT-A6-6) (91%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=508.40. Step 7. Preparation of 5-[5-(2-{[2-(2-amino-1,3-benzodiazol-1-yl)propyl](ethyl)amino}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A6-7). To a stirred solution of 5-[5-(2-{[2-(2-amino-1,3-benzodiazol-1-yl)propyl](ethyl)amino}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (240 mg, 0.47 mmol, 1 equiv.) in tetrahydrofuran (6 mL) was added LiOH.H 2O (33 mg, 1.41 mmol, 3 equiv) of H 2O (1.5 mL). The resulting mixture was stirred at room temperature for 2 h and concentrated under reduced pressure. The residue was purified by reverse flash chromatography using a C18 silica gel column using water and acetonitrile as mobile phases. The fractions containing the desired compound were evaporated to dryness to give 200 mg [5-(2-{[2-(2-amino-1,3-benzodiazol-1-yl)propyl](ethyl)amino}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A6-7) (85%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=494.40. Step 8. Preparation of 10-ethyl-5,12,26-trimethyl-7-oxa-4,5,10,13,20, 22,26-heptaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octacosa-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (Example A6). 5-[5-(2-{[2-(2-amino-1,3-benzodiazol-1-yl)propyl](ethyl)amino}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid (110 mg, 0.22 mmol, 1 equivalent) and N,N-Diisopropylethylamine (115 mg, 0.89 mmol, 4 equivalents) was added to the stirred mixture in 1,4-dioxane (6 mL) N, N, N, N-Tetramethyl- O-(7-Azabenzotriazol-1-yl)uronium hexafluorophosphate (169 mg, 0.44 mmol, 2 equiv). The resulting mixture was stirred at 60 °C for 2 h, cooled to room temperature, and concentrated under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (12/1) and by Prep-HPLC using an XBridge Shield RP18 OBD column with 0.1% NH4HCO 3and 0.1% NH 3.H 2O water and acetonitrile for further purification. The fractions containing the desired compound were evaporated to dryness to give 28.8 mg of 10-ethyl-5,12,26-trimethyl-7-oxa-4,5,10,13,20,22,26-heptaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octacosa-1(28),2(6), 3,14,16,18,20,24-octaene-23,27-dione (Example A6) (27%) as a white solid. LCMS: m/z (ESI), [M+H] +=476.30. 1H NMR (DMSO-d 6, 400 MHz) δ 1.07 (3H, t), 1.62 (3H, d), 2.53-2.65 (2H, m), 2.75-2.82 (2H, m), 3.33-3.58 (2H, m), 3.62 (3H, s), 3.75 (3H, s), 4.09 (1H, s), 4.28-4.37 (1H, m), 5.15 (1H, s), 7.14-7.29 (2H, m), 7.47-7.60 (1H, m), 7.60-7.71 (1H, m), 8.17 (1H, s), 8.27 (1H, d), 8.88 (1H, d), 12.54 (1H, s). Example A7 10-ethyl-5,12,12,26-tetramethyl-7-oxa-4,5,10,13,20,22,26-heptaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octadecene-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione Step 1. NPreparation of tert-butyl-{1-[ethyl(2-hydroxyethyl)amino]-2-methylpropan-2-yl}carbamate (INT-A7-1).NA mixture of tert-butyl-(2-methyl-1-oxopropan-2-yl)carbamate (5 g, 26.70 mmol, 1 eq.), triethylamine (8 g, 80.11 mmol, 3 eq.) and ethylethanolamine (2.4 g, 26.70 mmol, 1.00 eq.) in dichloromethane (50 mL) was stirred at room temperature under nitrogen atmosphere for 1 h. NaBH(OAc) was then added.3(17 g, 80.11 mmol, 3 equiv.), and the mixture was stirred at room temperature under nitrogen atmosphere overnight. The reaction was quenched with water (300 mL), and the mixture was extracted with ethyl acetate (3×200 mL). The combined organic layers were washed with brine (3×300 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue is purified by passing through a C18 column using water and acetonitrile as mobile phase. The fractions containing the desired compound are evaporated to dryness to obtain 5 g of N-tert-butyl {1-[ethyl(2-hydroxyethyl)amino]-2-methylpropan-2-yl}carbamate (INT-A7-1) (72%). LCMS: m/z (ESI), [M+H] +=261.10. 1H NMR (DMSO- d 6, 400 MHz) δ 0.93 (3H, t), 1.13-1.14 (6H, s), 1.36-1.39 (9H, d), 2.49-2.53 (3H, m), 2.53-2.56 (3H, m), 3.42-3.43 (2H, m), 4.31 (1H, s), 6.25 (1H, s). Step 2. Preparation of 2-((2-amino-2-methylpropyl)(ethyl)amino)ethan-1-ol (INT-A7-2). NA mixture of tert-butyl-{1-[ethyl(2-hydroxyethyl)amino]-2-methylpropan-2-yl}carbamate (2 g, 3.84 mmol, 1 equiv) in trifluoroacetic acid (5 mL) and dichloromethane (15 mL) was stirred at room temperature under nitrogen atmosphere for 2 h. The resulting mixture was concentrated under reduced pressure. The residue was treated with saturated NaHCO3(aqueous solution) solution (100 mL) and extracted with dichloromethane (3×100 mL). The combined organic layers were washed with brine (3×200 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure to give 1.23 g of 2-((2-amino-2-methylpropyl)(ethyl)amino)ethan-1-ol (INT-A7-2) as a colorless oil. This material was used in the next step without further purification. LCMS: m/z (ESI), [M+H] +=161.10. Step 3. Preparation of 2-[ethyl({2-methyl-2-[(2-nitrophenyl)amino]propyl})amino]ethanol (INT-A7-3). 2-[(2-amino-2-methylpropyl) (ethyl)amino]ethanol (2 g, 12.48 mmol, 1 equivalent), K 2CO 3A mixture of (5 g, 37.44 mmol, 3 equiv.) and o-fluoronitrobenzene (1.7 g, 12.48 mmol, 1 equiv.) in acetonitrile (20 mL) was stirred at 80 °C overnight under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (100 mL), and the mixture was extracted with dichloromethane (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by prep-TLC using dichloromethane/methanol (20/1) to give 1.2 g of 2-[ethyl({2-methyl-2-[(2-nitrophenyl)amino]propyl})amino]ethanol (INT-A7-3) (34%) as a yellow oil. LCMS: m/z (ESI), [M+H] +=282.05. 1H NMR (DMSO- d 6, 400 MHz) δ 0.97 (3H, t), 1.41 (6H, s), 2.62-2.66 (3H, m), 2.69 (2H, s), 3.17 (1H, d), 3.44-3.50 (2H, m), 4.37 (1H, t), 6.62-6.68 (1H, m), 7.26-7.33 (1H, m), 7.44-7.52 (1H, m), 8.04-8.10 (1H, m), 8.62-8.68 (1H, s). Step 4. Preparation of methyl 5-(5-(2-(ethyl(2-methyl-2-((2-nitrophenyl)amino)propyl)amino)ethoxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxo-1,6-dihydropyridine-3-carboxylate (INT-A7-4). To a stirred mixture of methyl 5-(5-hydroxy-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (M2, 281 mg, 1.06 mmol, 1 eq), triphenylphosphine (839 mg, 3.19 mmol, 3 eq) in tetrahydrofuran (10 mL) under nitrogen atmosphere at 0 °C were added diisopropyl azodicarboxylate (646 mg, 3.19 mmol, 3 eq) and 2-[ethyl({2-methyl-2-[(2-nitrophenyl)amino]propyl})amino]ethanol (300 mg, 1.06 mmol, 1 eq). The mixture was stirred for 1 h, treated with water (100 mL), and extracted with dichloromethane (2 x 100 mL). The combined organic layers were washed with brine (2×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by prep-TLC using dichloromethane/methanol (12/1) to give 230 mg of 5-(5-(2-(ethyl(2-methyl-2-((2-nitrophenyl)amino)propyl)amino)ethoxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxo-1,6-dihydropyridine-3-carboxylic acid methyl ester (INT-A7-4) (41%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=527.20. 1H NMR (DMSO- d 6, 400 MHz) δ 0.98 (3H, t), 1.39 (6H, s), 2.67-2.69 (2H, m), 2.74 (2H, s), 2.98 (2H, t), 3.56 (3H, s), 3.65 (3H, s), 3.77 (3H, s), 4.00 (2H, t), 5.77 (1H, s), 6.62-6.67 (1H, m), 7.23-7.28 (1H, d), 7.41-7.48 (1H, m), 7.90-7.93 (1H, s), 8.00-8.04 (2H, m), 8.40 (1H, d), 8.58 (1H, s) Step 5. Preparation of 5-{5-[2-({2-[(2-aminophenyl)amino]-2-methylpropyl} (ethyl)amino)ethoxy]-1-methylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A7-5). To a stirred mixture of 5-(5-{2-[ethyl({2-methyl-2-[(2-nitrophenyl)amino]propyl})amino]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (230 mg, 0.44 mmol, 1 equiv.) and ranyl nickel (50 mg) in methanol (2 mL) was added NH 2NH 2 .H 2O (22 mg, 0.44 mmol, 1 eq.). The mixture was stirred at 0 °C for 1 h. The reaction was quenched with water (50 mL), and the mixture was extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by prep-TLC using dichloromethane/methanol (10/1) to give 110 mg of 5-{5-[2-({2-[(2-aminophenyl)amino]-2-methylpropyl}(ethyl)amino)ethoxy]-1-methylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A7-5) (51%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=497.40. 1H NMR (DMSO- d 6, 400 MHz) δ 0.97 (3H, t), 1.14 (6H, s), 2.62-2.69 (2H, m), 2.94 (2H, t), 3.18 (2H, d), 3.58 (3H, s), 3.67 (4H, s), 3.80 (3H, s), 4.00 (2H, t), 4.38 (5H, s), 4.47 (2H, s), 6.35-6.39 (2H, m), 6.47-6.54 (4H, m), 6.72 (1H, d), 7.96 (1H, s), 8.11 (1H, d), 8.46 (1H, d). Step 6. Preparation of methyl 5-(5-(2-((2-(2-amino-1H-benzo[d]imidazol-1-yl)-2-methylpropyl)(ethyl)amino)ethoxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxo-1,6-dihydropyridine-3-carboxylate (INT-A7-6). A mixture of methyl 5-{5-[2-({2-[(2-aminophenyl)amino]-2-methylpropyl}(ethyl)amino)ethoxy]-1-methylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylate (200 mg, 0.40 mmol, 1 eq) and BrCN (51 mg, 0.48 mmol, 1.2 eq) in dichloromethane (5 mL) was stirred under nitrogen atmosphere at room temperature for 2 h and concentrated under vacuum. The residue was purified by prep-TLC using dichloromethane/methanol (12/1) to give 120 mg of 5-(5-(2-((2-(2-amino-1H-benzo[d]imidazol-1-yl)-2-methylpropyl)(ethyl)amino)ethoxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxo-1,6-dihydropyridine-3-carboxylic acid methyl ester (INT-A7-6) (61%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=522.30. 1H NMR (DMSO- d 6, 400 MHz) δ 0.82 (3H, t), 1.81 (6H, s), 2.44-2.48 (3H, m), 2.79 (2H, t), 3.02 (2H, s), 3.58 (6H, d), 3.78-3.82 (5H, m), 6.88 (1H, t), 6.99 (1H, t), 7.19 (1H, s), 7.55 (1H, d), 7.95 (1H, s), 8.05 (1H, d), 8.47 (1H, d) Step 7. Preparation of 5-(5-(2-((2-(2-amino-1H-benzo[d]imidazol-1-yl)-2-methylpropyl)(ethyl)amino)ethoxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxo-1,6-dihydropyridine-3-carboxylic acid (INT-A7-7). 5-[5-(2-{[2-(2-amino-1,3-benzodiazol-1-yl)-2-methylpropyl](ethyl)amino}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (120 mg, 0.23 mmol, 1 equiv) and LiOH .H 2O (39 mg, 0.92 mmol, 4 equivalents) in tetrahydrofuran/H 2O (2 mL/0.5 mL) was stirred at 40 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by C18 column using water and acetonitrile as mobile phase. The fractions containing the desired compound were evaporated to dryness to give 65 mg of 5-(5-(2-((2-(2-amino-1H-benzo[d]imidazol-1-yl)-2-methylpropyl)(ethyl)amino)ethoxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxo-1,6-dihydropyridine-3-carboxylic acid (INT-A7-7) (56%) as a white solid. LCMS: m/z (ESI), [M+H] +=508.15. 1H NMR (DMSO- d 6, 400 MHz) δ 0.77 (3H, t), 1.79 (6H, s), 2.35-2,42 (3H, m), 2.67-2.69 (1H, m), 2.81 (2H, t), 2.99 (2H, s), 3.48 (3H, s), 3.56 (3H, s), 3.82 (2H, t), 6.30 (2H, s), 6.78 (1H, t), 6.86-6.93 (1H, m), 7.06-7.11 (1H, m), 7.46 (1H, d), 7.86 (1H, s), 7.96 (1H, s), 8.17 (1H, d) Step 8. Preparation of 10-ethyl-5,12,12,26-tetramethyl-7-oxa-4,5,10,13,20,22,26-heptaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octacosa-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (Example A7). 5-[5-(2-{[2-(2-amino-1,3-benzodiazol-1-yl)-2-methylpropyl](ethyl)amino}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid (65 mg, 0.13 mmol, 1 equivalent), N,N,N,N-tetramethyl-O-(7-azabenzotriazol-1-yl)uronium hexafluorophosphate (97 mg, 0.25 mmol, 2 equivalents) andN,N- A mixture of diisopropylethylamine (33 mg, 0.25 mmol, 2 equiv.) in 1,4-dioxane (2 mL) was stirred at 60 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (50 mL), and the mixture was extracted with dichloromethane (3×50 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was subjected to prep-HPLC using an XBridge Shield RP18 OBD column with 10 mmol/L NH4HCO 3and 0.1% NH 3 .H 2O water and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 13.6 mg of 10-ethyl-5,12,12,26-tetramethyl-7-oxa-4,5,10,13,20,22,26-heptaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octacosa-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (Example A7) (22%) as a white solid. LCMS: m/z (ESI), [M+H] += 490.20. 1H NMR (DMSO- d 6, 400 MHz) δ 0.99 (3H, t), 1.94 (6H, s), 2.68 (2H, d), 3.08-3.22 (2H, m), 3.60-3.64 (5H, m), 3.73 (3H, s), 4.28 (2H, d), 7.17 (2H, t), 7.56-7.63 (1H, m), 7.83 (1H, d), 8.13 (1H, d), 8.29 (1H, d), 8.67 (1H, d), 12.93 (1H, s). Example A8 15,21-Dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazahexacyclo[24.4.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontria-3,5,7,9,13,17(32),18(22),19-octaene-12,16-dione Step 1. Preparation of tert-butyl 3-((2-nitrophenyl)amino)azoloheptane-1-carboxylate (INT-A8-1). To a stirred mixture of o-fluoronitrobenzene (5 g, 35.43 mmol, 1 eq.) and tert-butyl 3-aminoazoloheptane-1-carboxylate (7.9 g, 37.20 mmol, 1.05 eq.) in acetonitrile (80 mL) was added K 2CO 3(9.7 g, 70.87 mmol, 2 eq.). The mixture was stirred at 80 °C for 3 h under nitrogen atmosphere and cooled to room temperature. The reaction was quenched with water (100 mL) and the mixture was washed with CH2Cl 2(3×100 mL) extraction. The combined organic layers were washed with brine (3×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column and eluted with petroleum ether/ethyl acetate (10:1) to give 10.6 g of tert-butyl 3-[(2-nitrophenyl)amino]azanacycloheptane-1-carboxylate (INT-A8-1) (89%) as a brown oil. LCMS: m/z (ESI), [M+H] +=336.15. 1H NMR (DMSO-d 6, 400 MHz) δ 1.37 (9H, d), 1.47-2.03 (6H, m), 3.34-3.49 (3H, m), 3.51-3.62 (1H, m), 3.93 (1H, s), 6.61-6.75 (1H, m), 7.20 (1H, m), 7.47-7.55 (1H, m), 7.97-8.22 (2H, m). Step 2. N- Preparation of (2-nitrophenyl) azacycloheptane-3-amine (INT-A8-2). A mixture of tert-butyl 3-[(2-nitrophenyl) amino] azacycloheptane-1-carboxylate (12 g, 35.77 mmol, 1 eq.) and HCl in 1,4-dioxane (100 mL) was stirred at room temperature for 1.5 h. The mixture was concentrated under reduced pressure and treated with CH2Cl 2(60 mL). The mixture was treated with saturated NaHCO3(aqueous) solution (50 mL) and treated with CH 2Cl 2(3×100 mL) extraction. The combined organic layers were purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure to obtain 8 g of yellow oil.N-(2-Nitrophenyl)azinecycloheptyl-3-amine (INT-A8-2). This material was used in the next reaction without further purification. LCMS: m/z (ESI), [M+H] +=236.20. Step 3. 1-(2-((tert-butyldimethylsilyl)oxy)ethyl)- N-(2-nitrophenyl) azacycloheptyl-3-amine (INT-A8-3). To N-(2-nitrophenyl) azacycloheptyl-3-amine (1 g, 4.25 mmol, 1 equivalent) and (2-bromoethoxy)(tert-butyl)dimethylsilane (2.03 g, 8.50 mmol, 2 equivalents) were added to N, N-Dimethylformamide (15 mL) was added to the stirred mixture.2CO 3(1.17 g, 8.50 mmol, 2 equiv) and KI (1.41 g, 8.50 mmol, 2 equiv). The resulting mixture was stirred at 60 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (100 mL), and the mixture was washed with CH2Cl 2(3×100 mL) extraction. The combined organic layers were washed with brine (3×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column and eluted with ethyl acetate/petroleum ether (1/10) to give 819 mg of 1-{2-[(tert-butyldimethylsilyl)oxy]ethyl}- N-(2-Nitrophenyl) azacycloheptyl-3-amine (INT-A8-3) (48%). LCMS: m/z (ESI), [M+H] +=394.20. 1H NMR (DMSO-d 6, 400 MHz) δ -0.02 (6H, d), 0.81 (9H, s), 1.35-1.65 (3H, m), 1.65-1.85 (3H, m), 2.57-2.80 (3H, m), 2.81-3.03 (3H, m), 3.61-3.82 (2H, m), 3.93 (1H, s), 6.64-6.66 (1H, m), 7.04 (1H, d), 7.45-7.55 (1H, m), 8.02-8.09 (1H, m), 8.73 (1H, d). Step 4. Preparation of 2-(3-((2-nitrophenyl)amino)azoloheptan-1-yl)ethan-1-ol (INT-A8-4). To 1-{2-[(tert-butyldimethylsilyl)oxy]ethyl}- NTo a solution of -(2-nitrophenyl)azanidinecycloheptyl-3-amine (447 mg, 1.13 mmol, 1 eq.) in tetrahydrofuran (10 mL) was added tetrabutylammonium fluoride (593 mg, 2.272 mmol, 2 eq.). The mixture was stirred at room temperature for 2 h. The reaction was quenched with water (50 mL), and the mixture was extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure to give 447 mg of 2-{3-[(2-nitrophenyl)amino]azanacycloheptan-1-yl}ethanol (INT-A8-4) as a yellow oil. This material was used in the next reaction without further purification. LCMS: m/z (ESI), [M+H] +=280.05. Step 5. Preparation of 1-methyl-5-(1-methyl-5-(2-(3-((2-nitrophenyl)amino)azinecyclohept-1-yl)ethoxy)-1H-pyrazol-4-yl)-6-oxo-1,6-dihydropyridine-3-carboxylic acid methyl ester (INT-A8-5). To a stirred mixture of methyl 5-(5-hydroxy-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (M2, 207 mg, 0.78 mmol, 1.1 eq.) and triphenylphosphine (563 mg, 2.14 mmol, 3 eq.) in tetrahydrofuran (10 mL) at 0° C. under nitrogen atmosphere were added diisopropyl azodicarboxylate (434 mg, 2.14 mmol, 3 eq.) and 2-{3-[(2-nitrophenyl)amino]azanacycloheptan-1-yl}ethanol (200 mg, 0.71 mmol, 1 eq.). The resulting mixture was stirred at room temperature overnight. The reaction was quenched with water (50 mL), and the mixture was extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using ethyl acetate to give 301 mg of 1-methyl-5-[1-methyl-5-(2-{3-[(2-nitrophenyl)amino]azanacyclohept-1-yl}ethoxy)pyrazol-4-yl]-6-oxopyridine-3-carboxylic acid methyl ester (INT-A8-5) (80%) as an orange solid. LCMS: m/z (ESI), [M+H] +=525.30 1H NMR (DMSO-d 6, 400 MHz) δ 1.22-1.63 (7H, m), 2.45-2.68 (1H, m), 2.79-2.89 (2H, m), 2.95-3.05 (3H, m), 3.55 (3H, s), 3.68 (3H, s), 3.73 (3H, s), 4.05-4.10 (2H, m), 6.60-6.70 (1H, m), 6.95-7.09 (1H, m), 7.40-7.53 (1H,m), 7.94 (1H, s), 7.99-8.05 (2H, m), 8.32 (1H, d), 8.61 (1H, d) Step 6. Preparation of 5-(5-(2-(3-((2-aminophenyl)amino)azepan-1-yl)ethoxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A8-6). To a stirred mixture of ranyl nickel (100 mg) and 1-methyl-5-[1-methyl-5-(2-{3-[(2-nitrophenyl)amino]azepan-1-yl}ethoxy)pyrazol-4-yl]-6-oxopyridine-3-carboxylic acid methyl ester (300 mg, 0.54 mmol, 1 equivalent) in methanol (10 mL) at 0°C was added NH 2NH 2 .H 2O (84 mg, 0.28 mmol, 3 eq.). The mixture was stirred at room temperature for 2 h and filtered. The filter cake was washed with methanol (3×5 mL), and the solution was concentrated under reduced pressure. The residue was purified by Prep-TLC and eluted with dichloromethane/methanol (10/1) to give 89 mg of 5-[5-(2-{3-[(2-aminophenyl)amino]azanacyclohept-1-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A8-6) (44%) as a yellow oil. LCMS: m/z (ESI), [M+H] +=495.30 Step 7. Preparation of 5-(5-(2-(3-(2-amino-1H-benzo[d]imidazol-1-yl)azanthene-1-yl)ethoxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxo-1,6-dihydropyridine-3-carboxylic acid methyl ester (INT-A8-7). 5-[5-(2-{3-[(2-aminophenyl)amino]azanthene-1-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (89 mg, 0.18 mmol, 1 equiv) and BrCN (20 mg, 0.19 mmol, 1.10 equiv) were mixed in CH 2Cl 2The mixture in 4% paraformaldehyde (5 mL) was stirred at room temperature for 5 h and concentrated under reduced pressure. The residue was purified by Prep-TLC and eluted with dichloromethane/methanol (10/1) to give 21 mg of 5-(5-{2-[3-(2-amino-1,3-benzodiazol-1-yl)azinecyclohept-1-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid dimethyl ester (INT-A8-7) (22%) as a white solid. LCMS: m/z (ESI), [M+H] +=520.25 Step 8. Preparation of 5-(5-(2-(3-(2-amino-1H-benzo[d]imidazol-1-yl)azinecyclohept-1-yl)ethoxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxo-1,6-dihydropyridine-3-carboxylic acid (INT-A8-8). To 5-(5-{2-[3-(2-amino-1,3-benzodiazol-1-yl)azinecyclohept-1-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (60 mg, 0.11 mmol, 1 equivalent) in tetrahydrofuran (4 mL) and H 2Add LiOH.H O(1 mL) to the stirred mixture.2O (8 mg, 0.34 mmol, 3 eq.). The resulting mixture was stirred at room temperature for 8 h and concentrated under reduced pressure. The mixture was purified by reverse flash chromatography using a C18 silica gel column using water and acetonitrile as mobile phases. The fractions containing the desired compound were evaporated to dryness to give 16.1 mg of 5-(5-{2-[3-(2-amino-1,3-benzodiazol-1-yl)azanylcyclohept-1-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A8-8) (42%) as a white solid. LCMS: m/z (ESI), [M+H] +=506.20 Step 9. Preparation of 15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazahexacyclo[24.4.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontria-3,5,7,9,13,17(32),18(22),19-octaene-12,16-dione (Example A8). To a stirred mixture of 5-(5-{2-[3-(2-amino-1,3-benzodiazol-1-yl)azanylcycloheptyl-1-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (15 mg, 0.03 mmol, 1 eq.) and N,N,N,N-tetramethyl-O-(7-azanylbenzotriazol-1-yl)uronium hexafluorophosphate (33 mg, 0.09 mmol, 3 eq.) in 1,4-dioxane (2 mL) was added N,N-Diisopropylethylamine (11 mg, 0.09 mmol, 3 equiv.). The resulting mixture was stirred at room temperature for 3 h and concentrated. The residue was subjected to Prep-HPLC using an XBridge Shield RP18 OBD column with 0.1% NH4HCO 3of water and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 4 mg of 15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazahexacyclo[24.4.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontidine-3,5,7,9,13,17(32), 18(22),19-octaene-12,16-dione (Example A8) as a white solid (27%). LCMS: m/z (ESI), [M+H] +=488.15. 1H NMR (DMSO-d 6, 400 MHz) δ 1.65-2.03 (5H, m), 2.54-2.72 (1H, m), 2.72-3.03 (5H, m), 3.08-3.22 (1H, m), 3.63 (3H, s), 3.73 (3H, s), 3.80-3.94 (1H, m), 4.34-4.60 (1H, m), 5.14 (1H, s), 7.16-7.26 (2H, m), 7.50-7.57 (1H, m), 7.62 (1H, d), 8.27 (1H, d), 8.31 (1H, s), 8.98 (1H, s), 12.50 (1H, s). Example A9 15,21-Dimethyl-5-(morpholin-4-yl)-23-oxo-2,9,11,15,20,21, 26-heptaazaheptacyclic [24.4.1.1^{1,27}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]tritriacontahedral-3,5,7,9,13,17(33), 18(22),19-octaene-12,16-dione Step 1. Preparation of tert-butyl 1-[(5-bromo-2-nitrophenyl)amino]-6-azabicyclo[3.2.1]octane-6-carboxylate (INT-A9-1). tert-butyl 1-amino-6-azabicyclo[3.2.1]octane-6-carboxylate hydrochloride (1 g, 3.81 mmol, 1 equivalent), 4-bromo-2-fluoro-1-nitrobenzene (2.51 g, 11.42 mmol, 3 equivalents) and K 2CO 3(2.10 g, 15.22 mmol, 4 equiv.) in acetonitrile (20 mL) was stirred at 60 °C for 2 h. After cooling to room temperature, the reaction was treated with water (200 mL), and the mixture was extracted with ethyl acetate (3×200 mL). The combined organic layers were washed with brine (3×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by silica gel chromatography using petroleum ether/ethyl acetate (4/1) to give 1.25 g of tert-butyl 1-[(5-bromo-2-nitrophenyl)amino]-6-azabicyclo[3.2.1]octane-6-carboxylate (INT-A9-1) (77%) as a yellow solid. LCMS: m/z (ESI), [M+H - 56] +=371.85. 1H NMR (DMSO-d 6, 400 MHz) δ 1.40-1.44 (10H, m), 1.48-1.75 (2H, m), 1.79-1.82 (1H, m), 1.98-2.00 (1H, m), 2.13-2.29 (3H, m), 3.23-3.33 (1H, m), 4.01-4.12 (2H, m), 6.92 (1H, d), 7.20 (1H, s), 8.03 (1H, d), 8.28 (1H, s). Step 2. N-Preparation of (5-bromo-2-nitrophenyl)-6-azabicyclo[3.2.1]octan-1-amine (INT-A9-2). To a stirred mixture of tert-butyl 1-[(5-bromo-2-nitrophenyl)amino]-6-azabicyclo[3.2.1]octane-6-carboxylate (1.28 g, 3.00 mmol, 1 eq.) in dichloromethane (10 mL) was added TFA (10 mL) at room temperature. The resulting mixture was stirred at room temperature for 2 h and concentrated under reduced pressure. The residue was treated with saturated NaHCO 3(aqueous solution) solution (200 mL) and extracted with dichloromethane (3×200 mL). The combined organic layers were washed with anhydrous Na 2SO 4Dry and concentrate under reduced pressure to give 950 mg of a pale yellow solid.N-(5-Bromo-2-nitrophenyl)-6-azabicyclo[3.2.1]octan-1-amine (INT-A9-2) (97%). LCMS: m/z (ESI), [M+H] += 327.90. 1H NMR (CD 3OD, 400 MHz) δ 1.51-1.64 (1H, m), 1.72-1.76 (2H, m), 1.80-1.95 (2H, m), 1.97 (1H, d), 2.21-2.33 (2H, m), 3.02-3.06 (1H, m), 3.54-3.65 (2H, m), 6.83-6.86 (1H, m), 7.14 (1H, d), 8.07 (1H, d). Step 3. N-(5-bromo-2-nitrophenyl)-6-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-6-azabicyclo[3.2.1]octan-1-amine (INT-A9-3) preparation.N-(5-Bromo-2-nitrophenyl)-6-azabicyclo[3.2.1]octan-1-amine (580 mg, 1.78 mmol, 1 equivalent), (2-bromoethoxy)(tert-butyl)dimethylsilane (850 mg, 3.56 mmol, 2 equivalents), NaI (533 mg, 3.56 mmol, 2 equivalents) and K 2CO 3(491 mg, 3.56 mmol, 2 equivalents) in N,N-The mixture in dimethylformamide (10 mL) was stirred at 60 °C for 2 h under nitrogen atmosphere. After cooling to room temperature, the reaction was quenched with water (100 mL), and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was further purified by reverse phase flash chromatography using 20% to 100% acetonitrile/water to give 600 mg N-(5-Bromo-2-nitrophenyl)-6-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-6-azabicyclo[3.2.1]octan-1-amine (INT-A9-3) (69%). LCMS: m/z (ESI), [M+H] +=484.51. 1H NMR (CD 3OD, 400 MHz) δ 0.11 (6H, d), 0.93 (9H, s), 1.38-1.57 (2H, m), 1.62-1.78 (2H, m), 1.83-1.86 (2H, m), 2.00 (1H, d), 2.28 (1H, d), 2.33-2.42 (1H, m), 2.91 (2H, t), 3.36-3.42 (2H, m), 3.79-3.83 (2H, m), 6.82-6.86 (1H, m), 7.09 (1H, d), 8.07 (1H, d). Step 4. Preparation of 2-{1-[(5-bromo-2-nitrophenyl)amino]-6-azabicyclo[3.2.1]octan-6-yl}ethanol (INT-A9-4). At room temperature,N-To a stirred mixture of (5-bromo-2-nitrophenyl)-6-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-6-azabicyclo[3.2.1]octan-1-amine (598 mg, 1.23 mmol, 1 equiv) in tetrahydrofuran (9 mL) was added tetrabutylammonium fluoride (2 mL, 1 M in tetrahydrofuran). After stirring at room temperature for 2 h, the reaction was quenched with water (100 mL), and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (5×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure to obtain 360 mg of 2-{1-[(5-bromo-2-nitrophenyl)amino]-6-azabicyclo[3.2.1]octan-6-yl}ethanol (INT-A9-4) (78%) as a yellow solid. LCMS: m/z (ESI), [M+H] += 370.24. Step 5. Preparation of 5-[5-(2-{1-[(5-bromo-2-nitrophenyl)amino]-6-azabicyclo[3.2.1]oct-6-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A9-5). To a stirred mixture of methyl 5-(5-hydroxy-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (M2, 268 mg, 1.02 mmol, 1 eq) and triphenylphosphine (714 mg, 2.72 mmol, 2.8 eq) in tetrahydrofuran (10 mL) at 0 °C under nitrogen atmosphere was added diisopropyl azodicarboxylate (491 mg, 2.43 mmol, 2.5 eq) followed by 2-{1-[(5-bromo-2-nitrophenyl)amino]-6-azabicyclo[3.2.1]octan-6-yl}ethanol (360 mg, 0.97 mmol, 1 eq) in tetrahydrofuran (3 mL). After stirring at 0°C for 30 minutes, the reaction was quenched with water (100 mL), and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by silica gel chromatography using petroleum ether/ethyl acetate (1/9) to give 470 mg of 5-[5-(2-{1-[(5-bromo-2-nitrophenyl)amino]-6-azabicyclo[3.2.1]octan-6-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A9-5) (78%) as a light yellow solid. LCMS: m/z (ESI), [M+H] +=617.15. 1H NMR (CD 3OD, 400 MHz) δ 1.39-1.51 (1H, m), 1.62-1.74 (2H, m), 1.85 (3H, q), 2.24-2.28 (2H, m), 3.12-3.16 (2H, m), 3.18-3.25 (3H, m), 3.64 (3H, s), 3.79 (3H, s), 3.83 (3H, s), 4.12-4.16 (2H, m), 6.82-6.86 (1H, m), 7.06 (1H, d), 7.96 (1H, s), 8.08 (1H, d), 8.25-8.33 (2H, m) Step 6. Preparation of 5-[5-(2-{1-[(2-amino-5-bromophenyl)amino]-6-azabicyclo[3.2.1]oct-6-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A9-6). To a stirred mixture of 5-[5-(2-{1-[(5-bromo-2-nitrophenyl)amino]-6-azabicyclo[3.2.1]oct-6-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (460 mg, 0.75 mmol, 1 eq.) and ranyl nickel (128 mg, 1.49 mmol, 2 eq.) in methanol (20 mL) at 0°C was added NH 2NH 2 .H 2O (187 mg, 3.74 mmol, 5 equiv). After stirring at room temperature for 2 h, the reaction mixture was quenched by adding water (100 mL) and extracted with dichloromethane (3×100 mL). The combined organic layers were washed with brine (3×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by silica gel chromatography using dichloromethane/methanol (20/1) to give 370 mg of 5-[5-(2-{1-[(2-amino-5-bromophenyl)amino]-6-azabicyclo[3.2.1]octan-6-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A9-6) (84%) as a reddish brown solid. LCMS: m/z (ESI), [M+H] +=586.95. Step 7. Preparation of 5-(5-{2-[1-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-6-azabicyclo[3.2.1]oct-6-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A9-7). A mixture of methyl 5-[5-(2-{1-[(2-amino-5-bromophenyl)amino]-6-azabicyclo[3.2.1]octan-6-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylate (340 mg, 0.58 mmol, 1 eq) and BrCN (92 mg, 0.87 mmol, 1.5 eq) in EtOH (5 mL) was stirred at room temperature for 4 h. The reaction was stirred by saturated NaHCO3The solution (100 mL) was quenched and extracted with dichloromethane (3×100 mL). The combined organic layers were washed with brine (3×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column using dichloromethane/methanol (20/1) to give 120 mg of 5-(5-{2-[1-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-6-azabicyclo[3.2.1]octan-6-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A9-7) (33%) as a reddish brown solid. LCMS: m/z (ESI), [M+H] +=612.15. Step 8. Preparation of 5-(5-{2-[1-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-6-azabicyclo[3.2.1]oct-6-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A9-8). To a stirred solution of 5-(5-{2-[1-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-6-azabicyclo[3.2.1]octan-6-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (120 mg, 0.20 mmol, 1 eq.) in THF (4 mL) at room temperature was added LiOH (28 mg, 1.18 mmol, 6 eq.) in H 2O (1 mL). The resulting mixture was stirred at room temperature for 2 h and concentrated under reduced pressure. The residue was purified by reverse-phase flash chromatography using 20% to 100% acetonitrile/water elution to give 75 mg of 5-(5-{2-[1-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-6-azabicyclo[3.2.1]octan-6-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A9-8) (63%) as a white solid. LCMS: m/z (ESI), [M+H] += 598.20. Step 9. Preparation of 5-bromo-15,21-dimethyl-23-oxo-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,27}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]tris-3,5,7,9,13,17(33), 18(22),19-octaene-12,16-dione (INT-A9-9). To a stirred solution of 5-(5-{2-[1-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-6-azabicyclo[3.2.1]oct-6-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (70 mg, 0.12 mmol, 1 equiv) in dioxane (2 mL) was added at room temperature N,N-Diisopropylethylamine (45 mg, 0.35 mmol, 3 equivalents) and N,N,N,N-Tetramethyl- O-(7-Azabenzotriazol-1-yl)urea hexafluorophosphate (66 mg, 0.17 mmol, 1.5 equiv). After stirring at room temperature for 2 h, the reaction was quenched with water (80 mL), and the mixture was extracted with dichloromethane (2×80 mL). The combined organic layers were washed with brine (3×80 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by prep-TLC eluting with dichloromethane/methanol (20/1) to give 65 mg of 5-bromo-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,27}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]tritriacontahedral-3,5,7,9,13,17(33), 18(22),19-octaene-12,16-dione (INT-A9-9) (95%) as a brown solid. LCMS: m/z (ESI), [M+H] += 580.00. Step 10. Preparation of 15,21-dimethyl-5-(morpholin-4-yl)-23-oxo-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,27}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tris-3,5,7,9,13,17(33), 18(22),19-octaene-12,16-dione (Example A9). 5-Bromo-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,27}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]tritriacontria-3,5,7,9,13,17(33), 18(22),19-octaene-12,16-dione (50 mg, 0.09 mmol, 1 eq.), morpholine (30 mg, 0.34 mmol, 4 eq.) and BrettPhos Pd G 3To a stirred mixture of 2-nitropropene (23 mg, 0.03 mmol, 0.3 equiv) in dioxane (3 mL) was added LiHMDS (0.34 mL, 0.34 mmol, 4 equiv) dropwise. The reaction mixture was stirred at 60 °C for 30 min. After cooling to room temperature, the reaction was quenched with water (50 mL), and the mixture was extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was subjected to prep-HPLC using an XBridge Shield RP18 OBD column with 10 mmol/L NH4HCO 3and 0.1% NH 3 .H 2O water and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 13 mg of 15,21-dimethyl-5-(morpholin-4-yl)-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,27}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontahedral-3,5,7,9,13,17(33), 18(22),19-octaene-12,16-dione (Example A9) (25%) as a white solid. LCMS: m/z (ESI), [M+H] +=585.35. 1H NMR (CD 3OD, 400 MHz) δ 1.63-1.69 (1H, m), 1.85-1.92 (2H, m), 2.02-2.08 (1H, m), 2.15 (1H, d), 2.26 (1H, q), 2.60-2.65 (1H, m), 2.82 (1H, d), 3.15-3.23 (4H, m), 3.39 (1H, m), 3.47-3.58 (2H, m), 3.68 (3H, s), 3.85 (3H, s), 3.90 (4H, t), 4.41-4.49 (3H, m), 4.61-4.68 (1H, m), 7.00 (1H, m), 7.25 (1H, d), 7.38 (1H, d), 8.20 (1H, s), 8.49 (1H, s), 9.05 (1H, d). Example A10 1,15,21-Trimethyl-5-(morpholin-4-yl)-23-oxo-2,9,11,15, 20,21,26-heptaazahexacyclo[24.3.1.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]tri-11-3,5,7,9,13,17(31),18(22),19-octaene-12,16-dione Step 1. Preparation of tert-butyl 3-((5-bromo-2-nitrophenyl)amino)-3-methylpiperidine-1-carboxylate (INT-A10-1). 4-bromo-2-fluoro-1-nitrobenzene (6.16 g, 27.99 mmol, 3 equivalents), K 2CO 3(3.8 g, 27.99 mmol, 3 equivalents) and tert-butyl 3-amino-3-methylpiperidine-1-carboxylate (2 g, 9.33 mmol, 1.00 equivalents) in N, N-The mixture in dimethylacetamide (40 mL) was stirred at 100 °C for 12 h under nitrogen atmosphere and then cooled to 25 °C. The reaction was quenched with water (300 mL), and the mixture was extracted with ethyl acetate (3×200 mL). The combined organic layers were washed with brine (3×300 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by prep-TLC using petroleum ether/ethyl acetate (3/1) to give 3.5 g of tert-butyl 3-[(5-bromo-2-nitrophenyl)amino]-3-methylpiperidine-1-carboxylate (INT-A10-1) (91%) as a yellow oil. 1H NMR (DMSO-d 6, 400 MHz) δ 1.23 (9H, s), 1.44 (3H, s), 1.54 (2H, s), 1.64 (1H, s), 2.15 (1H, s), 2.78 (1H, s), 2.98 (1H , s), 3.88 (1H, s), 4.16-4.14 (1H, d), 6.86-6.93 (1H, m), 7.30 (1H, d), 8.04-7.98 (1H, d), 8.24 (1H, s ). Step 2. N-Preparation of (5-bromo-2-nitrophenyl)-3-methylpiperidin-3-amine (INT-A10-2). Trifluoroacetic acid (5 mL, 67.31 mmol, 7 equivalents) and tert-butyl 3-[(5-bromo-2-nitrophenyl)amino]-3-methylpiperidin-1-carboxylate (3.5 g, 8.44 mmol, 1 equivalent) were mixed in CH2Cl 2The mixture in 4% paraformaldehyde (15 mL) was stirred at room temperature under nitrogen atmosphere for 2 h and concentrated under reduced pressure. The residue was treated with saturated NaHCO3(aqueous solution) (100 mL) and extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure to obtain 2.4 g of a yellow solid.N-(5-bromo-2-nitrophenyl)-3-methylpiperidin-3-amine (INT-A10-2) (89%). 1H NMR (DMSO-d 6, 400 MHz) δ 1.51 (3H, s), 1.85-1.83 (3H, m), 2.03-2.0 (1H, t), 2.92-2.89 (1H, m), 3.09-3.05 (1H, d), 3.17-3.14 (1H, m), 3.85-3.82 (1H, d), 6.96-6.95 (1H, m), 7.34-7.31 (1H, d), 8.07-8.05 (1H, d), 8.18 (1H, s) Step 3. NPreparation of -(5-bromo-2-nitrophenyl)-1-(2-((tert-butyldimethylsilyl)oxy)ethyl)-3-methylpiperidin-3-amine (INT-A10-3). In a nitrogen atmosphere at 0°C to N-(5-bromo-2-nitrophenyl)-3-methylpiperidin-3-amine (1 g, 3.81 mmol, 1 equiv), 2-[(tert-butyldimethylsilyl)oxy]acetaldehyde (2 g, 15.27 mmol, 4 equiv) and trimethylamine (1.5 g, 15.27 mmol, 4 equiv) in CH 2Cl 2(50 mL) was added to the stirred mixture. 3(1.6 g, 7.63 mmol, 2 equiv.). The mixture was stirred at room temperature for 2 h. The reaction was quenched with water (100 mL), and the mixture was extracted with dichloromethane (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue is purified by silica gel chromatography eluting with petroleum ether/ethyl acetate (5/2) to give 840 mg of yellow oil.N-(5-bromo-2-nitrophenyl)-1-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-3-methylpiperidin-3-amine (INT-A10-3) (46%). 1H NMR (DMSO-d 6, 400 MHz) δ 0.06 (6H, s), 0.86 (9H, s), 1.28-1.25 (1H, td), 1.39 (5H, s), 2.00-2.17 (2H, m), 2.23-2.20 (1H, d), 2.83-2.81 (1H, d), 2.93-2.9 (1H, d), 3.77-3.74(2H, td), 6.82-6.80 (1H, m), 7.22-7.19 (1H, d), 8.01-7.98 (1H, d), 8.78 (1H, s). Step 4. Preparation of 2-(3-((5-bromo-2-nitrophenyl)amino)-3-methylpiperidin-1-yl)ethan-1-ol (INT-A10-4).NA mixture of -(5-bromo-2-nitrophenyl)-1-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-3-methylpiperidin-3-amine (700 mg, 1.48 mmol, 1 equiv) and tetrabutylammonium fluoride (774.7 mg, 2.96 mmol, 2.0 equiv) in tetrahydrofuran (7 mL) was stirred at room temperature under nitrogen atmosphere for 4 h. The reaction mixture was treated with water (30 mL) and extracted with dichloromethane (3×30 mL). The combined organic layers were washed with brine (3×30 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was subjected to prep-TLC using CH2Cl 2/methanol (12/1) for purification to obtain 500 mg of 2-{3-[(5-bromo-2-nitrophenyl)amino]-3-methylpiperidin-1-yl}ethanol (INT-A10-4) (94%) as a brown solid. 1H NMR (DMSO-d 6, 400 MHz) δ 1.19-1.16 (1H, t), 1.29-1.26 (1H, t), 1.41 (4H, s), 1.98-2.13 (3H, m), 2.25-2.23 (1H, d), 2.85-2.83 (2H, d), 3.59-3.57 (2H, t), 4.40-4.36 (1H, t), 6.83-6.80 (1H, d), 7.23-7.21 (1H, d), 8.03-8.01 (1H, d), 8.80 (1H, s). Step 5. Preparation of methyl 5-[5-(2-{3-[(5-bromo-2-nitrophenyl)amino]-3-methylpiperidin-1-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylate (INT-A10-5). To a stirred mixture of methyl 5-(5-hydroxy-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (M2, 350 mg, 1.33 mmol, 1 eq.) and triphenylphosphine (1046 mg, 3.99 mmol, 3 eq.) in tetrahydrofuran (10 mL) was added diisopropyl azodicarboxylate (806 mg, 3.99 mmol, 3 eq.) and 2-{3-[(5-bromo-2-nitrophenyl)amino]-3-methylpiperidin-1-yl}ethanol (523 mg, 1.463 mmol, 1.1 eq.) at 0 °C under nitrogen atmosphere. The mixture was stirred for 2 h. The reaction was quenched with water (100 mL), and the mixture was extracted with dichloromethane (3×100 mL). The combined organic layers were washed with brine (3×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue is purified by C18 FLASH using water and acetonitrile as mobile phase. The fractions containing the desired compound are evaporated to dryness to give 450 mg of 5-[5-(2-{3-[(5-bromo-2-nitrophenyl)amino]-3-methylpiperidin-1-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A10-5) (56%) as a yellow solid. 1H NMR (DMSO-d 6, 400 MHz) δ 1.41 (6H, s), 2.06-2.29 (3H, m), 2.74-2.94 (3H, m), 3.58 (3H, s), 3.69 (3H, s), 3.76 (3H, s), 4.10-4.06 (2H, m), 6.83-6.78 (1H, d), 7.23-7.20 (1H, d), 7.95 (1H, s), 8.01-7.98 (1H, d), 8.11-8.02 (1H, d), 8.44 (1H, d), 8.74 (1H, s). Step 6. Preparation of 5-(5-(2-(3-((2-amino-5-bromophenyl)amino)-3-methylpiperidin-1-yl)ethoxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxo-1,6-dihydropyridine-3-carboxylic acid methyl ester (INT-A10-6). 5-[5-(2-{3-[(5-bromo-2-nitrophenyl)amino]-3-methylpiperidin-1-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A10-5, 430 mg, 0.71 mmol, 1 equivalent), ranyl nickel (61 mg, 0.71 mmol, 1 equivalent) and NH 2NH 2 .H 2A mixture of 1,4-dihydro-2-nitropropene (71 mg, 1.42 mmol, 2 equiv.) in methanol (6 mL) was stirred at 0 °C under nitrogen atmosphere for 0.5 h. The resulting mixture was filtered. The filter cake was washed with methanol (50 mL). The solution was concentrated under reduced pressure to give 340 mg of 5-[5-(2-{3-[(2-amino-5-bromophenyl)amino]-3-methylpiperidin-1-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A10-6) (83%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=573.20. Step 7. Preparation of 5-(5-(2-(3-(2-amino-6-bromo-1H-benzo[d]imidazol-1-yl)-3-methylpiperidin-1-yl)ethoxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxo-1,6-dihydropyridine-3-carboxylic acid methyl ester (INT-A10-7). A mixture of methyl 5-[5-(2-{3-[(2-amino-5-bromophenyl)amino]-3-methylpiperidin-1-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylate (340 mg, 0.59 mmol, 1 eq.) and BrCN (69 mg, 0.65 mmol, 1.1 eq.) in EtOH (5 mL) was stirred at room temperature overnight under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by prep-TLC using CH2Cl 2/methanol (12/1) for purification to obtain 300 mg of 5-(5-{2-[3-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-methylpiperidin-1-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A10-7) (84%) as a brown solid. 1H NMR (DMSO-d 6, 400 MHz) δ 0.85-0.84 (1H, m), 0.98-1.09 (1H, m), 1.25-1.23 (4H, m), 1.49-1.47 (1H, m), 1.59 (4H, s), 2.20-2.07 (1H, m), 2.44-2.40 (1H, d), 2.79-2.67 (2H, m), 2.94-2.90 (1H, m), 3.15-3.12 (2H, m), 3.59 (3H, s), 3.71 (3H, s), 3.76 (3H, s), 4.21-4.06 (2H, m), 7.03-7.12 (2H, m), 7.35 (2H, s), 7.50-7.45 (1H, d), 7.95 (1H, s), 8.11-8.05 (1H, d), 8.47-8.45 (1H, d). Step 8. Preparation of 5-(5-(2-(3-(2-amino-6-bromo-1H-benzo[d]imidazol-1-yl)-3-methylpiperidin-1-yl)ethoxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxo-1,6-dihydropyridine-3-carboxylic acid (INT-A10-8). 5-(5-{2-[3-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-methylpiperidin-1-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (290 mg, 0.48 mmol, 1 equivalent) and LiOH .H 2O (40 mg, 0.970 mmol, 2 eq.) in THF/H 2O (2 mL/0.5 mL) was stirred at room temperature for 2 h under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by C18FLASH using water and acetonitrile as mobile phase. The fractions containing the desired compound were evaporated to dryness to give 230 mg of 5-(5-{2-[3-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-methylpiperidin-1-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A10-8) (81%) as a brown solid. 1H NMR (400 MHz, DMSO-d 6) δ 1.21-1.31 (1H, m), 1.49 (1H, t), 1.58 (4H, s), 2.14 (1H, t), 2.42 (1H, d), 2.70-2.84 (2H, m), 2.87-2.98 (1H, m), 3.13 (1H, d), 3.50 (3H, s), 3.69 (3H, s), 4.04-4.17 (2H, m), 7.08 (2H, d), 7.48 (1H, s), 7.54 (2H, s), 7.88 (1H, s), 7.99 (1H, t), 8.18 (1H, d). Step 9. Preparation of 5-bromo-1,15,21-trimethyl-23-oxa-2,9,11,15,20,21,26-heptaazahexacyclo[24.3.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontene-3,5,7,9,13,17(31),18(22),19-octaene-12,16-dione (INT-A10-9). 5-(5-{2-[3-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-methylpiperidin-1-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (230 mg, 0.39 mmol, 1 equivalent), N,N-Diisopropylethylamine (152.58 mg, 1.182 mmol, 3 equivalents) and N,N,N,N-Tetramethyl- OA mixture of -(7-azabenzotriazol-1-yl)uronium hexafluorophosphate (224.44 mg, 0.59 mmol, 1.5 eq.) in dioxane (5 mL) was stirred at 60° C. for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by P\prep-TLC using dichloromethane/methanol (10/1) to give 180 mg of 5-bromo-1,15,21-trimethyl-23-oxa-2,9,11,15,20,21,26-heptaazahexacyclo[24.3.1.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]triacontene-3,5,7,9,13,17(31),18(22),19-octaene-12,16-dione (INT-A10-9) (80%) as a brown solid. LCMS: m/z (ESI), [M+H] +=566.10. Step 10. Preparation of 1,15,21-trimethyl-5-(morpholin-4-yl)-23-oxo-2,9,11,15,20,21,26-heptaazahexacyclo[24.3.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontene-3,5,7,9,13,17(31),18(22),19-octaene-12,16-dione (Example A10). In a nitrogen atmosphere at room temperature, 5-bromo-1,15,21-trimethyl-23-oxa-2,9,11,15,20,21,26-heptaazahexacyclo[24.3.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tri-undecene-3,5,7,9,13,17(31),18(22),19-octaene-12,16-dione (90 mg, 0.16 mmol, 1 equivalent), BrettPhos Pd G 3To a stirred mixture of 2-nitropropene (43 mg, 0.05 mmol, 0.3 equiv.) and morpholine (55 mg, 0.63 mmol, 4 equiv.) in dioxane (4 mL) was added LiHMDS (0.95 mL, 0.95 mmol, 6.0 equiv.). The mixture was stirred at 60 °C for 1 h, cooled to room temperature, treated with water (30 mL), and extracted with ethyl acetate (3×30 mL). The combined organic layers were washed with brine (3×30 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was subjected to prep-HPLC using an XBridge Shield RP18 OBD column with 10 mmol/L NH4HCO 3+ 0.1%NH 3 .H 2O was used for purification with water and acetonitrile as the mobile phase. The fractions containing the desired compound were evaporated to dryness to give 12 mg of 1,15,21-trimethyl-5-(morpholin-4-yl)-23-oxa-2,9,11,15,20,21,26-heptaazahexacyclo[24.3.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tri-undecene-3,5,7,9,13,17(31),18(22),19-octaene-12,16-dione (Example A10) as a white solid (13%). LCMS: m/z (ESI), [M+H] +=573.50. 1H NMR (DMSO-d 6, 400 MHz) δ 1.89-1.85 (5H, m), 2.12-2.08 (1H, m), 2.30 (1H, s), 2.72-2.69 (1H, m), 2.92 (1H, d), 3.08-3.06 (3H, m), 3.29 (2H, s), 3.62 (3H, s), 3.72 (2H, s), 3.76-3.74 (3H, m), 4.62 (1H, t), 5.42-5.40 (1H, d), 6.93-6.90 (1H, m), 7.32 (1H, d), 7.48 (1H, d), 8.21-8.37 (1H, m), 8.74 (1H, d), 12.80 (1H, s). Example A11 5,13,27-Trimethyl-17-(morpholin-4-yl)-7-oxa-4,5,10,14,21,23,27-heptaazahexacyclo[23.3.1.1^{10,13}.0^{2,6}.0^{14,22}.0^{15, 20}]triazine-1(29),2(6),3,15,17,19,21,25-octaene-24,28-dione Step 1. Preparation of tert-butyl 3-[(5-bromo-2-nitrophenyl)amino]-3-methylpyrrolidine-1-carboxylate (INT-A11-1). 4-bromo-2-fluoro-1-nitrobenzene (2 g, 9.1 mmol, 3 equivalents), K 2CO 3A mixture of tert-butyl 3-amino-3-methylpyrrolidine-1-carboxylate (1.26 g, 9.09 mmol, 3 equiv.) and tert-butyl 3-amino-3-methylpyrrolidine-1-carboxylate (0.61 g, 3.03 mmol, 1 equiv.) in acetonitrile (20 mL) was stirred at 100 °C for 2 h under nitrogen atmosphere and cooled to room temperature. The reaction was quenched with water (200 mL), and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using petroleum ether/ethyl acetate (4/1) to give 1.2 g of tert-butyl 3-[(5-bromo-2-nitrophenyl)amino]-3-methylpyrrolidine-1-carboxylate (INT-A11-1) (80%) as a yellow solid. LCMS: m/z (ES+), [M+H - 56] +=344.00. Step 2. N-Preparation of (5-bromo-2-nitrophenyl)-3-methylpyrrolidin-3-amine (INT-A11-2). A mixture of tert-butyl 3-[(5-bromo-2-nitrophenyl)amino]-3-methylpyrrolidine-1-carboxylate (2.1 g, 5.25 mmol, 1 equiv) in trifluoroacetic acid (4 mL) and dichloromethane (16 mL) was stirred at room temperature under nitrogen atmosphere for 2 h. The mixture was concentrated under reduced pressure and treated with saturated NaHCO3(aqueous solution) solution (100 mL). The mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (10/1) to give 1.3 g of yellow solid.N-(5-bromo-2-nitrophenyl)-3-methylpyrrolidin-3-amine (INT-A11-2) (74%). LCMS: m/z (ES+), [M+H] +=300.20. Step 3. NPreparation of -(5-bromo-2-nitrophenyl)-1-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-3-methylpyrrolidin-3-amine (INT-A11-3). In a nitrogen atmosphere at room temperature to NTo a stirred solution of -(5-bromo-2-nitrophenyl)-3-methylpyrrolidin-3-amine (1.2 g, 4 mmol, 1 eq.) and 2-[(tert-butyldimethylsilyl)oxy]acetaldehyde (2.8 g, 16 mmol, 4 eq.) in dichloromethane (10 mL) were added trimethylamine (1.62 g, 16 mmol, 4 eq.) and NaBH(OAc) 3(1.69 g, 8 mmol, 2 equiv.). The resulting mixture was stirred at 60 °C overnight and cooled to room temperature. The reaction was quenched with water (10 mL), and the mixture was extracted with ethyl acetate (3×10 mL). The combined organic layers were washed with brine (3×10 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was subjected to prep-HPLC using an XBridge Shield RP18 OBD column with 10 mmol/L NH4HCO 3and 0.1% NH 3 .H 2O was purified using water and acetonitrile as the mobile phase. The fractions containing the desired compound were evaporated to dryness to give 1.24 g of yellow oil.N-(5-bromo-2-nitrophenyl)-1-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-3-methylpyrrolidin-3-amine (INT-A11-3) (61%). LCMS: m/z (ES+), [M+H] +=458.50 Step 4. Preparation of 2-{3-[(5-bromo-2-nitrophenyl)amino]-3-methylpyrrolidin-1-yl}ethanol (INT-A11-4).NA mixture of -(5-bromo-2-nitrophenyl)-1-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-3-methylpyrrolidin-3-amine (1.24 g, 2.71 mmol, 1 equiv) and tetrabutylammonium fluoride (1.41 g, 5.41 mmol, 2 equiv) in tetrahydrofuran (12 mL) was stirred at room temperature under nitrogen atmosphere for 2 h. The reaction was heated with saturated NaHCO3(aqueous solution) solution (50 mL) was quenched at room temperature, and the mixture was extracted with dichloromethane (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (10/1) to give 800 mg of 2-{3-[(5-bromo-2-nitrophenyl)amino]-3-methylpyrrolidin-1-yl}ethanol (INT-A11-4) (69%) as a yellow oil. LCMS: m/z (ES+), [M+H] += 344.00. Step 5. Preparation of 5-[5-(2-{3-[(5-bromo-2-nitrophenyl)amino]-3-methylpyrrolidin-1-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A11-5). To a stirred mixture of 2-{3-[(5-bromo-2-nitrophenyl)amino]-3-methylpyrrolidin-1-yl}ethanol (800 mg, 2.32 mmol, 1 eq) and triphenylphosphine (1829 mg, 7 mmol, 3 eq) in tetrahydrofuran (15 mL) was added methyl 5-(5-hydroxy-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (M2, 611 mg, 2.32 mmol, 1.0 eq) and diisopropyl azodicarboxylate (1410 mg, 7 mmol, 3 eq) at room temperature under nitrogen atmosphere. The resulting mixture was stirred at room temperature for 2 h. The reaction was quenched with water (100 mL), and the mixture was extracted with dichloromethane (3×100 mL). The combined organic layers were washed with brine (3×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (10/1) to give 380 mg of 5-[5-(2-{3-[(5-bromo-2-nitrophenyl)amino]-3-methylpyrrolidin-1-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A11-5) (25%) as a yellow oil. LCMS: m/z (ES+), [M+H] +=589.00. Step 6. Preparation of 5-[5-(2-{3-[(2-amino-5-bromophenyl)amino]-3-methylpyrrolidin-1-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A11-6). To a stirred mixture of 5-[5-(2-{3-[(5-bromo-2-nitrophenyl)amino]-3-methylpyrrolidin-1-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (380 mg, 0.65 mmol, 1 eq.) and ranyl nickel (55.23 mg, 0.65 mmol, 1 eq.) in methanol (6 mL) was added dropwise NH 2NH 2 .H 2O (129.09 mg, 2.58 mmol, 4 equiv.). The resulting mixture was stirred at room temperature for 1.5 h. The reaction was quenched with water (30 mL), and the mixture was extracted with dichloromethane (3×50 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by silica gel chromatography using dichloromethane/methanol (25/1) to give 240 mg of 5-[5-(2-{3-[(2-amino-5-bromophenyl)amino]-3-methylpyrrolidin-1-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A11-6) (67%) as a brown solid. LCMS: m/z (ES+), [M+H] +=559.05. Step 7. Preparation of 5-(5-{2-[3-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-methylpyrrolidin-1-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A11-7). To a stirred mixture of 5-[5-(2-{3-[(2-amino-5-bromophenyl)amino]-3-methylpyrrolidin-1-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (193 mg, 0.35 mmol, 1 eq.) in ethanol (10 mL) was added BrCN (40.19 mg, 0.38 mmol, 1.1 eq.) in portions at room temperature. The resulting mixture was stirred at 40 °C for 1 h. The mixture was cooled to room temperature. The reaction was quenched with water (30 mL), and the mixture was extracted with dichloromethane (3×30 mL). The combined organic layers were washed with brine (3×30 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (12/1) to give 140 mg of 5-(5-{2-[3-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-methylpyrrolidin-1-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A11-7) (69%) as a brown solid. LCMS: m/z (ES+), [M+H] +=586.00. 1H NMR (400 MHz, DMSO- d 6) δ 1.24 (1H, s), 1.65 (3H, s), 2.35-2.48 (2H, m), 2.69-2.73 (1H, m), 2.80-2.90 (2H, m), 2.94-2.98 (1H, m), 3.18-3.22 (1H, m), 3.58 (3H, s), 3.75 (6H, d), 4.08-4.15 (2H, m), 6.52 (1H, s), 7.07 (1H, s), 7.43 (1H, s), 7.97 (1H, s), 8.13 (1H, d), 8.44 (1H, d). Step 8. Preparation of 5-(5-{2-[3-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-methylpyrrolidin-1-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A11-8). 5-(5-{2-[3-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-methylpyrrolidin-1-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (170 mg, 0.29 mmol, 1 equivalent) and LiOH .H 2A mixture of O (50 mg, 1.16 mmol, 4 equiv.) in tetrahydrofuran (4 mL) and water (1 mL) was stirred at room temperature overnight under a nitrogen atmosphere and concentrated under reduced pressure. The residue was purified by prep-HPLC using an XBridge Shield RP18 OBD column with 10 mmol/L NH4HCO 3and 0.1% NH 3 .H 2O water and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 90 mg of 5-(5-{2-[3-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-methylpyrrolidin-1-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A11-8) (54%) as a white solid. LCMS: m/z (ES+), [M+H] +=572.35. Step 9. Preparation of 17-bromo-5,13,27-trimethyl-7-oxa-4,5,10,14,21, 23,27-heptaazahexacyclo[23.3.1.1^{10,13}.0^{2,6}.0^{14,22}. 0^{15,20}]triaconta-1(29),2(6),3,15,17,19,21,25-octaene-24,28-dione (INT-A11-9). 5-(5-{2-[3-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-methylpyrrolidin-1-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (90 mg, 0.18 mmol, 1 equivalent), N,N-Diisopropylethylamine (45 mg, 0.35 mmol, 2 equivalents) and N,N,N,N-Tetramethyl- OA mixture of -(7-azabenzotriazol-1-yl)urea hexafluorophosphate (133 mg, 0.35 mmol, 2 equiv.) in dioxane (3 mL) was stirred at room temperature overnight under nitrogen atmosphere. The reaction was quenched with water (30 mL), and the mixture was extracted with dichloromethane (3×30 mL). The combined organic layers were washed with brine (3×30 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (12/1) to give 70 mg of 17-bromo-5,13,27-trimethyl-7-oxa-4,5,10,14,21,23,27-heptaazahexacyclo[23.3.1.1^{10, 13}.0^{2,6}.0^{14,22}.0^{15,20}]triac-1(29),2(6),3,15,17, 19,21,25-octaene-24,28-dione (INT-A11-9) (72%) as a white solid. LCMS: m/z (ES+), [M+H] +=554.00. Step 10. Preparation of 5,13,27-trimethyl-17-(morpholin-4-yl)-7-oxa-4,5, 10,14,21,23,27-heptaazahexacyclo[23.3.1.1^{10,13}.0^{2,6}.0^{14,22}.0^{15,20}]tris-1(29),2(6),3,15,17,19,21,25-octaene-24,28-dione (Example A11). To a stirred mixture of 17-bromo-5,13,27-trimethyl-7-oxa-4,5,10,14,21,23,27-heptaazahexacyclo[23.3.1.1^{10,13}.0^{2,6}.0^{14,22}.0^{15,20}]triaconta-1(29),2(6),3,15,17,19,21,25-octaene-24,28-dione (70 mg, 0.13 mmol, 1 eq), BrettPhos (35 mg, 0.065 mmol, 0.5 eq) and morpholine (35 mg, 0.40 mmol, 3.2 eq) in dioxane (5 mL) was added LiHMDS (2.5 mL, 2.5 mmol, 19 eq) dropwise at room temperature under nitrogen atmosphere. The mixture was stirred at 60 °C for 1 h under nitrogen atmosphere and then cooled to room temperature. The reaction was quenched with water (5 mL), and the mixture was extracted with dichloromethane (3×20 mL). The combined organic layers were washed with brine (3×20 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (10/1) to give 10.8 mg of 5,13,27-trimethyl-17-(morpholin-4-yl)-7-oxa-4,5,10,14,21,23,27-heptaazahexacyclo[23.3.1.1^{10,13}.0^{2,6}.0^{14,22}.0^{15,20}]triazine-1(29),2(6),3,15,17,19, 21,25-octaene-24,28-dione (Example A11) (15%) as an off-white solid. LCMS: m/z (ES+), [M+H] +=559.20. 1H NMR (DMSO- d 6, 400 MHz) δ 1.65 (3H, s), 2.65-2.84 (3H, m), 2.97-3.03 (2H, m), 3.01-3.24 (4H, m), 3.31-3.37 (1H, m), 3.62 (3H, s), 3.75 (3H, s), 3.77-3.83 (5H, m), 4.14-4.20 (1H, m), 4.35-4.40 (1H, m), 5.16 (1H, d), 6.93 (1H, dd), 6.98 (1H, s), 7.45 (1H, d), 8.22 (1H, d), 8.31 (1H, s), 8.89 (1H, d), 12.63 (1H, s). Example A12 15,21,27,27-Tetramethyl-5-(morpholin-4-yl)-23-oxo-2,9,11,15, 20,21,26-heptaazahexacyclo[24.3.1.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]tri-11-3,5,7,9,13,17(31),18(22),19-octaene-12,16-dione Step 1. NPreparation of tert-butyl-(6,6-dimethylpiperidin-3-yl)carbamate (INT-A12-1).NA solution of tert-butyl-(6-oxopiperidin-3-yl)carbamate (4 g, 18.66 mmol, 1 eq.) in THF (50 mL) was treated with zirconium tetrachloride (10.44 g, 44.80 mmol, 2.4 eq.) at 10 °C under nitrogen atmosphere for 0.5 h, followed by the dropwise addition of CH 3MgBr (28.94 g, 242.68 mmol, 13 equiv). The resulting mixture was stirred at room temperature under nitrogen atmosphere for 16 h. The reaction was quenched with water (500 mL), and the mixture was extracted with ethyl acetate (3×400 mL). The combined organic layers were washed with brine (3×400 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue is purified by silica gel chromatography eluting with dichloromethane/methanol (30/1) to give 3.74 g of a yellow solid.N-(6,6-dimethylpiperidin-3-yl)carbamic acid tert-butyl ester (INT-A12-1) (87%). LCMS: m/z (ESI), [M+H] +=229.05. Step 2. NPreparation of tert-butyl-(1-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-6,6-dimethylpiperidin-3-yl)carbamate (INT-A12-2). At room temperature,N-(6,6-dimethylpiperidin-3-yl)carbamic acid tert-butyl ester (INT-A12-1, 234 mg, 1.02 mmol, 1 equivalent) and K 2CO 3(354 mg, 2.56 mmol, 2.5 equiv) was added dropwise to a stirred mixture of (2-bromomethoxy)(tert-butyl)dimethylsilane (294 mg, 1.23 mmol, 1.2 equiv) in DMSO (10 mL). The reaction mixture was stirred at 60 °C for 3 h. After cooling to room temperature, the reaction mixture was treated with water (100 mL) and extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue is purified by silica gel chromatography eluting with petroleum ether/ethyl acetate (10/1) to give 128 mg of yellow oil.N-tert-Butyl (1-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-6,6-dimethylpiperidin-3-yl)carbamate (INT-A12-2) (32%). LCMS: m/z (ESI), [M+H] +=387.15. Step 3. Preparation of 2-(5-amino-2,2-dimethylpiperidin-1-yl)ethanol (INT-A12-3). NA solution of tert-butyl-(1-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-6,6-dimethylpiperidin-3-yl)carbamate (INT-A12-2, 20 g, 50 mmol, 1 equivalent) and trifluoroacetic acid (60 mL) in dichloromethane (200 mL) was stirred at room temperature for 2 h. The resulting mixture was concentrated under reduced pressure. The crude product 2-(5-amino-2,2-dimethylpiperidin-1-yl)ethanol (INT-A12-3) as a yellow oil was directly used in the next step without further purification. LCMS: m/z (ESI), [M+H] +=173.00. Step 4. Preparation of 2-{5-[(5-bromo-2-nitrophenyl)amino]-2,2-dimethylpiperidin-1-yl}ethanol (INT-A12-4). 2-(5-amino-2,2-dimethylpiperidin-1-yl)ethanol (crude product, INT-A12-3), 4-bromo-2-fluoro-1-nitrobenzene (10.33 g, 60 mmol, 1.2 equivalents) and K 2CO 3(20.67 g, 52.24 mmol, 3 equiv.) in acetonitrile (150 mL) was stirred at 60 °C for 2 h. After cooling to room temperature, the reaction was quenched with water (800 mL), and the mixture was extracted with ethyl acetate (3×600 mL). The combined organic layers were washed with brine (3×600 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by silica gel column chromatography using petroleum ether/ethyl acetate (5/1) to give 9.3 g of 2-{5-[(5-bromo-2-nitrophenyl)amino]-2,2-dimethylpiperidin-1-yl}ethanol (INT-A12-4) (41%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=374.05. 1H NMR (DMSO-d 6, 400 MHz) δ 0.99 (3H, s), 1.05 (3H, s), 1.26-1.46 (2H, m), 1.63-1.70 (2H, m), 2.17-2.25 (1H, m), 2.50-2.60 (3H, m), 3.42 (2H, q), 3.86-3.92 (1H, m), 4.32 (1H, t), 6.76-7.82 (1H, m), 7.27 (1H, d), 8.00 (1H, d), 8.60 (1H, d). Step 5. Preparation of methyl 5-[5-(2-{5-[(5-bromo-2-nitrophenyl)amino]-2,2-dimethylpiperidin-1-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylate (INT-A12-5). A mixture of 2-{5-[(5-bromo-2-nitrophenyl)amino]-2,2-dimethylpiperidin-1-yl}ethanol (INT-A12-4, 350 mg, 0.94 mmol, 1 eq), methyl 5-(5-hydroxy-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (M2, 247 mg, 0.94 mmol, 1 eq) and 2-(tributyl-1^[5]-phosphinilidene)acetonitrile (1.13 g, 4.70 mmol, 5 eq) in toluene (8 mL) was stirred at 100 °C for 3 h. After cooling to room temperature, the reaction was quenched with water (100 mL), and the mixture was extracted with ethyl acetate (3 x 100 mL). The combined organic layers were washed with brine (3×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by reverse phase flash chromatography using 10%-50% acetonitrile/water to give 350 mg of 5-[5-(2-{5-[(5-bromo-2-nitrophenyl)amino]-2,2-dimethylpiperidin-1-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A12-5) (60%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=617.20. Step 6. Preparation of 5-[5-(2-{5-[(2-amino-5-bromophenyl)amino]-2,2-dimethylpiperidin-1-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A12-6). A mixture of 5-[5-(2-{5-[(5-bromo-2-nitrophenyl)amino]-2,2-dimethylpiperidin-1-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A12-5, 400 mg, 0.64 mmol, 1 eq.), ranyl nickel (5 mg, 0.15 mmol, 0.5 eq.) and hydrazine (62 mg, 1.94 mmol, 3 eq.) in methanol (10 mL) was stirred at room temperature for 2 h under nitrogen atmosphere. The reaction was quenched with water (100 mL), and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by prep-TLC using dichloromethane/methanol (20/1) to give 350 mg of 5-[5-(2-{5-[(2-amino-5-bromophenyl)amino]-2,2-dimethylpiperidin-1-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A12-6) (91%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=589.35. Step 7. Preparation of 5-(5-{2-[5-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-2,2-dimethylpiperidin-1-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A12-7). A mixture of methyl 5-[5-(2-{5-[(2-amino-5-bromophenyl)amino]-2,2-dimethylpiperidin-1-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylate (INT-A12-6, 350 mg, 0.59 mmol, 1 eq.) and cyanogen bromide (69 mg, 0.65 mmol, 1.1 eq.) in EtOH (10 mL) was stirred at room temperature under nitrogen atmosphere for 3 h. The reaction mixture was treated with saturated NaHCO3solution (100 mL) and extracted with ethyl acetate (3×100 mL). The combined organic layers were purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by prep-TLC using dichloromethane/methanol (20/1) to give 350 mg of 5-(5-{2-[5-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-2,2-dimethylpiperidin-1-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A12-7) (95%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=614.05. Step 8. Preparation of 5-(5-{2-[5-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-2,2-dimethylpiperidin-1-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A12-8). To a stirred solution of 5-(5-{2-[5-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-2,2-dimethylpiperidin-1-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A12-7, 240 mg, 0.39 mmol, 1 eq.) in tetrahydrofuran (4 mL) was added lithium hydroxide (28 mg, 1.17 mmol, 3 eq.) in H 2O (1 mL). The resulting mixture was stirred at room temperature for 2 h. The reaction mixture was concentrated under reduced pressure, and the residue was purified by reverse-phase flash chromatography using 10% to 40% acetonitrile/water elution to give 190 mg of 5-(5-{2-[5-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-2,2-dimethylpiperidin-1-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A12-8) (81%) as a white solid. LCMS: m/z (ESI), [M+H] +=598.30. Step 9. Preparation of 5-bromo-15,21,27,27-tetramethyl-23-oxa-2,9,11,15,20,21,26-heptaazahexacyclo[24.3.1.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]triacontene-3,5,7,9,13,17(31),18(22),19-octaene-12,16-dione (INT-A12-9). 5-(5-{2-[5-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-2,2-dimethylpiperidin-1-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A12-8, 190 mg, 0.31 mmol, 1 equivalent), N,N-Diisopropylethylamine (82 mg, 0.63 mmol, 2 equivalents) and N,N,N,N-Tetramethyl- OA solution of -(7-azabenzotriazol-1-yl)urea hexafluorophosphate (181 mg, 0.47 mmol, 1.5 equiv) in dioxane (5 mL) was stirred at room temperature under nitrogen atmosphere for 2 h. The reaction was quenched with water (100 mL), and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by prep-TLC using dichloromethane/methanol (20/1) to give 100 mg of 5-bromo-15,21,27,27-tetramethyl-23-oxa-2,9,11,15,20,21,26-heptaazahexacyclo[24.3.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontene-3,5,7,9,13,17(31),18(22),19-octaene-12,16-dione (INT-A12-9) (27%) as a white solid. LCMS: m/z (ESI), [M+H] +=580.35. Step 10. Preparation of 15,21,27,27-tetramethyl-5-(morpholin-4-yl)-23-oxo-2,9,11,15,20,21,26-heptaazahexacyclo[24.3.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontene-3,5,7,9,13,17(31),18(22),19-octaene-12,16-dione (Example A12). 5-Bromo-15,21,27,27-tetramethyl-23-oxa-2,9,11,15,20,21,26-heptaazahexacyclo[24.3.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontene-3,5,7,9,13,17(31),18(22),19-octaene-12,16-dione (INT-A12-9, 60 mg, 0.10 mmol, 1 equiv), morpholine (36 mg, 0.41 mmol, 4 equiv) and BrettPhos Pd G 3To a stirred mixture of 46 mg, 0.05 mmol, 0.5 equiv.) in dioxane (3 mL) was added LiHMDS (0.5 mL, 0.51 mmol, 5 equiv.) dropwise. The reaction mixture was stirred at 60 °C under nitrogen atmosphere for 1 h. After cooling to room temperature, the reaction was heated with saturated NH4Cl solution (50 mL), and the mixture was extracted with dichloromethane (3×50 mL). The combined organic layers were washed with brine (3×50 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by prep-TLC using dichloromethane/methanol (18/1). The product was purified by prep-HPLC using an XBridge Shield RP18 OBD column containing 10 mmol/L NH4HCO and 0.1% NH 3 .H 2The mixture was further purified by using water and acetonitrile as the mobile phase. The fractions containing the desired compound were evaporated to dryness to give 8.9 mg of 515,21,27,27-tetramethyl-5-(morpholin-4-yl)-23-oxa-2,9,11,15,20,21,26-heptaazahexacyclo[24.3.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tri-11-3,5,7,9,13,17(31), 18(22),19-octaene-12,16-dione (Example A12) as a white solid (14%). LCMS: m/z (ESI), [M+H] +=587.40. 1H NMR (DMSO-d 6, 400 MHz) δ 1.12 (3H, s), 1.20-1.28 (1H, m), 1.30 (3H, s), 1.61-1.85 (2H, m), 2.17-2.29 (2H, m), 2.42 (1H, t), 3.05-3.18 (4H, m), 3.39-3.60 (3H, m), 3.62 (3H, s), 3.69 (3H, s), 3.78 (4H, t), 4.48-4.69 (2H, m), 6.87-6.94 (1H, m), 7.18 (1H, s), 7.43 (1H, d), 8.29 (1H, s), 8.37 (1H, s), 8.80 (1H, d), 11.81-12.75 (1H, m) Example A13 1,15,21-Trimethyl-5-(morpholin-4-yl)-23-oxo-2,9,11,15,20, 21,26-heptaazahexacyclo[24.4.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18, 22}]triacontidine-3,5,7,9,13,17(32),18(22),19-octaene-12,16-dione Step 1. Preparation of 1-benzyl-3-methylazacycloheptan-3-ol (INT-A13-1). To a stirred mixture of 1-benzylazacycloheptan-3-one (3.0 g, 14.75 mmol, 1 eq.) in tetrahydrofuran (30 mL) was added MeMgBr (10.5 g, 88.54 mmol, 6 eq.) dropwise at -78 °C under nitrogen atmosphere. The mixture was stirred at 60 °C for 2 h. The reaction was heated with saturated NH4Cl(aq) solution (300 mL) was quenched, and the mixture was extracted with dichloromethane (3×300 mL). The combined organic layers were washed with brine (3×300 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure to give 3.1 g of 1-benzyl-3-methylazacycloheptan-3-ol (INT-A13-1) as a brown oil. This material was used in the next reaction without purification. LCMS: m/z (ESI), [M+H] +=220.05. Step 2. N- Preparation of (1-benzyl-3-methylazacycloheptan-3-yl)acetamide (INT-A13-2). To a stirred mixture of 1-benzyl-3-methylazacycloheptan-3-ol (3.1 g, about 14 mmol, about 1 equivalent) in acetonitrile (30 mL) was added dropwise fumed H 2SO 4(2.5 g, 14.13 mmol, 1 eq.). The mixture was stirred for 2 h. The reaction was quenched with water (300 mL) at room temperature. The mixture was treated with K 2CO 3Adjust to pH 9 and extract with dichloromethane (3×300 mL). Wash the combined organic layers with brine (3×300 mL) and precipitate with anhydrous Na 2SO 4Dry and concentrate under reduced pressure to obtain 1.1 g of a brown solid.N-(1-Benzyl-3-methylazacyclohept-3-yl)acetamide (INT-A13-2). 1H NMR (400 MHz, DMSO-d 6) δ 1.18 (3H, s), 1.35-1.67 (5H, m), 1.72 (3H, s), 1.90-2.00 (1H, m), 2.61 (1H, d), 2.87 (1H, d), 3.61 (2H, d), 7.20-7.37 (5H, m). Step 3. N-(3-methylazacycloheptyl-3-yl)acetamide (INT-A13-3). NA mixture of -(1-benzyl-3-methylazacyclohept-3-yl)acetamide (INT-A13-2, 1.1 g, 4.22 mmol, 1 eq.) and Pd/C (0.4 g, 4.22 mmol, 1 eq.) in methanol (10 mL) was stirred at room temperature overnight under nitrogen atmosphere. The mixture was filtered. The filter cake was washed with methanol (3×100 mL). The resulting solution was concentrated under reduced pressure. The crude product was directly used in the next step without further purification. 1H NMR (400 MHz, DMSO-d 6) δ 1.20 (3H, s), 1.32-1.58 (5H, m), 1.78 (3H, s), 1.90-2.10 (1H, m), 2.61 (1H, d), 2.65-2.77 (2H, m), 2.85 (1H, d), 7.23 (1H, s). Step 4. N-(1-(2-((tert-butyldimethylsilyl)oxy)ethyl)-3-methylazinecyclohept-3-yl)acetamide (INT-A13-4). To N-(3-Methylazacyclohept-3-yl)acetamide (400 mg, about 2.3 mmol, 1 equivalent) and (2-bromoethoxy)(tert-butyl)dimethylsilane (1124 mg, 4.69 mmol, 2 equivalents) were added to N,N-Dimethylformamide (8 mL) was added to the stirred mixture.2CO 3(649.4 mg, 4.69 mmol, 2 eq.) and NaI (704 mg, 4.69 mmol, 2 eq.). The mixture was stirred at 60 °C for 2 h and then cooled to room temperature. The reaction was quenched with water (100 mL), and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue is purified by C18FLASH using water and acetonitrile as mobile phase. The fractions containing the desired compound are evaporated to dryness to give 750 mg N-(1-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-3-methylazacyclohept-3-yl)acetamide (INT-A13-4) (96%). 1H NMR (400 MHz, DMSO-d 6) δ 0.01 (6H, s), 0.83 (9H, s), 1.19 (3H, s), 1.28-1.62 (6H, m), 1.73 (3H, s), 1.86 (1H, d), 2.51-2.64 (5H, m), 2.72-2.80 (1H, m), 3.55-3.63 (2H, m). Step 5. Preparation of 2-(3-amino-3-methylazinocycloheptan-1-yl)ethanol (INT-A13-5). NA mixture of 1-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-3-methylazacycloheptan-3-yl)acetamide (1.4 g, 4.26 mmol, 1 eq.) and HCl (6 M) (140 mL, 460.77 mmol, 108 eq.) was stirred at 100 °C overnight under a nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure to give 500 mg of 2-(3-amino-3-methylazacycloheptan-1-yl)ethanol (INT-A13-5) as a brown oil. 1H NMR (400 MHz, DMSO-d 6) δ 1.11-1.39 (3H, m), 1.48 (2H, d), 1.70 (1H, d), 1.76-2.08 (6H, m), 2.19 (1H, t), 3.18 (2H, t), 3.34-3.52 (1H, m), 3.52-3.95 (6H, m), 8.64 (2H, s). Step 6. Preparation of 2-(3-((5-bromo-2-nitrophenyl)amino)-3-methylazinocycloheptan-1-yl)ethanol (INT-A13-6). To a stirred mixture of 2-(3-amino-3-methylazinylcycloheptan-1-yl)ethanol (460 mg, 2.67 mmol, 1 eq) and 4-bromo-2-fluoro-1-nitrobenzene (1174 mg, 5.34 mmol, 2 eq) in acetonitrile (5 mL) was added K 2CO 3(738 mg, 5.34 mmol, 2 eq.). The mixture was stirred at 80 °C overnight and then cooled to room temperature. The reaction was quenched with water (200 mL), and the mixture was extracted with ethyl acetate (3×200 mL). The combined organic layers were washed with brine (3×200 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue is purified by silica gel chromatography using petroleum ether/ethyl acetate (1/10) to give 530 mg of 2-{3-[(5-bromo-2-nitrophenyl)amino]-3-methylazinocycloheptan-1-yl}ethanol (INT-A13-6) (53%) as a yellow oil. 1H NMR (400 MHz, DMSO-d 6) δ 1.31-1.63 (7H, m), 1.64-1.77 (1H, m), 2.21 (1H, d), 2.58-2.86 (6H, m), 3.60 (2H, d), 4.44 (1H, t), 6.81 (1H, d), 7.41 (1H, d), 8.01 (1H, d), 9.07 (1H, s). Step 7. Preparation of 5-[5-(2-{3-[(5-bromo-2-nitrophenyl)amino]-3-methylazinocycloheptan-1-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A13-7). To a stirred mixture of methyl 5-(5-hydroxy-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (M2, 405 mg, 1.53 mmol, 1 eq.) and triphenylphosphine (1210 mg, 4.614 mmol, 3 eq.) in tetrahydrofuran (5 mL) was added diisopropyl azodicarboxylate (933 mg, 4.61 mmol, 3 eq.) and 2-{3-[(5-bromo-2-nitrophenyl)amino]-3-methylazanylcycloheptan-1-yl}ethanol (629 mg, 1.69 mmol, 1.1 eq.) at 0°C under nitrogen atmosphere. The mixture was stirred at room temperature for 2 h. The reaction was quenched with water (100 mL), and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was subjected to Prep-TLC using CH2Cl 2/methanol (10/1) for purification to obtain 550 mg of 5-[5-(2-{3-[(5-bromo-2-nitrophenyl)amino]-3-methylazinocyclohept-1-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A13-7) (57%) as a yellow oil. 1H NMR (400 MHz, DMSO-d 6) δ 1.37 (3H, s), 1.44-1.50 (1H, m), 1.65-1.75 (1H, m), 2.17-2.25 (1H, m), 2.63-2.77 (2H, m), 2.83 (1H, s), 2.91 (1H, d), 3.10 (1H, d), 3.10-3.20 (1H, m), 3.33 (2H, s), 3.71 (6H, d), 4.06-4.22 (2H, m), 5.76 (1H, s), 6.80 (1H, d), 7.20-7.30 (2H, m), 7.33-7.50 (5H, m), 7.94 (1H, s), 7.98 (1H, d), 8.07 (1H, d), 8.40 (1H, d), 9.01 (1H, s). Step 8. Preparation of 5-(5-(2-(3-((2-amino-5-bromophenyl)amino)-3-methylazinocycloheptan-1-yl)ethoxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxo-1,6-dihydropyridine-3-carboxylic acid methyl ester (INT-A13-8). 5-[5-(2-{3-[(5-bromo-2-nitrophenyl)amino]-3-methylazinocyclohept-1-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (540 mg, 0.87 mmol, 1 equivalent), ranyl nickel (74 mg, 0.87 mmol, 1 equivalent) and NH 2NH 2 .H 2A stirred mixture of 2-[4-[(2-amino-5-bromophenyl)amino]-3-methylazinocycloheptan-1-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylate (INT-A13-8) was stirred at 0 °C for 1 h under nitrogen atmosphere. The resulting mixture was filtered. The filter cake was washed with methanol (50 mL). The resulting solution was concentrated under reduced pressure to give 300 mg of 5-[5-(2-{3-[(2-amino-5-bromophenyl)amino]-3-methylazinocycloheptan-1-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylate (INT-A13-8) as a brown oil. This material was used in the next reaction without purification. LCMS: m/z (ESI), [M+H]+=589.20. Step 9. Preparation of methyl 5-(5-{2-[3-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-methylazanol-1-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (INT-A13-9). A mixture of methyl 5-[5-(2-{3-[(2-amino-5-bromophenyl)amino]-3-methylazanol-1-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylate (280 mg, 0.47 mmol, 1 eq) and BrCN (50 mg, 0.47 mmol, 1 eq) in EtOH (4 mL) was stirred at 600° C. for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature and concentrated under reduced pressure. The residue was separated by Prep-TLC using CH2Cl 2/methanol (10/1) for purification to obtain 210 mg of 5-(5-{2-[3-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-methylazinocyclohept-1-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A13-9) (71%) as a brown oil. 1H NMR (400 MHz, DMSO-d 6) δ 1.19-1.23 (1H, m), 1.43-1.49 (1H, m), 1.60-1.64 (2H, m), 1.73-1.90 (5H, m), 2.65-2.81 (1H, m), 2.83-2.90 (1H, m), 2.90-3.05 (2H, m), 3.17 (3H, d), 3.55 (3H, s), 3.66 (3H, s), 3.74 (3H, s), 4.00-4.18 (3H, m), 6.95-7.11 (2H, m), 7.54 (1H, d), 7.70 (2H, s), 7.94 (1H, s), 8.06 (1H, d), 8.37 (1H, d). Step 10. Preparation of 5-(5-{2-[3-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-methylazanyl-1-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A13-10). 5-(5-{2-[3-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-methylazanyl-1-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (200 mg, 0.32 mmol, 1 eq.) and LiOH .H 2O (27 mg, 0.65 mmol, 2 equivalents) in tetrahydrofuran/H 2The mixture in 400 mL/mL was stirred at room temperature under nitrogen atmosphere for 2 h. The mixture was concentrated under reduced pressure. The residue was purified by C18FLASH using water and acetonitrile as mobile phase. The fractions containing the desired compound were evaporated to dryness to give 150 mg of 5-(5-{2-[3-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-methylazinocyclohept-1-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A13-10) (76%) as a brown solid. 1H NMR (400 MHz, DMSO-d 6) δ 1.43-1.68 (2H, m), 1.81 (3H, d), 2.70-2.74 (1H, m), 2.82-3.02 (2H, m), 3.06-3.12 (1H, m), 3.38-3.44 (4H, m), 3.62 (2H, s), 3.97-4.10 (1H, m), 6.94-7.08 (1H, m), 7.54 (1H, d), 7.82 (1H, s), 7.88 (1H, s), 8.02 (1H, s), 8.16 (1H, d). Step 11. Preparation of 5-bromo-1,15,21-trimethyl-23-oxa-2,9,11,15,20, 21,26-heptaazahexacyclo[24.4.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontria-3,5,7,9,13,17(32),18(22),19-octaene-12,16-dione (INT-A13-11). To a stirred mixture of 5-(5-{2-[3-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-methylazacyclohept-1-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (140 mg, 0.23 mmol, 1 eq.) and N,N,N,N-tetramethyl-O-(7-azabenzotriazol-1-yl)uronium hexafluorophosphate (133 mg, 0.35 mmol, 1.5 eq.) in dioxane (4 mL) was added under nitrogen atmosphere.N,N-Diisopropylethylamine (90 mg, 0.70 mmol, 3.0 equiv.). The resulting mixture was stirred at 60 °C for 2 h, cooled to room temperature, and concentrated under reduced pressure. The residue was treated with water (30 mL), and the mixture was extracted with dichloromethane (3×30 mL). The combined organic layers were washed with brine (3×30 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was subjected to Prep-TLC using CH2Cl 2/methanol (12/1) for purification to obtain 100 mg of 5-bromo-1,15,21-trimethyl-23-oxa-2,9,11,15,20,21,26-heptaazahexacyclo[24.4.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontria-3,5,7,9,13,17(32),18(22),19-octaene-12,16-dione (INT-A13-11) (73%) as a brown solid. 1H NMR (400 MHz, DMSO-d 6) δ 1.23-1.29 (7H, m), 1.68-1.72 (2H, m), 1.78-1.86 (5H, m), 1.87-1.91 (1H, m), 2.02-2.13 (1H, m), 2.69 (3H, s), 3.10-3.21 (3H, m), 3.23 (1H, t), 3.41 (1H, d), 3.60-3.66 (4H, m), 3.72 (3H, s), 4.30-4.47 (3H, m), 7.35-7.44 (1H, m), 7.57 (1H, d), 7.69 (1H, d), 8.10 (1H, s), 8.30 (1H, d), 8.62 (1H, d), 13.06 (1H, s). Step 12. Preparation of 1,15,21-trimethyl-5-(morpholin-4-yl)-23-oxo-2,9, 11,15,20,21,26-heptaazahexacyclo[24.4.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontidine-3,5,7,9,13,17(32),18(22),19-octaene-12,16-dione (Example A13). To 5-bromo-1,15,21-trimethyl-23-oxa-2,9,11,15,20,21,26-heptaazahexacyclo[24.4.1.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]triacontrine-3,5,7,9,13,17(32), 18(22),19-octaene-12,16-dione (90 mg, 0.15 mmol, 1 equivalent) and BrettPhos Pd G 3To a stirred mixture of 1,4-dioxane (3 mL) and morpholine (54 mg, 0.62 mmol, 4 equiv.) (42 mg, 0.046 mmol, 0.3 equiv.) was added LiHMDS (0.93 mL, 0.93 mmol, 6 equiv.). The mixture was stirred at 60 °C for 1 h and then cooled to room temperature. The reaction was quenched with water (30 mL), and the mixture was extracted with ethyl acetate (3×30 mL). The combined organic layers were washed with brine (3×30 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was subjected to Prep-TLC using CH2Cl 2/methanol (25/1) for purification to obtain 25.8 mg of 1,15,21-trimethyl-5-(morpholin-4-yl)-23-oxa-2,9,11,15,20,21,26-heptaazahexacyclo[24.4.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontria-3,5,7,9,13,17(32),18(22),19-octaene-12,16-dione (A13) (28%) as a white solid. LCMS: m/z (ESI), [M+H] +=587.40. 1H NMR (400 MHz, DMSO-d 6) δ 1.60-1.75 (3H, m) 1.80-1.88 (4H, s), 1.96-2.06 (1H, m), 2.57-2.67 (1H, m), 2.85-2.93 (1H, m), 2.95-3.09 (3H, m), 3.06-3.15 (2H, m), 3.21-3.29 (2H, m), 3.61 (3H, s), 3.70-3.80 (8H, m), 4.32 (1H, d), 4.39 (1H, t), 4.53 (1H, d), 6.94 (1H, d), 7.04 (1H, d), 7.49 (1H, d), 8.09 (1H, s), 8.26 (1H, d), 8.59 (1H, d), 12.85 (1H, s). Example A14 (11 R)-26-ethyl-5,11-dimethyl-16-(4-methylpiperazine-1-yl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]twenty-eight-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione Step 1. Preparation of 5-bromo-1-ethyl-6-oxo-1,6-dihydropyridine-3-carboxylic acid methyl ester (INT-A14-1). At 0°C, 5-bromo-6-hydroxypyridine-3-carboxylic acid methyl ester (10 g, 43.09 mmol, 1 equivalent) was added to N,NTo the stirred mixture in dimethylformamide (200 mL) was added NaH (3.45 g, 86.19 mmol, 2 eq., 60%). The reaction mixture was stirred at 60 °C for 30 min before the addition of ethyl iodide (10.08 g, 64.64 mmol, 1.5 eq.). The mixture was stirred for an additional 2 h at room temperature, treated with water (1 L), and extracted with ethyl acetate (3×1 L). The combined organic layers were washed with brine (3×1 L), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by silica gel chromatography using petroleum ether/ethyl acetate (3/1) to give 5 g of methyl 5-bromo-1-ethyl-6-oxopyridine-3-carboxylate (INT-A14-1) (37%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=261.90. 1H NMR (400 MHz, DMSO-d 6) δ 1.25 (3H, t), 3.81 (3H, s), 4.08 (2H, q), 8.17 (1H, d), 8.61 (1H, d) Step 2. Preparation of methyl-1-ethyl-5-(2-methyl-3-oxo-1-((2-(trimethylsilyl)ethoxy)methyl)-2,3-dihydro-1H-pyrazol-4-yl)-6-oxo-1,6-dihydropyridine-3-carboxylate (INT-A14-2). 5-Bromo-1-ethyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A14-1, 2.0 g, 7.69 mmol, 1 equivalent), 4,4,5,5-tetramethyl-2-(tetramethyl-1,3,2-dioxaborolane-2-yl)-1,3,2-dioxaborolane (2.93 g, 11.53 mmol, 1.5 equivalent), Pd(dppf)Cl 2A mixture of (0.56 g, 0.77 mmol, 0.1 eq.) and KOAc (2.26 g, 23.07 mmol, 3.0 eq.) in dioxane (25 mL) was stirred at 60 °C for 1 h under nitrogen atmosphere. The mixture was cooled to room temperature, followed by the addition of 4-iodo-2-methyl-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-3-one (2.72 g, 7.69 mmol, 1.0 eq.), Pd(dppf)Cl 2(1.13 g, 1.54 mmol, 0.2 equivalent), K 2CO 3(3.19 g, 23.07 mmol, 3.0 equiv) and H 2O (8 mL). The reaction mixture was stirred at 80 °C under nitrogen atmosphere for 2 h. After cooling to room temperature, the reaction was quenched with water (300 mL), and the mixture was extracted with ethyl acetate (3×300 mL). The combined organic layers were washed with brine (3×300 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by silica gel chromatography using dichloromethane/methanol (12/1) to give 1 g of methyl 1-ethyl-5-(2-methyl-3-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-4-yl)-6-oxopyridine-3-carboxylate (INT-A14-2) (31%) as a brown solid. LCMS: m/z (ESI), [M+H] +=408.15. 1H NMR (400 MHz, DMSO-d 6) δ 0.05 (9H, s), 0.78-0.86 (2H, m), 1.27 (3H, t), 3.37 (3H, s), 3.43-3.50 (2H, m), 3.82 (3H, s), 4.08-4.12 (2H, m), 5.32 (2H, s), 8.41 (1H, d), 8.87 (1H, s), 9.20 (1H, d). Step 3. Preparation of 1-ethyl-5-(5-hydroxy-1-methyl-1H-pyrazol-4-yl)-6-oxo-1,6-dihydropyridine-3-carboxylic acid methyl ester (INT-A14-3). A mixture of methyl 1-ethyl-5-(2-methyl-3-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-4-yl)-6-oxopyridine-3-carboxylate (1.0 g, 2.45 mmol, 1 equiv) and HCl (10 mL, 4 M in 1,4-dioxane) was stirred at room temperature overnight. The mixture was concentrated under vacuum. The residue was washed with CH2Cl 2(20 mL) and saturated NaHCO 3(aqueous solution) (50 mL). The resulting mixture was extracted with dichloromethane (3×100 mL). The combined organic layers were washed with brine (3×50 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by reverse phase flash chromatography using 0% to 100% acetonitrile/water to give 520 mg of methyl 1-ethyl-5-(5-hydroxy-1-methylpyrazol-4-yl)-6-oxopyridine-3-carboxylate (INT-A14-3) (76%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=278.05. 1H NMR (400 MHz, DMSO-d 6) δ 1.29 (3H, t), 3.33 (3H, s), 3.84 (3H, s), 4.13-4.17 (2H, m), 7.96 (1H, s), 8.45 (2H, d), 12.72-13.41 (1H, br s). Step 4. Methyl-( RPreparation of )-5-(5-((5-((5-bromo-2-nitrophenyl)amino)-4-methylpentyl)oxy)-1-methyl-1H-pyrazol-4-yl)-1-ethyl-6-oxopyridine-3-carboxylate (INT-A14-4). To a stirred mixture of 1-ethyl-5-(5-hydroxy-1-methylpyrazol-4-yl)-6-oxopyridine-3-carboxylic acid methyl ester (500 mg, 1.80 mmol, 1 equiv) and triphenylphosphine (1.41 g, 5.41 mmol, 3 equiv) in tetrahydrofuran (8 mL) at 0°C under nitrogen atmosphere were added diisopropyl azodicarboxylate (1.09 g, 5.41 mmol, 3 equiv) and (4 R)-5-[(5-bromo-2-nitrophenyl)amino]-4-methylpentan-1-ol (M3-7, 1.14 g, 3.60 mmol, 2.0 equiv.). The resulting mixture was stirred at 0 °C for 2 h. The reaction was quenched with water (100 mL), and the mixture was extracted with dichloromethane (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by reverse phase flash chromatography using 0% to 100% acetonitrile/water to give 370 mg of methyl-( R)-5-(5-((5-((5-bromo-2-nitrophenyl)amino)-4-methylpentyl)oxy)-1-methyl-1H-pyrazol-4-yl)-1-ethyl-6-oxo-1,6-dihydropyridine-3-carboxylate (INT-A14-4) (35%). LCMS: m/z (ESI), [M+H] +=577.90. 1H NMR (DMSO-d 6, 400 MHz) δ 0.96 (3H, d), 1.27 (3H, t), 1.29-1.45 (1H, m), 1.58-1.64 (1H, m), 1.69-1.96 (3H, m), 3.16-3.27 (1H, m), 3.27-3.38 (1H, m), 3.68 (3H, s), 3.78 (3H, s), 3.96 (2H, t), 4.08 (2H, q), 6.80-6.86 (1H, m), 7.26 (1H, d), 7.95-8.02 (2H, m), 8.13 (1H, d), 8.21 (1H, t), 8.43 (1H, d) Step 5. ( RPreparation of methyl 5-(5-((5-((2-amino-5-bromophenyl)amino)-4-methylpentyl)oxy)-1-methyl-1H-pyrazol-4-yl)-1-ethyl-6-oxo-1,6-dihydropyridine-3-carboxylate (INT-A14-5). Raney nickel (53 mg, 0.62 mmol, 1 equivalent) and methyl-( R)-5-(5-((5-((5-bromo-2-nitrophenyl)amino)-4-methylpentyl)oxy)-1-methyl-1H-pyrazol-4-yl)-1-ethyl-6-oxo-1,6-dihydropyridine-3-carboxylate (INT-A14-4, 360 mg, 0.62 mmol, 1 equivalent) was added to a stirring mixture of methanol (5 mL) with NH 2NH 2 .H 2O (62 mg, 1.25 mmol, 2 eq.). After stirring at 60 °C for 1 h, the resulting mixture was filtered and the organic solution was concentrated under reduced pressure to give 350 mg ( R)-5-(5-((5-((2-amino-5-bromophenyl)amino)-4-methylpentyl)oxy)-1-methyl-1H-pyrazol-4-yl)-1-ethyl-6-oxo-1,6-dihydropyridine-3-carboxylic acid methyl ester (INT-A14-5). This material was used directly in the next reaction without purification. Step 6. Methyl-( RPreparation of methyl 5-(5-((5-(2-amino-6-bromo-1H-benzo[d]imidazol-1-yl)-4-methylpentyl)oxy)-1-methyl-1H-pyrazol-4-yl)-1-ethyl-6-oxo-1,6-dihydropyridine-3-carboxylate (INT-A14-6). Freshly prepared ( R)-5-(5-((5-((2-amino-5-bromophenyl)amino)-4-methylpentyl)oxy)-1-methyl-1H-pyrazol-4-yl)-1-ethyl-6-oxo-1,6-dihydropyridine-3-carboxylate (INT-A14-5, 350 mg) in alcohol (5 mL) was added BrCN (83 mg, 0.79 mmol, 1.5 equiv). After stirring at 60 °C for 1 h under nitrogen atmosphere, the mixture was concentrated under vacuum. The residue was purified by prep-TLC eluting with petroleum ether/ethyl acetate (5/1) to obtain 260 mg of methyl-( R)-5-(5-((5-(2-amino-6-bromo-1H-benzo[d]imidazol-1-yl)-4-methylpentyl)oxy)-1-methyl-1H-pyrazol-4-yl)-1-ethyl-6-oxo-1,6-dihydropyridine-3-carboxylate (INT-A14-6) (69%). LCMS: m/z (ESI), [M+H] +=573.25. 11H NMR (DMSO-d 6, 400 MHz) δ 0.83 (3H, d), 1.26 (3H, t), 1.28-1.40 (1H, m), 1.49-1.53 (1H, m), 1.67-1.71 (1H, m), 1.78-1.94 (1H, m), 1.96-2.02 (1H, m), 3.61 (3H, s), 3.79 (3H, s), 3.80-3.97 (4H, m), 4.08 (2H, q), 6.56 (2H, s), 7.04 (2H, d), 7.36 (1H, d), 7.99 (1H, s), 8.13 (1H, d), 8.45 (1H, d). Step 7. 5-(5-{[(4 RPreparation of 5-(5-{[(4 )-4-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]pentyl]oxy}-1-methylpyrazol-4-yl)-1-ethyl-6-oxopyridine-3-carboxylic acid (INT-A14-7).R)-4-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]pentyl]oxy}-1-methylpyrazol-4-yl)-1-ethyl-6-oxopyridine-3-carboxylic acid methyl ester (260 mg, 0.45 mmol, 1 equivalent) was added to a stirred mixture in THF (2 mL) containing LiOH .H 2O (38 mg, 0.91 mmol, 2.0 equiv) of H 2O (0.5 mL). After stirring at 60 °C for 30 minutes under nitrogen atmosphere. The mixture was cooled to room temperature and concentrated under vacuum. The residue was purified by reverse flash chromatography using 0% to 20% acetonitrile/water elution to give 160 mg 5-(5-{[(4 R)-4-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]pentyl]oxy}-1-methylpyrazol-4-yl)-1-ethyl-6-oxopyridine-3-carboxylic acid (INT-A14-7) (63%). LCMS: m/z (ESI), [M+H] += 559.15. Step 8. (11 R)-16-bromo-26-ethyl-5,11-dimethyl-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]octadecene-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (INT-A14-8) was prepared by heating 5-(5-{[(4 R)-4-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]pentyl]oxy}-1-methylpyrazol-4-yl)-1-ethyl-6-oxopyridine-3-carboxylic acid (160 mg, 0.28 mmol, 1 equivalent) and N,N,N,N-Tetramethyl- O-(7-Azabenzotriazol-1-yl)urea hexafluorophosphate (163 mg, 0.43 mmol, 1.5 equivalents) was added to the stirred mixture in dioxane (2 mL) N,N-dDiisopropylethylamine (111 mg, 0.86 mmol, 3.0 equiv). The reaction mixture was stirred at room temperature for 1 h. The reaction was quenched with water (50 mL), and the mixture was extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by prep-TLC eluting with dichloromethane/methanol (12/1) to give 110 mg (11 R)-16-bromo-26-ethyl-5,11-dimethyl-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]octadecene-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (INT-A14-8) (71%). LCMS: m/z (ESI), [M+H] += 541.05. Step 9. (11 R)-26-ethyl-5,11-dimethyl-16-(4-methylpiperazine-1-yl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]twenty-eight-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (Example A14). In a nitrogen atmosphere, (11 R)-16-bromo-26-ethyl-5,11-dimethyl-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octadecene-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (120 mg, 0.22 mmol, 1 equiv), 1-methylpiperazine (89 mg, 0.89 mmol, 4 equiv) and BrettPhos Pd G 3(60 mg, 0.07 mmol, 0.3 equiv) in dioxane (5 mL) was added dropwise LiHMDS (223 mg, 1.33 mmol, 6 equiv). The reaction mixture was stirred at 60 °C for 30 min. After cooling to room temperature, the reaction was quenched with water (100 mL), and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by prep-TLC eluting with dichloromethane/methanol (15/1) to give 50 mg (11 R)-26-ethyl-5,11-dimethyl-16-(4-methylpiperazin-1-yl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octadecene-1(28),2(6),3, 14,16,18,20,24-octaene-23,27-dione (Example A14) (39%). LCMS: m/z (ESI), [M+H] +=559.35. 1H NMR (400 MHz, DMSO-d 6) δ 0.80-0.86 (4H, m), 1.13-1.34 (4H, m), 1.40-1.46 (2H, m), 1.91 (2H, d), 2.26-2.34 (5H, m), 2.80 (1H, s), 3.16 (4H, t), 3.72 (3H, s), 3.94 (2H, m), 4.05-4.18 (3H, m), 4.35 (1H, t), 6.82-6.88 (1H, m), 7.13 (1H, d), 7.36 (1H, d), 8.26 (1H, d), 8.35 (1H, s), 8.80 (1H, d), 12.35 (1H, s). Example A15 (11 R)-26-cyclopropyl-5,11-dimethyl-16-(4-methylpiperazine-1-yl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]twenty-eight-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione Step 1. Preparation of 5-bromo-1-cyclopropyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A15-1). 5-bromo-6-hydroxypyridine-3-carboxylic acid methyl ester (5 g, 21.55 mmol, 1 equivalent), cyclopropylboronic acid (4.07 g, 47.41 mmol, 2.2 equivalents), Cu(OAc) 2A mixture of (4.31 g, 23.70 mmol, 1.1 equiv) and trimethylamine (4.36 g, 43.10 mmol, 2 equiv) in acetonitrile (100 mL) and ethanol (5 mL) was stirred at 80 °C for 12 h under nitrogen atmosphere. The mixture was cooled to room temperature and filtered. The filter cake was washed with ethyl acetate (3×400 mL), and the resulting solution was concentrated under reduced pressure. The residue was purified on a silica gel column using petroleum ether/ethyl acetate (1/1) to give 350 mg of 5-bromo-1-cyclopropyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A15-1) (30%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=271.90. 1H NMR (DMSO-d 6, 400 MHz) δ 0.90-0.97 (2H, m), 1.01-1.09 (2H, m), 3.38-3.42 (1H, m), 3.80 (3H, s), 8.17 (1H, d), 8.22 (1H, d). Step 2. Preparation of 1-cyclopropyl-5-(2-methyl-3-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-4-yl)-6-oxopyridine-3-carboxylic acid methyl ester (INT-A15-2). 5-Bromo-1-cyclopropyl-6-oxopyridine-3-carboxylic acid methyl ester (400 mg, 1.47 mmol, 1 equivalent) and 4,4,5,5-tetramethyl-2-(tetramethyl-1,3,2-dioxaborolane-2-yl)-1,3,2-dioxaborolane (560 mg, 2.20 mmol, 1.5 equivalent), Pd(dppf)Cl 2CH 2Cl 2A mixture of (239 mg, 0.29 mmol, 0.2 eq) and KOAc (432 mg, 4.41 mmol, 3 eq) in dioxane (8 mL) was stirred at 80 °C for 1 h under nitrogen atmosphere and then cooled to room temperature. To the above mixture were added 4-iodo-2-methyl-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-3-one (520.79 mg, 1.470 mmol, 1 eq), K 2CO 3(406 mg, 2.94 mmol, 2 equiv.) and water (2 mL). The resulting mixture was stirred at 80 °C for another 2 h. After cooling to room temperature, the reaction was quenched with water (100 mL), and the mixture was extracted with dichloromethane (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by prep-TLC using dichloromethane/methanol (1/1) to give 210 mg of 1-methyl-5-(2-cyclopropyl-3-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-4-yl)-6-oxopyridine-3-carboxylic acid methyl ester (INT-A15-2) (34%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=420.15. 1H NMR (DMSO-d 6, 400 MHz) δ 0.06 (9H, s), 0.83 (2H, t), 0.89-0.95 (2H, m), 1.04-1.10 (2H, m), 3.24-3.26 (1H, m), 3.37 (3H, s), 3.45-3.50 (2H, m), 3.81(3H, d), 5.32 (2H, s), 8.06 (1H, d), 8.89 (1H, s), 9.18(1H, d). Step 3. Preparation of 1-cyclopropyl-5-(5-hydroxy-1-methylpyrazol-4-yl)-6-oxopyridine-3-carboxylic acid methyl ester (INT-A15-3). A solution of methyl 1-cyclopropyl-5-(2-methyl-3-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-4-yl)-6-oxopyridine-3-carboxylate (210 mg, 0.50 mmol, 1 eq.) and HCl (5 mL, 4 M in 1,4-dioxane) was stirred at room temperature for 6 h. The resulting mixture was concentrated under reduced pressure. The residue was treated with CH2Cl 2(20 mL) and saturated NaHCO 3(aqueous solution) (50 mL). The mixture was extracted with dichloromethane (3×100 mL). The combined organic layers were washed with brine (3×50 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure to give 190 mg of methyl 1-cyclopropyl-5-(5-hydroxy-1-methylpyrazol-4-yl)-6-oxopyridine-3-carboxylate (INT-A15-3), which was used directly in the next step without further purification. LCMS: m/z (ESI), [M+H] +=290.00. 1H NMR (DMSO-d 6, 400 MHz) δ 0.95-1.00 (2H, m), 1.08-1.12 (2H, m), 3.45-3.49 (1H, m), 3.51 (3H, s), 3.83 (3H, s), 7.46-7.69 (1H, m), 8.03 (1H, s), 8.08 (1H, d), 8.38 (1H, d). Step 4. 5-(5-{[(4 RPreparation of methyl 1-cyclopropyl-5-(5-hydroxy-1-methylpyrazol-4-yl)-6-oxopyridine-3-carboxylate (INT-A15-4). To a solution of methyl 1-cyclopropyl-5-(5-hydroxy-1-methylpyrazol-4-yl)-6-oxopyridine-3-carboxylate (crude, 190 mg, 0.66 mmol, 1 eq) and triphenylphosphine (516 mg, 1.97 mmol, 3 eq) in tetrahydrofuran (10 mL) was added diisopropyl azodicarboxylate (398 mg, 1.97 mmol, 3 eq) and (4 R)-5-[(5-bromo-2-nitrophenyl)amino]-4-methylpentan-1-ol (208 mg, 0.66 mmol, 1 eq.). The resulting mixture was stirred at 0 °C for 3 h. The reaction was quenched with water (100 mL), and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using ethyl acetate to give 135 mg of 5-(5-{[(4 R)-5-[(5-bromo-2-nitrophenyl)amino]-4-methylpentyl]oxy}-1-methylpyrazol-4-yl)-1-cyclopropyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A15-4) (33%). LCMS: m/z (ESI), [M+H] +=589.90. 1H NMR (DMSO-d 6, 400 MHz) δ 0.85-1.00 (5H, m), 1.02-1.12 (2H, m), 1.15-1.30 (2H, m), 1.30-1.52 (1H, m), 1.53-1.96 (3H, m), 3.19-3.23 (1H, m), 3.38-3.42 (1H, m), 3.68 (3H, s), 3.77 (3H, s), 3.95 (2H, t), 6.80-6.86 (1H, m), 7.26 (1H, d), 7.97-8.01 (2H, m), 8.06 (1H, d), 8.11 (1H, d), 8.20 (1H, t) Step 5. 5-(5-{[(4 RPreparation of methyl 5-[(2-amino-5-bromophenyl)amino]-4-methylpentyl]oxy}-1-methylpyrazol-4-yl)-1-cyclopropyl-6-oxopyridine-3-carboxylate (INT-A15-5). Raney nickel (60 mg, 0.70 mmol, 3 equivalents) and 5-(5-{[(4 RTo a mixture of methyl 5-[(5-bromo-2-nitrophenyl)amino]-4-methylpentyl]oxy}-1-methylpyrazol-4-yl)-1-cyclopropyl-6-oxopyridine-3-carboxylate (135 mg, 0.2 mmol, 1 equiv) in methanol (3 mL) was added hydrazine hydrate (23 mg, 0.46 mmol, 2 equiv). The resulting mixture was stirred at room temperature for 2 h. The resulting mixture was filtered. The filter cake was washed with methanol (3×50 mL). The resulting solution was concentrated under reduced pressure. The residue was purified by Prep-TLC eluting with dichloromethane/methanol (16/1) to obtain 85 mg of 5-(5-{[(4 R)-5-[(2-amino-5-bromophenyl)amino]-4-methylpentyl]oxy}-1-methylpyrazol-4-yl)-1-cyclopropyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A15-5) (65%). LCMS: m/z (ESI), [M+H] +=558.05. 1H NMR (DMSO-d 6, 400 MHz) δ 0.80-1.12 (7H, m), 1.20-1.40 (2H, m), 1.52-1.95 (4H, m), 2.70-3.02 (2H, m), 3.68 (3H, s), 3.79 (3H, s), 3.96 (2H, t), 4.59-4.75 (3H, m), 6.41-6.54 (3H, m), 8.01 (1H, s), 8.09 (1H, d), 8.14 (1H, d) Step 6. 5-(5-{[(4 RPreparation of methyl 4-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]pentyl]oxy}-1-methylpyrazol-4-yl)-1-cyclopropyl-6-oxopyridine-3-carboxylate (INT-A15-6). 5-(5-{[(4 RA mixture of methyl 5-[(2-amino-5-bromophenyl)amino]-4-methylpentyl]oxy}-1-methylpyrazol-4-yl)-1-cyclopropyl-6-oxopyridine-3-carboxylate (85 mg, 0.15 mmol, 1 equiv) and cyanogen bromide (24 mg, 0.23 mmol, 1.5 equiv) in ethanol (18 mL) was stirred at room temperature for 3 h. The resulting mixture was concentrated under reduced pressure, and the residue was purified by Prep-TLC eluting with dichloromethane/methanol (12/1) to obtain 60 mg of 5-(5-{[(4 R)-4-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]pentyl]oxy}-1-methylpyrazol-4-yl)-1-cyclopropyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A15-6) (65%). LCMS: m/z (ESI), [M+H] +=585.05. Step 7. 5-(5-{[(4 RPreparation of 5-(5-{[(4 RTo a mixture of methyl 4-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]pentyl]oxy}-1-methylpyrazol-4-yl)-1-cyclopropyl-6-oxopyridine-3-carboxylate (70 mg, 0.12 mmol, 1 eq.) in tetrahydrofuran (4 mL) was added LiOH (11 mg, 0.48 mmol, 4 eq.) in water (1 mL). After stirring at room temperature for 2 h, the mixture was concentrated under reduced pressure. The residue was purified by reverse phase flash chromatography using 10% to 50% acetonitrile/water elution to give 50 mg of 5-(5-{[(4R)-4-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]pentyl]oxy}-1-methylpyrazol-4-yl)-1-cyclopropyl-6-oxopyridine-3-carboxylic acid (INT-A15-7) (73%) as an off-white solid. LCMS: m/z (ESI), [M+H] +=569.25. Step 8. (11 R)-16-bromo-26-cyclopropyl-5,11-dimethyl-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]octadecene-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (INT-A15-8). 5-(5-{[(4 R)-4-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]pentyl]oxy}-1-methylpyrazol-4-yl)-1-cyclopropyl-6-oxopyridine-3-carboxylic acid (60 mg, 0.10 mmol, 1 equivalent), N, N-Diisopropylethylamine (27 mg, 0.21 mmol, 2 equivalents) and N,N,N,N-Tetramethyl- OA mixture of -(7-azabenzotriazol-1-yl)urea hexafluorophosphate (80 mg, 0.21 mmol, 2 equiv.) in 1,4-dioxane (1 mL, 11.80 mmol, 112 equiv.) was stirred at room temperature under nitrogen atmosphere for 2 h. The reaction was quenched with water (50 mL), and the mixture was extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (25/1) to give 27 mg (11 R)-16-bromo-26-cyclopropyl-5,11-dimethyl-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]octadecene-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (INT-A15-8) (46%). LCMS: m/z (ESI), [M+H] +=553.10. Step 9. (11 R)-26-cyclopropyl-5,11-dimethyl-16-(4-methylpiperazine-1-yl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]twenty-eight-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (Example A15). In a nitrogen atmosphere, (11 R)-16-bromo-26-cyclopropyl-5,11-dimethyl-7-oxa-4,5,13, 20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]octadecene-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (20 mg, 0.04 mmol, 1 equiv), 1-methylpiperazine (14.53 mg, 0.144 mmol, 4 equiv) and BrettPhos Pd G 3(9.86 mg, 0.01 mmol, 0.3 eq.) was added dropwise to a stirred mixture of dioxane (1 mL) with LiHMDS (0.14 mL, 0.144 mmol, 4 eq.). The reaction mixture was stirred at 60 °C for 30 min. After cooling to room temperature, the reaction was quenched with water (50 mL), and the mixture was extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by reverse phase flash chromatography eluting with 10% to 50% acetonitrile/water to give 8.6 mg (11 R)-26-cyclopropyl-5,11-dimethyl-16-(4-methylpiperazin-1-yl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octadecene-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (Example A15) (41%). LCMS: m/z (ESI), [M+H] += 571.35. 1H NMR (DMSO-d 6, 400 MHz) δ 0.80 (3H, d), 0.87-0.89 (2H, m), 1.09-1.13 (2H, m), 1.42-1.48 (1H, m), 1.90-1.92 (2H, m), 2.18-2.28 (4H, m), 2.50 (4H, t), 2.78-2.82 (1H, m), 3.16 (4H, t), 3.42-3.46 (1H, m), 3.72 (3H, s), 3.87-3.94 (2H, m), 4.09 (1H, d), 4.33 (1H, t), 6.84-6.88 (1H, m), 7.14 (1H, d), 7.31-7.37 (1H, m), 8.04 (1H, d), 8.35 (1H, s), 8.77 (1H, d), 12.38 (1H, s). Example A16A and Example A16B 11-Ethyl-5,26-dimethyl-16-(4-methylpiperazin-1-yl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]octadecene-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione, isomer 1 (Example A16A) and isomer 2 (Example A16B) Step 1. N, N- Preparation of dibenzylbutyramide (INT-A16-1). To a stirred mixture of butyric acid (10 g, 113.50 mmol, 1 eq.) and dibenzylamine (22.39 g, 113.50 mmol, 1 eq.) in 1,4-dioxane (100 mL) was added under nitrogen atmosphere at room temperature.N,N,N,N-Tetramethyl- O-(7-Azabenzotriazol-1-yl)urea hexafluorophosphate (64.73 g, 170.25 mmol, 1.5 equivalents) and N,N-diisopropylethylamine (44.01 g, 340.50 mmol, 3 equiv.). The resulting mixture was stirred at room temperature overnight under nitrogen atmosphere. The reaction was quenched with water (300 mL), and the mixture was washed with CH2Cl 2(3×300 mL) extraction. The combined organic layers were washed with brine (3×200 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue is purified by passing through a C18 column using water and acetonitrile as mobile phase. The fractions containing the desired compound are evaporated to dryness to give 25 g of N, N-Dibenzylbutyramide (INT-A16-1) (82%). LCMS: m/z (ESI), [M+H] +=268.00. Step 2. N, N- Preparation of dibenzyl-2-ethylpent-4-enamide (INT-A16-2). In a nitrogen atmosphere at -78 °C to N, N-Dibenzylbutyramide (10 g, 37.40 mmol, 1 eq.) was added to a stirred mixture of tetrahydrofuran (100 mL) with LiHMDS (41.14 mL, 41.14 mmol, 1.1 eq.) and allyl bromide (6.79 g, 56.10 mmol, 1.5 eq.). The resulting mixture was stirred at -60 °C for 2 h under a nitrogen atmosphere and warmed to room temperature. The reaction was quenched with water (200 mL), and the mixture was extracted with ethyl acetate (3×200 mL). The combined organic layers were washed with brine (3×200 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue is purified by passing through a C18 column using water and acetonitrile as mobile phase. The fractions containing the desired compound are evaporated to dryness to obtain 8 g of N, N-Dibenzyl-2-ethylpent-4-enamide (INT-A16-2) (69%). LCMS: m/z (ESI), [M+H] += 308.15. 1H NMR (DMSO-d 6, 400 MHz) δ 0.75 (3H, t), 1.30-1.46 (1H, m), 1.46-1.63 (1H, m), 2.02-2.17 (1H, m), 2.21-2.31 (1H, m), 2.63- 2.79 (1H, m), 4.49-4.61 (4H, m), 4.86-5.07 (2H, m), 5.59-5.83 (1H, m), 7.12-7.49 (10H, m). Step 3. N, N- Preparation of dibenzyl-2-ethyl-5-hydroxypentanamide (INT-A16-3).N, NA mixture of 2-dibenzyl-2-ethylpent-4-enamide (6.0 g, 19.51 mmol, 1 eq.) and 9-boraherobicyclo[3.3.1]nonane (5.95 g, 48.79 mmol, 2.5 eq.) in THF (15 mL) was stirred at room temperature under nitrogen atmosphere for 1 h. Then, NaOH (1.17 g, 29.27 mmol, 1.5 eq.) in H 2O (1.50 mL, 83.33 mmol, 4.27 equiv) and H 2O 2(12.12 mL, 156.13 mmol, 8.0 equiv., 30%). The resulting mixture was stirred at room temperature for 2 h. The reaction was heated with saturated Na 2S 2O 3The solution (100 mL) was quenched, and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by passing through a C18 column using water and acetonitrile as mobile phase. The fractions containing the desired compound were evaporated to dryness to give 5.0 g of N, N-Dibenzyl-2-ethyl-5-hydroxypentanamide (INT-A16-3) (79%). LCMS: m/z (ESI), [M+H] +=326.15. 1H NMR (DMSO-d 6, 400 MHz) δ 0.75 (3H, t), 1.30-1.56 (6H, m), 2.60 (1H, t), 3.23-3.31 (2H, m), 4.56 (4H, d), 7.05-7.54 (10H, m). Step 4. Preparation of 4-[(dibenzylamino)methyl]hexan-1-ol (INT-A16-4). Under nitrogen atmosphere at 0 °C to N, N-dibenzyl-2-ethyl-5-hydroxypentanamide (5.0 g, 15.36 mmol, 1 equivalent) in tetrahydrofuran (50 mL) was added with LiAlH 4(1.16 g, 30.73 mmol, 2.0 equiv). The resulting mixture was stirred at 0 °C for 2 h. The reaction was stirred at room temperature with water (1.2 mL) and H containing NaOH (387.12 mg, 9.679 mmol, 0.63 equiv).2O (800 mg). The resulting mixture was filtered. The filter cake was washed with tetrahydrofuran (2×50 mL). The organic solution was concentrated under reduced pressure. The residue was purified by C18 column using water and acetonitrile as mobile phase. The fractions containing the desired compound were evaporated to dryness to give 3.0 g of 4-[(dibenzylamino)methyl]hexan-1-ol (INT-A16-4) (63%) as a colorless oil. LCMS: m/z (ESI), [M+H] +=312.15. Step 5. Preparation of methyl 4-(aminomethyl)hexan-1-ol (INT-A16-5). A mixture of 4-[(dibenzylamino)methyl]hexan-1-ol (3.0 g, 9.63 mmol, 1 eq.) and Pd/C (0.31 g, 2.89 mmol, 0.3 eq.) in MeOH (50 mL) was stirred at 20 °C for 2 h under a hydrogen atmosphere. The mixture was filtered. The filter cake was washed with methanol (300 mL). The solution was concentrated under reduced pressure to give 1.2 g of 4-(aminomethyl)hexan-1-ol (INT-A16-5) as a colorless oil. This material was used in the next reaction without further purification. 1H NMR (DMSO-d 6, 400 MHz) δ 0.82 (3H, t), 1.0-1.44 (7H, m), 2.18-2.48 (2H, m), 3.37 (2H, t). Step 6. Preparation of 4-{[(5-bromo-2-nitrophenyl)amino]methyl}hexan-1-ol (INT-A16-6). 4-(Aminomethyl)hexan-1-ol (140 mg, 1.07 mmol, 1 eq.), 4-bromo-2-fluoro-1-nitrobenzene (234.72 mg, 1.07 mmol, 1.0 eq.) and K 2CO 3(294.91 mg, 2.13 mmol, 2.0 equiv) in acetonitrile (5.0 mL) was stirred at 60 °C overnight under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (50 mL), and the mixture was extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue is purified by C18 column using water and acetonitrile as mobile phase. The fractions containing the desired compound are evaporated to dryness to give 300 mg of 4-{[(5-bromo-2-nitrophenyl)amino]methyl}hexan-1-ol (INT-A16-6) (85%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=330.90. 1H NMR (DMSO-d 6, 400 MHz) δ 0.90 (3H, t), 1.34-1.49 (6H, m), 1.60-1.70 (1H, m), 3.28 (2H, t), 3.39 (2H, d), 4.32-4.43 (1H, m), 6.84 (1H, d), 7.26 (1H, d), 7.99 (1H, d), 8.15 (1H, t) Step 7. Preparation of 5-{5-[(4-{[(5-bromo-2-nitrophenyl)amino]methyl}hexyl)oxy]-1-methylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A16-7). To a stirred mixture of methyl 5-(5-hydroxy-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (M2, 262.29 mg, 0.99 mmol, 1.00 equiv) and triphenylphosphine (784.00 mg, 2.99 mmol, 3 equiv) in tetrahydrofuran (5 mL) under nitrogen atmosphere at 0° C. were added diisopropyl azodicarboxylate (604.41 mg, 2.99 mmol, 3 equiv) and 4-{[(5-bromo-2-nitrophenyl)amino]methyl}hexan-1-ol (330 mg, 0.99 mmol, 1.00 equiv). The resulting mixture was stirred at 0° C. under nitrogen atmosphere for 2 h. The reaction was quenched with water (50 mL) and the mixture was extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×50 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by C18 column using water and acetonitrile as mobile phase to obtain 500 mg of 5-{5-[(4-{[(5-bromo-2-nitrophenyl)amino]methyl}hexyl)oxy]-1-methylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A16-7) (87%) as orange oil. LCMS: m/z (ESI), [M+H] +=577.85. Step 8. Preparation of 5-{5-[(4-{[(2-amino-5-bromophenyl)amino]methyl}hexyl)oxy]-1-methylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A16-8). A mixture of 5-{5-[(4-{[(5-bromo-2-nitrophenyl)amino]methyl}hexyl)oxy]-1-methylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (400 mg, 0.69 mmol, 1 eq.), ranyl nickel (297.25 mg, 3.47 mmol, 5 eq.) and hydrazine hydrate (98%) (52.11 mg, 1.04 mmol, 1.5 eq.) in methanol (5 mL) was stirred at 0 °C for 2 h under nitrogen atmosphere. The resulting mixture was filtered. The filter cake was washed with methanol (3×30 mL). The solution was concentrated under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (10/1) to give 300 mg of 5-{5-[(4-{[(2-amino-5-bromophenyl)amino]methyl}hexyl)oxy]-1-methylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylate (INT-A16-8) (79%) as a brown solid. LCMS: m/z (ESI), [M+H] +=546.00. Step 9. Preparation of 5-[5-({4-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]hexyl}oxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A16-9). 5-{5-[(4-{[(2-amino-5-bromophenyl)amino]methyl}hexyl)oxy]-1-methylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (300 mg, 0.55 mmol, 1 equiv.) and BrCN (69.78 mg, 0.66 mmol, 1.2 equiv.) were mixed in CH2Cl 2The mixture in 4% paraformaldehyde (5 mL) was stirred at room temperature overnight under nitrogen atmosphere and concentrated under reduced pressure. The residue was purified by Prep-TLC using petroleum ether/ethyl acetate (1/3) to give 220 mg of 5-[5-({4-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]hexyl}oxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A16-9) (90%) as a brown solid. LCMS: m/z (ESI), [M+H] +=572.95. Step 10. Preparation of 5-[5-({4-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]hexyl}oxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A16-10). 5-[5-({4-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]hexyl}oxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (170 mg, 0.23 mmol, 1 equivalent) and LiOH .H 2O (24.96 mg, 0.55 mmol, 2.5 equiv.) in tetrahydrofuran (10 mL) and H 2O (2.5 mL) was stirred at 20 ° C for 2 h under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by C18 column using water and acetonitrile as mobile phase. The fractions containing the desired compound were evaporated to dryness to give 150 mg of 5-[5-({4-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]hexyl}oxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A16-10) (87%) as an orange oil. LCMS: m/z (ESI), [M+H] +=559.00. Step 11. Preparation of methyl 16-bromo-11-ethyl-5,26-dimethyl-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]octacosa-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (INT-A16-11). 5-[5-({4-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]hexyl}oxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid (150 mg, 0.27 mmol, 1 equivalent), N,N,N,N-tetramethyl-O-(7-azabenzotriazol-1-yl)uronium hexafluorophosphate (204.63 mg, 0.54 mmol, 2.0 equivalent) andN,N- A mixture of diisopropylethylamine (104.33 mg, 0.81 mmol, 3.0 equiv.) in dioxane (5 mL) was stirred at 60 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (30 mL), and the mixture was extracted with ethyl acetate (3×30 mL). The combined organic layers were washed with brine (3×30 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (5/1) to give 100 mg of 16-bromo-11-ethyl-5,26-dimethyl-7-oxa-4,5,13,20,22,26-hexaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octacosa-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (INT-A16-11) (69%) as a light yellow solid. LCMS: m/z (ESI), [M+H] += 540.85. Step 12. Preparation of methyl 5-{5-[(4-{[(2-amino-5-bromophenyl)amino]methyl}hexyl)oxy]-1-methylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylate (A16). 1-Methylpiperazine (83.56 mg, 0.85 mmol, 5.0 equivalent), 16-bromo-11-ethyl-5,26-dimethyl-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]octadecanoic-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (90 mg, 0.17 mmol, 1.00 equivalent), BrettPhos Pd G 3A mixture of (45.37 mg, 0.05 mmol, 0.3 equiv) and LiHMDS (1.00 mL, 1.00 mmol, 6.0 equiv) in dioxane (5.0 mL) was stirred at 60 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was terminated by adding saturated NH4Cl solution (30 mL) was quenched, and the mixture was extracted with ethyl acetate (3×30 mL). The combined organic layers were washed with brine (3×30 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (8/1) to give 2.0 mg of 11-ethyl-5,26-dimethyl-16-(4-methylpiperazin-1-yl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclo[22.3.1. 0^{2,6}.0^{13,21}.0^{14,19}]octacosa-1(28),2(6),3,14,16, 18,20,24-octaene-23,27-dione (A16) (2%) as a light yellow solid. LCMS: m/z (ESI), [M+H] +=559.50. 1H NMR (CD 3OD, 400 MHz) δ 0.82 (3H, t), 1.17-1.47 (6H, m), 1.74 (2H, s), 2.08 (1H, s), 2.28 (1H, s), 2.71 (1H, d), 2.88 (3H, s), 3.43 (4H, s), 3.68 (3H, s), 3.77 (3H, s), 3.80-3.92 (2H, m), 4.04 (1H, s), 4.30 (1H, d), 4.42 (1H, d), 6.99 (1H, s), 7.01 (1H, s), 7.39 (1H, d), 8.24 (1H, s), 8.40 (1H, s), 8.87 (1H, s) Step 13. Preparation of 11-ethyl-5,26-dimethyl-16-(4-methylpiperazin-1-yl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclo[22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]octadecene-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione, isomer 1 (Example A16A) and isomer 2 (Example A16B). A racemic mixture of 11-ethyl-5,26-dimethyl-16-(4-methylpiperazin-1-yl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclo[22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]octadecano-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (A16, 25 mg, 0.04 mmol) was purified by Prep-chiral-HPLC using a CHIRALPAK IG column (2*25 cm, 5 μm) with tert-butyl methyl ether containing 0.1% diethylamine as mobile phase A and EtOH/CH2Cl 2(1/1) was used as mobile phase B to separate, to obtain 1.8 mg of isomer 1 (Example A16A, 7%) and 2.2 mg of isomer 2 (Example A16B, 9%) as white solids. Isomer 1: LCMS: m/z (ESI), [M+H] +=559.45. 1H NMR (CD 3OD, 400 MHz) δ 0.72-0.91 (3H, m), 1.16-1.45 (2H, m), 1.65-1.80 (2H, m), 2.03-2.12 (1H, m), 2.20-2.36 (1H, m), 2.60-2.74 (1H, m), 2.80 (4H, s), 3.15-3.26 (4H, m), 3.38 (4H, t), 3.56-3.71 (4H, m), 3.80 (3H, d), 4.00-4.06 (1H, m), 4.25-4.31 (1H, m), 4.39-4.46 (1H, m), 6.91 (1H, s), 7.02 (1H, d), 7.37 (1H, s), 8.12 (1H, s), 8.41 (1H, d), 8.83 (1H, s). Chiral-HPLC, Rt=4.108 min. Isomer 2: LCMS: m/z (ESI), [M+H] +=559.35. 1H NMR (CD 3OD, 400 MHz) δ 0.72-0.91 (3H, m), 1.16-1.45 (2H, m), 1.65-1.80 (2H, m), 2.03-2.12 (1H, m), 2.20-2.36 (1H, m), 2.60-2.74 (1H, m), 2.80 (4H, s), 3.15-3.26 (4H, m), 3.38 (4H, t), 3.56-3.71 (4H, m), 3.80 (3H, d), 4.00-4.06 (1H, m), 4.25-4.31 (1H, m), 4.39-4.46 (1H, m), 6.83 (1H, s), 6.98 (1H, d), 7.35 (1H, s), 8.05 (1H, s), 8.41 (1H, d), 8.77 (1H, s). Chiral-HPLC, Rt=5.613 minutes. Example A18 5',26'-Dimethyl-16'-(4-methylpiperazine-1-yl)-7'-oxa-4',5',13',20',22',26'-hexaazaspiro[cyclopropane-1,11'-pentacyclic[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]twenty-eight]-1'(28'), 2'(6'),3',14',16',18',20',24'-octaene-23',27'-dione Step 1. Preparation of 3-(1-(((5-bromo-2-nitrophenyl)amino)methyl)cyclopropyl)propan-1-ol (INT-A18-1). 3-[1-(aminomethyl)cyclopropyl]propan-1-ol (500 mg, 3.87 mmol, 1 equivalent), K 2CO 3A mixture of (1069 mg, 7.74 mmol, 2 eq.) and 4-bromo-2-fluoro-1-nitrobenzene (936 mg, 4.25 mmol, 1.1 eq.) in acetonitrile (5 mL) was stirred at 60 °C for 2 h under a nitrogen atmosphere. The mixture was cooled to room temperature. The resulting mixture was concentrated under vacuum. The residue was loaded on a silica gel column and eluted with petroleum ether/ethyl acetate (5:1) to give 1.0 g of 3-(1-{[(5-bromo-2-nitrophenyl)amino]methyl}cyclopropyl)propan-1-ol (INT-A18-1) (78%) as a colorless oil. 1H NMR (DMSO-d 6, 400 MHz) δ 0.37-0.55 (4H, m), 1.31-1.44 (2H, m), 1.45-1.60 (2H, m), 3.25 (2H, d), 3.36-3.39 (2H, m), 4.45 (1H, s), 6.78-6.89 (1H, m), 7.26 (1H, d), 8.00 (1H, d), 8.08 (1H, t) Step 2. Preparation of methyl 5-(5-(3-(1-(((5-bromo-2-nitrophenyl)amino)methyl)cyclopropyl)propoxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxo-1,6-dihydropyridine-3-carboxylate (INT-A18-2). To a stirred mixture of methyl 5-(5-hydroxy-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (M2, 700 mg, 2.66 mmol, 1 eq) and triphenylphosphine (2092 mg, 7.97 mmol, 3 eq) in tetrahydrofuran (10 mL) was added diisopropyl azodicarboxylate (1613 mg, 7.97 mmol, 3 eq) and 3-(1-{[(5-bromo-2-nitrophenyl)amino]methyl}cyclopropyl)propan-1-ol (875 mg, 2.66 mmol, 1 eq) at 0°C under nitrogen atmosphere. The mixture was stirred at room temperature for 2 h. The reaction was quenched with water (80 mL) and the mixture was extracted with ethyl acetate (3×60 mL). The combined organic layers were washed with brine (3×50 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was loaded on a silica gel column and purified using petroleum ether/ethyl acetate (1/10) to give 700 mg of 5-{5-[3-(1-{[(5-bromo-2-nitrophenyl)amino]methyl}cyclopropyl)propoxy]-1-methylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A18-2) (45%) as a yellow oil. 1H NMR (DMSO-d 6, 400 MHz) δ 0.42-0.47 (m, 2H), 0.52 (2H, d), 1.50-1.58 (m, 2H), 1.80-1.88 (2H, m), 3.20-3.29 (2H, m), 3.57 (3H, s), 3.66 (3H, s), 3.77 (3H, s), 3.88-3.96 (2H, m), 6.79-6.84 (1H, m), 7.25 (1H, d), 7.55-7.66 (1H, m), 7.98 (1H, d), 8.07 (1H, d), 8.10 (1H, d), 8.42 (1H, d). Step 3. Preparation of 5-(5-(3-(1-(((2-amino-5-bromophenyl)amino)methyl)cyclopropyl)propoxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxo-1,6-dihydropyridine-3-carboxylic acid methyl ester (INT-A18-3). To a stirred mixture of 5-{5-[3-(1-{[(5-bromo-2-nitrophenyl)amino]methyl}cyclopropyl)propoxy]-1-methylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A18-2, 700 mg, 1.22 mmol, 1 equiv.) and ranyl nickel (50 mg) in methanol (10 mL) was added portionwise with NH 2NH 2 .H 2O (122 mg, 2.43 mmol, 2 eq). The mixture was stirred at 0 °C for 1 h. The resulting mixture was filtered. The filter cake was washed with methanol (3×30 mL), and the solution was concentrated under vacuum. The residue was used directly in the next step. Step 4. Preparation of 5-(5-(3-(1-((2-amino-6-bromo-1H-benzo[d]imidazol-1-yl)methyl)cyclopropyl)propoxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxo-1,6-dihydropyridine-3-carboxylic acid methyl ester (INT-A18-4). A mixture of methyl 5-{5-[3-(1-{[(2-amino-5-bromophenyl)amino]methyl}cyclopropyl)propoxy]-1-methylpyrazol-4-yl}-1-methyl-6-oxopyridine-3-carboxylate (580 mg, 1.06 mmol, 1 eq) and BrCN (135 mg, 1.27 mmol, 1.2 eq) in ethanol (6 mL) was stirred under nitrogen atmosphere at room temperature for 2 h. The mixture was concentrated under vacuum. The residue was loaded on a silica gel column and purified using methylene chloride/methanol (12/1) elution to give 600 mg of 5-[5-(3-{1-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]cyclopropyl}propoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A18-4) (98%) as a brown solid. 1H NMR (DMSO-d 6, 400 MHz) δ 0.36 (2H, t), 0.45-0.54 (2H, m), 1.40-1.50 (2H, m), 1.78-1.88 (2H, m), 3.58 (3H, s), 3.65 (3H, s), 3.74 (3H, s), 3.88 (2H, t), 4.08 (2H, s), 6.65 (2H, s), 7.06 (2H, s), 7.35 (1H, s), 7.97 (1H, s), 8.08 (1H, d), 8.43 (1H, d) Step 5. Preparation of 5-(5-(3-(1-((2-amino-6-bromo-1H-benzo[d]imidazol-1-yl)methyl)cyclopropyl)propoxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxo-1,6-dihydropyridine-3-carboxylic acid (INT-A18-5). 5-[5-(3-{1-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]cyclopropyl}propoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (580 mg, 1.02 mmol, 1 equivalent) and LiOH .H 2O (85 mg, 2.03 mmol, 2 equivalents) in tetrahydrofuran/H 2O (0.8 mL/0.2 mL) was stirred at room temperature for 2 h under nitrogen atmosphere. The mixture was concentrated under vacuum. The residue was purified by C18 column using water and acetonitrile as mobile phase. The fractions containing the desired compound were evaporated to dryness to give 400 mg of 5-[5-(3-{1-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]cyclopropyl}propoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A18-5) (70%) as a brown solid. 1H NMR (DMSO-d 6, 400 MHz) δ 0.24-0.35 (2H, m), 0.42 (2H, t), 1.40-1.59 (2H, m), 1.89-1.96 (2H, m), 3.51 (3H, s), 3.63 (3H, s), 3.90 (2H, t), 4.17 (2H, s), 6.94 (2H, s), 7.02 (2H, d), 7.33-7.37 (1H, m),7.94(1H, s), 8.07 (1H, s), 8.38 (1H, d). Step 6. Preparation of 16'-bromo-5',26'-dimethyl-7'-oxa-4',5',13',20',22', 26'-hexaazaspiro[cyclopropane-1,11'-pentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octadeca]-1'(28'),2'(6'),3',14',16',18',20',24'-octaene-23',27'-dione (INT-A18-6). Under nitrogen atmosphere at room temperature, 5-[5-(3-{1-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]cyclopropyl}propoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid (330 mg, 0.59 mmol, 1 equivalent) andN,N,N,N-Tetramethyl-O-(7-azabenzotriazol-1-yl)urea hexafluorophosphate (338 mg, 0.89 mmol, 1.5 equivalents) was added to a mixture of dioxane (5 mL)N,N-Diisopropylethylamine (230 mg, 1.78 mmol, 3 equiv.). The mixture was stirred at the temperature for 3 h. The reaction was quenched with water (80 mL), and the mixture was extracted with ethyl acetate (3×60 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC and eluted with methylene dichloride/methanol (12/1) to give 300 mg of 16'-bromo-5',26'-dimethyl-7'-oxa-4',5',13',20',22',26'-hexaazaspiro[cyclopropane-1,11'-pentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octadeca]-1'(28'), 2'(6'),3',14',16',18',20',24'-octaene-23',27'-dione (INT-A18-6) (93%) as a brown solid. 1H NMR (DMSO-d 6, 400 MHz) δ 0.39 (2H, t), 0.63 (2H, t), 1.78 (2H, s), 2.20-2.28 (2H, m), 3.10-3.18 (2H, m), 3.65 (3H, d), 3.73 (3H, s), 4.15-4.28 (2H, m), 7.34-7.51 (2H, m), 7.75 (1H, d), 8.24-8.36 (2H, m), 8.71 (1H, d), 12.82 (1H, s) Step 7. Preparation of 5',26'-dimethyl-16'-(4-methylpiperazin-1-yl)-7'-oxa-4',5',13',20',22',26'-hexaazaspiro[cyclopropane-1,11'-pentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octaconic]-1'(28'), 2'(6'),3',14',16',18',20',24'-octaene-23',27'-dione (Example A18). 16'-Bromo-5',26'-dimethyl-7'-oxa-4',5',13',20',22',26'-hexaazaspiro[cyclopropane-1,11'-pentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octadecene]-1'(28'), 2'(6'),3',14',16',18',20',24'-octaene-23',27'-dione (100 mg, 0.18 mmol, 1 eq.), 1-methylpiperazine (74 mg, 0.74 mmol, 4.0 eq.) and BrettPhos Pd G 3To a stirred mixture of 1,4-dioxane (4 mL) was added LiHMDS (1.12 mL, 1.12 mmol, 6 eq.). The mixture was stirred at 60 °C for 1 h and then cooled to room temperature. The reaction was heated with saturated NH4Cl(aq) solution (20 mL) was quenched, and the mixture was extracted with ethyl acetate (3×20 mL). The combined organic layers were washed with brine (3×30 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC and eluted with methylene dichloride/methanol (12/1) to give 18 mg of 5',26'-dimethyl-16'-(4-methylpiperazin-1-yl)-7'-oxa-4',5',13',20',22',26'-hexaazaspiro[cyclopropane-1,11'-pentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octaconic]-1'(28'),2'(6'),3',14',16',18',20',24'-octaene-23',27'-dione (Example A18) as a white solid (17%). LCMS: m/z (ESI), [M+H] +=557.35. 1H NMR (DMSO-d 6, 400 MHz) δ 0.35 (2H, t), 0.62 (2H, t), 1.73-1.81 (2H, m), 2.24 (5H, s), 2.45-2.49 (4H, m), 3.10-3.17 (4H, m), 3.62 (3H, s), 3.72 (3H, s), 4.18-4.29 (4H, m), 6.75-6.89 (1H, m), 6.95 (1H, d), 7.36 (1H, d), 8.29 (2H, d), 8.72 (1H, d), 12.51 (1H, s). Example A19 5,12,26-Trimethyl-16-(4-methylpiperazin-1-yl)-7-oxa-4,5, 13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]octadecene-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione Step 1. Preparation of 5-aminohexan-1-ol (INT-A19-1). 6-Hydroxy-hexan-2-one (1.0 g, 8.60 mmol, 1 equivalent), NH4OAc (3.32 g, 43.04 mmol, 5.0 equiv) and NaBH 3A mixture of CN (1.35 g, 21.52 mmol, 2.5 equiv.) in methanol (10 mL) was stirred at 80 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (200 mL), and the mixture was extracted with ethyl acetate (3×200 mL). The combined organic layers were washed with brine (3×200 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. Pass the residue through C18FLASH using 10 mmol/L NH4HCO 3and 0.1% NH 3 .H 2Purification was performed using water with 0.04% O and acetonitrile as mobile phase. The fractions containing the desired compound were evaporated to dryness to give 2.5 g of 5-aminohexan-1-ol (INT-A19-1) (74%) as a colorless oil. LCMS: m/z (ESI), [M+H] +=118.15. Step 2. Preparation of 5-((5-bromo-2-nitrophenyl)amino)hexan-1-ol (INT-A19-2). 5-aminohexan-1-ol (2.5 g, 21.33 mmol, 1 equivalent), K 2CO 3A mixture of (8.84 g, 63.99 mmol, 3 equiv.) and 4-bromo-2-fluoro-1-nitrobenzene (14.08 g, 63.99 mmol, 3 equiv.) in acetonitrile (80 mL) was stirred at 60 °C overnight under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (100 mL), and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column using petroleum ether/ethyl acetate (5/1) to give 3.1 g of 5-[(5-bromo-2-nitrophenyl)amino]hexan-1-ol (INT-A19-2) (42%) as a yellow oil. LCMS: m/z (ESI), [M+H] +=319.05. 1H NMR (DMSO-d 6, 400 MHz) δ 1.18-1.26 (3H, m), 1.29-1.49 (4H, m), 1.50-1.68 (2H, m), 3.36-3.43 (2H, m), 3.81-3.90 (1H, m), 4.37 (1H, t), 6.78-6.86 (1H, m), 7.31 (1H, d), 7.94 (1H, d), 7.99 (1H, d). Step 3. Preparation of 5-(5-((5-((5-bromo-2-nitrophenyl)amino)hexyl)oxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxo-1,6-dihydropyridine-3-carboxylate (INT-A19-3). To a stirred mixture of methyl 5-(5-hydroxy-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (M2, 500 mg, 1.89 mmol, 1.00 equiv) and triphenylphosphine (1494 mg, 5.69 mmol, 3 equiv) in tetrahydrofuran (20 mL) was added diisopropyl azodicarboxylate (1152 mg, 5.69 mmol, 3 equiv) and 5-[(5-bromo-2-nitrophenyl)amino]hexan-1-ol (662 mg, 2.08 mmol, 1.1 equiv) in tetrahydrofuran (10 mL) at 0° C. under nitrogen atmosphere. The mixture was stirred at 0° C. for 2 h. The reaction was quenched with water (50 mL), and the mixture was extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column using petroleum ether/ethyl acetate (1/1) to give 510 mg of 5-[5-({5-[(5-bromo-2-nitrophenyl)amino]hexyl}oxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A19-3) (47%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=564.05. Step 4. Preparation of 5-(5-((5-((2-amino-5-bromophenyl)amino)hexyl)oxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxo-1,6-dihydropyridine-3-carboxylic acid methyl ester (INT-A19-4). To a stirred mixture of 5-[5-({5-[(5-bromo-2-nitrophenyl)amino]hexyl}oxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (490 mg, 0.87 mmol, 1 eq.) and ranyl nickel (599 mg, 6.96 mmol, 8 eq.) in methanol (5 mL) at 0°C was added hydrazine (87.23 mg, 1.742 mmol, 2.0 eq.). The mixture was stirred at 0 °C for 2 h under nitrogen atmosphere. The mixture was filtered. The filter cake was washed with methanol (3×50 mL), and the resulting solution was concentrated under reduced pressure to give 400 mg of 5-[5-({5-[(2-amino-5-bromophenyl)amino]hexyl}oxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A19-4) as a brown solid. This material was used in the next reaction without further purification. LCMS: m/z (ESI), [M+H] +=534.20. Step 5. Preparation of 5-(5-((5-(2-amino-6-bromo-1H-benzo[d]imidazol-1-yl)hexyl)oxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxo-1,6-dihydropyridine-3-carboxylic acid methyl ester (INT-A19-5). A mixture of 5-[5-({5-[(2-amino-5-bromophenyl)amino]hexyl}oxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (400 mg, 0.751 mmol, 1 eq.) and BrCN (95.49 mg, 0.901 mmol, 1.2 eq.) in ethanol (5 mL) was stirred at room temperature overnight under nitrogen atmosphere. The reaction was quenched with water (20 mL), and the mixture was extracted with dichloromethane (3×30 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on Prep-TLC using dichloromethane/methanol (12/1) to give 320 mg of 5-(5-{[5-(2-amino-6-bromo-1,3-benzodiazol-1-yl)hexyl]oxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A19-5) (76%) as a brown solid. LCMS: m/z (ESI), [M+H] +=557.20. Step 6. Preparation of 5-(5-((5-(2-amino-6-bromo-1H-benzo[d]imidazol-1-yl)hexyl)oxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxo-1,6-dihydropyridine-3-carboxylic acid (INT-A19-6). 5-(5-{[5-(2-amino-6-bromo-1,3-benzodiazol-1-yl)hexyl]oxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (300 mg, 0.54 mmol, 1 equivalent) and LiOH.H 2A mixture of O (67 mg, 1.61 mmol, 3.0 equiv.) in tetrahydrofuran (4 mL) and water (1 mL) was stirred at room temperature under nitrogen atmosphere for 2 h. The mixture was concentrated under reduced pressure. The residue was passed through C18FLASH using a 10 mmol/L NH4HCO 3and 0.1% NH 3.H 2O and acetonitrile as the mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 260 mg of 5-(5-{[5-(2-amino-6-bromo-1,3-benzodiazol-1-yl)hexyl]oxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A19-6) (88%) as a brown solid. LCMS: m/z (ESI), [M+H] +=545.20. 1H NMR (DMSO-d 6, 400 MHz) δ 1.37-1.39 (3H, m), 1.60-1.84 (1H, m), 1.90-1.97 (1H, m), 3.51 (3H, s), 3.61 (3H, s), 3.80-3.83 (1H, m), 3.89-3.92 (1H, m), 4.69-4.73 (1H, m), 7.02-7.04 (2H, m), 7.11 (2H, s), 7.37 (1H, t), 8.01 (2H, d), 8.34 (1H, d) Step 7. Preparation of 16-bromo-5,12,26-trimethyl-7-oxa-4,5,13,20,22,26-hexaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octadecene-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (INT-A19-7). 5-(5-{[5-(2-amino-6-bromo-1,3-benzodiazol-1-yl)hexyl]oxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (250 mg, 0.46 mmol, 1 equivalent), N, N, N, N-Tetramethyl- O-(7-Azabenzotriazol-1-yl)urea hexafluorophosphate (262 mg, 0.69 mmol, 1.5 equivalents) and N, N- A mixture of diisopropylethylamine (178 mg, 1.38 mmol, 3.0 equiv.) in 1,4-dioxane (5 mL) was stirred at room temperature overnight under nitrogen atmosphere. The reaction was quenched with water (20 mL), and the mixture was extracted with dichloromethane (3×30 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on Prep-TLC using dichloromethane/methanol (12/1) to give 170 mg of 16-bromo-5,12,26-trimethyl-7-oxa-4,5,13,20,22,26-hexaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octacosa-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (INT-A19-7) (70%) as a brown solid. LCMS: m/z (ESI), [M+H] +=525.15. Step 8. Preparation of 5,12,26-trimethyl-16-(4-methylpiperazin-1-yl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octacosa-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (Example A19). To a stirred mixture of 16-bromo-5,12,26-trimethyl-7-oxa-4,5,13,20,22,26-hexaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octacosa-1(28),2(6),3, 14,16,18,20,24-octaene-23,27-dione (100 mg, 0.19 mmol, 1 eq), 1-methylpiperazine (76 mg, 0.76 mmol, 4 eq) and BrettPhos Pd G3 (51 mg, 0.06 mmol, 0.3 eq) in 1,4-dioxane (4 mL) was added LiHMDS (4 mL, 1.14 mmol, 6 eq) at room temperature under nitrogen atmosphere. The mixture was stirred at 60 °C for 1 h and cooled to room temperature. The reaction was heated with saturated NH4Cl(aq) solution (10 mL) was quenched and the mixture was washed with CH2Cl 2(3×30 mL) extraction. The combined organic layers were purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was passed through prep-HPLC using an XBridge Shield RP18 OBD column using water 10 mmol/L NH4HCO 3and 0.1% NH 3.H 2O and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 11 mg of 5,12,26-trimethyl-16-(4-methylpiperazin-1-yl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octacosa-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (Example A19) (10%) as a white solid. LCMS: m/z (ESI), [M+H] += 545.25. 1H NMR (DMSO-d 6, 400 MHz) δ 1.54-1.62 (4H, m), 1.63-1.70 (1H, m), 1.73-1.79 (1H, m), 1.82-1.92 (1H, m), 2.12-2.20 (2H, m), 2.22-2.31 (1H, m), 2.38-2.45 (4H, m), 3.12-3.27 (4H, m), 3.31 (3H, s), 3.61 (3H, s), 3.71 (3H, s), 3.90-4.08 (1H, m), 4.23-4.27 (1H, m), 4.82-4.98 (1H, m), 6.87-6.86 (1H, m), 7.09 (1H, s), 7.35-7.32 (1H, m), 8.23-8.31 (2H, m), 8.89-8.86 (1H, m), 12.29 (1H, s). Example A20 (11 R)-11,26-dimethyl-16-(4-methylpiperazine-1-yl)-5-(2,2,2-trifluoroethyl)-7-oxo-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]twenty-eight-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione Step 1. Preparation of tert-butyl 4-(3-chloro-4-nitrophenyl)piperazine-1-carboxylate (INT-A20-1). 2-Chloro-4-fluoro-1-nitrobenzene (10 g, 56.967 mmol, 1 equivalent), K 2CO 3A mixture of tert-butyl piperazine-1-carboxylate (15.75 g, 113.934 mmol, 2 equiv.) and tert-butyl piperazine-1-carboxylate (11.67 g, 62.664 mmol, 1.1 equiv.) in acetonitrile (200 mL) was stirred at 60 °C overnight under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (800 mL), and the mixture was extracted with ethyl acetate (3×600 mL). The combined organic layers were washed with brine (3×500 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by silica gel chromatography using petroleum ether/ethyl acetate (1/1) to give 5.4 g of tert-butyl 4-(3-chloro-4-nitrophenyl)piperazine-1-carboxylate (INT-A20-1) (27%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=342.10. Step 2. ( RPreparation of tert-butyl 4-(3-((5-hydroxy-2-methylpentyl)amino)-4-nitrophenyl)piperazine-1-carboxylate (INT-A20-2). tert-butyl 4-(3-chloro-4-nitrophenyl)piperazine-1-carboxylate (1.4 g, 4.096 mmol, 1 equivalent), K 2CO 3(1132.20 mg, 8.192 mmol, 2 equiv.) and (4 R)-5-amino-4-methylpentan-1-ol (M3-6, 480.03 mg, 4.096 mmol, 1 equivalent) in N,N-The mixture in dimethylacetamide (15 mL) was stirred at 100 °C overnight under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (100 mL), and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using petroleum ether/ethyl acetate (1/1) to give 730 mg of tert-butyl 4-(3-{[(2R)-5-hydroxy-2-methylpentyl]amino}-4-nitrophenyl)piperazine-1-carboxylate (INT-A20-2) (42%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=423.25. Step 3. Preparation of 2-(2,2,2-trifluoroethyl)pyrazol-3-ol (INT-A20-3). A mixture of (2E)-3-methoxyprop-2-enoic acid methyl ester (5 g, 43.06 mmol, 1 eq.) and (2,2,2-trifluoroethyl)hydrazine HCl salt (7.37 g, 64.59 mmol, 1.5 eq.) in methanol (20 mL) was stirred at 80 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (200 mL), and the mixture was extracted with dichloromethane (3×200 mL). The combined organic layers were washed with brine (3×200 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by silica gel column eluting with dichloromethane/methanol (20/1) to obtain 6 g of 2-(2,2,2-trifluoroethyl)pyrazol-3-ol (INT-A20-3) (83%) as a yellow solid. 1H NMR (DMSO, 400 MHz) δ 4.71 (2H, q), 5.38 (1H, d), 7.25 (1H, s), 11.40 (1H, s) Step 4. Preparation of 2-(2,2,2-trifluoroethyl)-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-3-one (INT-A20-4). 2-(2,2,2-trifluoroethyl)pyrazol-3-ol (6 g, 36.12 mmol, 1 eq.) and K 2CO 3(9.98 g, 72.24 mmol, 2 eq.) in acetonitrile (100 mL) was added SEM-Cl (12.04 g, 72.24 mmol, 2 eq.). The mixture was stirred at 0 °C for 2 h under nitrogen atmosphere. The reaction was quenched with water (200 mL), and the mixture was extracted with dichloromethane (3×200 mL). The combined organic layers were washed with brine (3×200 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by silica gel column eluting with petroleum ether/ethyl acetate (1/3) to obtain 900 mg of 2-(2,2,2-trifluoroethyl)-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-3-one (INT-A20-4) (8%) as a brown solid. 1H NMR (DMSO, 400 MHz) δ 0.81 (2H, d), 1.06 (1H, t), 1.24 (1H, s), 3.18 (1H, d), 3.45 (2H, m), 4.64 (2H, q), 5.09 (2H, s), 5.44 (1H, d), 8.08 (1H, d) Step 5. Preparation of 4-iodo-2-(2,2,2-trifluoroethyl)-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-3-one (INT-A20-5). To a mixture of 2-(2,2,2-trifluoroethyl)-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-3-one (250 mg, 0.84 mmol, 1 eq.) in acetonitrile (5 mL) was added NIS (227 mg, 1.01 mmol, 1.2 eq.). The resulting mixture was stirred at 0 °C for 2 h under nitrogen atmosphere. The reaction was quenched with saturated sodium thiosulfate solution (aq.) (30 mL), and the mixture was extracted with dichloromethane (3×20 mL). The combined organic layers were washed with brine (3×30 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by silica gel column eluting with petroleum ether/ethyl acetate (1/3) to give 200 mg of 4-iodo-2-(2,2,2-trifluoroethyl)-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-3-one (INT-A20-5) (56%) as brown oil. 1H NMR (DMSO, 400 MHz) δ -0.05 (9H, s), 0.74-0.87 (2H, m), 3.42-3.50 (2H, m), 4.72 (2H, q), 5.10 (2H, s), 8.35 (1H, s). Step 6. Preparation of 1-methyl-6-oxo-5-[3-oxo-2-(2,2,2-trifluoroethyl)-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-4-yl]pyridine-3-carboxylic acid methyl ester (INT-A20-6). 5-Bromo-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (561 mg, 2.28 mmol mmol, 1 equivalent), Pd(dppf)Cl 2 .CH 2Cl 2A mixture of (167 mg, 0.23 mmol, 0.1 eq), bis(pinacol)diboron (868 mg, 3.42 mmol, 1.5 eq) and KOAc (671 mg, 6.84 mmol, 3 eq) in 1,4-dioxane (6 mL) was stirred at 80 °C overnight under nitrogen atmosphere. The mixture was cooled to room temperature, and then 4-iodo-2-(2,2,2-trifluoroethyl)-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-3-one (966 mg, 2.28 mmol, 1 eq), K 2CO 3(948 mg, 6.86 mmol, 3 equivalents), Pd(dppf)Cl 2(167 mg, 0.23 mmol, 0.1 eq), 1,4-dioxane (10 mL) and H 2O (5 mL). The reaction mixture was stirred at 80 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (50 mL), and the mixture was extracted with dichloromethane (3×50 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using petroleum ether/ethyl acetate (1/5) to give 1 g of 1-methyl-6-oxo-5-[3-oxo-2-(2,2,2-trifluoroethyl)-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-4-yl]pyridine-3-carboxylic acid methyl ester (INT-A20-6) (94%) as a brown solid. LCMS: m/z (ESI), [M+H] +=462.00. Step 7. Preparation of methyl 5-[5-hydroxy-1-(2,2,2-trifluoroethyl)pyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylate (INT-A20-7). A mixture of methyl 1-methyl-6-oxo-5-[3-oxo-2-(2,2,2-trifluoroethyl)-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-4-yl]pyridine-3-carboxylate (1 g, 2.17 mmol, 1 eq.) and HCl in 1,4-dioxane (20 mL) was stirred at room temperature for 2 h under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was treated with saturated NaHCO 3(aqueous solution) solution (100 mL) and extracted with dichloromethane (3×100 mL). The combined organic layers were washed with brine (3×200 mL) and purified by anhydrous Na 2SO 4Drying and concentrating under reduced pressure gave 700 mg of methyl 5-[5-hydroxy-1-(2,2,2-trifluoroethyl)pyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylate (INT-A20-7) (97%) as a brown solid. 1H NMR (DMSO, 400 MHz) δ 3.67 (3H, s), 3.84 (3H, s), 4.78 (2H, q), 8.11 (1H, s), 8.33 (1H, s), 8.50 (1H, d). Step 8. 4-(3-{[(2 RPreparation of tert-butyl 5-({4-[5-(methoxycarbonyl)-1-methyl-2-oxopyridin-3-yl]-2-(2,2,2-trifluoroethyl)pyrazol-3-yl}oxy)-2-methylpentyl]amino}-4-nitrophenyl)piperazine-1-carboxylate (INT-A20-8). To a stirred solution of methyl 5-[5-hydroxy-1-(2,2,2-trifluoroethyl)pyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylate (970 mg, 2.93 mmol, 1.00 equiv) and triphenylphosphine (2.304 g, 8.78 mmol, 3 equiv) in tetrahydrofuran (15 mL) under nitrogen atmosphere at 0° C. was added tert-butyl 4-(3-{[(2R)-5-hydroxy-2-methylpentyl]amino}-4-nitrophenyl)piperazine-1-carboxylate (INT-A20-2, 1.237 g, 2.93 mmol, 1.00 equiv) and diisopropyl azodicarboxylate (1.776 g, 8.78 mmol, 3 equiv). The resulting mixture was stirred at 0° C. for 2 h under nitrogen atmosphere. The reaction was quenched with water (100 mL), and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using petroleum ether/ethyl acetate to give 1.2 g of 4-(3-{[(2 R)-5-({4-[5-(methoxycarbonyl)-1-methyl-2-oxopyridin-3-yl]-2-(2,2,2-trifluoroethyl)pyrazol-3-yl}oxy)-2-methylpentyl]amino}-4-nitrophenyl)piperazine-1-carboxylic acid tert-butyl ester (INT-A20-8) (55%). LCMS: m/z (ESI), [M+H] +=736.25. Step 9. 4-(4-amino-3-{[(2 RPreparation of tert-butyl 5-({4-[5-(methoxycarbonyl)-1-methyl-2-oxopyridin-3-yl]-2-(2,2,2-trifluoroethyl)pyrazol-3-yl}oxy)-2-methylpentyl]amino}phenyl)piperazine-1-carboxylate (INT-A20-9). In a nitrogen atmosphere at 0 ° C, 4-(3-{[(2 RTo a stirred mixture of tert-butyl 5-({4-[5-(methoxycarbonyl)-1-methyl-2-oxopyridin-3-yl]-2-(2,2,2-trifluoroethyl)pyrazol-3-yl}oxy)-2-methylpentyl]amino}-4-nitrophenyl)piperazine-1-carboxylate (600 mg, 0.81 mmol, 1 eq.) and ranyl nickel (699 mg, 8.15 mmol, 10 eq.) in methanol (5 mL) was added hydrazine hydrate (61 mg, 1.22 mmol, 1.5 eq.). The resulting mixture was stirred at room temperature for 2 h. The resulting mixture was filtered. The filter cake was washed with methanol (3×30 mL), and the solution was concentrated under reduced pressure to obtain 575 mg of 4-(4-amino-3-{[(2 R)-5-({4-[5-(methoxycarbonyl)-1-methyl-2-oxopyridin-3-yl]-2-(2,2,2-trifluoroethyl)pyrazol-3-yl}oxy)-2-methylpentyl]amino}phenyl)piperazine-1-carboxylate (INT-A20-9). This material was used in the next reaction without further purification. LCMS: m/z (ESI), [M+H] +=706.45. Step 10. 4-{2-amino-3-[(2 RPreparation of tert-butyl 5-({4-[5-(methoxycarbonyl)-1-methyl-2-oxopyridin-3-yl]-2-(2,2,2-trifluoroethyl)pyrazol-3-yl}oxy)-2-methylpentyl]-1,3-benzodiazol-5-yl}piperazine-1-carboxylate (INT-A20-10). A solution of tert-butyl 4-(4-amino-3-{[(2R)-5-({4-[5-(methoxycarbonyl)-1-methyl-2-oxopyridin-3-yl]-2-(2,2,2-trifluoroethyl)pyrazol-3-yl}oxy)-2-methylpentyl]amino}phenyl)piperazine-1-carboxylate (600 mg, 0.85 mmol, 1 eq) and BrCN (108 mg, 1.02 mmol, 1.2 eq) in dichloromethane (8 mL) was stirred at room temperature overnight under nitrogen atmosphere. The resulting mixture was concentrated under vacuum. The residue was purified by Prep-TLC eluting with dichloromethane/methanol (10/1) to give 180 mg of 4-{2-amino-3-[(2 R)-5-({4-[5-(methoxycarbonyl)-1-methyl-2-oxopyridin-3-yl]-2-(2,2,2-trifluoroethyl)pyrazol-3-yl}oxy)-2-methylpentyl]-1,3-benzodiazol-5-yl}piperazine-1-carboxylic acid tert-butyl ester (INT-A20-10) (29%). 1H NMR (DMSO, 400 MHz) δ 0.81 (4H, d), 1.15-1.34 (4H, m), 1.36-1.48 (11H, m), 1.70-1.76 (1H, m), 1.85-1.91 (1H, m), 1.97-2.05 (1H, m), 2.93-2.99 (4H, m), 3.30-3.37 (3H, m), 3.55-3.63 (3H, m), 3.75-3.83 (3H, m), 3.82-3.98 (5H, m), 4.86-4.96 (2H, m), 5.77 (6H, s), 6.67-6.77 (1H, m), 6.90 (1H, s), 6.97-7.16 (3H, m), 8.03 (1H, s), 8.05 (1H, d), 8.52 (1H, d). Step 11. 5-(5-{[(4 R)-4-({2-amino-6-[4-(tert-butoxycarbonyl)piperazine-1-yl]-1,3-benzodiazol-1-yl}methyl)pentyl]oxy}-1-(2,2,2-trifluoroethyl)pyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A20-11). 4-{2-amino-3-[(2 R)-5-({4-[5-(methoxycarbonyl)-1-methyl-2-oxopyridin-3-yl]-2-(2,2,2-trifluoroethyl)pyrazol-3-yl}oxy)-2-methylpentyl]-1,3-benzodiazol-5-yl}piperazine-1-carboxylic acid tert-butyl ester (150 mg, 0.20 mmol, 1 equivalent) and LiOH.H 2O (13 mg, 0.31 mmol, 1.5 equivalents) in tetrahydrofuran (4 mL) and H 2The mixture in 4% O (1 mL) was stirred at room temperature under nitrogen atmosphere for 2 h. The resulting mixture was concentrated under reduced pressure. The residue was purified by reverse phase flash chromatography on a C18 silica gel column eluted with acetonitrile/water (0% to 100% gradient) to give 70 mg of 5-(5-{[(4 R)-4-({2-amino-6-[4-(tert-butoxycarbonyl)piperazine-1-yl]-1,3-benzodiazol-1-yl}methyl)pentyl]oxy}-1-(2,2,2-trifluoroethyl)pyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A20-11) (47%). 1H NMR (DMSO, 400 MHz) δ 0.76 (3H, d), 1.25 (1H, s), 1.42 (9H, s), 1.54-1.61 (1H, m), 1.70-1.76 (1H, m), 1.85-1.91 (1H, m), 1.97-2.05 (1H, s), 2.93-2.99 (4H, m), 3.30-3.37 (3H, m), 3.55-3.63 (3H, m), 3.75-3.83 (3H, m), 3.82-3.98 (5H, m), 4.88 (2H, m), 6.54 (2H, s), 6.60 (1H, q), 6.79 (1H, d), 6.96 (1H, d), 8.02 (1H, s), 8.04 (1H, d), 8.25 (1H, d). Step 12. Preparation of tert-butyl 4-[(11R)-11,26-dimethyl-23,27-dioxo-5-(2,2,2-trifluoroethyl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octadecano-1(28),2(6),3, 14,16,18,20,24-octan-16-yl]piperazine-1-carboxylate (INT-A20-12). N,N-diisopropylethylamine (27 mg, 0.21 mmol, 3 equivalents), 5-(5-{[(4R)-4-({2-amino-6-[4-(tert-butoxycarbonyl)piperazin-1-yl]-1,3-benzodiazol-1-yl}methyl)pentyl]oxy}-1-(2,2,2-trifluoroethyl)pyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (50 mg, 0.07 mmol, 1 equivalent) and N,N,N,N-Tetramethyl- OA mixture of -(7-azabenzotriazol-1-yl)urea hexafluorophosphate (40 mg, 0.10 mmol, 1.5 equiv.) in 1,4-dioxane (2 mL) was stirred at 60 °C overnight under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (30 mL), and the mixture was extracted with ethyl acetate (3×20 mL). The combined organic layers were washed with brine (3×20 mL), purified by anhydrous Na 2SO 4The residue was purified by Prep-TLC using dichloromethane/methanol (10/1) to give 48 mg of tert-butyl 4-[(11R)-11,26-dimethyl-23,27-dioxo-5-(2,2,2-trifluoroethyl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octacos-1(28), 2(6),3,14,16,18,20,24-octan-16-yl]piperazine-1-carboxylate (INT-A20-12) (98%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=699.45. Step 13. (11 RPreparation of )-11,26-dimethyl-16-(piperazin-1-yl)-5-(2,2,2-trifluoroethyl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octacosa-1(28),2(6),3, 14,16,18,20,24-octaene-23,27-dione (INT-A20-13). A mixture of tert-butyl 4-[(11R)-11,26-dimethyl-23,27-dioxo-5-(2,2,2-trifluoroethyl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octacos-1(28),2(6),3,14,16,18,20,24-octan-16-yl]piperazine-1-carboxylate (INT-A20-13 40 mg, 0.06 mmol, 1 eq.) and trifluoroacetic acid (0.5 mL) in dichloromethane (2 mL) was stirred under nitrogen atmosphere at room temperature for 1 h. The resulting mixture was concentrated under reduced pressure. Saturate the residue with NaHCO3(aqueous solution) solution (100 mL) and extracted with dichloromethane (3×100 mL). The combined organic layers were washed with brine (3×200 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The crude product was used directly in the next step without further purification. LCMS: m/z (ESI), [M+H] +=599.15. Step 14. (11 R)-11,26-dimethyl-16-(4-methylpiperazine-1-yl)-5-(2,2,2-trifluoroethyl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]twenty-eight-1(28),2(6),3, 14,16,18,20,24-octaene-23,27-dione (Example A20). At room temperature, (11 RTo a stirred mixture of 1,1,26-dimethyl-16-(piperazin-1-yl)-5-(2,2,2-trifluoroethyl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octacos-1(28),2(6),3, 14,16,18,20,24-octaene-23,27-dione (40 mg, 0.07 mmol, 1 eq), trimethylamine (20 mg, 0.20 mmol, 3 eq) and paraformaldehyde (6 mg, 0.06 mmol, 1 eq) in dichloromethane (2 mL) was added sodium acetyloxyborohydride (42 mg, 0.20 mmol, 3 eq). The resulting mixture was stirred at room temperature for 2 h under nitrogen atmosphere and concentrated under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (5/1) to give 6.7 mg (11 R)-11,26-dimethyl-16-(4-methylpiperazine-1-yl)-5-(2,2,2-trifluoroethyl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]twenty-eight-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (Example A20) (16%). LCMS: m/z (ESI), [M+H] +=613.30. 1H NMR (DMSO-d 6, 400 MHz) δ 0.77-0.89 (5H, m), 1.11-1.29 (9H, m), 1.40-1.50 (2H, m), 1.85-1.92 (2H, m), 2.15-2.21 (2H, m), 2.28 (3H, s), 2.75-2.80 (1H, m), 3.14-3.20 (3H, m), 3.63 (3H, s), 3.90-3.96 (2H, m), 4.07-4.11 (1H, m), 4.43-4.48 (1H, m), 4.93-5.00 (1H, m), 6.87 (1H, d), 7.14 (1H, s), 7.36 (1H, d), 8.31 (1H, d), 8.46 (1H, s), 8.73 (1H, d), 12.37 (1H, s) Example A21 (11 R)-5-ethyl-11,26-dimethyl-16-(4-methylpiperazine-1-yl)-7-oxo-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]twenty-eight-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione Step 1. Preparation of 2-ethylpyrazol-3-ol (INT-A21-1). A mixture of ethylhydrazine dihydrochloride (5.7 g, 43.06 mmol, 1 equivalent) and (2E)-3-methoxyprop-2-enoic acid methyl ester (5 g, 43.06 mmol, 1 equivalent) in methanol (50 mL) was stirred at 80 ° C overnight under a nitrogen atmosphere. After cooling to room temperature, the reaction mixture was concentrated under reduced pressure, and the residue was purified by silica gel chromatography using petroleum ether/ethyl acetate (1/1) to obtain 4 g of 2-ethylpyrazol-3-ol (INT-A21-1) (82%) as a white solid. 1H NMR (DMSO-d 6, 400 MHz) δ 1.26 (3H, t), 3.94-3.98 (2H, m), 5.82 (1H, m), 7.48 (1H, d), 10.99 (1H, s). Step 2. Preparation of 2-ethyl-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-3-one (INT-A21-2). 2-Ethylpyrazol-3-ol (5 g, 44.59 mmol, 1 eq.), 2-(trimethylsilyl)ethoxymethyl chloride (11.15 g, 66.88 mmol, 1.5 eq.) and K 2CO 3A mixture of (15.41 g, 111.47 mmol, 2.5 equiv) in acetonitrile (50 mL) was stirred at room temperature under nitrogen atmosphere for 0.5 h. The reaction was quenched with water (500 mL), and the mixture was extracted with ethyl acetate (3×500 mL). The combined organic layers were washed with brine (3×500 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue is purified by silica gel chromatography using petroleum ether/ethyl acetate (1/1) to give 1.5 g of 2-ethyl-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-3-one (INT-A21-2) (13%) as a yellow oil. 1H NMR (DMSO-d 6, 400 MHz) δ 0.00 (9H, s), 0.79-0.84 (2H, m), 1.04 (3H, t), 3.38-3.47 (2H, m), 3.77 (2H, q), 5.08 (2H, s), 5.27 (1H, d), 7.89 (1H, d) Step 3. Preparation of 2-ethyl-4-iodo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-3-one (INT-A21-3). 2-Ethyl-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-3-one (1.15 g, 4.74 mmol, 1 equivalent) and N-iodosuccinimide (1.28 g, 5.69 mmol, 1.2 equivalents) in acetonitrile (15 mL) was stirred at 0 °C for 3 h under nitrogen atmosphere. The reaction was quenched with water (100 mL), and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by silica gel chromatography using petroleum ether/ethyl acetate (1/1) to give 1.9 g of 2-ethyl-4-iodo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-3-one (INT-A21-3) (97%) as a yellow solid. 1H NMR (DMSO-d 6, 400 MHz) δ 0.03 (9H, s), 0.81 (2H, t), 1.05 (3H, t), 3.43 (2H, t), 3.83 (2H, q), 5.09 (2H, s), 8.17 (1H, s). Step 4. Preparation of 5-(2-ethyl-3-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A21-4). 5-Bromo-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (1.4 g, 5.69 mmol, 1 equivalent), bis(pinacol)diboron (1.73 g, 6.82 mmol, 1.2 equivalent), KOAc (1.4 g, 14.22 mmol, 2.5 equivalent) and Pd(dppf)Cl 2A mixture of dichloromethane (0.46 g, 0.57 mmol, 0.1 eq.) in dioxane (16 mL) was stirred at 80 °C for 2 h under a nitrogen atmosphere. 2-ethyl-4-iodo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-3-one (1.7 g, 4.61 mmol, 0.9 eq.), Pd(dppf)Cl 2Dichloromethane (0.28 g, 0.51 mmol, 0.1 equivalent), K 2CO 3(1.91 g, 13.82 mmol, 2.7 eq.) and H 2O (4 mL), the reaction mixture was cooled to room temperature. The reaction was stirred at 80 °C for another 2 h under nitrogen atmosphere. After cooling to room temperature, the reaction was quenched with water (100 mL), and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by silica gel chromatography using dichloromethane/methanol (10/1) to obtain 1.6 g of ethyl 5-(2-ethyl-3-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (INT-A21-4) (76%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=408.10. 1H NMR (DMSO-d 6, 400 MHz) δ 0.05 (9H, s), 0.82 (2H, t), 1.10-1.13 (3H, m), 3.47 (2H, t), 3.60 (3H, s), 3.81 (3H, s), 3.89 (2H, q), 5.29 (2H, s), 8.43 (1H, d), 8.88 (1H, s), 9.21 (1H, d). Step 5. Preparation of 5-(1-ethyl-5-hydroxypyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A21-5). A solution of methyl 5-(2-ethyl-3-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (1.6 g, 3.92 mmol, 1 eq) and HCl (20 mL, 4 M in 1,4-dioxane) was stirred at room temperature for 2 h. The residue was treated with dichloromethane (30 mL) and K 2CO 3Work up. The mixture was filtered. The filter cake was washed with dichloromethane (3×20 mL). The resulting solution was concentrated under reduced pressure to give 640 mg of methyl 5-(1-ethyl-5-hydroxypyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (INT-A21-5) as a yellow solid. This material was used in the next reaction without further purification. LCMS: m/z (ESI), [M+H] +=278.00. Step 6. 5-(5-{[(4 RPreparation of methyl 5-(1-ethyl-5-hydroxypyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (INT-A21-6). A mixture of methyl 5-(1-ethyl-5-hydroxypyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (670 mg, 2.41 mmol, 1 eq.) and triphenylphosphine (1.58 g, 6.04 mmol, 2.5 eq.) in tetrahydrofuran (10 mL) was treated with diisopropyl azodicarboxylate (0.98 g, 4.83 mmol, 2 eq.) at 0°C under nitrogen atmosphere for 1 h, followed by the addition of (4 R)-5-[(5-bromo-2-nitrophenyl)amino]-4-methylpentan-1-ol (M3-7, 766 mg, 2.41 mmol, 1 equiv). After stirring for another 4 h at 0 °C under nitrogen atmosphere, the reaction was quenched with water (100 mL), and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (10/1) to give 720 mg of 5-(5-{[(4 R)-5-[(5-bromo-2-nitrophenyl)amino]-4-methylpentyl]oxy}-1-ethylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A21-6) (51%). LCMS: m/z (ESI), [M+H] +=578.00. Step 7. 5-(5-{[(4 RPreparation of 5-(5-{[(4 )-5-[(2-amino-5-bromophenyl)amino]-4-methylpentyl]oxy}-1-ethylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A21-7).RA mixture of methyl 5-[(5-bromo-2-nitrophenyl)amino]-4-methylpentyl]oxy}-1-ethylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (720 mg, 1.25 mmol, 1 equiv), ranyl nickel (54 mg, 0.62 mmol, 0.5 equiv) and hydrazine (120 mg, 3.74 mmol, 3 equiv) in methanol (15 mL) was stirred at room temperature for 2 h. The resulting mixture was filtered and the filter cake was washed with methanol (3×20 mL). The resulting solution was concentrated under reduced pressure to give 600 mg of 5-(5-{[(4 R)-5-[(2-amino-5-bromophenyl)amino]-4-methylpentyl]oxy}-1-ethylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A21-7). This material was used in the next reaction without further purification. LCMS: m/z (ESI), [M+H] +=546.20. Step 8. 5-(5-{[(4 RPreparation of methyl 4-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]pentyl]oxy}-1-ethylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (INT-A21-8). 5-(5-{[(4 RA mixture of methyl 5-[(2-amino-5-bromophenyl)amino]-4-methylpentyl]oxy}-1-ethylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (600 mg, 1.10 mmol, 1 equiv) and cyanogen bromide (232 mg, 2.20 mmol, 2 equiv) in ethanol (15 mL) was stirred at room temperature for 2 h and concentrated. The residue was purified by silica gel chromatography using dichloromethane/methanol (10/1) to give 400 mg of 5-(5-{[(4 R)-4-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]pentyl]oxy}-1-ethylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A21-8) (63%). LCMS: m/z (ESI), [M+H] +=573.30. Step 9. 5-(5-{[(4 RPreparation of 5-(5-{[(4 RTo a stirred mixture of methyl 4-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]pentyl]oxy}-1-ethylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (700 mg, 1.22 mmol, 1 eq.) in tetrahydrofuran (15 mL) was added LiOH (58 mg, 2.45 mmol, 2 eq.) in H 2O (3 mL). The resulting mixture was stirred at room temperature for 2 h and concentrated under reduced pressure. The residue was purified by reverse phase flash chromatography using 10% to 50% acetonitrile/water to give 600 mg of 5-(5-{[(4 R)-4-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]pentyl]oxy}-1-ethylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A21-9) (87%). LCMS: m/z (ESI), [M+H] +=559.30. Step 10. (11 R)-16-bromo-5-ethyl-11,26-dimethyl-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]octadecene-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (INT-A21-10). 5-(5-{[(4 R)-4-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]pentyl]oxy}-1-ethylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (460 mg, 0.82 mmol, 1 equivalent), N,N,N,N-Tetramethyl-O-(7-azabenzotriazol-1-yl)urea hexafluorophosphate (627 mg, 1.65 mmol, 2 equivalents) and N, N- A mixture of diisopropylethylamine (213 mg, 1.65 mmol, 2 equiv.) in dioxane (10 mL) was stirred at room temperature under nitrogen atmosphere for 2 h. The reaction was quenched with water (100 mL), and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC eluting with dichloromethane/methanol (15/1) to give 430 mg (11 R)-16-bromo-5-ethyl-11,26-dimethyl-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octadecene-1(28),2(6),3, 14,16,18,20,24-octaene-23,27-dione (INT-A21-10) (96%). LCMS: m/z (ESI), [M+H] +=541.00. Step 11. (11 R)-5-ethyl-11,26-dimethyl-16-(4-methylpiperazine-1-yl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]twenty-eight-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (Example A21). In a nitrogen atmosphere at room temperature to (11 R)-16-bromo-5-ethyl-11,26-dimethyl-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]octadecene-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (100 mg, 0.18 mmol, 1 equivalent), BrettPhos Pd G 3To a mixture of 1,4-dioxane (5 mL) and 1-methylpiperazine (84.02 mg, 0.09 mmol, 0.5 eq.) was added LiHMDS (1.1 mL, 1.11 mmol, 6 eq.) dropwise. The reaction mixture was stirred at 60 °C for 30 min. After cooling to room temperature, the reaction was heated with saturated NH4Cl solution (100 mL) was quenched, and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (18/1) to give 29 mg (11 R)-5-ethyl-11,26-dimethyl-16-(4-methylpiperazine-1-yl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]twenty-eight-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (Example A21) (28%). LCMS: m/z (ESI), [M+H] +=559.35. 1H NMR (DMSO-d 6, 400 MHz) δ 0.80 (3H, d), 1.37 (3H, t), 1.45 (1H, t), 1.90 (2H, d), 2.17-2.25 (4H, m), 2.51 (4H, t), 2.79 (1H, s), 3.15 (4H, t), 3.61 (3H, s), 3.86-4.13 (5H, m), 4.36-4.40 (1H, m), 6.86 (1H, d), 7.12 (1H, d), 7.35 (1H, d), 8.26 (1H, d), 8.37 (1H, s), 8.78 (1H, d), 12.35 (1H, s). Example A22 (11 R)-5-cyclopropyl-11,26-dimethyl-16-(4-methylpiperazine-1-yl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]twenty-eight-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione Step 1. 5-(5-{[(4 RPreparation of methyl 5-[(5-bromo-2-nitrophenyl)amino]-4-methylpentyl]oxy}-1-cyclopropylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (INT-A22-1). To a stirred mixture of methyl 5-(1-cyclopropyl-5-hydroxypyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (INT-2-7, 750 mg, 2.59 mmol, 1 eq) and triphenylphosphine (1360 mg, 5.19 mmol, 2 eq) in tetrahydrofuran (10 mL) at 0°C, diisopropyl azodicarboxylate (1048 mg, 5.19 mmol, 2 eq) and (4 R)-5-[(5-bromo-2-nitrophenyl)amino]-4-methylpentan-1-ol (M3-7, 905 mg, 2.85 mmol, 1.1 equiv). The resulting mixture was stirred at 0 °C for 2 h under a nitrogen atmosphere. The reaction was quenched with water (30 mL), and the mixture was extracted with dichloromethane (3×30 mL). The combined organic layers were purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column using petroleum ether/ethyl acetate (1/1) to give 700 mg of 5-(5-{[(4 R)-5-[(5-bromo-2-nitrophenyl)amino]-4-methylpentyl]oxy}-1-cyclopropylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A22-1) (45%). LCMS: m/z (ESI), [M+H] +=590.10 Step 2. 5-(5-{[(4 RPreparation of methyl 5-[(2-amino-5-bromophenyl)amino]-4-methylpentyl]oxy}-1-cyclopropylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (INT-A22-2). At 0°C, 5-(5-{[(4 R)-5-[(5-bromo-2-nitrophenyl)amino]-4-methylpentyl]oxy}-1-cyclopropylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (700 mg, 1.19 mmol, 1 equiv) and ranyl nickel (51 mg, 0.60 mmol, 0.5 equiv) were added to a stirring mixture of methanol (15 mL) with NH 2NH 2.H 2O (90 mg, 1.79 mmol, 1.5 equiv). The mixture was stirred at 0 °C for 1 h under nitrogen atmosphere. The mixture was filtered. The filter cake was washed with methanol (2×20 mL). The resulting solution was concentrated under reduced pressure. The residue was purified on a silica gel column using dichloromethane/methanol (10/1) to obtain 570 mg 5-(5-{[(4 R)-5-[(2-amino-5-bromophenyl)amino]-4-methylpentyl]oxy}-1-cyclopropylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A22-2) (85%). LCMS: m/z (ESI), [M+H] +=560.40 Step 3. 5-(5-{[(4 RPreparation of methyl 4-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]pentyl]oxy}-1-cyclopropylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (INT-A22-3). 5-(5-{[(4 RA mixture of methyl 5-[(2-amino-5-bromophenyl)amino]-4-methylpentyl]oxy}-1-cyclopropylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (570 mg, 1.02 mmol, 1 eq.) and BrCN (162 mg, 1.53 mmol, 1.5 eq.) in ethanol (5 mL) was stirred at room temperature for 2 h under a nitrogen atmosphere. The mixture was concentrated under reduced pressure, and the residue was purified on a silica gel column using dichloromethane/methanol (10/1) for elution to obtain 550 mg of 5-(5-{[(4 R)-4-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]pentyl]oxy}-1-cyclopropylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A22-3) (92%). LCMS: m/z (ESI), [M+H] +=585.15 Step 4. 5-(5-{[(4 RPreparation of 5-(5-{[(4 )-4-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]pentyl]oxy}-1-cyclopropylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A22-4).RTo a stirred mixture of methyl 4-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]pentyl]oxy}-1-cyclopropylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (480 mg, 0.823 mmol, 1 eq.) in tetrahydrofuran (20 mL) was added LiOH (78.81 mg, 3.292 mmol, 4 eq.) and H 2O (5 mL). The resulting mixture was stirred at 60 °C for 30 min. The mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by reverse phase flash chromatography using 10% to 40% acetonitrile/water elution to give 380 mg of 5-(5-{[(4 R)-4-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]pentyl]oxy}-1-cyclopropylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A22-4) (81%). LCMS: m/z (ESI), [M+H] += 571.00. Step 5. (11 R)-16-bromo-5-cyclopropyl-11,26-dimethyl-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]octadecene-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (INT-A22-5). 5-(5-{[(4 R)-4-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]pentyl]oxy}-1-cyclopropylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (375 mg, 0.66 mmol, 1 equivalent), N, N-Diisopropylethylamine (255 mg, 1.98 mmol, 3 equiv.) and N,N,N,N-Tetramethyl- OA mixture of -(7-azabenzotriazol-1-yl)urea hexafluorophosphate (375 mg, 0.98 mmol, 1.5 equiv.) in 1,4-dioxane (10 mL) was stirred at 60 °C for 2 h under nitrogen atmosphere. The reaction mixture was cooled to room temperature and saturated with NH4Cl(aqueous solution) (20 mL) and extracted with dichloromethane (3×20 mL). The combined organic layers were washed with brine (3×30 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column eluting with dichloromethane/methanol (10/1) to give 330 mg (11 R)-16-bromo-5-cyclopropyl-11,26-dimethyl-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]octadecene-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (INT-A22-5) (90%). LCMS: m/z (ESI), [M+H] += 553.10. Step 6. (11 R)-5-cyclopropyl-11,26-dimethyl-16-(4-methylpiperazine-1-yl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]twenty-eight-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (Example A22). In a nitrogen atmosphere at room temperature to (11 R)-16-bromo-5-cyclopropyl-11,26-dimethyl-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]octadecene-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (90 mg, 0.16 mmol, 1 equivalent), BrettPhos Pd G 3To a stirred mixture of 4-(44 mg, 0.05 mmol, 0.3 eq.) and 1-methylpiperazine (49 mg, 0.49 mmol, 3 eq.) in 1,4-dioxane (9 mL) was added LiHMDS (2.7 mL, 2.7 mmol, 16.9 eq.). The mixture was stirred at 60 °C for 30 min under nitrogen atmosphere and cooled to room temperature. The reaction was heated with saturated NH4Cl(aq) solution (30 mL) was quenched, and the mixture was extracted with dichloromethane (3×20 mL). The combined organic layers were washed with brine (3×30 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column using dichloromethane/methanol (15/1) to give 24.7 mg of the title compound (Example A22) as an off-white solid (26%). LCMS: m/z (ESI), [M+H] +=571.40. 1H NMR (DMSO-d 6, 400 MHz) δ 0.81 (3H, d), 0.971.06 (4H, m), 1.37-1.54 (1H, m), 1.83-2.03 (2H, m), 2.17-2.23 (1H, m), 2.24 (3H, s), 2.50 (4H, t), 2.82 (1H, br s), 3.15 (4H, br s), 3.53-3.60 (1H, m), 3.61 (3H, s), 3.85-4.20 (3H, m), 4.30-4.47 (1H, m), 6.86 (1H, dd), 7.13 (1H, d), 7.35 (1H, d), 8.26 (1H, d), 8.30 (1H, s), 8.84 (1H, d), 12.35 (1H, s). Example A23 (11 R)-15-chloro-5,11,26-trimethyl-16-(4-methylpiperazine-1-yl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]twenty-eight-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione Step 1. (4 RPreparation of )-5-[(2-chloro-3-fluoro-6-nitrophenyl)amino]-4-methylpentan-1-ol (INT-A23-1). 2-Chloro-1,3-difluoro-4-nitrobenzene (1.73 g, 8.96 mmol, 1.05 equivalents) and (4 R)-5-amino-4-methylpentan-1-ol (M3-6, 1 g, 8.53 mmol, 1 equivalent) was added in portions to a stirred mixture in acetonitrile (25 mL) with K 2CO 3(2.36 g, 17.06 mmol, 2 eq.). The mixture was stirred at 60 °C for 2 h under nitrogen atmosphere and cooled to room temperature. The reaction was quenched with water (100 mL) and the mixture was washed with CH2Cl 2(3×100 mL) extraction. The combined organic layers were washed with brine (3×50 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue is purified by silica gel chromatography eluting with petroleum ether/ethyl acetate (3/1) to give 2 g (4 R)-5-[(2-chloro-3-fluoro-6-nitrophenyl)amino]-4-methylpentan-1-ol (INT-A23-1) (80%). LCMS: m/z (ESI), [M+H] +=290.95. 1H NMR (DMSO-d6, 400 MHz) δ 0.82 (3H, d), 1.01 - 1.30 (2H, m), 1.31-1.72 (3H, m), 2.78-2.89 (1H, m), 2.94-3.09 (1H, m), 3.33 (2H, d), 4.37 (1H, t), 6.59 (1H, t), 7.22 (1H, d), 7.76 (1H, d). Step 2. 4-(2-chloro-3-{[(2 RPreparation of tert-butyl 5-hydroxy-2-methylpentyl]amino}-4-nitrophenyl)piperazine-1-carboxylate (INT-A23-2). At room temperature, (4 R)-5-[(2-chloro-3-fluoro-6-nitrophenyl)amino]-4-methylpentan-1-ol (2 g, 6.87 mmol, 1 eq.) and tert-butyl piperazine-1-carboxylate (2.56 g, 13.75 mmol, 2 eq.) were added dropwise to a stirred mixture in dimethyl sulfoxide (30 mL).N,N-Diisopropylethylamine (1.78 g, 13.75 mmol, 2 equiv.). The mixture was stirred at 120 °C for 4 h under nitrogen atmosphere and cooled to room temperature. The reaction was quenched with water (50 mL), and the mixture was extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by silica gel chromatography using petroleum ether/ethyl acetate (10/1) to give 3.1 g of 4-(2-chloro-3-{[(2 R)-5-hydroxy-2-methylpentyl]amino}-4-nitrophenyl)piperazine-1-carboxylic acid tert-butyl ester (INT-A23-2) (98%). LCMS: m/z (ESI), [M+H] +=457.15. Step 3. 4-(2-chloro-3-{[(2 RPreparation of tert-butyl 5-({4-[5-(methoxycarbonyl)-1-methyl-2-oxopyridin-3-yl]-2-methylpyrazol-3-yl}oxy)-2-methylpentyl]amino}-4-nitrophenyl)piperazine-1-carboxylate (INT-A23-3). To a stirred mixture of methyl 5-(5-hydroxy-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (M2, 576.04 mg, 1.09 mmol, 1 eq.) and triphenylphosphine (1.7 g, 3.28 mmol, 3 eq.) in tetrahydrofuran (10 mL) at 0°C were added diisopropyl azodicarboxylate (1.3 g, 3.28 mmol, 3 eq.) and 4-(2-chloro-3-{[(2 R)-5-hydroxy-2-methylpentyl]amino}-4-nitrophenyl)piperazine-1-carboxylic acid tert-butyl ester (1 g, 1.09 mmol, 1 equiv). The mixture was stirred at 0 °C for 1 h under nitrogen atmosphere. The reaction was quenched with water (50 mL), and the mixture was extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC and eluted with dichloromethane/methanol (25/1) to give 1.1 g of 4-(2-chloro-3-{[(2 R)-5-({4-[5-(methoxycarbonyl)-1-methyl-2-oxopyridin-3-yl]-2-methylpyrazol-3-yl}oxy)-2-methylpentyl]amino}-4-nitrophenyl)piperazine-1-carboxylic acid tert-butyl ester (INT-A23-3) (66%). LCMS: m/z (ESI), [M+H] +=702.15. Step 4. 4-(4-amino-2-chloro-3-{[(2 RPreparation of tert-butyl 5-({4-[5-(methoxycarbonyl)-1-methyl-2-oxopyridin-3-yl]-2-methylpyrazol-3-yl}oxy)-2-methylpentyl]amino}phenyl)piperazine-1-carboxylate (INT-A23-4). Raney nickel (500 mg) and 4-(2-chloro-3-{[(2 R)-tert-butyl 5-({4-[5-(methoxycarbonyl)-1-methyl-2-oxopyridin-3-yl]-2-methylpyrazol-3-yl}oxy)-2-methylpentyl]amino}-4-nitrophenyl)piperazine-1-carboxylate (1.1 g, 1.56 mmol, 1 equivalent) was added to a stirring mixture of NH 2NH 2 .H 2O (235.26 mg, 4.69 mmol, 3 equiv.). The mixture was stirred at 0 °C for 1 h and filtered. The filter cake was washed with methanol (2×20 mL). The resulting solution was concentrated under reduced pressure. The residue was purified by Prep-TLC and eluted with dichloromethane/methanol (10/1) to give 610 mg of 4-(4-amino-2-chloro-3-{[(2 R)-5-({4-[5-(methoxycarbonyl)-1-methyl-2-oxopyridin-3-yl]-2-methylpyrazol-3-yl}oxy)-2-methylpentyl]amino}phenyl)piperazine-1-carboxylic acid tert-butyl ester (INT-A23-4) (52%). LCMS: m/z (ESI), [M+H] +=672.20. 1H NMR (DMSO- d 6, 400 MHz) δ 0.95 (3H, d), 1.95-1.25 (1H, m), 1.36-1.45 (11H, m), 1.59-1.75 (2H, m), 2.65-2.78 (5H, m), 2.81 (1H, d), 3.57 (4H, s), 3.62-3.76 (7H, m), 3.81 (1H, d), 3.92 (2H, t), 5.76 (3H, s), 6.49-6.59 (2H, m), 7.58-7.69 (1H, m), 8.00 (1H, s), 8.14 (1H, d), 8.45 (1H, d). Step 5. 4-{2-amino-4-chloro-3-[(2 RPreparation of tert-butyl 5-({4-[5-(methoxycarbonyl)-1-methyl-2-oxopyridin-3-yl]-2-methylpyrazol-3-yl}oxy)-2-methylpentyl]-1,3-benzodiazol-5-yl}piperazine-1-carboxylate (INT-A23-5). 4-(4-amino-2-chloro-3-{[(2 RA mixture of tert-butyl 5-({4-[5-(methoxycarbonyl)-1-methyl-2-oxopyridin-3-yl]-2-methylpyrazol-3-yl}oxy)-2-methylpentyl]amino}phenyl)piperazine-1-carboxylate (600 mg, 0.89 mmol, 1 eq.) and BrCN (94.54 mg, 0.89 mmol, 1 eq.) in ethanol (8 mL) was stirred at room temperature for 2 h under nitrogen atmosphere and concentrated under vacuum. The residue was purified by Prep-TLC and eluted with dichloromethane/methanol (12/1) to give 480 mg of 4-{2-amino-4-chloro-3-[(2 R)-5-({4-[5-(methoxycarbonyl)-1-methyl-2-oxopyridin-3-yl]-2-methylpyrazol-3-yl}oxy)-2-methylpentyl]-1,3-benzodiazol-5-yl}piperazine-1-carboxylic acid tert-butyl ester (INT-A23-5) (76%). LCMS: m/z (ESI), [M+H] +=697.20. 1H NMR (DMSO- d 6, 400 MHz) δ 0.01 (5H, s), 1.24 (2H, s), 1.78-1.92 (8H, m), 2.60-2.79 (2H, m), 2.81-2.95 (4H, m), 3.17 (3H, d), 3.57 (3H, s), 3.71 (6H, d), 4.01-4.22 (2H, m), 5.95 (2H, s), 7.04 (2H, s), 7.47 (1H, s), 7.96 (1H, s), 8.12 (1H, d), 8.39 (1H, d). Step 6 and Step 7. 4-[(11 RPreparation of tert-butyl 1-(28),2(6),3,14,16,18,20,24-octan-16-yl)piperazine-1-carboxylate (INT-A23-7). 4-{2-amino-4-chloro-3-[(2 R)-tert-butyl 5-({4-[5-(methoxycarbonyl)-1-methyl-2-oxopyridin-3-yl]-2-methylpyrazol-3-yl}oxy)-2-methylpentyl]-1,3-benzodiazol-5-yl}piperazine-1-carboxylate (INT-A23-5, 450 mg, 0.76 mmol, 1 equiv) and LiOH (18 mg, 0.71 mmol, 1.2 equiv) in tetrahydrofuran/H 2O (2 mL/1 mL) was stirred at 80 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature and concentrated under vacuum. To the residue was added N,N,N,N-Tetramethyl- O-(7-Azabenzotriazole-1-yl)urea hexafluorophosphate (166.96 mg, 0.44 mmol, 1.5 equivalents)) , N,N-Diisopropylethylamine (113.51 mg, 0.87 mmol, 3 equivalents) and 1,4-dioxane (6 mL). The mixture was stirred under nitrogen atmosphere for 3 h. The mixture was treated with water (10 mL), extracted with ethyl acetate (3×10 mL), and washed with anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC and eluted with methylene dichloride/methanol (12/1) to give 160 mg 4-[(11 R)-15-chloro-5,11,26-trimethyl-23,27-dioxo-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octadecano-1(28),2(6),3,14,16,18,20,24-octan-16-yl]piperazine-1-carboxylic acid tert-butyl ester (INT-A23-7) (78%). LCMS: m/z (ESI), [M+H] +=665.15. Step 8. (11 R)-15-chloro-5,11,26-trimethyl-16-(piperazine-1-yl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]twenty-eight-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (Example A23). 4-[(11 R)-15-chloro-5,11,26-trimethyl-23,27-dioxo-7-oxa-4,5,13,20,22,26-hexaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octadecano-1(28),2(6),3,14,16,18,20,24-octan-16-yl]piperazine-1-carboxylic acid tert-butyl ester (150 mg, 0.22 mmol, 1 equivalent) in trifluoroacetic acid (1 mL) and CH 2Cl 2The mixture in (4 mL) was stirred at room temperature under nitrogen atmosphere for 1 h. The mixture was concentrated under reduced pressure. The residue was treated with saturated NaHCO3Solution treatment and CH 2Cl 2(3×10 mL) extraction. The combined organic layers were washed with brine (3×10 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC and eluted with methylene dichloride/methanol (12/1) to give 13.5 mg (11 R)-15-chloro-5,11,26-trimethyl-16-(piperazine-1-yl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octadecene-1(28),2(6),3, 14,16,18,20,24-octaene-23,27-dione (Example A23) (56%). LCMS: m/z (ESI), [M+H] +=565.35. 1H NMR (DMSO-d 6, 400 MHz) δ1.23-1.32 (3H, m), 1.52-1.59 (2H, m), 1.73 (1H, s), 1.95 (1H, s), 2.12-2.19 (1H, m), 2.89-2.97 (9H, m), 3.62-3.69 (3H, m), 3.71-3.76 (3H, m), 3.90 (1H, s), 4.19 (1H, m), 4.32-4.39 (1H, m), 4.70-4.79 (1H, m), 7.09-7.27 (1H, m), 7.50 (1H, d), 8.28-8.45 (2H, m), 8.65-8.76 (1H, m). Example A25 (11 R)-5,11,26-trimethyl-16-(4-methylpiperazine-1-yl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]twenty-eight-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione Step 1. Preparation of 1-(3-chloro-4-nitrophenyl)-4-methylpiperazine (INT-A25-1). 2-Chloro-4-fluoro-1-nitrobenzene (4 g, 22.79 mmol, 1 equivalent) and 1-methylpiperazine-(2.28 g, 22.79 mmol, 1 equivalent) and K 2CO 3(6.3 g, 45.57 mmol, 2 equiv.) in acetonitrile (60 mL) was stirred at 60 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature, treated with water (100 mL), and extracted with dichloromethane (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using petroleum ether/ethyl acetate (1/1) to give 5.6 g of 1-(3-chloro-4-nitrophenyl)-4-methylpiperazine (INT-A25-1) (96%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=256.05. 1H NMR (DMSO-d 6, 400 MHz) δ 2.22 (3H, s), 2.41 (4H, t), 3.45 (4H, t), 7.00-7.03 (1H, m), 7.10 (1H, d), 8.02 (1H, d). Step 2. (4 RPreparation of )-4-methyl-5-{[5-(4-methylpiperazine-1-yl)-2-nitrophenyl]amino}pentan-1-ol (INT-A25-1). 1-(3-chloro-4-nitrophenyl)-4-methylpiperazine (1.5 g, 5.87 mmol, 1 equivalent), K 2CO 3(1.62 g, 11.73 mmol, 2 equiv.) and (4 R)-5-amino-4-methylpentan-1-ol (M3-6, 0.82 g, 7.04 mmol, 1.2 equivalents) in N,N-The mixture in dimethylformamide (10 mL) was stirred at 120 °C for 16 h under nitrogen atmosphere. The reaction mixture was cooled to room temperature, treated with water (80 mL), and extracted with dichloromethane (3×70 mL). The combined organic layers were washed with brine (3×500 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by prep-TLC eluting with petroleum ether/ethyl acetate (1/1) to give 1.5 g (4 R)-4-methyl-5-{[5-(4-methylpiperazine-1-yl)-2-nitrophenyl]amino}pentan-1-ol (INT-A25-2) (76%). LCMS: m/z (ESI), [M+H] +=337.15. Step 3. 1-methyl-5-(1-methyl-5-{[(4 RPreparation of methyl 5-{[5-(4-methylpiperazine-1-yl)-2-nitrophenyl]amino}pentyl]oxy}pyrazol-4-yl)-6-oxopyridine-3-carboxylate (INT-A25-3). To a stirred mixture of triphenylphosphine (1.57 g, 5.97 mmol, 3 equiv.) and methyl 5-(5-hydroxy-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (M2, 498.05 mg, 1.89 mmol, 0.95 equiv.) in tetrahydrofuran (20 mL) was added (4 R)-4-methyl-5-{[5-(4-methylpiperazin-1-yl)-2-nitrophenyl]amino}pentan-1-ol (670 mg, 1.99 mmol, 1 equiv) and diisopropyl azodicarboxylate (1.21 g, 5.97 mmol, 3 equiv). The mixture was stirred at room temperature for 2 h, treated with water (80 mL), and extracted with ethyl acetate (3×80 mL). The combined organic layers were washed with water (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column eluting with dichloromethane/methanol (35/1) to give 500 mg of 1-methyl-5-(1-methyl-5-{[(4R)-4-methyl-5-{[5-(4-methylpiperazin-1-yl)-2-nitrophenyl]amino}pentyl]oxy}pyrazol-4-yl)-6-oxopyridine-3-carboxylic acid methyl ester (INT-A25-3) (43%) as a light yellow solid. LCMS: m/z (ESI), [M+H] +=582.20. Step 4. 5-(5-{[(4 RPreparation of methyl 5-{[2-amino-5-(4-methylpiperazine-1-yl)phenyl]amino}-4-methylpentyl]oxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (INT-A25-4). In a nitrogen atmosphere at 0 ° C, 1-methyl-5-(1-methyl-5-{[(4 RTo a stirred mixture of methyl 4-(4-methyl-5-{[5-(4-methylpiperazin-1-yl)-2-nitrophenyl]amino}pentyl]oxy}pyrazol-4-yl)-6-oxopyridine-3-carboxylate (700 mg, 1.20 mmol, 1 eq.) in methanol (6 mL) were added Ranyl nickel (349.52 mg, 4.08 mmol, 3.4 eq.) and NH 2NH 2 .H 2O (90.37 mg, 1.80 mmol, 1.5 equiv). The resulting mixture was stirred at room temperature for 1 h, treated with water (20 mL), and extracted with dichloromethane (3×20 mL). The combined organic layers were washed with water (3×10 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure to obtain 510 mg of 5-(5-{[(4 R)-5-{[2-amino-5-(4-methylpiperazin-1-yl)phenyl]amino}-4-methylpentyl]oxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A25-4) (77%). This material was used in the next reaction without further purification. LCMS: m/z (ESI), [M+H] +=552.25. Step 5. 5-(5-{[(4 RPreparation of methyl 4-{[2-amino-6-(4-methylpiperazine-1-yl)-1,3-benzodiazol-1-yl]methyl}pentyl]oxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (INT-A25-5). In a nitrogen atmosphere, 5-(5-{[(4 RTo a stirred mixture of methyl 2-(2-{[2-amino-5-(4-methylpiperazin-1-yl)phenyl]amino}-4-methylpentyl]oxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (480 mg, 0.87 mmol, 1 equiv) in dichloromethane (8 mL) was added cyanogen bromide (138.24 mg, 1.30 mmol, 1.5 equiv). The resulting mixture was stirred at room temperature under nitrogen atmosphere for 2 h, treated with water (20 mL), and extracted with dichloromethane (3×20 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by silica gel chromatography using dichloromethane/methanol (15/1) to give 170 mg of 5-(5-{[(4 R)-4-{[2-amino-6-(4-methylpiperazine-1-yl)-1,3-benzodiazol-1-yl]methyl}pentyl]oxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A25-5) (34%). LCMS: m/z (ESI), [M+H] +=577.40. Step 6. 5-(5-{[(4 R)-4-{[2-amino-6-(4-methylpiperazine-1-yl)-1,3-benzodiazol-1-yl]methyl}pentyl]oxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A25-6). At room temperature, 5-(5-{[(4 RTo a stirred mixture of methyl 4-{[2-amino-6-(4-methylpiperazin-1-yl)-1,3-benzodiazol-1-yl]methyl}pentyl]oxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (140 mg, 0.24 mmol, 1 eq.) in tetrahydrofuran (4 mL) was added water (0.3 mL) containing LiOH (11.63 mg, 0.49 mmol, 2 eq.). The resulting mixture was stirred at room temperature for 1 h and concentrated under reduced pressure. The residue was purified by reverse-phase flash chromatography eluting with acetonitrile/water (0% to 100%) to give 37 mg of 5-(5-{[(4 R)-4-{[2-amino-6-(4-methylpiperazine-1-yl)-1,3-benzodiazol-1-yl]methyl}pentyl]oxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A25-6) (27%). LCMS: m/z (ESI), [M+H] +=563.35. Step 7. (11 R)-5,11,26-trimethyl-16-(4-methylpiperazine-1-yl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]twenty-eight-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (Example A25). At room temperature, 5-(5-{[(4 R)-4-{[2-amino-6-(4-methylpiperazine-1-yl)-1,3-benzodiazol-1-yl]methyl}pentyl]oxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (34 mg, 0.06 mmol, 1 equivalent) was added to the stirred mixture in dioxane (3 mL) N,N-Diisopropylethylamine (23 mg, 0.18 mmol, 3 equivalents ) and N,N,N,N-Tetramethyl-O-(7-azabenzotriazol-1-yl)uronium hexafluorophosphate (34 mg, 0.09 mmol, 1.5 equiv). The resulting mixture was stirred at 60 °C for 1 h under a nitrogen atmosphere, cooled to room temperature, and concentrated under reduced pressure. The residue was passed through Prep-HPLC using an XBridge Shield RP18 OBD column containing 10 mmol/L NH4HCO 3of water and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 14.4 mg (11 R)-5,11,26-trimethyl-16-(4-methylpiperazin-1-yl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octadecene-1(28),2(6),3, 14,16,18,20,24-octaene-23,27-dione (Example A25) (43%). LCMS: m/z (ESI), [M+H] +=545.35. 1H NMR (DMSO-d 6, 400 MHz) δ 0.81 (3H, d), 1.44 (1H, q), 1.92 (2H, d), 2.12-2.25 (1H, m), 2.25 (3H, s), 2.50 (4H, t), 2.80 (1H, br s), 3.16 (4H, t), 3.62 (3H, s), 3.72 (3H, s), 3.88-3.99 (2H, m), 4.10 (1H, d), 4.33-4.38 (1H, m), 6.86 (1H, dd), 7.13 (1H, d), 7.36 (1H, d), 8.27 (1H, d), 8.36 (1H, s), 8.81 (1H, d), 12.35 (1H, s). Example A26 (11 R)5,11,26-trimethyl-16-(morpholin-4-yl)-7-oxa-4,5,13,20,22, 26-hexaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]twenty-eight-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione Step 1. Preparation of 4-(3-chloro-4-nitrophenyl)morpholine (INT-A26-1). 2-Chloro-4-fluoro-1-nitrobenzene (3 g, 17.09 mmol, 1 equivalent), K 2CO 3A mixture of 1,4-dimethoxy- ...2SO 4Dry and concentrate under reduced pressure. The residue is purified on a silica gel column eluted with petroleum ether/ethyl acetate (5/1) to give 2.7 g of 4-(3-chloro-4-nitrophenyl)morpholine (INT-A26-1) (65%) as a yellow solid. 1H NMR (DMSO-d 6, 400 MHz) δ 3.38-3.49 (4H, m), 3.63-3.75 (4H, m,), 7.00-7.04 (1H, m), 7.12 (1H, d,), 8.04 (1H, d,). Step 2. ( RPreparation of )-4-methyl-5-((5-morpholino-2-nitrophenyl)amino)pentan-1-ol (INT-A26-2). 4-(3-chloro-4-nitrophenyl)morpholine (1 g, 4.12 mmol, 1 equivalent), Cs 2CO 3(2.69 g, 8.24 mmol, 2 equiv.) and (4 RA mixture of 5-amino-4-methylpentan-1-ol (0.58 g, 4.94 mmol, 1.2 equiv.) in dimethylacetamide (15 mL) was stirred at 120 °C for 4 h under nitrogen atmosphere and cooled to room temperature. The reaction was quenched with water (100 mL), and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by reverse phase flash chromatography using a C18 silica gel column with acetonitrile/water (0% to 100% gradient) as mobile phase to give 730 mg (4 R)-4-methyl-5-{[5-(oxo-4-yl)-2-nitrophenyl]amino}pentan-1-ol (INT-A26-2) (54%). LCMS: m/z (ESI), [M+H] +=324.20. 1HNMR (DMSO-d 6, 400 MHz) δ 0.97 (3H, d), 1.21-1.29 (1H, m), 1.40-1.47 (3H, m), 1.78-1.87 (1H, m), 3.09-3.19 (1H, m), 3.21- 3.28 (1H, m), 3.45-3.50 (4H, m) 3.69-3.73 (6H, m), 4.40 (1H, s), 6.40-6.43 (1H, m), 7.00-7.10 (1H, m), 7.92 (1H, d), 8.02-8.12 (1H, m). Step 3. ( RPreparation of methyl 5-(1-methyl-5-((4-methyl-5-((5-oxolino-2-nitrophenyl)amino)pentyl)oxy)-1H-pyrazol-4-yl)-6-oxo-1,6-dihydropyridine-3-carboxylate (INT-A26-3). To a stirred mixture of methyl 5-(5-hydroxy-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (M2, 260 mg, 0.98 mmol, 1 eq) and triphenylphosphine (777 mg, 2.964 mmol, 3.0 eq) in tetrahydrofuran (3 mL) at 0°C were added diisopropyl azodicarboxylate (599 mg, 2.96 mmol, 3 eq) and (4 R)-4-methyl-5-{[5-(morpholin-4-yl)-2-nitrophenyl]amino}pentan-1-ol (638 mg, 1.97 mmol, 2 equiv.). The mixture was stirred at 0 °C for 2 h under nitrogen atmosphere, treated with water (100 mL), and extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC and eluted with petroleum ether/ethyl acetate (1/1) to give 130 mg of 1-methyl-5-(1-methyl-5-{[(4 R)-4-methyl-5-{[5-(pyridine-4-yl)-2-nitrophenyl]amino}pentyl]oxy}pyrazol-4-yl)-6-oxopyridine-3-carboxylic acid methyl ester (INT-A26-3) (23%). LCMS: m/z (ESI), [M+H] +=569.30. 1H NMR (DMSO-d 6, 400 MHz) δ 0.98 (3H, d), 1.31-1.45 (1H, m), 1.60 (1H, m), 1.74-1.92 (3H, m), 3.16 (1H, m), 3.23-3.30 (1H, m), 3.37-3.43 (4H, m), 3.57 (3H, s), 3.65-3.73 (7H, m), 3.77 (3H, s), 3.96 (2H, t), 6.01 (1H, d), 6.43-6.48 (1H, m), 7.92 (1H, d), 7.99 (1H, s), 8.14 (1H, d), 8.41-8.46 (2H, m). Step 4. ( RPreparation of methyl 5-(5-((5-((2-amino-5-oxolinophenyl)amino)-4-methylpentyl)oxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxo-1,6-dihydropyridine-3-carboxylate (INT-A26-4). Raney nickel (40 mg, 0.47 mmol, 1 equivalent) and 1-methyl-5-(1-methyl-5-{[(4 R)-4-methyl-5-{[5-(pyridine-4-yl)-2-nitrophenyl]amino}pentyl]oxy}pyrazol-4-yl)-6-oxopyridine-3-carboxylic acid methyl ester (INT-A26-3, 270 mg, 0.47 mmol, 1 equivalent) was added to a stirring mixture in methanol (4 mL) with NH 2NH 2 .H 2O (47 mg, 0.95 mmol, 2 eq.). The mixture was stirred at 0 °C for 1 h and filtered. The filter cake was washed with methanol (2×50 mL) and the resulting solution was concentrated under reduced pressure. The crude product was used directly in the next step without further purification. LCMS: m/z (ESI), [M+H] +=539.30. Step 5. Methyl-( RPreparation of 5-(5-((5-(2-amino-6-oxo-1H-benzo[d]imidazol-1-yl)-4-methylpentyl)oxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxo-1,6-dihydropyridine-3-carboxylic acid methyl ester (INT-A26-5). 5-(5-{[(4 RA mixture of methyl 5-{[2-amino-5-(morpholin-4-yl)phenyl]amino}-4-methylpentyl]oxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (190 mg, 0.35 mmol, 1 eq.) and BrCN (41 mg, 0.38 mmol, 1.1 eq.) in ethanol (3 mL) was stirred at room temperature for 2 h under nitrogen atmosphere. The mixture was concentrated under vacuum. The residue was purified by Prep-TLC and eluted with dichloromethane/methanol (12/1) to give 100 mg of 5-(5-{[(4 R)-4-{[2-amino-6-(pyridine-4-yl)-1,3-benzodiazol-1-yl]methyl}pentyl]oxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A26-5) (50%). 1H NMR (DMSO-d 6, 400 MHz) δ 0.84 (d, 3H), 1.27-1.40 (1H, m), 1.44-1.60 (1H, m), 1.67-1.71 (1H, m), 1.80-1.94 (1H, m), 2.04 (1H, s), 2.94-3.03 (3H, m), 3.17 (1H, d), 3.55-3.61 (5H, m), 3.69-3.76 (4H, m), 3.78 (3H, s), 3.88-3.91 (2H, m), 6.21 (1H, s), 6.59-6.63 (1H, m), 6.76 (2H, d), 7.00 (1H, d), 7.99 (1H, s), 8.13 (1H, d), 8.45 (1H, d). Step 6 ( R)-5-(5-((5-(2-amino-6-oxolino-1H-benzo[d]imidazol-1-yl)-4-methylpentyl)oxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxo-1,6-dihydropyridine-3-carboxylic acid (INT-A26-6). 5-(5-{[(4 R)-4-{[2-amino-6-(pyridine-4-yl)-1,3-benzodiazol-1-yl]methyl}pentyl]oxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (100 mg, 0.17 mmol, 1 equiv) and LiOH (8.5 mg, 0.35 mmol, 2 equiv) in tetrahydrofuran/H 2O (4 mL/1 mL) was stirred at room temperature for 2 h under nitrogen atmosphere and concentrated under vacuum. The residue was purified by reverse phase flash chromatography using a C18 silica gel column and acetonitrile/water (0% to 100% gradient) as the mobile phase to obtain 85 mg of 5-(5-{[(4 R)-4-{[2-amino-6-(pyrrolidine-4-yl)-1,3-benzodiazol-1-yl]methyl}pentyl]oxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A26-6) (87%). 1H NMR (DMSO-d 6, 400 MHz) δ 0.78 (3H, d), 1.43 (1H, s), 1.54-1.75 (2H, m), 1.83 (1H, d), 2.03 (1H, s), 2.89-3.04 (4H, m) , 3.50 (3H, s), 3.60 (3H, s), 3.70-3.75 (4H, m), 3.79-3.82 (1H, m), 3.94 (3H, m), 6.56-6.62 (2H, m), 6.76 (1H, d), 6.96 (1H, d), 8.00 (2H, d), 8.01-8.03 (1H, s), 8.31 (1H, d). Step 7 (11 R)5,11,26-trimethyl-16-(morpholin-4-yl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]twenty-eight-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (Example A26). 5-(5-{[(4 R)-4-{[2-amino-6-(pyrrolidine-4-yl)-1,3-benzodiazol-1-yl]methyl}pentyl]oxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (50 mg, 0.09 mmol, 1 equivalent), N,N-Diisopropylethylamine (35 mg, 0.27 mmol, 3 equivalents) and N,N,N,N-Tetramethyl- OA mixture of -(7-azabenzotriazol-1-yl)urea hexafluorophosphate (51 mg, 0.13 mmol, 1.5 equiv) in 1,4-dioxane (3 mL) was stirred at 60 °C for 1 h under nitrogen atmosphere. The mixture was cooled to room temperature and concentrated under vacuum. The residue was purified by Prep-HPLC using an XBridge Prep OBD C18 column with 10 mmol/L NH4HCO 3of water and acetonitrile as mobile phase. The fractions containing the desired compound were evaporated to dryness to give 16 mg (11 R)5,11,26-trimethyl-16-(morpholin-4-yl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octadecene-1(28),2(6),3, 14,16,18,20,24-octaene-23,27-dione (Example A26) (34%). LCMS: m/z (ESI), [M+H] +=532.30. 1H NMR (DMSO-d 6, 400 MHz) δ 0.81 (3H, d), 1.43 (1H, q), 1.92 (2H, d), 2.19 (1H, br s), 2.80 (1H, br s), 3.14 (4H, t), 3.62 (3H, s), 3.72 (3H, s), 3.78 (4H, t), 3.89-3.97 (2H, m), 4.06-4.14 (1H, m), 4.31-4.41 (1H, m), 6.87 (1H, dd), 7.15 (1H, d), 7.38 (1H, d), 8.27 (1H, d), 8.35 (1H, s), 8.81 (1H, d), 12.37 (1H, s). Example A27 (11 R)-16-[3-(dimethylamino)pyrrolidin-1-yl]-5,11,26-trimethyl-7-oxa-4,5,13,20,22,26-hexaazapentacyclic[22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]twenty-eight-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione Step 1. (11 R)-16-[3-(dimethylamino)pyrrolidin-1-yl]-5,11,26-trimethyl-7-oxa-4,5,13,20,22,26-hexaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octacosa-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (Example A27). Under nitrogen atmosphere at room temperature, (11R)-16-bromo-5,11,26-trimethyl-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]octadecene-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (M3, 40 mg, 0.07 mmol, 1 equivalent), N,N-dimethylpyrrolidin-3-amine (34 mg, 0.30 mmol, 4 equiv) and BrettPhos Pd G 3To a stirred mixture of 2,4-dioxane (5 mL) and 1,4-dioxane (20 mg, 0.02 mmol, 0.3 eq.) was added LiHMDS (0.22 mL, 0.22 mmol, 3 eq.) dropwise. The reaction mixture was stirred at 60 °C for 30 min. After cooling to room temperature, the reaction was heated with saturated NH4Cl solution (30 mL), and the mixture was extracted with dichloromethane (3×20 mL). The combined organic layers were washed with brine (3×30 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by prep-TLC using dichloromethane/methanol (15/1) for elution, and the product was purified by Prep-HPLC using an XBridge Prep OBD C18 column with 10 mmol/L NH4HCO 3The product was further purified using water and acetonitrile as the mobile phase. The fractions containing the desired compound were evaporated to dryness to give 5 mg (11 R)-16-[3-(dimethylamino)pyrrolidin-1-yl]-5,11,26-trimethyl-7-oxa-4,5,13,20,22,26-hexaazapentacyclic[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octadecene-1(28),2(6),3, 14,16,18,20,24-octaene-23,27-dione (Example A27) (13%). LCMS: m/z (ESI), [M+H] +=559.50. 1H NMR (DMSO-d 6, 400 MHz) δ 0.81 (3H, d), 1.40-1.50 (1H, m), 1.88-1.95 (3H, m), 2.12-2.31 (8H, m), 2.79-2.84 (2H, m), 3.06-3.17 (1H, m), 3.23-3.49 (3H, m), 3.62 (3H, s), 3.72 (3H, s), 3.84-3.97 (2H, m), 4.06-4.15 (1H, m), 4.32-4.41 (1H, m), 6.47 (1H, d), 6.66 (1H, s), 7.33 (1H, d), 8.26 (1H, d), 8.36 (1H, s), 8.81 (1H, d), 12.28 (1H, s). Using similar conditions as described above for the synthesis of Example A27, the following Examples were prepared from M3: Example A28, Example A29, Example A30, Example A31, Example A32, Example A33, Example A34, Example A35, Example A36, Example A37, Example A38, Example A29, Example A40, Example A41, Example A42, Example A43, Example A44, Example A45, Example A46, Example A47, Example A48, Example A49, Example A50, Example A51, Example A52, Example A53, Example A54, Example A55, Example A56, Example A57, and Example A58. Structure and m/z and 1The H NMR data are listed in Table 1. Example A59, Example A60A and Example A60B 6-Bromo-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontahedral-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione (Example A59), 15,21-Dimethyl-6-(4-methylpiperazin-1-yl)-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]tris-3,5,7,9,13,17(33), 18(22),19-octaene-12,16-dione, isomer 1 (Example A60A) and isomer 2 (Example A60B) Step 1. Preparation of 1-[(4-bromo-2-nitrophenyl)amino]-3-azabicyclo[3.2.1]octane-3-carboxylic acid tert-butyl ester (INT-A60-1). 1-Amino-3-azabicyclo[3.2.1]octane-3-carboxylic acid tert-butyl ester (2.0 g, 8.80 mmol, 1 equivalent), K 2CO 3A mixture of (2.44 g, 17.6 mmol, 2 equiv.) and 4-bromo-1-fluoro-2-nitrobenzene (3.88 g, 17.6 mmol, 2 equiv.) in DMSO (40 mL) was stirred at 120 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (200 mL), and the mixture was extracted with ethyl acetate (3×200 mL). The combined organic layers were washed with brine (3×200 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue is purified by C18 column using water and acetonitrile as mobile phase. The fractions containing the desired compound are evaporated to dryness to give 800 mg of tert-butyl 1-[(4-bromo-2-nitrophenyl)amino]-3-azabicyclo[3.2.1]octane-3-carboxylate (INT-A60-1) (21%) as a yellow oil. LCMS: m/z (ESI), [M+H] +=372.05. Step 2. N-Preparation of (4-bromo-2-nitrophenyl)-3-azabicyclo[3.2.1]octan-1-amine (INT-A60-2). A mixture of tert-butyl 1-[(4-bromo-2-nitrophenyl)amino]-3-azabicyclo[3.2.1]octane-3-carboxylate (550 mg, 1.29 mmol, 1 equivalent) and trifluoroacetic acid (2 mL) in dichloromethane (6 mL) was stirred at room temperature for 2 h under nitrogen atmosphere and saturated with NaHCO 3(aqueous) solution (50 mL) and treated with CH 2Cl 2(3×50 mL) extraction. The combined organic layers were washed with brine (3×30 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure to give 435 mg of a yellow solid.N-(4-bromo-2-nitrophenyl)-3-azabicyclo[3.2.1]octan-1-amine (INT-A60-2) (100%). LCMS: m/z (ESI), [M+H] +=328.00. Step 3. N-(4-bromo-2-nitrophenyl)-3-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-3-azabicyclo[3.2.1]octan-1-amine (INT-A60-3) preparation.N-(4-Bromo-2-nitrophenyl)-3-azabicyclo[3.2.1]octan-1-amine (435 mg, 1.34 mmol, 1 equivalent), (2-bromoethoxy)(tert-butyl)dimethylsilane (652 mg, 2.72 mmol, 2 equivalents), NaI (408 mg, 2.72 mmol, 2 equivalents) and K 2CO 3(377 mg, 2.72 mmol, 2 equiv) in DMF (5 mL) was stirred at 60 °C for 4 h under nitrogen atmosphere. The mixture was cooled to room temperature, treated with water (50 mL), and extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by C18 column using water and acetonitrile as mobile phase. The fractions containing the desired compound were evaporated to dryness to give 360 mg of N-(4-bromo-2-nitrophenyl)-3-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-3-azabicyclo[3.2.1]octan-1-amine (INT-A60-3) (54%). LCMS: m/z (ESI), [M+H] +=485.90. Step 4. Preparation of 2-{1-[(4-bromo-2-nitrophenyl)amino]-3-azabicyclo[3.2.1]octan-3-yl}ethanol (INT-A60-4). NA mixture of -(4-bromo-2-nitrophenyl)-3-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-3-azabicyclo[3.2.1]octan-1-amine (360 mg, 0.74 mmol, 1 eq) and tetrabutylammonium fluoride (388 mg, 1.48 mmol, 2 eq) in tetrahydrofuran (5 mL) was stirred at room temperature overnight under a nitrogen atmosphere. The mixture was concentrated under reduced pressure. The residue was loaded onto a silica gel column and eluted with dichloromethane/methanol (10/1) to give 260 mg of 2-{1-[(4-bromo-2-nitrophenyl)amino]-3-azabicyclo[3.2.1]octan-3-yl}ethanol (INT-A60-4) (94%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=370.10. 1H NMR (DMSO-d 6, 400 MHz) δ 1.61-1.71 (3H, m), 1.79-1.83 (1H, m), 1.91-1.97 (1H, m), 2.11-2.17 (1H, m), 2.18-2.24 (1H, m), 2.30-2.49 (5H, m), 3.11-3.18 (1H, m), 3.45-3.55 (2H, m), 4.34 (1H, t), 7.17(1H, d), 7.57-7.67 (1H, m), 8.18 (1H, d), 8.29 (1H, s). Step 5. Preparation of methyl 5-[5-(2-{1-[(4-bromo-2-nitrophenyl)amino]-3-azabicyclo[3.2.1]oct-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylate (INT-A60-5). To a stirred mixture of methyl 5-(5-hydroxy-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (M2, 260 mg, 1.25 mmol, 1.97 equiv) and triphenylphosphine (782 mg, 2.98 mmol, 4.70 equiv) in tetrahydrofuran (5 mL) under nitrogen atmosphere at 0° C. was added 2-{1-[(4-bromo-2-nitrophenyl)amino]-3-azabicyclo[3.2.1]octan-3-yl}ethanol (235 mg, 0.63 mmol, 1 equiv) and diisopropyl azodicarboxylate (451 mg, 2.23 mmol, 3.5 equiv). The mixture was stirred at 0° C. for 2 h, treated with water (50 mL), and extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×50 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue is purified by C18 column using water and acetonitrile as mobile phase. The fractions containing the desired compound are evaporated to give 260 mg of 5-[5-(2-{1-[(4-bromo-2-nitrophenyl)amino]-3-azabicyclo[3.2.1]octan-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A60-5) (66%) as an orange oil. LCMS: m/z (ESI), [M+H] +=615.15. 1H NMR (DMSO-d 6, 400 MHz) δ 1.47-1.63 (2H, m), 1.66-1.70 (1H, m), 1.72-1.85 (1H, m), 1.88-1.94 (1H, m), 2.04-2.23 (4H, m), 2.66-2.76 (2H, m), 2.80 -2.89 (1H, m), 3.07-3.11 (1H, m), 3.58 (3H, s),3.73 (3H, s), 3.78 (3H, s), 4.10 (1H, s), 4.11 (1H, s), 6.94 (1H, d), 7.56-7.66 (1H, m), 8.02 (1H, s), 8.12 (1H, d), 8.17 (1H, d), 8.26 (1H, s), 8.43 (1H, d). Step 6. Preparation of 5-(5-(2-(1-((2-amino-4-bromophenyl)amino)-3-azabicyclo[3.2.1]oct-3-yl)ethoxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxo-1,6-dihydropyridine-3-carboxylic acid methyl ester (INT-A60-6). 5-[5-(2-{1-[(4-bromo-2-nitrophenyl)amino]-3-azabicyclo[3.2.1]oct-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (240 mg, 0.39 mmol, 1.00 equivalent), NH 2NH 2 .H 2A mixture of O (39 mg, 0.78 mmol, 2 eq.) and Raney nickel (33 mg, 0.39 mmol, 1 eq.) in methanol (2 mL) was stirred at 0 °C for 2 h under nitrogen atmosphere. The mixture was filtered. The filter cake was washed with methanol (2×50 mL). The resulting solution was concentrated under reduced pressure. The residue was loaded onto a silica gel column and eluted with dichloromethane/methanol (10/1) to give 200 mg of 5-(5-(2-(1-((2-amino-4-bromophenyl)amino)-3-azabicyclo[3.2.1]octan-3-yl)ethoxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxo-1,6-dihydropyridine-3-carboxylic acid methyl ester (INT-A60-6) (84%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=585.10. 1H NMR (DMSO-d 6, 400 MHz) δ 1.43-1.51 (4H, m), 1.63 -1.75 (1H, m), 1.77-1.81 (1H, m), 1.87-1.91 (1H, m), 1.95-1.99 (1H, m), 2.01-2.05 (1H, m), 2.09-2.13 (1H, m), 2.67-2.77 (3H, m), 3.03-3.09 (1H, m), 3.58 (3H, s), 3.71(3H, s), 3.79 (3H, s), 3.97-4.14 (2H, m), 4.87 (2H, s), 6.38 (1H, d), 6.46-6.56 (1H, m), 6.66 (1H, d), 8.00 (1H, s), 8.13 (1H, d), 8.45 (1H, d). Step 7. Preparation of 5-(5-{2-[1-(2-amino-5-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A60-7). A mixture of 5-(5-{2-[1-(2-amino-5-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]octan-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (200 mg, 0.32 mmol, 1 eq.) and BrCN (38 mg, 0.36 mmol, 1.1 eq.) in ethanol (2 mL) was stirred at 60 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature, treated with water (50 mL), and extracted with dichloromethane (2×50 mL). The combined organic layers were washed with brine (3×30 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC eluting with (dichloromethane/methanol 12/1) to give 150 mg of 5-(5-{2-[1-(2-amino-5-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]octan-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A60-7) (75%) as a brown solid. LCMS: m/z (ESI), [M+H] +=611.90. 1H NMR (DMSO-d 6, 400 MHz) δ 1.04-1.07 (1H, m), 1.22-1.26 (1H, m), 1.48-1.53 (1H, m), 1.71-1.84 (2H, m), 2.20-2.24 (2H, m), 2.27-2.33 (2H, m), 2.62 -2.76 (2H, m), 2.76 -2.84 (1H, m), 3.02 -3.08 (1H, m), 3.58 (3H, s), 3.73 (3H, s), 3.77 (3H, s), 4.09-4.13 (2H, m), 6.08 (2H, s), 6.93-6.97 (1H, m), 7.19 (1H, d), 7.25 (1H, d), 8.01 (1H, s), 8.11 (1H, d), 8.42 (1H, d). Step 8. Preparation of 5-(5-{2-[1-(2-amino-5-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A60-8). 5-(5-{2-[1-(2-amino-5-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (140 mg, 0.22 mmol, 1 equivalent) and LiOH .H 2O (48 mg, 1.14 mmol, 5 equivalents) in tetrahydrofuran/H 2O (2 mL/0.5 mL) was stirred at 60 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature and concentrated. The residue was purified by C18 column using water and acetonitrile as mobile phase. The fractions containing the desired compound were evaporated to dryness to give 100 mg of 5-(5-{2-[1-(2-amino-5-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A60-8) (73%) as a white solid. LCMS: m/z (ESI), [M+H] +=595.95. 1H NMR (DMSO-d 6, 400 MHz) δ 1.54 (1H, m), 1.73-1.79 (2H, m), 2.15-2.21 (1H, m), 2.28-2.34 (2H, m), 2.63-2.69 (3H, m), 2.72-2.78 (1H, m), 2.85-2.91 (2H, m), 3.05-3.11 (1H, m), 3.49 (3H, s), 3.68 (3H, s), 4.01-4.19 (2H, m), 6.29 (2H, s), 6.90-6.99 (1H, m), 7.22 (1H, d), 7.27 (1H, d), 7.95 (2H, d), 8.24 (1H, d). Step 9. Preparation of 6-bromo-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontria-3,5,7,9,13,17(33), 18(22),19-octaene-12,16-dione (Example A59). 5-(5-{2-[1-(2-amino-5-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (90 mg, 0.15 mmol, 1 equivalent), N,N-Diisopropylethylamine (58 mg, 0.45 mmol, 3 equivalents) and N,N,N,N-Tetramethyl- OA mixture of -(7-azabenzotriazol-1-yl)urea hexafluorophosphate (114 mg, 0.30 mmol, 2 equiv.) and 1,4-dioxane (2 mL) was stirred at 60 °C for 30 min under a nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (30 mL), and the mixture was extracted with dichloromethane (2×30 mL). The combined organic layers were washed with brine (3×30 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (12/1) to give 80 mg of 6-bromo-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontahedral-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione (Example A59) (91%) as a white solid. LCMS: m/z (ESI), [M+H] +=580.20. 1H NMR (DMSO-d 6, 400 MHz) δ δ 1.53-1.77 (3H, m), 2.00 (1H, d), 2.39-2.63 (3H, m), 2.64-2.77 (2H, m), 3.16-3.28 (1H, m), 3.29-3.39 (2H, m), 3.62 (3H, s), 3.63-3.69 (1H, m), 3.72 (3H, s), 4.45-4.70 (2H, m), 7.28 (1H, d), 7.72 (1H, d), 8.02 (1H, s), 8.39-8.43 (2H, m), 8.78 (1H, s), 12.81 (1H, s). Step 10. Preparation of 15,21-dimethyl-6-(4-methylpiperazin-1-yl)-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontahedral-3,5,7,9,13,17(33), 18(22),19-octaene-12,16-dione (A60). 6-Bromo-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriaconta-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione (75 mg, 0.13 mmol, 1 eq.), 1-methylpiperazine (38 mg, 0.39 mmol, 3 eq.) and BrettPhos Pd G 3To a stirred mixture of 1,4-dioxane (3 mL) and 2-nitropropene (35 mg, 0.03 mmol, 0.3 equiv.) was added LiHMDS (1.3 mL, 1.30 mmol, 10 equiv.). The resulting mixture was stirred at 60 °C under nitrogen atmosphere for 2 h and cooled to room temperature. The reaction was heated with saturated NH4Cl(aq) solution (50 mL) was quenched, and the mixture was extracted with dichloromethane (2×50 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4The residue was purified by Prep-TLC using dichloromethane/methanol (10/1) to give 22.3 mg of 15,21-dimethyl-6-(4-methylpiperazin-1-yl)-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontria-3,5,7,9,13, 17(33),18(22),19-octaene-12,16-dione (A60) as a yellow solid (28%). LCMS: m/z (ESI), [M+H] +=598.35. 1H NMR (DMSO-d 6, 400 MHz) δ 1.50-1.80 (3H, m), 1.97 (1H, d), 2.26-2.58 (10H, m), 2.55-2.80 (3H, m), 3.03-3.43 (6H, m), 3.63 (3H, s), 3.64-3.70 (1H, m), 3.72 (3H, s), 4.57 (1H, t), 4.69 (1H, d), 6.86 (1H, dd), 7.17 (1H, s), 7.57 (1H, d), 8.29 (1H, d), 8.42 (1H, s), 8.83 (1H, d), 12.56 (1H, s). Step 11. Preparation of 15,21-dimethyl-6-(4-methylpiperazin-1-yl)-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontahedral-3,5,7,9,13,17(33), 18(22),19-octaene-12,16-dione, isomer 1 (Example A60A) and isomer 2 (Example A60B). A racemic mixture of 15,21-dimethyl-6-(4-methylpiperazin-1-yl)-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tris-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione (16.9 mg, 0.02 mmol) was purified by Pre-SFC using a chiral ART cellulose-SB column (2*25 cm, 5 μm) with tert-butyl methyl ether containing 0.1% triethylamine as mobile phase A and ethanol/dichloromethane (1: 1) as mobile phase B to separate to obtain 1 mg of isomer 1 (5%) and 1 mg of isomer 2 (5%) as yellow solid. Isomer 1: LCMS: m/z (ESI), [M+H] +=598.40. 1H NMR (DMSO-d 6, 400 MHz) δ 1.50-1.80 (3H, m), 1.97 (1H, d), 2.26-2.58 (10H, m), 2.55-2.80 (3H, m), 3.03-3.43 (6H, m), 3.63 (3H, s), 3.64-3.70 (1H, m), 3.72 (3H, s), 4.55 (1H, t), 4.69 (1H, d), 6.84 (1H, dd), 7.16 (1H, s), 7.55 (1H, d), 8.28 (1H, d), 8.41 (1H, s), 8.83 (1H, d), 12.54 (1H, s). Chiral-HPLC, Rt=3.374 minutes Isomer 2: LCMS: m/z (ESI), [M+H] +=598.35. 1H NMR (DMSO-d 6, 400 MHz) δ 1.50-1.80 (3H, m), 1.97 (1H, d), 2.26-2.58 (10H, m), 2.55-2.80 (3H, m), 3.03-3.43 (6H, m), 3.63 (3H, s), 3.64-3.70 (1H, m), 3.72 (3H, s), 4.55 (1H, t), 4.69 (1H, d), 6.85 (1H, dd), 7.16 (1H, s), 7.56 (1H, d), 8.28 (1H, d), 8.41 (1H, s), 8.83 (1H, d), 12.55 (1H, s). Chiral-HPLC, Rt = 3.923 minutes Example A61A and Example A61B 15,21-Dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontahedral-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione, isomer 1 (Example A61A) and isomer 2 (Example A61B) Step 1. Preparation of tert-butyl 1-[(2-nitrophenyl)amino]-3-azabicyclo[3.2.1]octane-3-carboxylate (INT-A61-1). tert-butyl 1-amino-3-azabicyclo[3.2.1]octane-3-carboxylate (500 mg, 2.21 mmol, 1 equivalent), o-fluoronitrobenzene (312 mg, 2.20 mmol, 1 equivalent) and K 2CO 3A mixture of (610.66 mg, 4.42 mmol, 2 equiv.) in dimethyl sulfoxide (10 mL) was stirred at 120 °C overnight under nitrogen atmosphere and cooled to room temperature. The reaction was quenched with water (50 mL), and the mixture was extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using petroleum ether/ethyl acetate (5/1) to give 600 mg of tert-butyl 1-[(2-nitrophenyl)amino]-3-azabicyclo[3.2.1]octane-3-carboxylate (INT-A61-1) (78%) as an orange oil. LCMS: m/z (ESI), [M+H] += 291.95. Step 2. N-Preparation of (2-nitrophenyl)-3-azabicyclo[3.2.1]octan-1-amine (INT-A61-2). A mixture of tert-butyl 1-[(2-nitrophenyl)amino]-3-azabicyclo[3.2.1]octane-3-carboxylate (550 mg, 1.58 mmol, 1 equiv) in trifluoroacetic acid (2 mL) and dichloromethane (6 mL) was stirred at room temperature under nitrogen atmosphere for 2 h. The mixture was treated with water (50 mL) and saturated NaHCO3(aqueous solution). The mixture was extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×50 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure to give 380 mg of orange oil.N-(2-Nitrophenyl)-3-azabicyclo[3.2.1]octan-1-amine (INT-A61-2). This material was used in the next reaction without further purification. LCMS: m/z (ESI), [M+H] +=248.00. Step 3. 3-{2-[(tert-butyldimethylsilyl)oxy]ethyl}- N-(2-nitrophenyl)-3-nitrobicyclo[3.2.1]octan-1-amine (INT-A61-3).N-(2-Nitrophenyl)-3-azabicyclo[3.2.1]octan-1-amine (380 mg, 1.54 mmol, 1 equivalent), (2-bromoethoxy)(tert-butyl)dimethylsilane (735 mg, 3.07 mmol, 2 equivalents) and K 2CO 3(425 mg, 3.07 mmol, 2 equivalents) in N,N-The mixture in dimethylformamide (5 mL) was stirred at 60 °C for 2 h under nitrogen atmosphere and cooled to room temperature. The reaction was quenched with water (50 mL), and the mixture was extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using petroleum ether/ethyl acetate (5/1) to give 450 mg of 3-{2-[(tert-butyldimethylsilyl)oxy]ethyl}- N-(2-Nitrophenyl)-3-azabicyclo[3.2.1]octan-1-amine (INT-A61-3) (72%). LCMS: m/z (ESI), [M+H] +=406.15. Step 4. Preparation of 2-{1-[(2-nitrophenyl)amino]-3-azabicyclo[3.2.1]oct-3-yl}ethanol (INT-A61-4). 3-{2-[(tert-butyldimethylsilyl)oxy]ethyl}- NA mixture of -(2-nitrophenyl)-3-azabicyclo[3.2.1]octan-1-amine (400 mg, 1 mmol, 1 equiv) and tetrabutylammonium fluoride (516 mg, 2 mmol, 2 equiv) in tetrahydrofuran (10 mL) was stirred at room temperature under nitrogen atmosphere for 2 h. The reaction was heated with saturated NaHCO3(aqueous solution) (100 mL) was quenched, and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure to give 280 mg of 2-{1-[(2-nitrophenyl)amino]-3-azabicyclo[3.2.1]octan-3-yl}ethanol (INT-A61-4) as an orange oil. This material was used in the next reaction without further purification. LCMS: m/z (ESI), [M+H] +=292.00. Step 5. Preparation of 1-methyl-5-[1-methyl-5-(2-{1-[(2-nitrophenyl)amino]-3-azabicyclo[3.2.1]oct-3-yl}ethoxy)pyrazol-4-yl]-6-oxopyridine-3-carboxylic acid methyl ester (INT-A61-5). To a stirred mixture of 2-{1-[(2-nitrophenyl)amino]-3-azabicyclo[3.2.1]octan-3-yl}ethanol (330 mg, 1.13 mmol, 1 eq), diisopropyl azodicarboxylate (687 mg, 3.40 mmol, 3 eq) and triphenylphosphine (891 mg, 3.40 mmol, 3 eq) in tetrahydrofuran (10 mL) under nitrogen atmosphere was added methyl 5-(5-hydroxy-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (M2, 298 mg, 1.13 mmol, 1 eq). After stirring at 0 °C under nitrogen atmosphere for 2 h, the reaction was quenched with water (100 mL) and the mixture was extracted with dichloromethane (3 x 100 mL). The combined organic layers were washed with brine (3×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using petroleum ether/ethyl acetate (1/1) to give 400 mg of 1-methyl-5-[1-methyl-5-(2-{1-[(2-nitrophenyl)amino]-3-azabicyclo[3.2.1]octan-3-yl}ethoxy)pyrazol-4-yl]-6-oxopyridine-3-carboxylic acid methyl ester (INT-A61-5) (66%) as an orange solid. Step 6. Preparation of 5-[5-(2-{1-[(2-aminophenyl)amino]-3-azabicyclo[3.2.1]oct-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A61-6). To a stirred mixture of 1-methyl-5-[1-methyl-5-(2-{1-[(2-nitrophenyl)amino]-3-azabicyclo[3.2.1]oct-3-yl}ethoxy)pyrazol-4-yl]-6-oxopyridine-3-carboxylic acid methyl ester (340 mg, 0.634 mmol, 1 eq.) and ranyl nickel (54.29 mg, 0.634 mmol, 1.0 eq.) in methanol (20 mL) at 0°C was added NH 2NH 2 .H 2O (63.44 mg, 1.268 mmol, 2.0 equiv). The mixture was stirred at room temperature under nitrogen atmosphere for 1 h and filtered. The filter cake was washed with methanol (3×10 mL). The resulting solution was concentrated under reduced pressure. The residue was purified by Prep-TLC eluting with dichloromethane/methanol (12/1) to give 240 mg of 5-[5-(2-{1-[(2-aminophenyl)amino]-3-azabicyclo[3.2.1]octan-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A61-6) (63%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=507.25. Step 7. Preparation of 5-(5-{2-[1-(2-amino-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A61-7). A mixture of methyl 5-[5-(2-{1-[(2-aminophenyl)amino]-3-azabicyclo[3.2.1]octan-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylate (300 mg, 0.59 mmol, 1 eq) and BrCN (94 mg, 0.89 mmol, 1.5 eq) in ethanol (5 mL) was stirred at room temperature overnight under nitrogen atmosphere. The mixture was concentrated under reduced pressure, and the residue was purified by Prep-TLC using dichloromethane/methanol (12/1) to give 200 mg of 5-(5-{2-[1-(2-amino-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A61-7) (64%) as a brownish yellow solid. LCMS: m/z (ESI), [M+H] +=532.25. Step 8. Preparation of 5-(5-(2-(-1-(2-amino-1H-benzo[d]imidazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl)ethoxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxo-1,6-dihydropyridine-3-carboxylic acid methyl ester, isomer 1 (INT-A61-7A) and isomer 2 (INT-A61-7B). A racemic mixture of methyl 5-(5-{2-[1-(2-amino-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]octan-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (200 mg) was separated using an SFC DZ-CHIRALPAK IC-3 column (4.6*50 mm, 3.0 μm), tert-butyl methyl ether containing 0.2% diethylamine as mobile phase A and ethanol/dichloromethane (1/1) as mobile phase B to obtain 70 mg of isomer 1 (35%) and 70 mg of isomer 2 (35%) as yellow solids (35%). Step 9. Preparation of 5-(5-{2-[1-(2-amino-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A61-8A). 5-(5-{2-[1-(2-amino-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A61-7A, 70 mg, 0.13 mmol, 1 equivalent) and LiOH .H 2A solution of O (22 mg, 0.53 mmol, 4 equiv.) in water (1 mL) and tetrahydrofuran (4 mL) was stirred at room temperature overnight under nitrogen atmosphere. The mixture was concentrated under reduced pressure. The residue was purified by prep-HPLC on an XBridge Shield RP18 OBD column using a column containing 10 mmol/L NH4HCO 3and 0.1% NH 3 .H 2O water and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 45 mg of 5-(5-{2-[1-(2-amino-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A61-8A) (70%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=518.10. Step 10. Preparation of 5,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]tris-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione, isomer 1 (Example A61A). 5-(5-{2-[1-(2-amino-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (45 mg, 0.087 mmol, 1 equivalent), N,N-Diisopropylethylamine (33.9, 0.261 mmol, 3 equivalents) and N,N,N,N-Tetramethyl- OA mixture of -(7-azabenzotriazol-1-yl)uronium hexafluorophosphate (66 mg, 0.17 mmol, 2 eq.) in dioxane (2 mL) was stirred at room temperature overnight under nitrogen atmosphere. After the mixture was concentrated under reduced pressure, the residue was purified by Prep-TLC using dichloromethane/methanol (12/1) to give 26.8 mg of 15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontahedral-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione, isomer 1 (Example A61A) as an off-white solid (61%). LCMS: m/z (ESI), [M+H] += 500.15. 1H NMR (DMSO- d 6, 400 MHz) δ 1.53-1.77 (3H, m), 2.00 (1H, d), 2.39-2.63 (3H, m), 2.64-2.77 (2H, m), 3.16-3.28 (1H, m), 3.29-3.39 (2H, m), 3.62 (3H, s), 3.63-3.69 (1H, m), 3.72 (3H, s), 4.55 (1H, t), 4.68 (1H, d), 7.12-7.23 (2H, m), 7.55-7.60 (1H, m), 7.69-7.75 (1H, m), 8.29 (1H, d), 8.41 (1H, d), 8.83 (1H, d), 12.76 (1H, s). Step 11. Preparation of 5-(5-{2-[1-(2-amino-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A61-8B). 5-(5-{2-[1-(2-amino-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A61-7B, 70 mg, 0.13 mmol, 1 equiv.) and LiOH .H 2A mixture of O (22 mg, 0.53 mmol, 4 equiv.) in water (1 mL) and tetrahydrofuran (4 mL) was stirred at room temperature overnight under a nitrogen atmosphere. After the mixture was concentrated under reduced pressure, the residue was subjected to prep-HPLC using an XBridge Shield RP18 OBD column with 10 mmol/L NH4HCO 3and 0.1% NH 3.H 2O water and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 45 mg of 5-(5-{2-[-1-(2-amino-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A61-8B) (70%) as an off-white solid. LCMS: m/z (ESI), [M+H] += 518.15. Step 12. Preparation of 15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]tris-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione (isomer 2 Example A61B). 5-(5-{2-[1-(2-amino-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (45 mg, 0.087 mmol, 1 equivalent), N,N-Diisopropylethylamine (33.9, 0.261 mmol, 3 equivalents) and N,N,N,N-Tetramethyl- OA mixture of -(7-azabenzotriazol-1-yl)uronium hexafluorophosphate (66 mg, 0.17 mmol, 2 eq.) in dioxane (2 mL) was stirred at room temperature overnight under nitrogen atmosphere. After the mixture was concentrated under reduced pressure, the residue was purified by Prep-TLC using dichloromethane/methanol (12/1) to give 25.8 mg of 15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontahedral-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione, isomer 2 (Example A61B) as an off-white solid (57%). LCMS: m/z (ESI), [M+H] += 500.15. 1H NMR (DMSO- d 6, 400 MHz) δ 1.53-1.77 (3H, m), 2.00 (1H, d), 2.39-2.63 (3H, m), 2.64-2.77 (2H, m), 3.16-3.28 (1H, m), 3.29-3.39 (2H, m), 3.62 (3H, s), 3.63-3.69 (1H, m), 3.72 (3H, s), 4.55 (1H, t), 4.68 (1H, d), 7.12-7.23 (2H, m), 7.55-7.60 (1H, m), 7.69-7.75 (1H, m), 8.29 (1H, d), 8.41 (1H, d), 8.83 (1H, d), 12.76 (1H, s). Example A62A 15,21-Dimethyl-5-(morpholin-4-yl)-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]tritriacontahedral-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione, isomer 1 (Example A62A) Step 1. Preparation of 15,21-dimethyl-5-(morpholin-4-yl)-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontahedral-3,5,7,9,13,17(33), 18(22),19-octaene-12,16-dione, Isomer 1 (Example A62A). 5-Bromo-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]tris-3,5,7,9,13,17(33), 18(22),19-octaene-12,16-dione (M4A, 50 mg, 0.09 mmol, 1 eq.), morpholine (30 mg, 0.34 mmol, 4 eq.) and BrettPhos Pd G 3To a stirred mixture of 2-nitropropene (23 mg, 0.03 mmol, 0.3 equiv) in dioxane (10 mL) was added LiHMDS (0.34 mL, 0.34 mmol, 4 equiv) dropwise. The reaction mixture was stirred at 60 °C for 30 min. After cooling to room temperature, the reaction was heated with saturated NH4Cl solution (30 mL), and the mixture was extracted with dichloromethane (3×20 mL). The combined organic layers were washed with brine (3×30 mL) and purified by anhydrous Na 2SO 4The residue was purified by Prep-TLC using dichloromethane/methanol (20/1) to give 22 mg of 15,21-dimethyl-5-(morpholin-4-yl)-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontria-3,5,7,9,13, 17(33),18(22),19-octaene-12,16-dione, Isomer 1 (Example A62A) as a white solid (43%). LCMS: m/z (ESI), [M+H] +=585.35. 1H NMR (DMSO-d 6, 400 MHz) δ 1.53-1.82 (3H, m), 2.02 (1H, d), 2.39-2.61 (3H, m), 2.65-2.77 (3H, m), 3.10 (4H, q), 3.17-3.43 (2H, m), 3.62 (3H, s), 3.63-3.69 (1H, m), 3.72 (3H, s), 3.77 (4H, t), 4.54 (1H, t), 4.69 (1H, d), 6.91 (1H, dd), 7.20 (1H, s), 7.44 (1H, d), 8.28 (1H, d), 8.42 (1H, s), 8.83 (1H, d), 12.57 (1H, s). Using similar reaction conditions described above for the synthesis of Example A62A, the following examples were prepared from intermediate M4A: Example A63A, Example A64A, Example A65A, Example A66A, Example A66C, Example A67A, Example A68A, Example A69A, Example A70A, Example A71A, Example A72A, Example A73A, Example A76A, Example A77A, Example A78A, Example A79A, Example A81A, Example A82A, Example Example A83A, Example A85A, Example A86A, Example A87A, Example A88A, Example A89A, Example A90A, Example A91A, Example A92A, Example A95A, Example A97A, Example A98A, Example A99A, Example A104A, Example A105A, Example A106A, Example A107A, Example A108A. Example A109A. Structure and m/z and 1The H NMR data are listed in Table 1. Example A62B 15,21-Dimethyl-5-(morpholin-4-yl)-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]tritriacontahedral-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione, isomer 2 (Example A62B) Step 1. Preparation of 15,21-dimethyl-5-(morpholin-4-yl)-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontahedral-3,5,7,9,13,17(33), 18(22),19-octaene-12,16-dione, isomer 2 (Example A62B). 5-Bromo-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]tritriacontria-3,5,7,9,13,17(33), 18(22),19-octaene-12,16-dione (M4B, 50 mg, 0.09 mmol, 1 eq.), morpholine (30 mg, 0.34 mmol, 4 eq.) and BrettPhos Pd G 3To a stirred mixture of 2-nitropropene (23 mg, 0.03 mmol, 0.3 equiv) in dioxane (5 mL) was added LiHMDS (0.34 mL, 0.34 mmol, 4 equiv) dropwise. The reaction mixture was stirred at 60 °C for 30 min. After cooling to room temperature, the reaction was heated with saturated NH4Cl solution (30 mL), and the mixture was extracted with dichloromethane (3×20 mL). The combined organic layers were washed with brine (3×30 mL) and purified by anhydrous Na 2SO 4The residue was purified by Prep-TLC using dichloromethane/methanol (20/1) to give 6.4 mg of 15,21-dimethyl-5-(morpholin-4-yl)-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontria-3,5,7,9,13, 17(33),18(22),19-octaene-12,16-dione, Isomer 2 (Example A62B) as a white solid (12%). LCMS: m/z (ESI), [M+H] +=585.35. 1H NMR (DMSO-d 6, 400 MHz) δ 1.53-1.82 (3H, m), 2.02 (1H, d), 2.39-2.61 (3H, m), 2.65-2.77 (3H, m), 3.10 (4H, q), 3.17-3.43 (2H, m), 3.62 (3H, s), 3.63-3.69 (1H, m), 3.72 (3H, s), 3.77 (4H, t), 4.54 (1H, t), 4.69 (1H, d), 6.91 (1H, dd), 7.20 (1H, s), 7.44 (1H, d), 8.28 (1H, d), 8.42 (1H, s), 8.83 (1H, d), 12.57 (1H, s). Using similar reaction conditions described above for the synthesis of Example A62B, the following examples were prepared from intermediate M4B: Example A63B, Example A64B, Example A65B, Example A66B, Example A66D, Example A67B, Example A68B, Example A69B, Example A70B, Example A71B, Example A72B, Example A73B, Example A76B, Example A77B, Example A78B, Example A79B, Example A81B, Example A82B, Example Example A83B, Example A85B, Example A86B, Example A87B, Example A88B, Example A89B, Example A90B, Example A91B, Example A92B, Example A95B, Example A97B, Example A98B, Example A99B, Example A104B, Example A105B, Example A106B, Example A107B, Example A108B. Example A109B. Structure and m/z and 1The H NMR data are listed in Table 1. Example A74 15,21-Dimethyl-5-(morpholin-4-yl)-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]triacontria-3,5,7,9,13,17(34),18(22),19-octaene-12,16-dione Step 1. N-(5-bromo-2-nitrophenyl)-3-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-3-azabicyclo[3.2.2]nonan-1-amine (INT-A74-1). 3-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-3-azabicyclo[3.2.2]nonan-1-amine (1 g, 3.35 mmol, 1 equivalent), 4-bromo-2-fluoro-1-nitrobenzene (1.84 g, 8.37 mmol, 2.5 equivalents) and K 2CO 3A mixture of (1.39 g, 10.05 mmol, 3 equiv.) in dimethyl sulfoxide (10 mL) was stirred at 120 °C for 4 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (50 mL), and the mixture was extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue is purified by silica gel chromatography eluting with petroleum ether/ethyl acetate (5/1) to give 1.5 g of yellow oil.N-(5-bromo-2-nitrophenyl)-3-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-3-azabicyclo[3.2.2]nonan-1-amine (INT-A74-1) (85%). LCMS: m/z (ESI), [M+H]+=498.15. Step 2. Preparation of 2-{1-[(5-bromo-2-nitrophenyl)amino]-3-azabicyclo[3.2.2]nonan-3-yl}ethanol (INT-A74-2). NA solution of -(5-bromo-2-nitrophenyl)-3-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-3-azabicyclo[3.2.2]nonan-1-amine (1.5 g, 3.00 mmol, 1 eq.) and tetrabutylammonium fluoride (1.57 g, 6.01 mmol, 2 eq.) in tetrahydrofuran (15 mL) was stirred at room temperature for 2 h under nitrogen atmosphere. The reaction was heated with saturated NaHCO3(aqueous solution) (100 mL) was quenched, and the mixture was extracted with dichloromethane (3×100 mL). The combined organic layers were washed with brine (3×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column eluting with dichloromethane/methanol (10/1) to give 1.2 g of 2-{1-[(5-bromo-2-nitrophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethanol (INT-A74-2) (93%) as a yellow oil. LCMS: m/z (ESI), [M+H] +=384.00. Step 3. Preparation of 5-[5-(2-{1-[(5-bromo-2-nitrophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A74-3). To a stirred mixture of methyl 5-(5-hydroxy-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (M2, 0.66 g, 2.49 mmol, 0.8 eq.) and triphenylphosphine (2.46 g, 9.369 mmol, 3 eq.) in tetrahydrofuran (10 mL) were added diisopropyl azodicarboxylate (1.89 g, 9.36 mmol, 3 eq.) and 2-{1-[(5-bromo-2-nitrophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethanol (1.20 g, 3.11 mmol, 1 eq.) at 0° C. The mixture was stirred at 0° C. for 2 h under nitrogen atmosphere. The reaction was quenched with water (50 mL), and the mixture was extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC and eluted with petroleum ether/ethyl acetate (1/1) to give 1.2 g of 5-[5-(2-{1-[(5-bromo-2-nitrophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A74-3) (57%) as a yellow oil. LCMS: m/z (ESI), [M+H] +=629.25. 1H NMR (DMSO-d 6, 400 MHz) δ 1.58-1.76 (9H, m), 2.56-2.79 (2H, m), 2.90 (4H, q), 3.56 (3H, s), 3.72 (6H, d), 4.13 (2H, t), 6.70-6.85 (1H, m), 7.01 (1H, d), 7.95-8.12 (4H, m), 8.28 (1H, d). Step 4. Preparation of 5-[5-(2-{1-[(2-amino-5-bromophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A74-4). To a stirred mixture of ranyl nickel (0.90 g) and methyl 5-[5-(2-{1-[(5-bromo-2-nitrophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylate (1.1 g, 1.74 mmol, 1 equiv) in methanol (8 mL) at 0°C under nitrogen atmosphere was added NH2NH 2.H 2O (47 mg, 0.95 mmol, 2 eq.). The mixture was stirred at 0 °C for 1 h. The mixture was filtered and the filter cake was washed with methanol (2×20 mL). The resulting solution was concentrated under reduced pressure. The residue was purified by Prep-TLC and eluted with dichloromethane/methanol (10/1) to give 910 mg of 5-[5-(2-{1-[(2-amino-5-bromophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A74-4) (76%) as a light yellow solid. LCMS: m/z (ESI), [M+H] += 599.20. Step 5. Preparation of 5-(5-{2-[1-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.2]non-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A74-5). A mixture of methyl 5-[5-(2-{1-[(2-amino-5-bromophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylate (900 mg, 1.50 mmol, 1 eq) and BrCN (174 mg, 1.65 mmol, 1.1 eq) in ethanol (10 mL) was stirred under nitrogen atmosphere at room temperature for 2 h. The resulting mixture was concentrated under vacuum. The residue was purified by Prep-TLC and eluted with dichloromethane/methanol (12/1) to give 640 mg of 5-(5-{2-[1-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.2]non-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A74-5) (68%) as a light yellow solid. LCMS: m/z (ESI), [M+H] +=624.10. 1H NMR (DMSO-d 6, 400 MHz) δ 1.24 (1H, s), 1.78-1.95 (8H, m), 2.60-2.74 (2H, m), 2.85-2.95 (4H, m), 3.57 (3H, s), 3.71 (6H, d), 4.08-4.17 (2H, m), 5.95 (2H, s), 7.04 (2H, s), 7.47 (1H, s), 7.96 (1H, s), 8.12 (1H, ), 8.39 (1H, d). Step 6. 5-(5-{2-[1-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.2]non-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A74-6). 5-(5-{2-[1-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.2]non-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (620 mg, 0.99 mmol, 1 equiv) and LiOH (28 mg, 1.19 mmol, 1.2 equiv) were dissolved in tetrahydrofuran/H 2O (4 mL/1 mL) was stirred at 80 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature and concentrated under vacuum to give 550 mg of 5-(5-{2-[1-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.2]non-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A74-6) (90%) as a white solid. This material was used in the next reaction without purification. Step 7. Preparation of 5-bromo-15,21-dimethyl-23-oxa-2,9,11,15,20,21, 26-heptaazaheptacyclo[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontriacontria-3,5,7,9,13,17(34),18(22),19-octaene-12,16-dione (INT-A74-7). 5-(5-{2-[1-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.2]non-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (550 mg, 0.90 mmol, 1 equivalent), N,N,N,N-Tetramethyl-O-(7-azabenzotriazol-1-yl)urea hexafluorophosphate (1027 mg, 2.70 mmol, 3 equivalents) and N,N- A mixture of diisopropylethylamine (174 mg, 1.35 mmol, 1.5 equiv.) in dioxane (6 mL) was stirred at 60 °C for 2 h under a nitrogen atmosphere. The mixture was cooled to room temperature and concentrated under vacuum. The residue was purified by prep-HPLC using an XBridge Prep OBD C18 column with 10 mmol/L NH4HCO 3of water and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 500 mg of 5-bromo-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontriacontria-3,5,7,9,13,17(34),18(22),19-octaene-12,16-dione (INT-A74-7) (88%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=592.15. Step 8. Preparation of 15,21-dimethyl-5-(morpholin-4-yl)-23-oxo-2,9,11, 15,20,21,26-heptaazaheptacyclo[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontriacontria-3,5,7,9,13,17(34), 18(22),19-octaene-12,16-dione (Example A74). 5-Bromo-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic [26.2.2.1^{1,26}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]triacontria-3,5,7,9,13,17(34), 18(22),19-octaene-12,16-dione (130 mg, 0.21 mmol, 1 equiv), morpholine (57 mg, 0.65 mmol, 3 equiv) and BrettPhos Pd G 3To a stirred mixture of 1,4-dioxane (59 mg, 0.06 mmol, 0.3 eq.) and 1,4-dioxane (12 mL) was added LiHMDS (5.2 mL, 5.20 mmol, 6 eq.). The mixture was stirred at 60 °C for 1 h and cooled to room temperature. The reaction was heated with saturated NH4Cl(aq) (15 mL) was quenched and the mixture was washed with CH 2Cl 2(3×30 mL) extraction. The combined organic layers were purified by anhydrous Na 2SO 4The residue was purified by Prep-TLC and eluted with methylene dichloride/methanol (12/1) to give 38.3 mg of 15,21-dimethyl-5-(morpholin-4-yl)-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tetracontriacontria-3,5,7,9,13,17(34),18(22),19-octaene-12,16-dione (Example A74) as a white solid (28%). LCMS: m/z (ESI), [M+H] += 599.25. 1H NMR (DMSO-d 6, 400 MHz) δ 1.82-2.02 (4H, m), 2.02-2.12 (3H, m), 2.80 (2H, d), 2.87 (2H, t), 3.08 (4H, t), 3.38-3.52 (4H, m), 3.62 (3H, s), 3.75 (3H, s), 3.77-3.85 (4H, m), 4.28 (2H, t), 6.93 (1H, dd), 7.34 (1H, d), 7.49 (1H, d), 8.31 (2H, d), 9.12 (1H, d), 13.20 (1H, s). Example A75 15,21-Dimethyl-5-(4-methylpiperazin-1-yl)-23-oxo-2,9,11,15, 20,21,26-heptaazaheptacyclic [26.2.2.1^{1,26}.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]triacontria-3,5,7,9,13,17(34),18(22),19-octaene-12,16-dione Step 1. Preparation of 15,21-dimethyl-5-(4-methylpiperazin-1-yl)-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontria-3,5,7,9,13,17(34), 18(22),19-octaene-12,16-dione (Example A75). 5-Bromo-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic [26.2.2.1^{1,26}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]triacontria-3,5,7,9,13,17(34), 18(22),19-octaene-12,16-dione (130 mg, 0.21 mmol, 1 equiv), 1-methylpiperazine (65.93 mg, 0.65 mmol, 3 equiv) and BrettPhos Pd G 3To a stirred mixture of 2-nitropropene (59.67 mg, 0.06 mmol, 0.3 eq.) and 1,4-dioxane (13 mL) was added LiHMDS (5.2 mL, 5.2 mmol, 6 eq.). The mixture was stirred at 60 °C for 1 h and cooled to room temperature. The mixture was washed with saturated NH4Cl(aqueous solution) (30 mL) was treated and the mixture was extracted with dichloromethane (3×20 mL). The combined organic layers were washed with brine (3×30 mL) and purified by anhydrous Na 2SO 4The residue was purified by Prep-TLC and eluted with methylene dichloride/methanol (12/1) to give 31 mg of 15,21-dimethyl-5-(4-methylpiperazin-1-yl)-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tetracontriacontria-3,5,7,9,13,17(34),18(22),19-octaene-12,16-dione (Example A75) as a light yellow solid (22%). LCMS: m/z (ESI), [M+H] +=612.30. 1H NMR (DMSO-d 6, 400 MHz) δ 1.85-1.99 (4H, m), 2.05-2.09 (3H, m), 2.24 (3H, t), 2.46-2.50 (4H, m), 2.80 (2H, d), 2.87 (2H, t), 3.09 (4H, t), 3.52-3.59 (4H, m), 3.62 (3H, s), 3.75 (3H, s), 4.28 (2H, t), 6.92 (1H, dd), 7.33 (1H, d), 7.47 (1H, d), 8.31 (2H, d), 9.12 (1H, d), 13.19 (1H, s). Example A80A, Example A80A1, Example A80A2 15,21-Dimethyl-5-[3-(4-methylpiperazin-1-yl)pyrrolidin-1-yl]-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic[24.4.1.1^{1,28}. 1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tris-3,5,7,9,13, 17(33),18(22),19-octaene-12,16-dione, isomer A (Example A80A), isomer A1 (Example A80A1) and isomer A2 (Example A80A2) Step 1. Preparation of 15,21-dimethyl-5-[3-(4-methylpiperazin-1-yl)pyrrolidin-1-yl]-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontahedral-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione, Isomer A (Example A80A). 5-Bromo-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriaconta-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione (M4A, 90 mg, 0.15 mmol, 1 eq.), 1-methyl-4-(pyrrolidin-3-yl)piperazine (79.01 mg, 0.46 mmol, 3 eq.) and BrettPhos Pd G 3To the stirred mixture of 4-nitropropene (42.31 mg, 0.04 mmol, 0.3 equiv) was added LiHMDS (3.6 mL, 3.6 mmol, 24 equiv) dropwise. The mixture was stirred at 60 °C for 1 h and then cooled to room temperature. The reaction was heated with saturated NH4Cl solution (aqueous solution) (10 mL) was quenched, and the mixture was extracted with dichloromethane (3×20 mL). The combined organic layers were washed with brine (3×30 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was subjected to Prep-HPLC using an XBridge Shield RP18 OBD column with 10 mmol/L NH4HCO 3and 0.1% NH 3.H 2The mixture was purified by using water and acetonitrile as the mobile phase. The fractions containing the desired compound were evaporated to dryness to give 2.6 mg of 15,21-dimethyl-5-[3-(4-methylpiperazin-1-yl)pyrrolidin-1-yl]-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontria-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione (Isomer A, Example A80A) as a yellow solid (2%). LCMS: m/z (ESI), [M+H]+= 667.40. 1H NMR (DMSO-d 6, 400 MHz) δ 1.57-1.87 (4H, m), 2.00 (1H, s), 2.16 (3H, s), 2.23-2.63 (12H, m), 2.66-2.76 (2H, m), 2.91 (1H, t), 3.07 (1H, q), 3.20-3.59 (6H, m), 3.62 (3H, s), 3.63-3.69 (1H, m), 3.72 (3H, s), 4.57 (1H, t), 4.69 (1H, d), 6.49 (1H, dd), 6.75 (1H, s), 7.38 (1H, d), 8.26 (1H, d), 8.41 (1H, s), 8.81 (1H, d), 12.51 (1H, s). SFC-HPLC, Rt=1.029 minutes. Step 2. Preparation of 15,21-dimethyl-5-[3-(4-methylpiperazin-1-yl)pyrrolidin-1-yl]-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontahedral-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione, isomer A1 (Example A80A1) and isomer A2 (Example A80A2). A mixture of 15,21-dimethyl-5-[3-(4-methylpiperazin-1-yl)pyrrolidin-1-yl]-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,28}. 1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tris-3,5,7,9,13, 17(33),18(22),19-octaene-12,16-dione (Example A80A, 20 mg, 0.17 mmol) was chromatographed by Prep-chiral-HPLC using an XBridge Shield RP18 OBD column (30*150 mm, 5μm) containing 10 mmol/L NH4HCO 3and 0.1% NH 3.H 2O water and acetonitrile as the mobile phase to obtain 8.5 mg of isomer A1 (Example A80A1, 18%) and 3.2 mg of isomer A2 (Example A80A2, 6%) as yellow solids. Isomer A1: LCMS: m/z (ESI), [M+H] +=667.45. 1H NMR (DMSO-d 6, 400 MHz) δ 1.57-1.87 (4H, m), 2.00 (1H, s), 2.16 (3H, s), 2.23-2.63 (12H, m), 2.66-2.76 (2H, m), 2.91 (1H, t), 3.07 (1H, q), 3.20-3.59 (6H, m), 3.62 (3H, s), 3.63-3.69 (1H, m), 3.72 (3H, s), 4.57 (1H, t), 4.69 (1H, d), 6.49 (1H, dd), 6.76 (1H, s), 7.38 (1H, d), 8.26 (1H, d), 8.42 (1H, s), 8.82 (1H, d), 12.53 (1H, s). Chiral-HPLC, Rt=5.235 min. Isomer A2: LCMS: m/z (ESI), [M+H]+=667.45. 1H NMR (DMSO-d 6, 400 MHz) δ 1.57-1.87 (4H, m), 2.00 (1H, s), 2.16 (3H, s), 2.23-2.63 (12H, m), 2.66-2.76 (2H, m), 2.91 (1H, t), 3.07 (1H, q), 3.20-3.59 (6H, m), 3.62 (3H, s), 3.63-3.69 (1H, m), 3.72 (3H, s), 4.57 (1H, t), 4.69 (1H, d), 6.47 (1H, dd), 6.76 (1H, s), 7.37 (1H, d), 8.26 (1H, d), 8.42 (1H, s), 8.82 (1H, d), 12.49 (1H, s). Chiral-HPLC, Rt=6.080 minutes Example A80B, Example A80B1, Example A80B2 15,21-Dimethyl-5-[3-(4-methylpiperazine-1-yl)pyrrolidin-1-yl]-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic[24.4.1.1^{1,28}. 1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tri-3,5,7,9,13, 17(33),18(22),19-octaene-12,16-dione, isomer B (Example A80B), isomer B1 (Example A80B1) and isomer B2 (Example A80B2) Step 1. Preparation of 15,21-dimethyl-5-[3-(4-methylpiperazin-1-yl)pyrrolidin-1-yl]-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontahedral-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione, Isomer B (Example A80B). 5-Bromo-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tris-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione (M4B, 90 mg, 0.15 mmol, 1 eq.), 1-methyl-4-(pyrrolidin-3-yl)piperazine (79.01 mg, 0.46 mmol, 3 eq.) and BrettPhos Pd G 3To the stirred mixture of 4-(42.31 mg, 0.04 mmol, 0.3 equiv) was added LiHMDS (3.6 mL, 3.60 mmol, 24 equiv) dropwise. The mixture was stirred at 60 °C for 1 h and then cooled to room temperature. The reaction was heated with saturated NH4Cl solution (aqueous solution) (10 mL) was quenched, and the mixture was extracted with dichloromethane (3×20 mL). The combined organic layers were washed with brine (3×30 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-HPLC using an XBridge Shield RP18 OBD column containing 10 mmol/L NH4HCO 3and 0.1% NH 3.H 2O water and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 1.6 mg of 15,21-dimethyl-5-[3-(4-methylpiperazin-1-yl)pyrrolidin-1-yl]-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tris-3,5,7,9,13, 17(33),18(22),19-octaene-12,16-dione, isomer B (Example A80B) as a yellow solid (1%). LCMS: m/z (ESI), [M+H] +=667.40 1H NMR (DMSO-d 6, 400 MHz) δ 1.57-1.87 (4H, m), 2.00 (1H, s), 2.16 (3H, s), 2.23-2.63 (12H, m), 2.66-2.76 (2H, m), 2.91 (1H, t), 3.07 (1H, q), 3.20-3.59 (6H, m), 3.62 (3H, s), 3.63-3.69 (1H, m), 3.72 (3H, s), 4.57 (1H, t), 4.69 (1H, d), 6.49 (1H, d), 6.75 (1H, s), 7.38 (1H, d), 8.26 (1H, d), 8.41 (1H, s), 8.81 (1H, d), 12.51 (1H, s). SFC-HPLC, Rt=0.869, Step 2. Preparation of 15,21-dimethyl-5-[3-(4-methylpiperazin-1-yl)pyrrolidin-1-yl]-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tris-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione, isomer B1 (Example A80B1) and isomer B2 (Example A80B2). A mixture of 15,21-dimethyl-5-[3-(4-methylpiperazin-1-yl)pyrrolidin-1-yl]-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tris-3,5,7,9,13, 17(33),18(22),19-octaene-12,16-dione (Example A80B, 18 mg, 0.17 mmol) was purified by Prep-chiral-HPLC using an XBridge Prep OBD C18 column (30*150 mm, 5 μm) containing 10 mmol/L NH4HCO3 water and acetonitrile were used as mobile phases for separation to obtain 2.4 mg of isomer B1 (Example A80B1, 5%) and 1.2 mg of isomer B2 (Example A80B2, 2%) as yellow solids. Isomer B1: LCMS: m/z (ESI), [M+H] +=667. 1H NMR (DMSO-d 6, 400 MHz) δ 1.57-1.87 (4H, m), 2.00 (1H, s), 2.16 (3H, s), 2.23-2.63 (12H, m), 2.66-2.76 (2H, m), 2.91 (1H, t), 3.07 (1H, q), 3.20-3.59 (6H, m), 3.62 (3H, s), 3.63-3.69 (1H, m), 3.72 (3H, s), 4.57 (1H, t), 4.69 (1H, d), 6.49 (1H, d), 6.75 (1H, s), 7.38 (1H, d), 8.26 (1H, d), 8.41 (1H, s), 8.81 (1H, d), 12.51 (1H, s). Chiral HPLC, Rt=3.906 min. Isomer B2: LCMS: m/z (ESI), [M+H] +=667.45. 1H NMR (DMSO-d 6, 400 MHz) δ 1.57-1.87 (4H, m), 2.00 (1H, s), 2.16 (3H, s), 2.23-2.63 (12H, m), 2.66-2.76 (2H, m), 2.91 (1H, t), 3.07 (1H, q), 3.20-3.59 (6H, m), 3.62 (3H, s), 3.63-3.69 (1H, m), 3.72 (3H, s), 4.57 (1H, t), 4.69 (1H, d), 6.49 (1H, d), 6.75 (1H, s), 7.38 (1H, d), 8.26 (1H, d), 8.41 (1H, s), 8.81 (1H, d), 12.51 (1H, s). Chiral HPLC, Rt = 5.026 minutes. Example A84A and Example A84B 15,21-dimethyl-5-[4-fluoro-3-( N, N-dimethylamino)pyrrolidinyl]-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,28}. 1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tris-3,5,7,9,13, 17(33),18(22),19-octaene-12,16-dione, isomer 1 (Example A84A) and isomer 2 (Example A84B) Step 1. Preparation of tert-butyl 3-(dimethylamino)-4-fluoropyrrolidine-1-carboxylate (INT-A84-1). tert-butyl 3-amino-4-fluoropyrrolidine-1-carboxylate (500 mg, 2.44 mmol, 1 eq.), polyoxymethylene (4410.29 mg, 48.96 mmol, 20 eq.) and NaBH(OAc) were added at 0 °C under nitrogen atmosphere.3(2075.35 mg, 9.79 mmol, 4 equivalents) in Cl 2Cl 2Trimethylamine (990.90 mg, 9.79 mmol, 4 equiv.) was added dropwise to the stirred mixture in 4% paraformaldehyde (20 mL). The mixture was stirred at 0 °C for 2 h. The reaction was quenched with water (10 mL), and the mixture was washed with CH2Cl 2(3×20 mL) extraction. The combined organic layers were washed with brine (3×20 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. Apply the residue on a silica gel column using petroleum CH 2Cl 2/methanol (10/1) for purification to obtain 500 mg of tert-butyl 3-(dimethylamino)-4-fluoropyrrolidine-1-carboxylate (INT-A84-1) (79%) as a brown oil. 1H NMR (DMSO- d 6, 400 MHz) δ 1.40 (9H, s), 2.10-2.23 (5H, m), 2.85-2.95 (1H, m), 3.25 (1H, d), 3.47-3.64 (4H, m), 5.11-5.23 (1H, m). Step 2. 4-Fluoro- N, N- Preparation of dimethylpyrrolidin-3-amine (INT-A84-2). A mixture of tert-butyl 3-(dimethylamino)-4-fluoropyrrolidine-1-carboxylate (500 mg, 2.152 mmol, 1 eq.) and HCl in 1,4-dioxane (4 mL) was stirred at room temperature for 2 h under nitrogen atmosphere. Solid K was added to the reaction solution.2CO 3After filtration, the filtrate was concentrated under reduced pressure, and 300 mg of 4-fluoro- N, N-Dimethylpyrrolidin-3-amine (INT-A84-2). This material was used in the next reaction without further purification. 1H NMR (DMSO- d 6, 400 MHz) δ 2.05-2.25 (6H, m), 2.40-2.65 (2H, m), 2.76-3.48 (4H, m), 4.94-5.08 (1H, m) Step 3. 15,21-dimethyl-5-[4-fluoro-3-( N, N-dimethylamino)pyrrolidinyl]-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triathrine-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione, preparation of isomer 1 (Example A84A) and isomer 2 (Example A84B). 5-Bromo-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]tris-3,5,7,9,13,17(33), 18(22),19-octaene-12,16-dione (M4A, 150 mg, 0.25 mmol, 1.00 equivalent), BrettPhos Pd G 3(70.52 mg, 0.07 mmol, 0.3 eq.) and 4-fluoro- N, NTo a stirred mixture of 2-dimethylpyrrolidin-3-amine (102.83 mg, 0.77 mmol, 3 equiv) in 1,4-dioxane (15 mL) was added LiHMDS (6 mL, 6.00 mmol, 24 equiv) dropwise. The mixture was stirred at 60 °C for 1 h and then cooled to room temperature. The reaction was heated with saturated NH4Cl(aq) solution (15 mL) was quenched, and the mixture was extracted with dichloromethane (3×20 mL). The combined organic layers were washed with brine (3×30 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was passed through Prep-HPLC using an XBridge Shield RP18 OBD column with 0.1% NH3.H 2O water and acetonitrile as the mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to obtain 9.8 mg of 15,21-dimethyl-5-[4-fluoro-3-( N, N-dimethylamino)pyrrolidinyl]-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,28}. 1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriaconta-3,5,7,9,13, 17(33),18(22),19-octaene-12,16-dione, isomer 1 (Example A84A) (6%) and 10.0 mg of isomer 2 (Example A84B) (6%) as a yellow solid. Isomer 1: LCMS: m/z (ESI), [M+H] +=630.40. 1H NMR (DMSO-d 6, 400 MHz) δ 1.48-1.68 (4H, m), 2.01-2.08 (1H, m), 2.30 (6H, s), 2.41-2.58 (3H, m), 2.55-2.79 (4H, m), 3.04-3.10 (2H, m), 3.22-3.32 (1H, m), 3.54-3.69 (3H, m), 3.62 (3H, s), 3.72 (3H, s), 4.55 (1H, t), 4.62-4.73 (1H, m), 5.25-5.35 (1H, m), 6.50 (1H, dd), 6.76 (1H, s), 7.40 (1H, d), 8.26 (1H, d), 8.42 (1H, s), 8.82 (1H, d), 12.52 (1H, s). SFC-HPLC, Rt=4.025 minutes. Isomer 2: LCMS: m/z (ESI), [M+H] +=630.40. 1H NMR (DMSO-d 6, 400 MHz) δ 1.48-1.68 (4H, m), 2.01-2.08 (1H, m), 2.30 (6H, s), 2.41-2.58 (3H, m), 2.55-2.79 (4H, m), 3.04-3.10 (2H, m), 3.22-3.32 (1H, m), 3.54-3.69 (3H, m), 3.62 (3H, s), 3.72 (3H, s), 4.55 (1H, t), 4.62-4.73 (1H, m), 5.25-5.35 (1H, m), 6.59 (1H, dd), 6.86 (1H, s), 7.41 (1H, d), 8.27 (1H, d), 8.42 (1H, s), 8.82 (1H, d), 12.51 (1H, s). SFC-HPLC, Rt = 3.757 minutes. Example A84C and Example A84D 15,21-dimethyl-5-[4-fluoro-3-( N, N-dimethylamino)pyrrolidinyl]-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,28}. 1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tris-3,5,7,9,13, 17(33),18(22),19-octaene-12,16-dione, isomer 3 (Example A84C) and isomer 4 (Example A84B) Step 1. 15,21-dimethyl-5-[4-fluoro-3-( N, N-dimethylamino)pyrrolidinyl]-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triathrine-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione, preparation of isomer 3 (Example A84C) and isomer 4 (Example A84D). Under nitrogen atmosphere at 0 ° C, 5-bromo-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]tritriacontria-3,5,7,9,13,17(33), 18(22),19-octaene-12,16-dione (M4B, 150 mg, 0.25 mmol, 1 equivalent), 4-fluoro- N, N-dimethylpyrrolidin-3-amine (102.83 mg, 0.77 mmol, 3 equiv) and BrettPhos Pd G 3To the stirred mixture of 2-nitropropene (70.52 mg, 0.07 mmol, 0.3 equiv) was added LiHMDS (6 mL, 6.00 mmol, 24 equiv) dropwise. The mixture was stirred at 60 °C for 1 h and then cooled to room temperature. The reaction was heated with saturated NH4Cl(aq) solution (10 mL) was quenched, and the mixture was extracted with dichloromethane (3×20 mL). The combined organic layers were washed with brine (3×30 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was passed through Prep-HPLC using an XBridge Shield RP18 OBD column with 0.1% NH3.H 2O water and acetonitrile as the mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to obtain 11.8 mg of 15,21-dimethyl-5-[4-fluoro-3-( N, N-dimethylamino)pyrrolidinyl]-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriaconta-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione, isomer 3 (Example A84C) (7%) and 6.0 mg of isomer 4 (Example A84D) (3%) as a yellow solid. Isomer 3: LCMS: m/z (ESI), [M+H] +=630.40. 1H NMR (DMSO-d 6, 400 MHz) δ 1.48-1.68 (4H, m), 2.01-2.08 (1H, m), 2.30 (6H, s), 2.41-2.58 (3H, m), 2.55-2.79 (4H, m), 3.04-3.10 (2H, m), 3.22-3.32 (1H, m), 3.54-3.69 (3H, m), 3.62 (3H, s), 3.72 (3H, s), 4.55 (1H, t), 4.62-4.73 (1H, m), 5.25-5.35 (1H, m), 6.50 (1H, dd), 6.76 (1H, s), 7.40 (1H, d), 8.26 (1H, d), 8.42 (1H, s), 8.82 (1H, d), 12.53 (1H, s). SFC-HPLC, Rt=3.983min Isomer 4: LCMS: m/z (ESI), [M+H] +=630.40. 1H NMR (DMSO-d 6, 400 MHz) δ 1.48-1.68 (4H, m), 2.01-2.08 (1H, m), 2.30 (6H, s), 2.41-2.58 (3H, m), 2.55-2.79 (4H, m), 3.04-3.10 (2H, m), 3.22-3.32 (1H, m), 3.54-3.69 (3H, m), 3.62 (3H, s), 3.72 (3H, s), 4.55 (1H, t), 4.62-4.73 (1H, m), 5.25-5.35 (1H, m), 6.59 (1H, dd), 6.86 (1H, s), 7.41 (1H, d), 8.27 (1H, d), 8.42 (1H, s), 8.82 (1H, d), 12.55 (1H, s). SFC-HPLC, Rt = 3.724 minutes. Example A94A, Example 94A1 and Example A94A2 5-[3-(3-fluoroaziridocyclobutane-1-yl)pyrrolidin-1-yl]-15,21-dimethyl-23-oxadiazol-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}. 1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tri-3,5,7,9,13, 17(33),18(22),19-octaene-12,16-dione, isomer A (Example A94A), isomer A1 (Example A94A1) and isomer A2 (Example A94A2) Step 1. Preparation of tert-butyl 3-(3-fluoroaziridocyclobutane-1-yl)pyrrolidine-1-carboxylate (INT-A94A-1). tert-butyl 3-oxopyrrolidine-1-carboxylate (1500 mg, 8.10 mmol, 1 equivalent) and 3-fluoroaziridocyclobutane hydrochloride (903 mg, 8.10 mmol, 1 equivalent) were mixed in CH2Cl 2The mixture in (50 mL) was stirred at room temperature under air atmosphere for 1 h. NaBH(OAc) was added to the above mixture in portions at 0°C within 2 minutes.3(8582 mg, 40.49 mmol, 5 equiv.). The resulting mixture was stirred for another 2 h. The reaction was treated with saturated NH4Cl(aq) (50 mL) was quenched and the mixture was washed with CH 2Cl 2(3×100 mL) extraction. The combined organic layers were washed with brine (3×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column using dichloromethane/methanol (12/1) to give 1.5 g of tert-butyl 3-(3-fluoroaziridocyclobutane-1-yl)pyrrolidine-1-carboxylate (INT-A94A-1) (75%) as a yellow oil. 1H NMR (CDCl 3, 400 MHz) δ 1.46 (9H, d), 1.68-1.83 (1H, m), 1.85-1.95 (1H, m), 3.05-3.15 (1H, m), 3.18-3.48 (6H, m), 3.72-3.87 (2H, m) Step 2. Preparation of 3-(3-fluoro-1-nitro-2-cyclobutane-1-yl)pyrrolidine (INT-A94A-2). 3-(3-fluoro-1-nitro-2-cyclobutane-1-yl)pyrrolidine-1-carboxylic acid tert-butyl ester (500 mg, 2.05 mmol, 1 equivalent) and trifluoroacetic acid (3 mL) were mixed in CH 2Cl 2The solution in (9 mL) was stirred at room temperature for 2 h. The mixture was concentrated under reduced pressure, treated with dichloromethane/methanol (10/1) (30 mL), and K 2CO 3Adjust to pH 7. The mixture was filtered and the filter cake was washed with methanol (2×10 mL). The resulting solution was concentrated under reduced pressure to give 150 mg of 3-(3-fluoroaziridocyclobutane-1-yl)pyrrolidine (INT-A94A-2) as a yellow oil. This material was used in the next reaction without further purification. 1H NMR (DMSO-d 6, 400 MHz) δ 1.75-1.84 (2H, m), 2.70-2.81 (2H, m), 2.82-2.95 (2H, m), 3.05-3.21 (3H, m), 3.59-3.62 (2H, m), 4.98-5.18 (2H, m) Step 3. Preparation of 5-[3-(3-fluoro-1-nitro-2-cyclobutane-1-yl)pyrrolidin-1-yl]-15,21-dimethyl-23-oxo-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tris-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione (isomer A, Example A94A). 3-(3-fluoro-1-nitro-2-cyclobutane-1-yl)pyrrolidine (125 mg, 0.86 mmol, 5 equivalents), BrettPhos Pd G 3To a stirred mixture of 5-bromo-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontria-3,5,7,9,13,17(33), 18(22),19-octaene-12,16-dione (M4A, 100 mg, 0.173 mmol, 1 eq) in 1,4-dioxane (10 mL) was added LiHMDS (2.1 mL, 2.07 mmol, 12 eq). The resulting mixture was stirred at 60 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was treated with saturated NH4Cl(aq) (15 mL) was quenched and the mixture was washed with CH 2Cl 2(3×50 mL) extraction. The combined organic layers were washed with brine (3×30 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (10:1). The product was purified by Prep-HPLC using an XBridge Prep Phenyl OBD column with 0.1% NH4HCO 3The product was further purified by using water and acetonitrile as mobile phase. The fractions containing the desired compound were evaporated to dryness to give 13 mg of 5-[3-(3-fluoroazidocyclobutane-1-yl)pyrrolidin-1-yl]-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontria-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione, Isomer A (Example A94A) as an off-white solid (12%). LCMS: m/z (ESI), [M+H] += 642.40. 1H NMR (DMSO-d 6, 400 MHz) δ 1.48-1.85 (4H, m), 1.89-2.05 (2H, m), 2.35-2.56 (4H, m), 2.68-2.76 (2H, m), 2.96 (1H, t), 3.07-3.20 (4H, m), 3.20-3.43 (4H, m), 3.45-3.70 (3H, m), 3.62 (3H, s), 3.72 (3H, s), 4.55 (1H, t), 4.63-4.73 (1H, m), 4.95-5.31 (1H, m), 6.44 (1H, d), 6.73 (1H, s), 7.37 (1H, d), 8.25 (1H, d), 8.42 (1H, s), 8.82 (1H, d), 12.50 (1H, s). Step 4. Preparation of 5-[3-(3-fluoroazidocyclobutane-1-yl)pyrrolidin-1-yl]-15,21-dimethyl-23-oxo-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontahedral-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione, isomer A1 (Example A94A1) and isomer A2 (Example A94A2). A mixture of 5-[3-(3-fluoroaziridocyclobutane-1-yl)pyrrolidin-1-yl]-15,21-dimethyl-23-oxadiazol-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}. 1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tris-3,5,7,9,13, 17(33),18(22),19-octaene-12,16-dione (Example A94A, 10.5 mg) was chromatographed by Prep-chiral-HPLC using a CHIRALPAK IG column (2×25 cm, 5 μm) with 0.2% diethylamine and ethanol/CH2Cl 2The mixture was separated by using (1/1) hexane as the mobile phase to obtain 2.3 mg of 5-[3-(3-fluoroazidocyclobutane-1-yl)pyrrolidin-1-yl]-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriaconta-3,5,7,9,13,17(33), 18(22),19-octaene-12,16-dione, isomer A1 (Example A94A1) as a yellow solid (21%) and 3.4 mg Isomer A2 (Example A94A2) (32%). Isomer A1: LCMS: m/z (ESI), [M+H] +=642.35 1H NMR (DMSO-d 6, 400 MHz) δ 1.48-1.85 (4H, m), 1.89-2.05 (2H, m), 2.35-2.56 (4H, m), 2.68-2.76 (2H, m), 2.96 (1H, t), 3.07-3.20 (4H, m), 3.20-3.43 (4H, m), 3.45-3.70 (3H, m), 3.62 (3H, s), 3.72 (3H, s), 4.55 (1H, t), 4.63-4.73 (1H, m), 4.95-5.31 (1H, m), 6.44 (1H, d), 6.73 (1H, s), 7.37 (1H, d), 8.25 (1H, d), 8.42 (1H, s), 8.82 (1H, d), 12.50 (1H, s). Chiral HPLC, Rt=2.381 min. Isomer A2: LCMS: m/z (ESI), [M+H] +=642.55. 1H NMR (DMSO-d 6, 400 MHz) δ 1.48-1.85 (4H, m), 1.89-2.05 (2H, m), 2.35-2.56 (4H, m), 2.68-2.76 (2H, m), 2.96 (1H, t), 3.07-3.20 (4H, m), 3.20-3.43 (4H, m), 3.45-3.70 (3H, m), 3.62 (3H, s), 3.72 (3H, s), 4.55 (1H, t), 4.63-4.73 (1H, m), 4.95-5.31 (1H, m), 6.44 (1H, d), 6.73 (1H, s), 7.37 (1H, d), 8.25 (1H, d), 8.42 (1H, s), 8.82 (1H, d), 12.50 (1H, s). Chiral HPLC, Rt=2.959 minutes. Example A94B, Example 94B1 and Example A94B2 5-[3-(3-fluoroazolocyclobutane-1-yl)pyrrolidin-1-yl]-15,21-dimethyl-23-oxo-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tris-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione, isomer B (Example A94B), isomer B1 (Example A94B1) and isomer B2 (Example A94B2) Step 1. Preparation of 5-[3-(3-fluoroazidocyclobutane-1-yl)pyrrolidin-1-yl]-15,21-dimethyl-23-oxo-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontahedral-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione (isomer B, Example A94B). To 3-(3-fluoroaziridine cyclobutane-1-yl)pyrrolidine (INT-A94A-2, 125 mg, 0.86 mmol, 5 equivalents), BrettPhos Pd G 3To a stirred mixture of 5-bromo-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontria-3,5,7,9,13, 17(33),18(22),19-octaene-12,16-dione (M4B, 100 mg, 0.173 mmol, 1 eq) in 1,4-dioxane (10 mL) was added LiHMDS (2.1 mL, 2.07 mmol, 12 eq). The resulting mixture was stirred at 60 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was treated with saturated NH4Cl(aq) (15 mL) was quenched and the mixture was washed with CH 2Cl 2(3×50 mL) extraction. The combined organic layers were washed with brine (3×30 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (10/1). The product was purified by Prep-HPLC using an XBridge Prep OBD C18 column with 0.1% NH4HCO 3The product was purified by using water and acetonitrile as mobile phase. The fractions containing the desired compound were evaporated to dryness to give 15.3 mg of 5-[3-(3-fluoroazidocyclobutane-1-yl)pyrrolidin-1-yl]-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontria-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione, Isomer B (Example A94B) as an off-white solid (13%). LCMS: m/z (ESI), [M+H] += 642.40. 1H NMR (DMSO-d 6, 400 MHz) δ 1.48-1.85 (4H, m), 1.89-2.05 (2H, m), 2.35-2.56 (4H, m), 2.68-2.76 (2H, m), 2.96 (1H, t), 3.07-3.20 (4H, m), 3.20-3.43 (4H, m), 3.45-3.70 (3H, m), 3.62 (3H, s), 3.72 (3H, s), 4.55 (1H, t), 4.63-4.73 (1H, m), 4.95-5.31 (1H, m), 6.44 (1H, d), 6.73 (1H, s), 7.37 (1H, d), 8.25 (1H, d), 8.42 (1H, s), 8.82 (1H, d), 12.50 (1H, s). Step 2. Preparation of 5-[3-(3-fluoroazidocyclobutane-1-yl)pyrrolidin-1-yl]-15,21-dimethyl-23-oxo-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontahedral-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione, isomer B1 (Example A94B1) and isomer B2 (Example A94B2). A mixture of 5-[3-(3-fluoroaziridocyclobutane-1-yl)pyrrolidin-1-yl]-15,21-dimethyl-23-oxadiazol-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}. 1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tris-3,5,7,9,13, 17(33),18(22),19-octaene-12,16-dione (Example A94B, 11 mg) was subjected to Prep-chiral-HPLC using a CHIRALPAK IG column (2×25 cm, 5 μm) with 0.2% diethylamine as mobile phase A and ethanol/CH2Cl 2(1/1) was separated using hexane as mobile phase B to give 3.7 mg of 5-[3-(3-fluoroazidocyclobutane-1-yl)pyrrolidin-1-yl]-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriaconta-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione, isomer B1 (Example A94B1) (28%) as a yellow solid and 4.5 mg Isomer B2 (Example A94B2) (34%). Isomer B1: LCMS: m/z (ESI), [M+H] +=642.30. 1H NMR (DMSO-d 6, 400 MHz) δ 1.48-1.85 (4H, m), 1.89-2.05 (2H, m), 2.35-2.56 (4H, m), 2.68-2.76 (2H, m), 2.96 (1H, t), 3.07-3.20 (4H, m), 3.20-3.43 (4H, m), 3.45-3.70 (3H, m), 3.62 (3H, s), 3.72 (3H, s), 4.55 (1H, t), 4.63-4.73 (1H, m), 4.95-5.31 (1H, m), 6.44 (1H, d), 6.73 (1H, s), 7.37 (1H, d), 8.25 (1H, d), 8.42 (1H, s), 8.82 (1H, d), 12.50 (1H, s). Chiral HPLC, Rt=1.608 min. Isomer B2: LCMS: m/z (ESI), [M+H] +=642.55. 1H NMR (DMSO-d 6, 400 MHz) δ 1.48-1.85 (4H, m), 1.89-2.05 (2H, m), 2.35-2.56 (4H, m), 2.68-2.76 (2H, m), 2.96 (1H, t), 3.07-3.20 (4H, m), 3.20-3.43 (4H, m), 3.45-3.70 (3H, m), 3.62 (3H, s), 3.72 (3H, s), 4.55 (1H, t), 4.63-4.73 (1H, m), 4.95-5.31 (1H, m), 6.48 (1H, d), 6.76 (1H, s), 7.38 (1H, d), 8.26 (1H, d), 8.42 (1H, s), 8.82 (1H, d), 12.52 (1H, s). Chiral HPLC, Rt = 2.068 minutes. Example A100 15,21-Dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic [26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontria-3,5,7,9,13,17(34),18(22),19-octaene-12,16-dione Step 1. Preparation of 2-{1-[(2-nitrophenyl)amino]-3-azabicyclo[3.2.2]nonan-3-yl}ethanol (INT-A100-1). 3-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-3-azabicyclo[3.2.2]nonan-1-amine (250 mg, 0.83 mmol, 1 equivalent), K 2CO 3A mixture of 2-fluoro-1-nitro-2-nitro-1-yl (347 mg, 2.51 mmol, 3 equiv.) and o-fluoronitrobenzene (295 mg, 2.09 mmol, 2.5 equiv.) in acetonitrile (8 mL) was stirred at 100 °C for 8 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (20 mL), and the mixture was extracted with ethyl acetate (3×30 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column using petroleum ether/ethyl acetate (5/1) to give 110 mg of 2-{1-[(2-nitrophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethanol (INT-A100-1) (38%) as a yellow oil. LCMS: m/z (ESI), [M+H] +=306.15. Step 2. Preparation of 1-methyl-5-[1-methyl-5-(2-{1-[(2-nitrophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethoxy)pyrazol-4-yl]-6-oxopyridine-3-carboxylic acid methyl ester (INT-A100-2). To a stirred mixture of 2-{1-[(2-nitrophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethanol (100 mg, 0.32 mmol, 1 eq) and triphenylphosphine (257 mg, 0.98 mmol, 3 eq) in tetrahydrofuran (20 mL) was added diisopropyl azodicarboxylate (198 mg, 0.98 mmol, 3 eq) and methyl 5-(5-hydroxy-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (M2, 68 mg, 0.26 mmol, 0.8 eq) in tetrahydrofuran (5 mL) dropwise under nitrogen atmosphere at 0° C. The mixture was stirred at 0° C. for 2 h. The reaction was quenched with water (20 mL), and the mixture was extracted with ethyl acetate (3×30 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column eluting with petroleum ether/ethyl acetate (1/1) to give 150 mg of 1-methyl-5-[1-methyl-5-(2-{1-[(2-nitrophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethoxy)pyrazol-4-yl]-6-oxopyridine-3-carboxylic acid methyl ester (INT-A100-2) (74%) as a yellow oil. LCMS: m/z (ESI), [M+H] +=551.20. Step 3. Preparation of 5-[5-(2-{1-[(2-aminophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A100-3). To a stirred mixture of methyl 1-methyl-5-[1-methyl-5-(2-{1-[(2-nitrophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethoxy)pyrazol-4-yl]-6-oxopyridine-3-carboxylate (140 mg, 0.25 mmol, 1 eq) and ranyl nickel (69 mg) in methanol (5 mL) was added hydrazine hydrate (51 mg, 1.01 mmol, 4.00 eq) at 0 °C. The mixture was stirred at 0 °C under nitrogen atmosphere for 2 h. The resulting mixture was filtered, and the filter cake was washed with methanol (3 x 10 mL). The resulting solution was concentrated under reduced pressure to give 90 mg of 5-[5-(2-{1-[(2-aminophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A100-3) (58%) as a light yellow solid. LCMS: m/z (ESI), [M+H] +=521.30. Step 4. Preparation of 5-(5-{2-[1-(2-amino-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.2]non-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A100-4). A mixture of 5-[5-(2-{1-[(2-aminophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (90 mg, 0.17 mmol, 1 eq.) and BrCN (21 mg, 0.20 mmol, 1.20 eq.) in ethanol (5 mL) was stirred at room temperature overnight under nitrogen atmosphere. The reaction was quenched with water (20 mL), and the mixture was extracted with ethyl acetate (3×30 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using petroleum ether/ethyl acetate (1/1) to give 60 mg of 5-(5-{2-[1-(2-amino-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.2]non-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A100-4) (57%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=546.35. Step 5. Preparation of 5-(5-{2-[1-(2-amino-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.2]non-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A100-5). 5-(5-{2-[1-(2-amino-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.2]non-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (50 mg, 0.09 mmol, 1 equivalent) and LiOH.H 2A mixture of O (7 mg, 0.27 mmol, 3 equiv.) in tetrahydrofuran (4 mL) and water (1 mL) was stirred at room temperature under nitrogen atmosphere for 2 h. The mixture was concentrated under reduced pressure. The residue was passed through C18FLASH using a 10 mmol/L NH4HCO 3and 0.1% NH 3.H 2O and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 25 mg of 5-(5-{2-[1-(2-amino-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.2]non-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A100-5) (50%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=532.25. Step 6. Preparation of 15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]triacontria-3,5,7,9,13,17(34),18(22),19-octaene-12,16-dione (Example A100). 5-(5-{2-[1-(2-amino-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.2]non-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (30 mg, 0.05 mmol, 1 equivalent), N, N, N, N-Tetramethyl- O-(7-Azabenzotriazol-1-yl)urea hexafluorophosphate (32 mg, 0.08 mmol, 1.5 equivalents) and N, N- A mixture of diisopropylethylamine (21 mg, 0.16 mmol, 3 equiv.) in 1,4-dioxane (5 mL) was stirred at 60 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (10 mL), and the mixture was extracted with ethyl acetate (3×20 mL). The combined organic layers were washed with brine (3×30 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was subjected to Prep-HPLC using an XBridge Shield RP18 OBD column with 10 mmol/L NH4HCO 3and 0.1% NH 3 .H 2O water and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 26.8 mg of 15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontria-3,5,7,9,13,17(34),18(22),19-octaene-12,16-dione (Example A100) (92%) as a white solid. LCMS: m/z (ESI), [M+H] += 514.20. 1H NMR (DMSO-d 6 , 400 MHz) δ 1.85-1.98 (4H, m), 2.02-2.18 (3H, m), 2.78-2.88 (2H, m), 2.88-2.95 (2H, m), 3.40-3.52 (4H, m), 3.63 (3H, s), 3.75 (3H, s), 4.29 (2H, s), 7.12-7.21 (2H, m), 7.63 (1H, d), 7.89 (1H, d), 8.38-8.24 (2H, m), 9.12 (1H, d), 13.30 (1H, s). Example A101 (11R)-15-chloro-5,11,26-trimethyl-16-(4-methylpiperazin-1-yl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]octadecene-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione Step 1. (11 R)-15-chloro-5,11,26-trimethyl-16-(4-methylpiperazine-1-yl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]twenty-eight-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (Example A101). In a nitrogen atmosphere at 0°C to (11 R)-15-chloro-5,11,26-trimethyl-16-(piperazin-1-yl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]octadecene-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (Example A23, 80 mg, 0.14 mmol, 1 equivalent), paraformaldehyde (38 mg, 0.42 mmol, 3 equivalents) and NaBH(OAc) 3(60 mg, 0.28 mmol, 2 eq.) in CH 2Cl 2(20 mL) was added dropwise to the stirring mixture N, N-Diisopropylethylamine (36 mg, 0.28 mmol, 2 eq.). The mixture was stirred at 0 °C for 2 h. The reaction was quenched with water (10 mL), and the mixture was washed with CH2Cl 2(3×10 mL) extraction. The combined organic layers were washed with brine (3×10 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was subjected to Prep-HPLC using an XBridge Shield RP18 OBD column with 10 mmol/L NH4HCO 3and 0.1% NH 3.H 2O water and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 32.4 mg (11 R)-15-chloro-5,11,26-trimethyl-16-(4-methylpiperazin-1-yl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octadecene-1(28),2(6),3, 14,16,18,20,24-octaene-23,27-dione (Example A101) (39%). LCMS: m/z (ESI), [M+H] +=579.20. 1H NMR (DMSO- d 6, 400 MHz) δ 0.86 (3H, d), 1.51-1.59 (1H, m), 1.86-2.08 (2H, m), 2.21-2.29 (5H, m), 2.51-2.64 (4H, m), 2.80-2.94 (4H, m), 3.62 (3H, s), 3.71 (3H, s), 3.91-4.03 (1H, m), 4.18 (1H, dd), 4.32-4.49 (1H, m), 4.75-4.97 (1H, m), 7.15 (1H, d), 7.58 (1H, d), 8.31-8.47 (2H, m), 8.85 (1H, d), 12.73 (1H, s). Example A102 and Example A103 (11 R)-5,11,30-trimethyl-7-oxa-4,5,13,18,24,26,30-heptaazahexacyclo[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]triacontidine-1(32),2(6),3,14,20,22,24,28-octaene-27,31-dione (Example A102) and (11 R)-5,11,18,30-tetramethyl-7-oxa-4,5,13,18,24,26, 30-heptaazahexacyclo[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}. 0^{15,20}]triacontidine-1(32),2(6),3,14,20,22,24,28-octaene-27,31-dione (Example A103). Step 1. 2,2,2-Trifluoro- N- Preparation of (2-fluorophenylethyl) acetamide (INT-A102-1). A mixture of 2-(2-fluorophenyl) ethylamine (50 g, 359 mmol, 1 eq.), trimethylamine (73 g, 359 mmol, 2 eq.) and trifluoroacetic anhydride (75 g, 1.1 eq.) in dichloromethane (800 mL) was stirred at 0 °C for 3 h under a nitrogen atmosphere. The reaction was quenched with water (200 mL), and the mixture was extracted with dichloromethane (3×200 mL). The combined organic layers were washed with brine (3×200 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue is purified by silica gel chromatography using petroleum ether/ethyl acetate (5/1) to give 76 g of 2,2,2-trifluoro- N-[2-(2-fluorophenyl)ethyl]acetamide (INT-A102-1) (90%). LCMS: m/z (ESI), [M+H] +=236.05. Step 2. Preparation of 2,2,2-trifluoro-1-(5-fluoro-3,4-dihydroisoquinoline-2(1H)-yl)ethan-1-one (INT-A102-2). 2,2,2-trifluoro- N-[2-(2-fluorophenyl)ethyl]acetamide (76 g, 323.15 mmol, 1 equivalent), (HCHO) n(43 g, 969.5 mmol, 3 eq.) and H 2SO 4A mixture of (224 mL, 4201 mmol, 13 equiv) and AcOH (296 mL) was stirred at 0 °C for 3 h under nitrogen atmosphere. The reaction was quenched with water (800 mL) and neutralized to pH 7 with NaOH. The mixture was extracted with dichloromethane (3×500 mL). The combined organic layers were washed with brine (3×500 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by silica gel chromatography using petroleum ether/ethyl acetate (1/1) to give 10 g of 2,2,2-trifluoro-1-(5-fluoro-3,4-dihydro-1H-isoquinolin-2-yl)ethanone (INT-A102-2) (13%) as a brownish yellow oil. LCMS: m/z (ESI), [M+H] +=248.10. Step 3. Preparation of 2,2,2-trifluoro-1-(5-fluoro-6-nitro-3,4-dihydroisoquinolin-2(1H)-yl)ethan-1-one (INT-A102-3). 2,2,2-trifluoro-1-(5-fluoro-3,4-dihydro-1H-isoquinolin-2-yl)ethanone (10 g, 40.45 mmol, 1 equivalent) and KNO 3(4 g, 40.45 mmol, 1 eq.) in H 2SO 4The mixture in 4% paraformaldehyde (100 mL) was stirred at 0 °C overnight. The reaction was quenched with water (500 mL) and adjusted to pH 9 with NaOH. The mixture was extracted with dichloromethane (3×200 mL). The combined organic layers were washed with brine (3×200 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue is purified by silica gel chromatography eluting with petroleum ether/ethyl acetate (3/1) to give 1.4 g of 2,2,2-trifluoro-1-(5-fluoro-6-nitro-3,4-dihydro-1H-isoquinolin-2-yl)ethanone (INT-A102-3) (12%) as a yellow oil. 1H NMR (DMSO, 400 MHz) δ 2.91-2.99 (2H, m), 3.85-3.92 (2H, m), 4.91 (2H, d), 7.45 (1H, d), 8.12 (1H, d). Step 4. 2,2,2-Trifluoro-1-(5-{[(2 RPreparation of 2,2,2-trifluoro-1-(5-fluoro-6-nitro-3,4-dihydro-1H-isoquinolin-2-yl)ethanone (INT-A102-3, 15 g, 51.34 mmol, 1 equivalent), N,N-diisopropylethylamine (19.9 g, 154.01 mmol, 3 equivalents) and (4 RA mixture of 5-amino-4-methylpentan-1-ol (M3-6, 6016 mg, 51.34 mmol, 1 equivalent) in acetonitrile (150 mL) was stirred at 80 °C for 2 h. The mixture was cooled to room temperature. The reaction was quenched with water (500 mL), and the mixture was extracted with dichloromethane (3×500 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by eluting with petroleum ether/ethyl acetate (1/1) through a silica gel column to obtain 10 g of 2,2,2-trifluoro-1-(5-{[(2 R)-5-hydroxy-2-methylpentyl]amino}-6-nitro-3,4-dihydro-1H-isoquinolin-2-yl)ethanone (INT-A102-3) (50%). LCMS: m/z (ESI), [M+H] +=390.10. Step 5. (4 R)-4-methyl-5-[(6-nitro-1,2,3,4-tetrahydroisoquinolin-5-yl)amino]pentan-1-ol (INT-A102-5). 2,2,2-trifluoro-1-(5-{[(2 R)-5-hydroxy-2-methylpentyl]amino}-6-nitro-3,4-dihydro-1H-isoquinolin-2-yl)ethanone (10 g, 25.68 mmol, 1 equivalent) and K 2CO 3(10.6 g, 77.05 mmol, 3 equiv.) in methanol (80 mL) and H 2O (40 mL) was stirred at room temperature for 5 h. The reaction was treated with water (500 mL), and the mixture was extracted with dichloromethane (3×500 mL). The combined organic layers were washed with brine (3×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by passing through a silica gel column eluting with petroleum ether/ethyl acetate (1/1) to obtain 7 g (4 R)-4-methyl-5-[(6-nitro-1,2,3,4-tetrahydroisoquinolin-5-yl)amino]pentan-1-ol (INT-A102-5) (93%). LCMS: m/z (ESI), [M+H] +=294.15. 1H NMR (DMSO-d 6,400 MHz) δ 0.83-0.88 (3 H, m), 1.07-1.11 (1 H, m), 1.22-1.51 (3 H, m), 1.57-1.64 (1 H, m), 2.59 (2 H, t), 2.88-2.92 (3 H, m), 3.10-3.20 (1 H, m), 3.30-3.33 (2 H, m), 3.87 (2H, s), 4.37 (1 H, s), 6.56 (1 H, d), 6.69 (1 H, t), 7.26-7.37 (1 H, m), 7.70 (1 H, d). Step 6. 5-{[(2 RPreparation of tert-butyl 6-nitro-3,4-dihydro-1H-isoquinoline-2-carboxylate (INT-A102-6). (4 RA mixture of )-4-methyl-5-[(6-nitro-1,2,3,4-tetrahydroisoquinolin-5-yl)amino]pentan-1-ol (8 g, 27.27 mmol, 1 equiv), trimethylamine (5.5 g, 54.54 mmol, 2 equiv) and di-tert-butyl dicarbonate (7.1 g, 32.72 mmol, 1.2 equiv) in dichloromethane (100 mL) was stirred at room temperature for 2 h. The reaction was quenched with water (300 mL), and the mixture was extracted with dichloromethane (3×300 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by passing through a silica gel column eluting with petroleum ether/ethyl acetate (1/1) to obtain 6.5 g of 5-{[(2 R)-5-hydroxy-2-methylpentyl]amino}-6-nitro-3,4-dihydro-1H-isoquinoline-2-carboxylic acid tert-butyl ester (INT-A102-6) (60%). LCMS: m/z (ESI), [M+H] +=394.20. 1H NMR (DMSO-d 6400 MHz) δ 0.84 (3H, d), 1.03-1.17 (1H, m), 1.22-1.45 (2H, m), 1.45 (9H, s), 1.61 (1H, q), 2.75 (2H, t), 2.93-2.97 (1H, m), 3.08-3.12 (1H, m), 3.30-3.33 (2H, m), 3.51 (2H, t), 4.36 (1H, t), 4.52 (2H, s), 6.73 (1H, d), 6.83 (1H, s), 7.80 (1H, d) Step 7. 5-{[(2 RPreparation of tert-butyl 6-nitro-3,4-dihydro-1H-isoquinoline-2-carboxylate (INT-A102-7). In a nitrogen atmosphere, 5-{[(2 RTo a stirred mixture of tert-butyl 5-(5-hydroxy-2-methylpentyl]amino}-6-nitro-3,4-dihydro-1H-isoquinoline-2-carboxylate (INT-A102-6, 800 mg, 2.03 mmol, 1 eq.) and methyl 5-(5-hydroxy-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (M2, 535 mg, 2.03 mmol, 1.0 eq.) in toluene (30 mL) was added 2-(tributyl-1^[5]-phosphinilidene)acetonitrile (2 g, 10.16 mmol, 5 eq.). The resulting mixture was stirred at 100 °C for 2 h and then cooled to room temperature. The reaction was quenched with water (100 mL) and the mixture was extracted with ethyl acetate (3 x 100 mL). The combined organic layers were washed with brine (3×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by C18 column using water and acetonitrile as mobile phase. The fractions containing the desired compound were evaporated to give 700 mg 5-{[(2 R)-5-({4-[5-(methoxycarbonyl)-1-methyl-2-oxopyridin-3-yl]-2-methylpyrazol-3-yl}oxy)-2-methylpentyl]amino}-6-nitro-3,4-dihydro-1H-isoquinoline-2-carboxylic acid tert-butyl ester (INT-A102-7) (52%). LCMS: m/z (ESI), [M+H] += 639.30. 1H NMR (DMSO- d 6, 400 MHz) δ 0.87 (3H, d), 0.92-0.96 (1H, m), 1.27 (1H, d), 1.44 (11H, s), 2.72-2.77 (2H, m), 2.94-3.03 (1H, m), 3.09-3.16 (1H, m), 3.18 (1H, d), 3.50 (2H, s), 3.58 (3H, s), 3.66 (3H, s), 3.78 (3H, s), 3.88-3.92 (2H, t), 4.50 (2H, s), 6.72 (1H, d), 6.83 (1H, s), 7.78 (1H, d), 8.00 (1H, s), 8.11 (1H, d), 8.45 (1H, d). Step 8. 6-amino-5-{[(2 RPreparation of 5-({4-[5-(methoxycarbonyl)-1-methyl-2-oxopyridin-3-yl]-2-methylpyrazol-3-yl}oxy)-2-methylpentyl]amino}-3,4-dihydro-1H-isoquinoline-2-carboxylic acid tert-butyl ester (INT-A102-8). To a mixture of 5-{[(2R)-5-({4-[5-(methoxycarbonyl)-1-methyl-2-oxopyridin-3-yl]-2-methylpyrazol-3-yl}oxy)-2-methylpentyl]amino}-6-nitro-3,4-dihydro-1H-isoquinoline-2-carboxylic acid tert-butyl ester (700 mg, 1.09 mmol, 1 equiv.) and ranyl nickel (187 mg, 2.19 mmol, 2 equiv.) in methanol (8 mL), NH 2NH 2 .H 2O (109 mg, 2.19 mmol, 2 eq.). The mixture was stirred at 0 °C for 30 min under nitrogen atmosphere. The mixture was filtered and the filter cake was washed with methanol (3×40 mL). The resulting solution was concentrated under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (10/1) to give 400 mg of 6-amino-5-{[(2 R)-5-({4-[5-(methoxycarbonyl)-1-methyl-2-oxopyridin-3-yl]-2-methylpyrazol-3-yl}oxy)-2-methylpentyl]amino}-3,4-dihydro-1H-isoquinoline-2-carboxylic acid tert-butyl ester (INT-A102-8) (59%). LCMS: m/z (ESI), [M+H] +=609.30. Step 9. 5-(5-{[(4 RPreparation of methyl 6-oxopyridine-3-carboxylate (INT-A102-9). 6-amino-5-{[(2 RA mixture of tert-butyl 5-({4-[5-(methoxycarbonyl)-1-methyl-2-oxopyridin-3-yl]-2-methylpyrazol-3-yl}oxy)-2-methylpentyl]amino}-3,4-dihydro-1H-isoquinoline-2-carboxylate (400 mg, 0.65 mmol, 1 equiv) and BrCN (104 mg, 0.98 mmol, 1.5 equiv) in ethanol (5 mL) was stirred at room temperature under nitrogen atmosphere for 2 h. The reaction was quenched with water (50 mL), and the mixture was extracted with dichloromethane (3×50 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (12/1) to give 300 mg of 5-(5-{[(4 R)-4-{[2-amino-7-(tert-butoxycarbonyl)-6H,8H,9H-imidazo[4,5-f]isoquinolin-1-yl]methyl}pentyl]oxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A102-9) (72%). LCMS: m/z (ESI), [M+H] +=634.30. Step 10. 5-(5-{[(4 RPreparation of 5-(5-{[(4 R)-4-{[2-amino-7-(tert-butoxycarbonyl)-6H,8H,9H-imidazo[4,5-f]isoquinolin-1-yl]methyl}pentyl]oxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (300 mg, 0.47 mmol, 1 equivalent) and LiOH .H 2O (99 mg, 2.36 mmol, 5 equivalents) in tetrahydrofuran/H 2O (2 mL/0.5 mL) was stirred at room temperature for 2 h under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by C18 column using water and acetonitrile as mobile phase. The fractions containing the desired compound were evaporated to dryness to give 250 mg of 5-(5-{[(4R)-4-{[2-amino-7-(tert-butoxycarbonyl)-6H,8H,9H-imidazo[4,5-f]isoquinolin-1-yl]methyl}pentyl]oxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A102-10) (85%) as a white solid. LCMS: m/z (ESI), [M+H] +=620.40. Step 11. (11 RPreparation of 5-(5-{[(4 )-(5-{[(4 )-(5-{[(4 )-(5-{[(4 )-(5-{[(4 )-(5-{[(4 )-(5-{[(4 )-(5-{[(4 )-(4 )-(5-{[(4 )-(4 )-(5-{[(4 )-(4 )-(5-{[(4 )-(4 )-(5-{[(4 )-(4 )-(5-{[(4 )-(4 )-(5-{[(4 )-(4 )-(5-{[(4 )-(4 )-(5-{[(4 )-(4 )-(5-{[(4 )-(4 )-(4 )-(5-{[(4 )-(4 )-(5-{[(4 )R)-4-{[2-amino-7-(tert-butoxycarbonyl)-6H,8H,9H-imidazo[4,5-f]isoquinolin-1-yl]methyl}pentyl]oxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (200 mg, 0.32 mmol, 1 equivalent), N,N,N,N-Tetramethyl-O-(7-azabenzotriazol-1-yl)urea hexafluorophosphate (245 mg, 0.64 mmol, 2 equivalents) and N,N- A mixture of diisopropylethylamine (125 mg, 0.96 mmol, 3 equiv.) in 1,4-dioxane (5 mL) was stirred at 60 °C for 30 min under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (20 mL), and the mixture was extracted with dichloromethane (2×20 mL). The combined organic layers were washed with brine (3×30 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC eluting with (dichloromethane/methanol 12/1) to give 180 mg (11 R)-5,11,30-trimethyl-27,31-dioxo-7-oxa-4,5,13,18,24,26,30-heptaazahexacyclic [26.3.1.0^{2,6}.0^{13,25}. 0^{14,23}.0^{15,20}]triacontidine-1(32),2(6),3,14,20,22,24,28-octaene-18-carboxylic acid tert-butyl ester (INT-A102-11) (92%). LCMS: m/z (ESI), [M+H] +=602.30. Step 12. (11 R)-5,11,30-trimethyl-7-oxa-4,5,13,18,24, 26,30-heptaazahexacyclo[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]triacontac-1(32),2(6),3,14,20,22,24,28-octaene-27,31-dione (Example A102). A mixture of (11R)-5,11,30-trimethyl-27,31-dioxo-7-oxa-4,5,13,18,24,26,30-heptaazahexacyclo[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]triacontria-1(32),2(6),3,14,20,22,24,28-octaene-18-carboxylic acid tert-butyl ester (200 mg, 0.33 mmol, 1 eq.) in dichloromethane/trifluoroacetic acid (3 mL/1 mL) was stirred at room temperature for 30 min under nitrogen atmosphere. The reaction mixture was concentrated under reduced pressure. The residue was treated with saturated NaHCO3(aqueous solution) (50 mL) and extracted with dichloromethane (3×50 mL). The combined organic layers were washed with brine (3×50 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (10/1) to give 110 mg (11 R)-5,11,30-trimethyl-7-oxa-4,5,13,18,24,26,30-heptaazahexacyclo[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]triacontidine-1(32),2(6),3,14,20,22,24,28-octaene-27,31-dione (Example A102) (65%). LCMS: m/z (ESI), [M+H] +=502.45. 1H NMR (DMSO- d 6, 400 MHz) δ 0.81 (3H, d), 1.21-1.27 (1H, m), 1.41-1.45 (1H, m), 1.76-2.00 (4H, m), 2.35 (3H, s), 2.60-2.76 (2H, m), 3.52-3.58 (2H, m), 3.62 (3H, s), 3.71 (3H, s), 3.85-3.93 (1H, m), 4.11-4.27 (1H, m), 4.27-4.37 (1H, m), 6.91 (1H, d), 7.38 (1H, d), 8.28 (1H, d), 8.36 (1H, s), 8.77 (1H, d), 12.62 (1H, s). Step 13. Preparation of (11R)-5,11,18,30-tetramethyl-7-oxa-4,5,13,18, 24,26,30-heptaazahexacyclo[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}. 0^{15,20}]triacontidine-1(32),2(6),3,14,20,22,24,28-octaene-27,31-dione (Example A103). In a nitrogen atmosphere, at room temperature, (11 R)-5,11,30-trimethyl-7-oxa-4,5,13,18,24,26,30-heptaazahexacyclo[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]triacontidine-1(32),2(6),3,14,20,22,24,28-octaene-27,31-dione (Example A102, 50 mg, 0.10 mmol, 1 equivalent), paraformaldehyde (26 mg, 0.30 mmol, 3 equivalents) and N, N-Diisopropylethylamine (25 mg, 0.20 mmol, 2 equiv.) was stirred in dichloromethane (2 mL) and NaBH(OAc) was added.3(42 mg, 0.20 mmol, 2 equiv.). The reaction was quenched with water (50 mL), and the mixture was extracted with dichloromethane (3×50 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was subjected to prep-HPLC using an XBridge Prep OBD C18 column with 10 mmol/L NH4HCO 3of water and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 26 mg (11 R)-5,11,18,30-tetramethyl-7-oxa-4,5,13,18,24,26,30-heptaazahexacyclo[26.3.1.0^{2,6}. 0^{13,25}.0^{14,23}.0^{15,20}]triacontidine-1(32),2(6),3,14,20, 22,24,28-octaene-27,31-dione (Example A103) (50%). LCMS: m/z (ESI), [M+H] +=516.30 1H NMR (DMSO- d 6, 400 MHz) δ 0.81 (3H, d), 1.21-1.27 (1H, m), 1.41-1.45 (1H, m), 1.76-2.00 (3H, m), 2.35 (3H, s), 2.60-2.76 (2H, m), 3.21 (3H, s), 3.52-3.58 (2H, m), 3.62 (3H, s), 3.71 (3H, s), 3.85-3.93 (1H, m), 4.11-4.27 (1H, m), 4.27-4.37 (1H, m), 6.91 (1H, d), 7.38 (1H, d), 8.28 (1H, d), 8.36 (1H, s), 8.77 (1H, d), 12.62 (1H, s). Example A109 (12 R)-12,27-dimethyl-17-(4-methylpiperazine-1-yl)-8-oxo-14,21, 23,27-tetraazapentacyclic [23.3.1.0^{2,7}.0^{14,22}.0^{15,20}]twenty-nine-1(29),2(7),3,5,15,17,19,21,25-nonene-24,28-dione Step 1. Preparation of 5-(2-hydroxyphenyl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A109-1). 5-bromo-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (8 g, 32.51 mmol, 1 equivalent), 2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenol (7.87 g, 35.76 mmol, 1.1 equivalent), Pd(dppf)Cl 2(1.43 g, 1.95 mmol, 0.06 eq.) and K 2CO 3(8.99 g, 65.02 mmol, 2 equivalents) in 1,4-dioxane (240 mL) and H 2The mixture in 2% O (60 mL) was stirred at 80 °C under nitrogen atmosphere for 1.5 h. The mixture was cooled to room temperature. The reaction was quenched with water (500 mL), and the mixture was washed with CH2Cl 2(3×500 mL) extraction. The combined organic layers were washed with brine (3×500 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by prep-TLC using petroleum ether/ethyl acetate (3/1) to give 5.6 g of methyl 5-(2-hydroxyphenyl)-1-methyl-6-oxopyridine-3-carboxylate (INT-A109-1) (66%) as a brown solid. LCMS: m/z (ESI), [M+H] +=260.05. 1H NMR (DMSO-d 6, 400 MHz) δ 3.61 (3H, s), 3.82 (3H, s), 6.81-6.93 (2H, m), 7.18-7.22 (1H, m), 7.24-7.28 (1H, m), 7.86 (1H, d), 8.60 (1H, d), 9.39 (1H, s). Step 2. 5-(2-{[(4 RPreparation of methyl 5-[(5-bromo-2-nitrophenyl)amino]-4-methylpentyl]oxy}phenyl)-1-methyl-6-oxopyridine-3-carboxylate (INT-A109-2). 5-(2-Hydroxyphenyl)-1-methyl-6-oxopyridine-3-carboxylate (1 g, 3.86 mmol, 1 equivalent), 2-(tributyl-1^[5]-phosphinilidene)acetonitrile (4.65 g, 19.29 mmol, 5 equivalents) and (4 RA mixture of 5-[(5-bromo-2-nitrophenyl)amino]-4-methylpentan-1-ol (M3-7, 1.22 g, 3.86 mmol, 1 equiv.) in toluene (40 mL) was stirred at 130 °C for 1.5 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (200 mL), and the mixture was extracted with ethyl acetate (3×200 mL). The combined organic layers were washed with brine (3×200 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by prep-TLC using petroleum ether/ethyl acetate (3/1) to give 1.7 g of 5-(2-{[(4 R)-5-[(5-bromo-2-nitrophenyl)amino]-4-methylpentyl]oxy}phenyl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A109-2) (79%). LCMS: m/z (ESI), [M+H] +=558.10. Step 3. 5-(2-{[(4 RPreparation of methyl 5-[(2-amino-5-bromophenyl)amino]-4-methylpentyl]oxy}phenyl)-1-methyl-6-oxopyridine-3-carboxylate (INT-A109-3). At 0°C, 5-(2-{[(4RTo a stirred mixture of methyl 5-[(5-bromo-2-nitrophenyl)amino]-4-methylpentyl]oxy}phenyl)-1-methyl-6-oxopyridine-3-carboxylate (800 mg, 1.43 mmol, 1 equiv) and ranyl nickel (80 mg, 0.93 mmol, 0.65 equiv) in methanol (35 mL) was added NH2NH 2 .H 2O (143 mg, 2.87 mmol, 2 eq.). The resulting mixture was stirred at room temperature for 2 h. The reaction was quenched with water (100 mL), and the mixture was extracted with ethyl acetate (3×80 mL). The combined organic layers were washed with brine (3×80 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using petroleum ether/ethyl acetate (1/5) to give 690 mg of 5-(2-{[(4 R)-5-[(2-amino-5-bromophenyl)amino]-4-methylpentyl]oxy}phenyl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A109-3) (91%). LCMS: m/z (ESI), [M+H] +=528.00. Step 4. 5-(2-{[(4 RPreparation of methyl 4-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]pentyl]oxy}phenyl)-1-methyl-6-oxopyridine-3-carboxylate (INT-A109-4). 5-(2-{[(4 RA mixture of methyl 5-[(2-amino-5-bromophenyl)amino]-4-methylpentyl]oxy}phenyl)-1-methyl-6-oxopyridine-3-carboxylate (1.3 g, 2.46 mmol, 1 equiv) and BrCN (521 mg, 4.92 mmol, 2 equiv) in dichloromethane (30 mL) was stirred at room temperature for 4 h. The reaction was quenched with water (200 mL), and the mixture was extracted with dichloromethane (3×200 mL). The combined organic layers were washed with brine (3×200 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (20/1) to give 1.32 g of 5-(2-{[(4 R)-4-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]pentyl]oxy}phenyl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A109-4) (97%). LCMS: m/z (ESI), [M+H] +=554.95. Step 5. 5-(2-{[(4 RPreparation of 5-(2-{[(4 )-4-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]pentyl]oxy}phenyl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A109-5).RTo a stirred mixture of methyl 4-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]pentyl]oxy}phenyl)-1-methyl-6-oxopyridine-3-carboxylate (1.3 g, 2.35 mmol, 1 eq.) in tetrahydrofuran (10 mL) were added LiOH (225.02 mg, 9.39 mmol, 4 eq.) and H 2O (4 mL). The resulting mixture was stirred at room temperature for 1 h and concentrated under vacuum. The residue was purified by C18 FLASH chromatography using water and acetonitrile as mobile phases. The fractions containing the desired compound were evaporated to dryness to give 900 mg of 5-(2-{[(4 R)-4-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]pentyl]oxy}phenyl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A109-5) (71%). LCMS: m/z (ESI), [M+H] +=540.95. Step 6. (12 R)-17-bromo-12,27-dimethyl-8-oxa-14,21,23,27-tetraazapentacyclic [23.3.1.0^{2,7}.0^{14,22}.0^{15,20}]twenty-nine-1(29),2(7),3,5,15,17,19,21,25-nonene-24,28-dione (INT-A109-6). At room temperature, 5-(2-{[(4 R)-4-[(2-amino-6-bromo-1,3-benzodiazol-1-yl)methyl]pentyl]oxy}phenyl)-1-methyl-6-oxopyridine-3-carboxylic acid (450 mg, 0.83 mmol, 1 equivalent) was added to the stirred mixture in dioxane (10 mL) N,N-Diisopropylethylamine (323 mg, 2.50 mmol, 3 equiv.) and N,N,N,N-Tetramethyl- O-(7-Azabenzotriazole-1-yl)uronium hexafluorophosphate (476 mg, 1.25 mmol, 1.5 equiv). The resulting mixture was stirred at 60 °C for 1 h under a nitrogen atmosphere and cooled to room temperature. The reaction was quenched with water (100 mL), and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by prep-TLC eluting with methylenedichloromethane/methanol (30/1) to give 350 mg (12 R)-17-bromo-12,27-dimethyl-8-oxa-14,21,23,27-tetraazapentacyclic [23.3.1.0^{2,7}.0^{14,22}. 0^{15,20}]twenty-nine-1(29),2(7),3,5,15,17,19,21,25-nonene-24,28-dione (INT-A109-6) (80%). LCMS: m/z (ESI), [M+H] +=523.05. Step 7. (12 R)-12,27-dimethyl-17-(4-methylpiperazine-1-yl)-8-oxa-14,21,23,27-tetraazapentacyclic [23.3.1.0^{2,7}.0^{14,22}. 0^{15,20}]twenty-nine-1(29),2(7),3,5,15,17,19,21,25-nonene-24,28-dione (Example A109). In a nitrogen atmosphere at room temperature to (12 RTo a stirred mixture of )-17-bromo-12,27-dimethyl-8-oxa-14,21,23,27-tetraazapentacyclo[23.3.1.0^{2,7}.0^{14,22}.0^{15,20}]nonaconicotin-1(29),2(7), 3,5,15,17,19,21,25-nonene-24,28-dione (200 mg, 0.38 mmol, 1 eq), 1-methylpiperazine (127 mg, 1.27 mmol, 3.31 eq) and BrettPhos Pd G3 (90 mg, 0.10 mmol, 0.26 eq) in 1,4-dioxane (10 mL) was added LiHMDS (3.8 mL, 3.8 mmol, 10 eq) dropwise. The resulting mixture was stirred at 60 °C for 50 min and cooled to room temperature. The reaction was heated with saturated NH4Cl(aqueous solution) (100 mL) was quenched, and the mixture was extracted with dichloromethane (3×100 mL). The combined organic layers were washed with brine (3×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by prep-TLC using dichloromethane/methanol (30/1) for elution, and the product was purified by prep-HPLC using an XBridge Prep OBD C18 column with 10 mmol/L NH4HCO 3The product was further purified using water and acetonitrile as the mobile phase. The fractions containing the desired compound were evaporated to dryness to give 13.7 mg (12 R)-12,27-dimethyl-17-(4-methylpiperazin-1-yl)-8-oxa-14,21,23,27-tetraazapentacyclic [23.3.1.0^{2,7}.0^{14,22}.0^{15,20}]twenty-nine-1(29),2(7),3,5,15,17,19,21,25-nonene-24,28-dione (Example A109) (7%). LCMS: m/z (ESI), [M+H] +=541.35. 1H NMR (DMSO-d 6, 400 MHz) δ 0.92 (3H, d), 1.29-1.33 (1H, m), 1.60-1.66 (1H, m), 2.02-2.06 (2H, m), 2.24 (3H, s), 2.26-2.30 (1H, m), 2.49-2.51 (4H, m), 3.14 (4H, t), 3.60 (3H, s), 3.93-4.08 (3H, m), 4.20 (1H, t), 6.83-6.89 (1H, m), 6.86 (1H, dd), 6.92-6.96 (1H, m), 7.12 (1H, d), 7.25-7.29 (1H, m), 7.34 (1H, d), 7.69 (1H, dd), 8.33 (1H, d), 8.85 (1H, d), 12.27 (1H, s). Example A110 (12 R)-4-fluoro-12,27-dimethyl-17-(4-methylpiperazine-1-yl)-8-oxo-14,21,23,27-tetraazapentacyclic [23.3.1.0^{2,7}.0^{14,22}. 0^{15,20}]twenty-nine-1(29),2(7),3,5,15,17,19,21,25-nonene-24,28-dione Step 1. Preparation of 5-(5-fluoro-2-hydroxyphenyl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A110-1). 5-Bromo-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (2 g, 8.128 mmol, 1 equivalent), 4-fluoro-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenol (2.32 g, 9.754 mmol, 1.2 equivalents), Pd(dppf)Cl 2(0.48 g, 0.656 mmol, 0.08 eq.) and K 2CO 3(2.81 g, 20.320 mmol, 2.5 equiv.) in dioxane (40 mL) and H 2The mixture in 2% O (10 mL) was stirred at 80 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (150 mL), and the mixture was extracted with dichloromethane (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue is purified by silica gel chromatography eluting with dichloromethane/methanol (15/1) to give 1 g of methyl 5-(5-fluoro-2-hydroxyphenyl)-1-methyl-6-oxopyridine-3-carboxylate (INT-A110-1) (44%) as a yellow solid. 1H NMR (DMSO-d 6, 400 MHz) 3.60 (3H, s), 3.81 (3H, s), 6.84-6.95 (1H,m), 6.96-7.08 (1H, m), 7.14 (1H, d), 7.94 (1H, d), 8.61 (1H, d), 9.41 (1H, s) Step 2. 5-(5-fluoro-2-{[(4R)-4-methyl-5-{[5-(4-methylpiperazin-1-yl)-2-nitrophenyl]amino}pentyl]oxy}phenyl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A110-2). 5-(5-fluoro-2-hydroxyphenyl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (526 mg, 1.90 mmol, 1 equivalent), (4 RA mixture of )-4-methyl-5-{[5-(4-methylpiperazin-1-yl)-2-nitrophenyl]amino}pentan-1-ol (639.07 mg, 1.90 mmol, 1 equiv) and 2-(tributyl-1^[5]-phosphinilidene)acetonitrile (2.30 g, 9.50 mmol, 5 equiv) in toluene (5 mL) was stirred at 130 °C overnight under a nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (100 mL), and the mixture was extracted with dichloromethane (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using petroleum ether/ethyl acetate (1/1) to give 500 mg of 5-(5-fluoro-2-{[(4R)-4-methyl-5-{[5-(4-methylpiperazin-1-yl)-2-nitrophenyl]amino}pentyl]oxy}phenyl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A110-2) (44%) as a white solid. 1H NMR (DMSO-d 6 , 400 MHz) 0.90 (3H, s), 1.31-1.39 (4H, m), 2.20 (3H, s), 2.32-2.41 (4H, m), 2.44 (1H, t), 2.98-3.28 (2H, m), 3.40 (4H, t), 3.56 (3H, s), 3.76 (3H, s), 3.95 (2H, t), 5.77 (1H, s), 5.96 (1H, d), 6.43 (1H, d), 6.97-7.29 (4H, m), 7.83-7.95 (2H, m), 8.05 (1H, d), 8.39 (1H, t), 8.53 (1H, d). Step 3. 5-(2-{[(4 RPreparation of methyl 5-{[2-amino-5-(4-methylpiperazine-1-yl)phenyl]amino}-4-methylpentyl]oxy}-5-fluorophenyl)-1-methyl-6-oxopyridine-3-carboxylate (INT-A110-3). At 0°C, 5-(5-fluoro-2-{[(4 RTo a stirred mixture of methyl 4-(4-methyl-5-{[5-(4-methylpiperazin-1-yl)-2-nitrophenyl]amino}pentyl]oxy}phenyl)-1-methyl-6-oxopyridine-3-carboxylate (500 mg, 0.83 mmol, 1 eq.) and ranyl nickel (3.60 mg, 0.042 mmol, 0.05 eq.) in methanol (20 mL) was added hydrazine (126.06 mg, 2.51 mmol, 3 eq.). The mixture was stirred at 0 °C for 30 min under nitrogen atmosphere. The obtained mixture was filtered, and the filter cake was washed with methanol (3×30 mL). The obtained solution was concentrated under reduced pressure. The residue was purified by C18 FLASH chromatography using water and acetonitrile as mobile phase. The fractions containing the desired compound were evaporated to dryness to give 400 mg of methyl 5-(2-{[(4R)-5-{[2-amino-5-(4-methylpiperazin-1-yl)phenyl]amino}-4-methylpentyl]oxy}-5-fluorophenyl)-1-methyl-6-oxopyridine-3-carboxylate (INT-A110-3) (84%) as a yellow solid. LCMS: m/z (ESI), [M+H] += 566.15. Step 4. 5-(2-{[(4 RPreparation of 5-(2-{[(4 RA mixture of methyl 5-{[2-amino-5-(4-methylpiperazin-1-yl)phenyl]amino}-4-methylpentyl]oxy}-5-fluorophenyl)-1-methyl-6-oxopyridine-3-carboxylate (240 mg, 0.42 mmol, 1 eq.) and BrCN (51 mg, 0.46 mmol, 1.1 eq.) in dichloromethane (5 mL) was stirred at room temperature for 4 h under a nitrogen atmosphere and concentrated under reduced pressure. The residue was purified by Prep-TLC eluting with dichloromethane/methanol (1/1) to obtain 140 mg of 5-(2-{[(4 R)-4-{[2-amino-6-(4-methylpiperazine-1-yl)-1,3-benzodiazol-1-yl]methyl}pentyl]oxy}-5-fluorophenyl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A110-4) (55%). LCMS: m/z (ESI), [M+H] +=591.15. Step 5. 5-(2-{[(4 RPreparation of 5-(2-{[(4 R)-4-{[2-amino-6-(4-methylpiperazine-1-yl)-1,3-benzodiazol-1-yl]methyl}pentyl]oxy}-5-fluorophenyl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (75 mg, 0.12 mmol, 1 equivalent) and LiOH .H 2A mixture of 5-(2-[(4-(2-[(4-(2-[(4-(2-[(4-(2-[(4-(2-[(4-(2-[(4-(2-[(4-(2-[(4-(2-R)-4-{[2-amino-6-(4-methylpiperazine-1-yl)-1,3-benzodiazol-1-yl]methyl}pentyl]oxy}-5-fluorophenyl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A110-5) (54%). LCMS: m/z (ESI), [M+H] +=577.25. Step 6. (12 R)-4-fluoro-12,27-dimethyl-17-(4-methylpiperazine-1-yl)-8-oxa-14,21,23,27-tetraazapentacyclic [23.3.1.0^{2,7}. 0^{14,22}.0^{15,20}]twenty-nine-1(29),2(7),3,5,15,17,19,21, 25-nonene-24,28-dione (Example A110). 5-(2-{[(4 R)-4-{[2-amino-6-(4-methylpiperazine-1-yl)-1,3-benzodiazol-1-yl]methyl}pentyl]oxy}-5-fluorophenyl)-1-methyl-6-oxopyridine-3-carboxylic acid (35 mg, 0.06 mmol, 1 equivalent), N,N,N,N-Tetramethyl- O-(7-Azabenzotriazol-1-yl)urea hexafluorophosphate (34 mg, 0.09 mmol, 1.5 equivalents) and N,N- A mixture of diisopropylethylamine (23 mg, 0.18 mmol, 3 equiv.) in 1,4-dioxane (2 mL) was stirred at room temperature under nitrogen atmosphere for 2 h. The reaction was quenched with water (50 mL), and the mixture was extracted with dichloromethane (3×50 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was subjected to prep-HPLC using an XBridge Shield RP18 OBD column with 10 mmol/L NH4HCO 3and 0.1% NH 3.H 2O water and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to obtain 3.6 mg (12 R)-4-fluoro-12,27-dimethyl-17-(4-methylpiperazine-1-yl)-8-oxa-14,21,23,27-tetraazapentacyclic [23.3.1.0^{2,7}. 0^{14,22}.0^{15,20}]twenty-nine-1(29),2(7),3,5,15,17,19,21, 25-nonene-24,28-dione (Example A110) (10%). LCMS: m/z (ESI), [M+H] +=559.40. 1H NMR (DMSO-d 6 , 400 MHz) δ 0.94 (3H, d), 1.31-1.40 (1H, m), 1.64-1.70 (1H, m), 2.01-2.14 (2H, m), 2.21-2.30 (1H, m), 2.41 (3H, s), 2.66-2.73 (4H, m), 3.22 (4H, t), 3.60 (3H, s), 3.76-3.85 (1H, m), 3.99-4.09 (2H, m), 4.13 (1H, s), 6.87-6.97 (2H, m), 6.98-7.09 (2H, m), 7.24 (1H, d), 7.47-7.55 (1H, m), 8.23 (1H, s), 8.91 (1H, s). Example A111 (12 R)-4,5-difluoro-12,27-dimethyl-17-(4-methylpiperazine-1-yl)-8-oxo-14,21,23,27-tetraazapentacyclic [23.3.1.0^{2,7}.0^{14,22}. 0^{15,20}]twenty-nine-1(29),2(7),3,5,15,17,19,21,25-nonene-24,28-dione Step 1. Preparation of methyl 5-(4,5-difluoro-2-hydroxyphenyl)-1-methyl-6-oxopyridine-3-carboxylate (INT-A111-1). To a stirred mixture of methyl 5-bromo-1-methyl-6-oxopyridine-3-carboxylate (2.00 g, 8.12 mmol, 1 eq.) and 4,5-difluoro-2-hydroxyphenylboronic acid (2.12 g, 12.19 mmol, 1.5 eq.) in 1,4-dioxane (32 mL) and water (8 mL) were added portionwise 1,1'-bis(diphenylphosphino)ferrocene-palladium(II) (1.19 g, 1.62 mmol, 0.2 eq.) and K 2CO 3(2.81 g, 20.32 mmol, 2.5 equiv.). The resulting mixture was stirred at 80 °C for 2 h. The mixture was cooled to room temperature. The reaction was quenched with water (100 mL), and the mixture was extracted with dichloromethane (3×80 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was subjected to C18 FLASH chromatography using a 10 mmol/L NH4HCO 3and 0.1% NH 3 .H 2Purification was performed using water with 0.04% 2-nitropropane and acetonitrile as the mobile phase. The fractions containing the desired compound were evaporated to dryness to give 1.38 g of methyl 5-(4,5-difluoro-2-hydroxyphenyl)-1-methyl-6-oxopyridine-3-carboxylate (INT-A111-1) (57%) as a white solid. 1H NMR (DMSO-d 6, 400 MHz) δ 3.59 (3H, s), 3.81 (3H, s), 6.81-6.91 (1H, m), 7.33-7.41 (1H, d), 7.92 (1H, d), 8.61 (1H, d), 9.90 (1H, s). Step 2. 5-(4,5-difluoro-2-{[(4 RPreparation of methyl 5-(4,5-difluoro-2-hydroxyphenyl)-1-methyl-6-oxopyridine-3-carboxylate (INT-A111-2). Methyl 5-(4,5-difluoro-2-hydroxyphenyl)-1-methyl-6-oxopyridine-3-carboxylate (500 mg, 1.69 mmol, 1 equivalent) and (4 R)-4-methyl-5-{[5-(4-methylpiperazin-1-yl)-2-nitrophenyl]amino}pentan-1-ol (INT-A25-3, 569 mg, 1.69 mmol, 1 equiv) was added in portions to a stirred mixture of toluene (7 mL) with 2-(tributyl-λ5-phosphinidylidene)acetonitrile (2.04 g, 8.47 mmol, 5 equiv). The resulting mixture was stirred at 80 °C for 2 h. The mixture was cooled to room temperature. The reaction was quenched with water (200 mL), and the mixture was extracted with dichloromethane (3×300 mL). The combined organic layers were washed with brine (3×200 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. Pass the residue through C18 FLASH using 10 mmol/L NH4HCO 3and 0.1% NH 3 .H 2O and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 620 mg of 5-(4,5-difluoro-2-{[(4R)-4-methyl-5-{[5-(4-methylpiperazin-1-yl)-2-nitrophenyl]amino}pentyl]oxy}phenyl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A111-2) (59%) as a yellow oil. LCMS: m/z (ESI), [M+H] += 614.25. 1H NMR (DMSO-d 6, 400 MHz) δ 0.91 (4H, d), 1.20-1.32 (1H, m), 1.59-1.72 (3H, m), 1.75-1.87 (1H, m), 2.32 (3H, d), 3.04-3.13 (1H, m), 3.19-3.25 (1H, m), 3.320-3.36 (8H, m), 3.56 (3H, s), 3.76 (3H, s), 3.97 (3H, t), 5.76 (1H, s), 5.98 (1H, s), 6.40-6.46 (1H, m), 7.18-7.28 (1H, m), 7.40-7.50 (1H, m), 7.84-7.95 (2H, m), 8.37 (1H, t), 8.53 (1H, d). Step 3. 5-(2-{[(4 RPreparation of methyl 5-{[2-amino-5-(4-methylpiperazine-1-yl)phenyl]amino}-4-methylpentyl]oxy}-4,5-difluorophenyl)-1-methyl-6-oxopyridine-3-carboxylate (INT-A111-3). In a nitrogen atmosphere, 5-(4,5-difluoro-2-{[(4 RTo a stirred mixture of methyl 4-(2-(4-methylpiperazin-1-yl)-2-nitrophenyl]amino}pentyl]oxy}phenyl)-1-methyl-6-oxopyridine-3-carboxylate (600 mg, 0.97 mmol, 1 eq.) and ranyl nickel (251 mg, 2.93 mmol, 3 eq.) in methanol (5 mL) was added hydrazine hydrate (98%) (2.45 mg, 0.04 mmol, 3 eq.). The resulting mixture was stirred at 0 °C for 30 min under a nitrogen atmosphere. The mixture was filtered, and the filter cake was washed with methanol (3×10 mL). The resulting solution was concentrated under reduced pressure to give 310 mg of 5-(2-{[(4 R)-5-{[2-amino-5-(4-methylpiperazin-1-yl)phenyl]amino}-4-methylpentyl]oxy}-4,5-difluorophenyl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A111-3). This material was used in the next reaction without further purification. LCMS: m/z (ESI), [M+H] +=582.25. Step 4. 5-(2-{[(4 RPreparation of methyl 4-{[2-amino-6-(4-methylpiperazine-1-yl)-1,3-benzodiazol-1-yl]methyl}pentyl]oxy}-4,5-difluorophenyl)-1-methyl-6-oxopyridine-3-carboxylate (INT-A111-4). 5-(2-{[(4 RA mixture of methyl 5-{[2-amino-5-(4-methylpiperazin-1-yl)phenyl]amino}-4-methylpentyl]oxy}-4,5-difluorophenyl)-1-methyl-6-oxopyridine-3-carboxylate (700 mg, 1.20 mmol, 1 equiv) and cyanogen bromide (190 mg, 1.80 mmol, 1.5 equiv) in dichloromethane (10 mL) was stirred at room temperature under nitrogen atmosphere for 2 h. The reaction was quenched with water (10 mL), and the mixture was extracted with dichloromethane (3×60 mL). The combined organic layers were washed with brine (3×20 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. Pass the residue through C18FLASH using 10 mmol/L NH4HCO 3and 0.1% NH 3 .H 2O and acetonitrile water as the mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 120 mg of 5-(2-{[(4 R)-4-{[2-amino-6-(4-methylpiperazine-1-yl)-1,3-benzodiazol-1-yl]methyl}pentyl]oxy}-4,5-difluorophenyl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A111-4) (16%). LCMS: m/z (ESI), [M+H] +=609.15. Step 5. Preparation of 5-(2-{[(4R)-4-{[2-amino-6-(4-methylpiperazine-1-yl)-1,3-benzodiazol-1-yl]methyl}pentyl]oxy}-4,5-difluorophenyl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A111-5). 5-(2-{[(4 RA mixture of methyl 4-{[2-amino-6-(4-methylpiperazine-1-yl)-1,3-benzodiazol-1-yl]methyl}pentyl]oxy}-4,5-difluorophenyl)-1-methyl-6-oxopyridine-3-carboxylate (100 mg, 0.16 mmol, 1 equiv) and lithium hydroxide monohydrate (10 mg, 0.24 mmol, 1.5 equiv) in tetrahydrofuran (2 mL) and water (0.5 mL) was stirred at room temperature under nitrogen atmosphere for 2 h. The reaction was quenched with water (5 mL), and the mixture was extracted with dichloromethane (3×10 mL). The combined organic layers were washed with brine (3×5 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (10/1) to give 50 mg of 5-(2-{[(4 R)-4-{[2-amino-6-(4-methylpiperazine-1-yl)-1,3-benzodiazol-1-yl]methyl}pentyl]oxy}-4,5-difluorophenyl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A111-5) (51%). LCMS: m/z (ESI), [M+H] +=595.20. Step 6. (12 R)-4,5-difluoro-12,27-dimethyl-17-(4-methylpiperazine-1-yl)-8-oxa-14,21,23,27-tetraazapentacyclic [23.3.1.0^{2,7}. 0^{14,22}.0^{15,20}]twenty-nine-1(29),2(7),3,5,15,17,19,21, 25-nonene-24,28-dione (Example A111). 5-(2-{[(4 R)-4-{[2-amino-6-(4-methylpiperazine-1-yl)-1,3-benzodiazol-1-yl]methyl}pentyl]oxy}-4,5-difluorophenyl)-1-methyl-6-oxopyridine-3-carboxylic acid (40 mg, 0.06 mmol, 1 equivalent), N,N,N,N-Tetramethyl- O-(7-Azabenzotriazol-1-yl)urea hexafluorophosphate (38 mg, 0.10 mmol, 1.5 equivalents) and N,N- A mixture of diisopropylethylamine (26 mg, 0.20 mmol, 3 equiv.) in 1,4-dioxane (4 mL) was stirred at room temperature under nitrogen atmosphere for 3 h. The reaction was quenched with water (10 mL), and the mixture was extracted with dichloromethane (3×15 mL). The combined organic layers were washed with brine (3×10 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (5/1) to give 9 mg (12 R)-4,5-difluoro-12,27-dimethyl-17-(4-methylpiperazine-1-yl)-8-oxa-14,21,23,27-tetraazapentacyclic [23.3.1.0^{2,7}.0^{14,22}. 0^{15,20}]twenty-nine-1(29),2(7),3,5,15,17,19,21,25-nonene-24,28-dione (Example A111) (22%). LCMS: m/z (ESI), [M+H] +=577.30. 1H NMR (DMSO-d 6, 400 MHz) δ 0.92 (3H, d), 1.15-1.65 (3H, m), 2.00-2.10 (2H, m), 2.35-2.45 (4H, m), 2.50 (3H, s), 3.10-3.30 (4H, m), 3.32 (3H, s), 3.91-3.97 (2H, m), 4.05 (1H, d), 4.19 (1H, t), 6.88 (1H, dd), 7.11 (1H, s), 7.26 (1H, dd), 7.35 (1H, d), 7.76 (1H, dd), 8.38 (1H, d), 8.89 (1H, d), 12.31 (1H, s) Example A112 5,12,12,26-Tetramethyl-16-(4-methylpiperazin-1-yl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]octadecene-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione Step 1. NPreparation of tert-butyl-[(3E)-6-(benzyloxy)-2-methylhex-3-en-2-yl]carbamate (INT-A112-1). To a stirred mixture of (3-benzyloxypropyl)triphenylphosphonium bromide (14.5 g, 30 mmol, 1.1 equiv) in tetrahydrofuran (100 mL) at 0°C was added KHMDS (5.9 g, 30 mmol, 1.1 equiv) dropwise under nitrogen atmosphere. The resulting mixture was stirred at 0°C for 1 h under nitrogen atmosphere. To the above mixture was added N-tert-Butyl (2-methyl-1-oxopropan-2-yl)carbamate (5 g, 26.7 mmol, 1 equiv). The resulting mixture was stirred at room temperature overnight. The reaction was quenched with water (50 mL), and the mixture was extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using petroleum ether/ethyl acetate (5/1) to give 6 g of colorless oil.N-[(3E)-6-(Benzyloxy)-2-methylhex-3-en-2-yl]carbamic acid tert-butyl ester (INT-A112-1) (70%).1H NMR (CDCl 3, 400 MHz) δ 1.44 (9H, s), 1.46 (6H, s), 2.50-2.62 (2H, m), 3.45-3.52 (2H, m), 4.56 (2H, s), 5.32-5.42 (1H, m), 5.56-6.02 (1H, m), 7.29-7.39 (5H, m). Step 2. NPreparation of tert-butyl-(6-hydroxy-2-methylhexan-2-yl)carbamate (INT-A112-2).NA mixture of tert-butyl-[(3E)-6-(benzyloxy)-2-methylhex-3-en-2-yl]carbamate (2 g, 6.26 mmol, 1 equiv) and Pd/C (3.3 g, 31 mmol, 5 equiv) in methanol (40 mL) was stirred at room temperature under a hydrogen atmosphere for 16 h. The mixture was filtered and the filter cake was washed with methanol (3×40 mL). The resulting solution was concentrated under reduced pressure to give 1.4 g of N-tert-Butyl (6-hydroxy-2-methylhexan-2-yl)carbamate (INT-A112-2) (97%). 1H NMR (CDCl 3, 400 MHz) δ1.26 (6H, s), 1.32-1.41 (2H, m), 1.45 (9H, s), 1.60 (2H, d), 1.65-1.72 (2H, m), 3.62-3.73 (2H, m). Step 3. Preparation of 5-amino-5-methylhexan-1-ol (INT-A112-3). NA solution of tert-butyl-(6-hydroxy-2-methylhexan-2-yl)carbamate (1.3 g, 5.6 mmol, 1 equiv) in trifluoroacetic acid (4 mL) and dichloromethane (16 mL) was stirred at room temperature under nitrogen atmosphere for 2 h. The mixture was concentrated under reduced pressure. The residue was treated with saturated NaHCO3(aqueous solution) (30 mL) and extracted with dichloromethane (3×30 mL). The combined organic layers were washed with brine (3×30 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC eluting with dichloromethane/methanol (10/1) to give 680 mg of 5-amino-5-methylhexan-1-ol (INT-A112-3) (92%) as a colorless oil. 1H NMR (CD 3OD, 400 MHz) δ 1.35 (6H, d), 1.48-1.55 (2H, m), 1.65-1.72 (2H, m), 1.80-1.86 (2H, m), 4.40-4.49 (2H, m). Step 4. Preparation of 5-((5-bromo-2-nitrophenyl)amino)-5-methylhexan-1-ol (INT-A112-4). 5-Amino-5-methylhexan-1-ol (680 mg, 5.18 mmol, 1 equivalent), K 2CO 3A mixture of (2150 mg, 15.52 mmol, 3 eq.) and 4-bromo-2-fluoro-1-nitrobenzene (1140 mg, 5.18 mmol, 1 eq.) in acetonitrile was stirred at 60 °C for 16 h and cooled to room temperature. The resulting mixture was filtered and the filter cake was washed with dichloromethane (3×20 mL). The resulting solution was concentrated under reduced pressure. The residue was purified by Prep-TLC using petroleum ether/ethyl acetate (1/1) to give 1.3 g of 5-((5-bromo-2-nitrophenyl)amino)-5-methylhexan-1-ol (INT-A112-4) (76%) as a yellow oil. LCMS: m/z (ES+), [M+H ] +=331.00. Step 5. Preparation of 5-(5-((5-((5-bromo-2-nitrophenyl)amino)-5-methylhexyl)oxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxo-1,6-dihydropyridine-3-carboxylate (INT-A112-5). Methyl 5-(5-hydroxy-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (M2, 509 mg, 1.93 mmol, 1 equivalent) and PPh were added to the mixture at 0 °C under a nitrogen atmosphere.3To a stirred mixture of 5-((5-bromo-2-nitrophenyl)amino)-5-methylhexan-1-ol (638 mg, 1.93 mmol, 1 eq.) and diisopropyl azodicarboxylate (1172 mg, 5.80 mmol, 3 eq.) (1520 mg, 5.80 mmol, 3 eq.) in tetrahydrofuran (20 mL) were added. The resulting mixture was stirred at room temperature for 1 h. The reaction was quenched with water (100 mL), and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×60 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (20/1) to give 610 mg of 5-[5-({5-[(5-bromo-2-nitrophenyl)amino]-5-methylhexyl}oxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A112-5) (55%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=578.30. Step 6. Preparation of 5-(5-((5-((2-amino-5-bromophenyl)amino)-5-methylhexyl)oxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxo-1,6-dihydropyridine-3-carboxylic acid methyl ester (INT-A112-6). 5-[5-({5-[(5-bromo-2-nitrophenyl)amino]-5-methylhexyl}oxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (600 mg, 1.04 mmol, 1 equivalent) and NH 2NH 2 .H 2To a stirred mixture of 2-nitropropene (104 mg, 2.08 mmol, 2 eq.) in methanol (20 mL) was added Raney nickel (445.88 mg, 5.21 mmol, 5 eq.). The resulting mixture was stirred at room temperature for 2 h. The resulting mixture was filtered, and the filter cake was washed with methanol (3×20 mL). The resulting solution was concentrated under reduced pressure to give 450 mg of methyl 5-(5-((5-((2-amino-5-bromophenyl)amino)-5-methylhexyl)oxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxo-1,6-dihydropyridine-3-carboxylate (INT-A112-6) as a yellow solid. This material was used in the next reaction without further purification. LCMS: m/z (ESI), [M+H] +=546.05. Step 7. Preparation of 5-(5-((5-(2-amino-6-bromo-1H-benzo[d]imidazol-1-yl)-5-methylhexyl)oxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxo-1,6-dihydropyridine-3-carboxylic acid methyl ester (INT-A112-7). A mixture of 5-[5-({5-[(2-amino-5-bromophenyl)amino]-5-methylhexyl}oxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (450 mg, 0.82 mmol, 1 eq.) and BrCN (174 mg, 1.65 mmol, 2 eq.) in ethanol (15 mL) was stirred at room temperature for 2 h. The reaction was quenched with water (20 mL), and the mixture was extracted with dichloromethane (3×20 mL). The combined organic layers were washed with brine (3×20 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (10/1) to give 300 mg of 5-(5-{[5-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-5-methylhexyl]oxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A112-7) (64%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=573.25. 1H NMR (DMSO, 400 MHz) δ 1.24 (2H, s), 1.66-1.72 (2H, m), 1.77 (6H, s), 1.99-2.07 (2H, m), 3.58 (6H, s), 3.80 (3H, s), 3.85-3.90 (2H, m), 6.31 (2H, s), 7.10 (2H, d), 7.61 (1H, s), 7.97 (1H, s), 8.09 (1H, d), 8.45 (1H, d). Step 8. Preparation of 5-(5-((5-(2-amino-6-bromo-1,3-benzo[d]imidazol-1-yl)-5-methylhexyl)oxy)-1-methyl-1H-pyrazol-4-yl)-1-methyl-6-oxo-1,6-dihydropyridine-3-carboxylic acid (INT-A112-8). 5-[5-({5-[(aminomethyl)(3-bromophenyl)amino]-5-methylhexyl}oxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (280 mg, 0.5 mmol, 1 eq.) was added to THF (6 mL) and H 2Add LiOH to the stirred mixture in O (1.5 mL).H 2O (63 mg, 1.5 mmol, 3 equiv.). The resulting mixture was stirred at room temperature under a nitrogen atmosphere for 16 h and concentrated under reduced pressure. The residue was passed through a C18 FLASH column using a 5% HPLC-MS/MS column containing 10 mmol/L NH4HCO 3and 0.1% NH 3 .H 2O and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 150 mg of 5-(5-{[5-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-5-methylhexyl]oxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A112-8) (54%) as a white solid. LCMS: m/z (ESI), [M+H] +=557.25. Step 9. Preparation of 16-bromo-5,12,12,26-tetramethyl-7-oxa-4,5,13,20,22, 26-hexaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octadecene-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (INT-A112-9). 5-(5-{[5-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-5-methylhexyl]oxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (150 mg, 0.27 mmol, 1 equivalent), N,N-Diisopropylethylamine (104 mg, 0.81 mmol, 3 equivalents) and N,N,N,NA mixture of 2-tetramethyl-O-(7-azabenzotriazol-1-yl)uronium hexafluorophosphate (205 mg, 0.54 mmol, 2 equiv.) in dioxane (5 mL) was stirred at 60 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (20 mL), and the mixture was extracted with dichloromethane (3×20 mL). The combined organic layers were washed with brine (3×20 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by silica gel chromatography using dichloromethane/methanol (10/1) to give 140 mg of 16-bromo-5,12,12,26-tetramethyl-7-oxa-4,5,13,20,22,26-hexaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octacosa-1(28),2(6),3, 14,16,18,20,24-octaene-23,27-dione (INT-A112-9) (96%) as a white solid. LCMS: m/z (ESI), [M+H] +=539.20 Step 10. Preparation of 5,12,12,26-tetramethyl-16-(4-methylpiperazin-1-yl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octadeca-1(28),2(6),3,14,16,18,20,24-octaene-23,27-dione (Example A112). To a stirred mixture of 16-bromo-5,12,12,26-tetramethyl-7-oxa-4,5,13,20,22,26-hexaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octacosa-1(28),2(6),3, 14,16,18,20,24-octaene-23,27-dione (70 mg, 0.13 mmol, 1 eq), BrettPhos (7 mg, 0.013 mmol, 0.1 eq) and 1-methyl-piperazine (65 mg, 0.65 mmol, 5 eq) in dioxane (5 mL) was added LiHMDS (1.3 mL, 1.3 mmol, 10 eq) dropwise at room temperature under nitrogen atmosphere. The mixture was stirred at 60 °C for 2 h under nitrogen atmosphere and cooled to room temperature. The reaction was heated with saturated NH4Cl(aqueous solution) (10 mL) was quenched, and the mixture was extracted with dichloromethane (3×50 mL). The combined organic layers were washed with brine (3×50 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by silica gel chromatography using dichloromethane/methanol (12/1) to give 24.7 mg of 5,12,12,26-tetramethyl-16-(4-methylpiperazin-1-yl)-7-oxa-4,5,13,20,22,26-hexaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octacosa-1(28),2(6),3, 14,16,18,20,24-octaene-23,27-dione (Example 112) as an off-white solid (33%). LCMS: m/z (ESI), [M+H] +=559.30. 1H NMR (DMSO-d 6, 400 MHz) δ 1.20-1.26 (2H, m), 1.68-1.76 (4H, m), 1.92 (6H, s), 2.28 (3H, s), 2.40-2.60 (4H, m), 3.11 (4H, br s), 3.63 (3H, s), 3.70 (3H, s), 4.08 (2H, t), 6.92 (1H, dd), 7.27 (1H, d), 7.44 (1H, d), 8.27 (1H, d), 8.32 (1H, s), 8.81 (1H, d), 12.71 (1H, s). Example A113 6-Fluoro-15,21-dimethyl-5-(morpholin-4-yl)-23-oxo-2,9,11,15,20,21, 26-heptaazaheptacyclic [26.2.2.1^{1,26}.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]triacontria-3,5,7,9,13,17(34),18(22),19-octaene-12,16-dione Step 1. Preparation of 2-{1-[(5-bromo-4-fluoro-2-nitrophenyl)amino]-3-azabicyclo[3.2.2]nonan-3-yl}ethanol (INT-A113-1). To a stirred mixture of 3-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-3-azabicyclo[3.2.2]nonan-1-amine (M5-9, 700 mg, 2.35 mmol, 1 eq) in DMSO (10 mL) at room temperature were added trimethylamine (712 mg, 7.04 mmol, 3 eq) and 1-bromo-2,5-difluoro-4-nitrobenzene (1.12 g, 4.69 mmol, 2 eq). The resulting mixture was stirred at 100 °C for 2 h under nitrogen atmosphere and cooled to room temperature. The reaction was quenched with water (100 mL), and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue is purified by C18 FLASH using water and acetonitrile as mobile phase. The fractions containing the desired compound are evaporated to dryness to give 800 mg of 2-{1-[(5-bromo-4-fluoro-2-nitrophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethanol (INT-A113-1) (85%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=404.00. 1H NMR (DMSO-d 6, 400 MHz) δ 1.49-1.61 (2H, m), 1.74-1.83 (4H, m), 1.90 (1H, s), 2.35-2.46 (2H, m), 2.55 (2H, t), 2.64 (2H, d), 2.86 (2H, s), 3.58 (2H, q), 4.43 (1H, t), 7.25 (1H, d), 7.99 (1H, s), 8.07 (1H, d). Step 2. Preparation of methyl 5-[5-(2-{1-[(5-bromo-4-fluoro-2-nitrophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylate (INT-A113-2). To a stirred mixture of 2-{1-[(5-bromo-4-fluoro-2-nitrophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethanol (785 mg, 1.95 mmol, 1 eq) and methyl 5-(5-hydroxy-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylate (M2, 667 mg, 2.54 mmol, 1.3 eq) in toluene (20 mL) was added 2-(tributyl-λ5-phosphinidylidene)acetonitrile (2826 mg, 11.71 mmol, 6 eq) at room temperature under nitrogen atmosphere. The resulting mixture was stirred at 100 °C for 3 h. The mixture was cooled to room temperature. The reaction was quenched with water (100 mL) and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC and eluted with petroleum ether/ethyl acetate (1/2) to give 670 mg of 5-[5-(2-{1-[(5-bromo-4-fluoro-2-nitrophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A113-2) (53%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=647.15. Step 3. Preparation of 5-[5-(2-{1-[(2-amino-5-bromo-4-fluorophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A113-3). To a stirred mixture of 5-[5-(2-{1-[(5-bromo-4-fluoro-2-nitrophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (660 mg, 1.02 mmol, 1 eq) and ranyl nickel (175 mg, 2.04 mmol, 2 eq) in methanol (10 mL) was added NH 2NH 2.H 2O (153 mg, 3.06 mmol, 3 equiv.). The resulting mixture was stirred at room temperature for 1 h. The reaction was quenched with water (80 mL), and the mixture was extracted with dichloromethane (3×80 mL). The combined organic layers were washed with brine (3×80 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC and eluted with petroleum ether/ethyl acetate (1/32) to give 490 mg of 5-[5-(2-{1-[(2-amino-5-bromo-4-fluorophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A113-3) (78%) as a brown solid. LCMS: m/z (ESI), [M+H] += 617.10. Step 4. Preparation of 5-(5-{2-[1-(2-amino-6-bromo-5-fluoro-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.2]non-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A113-4). To a stirred mixture of 5-[5-(2-{1-[(2-amino-5-bromo-4-fluorophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (480 mg, 0.78 mmol, 1 eq) in ethanol (5 mL) at room temperature was added BrCN (165 mg, 1.55 mmol, 2 eq). The resulting mixture was stirred at 60 °C for 1 h and cooled to room temperature. The reaction was quenched with saturated sodium thiosulfate (aq.) (60 mL) and the mixture was extracted with dichloromethane (3×50 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC and eluted with dichloromethane/methanol (20/1) to give 250 mg of 5-(5-{2-[1-(2-amino-6-bromo-5-fluoro-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.2]non-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A113-4) (50%) as a reddish brown solid. LCMS: m/z (ESI), [M+H] +=642.15. 1H NMR (DMSO-d 6, 400 MHz) δ 1.04-1.30 (2H, m), 1.81 (1H, s), 1.80-1.98 (5H, m), 2.69 (2H, s), 2.81-2.94 (4H, m), 3.56 (3H, s), 3.65-3.77 (6H, m), 4.12 (2H, t), 6.11 (2H, s), 7.05 (1H, d), 7.48 (1H, d), 7.97 (1H, s), 8.10 (1H, d), 8.38 (1H, d). Step 5. Preparation of 5-(5-{2-[1-(2-amino-6-bromo-5-fluoro-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.2]non-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A113-5). To a stirred solution of 5-(5-{2-[1-(2-amino-6-bromo-5-fluoro-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.2]non-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (220 mg, 0.34 mmol, 1 eq.) in tetrahydrofuran (4 mL) at room temperature was added LiOH (21 mg, 0.86 mmol, 2.5 eq.) in H 2O (1 mL). The resulting mixture was stirred at 60 ° C for 1 h and cooled to room temperature. The mixture was concentrated under reduced pressure. The residue was purified by C18 FLASH chromatography using water and acetonitrile as mobile phases. The fractions containing the desired compound were evaporated to dryness to give 190 mg of 5-(5-{2-[1-(2-amino-6-bromo-5-fluoro-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.2]non-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A113-5) (88%) as a white solid. LCMS: m/z (ESI), [M+H] +=630.10. Step 6. Preparation of 5-bromo-6-fluoro-15,21-dimethyl-23-oxa-2,9,11,15,20, 21,26-heptaazaheptacyclo[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]triacontriacontria-3,5,7,9,13,17(34),18(22),19-octaene-12,16-dione (INT-A113-6). To a stirred solution of 5-(5-{2-[1-(2-amino-6-bromo-5-fluoro-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.2]non-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (190 mg, 0.30 mmol, 1 equiv.) in 1,4-dioxane (5 mL) was added at room temperature.N,N-Diisopropylethylamine (117 mg, 0.91 mmol, 3 equiv.) and N,N,N,N-Tetramethyl- O-(7-Azabenzotriazole-1-yl)uronium hexafluorophosphate (230 mg, 0.60 mmol, 2 equiv). The resulting mixture was stirred at 60 °C for 1 h under a nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (30 mL), and the mixture was extracted with dichloromethane (3×30 mL). The combined organic layers were washed with brine (3×30 mL), purified by anhydrous Na 2SO 4The residue was purified by Prep-TLC and eluted with dichloromethane/methanol (22/1) to give 150 mg of 5-bromo-6-fluoro-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tetracontriacontria-3,5,7,9,13,17(34), 18(22),19-octaene-12,16-dione (INT-A113-6) (81%) as a light yellow solid. LCMS: m/z (ESI), [M+H] +=610.30. Step 7. Preparation of 6-fluoro-15,21-dimethyl-5-(morpholin-4-yl)-23-oxo-2,9, 11,15,20,21,26-heptaazaheptacyclo[26.2.2.1^{1,26}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]triacontria-3,5,7,9,13,17(34), 18(22),19-octaene-12,16-dione (Example A113). 5-Bromo-6-fluoro-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic [26.2.2.1^{1,26}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]triacontria-3,5,7,9,13,17(34), 18(22),19-octaene-12,16-dione (50 mg, 0.08 mmol, 1 equivalent), BrettPhos Pd G 3To a stirred mixture of 1,4-dioxane (4 mL) and 2-nitropropene (23 mg, 0.03 mmol, 0.3 equiv) and morpholine (22 mg, 0.25 mmol, 3 equiv) was added LiHMDS (0.82 mL, 0.82 mmol, 10 equiv). The resulting mixture was stirred at 60 °C for 30 min under nitrogen atmosphere and cooled to room temperature. The reaction was heated with saturated NH4Cl(aqueous solution) (80 mL) was quenched, and the mixture was extracted with dichloromethane (3×80 mL). The combined organic layers were washed with brine (3×50 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC and eluted with dichloromethane/methanol (25/1). The product was purified by prep-HPLC using an XBridge Shield RP18 OBD column with 10 mmol/L NH4HCO 3The mixture was further purified by using water and acetonitrile as mobile phase. The fractions containing the desired compound were evaporated to dryness to give 1.2 mg of 6-fluoro-15,21-dimethyl-5-(morpholin-4-yl)-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontria-3,5,7,9,13,17(34),18(22),19-octaene-12,16-dione (Example A113) as a white solid (2%). LCMS: m/z (ESI), [M+H] +=617.35. 1H NMR (CDCl 3, 400 MHz) δ 2.00-2.20 (9H, m), 2.84 (2H, d), 2.95 (2H, t), 3.06 (4H, t), 3.40-3.50 (2H, m), 3.71 (3H, s), 3.80 (3H, s), 3.91 (4H, t), 4.23 (2H, t), 7.01 (1H, d), 7.39 (1H, d), 8.23 (1H, d), 8.50 (1H, s), 9.19 (1H, d), 13.50 (1H, s). Example A115 6-Fluoro-15,21-dimethyl-5-(4-methylpiperazin-1-yl)-23-oxo-2,9, 11,15,20,21,26-heptaazaheptacyclic [26.2.2.1^{1,26}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]triacontria-3,5,7,9,13,17(34), 18(22),19-octaene-12,16-dione 5-Bromo-6-fluoro-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic [26.2.2.1^{1,26}. 1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontria-3,5,7,9,13, 17(34),18(22),19-octaene-12,16-dione (INT-A113-6, 50 mg, 0.08 mmol, 1 equivalent), BrettPhos Pd G 3To a stirred mixture of 1,4-dioxane (4 mL) and 1-methylpiperazine (23 mg, 0.03 mmol, 0.3 equiv) was slowly added LiHMDS (0.82 mL, 0.82 mmol, 10 equiv). The resulting mixture was stirred at 60 °C for 30 min and then cooled to room temperature. The reaction was heated with saturated NH4Cl(aqueous solution) (80 mL) was quenched, and the mixture was extracted with dichloromethane (3×80 mL). The combined organic layers were washed with brine (3×50 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by prep-TLC and eluted with dichloromethane/methanol (12/1). The product was purified by prep-HPLC using an XBridge Prep OBD C18 column with 10 mmol/L NH4HCO 3and 0.1% NH 3.H 2The mixture was further purified by using water and acetonitrile as mobile phase. The fractions containing the desired compound were evaporated to dryness to give 0.8 mg of 6-fluoro-15,21-dimethyl-5-(4-methylpiperazin-1-yl)-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontria-3,5,7,9,13,17(34), 18(22),19-octaene-12,16-dione (Example A115) as a white solid (2%). LCMS: m/z (ESI), [M+H] +=630.40. 1H NMR (CD 3OD, 400 MHz) δ 2.02-2.07 (4H, m), 2.08-2.19 (4H, m), 2.40 (3H, s), 2.62-2.78 (4H, m), 2.85-2.97 (4H, m), 3.03-3.13 (3H, m), 3.43-3.55 (4H, m), 3.69 (3H, s), 3.79 (3H, s), 4.31 (2H, t), 7.23 (1H, s), 7.51 (1H, d), 8.33 (1H, s), 8.34 (1H, s), 9.27 (1H, s). Example A116 6-Fluoro-15,21-dimethyl-23-oxo-2,9,11,15,20,21,26-heptaazaheptacyclic [26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontria-3,5,7,9,13,17(34),18(22),19-octaene-12,16-dione 5-Bromo-6-fluoro-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic [26.2.2.1^{1,26}. 1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontria-3,5,7,9,13, 17(34),18(22),19-octaene-12,16-dione (INT-A113-6, 20 mg, 0.03 mmol, 1 equivalent) and BrettPhos Pd G 3To a stirred mixture of (9 mg, 0.01 mmol, 0.3 equiv) and 1,4-dioxane (2 mL) was slowly added LiHMDS (0.33 mL, 0.33 mmol, 10 equiv). The resulting mixture was stirred at 60 °C for 30 min and cooled to room temperature. The reaction was heated with saturated NH4Cl(aqueous solution) (50 mL) was quenched. The mixture was extracted with dichloromethane (3×50 mL). The combined organic layers were washed with brine (3×20 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by prep-TLC and eluted with dichloromethane/methanol (22/1). The product was purified by prep-HPLC using an XBridge Shield RP18 OBD column with 10 mmol/L NH4HCO 3The mixture was further purified by using water and acetonitrile as mobile phase. The fractions containing the desired compound were evaporated to dryness to give 0.9 mg of 6-fluoro-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18, 22}]triacontria-3,5,7,9,13,17(34),18(22),19-octaene-12,16-dione (Example A116) as a white solid (5%). LCMS: m/z (ESI), [M+H] +=532.15. 1H NMR (DMSO-d 6, 400 MHz) δ 1.88-1.94 (4H, m), 1.96-2.13 (3H, m), 2.79 (2H, d), 2.86 (2H, t), 3.39-3.42 (4H, m), 3.62 (3H, s), 3.74 (3H, s), 4.28 (2H, t), 6.91-7.01 (1H, m), 7.45 (1H, dd), 7.87 (1H, dd), 8.27-8.35 (2H, m), 9.10 (1H, d), 13.34 (1H, s). Example A117A, Example A117B, Example A118A and Example A118B 6-Fluoro-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontahedral-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione, isomer 1 (Example A117A) and isomer 2 (Example A117B); 6-Fluoro-15,21-dimethyl-5-(morpholin-4-yl)-23-oxa-2,9,11,15,20,21, 26-Heptazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]tris-3,5,7,9,13,17(33), 18(22),19-octaene-12,16-dione, isomer 1 (Example A118A) and isomer 2 (Example A118B). Step 1. Preparation of 3-(2-((tert-butyldimethylsilyl)oxy)ethyl)-3-azabicyclo[3.2.1]octan-1-amine (INT-A117-1).NA mixture of tert-butyl-(3-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-3-azabicyclo[3.2.1]octan-1-yl)carbamate (M4-9, 10 g, 26 mmol, 1 equivalent) in trifluoroacetic acid (35 mL) and dichloromethane (100 mL) was stirred at room temperature for 2 h. The resulting mixture was concentrated under reduced pressure. The residue was treated with saturated NaHCO3(aqueous solution) (100 mL) and extracted with dichloromethane (3×100 mL). The combined organic layers were washed with brine (3×200 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure to give 7.3 g of 3-(2-((tert-butyldimethylsilyl)oxy)ethyl)-3-azabicyclo[3.2.1]octan-1-amine (INT-A117-1) (98%) as a yellow solid. Use the crude product without purification. LCMS: m/z (ESI), [M+H] +=285.15. Step 2. Preparation of 2-{1-[(5-bromo-4-fluoro-2-nitrophenyl)amino]-3-azabicyclo[3.2.1]octan-3-yl}ethanol (INT-A117-2). A mixture of 3-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-3-azabicyclo[3.2.1]octan-1-amine (2 g, 7.03 mmol, 1 eq.), triethylamine (2.84 g, 28.11 mmol, 4 eq.) and 1-bromo-2,5-difluoro-4-nitrobenzene (5.01 g, 21.08 mmol, 3 eq.) in DMSO (30 mL) was stirred at 100 °C for 3 h. The mixture was cooled to room temperature. The reaction was quenched with water (50 mL), and the mixture was extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column and eluted with ethyl acetate/petroleum ether (1/1) to give 1.5 g of 2-{1-[(5-bromo-4-fluoro-2-nitrophenyl)amino]-3-azabicyclo[3.2.1]octan-3-yl}ethanol (INT-A117-2) (55%) as an orange oil. LCMS: m/z (ESI), [M+H] +=387.95. Step 3. Preparation of 5-[5-(2-{1-[(5-bromo-4-fluoro-2-nitrophenyl)amino]-3-azabicyclo[3.2.1]oct-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A117-3). A mixture of 2-{1-[(5-bromo-4-fluoro-2-nitrophenyl)amino]-3-azabicyclo[3.2.1]octan-3-yl}ethanol (700 mg, 1.80 mmol, 1 eq.), 5-(5-hydroxy-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (M2, 712 mg, 2.70 mmol, 1.5 eq.) and 2-(tributylphosphino)acetonitrile (2.61 g, 10.82 mmol, 6 eq.) in toluene (20 mL) was stirred at 100 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature and concentrated under reduced pressure. The residue was treated with water (50 mL), and the mixture was washed with CH 2Cl 2(3×50 mL) extraction. The combined organic layers were washed with brine (3×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was subjected to prep-TLC using CH 2Cl 2/methanol (25/1) for purification to obtain 700 mg of 5-[5-(2-{1-[(5-bromo-4-fluoro-2-nitrophenyl)amino]-3-azabicyclo[3.2.1]oct-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A117-3) (61%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=633.35. 1H NMR (DMSO-d 6, 400 MHz) δ 1.53-1.74 (4H, m), 1.75-1.87 (1H, m), 1.93 (1H, s), 2.16 (1H, d), 2.21 (2H, d), 2.70 (1H, d), 2.76-2.86 (2H, m), 3.16 (1H, d), 3.57 (3H, s), 3.76 (6H, d), 4.09 (2H, t), 7.26 (1H, d), 7.97 (1H, s), 8.04-8.11 (2H, m), 8.17 (1H, s), 8.41 (1H, d). Step 4. Preparation of methyl 5-[5-(2-{1-[(2-amino-5-bromo-4-fluorophenyl)amino]-3-azabicyclo[3.2.1]oct-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylate (INT-A117-4). To a stirred mixture of methyl 5-[5-(2-{1-[(5-bromo-4-fluoro-2-nitrophenyl)amino]-3-azabicyclo[3.2.1]octan-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylate (680 mg, 1.07 mmol, 1 eq) and ranyl nickel (184 mg, 2.15 mmol, 2 eq) in methanol (10 mL) was added hydrazine hydrate (107 mg, 2.15 mmol, 2 eq) at room temperature. The resulting mixture was stirred at room temperature for 1 h and filtered. The filter cake was washed with methanol (3×10 mL). The filtrate was concentrated under reduced pressure to obtain 600 mg of 5-[5-(2-{1-[(2-amino-5-bromo-4-fluorophenyl)amino]-3-azabicyclo[3.2.1]oct-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A117-4) (99%) as a white solid. The crude product was used in the next reaction without purification. LCMS: m/z (ESI), [M+H] += 603.10. Step 5. Preparation of 5-(5-{2-[1-(2-amino-6-bromo-5-fluoro-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A117-5). A mixture of 5-[5-(2-{1-[(2-amino-5-bromo-4-fluorophenyl)amino]-3-azabicyclo[3.2.1]octan-3-yl}ethoxy)-1-methylpyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (550 mg, 0.91 mmol, 1 eq) and BrCN (193 mg, 1.82 mmol, 2 eq) in ethanol (10 mL) was stirred at room temperature overnight under nitrogen atmosphere. The reaction mixture was concentrated under vacuum. The residue was purified by filtration with N, N-dimethylformamide (10 mL) for purification to obtain 280 mg of 5-(5-{2-[1-(2-amino-6-bromo-5-fluoro-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A117-5) (48%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=628.10. Step 6. Preparation of 5-(5-{2-[1-(2-amino-6-bromo-5-fluoro-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (isomer 1, INT-A117A-6 and isomer 2, INT-A117B-6). A racemic mixture of 5-(5-{2-[1-(2-amino-6-bromo-5-fluoro-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (200 mg) was purified by prep-chiral-HPLC using a CHIRALPAK IG column (2×25 cm, 5 μm) with a mobile phase of hexane (containing 0.1% diethylamine) and ethanol/CH2Cl 2(1/1) of mobile phase B to obtain 5-(5-{2-[1-(2-amino-6-bromo-5-fluoro-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]octan-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester, 90 mg of isomer 1 (INT-A117A-6, 45%) and 90 mg of isomer 2 (INT-A117B-6, 45%) as an off-white solid. INT-A117A-6: chiral-HPLC, Rt=3.619 minutes. INT-A117B-6: chiral-HPLC, Rt=4.508 minutes. Step 7. Preparation of 5-(5-{2-[1-(2-amino-6-bromo-5-fluoro-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A117A-7). 5-(5-{2-[1-(2-amino-6-bromo-5-fluoro-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A117A-6, 80 mg, 0.13 mmol, 1 eq.) and LiOH (6 mg, 0.25 mmol, 2 eq.) were dissolved in THF (2 mL) and H 2O (0.5 mL) was stirred at room temperature under nitrogen atmosphere for 2 h. The mixture was concentrated under reduced pressure to give 65 mg of 5-(5-{2-[1-(2-amino-6-bromo-5-fluoro-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A117A-7) (83%) as a yellow solid. This material was used in the next step without further purification. LCMS: m/z (ESI), [M+H] +=614.20. Step 8. Preparation of 5-bromo-6-fluoro-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]tris-3,5,7,9,13,17(33), 18(22),19-octaene-12,16-dione (INT-A117A-8). 5-(5-{2-[1-(2-amino-6-bromo-5-fluoro-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A117A-7, 60 mg, 0.10 mmol, 1 equivalent), N, N, N, N-Tetramethyl- O-(7-Azabenzotriazol-1-yl)urea hexafluorophosphate (74 mg, 0.20 mmol, 2 equivalents) and N,N- A mixture of diisopropylethylamine (38 mg, 0.30 mmol, 3 equiv.) in 1,4-dioxane (6 mL) was stirred at 60 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (20 mL), and the mixture was washed with CH2Cl 2(3×30 mL) extraction. The combined organic layers were purified by anhydrous Na 2SO 4The residue was purified by prep-TLC and eluted with dichloromethane/methanol (12/1) to give 50 mg of 5-bromo-6-fluoro-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontria-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione (INT-A117A-8) (85%) as a light yellow solid. LCMS: m/z (ESI), [M+H] +=594.05. Step 9. 6-Fluoro-15,21-dimethyl-23-oxo-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tris-3,5,7,9,13,17(33), 18(22),19-octaene-12,16-dione (isomer 1, Example A117A) and 6-fluoro-15,21-dimethyl-5-(morpholin-4-yl)-23-oxo-2,9,11,15,20,21, Preparation of 26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]tris-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione (isomer 1, Example A118A). 5-Bromo-6-fluoro-15,21-dimethyl-23-oxa-2,9,11,15,20,21, 26-heptaazaheptacyclic [24.4.1.1^{1,28}.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]tris-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione (INT-A117A-8, 50 mg, 0.08 mmol, 1 equivalent), morpholine (146 mg, 1.68 mmol, 20 equivalents), BrettPhos Pd G 3A mixture of (22 mg, 0.03 mmol, 0.3 equiv) and t-BuOK (113 mg, 1.01 mmol, 12 equiv) in 1,4-dioxane (4 mL) was stirred at 80 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (20 mL), and the mixture was washed with CH2Cl 2(3×30 mL) extraction. The combined organic layers were purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by prep-TLC and eluted with dichloromethane/methanol (12/1). The product was purified by Prep-HPLC using an XBridge Shield RP18 OBD column with 0.1% NH4HCO 3The mixture was further purified by using water and acetonitrile as the mobile phase. The fractions containing the desired compound were evaporated to dryness to give 5.5 mg of 6-fluoro-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontria-3,5,7,9,13,17(33), 18(22),19-octaene-12,16-dione (isomer 1, Example A117A, 8%) and 8.1 mg of 6-Fluoro-15,21-dimethyl-5-(morpholin-4-yl)-23-oxo-2,9,11,15,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tris-3,5,7,9,13,17(33), 18(22),19-octaene-12,16-dione (isomer 1, Example A118A, 11%). Example 117A: LCMS: m/z (ESI), [M+H] +=518.25. 1H NMR (DMSO-d 6, 400 MHz) δ 1.52-1.83 (3H, m), 2.02 (1H, d), 2.38-2.43 (2H, m), 2.51-2.65 (2H, m), 2.72-2.78 (1H, m), 3.21-3.27 (2H, m), 3.37-3.46 (1H, m), 3.63 (3H, s), 3.64-3.70 (1H, m), 3.72 (3H, s), 4.57 (1H, t), 4.71 (1H, d), 6.98-7.03 (1H, m), 7.37 (1H, dd), 7.72 (1H, dd), 8.29 (1H, d), 8.41 (1H, d), 8.83 (1H, s), 12.82 (1H, s). Example 118A: LCMS: m/z (ESI), [M+H] +=603.20. 1H NMR (DMSO-d 6, 400 MHz) δ 1.48-1.82 (3H, m), 2.02 (1H, d), 2.38-2.49 (2H, m), 2.51-2.56 (2H, m), 2.62-2.77 (2H, m), 2.85-3.07 (4H, m), 3.21-3.27 (1H, m), 3.37-3.46 (1H, m), 3.62 (3H, s), 3.63-3.70 (1H, m), 3.72 (3H, s), 3.77 (4H, t), 4.57 (1H, t), 4.71 (1H, d), 7.21-7.43 (2H, m), 8.28 (1H, d), 8.41 (1H, d), 8.81 (1H, s), 12.71 (1H, s). Example A117B, 6-fluoro-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tris-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione (isomer 2) was prepared from INT-A117B-6 as a light yellow solid using the same reaction sequence as described in the synthesis of Example A117A. LCMS: m/z (ESI), [M+H] +=518.30. 1H NMR (DMSO-d 6, 400 MHz) δ 1.56-1.82 (3H, m), 2.02 (1H, d), 2.38-2.45 (2H, m), 2.51-2.55 (1H, m), 2.62-2.78 (2H, m), 3.15-3.25 (2H, m), 3.37-3.46 (1H, m), 3.63 (3H, s), 3.64-3.70 (1H, m), 3.72 (3H, s), 4.53 (1H, t), 4.65 (1H, d), 6.98-7.04 (1H, m), 7.37 (1H, dd), 7.72 (1H, dd), 8.29 (1H, d), 8.41 (1H, d), 8.83 (1H, s), 12.82 (1H, s). Example A118B, 6-fluoro-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tris-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione (isomer 2) was prepared from INT-A117B-6 as a light yellow solid using the same reaction sequence as described in the synthesis of Example A118A. LCMS: m/z (ESI), [M+H] +=603.30. 1H NMR (DMSO-d 6, 400 MHz) δ 1.52-1.82 (3H, m), 2.03 (1H, d), 2.38-2.45 (2H, m), 2.51-2.62 (3H, m), 2.65-3.12 (6H, m), 3.21-3.27 (1H, m), 3.37-3.46 (1H, m), 3.62 (3H, s), 3.63-3.70 (1H, m), 3.72 (3H, s), 3.77 (4H, t), 4.55 (1H, t), 4.67 (1H, d), 7.29 (1H, d), 7.37 (1H, d), 8.27 (1H, s), 8.41 (1H, s), 8.81 (1H, s), 12.71 (1H, s). Example A119 5-[4-(Dimethylamino)piperidin-1-yl]-15,21-dimethyl-23-oxo-2,9,11,15,20,21,26-heptaazaheptacyclic[26.2.2.1^{1,26}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]triacontria-3,5,7,9,13,17(34), 18(22),19-octaene-12,16-dione Under nitrogen atmosphere at room temperature, 5-bromo-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic [26.2.2.1^{1,26}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]triacontria-3,5,7,9,13,17(34), 18(22),19-octaene-12,16-dione (M5, 100 mg, 0.16 mmol, 1 equivalent) was added.N,N-dimethylpiperidin-4-amine (64.92 mg, 0.50 mmol, 3 equiv) and BrettPhos Pd G 3To a stirred mixture of 4-(45.90 mg, 0.05 mmol, 0.3 eq.) and 1,4-dioxane (10 mL) was added LiHMDS (1 mL, 1.014 mmol, 6 eq.) dropwise. The mixture was stirred at 60 °C for 1 h and then cooled to room temperature. The reaction was heated with saturated NH4Cl(aqueous solution) (15 mL) was quenched, and the mixture was extracted with dichloromethane (3×20 mL). The combined organic layers were washed with brine (3×30 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was subjected to prep-HPLC using an XBridge Shield RP18 OBD column with 10 mmol/L NH4HCO 3and 0.1% NH 3.H 2O water and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 50.1 mg of 5-[4-(dimethylamino)piperidin-1-yl]-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontria-3,5,7,9,13,17(34), 18(22),19-octaene-12,16-dione (Example A119) (46%) as a light yellow solid. LCMS: m/z (ESI), [M+H] +=640.40. 1H NMR (DMSO-d 6, 400 MHz) δ 1.47-1.59 (2H, m), 1.81-1.99 (6H, m), 2.00-2.13 (3H, m), 2.22 (6H, s), 2.57-2.70 (2H, m), 2.75-2.82 (2H, m), 2.86 (2H, t), 3.38-3.50 (4H, m), 3.50-3.59 (3H, m), 3.62 (3H, s), 3.74 (3H, s), 4.27 (2H, t), 6.91 (1H, d), 7.34 (1H, s), 7.45 (1H, d), 8.30 (2H, d), 9.11 (1H, d), 13.18 (1H, s). Examples A120-A128 (Table 2) were prepared using the same reaction sequence described above for the synthesis of Example A119 from M5. Example A129B 21-Methyl-5-(morpholin-4-yl)-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18, 22}]tris-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione, isomer 2 (Example A129B) Step 1. Preparation of 1-benzyl-2-methylpyrazol-3-one (INT-A129-1). 2-Methyl-1H-pyrazol-3-one (10 g, 101.93 mmol, 1 equivalent), K 2CO 3A mixture of (42.26 g, 305.79 mmol, 3 equiv) and BnBr (18.31 g, 107.02 mmol, 1.05 equiv) in acetonitrile (100 mL) was stirred at room temperature under nitrogen atmosphere for 2 h. The reaction was quenched with water (300 mL), and the mixture was extracted with ethyl acetate (3×200 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue is purified by C18 FLASH using water and acetonitrile as mobile phase. The fractions containing the desired compound are evaporated to dryness to give 5.5 g of 1-benzyl-2-methylpyrazol-3-one (INT-A129-1) (28%) as a brown solid. LCMS: m/z (ESI), [M+H] +=189.10. 1H NMR (DMSO-d 6 , 400 MHz) δ 3.14 (3H, s), 4.97 (2H, s), 5.23 (1H, d), 7.14-7.22 (2H, m), 7.26-7.38 (3H, m), 7.92 (1H, d). Step 2. Preparation of 1-benzyl-4-iodo-2-methylpyrazol-3-one (INT-A129-2). To a stirred mixture of 1-benzyl-2-methylpyrazol-3-one (6.35 g, 33.73 mmol, 1 eq.) in acetonitrile (200 mL) was added NIS (8.35 g, 37.10 mmol, 1.1 eq.) in portions and stirred at 0 °C for 2 h under nitrogen atmosphere. The reaction was quenched with saturated sodium thiosulfate solution (200 mL), and the mixture was extracted with ethyl acetate (3×300 mL). The combined organic layers were washed with brine (3×300 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue is purified by C18 FLASH using water and acetonitrile as mobile phase. The fractions containing the desired compound are evaporated to dryness to give 9.5 g of 1-benzyl-4-iodo-2-methylpyrazol-3-one (INT-A129-2) (89%) as a reddish-brown semisolid. LCMS: m/z (ESI), [M+H] +=315.05. 1H NMR (DMSO-d 6 , 400 MHz) δ 3.22 (3H, s), 5.00 (2H, s), 7.16-7.41 (5H, m), 8.18 (1H, s). Step 3. Preparation of 5-bromo-6-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyridine-3-carboxylic acid methyl ester (INT-A129-3). Under nitrogen atmosphere at 0 °C, 5-bromo-6-hydroxypyridine-3-carboxylic acid methyl ester (10 g, 43.09 mmol, 1 equivalent) and K 2CO 3To a stirred mixture of 2-(trimethylsilyl)ethoxymethyl chloride (10.78 g, 64.64 mmol, 1.5 equiv.) (11.91 g, 86.194 mmol, 2 equiv.) in acetonitrile (200 mL) was added dropwise 2-(trimethylsilyl)ethoxymethyl chloride (10.78 g, 64.64 mmol, 1.5 equiv.). The resulting mixture was stirred at room temperature for 3 h. The reaction was quenched with water (200 mL), and the mixture was extracted with ethyl acetate (3×200 mL). The combined organic layers were washed with brine (3×200 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue is recrystallized from petroleum ether/ethyl acetate (20/1) to give 8 g of methyl 5-bromo-6-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyridine-3-carboxylate (INT-A129-3) (51%) as a white solid. LCMS: m/z (ESI), [M+H] +=364.10. 1H NMR (DMSO-d 6 , 400 MHz) δ 0.00 (9H, s), 0.89-0.94 (2H, m), 3.60-3.68 (2H, m), 3.84 (3H, s), 5.44 (2H, s), 8.24 (1H, d), 8.58 (1H, d). Step 4. Preparation of methyl 6-oxo-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1,6-dihydropyridine-3-carboxylate (INT-A129-4). 5-Bromo-6-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyridine-3-carboxylic acid methyl ester (6.89 g, 19.01 mmol, 1 equivalent), KOAc (5.60 g, 57.054 mmol, 3 equivalents), Pd(dppf)Cl 2CH 2Cl 2A mixture of (1.55 g, 1.90 mmol, 0.1 eq.) and bis(pinacol)diboron (7.24 g, 28.52 mmol, 1.5 eq.) in 1,4-dioxane (100 mL) was stirred at 80 °C for 2 h under nitrogen atmosphere and then cooled to room temperature. The mixture was used directly in the next step without further work-up. LCMS: m/z (ESI), [M+H] +=328.05. Step 5. Preparation of 5-(1-benzyl-2-methyl-3-oxopyrazol-4-yl)-6-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyridine-3-carboxylic acid methyl ester (INT-A129-5). Freshly prepared 6-oxo-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolane-2-yl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1,6-dihydropyridine-3-carboxylic acid methyl ester (INT-A129-4), K 2CO 3(5.28 g, 38.20 mmol, 3 equivalents), Pd(dppf)Cl 2CH 2Cl 2A mixture of (1.04 g, 1.27 mmol, 0.1 equiv) and 1-benzyl-4-iodo-2-methylpyrazol-3-one (4 g, 12.73 mmol, 1.00 equiv) in dioxane (100 mL) and water (25 mL) was stirred at 80 °C for 2 h under nitrogen atmosphere. The reaction mixture was cooled to room temperature, treated with water (100 mL), and extracted with dichloromethane (3×100 mL). The combined organic layers were washed with brine (3×300 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by reverse C18 FLASH chromatography using acetonitrile and water (0% to 100% gradient) as mobile phase to give 2.5 g of 5-(1-benzyl-2-methyl-3-oxopyrazol-4-yl)-6-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyridine-3-carboxylic acid methyl ester (INT-A129-5) (41%) as a brown solid. LCMS: m/z (ESI), [M+H] +=470.30. Step 6. Preparation of methyl 5-(5-hydroxy-1-methylpyrazol-4-yl)-6-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyridine-3-carboxylate (INT-A129-6). A mixture of methyl 5-(1-benzyl-2-methyl-3-oxopyrazol-4-yl)-6-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyridine-3-carboxylate (2.5 g, 5.32 mmol, 1.00 equiv) and Pd/C (2.83 g, 26.62 mmol, 5 equiv) in methanol (50 mL) was stirred at room temperature overnight under a hydrogen atmosphere. The resulting mixture was filtered. The filter cake was washed with methanol (3×100 mL). The filtrate was concentrated under reduced pressure to obtain 1.5 g of methyl 5-(5-hydroxy-1-methylpyrazol-4-yl)-6-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyridine-3-carboxylate (INT-A129-6) (74%) as a brown solid. LCMS: m/z (ESI), [M+H] +=380.00. Step 7. Preparation of 2-{1-[(5-fluoro-2-nitrophenyl)amino]-3-azabicyclo[3.2.1]octan-3-yl}ethanol (INT-A129-7). A mixture of 2-{1-amino-3-azabicyclo[3.2.1]octan-3-yl}ethanol dihydrochloride (4.8 g, 19.75 mmol, 1 eq.), trimethylamine (9.99 g, 98.77 mmol, 5 eq.) and 2,4-difluoro-1-nitrobenzene (4.71 g, 29.63 mmol, 1.5 eq.) in acetonitrile (100 mL) was stirred at 60 °C for 2 h under nitrogen atmosphere and cooled to room temperature. The reaction was quenched with water (300 mL), and the mixture was extracted with ethyl acetate (3×200 mL). The combined organic layers were washed with brine (3×300 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column using petroleum ether/ethyl acetate (2/1) to give 5.5 g of 2-{1-[(5-fluoro-2-nitrophenyl)amino]-3-azabicyclo[3.2.1]octan-3-yl}ethanol (INT-A129-7) (90 %) as an orange oil. LCMS: m/z (ESI), [M+H] +=310.10. 1H NMR (DMSO-d 6 , 400 MHz) δ 1.58-1.73 (3H, m), 1.76-1.98 (2H, m), 2.08-2.25 (4H, m), 2.36-2.51 (2H, m), 2.68 (1H, dd), 3.12-3.20 (1H, m), 3.44-3.58 (2H, m), 4.35 (1H, t), 6.53-6.62 (1H, m), 6.83 (1H, dd), 8.19 (1H, dd), 8.43-8.48 (1H, m). Step 8. Preparation of 2-{1-[(5-fluoro-2-nitrophenyl)amino]-3-azabicyclo[3.2.1]oct-3-yl}ethanol (isomer 2, INT-A129-7B). A racemic mixture of 2-{1-[(5-fluoro-2-nitrophenyl)amino]-3-azabicyclo[3.2.1]oct-3-yl}ethanol (15 g) was subjected to Prep_SFC (CHIRALPAK IG, 3*25 cm, 5 μm) with CO 2The mixture was separated by eluting with a mobile phase A of 1% and a mobile phase B of methanol to obtain 6.2 g (41%) of isomer 1 (INT-A129-7A) as an orange oil and 6.5 g of isomer 2 (INT-A129-7B) (43%) as a light yellow oil. Isomer 1: LCMS: m/z (ESI), [M+H] +=310.20. Rt= 1.429 min. Isomer 2: LCMS: m/z (ESI), [M+H] +=310.20. 1H NMR (DMSO-d 6 , 400 MHz) δ 1.59-1.73 (3H, m), 1.82 (1H, d), 1.94 (1H, s), 2.17 (2H, dd), 2.44 (2H, d), 2.68 (1H, d), 3.17 (2H, d), 3.51 (2H, s), 4.11 (1H, q), 4.35 (1H, d), 6.58 (1H, t), 6.83 (1H, d), 8.19 (1H, dd), 8.45 ( 1H, s). Rt=1.987 minutes. Step 9. 2-(1-{[5-(morpholin-4-yl)-2-nitrophenyl]amino}-3-azabicyclo[3.2.1]octan-3-yl)ethanol (INT -A129-9B). 2-{1-[(5-fluoro-2-nitrophenyl)amino]-3-azabicyclo[3.2.1]octan-3-yl}ethanol (INT- A129-7B, 6.5 g, 21.01 mmol, 1 eq.), morpholine (6.4 g, 73.54 mmol, 3.5 eq.) and trimethylamine (6.38 g, 63.03 mmol, 3 eq.) in dimethyl sulfoxide (100 mL) The mixture was stirred at 120 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (800 mL), and the mixture was extracted with ethyl acetate (3×500 mL). The combined organic layers were washed with brine (3×500 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure to obtain 7.4 g of 2-(1-{[5-(morpholin-4-yl)-2-nitrophenyl]amino}-3-azabicyclo[3.2.1]octan-3-yl)ethanol (INT-A129-9B) (93%) as an orange solid. LCMS: m/z (ESI), [M+H] +=377.25. 1H NMR (DMSO-d 6 , 400 MHz) δ 1.63-1.71 (3H, m), 1.83-1.87 (1H, m), 1.92-1.96 (1H, m), 2.05 (1H, d), 2.14-2.22 (2H, m), 2.32-2.51 (3H, m), 2.51-2.57 (1H, m), 2.65 (1H, dd), 3.31-3.37 (4H, m), 3.45-3.56 (2H, m), 3.73 (4H, t), 4.35 (1H, t), 6.09 (1H, d), 6.46 (1H, dd), 7.93 (1H, d), 8.67 (1H, s). Step 10. Preparation of methyl 5-(1-methyl-5-{2-[1-{[5-(oxolin-4-yl)-2-nitrophenyl]amino}-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}pyrazol-4-yl)-6-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyridine-3-carboxylate (INT-A129-10B). To a stirred mixture of methyl 5-(5-hydroxy-1-methylpyrazol-4-yl)-6-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyridine-3-carboxylate (INT-A129-6, 756 mg, 1.99 mmol, 1.5 equiv), 2-[1-{[5-(morpholin-4-yl)-2-nitrophenyl]amino}-3-azabicyclo[3.2.1]octan-3-yl]ethanol (INT-A129-9B, 500 mg, 1.32 mmol, 1.00 equiv) and triphenylphosphine (1045 mg, 3.98 mmol, 3 equiv) in tetrahydrofuran (10 mL) was added diisopropyl azodicarboxylate (805 mg, 3.98 mmol, 3 equiv) dropwise at 0 °C under nitrogen atmosphere. The resulting mixture was stirred at 0 °C for 2 h under nitrogen atmosphere. The reaction was quenched with water (100 mL), and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on Prep-TLC using petroleum ether/ethyl acetate (1/3) to give 400 mg of 5-(1-methyl-5-{2-[1-{[5-(oxolin-4-yl)-2-nitrophenyl]amino}-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}pyrazol-4-yl)-6-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyridine-3-carboxylic acid methyl ester (INT-A129-10B) (40%) as an orange solid. LCMS: m/z (ESI), [M+H] += 738.60. Step 11. Preparation of 5-(5-{2-[1-{[2-amino-5-(oxolin-4-yl)phenyl]amino}-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyridine-3-carboxylic acid methyl ester (INT-A129-11B). To a stirred mixture of methyl 5-(1-methyl-5-{2-[1-{[5-(morpholin-4-yl)-2-nitrophenyl]amino}-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}pyrazol-4-yl)-6-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyridine-3-carboxylate (INT-A129-10B, 380 mg, 0.51 mmol, 1 eq) and ranyl nickel (220.59 mg, 2.57 mmol, 5 eq) in methanol (10 mL) was added hydrazine hydrate (98%) (38.67 mg, 0.77 mmol, 1.5 eq) dropwise and stirred at 0 °C for 2 h under nitrogen atmosphere. The resulting mixture was filtered. The filter cake was washed with methanol (3×50 mL). The filtrate was concentrated under reduced pressure. The crude product was used directly in the next step without further purification. LCMS: m/z (ESI), [M+H] +=708.70. Step 12. Preparation of 5-(5-{2-[1-[2-amino-6-(oxo-4-yl)-1,3-benzodiazol-1-yl]-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyridine-3-carboxylic acid methyl ester (INT-A129-12B). A mixture of 5-(5-{2-[1-{[2-amino-5-(morpholin-4-yl)phenyl]amino}-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyridine-3-carboxylic acid methyl ester (INT-A129-11B) and BrCN (62.54 mg, 0.59 mmol, 1.1 equiv) in ethanol (10 mL) was stirred at room temperature overnight under nitrogen atmosphere. The reaction was heated with saturated NaHCO3The solution (50 mL) was quenched, and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC eluting with dichloromethane/methanol (15/1) to give 250 mg of 5-(5-{2-[1-[2-amino-6-(oxolin-4-yl)-1,3-benzodiazol-1-yl]-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyridine-3-carboxylic acid methyl ester (INT-A129-12B) (63%) as a brown solid. LCMS: m/z (ESI), [M+H] +=733.65. Step 13. Preparation of 5-(5-{2-[1-[2-amino-6-(oxo-4-yl)-1,3-benzodiazol-1-yl]-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyridine-3-carboxylic acid (INT-A129-13B). A mixture of methyl 5-(5-{2-[1-[2-amino-6-(morpholin-4-yl)-1,3-benzodiazol-1-yl]-3-azabicyclo[3.2.1]octan-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyridine-3-carboxylate (INT-A129-12B, 80 mg, 0.10 mmol, 1 eq) and LiOH (4 mg, 0.17 mmol, 1.5 eq) in tetrahydrofuran (2 mL) and water (0.5 mL) was stirred at room temperature overnight under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The resulting mixture was used directly in the next step without further purification. LCMS: m/z (ESI), [M+H] +=719.55. Step 14. Preparation of 21-methyl-5-(morpholin-4-yl)-15-{[2-(trimethylsilyl)ethoxy]methyl}-23-oxo-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tris-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione (INT-A129-14B). 5-(5-{2-[1-[2-amino-6-(morpholin-4-yl)-1,3-benzodiazol-1-yl]-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyridine-3-carboxylic acid (INT-A129-13B, 80 mg, 0.10 mmol, 1 equivalent), N,N,N,N-Tetramethyl- O-(7-Azabenzotriazol-1-yl)urea hexafluorophosphate (63 mg, 0.16 mmol, 1.5 equiv.) and N,N- A mixture of diisopropylethylamine (43 mg, 0.33 mmol, 3 equiv.) in dioxane (5 mL) was stirred at room temperature under nitrogen atmosphere for 2 h. The reaction was quenched with water (50 mL), and the mixture was extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC eluting with dichloromethane/methanol (20/1) to give 50 mg of 21-methyl-5-(morpholin-4-yl)-15-{[2-(trimethylsilyl)ethoxy]methyl}-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontria-3,5,7,9,13,17(33), 18(22),19-octaene-12,16-dione (INT-A129-14B) (64%) as a white solid. LCMS: m/z (ESI), [M+H] +=701.60. Step 15. Preparation of 21-methyl-5-(morpholin-4-yl)-23-oxo-2,9,11,15,20, 21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]tris-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione (Example A129B). A mixture of 21-methyl-5-(morpholin-4-yl)-15-{[2-(trimethylsilyl)ethoxy]methyl}-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]tris-3,5,7,9,13,17(33), 18(22),19-octaene-12,16-dione (INT-A129-14B, 45 mg, 0.06 mmol, 1 equiv) in 1,4-dioxane (2 mL) in HCl was stirred at room temperature for 2 h under nitrogen atmosphere. The mixture was treated with saturated NaHCO 3The solution was neutralized to pH 7. The mixture was extracted with dichloromethane (3×20 mL), and the combined organic layers were concentrated under reduced pressure. The residue was purified by Prep-HPLC (XBridge Prep OBD C18 column, 30*150 mm, 5μm) using water (containing 10 mmol/L NH4HCO 3and 0.05% NH 3H 2O) and mobile phase B of CAN to obtain 14.3 mg of 21-methyl-5-(morpholin-4-yl)-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18, 22}]tritriacontac-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione (39%) as a white solid. LCMS: m/z (ESI), [M+H] +=571.45. 1H NMR (DMSO-d 6 , 400 MHz) δ 1.50-1.83 (4H, m), 2.00 (1H, d), 2.71 (2H, s), 3.05-3.15 (5H, m), 3.17-3.23 (3H, m), 3.62-3.92 (9H, m), 4.56 (1H, t), 4.65-4.72 (1H, m), 6.90 (1H, d), 7.19 (1H, s), 7.41 (1H, d), 7.87 (1H, s), 8.40 (1H, s), 8.82 (1H, d), 11.96 (1H, s), 12.62 (1H, s). Example A130B 15-Methyl-5-(morpholin-4-yl)-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18, 22}]tris-3,5,7,9,13,17(33),18,21-octaene-12,16-dione, isomer 2 (Example A130B) Step 1. Preparation of tert-butyl 3-hydroxy-4-iodopyrazole-1-carboxylate (INT-A130-1). To a stirred mixture of tert-butyl 3-hydroxypyrazole-1-carboxylate (4 g, 21.71 mmol, 1 eq.) in acetonitrile (80 mL) was added NIS (5.86 g, 26.05 mmol, 1.2 eq.) in portions at 0 °C under nitrogen atmosphere. The resulting mixture was stirred at 0 °C under nitrogen atmosphere for 2 h. The reaction was quenched with saturated sodium thiosulfate solution, and the mixture was extracted with ethyl acetate (3×200 mL). The combined organic layers were washed with brine (3×200 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue is purified by C18 FLASH using water and acetonitrile as mobile phase. The fractions containing the desired compound are evaporated to dryness to give 2.5 g of tert-butyl 3-hydroxy-4-iodopyrazole-1-carboxylate (40%) as a brown solid. LCMS: m/z (ESI), [M+H] += 311.00. 1H NMR (DMSO-d 6 , 400 MHz) δ 1.53 (9H, s), 8.18 (1H, s), 11.63 (1H, s). Step 2. Preparation of tert-butyl 4-iodo-3-{2-[1-{[5-(oxo-4-yl)-2-nitrophenyl]amino}-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}pyrazole-1-carboxylate (INT-A130-2B). To a stirred mixture of 2-[1-{[5-(morpholin-4-yl)-2-nitrophenyl]amino}-3-azabicyclo[3.2.1]octan-3-yl]ethanol (INT-A129-9B, 500 mg, 1.32 mmol, 1.00 equiv) and triphenylphosphine (1045 mg, 3.98 mmol, 3 equiv) in tetrahydrofuran (10 mL) under nitrogen atmosphere at 0° C. was added diisopropyl azodicarboxylate (805 mg, 3.98 mmol, 3 equiv) and tert-butyl 3-hydroxy-4-iodopyrazole-1-carboxylate (617 mg, 1.99 mmol, 1.5 equiv). The resulting mixture was stirred at 0° C. for 2 h under nitrogen atmosphere. The reaction mixture was quenched with water (100 mL), and the mixture was extracted with dichloromethane (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using petroleum ether/ethyl acetate (1/1) to give 550 mg of tert-butyl 4-iodo-3-{2-[1-{[5-(oxolin-4-yl)-2-nitrophenyl]amino}-3-azabicyclo[3.2.1]octan-3-yl]ethoxy}pyrazole-1-carboxylate (61%) as a yellow solid. LCMS: m/z (ESI), [M+H] += 669.40. Step 3. Preparation of 5-[1-(tert-butoxycarbonyl)-3-{2-[1-{[5-(oxoline-4-yl)-2-nitrophenyl]amino}-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}pyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (INT-A130-3B). 4-iodo-3-{2-[1-{[5-(oxoline-4-yl)-2-nitrophenyl]amino}-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}pyrazole-1-carboxylic acid tert-butyl ester (INT-A130-3B, 550 mg, 0.823 mmol, 1 equivalent), K 2CO 3(341 mg, 2.46 mmol, 3 equivalents), Pd(dppf)Cl 2CH 2Cl 2A mixture of methyl 1-methyl-6-oxo-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine-3-carboxylate (67 mg, 0.08 mmol, 0.1 equiv) and methyl 1-methyl-6-oxo-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine-3-carboxylate (361 mg, 1.23 mmol, 1.5 equiv) in dioxane (20 mL) and water (5 mL) was stirred at 80 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature, treated with water (100 mL), and extracted with dichloromethane (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. Pass the residue through C18 FLASH using 10 mmol/L NH4HCO 3and 0.1% NH 3 .H 2O water and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 130 mg of 5-[1-(tert-butoxycarbonyl)-3-{2-[1-{[5-(oxolin-4-yl)-2-nitrophenyl]amino}-3-azabicyclo[3.2.1]octan-3-yl]ethoxy}pyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylic acid methyl ester (22%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=708.55. Step 4. Preparation of 5-(3-{2-[1-[2-amino-6-(oxo-4-yl)-1,3-benzodiazol-1-yl]-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1H-pyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A130-4B). To a stirred mixture of ranyl nickel (6.05 mg, 0.07 mmol, 1 eq.) and methyl 5-[1-(tert-butoxycarbonyl)-3-{2-[1-{[5-(morpholin-4-yl)-2-nitrophenyl]amino}-3-azabicyclo[3.2.1]octan-3-yl]ethoxy}pyrazol-4-yl]-1-methyl-6-oxopyridine-3-carboxylate (50 mg, 0.07 mmol, 1 eq.) in methanol (1 mL) at 0 °C under nitrogen atmosphere was added hydrazine hydrate (10.61 mg, 0.21 mmol, 3 eq.). The resulting mixture was stirred at room temperature for 2 h. The resulting mixture was filtered, and the filter cake was washed with methanol (3×5 mL). The filtrate was concentrated under reduced pressure. To this residue, ethanol (1 mL) and BrCN (8.23 mg, 0.07 mmol, 1.1 equiv) were added. The resulting mixture was stirred at room temperature for another 2 h and treated with LiOH.H 2O (5.93 mg, 0.14 mmol, 2 equivalents) in H 2O (0.2 mL). The resulting mixture was stirred at 40 °C for 2 h and concentrated under reduced pressure. The residue was purified by reverse-phase flash chromatography using 10% to 50% acetonitrile/water elution to give 15 mg of 5-(3-{2-[(1S,5S)-1-[2-amino-6-(oxolin-4-yl)-1,3-benzodiazol-1-yl]-3-azabicyclo[3.2.1]octan-3-yl]ethoxy}-1H-pyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (36%) as a white solid. LCMS: m/z (ESI), [M+H] +=589.40. Step 5. Preparation of 15-methyl-5-(morpholin-4-yl)-23-oxo-2,9,11,15,20, 21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]tris-3,5,7,9,13,17(33),18,21-octaene-12,16-dione (Example A130B). 5-(3-{2-[1-[2-amino-6-(morpholin-4-yl)-1,3-benzodiazol-1-yl]-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1H-pyrazol-4-yl)-1-methyl-6-oxopyridine-3-carboxylic acid (INT-A130-4B, 20 mg, 0.03 mmol, 1 equivalent), N,N,N,N-Tetramethyl- O-(7-Azabenzotriazol-1-yl)urea hexafluorophosphate (19.38 mg, 0.05 mmol, 1.5 equivalents) and N,N- A mixture of diisopropylethylamine (13.17 mg, 0.10 mmol, 3 equiv.) in 1,4-dioxane (2 mL) was stirred at 40 °C for 2 h under nitrogen atmosphere. The reaction was quenched with water (30 mL), and the mixture was extracted with ethyl acetate (3×30 mL). The combined organic layers were washed with brine (3×30 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column eluting with dichloromethane/methanol (25/1) to give 4.9 mg of 15-methyl-5-(morpholin-4-yl)-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}. 1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontac-3,5,7,9,13, 17(33),18,21-octaene-12,16-dione (25%) as a white solid. LCMS: m/z (ESI), [M+H] +=571.20. 1H NMR (DMSO-d 6, 400 MHz) δ 1.51-1.61 (2H, m), 1.68 (1H, d), 2.45-2.67 (6H, m), 3.09 (4H, q), 3.37-3.55 (3H, m), 3.62 (3H, s), 3.76 (4H, t), 4.14 (2H, d), 4.32 (1H, d), 6.87-6.93 (1H, m), 7.18 (1H, d), 7.42 (1H, d), 8.20 (1H, d), 8.44 (1H, d), 9.19 (1H, d), 11.95 (1H, s), 12.53 (1H, s). Example A132B 5-Hydroxy-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18, 22}]tris-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione, isomer 2 (Example A132B) 5-Bromo-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]tris-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione (M4B, 150 mg, 0.26 mmol, 1 equivalent), Pd 2(dba) 3(47.49 mg, 0.05 mmol, 0.2 eq.), t-BuXPhos (22.02 mg, 0.052 mmol, 0.2 eq.), KOH (58.19 mg, 1.036 mmol, 4 eq.) in 1,4-dioxane (3 mL) and H 2The mixture in 2% O (2 mL) was stirred at 100 °C for 2 h under nitrogen atmosphere. The reaction was quenched with water (50 mL), and the mixture was extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by C18 FLASH using water and acetonitrile as mobile phase. The fractions containing the desired compound were evaporated to dryness to give 4.8 mg of 5-hydroxy-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontria-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione (isomer 2) (3%) as a white solid. LCMS: m/z (ESI), [M+H] +=516.25. 1H NMR (DMSO-d 6, 400 MHz) δ 1.52-1.63 (1H, m), 1.63-1.79 (2H, m), 1.99 (1H, d), 2.42-2.72 (8H, m), 3.62 (3H, s), 3.63-3.69 (1H, m), 3.71 (3H, s), 4.54 (1H, t), 4.67 (1H, d), 6.63 (1H, dd), 7.16 (1H, d), 7.35 (1H, d), 8.26 (1H, d), 8.41 (1H, s), 8.81 (1H, d), 9.32 (1H, s), 12.55 (1H, s). Example A133B 5-[(2-Hydroxyethyl)amino]-15,21-dimethyl-23-oxo-2,9,11,15,20, 21,26-heptaazaheptacyclic[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]tris-3,5,7,9,13,17(33), 18(22),19-octaene-12,16-dione, isomer 2 Step 1. 5-({2-[(tert-butyldimethylsilyl)oxy]ethyl}amino)-15,21-dimethyl-23-oxo-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}] -3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione (INT-A133-1B). 5-Bromo-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,28}.1^{13,17}. A mixture of 0^{2,10}.0^{3,8}.0^{18,22}]tris-3,5,7,9,13,17(33), 18(22),19-octaene-12,16-dione (120 mg, 0.20 mmol, 1 eq.), (2-aminoethoxy)(tert-butyl)dimethylsilane (M4B, 109.12 mg, 0.62 mmol, 3 eq.), BrettPhos Pd G3 (56.41 mg, 0.06 mmol, 0.3 eq.) and LiHMDS (347.12 mg, 2.07 mmol, 10 eq.) in dioxane (5 mL) was stirred at 60 °C for 2 h under nitrogen atmosphere. The reaction was heated with NH 4Cl(aqueous solution) (50 mL) was quenched, and the mixture was extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×50 mL) and purified by anhydrous Na 2SO 4The residue was purified by silica gel column chromatography using dichloromethane/methanol (20:1) to give 65 mg of 5-({2-[(tert-butyldimethylsilyl)oxy]ethyl}amino)-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontria-3,5,7,9,13,17(33), 18(22),19-octaene-12,16-dione as a yellow solid (46%). LCMS: m/z (ESI), [M+H] +=673.30. Step 2. 5-[(2-Hydroxyethyl)amino]-15,21-dimethyl-23-oxo-2,9,11,15,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]tri-3,5,7,9,13,17(33), 18(22),19-octaene-12,16-dione (Example A133B). 5-({2-[(tert-butyldimethylsilyl)oxy]ethyl}amino)-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tris-3,5,7,9,13, 17(33),18(22),19-octaene-12,16-dione (INT-A133-1B, 60 mg, 0.09 mmol, 1 equivalent) was dissolved in HCl in 1,4-dioxane (2 The solution in 10% ethanol (20% ethanol) was stirred at 15 °C for 2 h under nitrogen atmosphere. The reaction was treated with NaHCO3The solution (50 mL) was quenched and the mixture was extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×50 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by C18 FLASH using water and acetonitrile as mobile phase. The fractions containing the desired compound were evaporated to dryness to give 32.3 mg of 5-[(2-hydroxyethyl)amino]-15,21-dimethyl-23-oxa-2,9,11,15,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18, 22}]tris-3,5,7,9,13,17(33),18(22),19-octaene-12,16-dione (60%) as a white solid. LCMS: m/z (ESI), [M+H] +=559.20. 1H NMR (DMSO-d 6, 400 MHz) δ 1.48-1.82 (3H, m), 1.98 (1H, d), 2.42 (3H, d), 2.56 (1H, s), 2.73 (1H, d), 3.10 (2H, s), 3.18-3.25 (1H, m), 3.31 (1H, s), 3.55-3.63 (6H, m), 3.66 (1H, d), 3.72 (3H, s), 4.55 (1H, t), 4.64-4.74 (2H, m), 5.47 (1H, s), 6.51 (1H, dd), 6.96 (1H, s), 7.28 (1H, d), 8.25 (1H, d), 8.41 (1H, s), 8.82 (1H, d), 12.47 (1H, s). Example B1 15,21-Dimethyl-23-oxo-2,9,11,16,20,21,26-heptaazaheptacyclic [26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontetrazol-3,5,7,9,13,15,17(34),18(22), 19-nonan-12-one Step 1. Preparation of methyl 2-methyl-6-[1-methyl-5-(2-{1-[(2-nitrophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethoxy)pyrazol-4-yl]pyridine-4-carboxylate (INT-B1-1). To a stirred mixture of 2-{1-[(2-nitrophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethanol (250 mg, 0.81 mmol, 1 eq) and methyl 2-(5-hydroxy-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylate (M1, 202 mg, 0.81 mmol, 1 eq) in toluene (100 mL) was added 2-(tributyl-λ5-phosphinilidene)acetonitrile (1580 mg, 6.55 mmol, 8 eq) under nitrogen atmosphere. The mixture was stirred at 100 °C for 4 h, cooled to room temperature, and concentrated under reduced pressure. The reaction was quenched with water (80 mL), and the mixture was extracted with ethyl acetate (3×150 mL). The combined organic layers were washed with brine (3×150 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (10/1) to give 420 mg of methyl 2-methyl-6-[1-methyl-5-(2-{1-[(2-nitrophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethoxy)pyrazol-4-yl]pyridine-4-carboxylate (INT-B1-1) (95%) as a yellow oil. LCMS: m/z (ESI), [M+H] +=535.30. Step 2. Preparation of methyl 2-[5-(2-{1-[(2-aminophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethoxy)-1-methylpyrazol-4-yl]-6-methylpyridine-4-carboxylate (INT-B1-2). To a stirred mixture of methyl 2-methyl-6-[1-methyl-5-(2-{1-[(2-nitrophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethoxy)pyrazol-4-yl]pyridine-4-carboxylate (400 mg, 0.75 mmol, 1 eq.) and ranyl nickel (512 mg, 5.98 mmol, 8 eq.) in MeOH (30 mL) was added NH 2NH 2.H 2O (74 mg, 1.50 mmol, 2 eq.). The resulting mixture was stirred at room temperature for 2 h and filtered. The filter cake was washed with methanol (3×20 mL). The resulting solution was concentrated under reduced pressure to give 360 mg of 2-[5-(2-{1-[(2-aminophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethoxy)-1-methylpyrazol-4-yl]-6-methylpyridine-4-carboxylic acid methyl ester (INT-B1-2) as a yellow solid. This material was used in the next reaction without further purification. LCMS: m/z (ESI), [M+H] +=505.30. Step 3. Preparation of methyl 2-(5-{2-[1-(2-amino-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.2]non-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylate (INT-B26-3). To a stirred mixture of methyl 2-[5-(2-{1-[(2-aminophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethoxy)-1-methylpyrazol-4-yl]-6-methylpyridine-4-carboxylate (360 mg, 0.71 mmol, 1 eq.) and BrCN (113 mg, 1.06 mmol, 1.5 eq.) in ethanol (30 mL) was added under nitrogen atmosphere at room temperature. The resulting mixture was stirred at room temperature for 1 h and concentrated under vacuum. The residue was purified by Prep-TLC using dichloromethane/methanol (15/1) to give 256 mg of methyl 2-(5-{2-[1-(2-amino-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.2]non-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylate (INT-B26-3) (67%) as a pink solid. LCMS: m/z (ESI), [M+H]+=530.35. Step 4. Preparation of 2-(5-{2-[1-(2-amino-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.2]non-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylic acid (INT-B1-4). 2-(5-{2-[1-(2-amino-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.2]non-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylic acid methyl ester (250 mg, 0.47 mmol, 1 equivalent) and LiOH.H 2O (39 mg, 0.94 mmol, 2 equivalents) in tetrahydrofuran (20 mL) and H 2The mixture in 4 mL of O was stirred at room temperature overnight and concentrated in vacuo. The residue was purified by C18 FLASH chromatography using 10 mmol/L NH4HCO 3and 0.1% NH 3 .H 2Purification was performed using water with 0.05% ethanol and acetonitrile as the mobile phase. The fractions containing the desired compound were evaporated to dryness to give 170 mg of 2-(5-{2-[1-(2-amino-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.2]non-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylic acid (INT-B1-4) (69%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=516.15. 1H NMR (CD 3OD, 400 MHz) δ 1.78-1.98 (7H, m), 2.43 (3H, s), 2.64 (2H, d), 2.82 (2H, t), 2.90 (2H, t), 3.16 (2H, s), 3.68 (3H, s), 4.26 (2H, t), 3.74-6.79 (1H, m), 6.89 (1H, t), 7.07-7.10 (1H, m), 7.35 (1H, d), 7.38 (1H, d), 7.73 (1H, s), 7.76 (1H, s). Step 5. Preparation of 15,21-dimethyl-23-oxa-2,9,11,16,20,21,26-heptaazaheptacyclo[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontria-3,5,7,9,13,15,17(34),18(22), 19-nonan-12-one (Example B1). Under nitrogen atmosphere at room temperature, 2-(5-{2-[1-(2-amino-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.2]non-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylic acid (160 mg, 0.31 mmol, 1 equivalent) andN, N, N, N-Tetramethyl- O-(7-Azabenzotriazol-1-yl)urea hexafluorophosphate (353 mg, 0.93 mmol, 3 equivalents) was added to a stirred mixture of 1,4-dioxane (10 mL) N, N-Diisopropylethylamine (160 mg, 1.24 mmol, 4 equiv.). The mixture was stirred at 60 °C for 3 h under air atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water at room temperature. The resulting mixture was extracted with dichloromethane (3×200 mL). The combined organic layers were washed with brine (3×150 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was subjected to Prep-HPLC using an XBridge Shield RP18 OBD column with 10 mmol/L NH4HCO 3and 0.1% NH 3.H 2O water and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 63.1 mg of 15,21-dimethyl-23-oxa-2,9,11,16,20,21,26-heptaazaheptacyclo[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontria-3,5,7,9,13,15,17(34),18(22), 19-nonan-12-one (Example B1) as a white solid (40%). LCMS: m/z (ESI), [M+H] +=498.20. 1H NMR (DMSO-d 6 ,400 MHz) δ 1.82-2.04 (4H, m), 2.04-2.15 (3H, m), 2.55 (3H, s), 2.82 (2H, s), 2.87 (2H, t), 3.42-3.55 (4H, m), 3.77 (3H, s), 4.38 (2H, t), 7.18 (2H, dt), 7.62 (1H, d), 7.65 (1H, d), 7.89 (1H, s), 7.91 (1H, d), 8.77 (1H, s), 13.32 (1H, s). Example B2 15,21-Dimethyl-5-(morpholin-4-yl)-23-oxo-2,9,11,16,20,21,26-heptaazaheptacyclic[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]triacontria-3,5,7,9,13,15,17(34),18(22), 19-nonan-12-one Step 1. Preparation of methyl 2-[5-(2-{1-[(5-bromo-2-nitrophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethoxy)-1-methylpyrazol-4-yl]-6-methylpyridine-4-carboxylate (INT-B2-1). A mixture of 2-{1-[(5-bromo-2-nitrophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethanol (380 mg, 0.98 mmol, 1 eq), 2-(tributyl-λ5-phosphinilidene)acetonitrile (1193 mg, 4.94 mmol, 5 eq) and methyl 2-(5-hydroxy-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylate (M1, 366 mg, 1.48 mmol, 1.5 eq) in toluene (5 mL) was stirred at 100 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (100 mL), and the mixture was extracted with dichloromethane (3×100 mL). The combined organic layers were washed with brine (3×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on Prep-TLC using petroleum ether/ethyl acetate (3/1) to give 500 mg of methyl 2-[5-(2-{1-[(5-bromo-2-nitrophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethoxy)-1-methylpyrazol-4-yl]-6-methylpyridine-4-carboxylate (INT-B2-1) (82%) as an orange oil. LCMS: m/z (ESI), [M+H] +=613.15. 1H NMR (DMSO-d 6 , 400 MHz) δ 1.44-1.47 (2H, m), 1.74-1.81 (6H, m), 1.89-1.96 (1H, m), 2.48 (3H, s), 2.66-2.72 (2H, m), 2.89-2.97 (4H, m), 3.74 (3H, s), 3.84 (3H, s), 4.39 (2H, t), 6.72-6.77 (1H, m), 7.01 (1H, d), 7.33 (1H, d), 7.77 (1H, d), 7.86 (1H, s), 7.95 (1H, d), 8.06 (1H, s). Step 2. Preparation of methyl 2-[5-(2-{1-[(2-amino-5-bromophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethoxy)-1-methylpyrazol-4-yl]-6-methylpyridine-4-carboxylate (INT-B2-2). To a stirred mixture of methyl 2-[5-(2-{1-[(5-bromo-2-nitrophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethoxy)-1-methylpyrazol-4-yl]-6-methylpyridine-4-carboxylate (480 mg, 0.78 mmol, 1 eq) and ranyl nickel (335 mg, 3.91 mmol, 5 eq) in methanol (5 mL) was added hydrazine hydrate (98%) (61 mg, 1.22 mmol, 1.5 eq) at 0 °C under nitrogen atmosphere. The mixture was stirred at 0 °C under nitrogen atmosphere for 2 h. The reaction was quenched with water (100 mL), and the mixture was extracted with dichloromethane (3×100 mL). The combined organic layers were washed with brine (3×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using petroleum ether/ethyl acetate (1/1) to give 370 mg of 2-[5-(2-{1-[(2-amino-5-bromophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethoxy)-1-methylpyrazol-4-yl]-6-methylpyridine-4-carboxylic acid methyl ester (INT-B2-2) (81 %) as a brown solid. LCMS: m/z (ESI), [M+H] +=583.20. 1H NMR (DMSO-d 6 , 400 MHz) δ 1.38-1.49 (2H, m), 1.69-1.76 (4H, m), 1.85-1.88 (1H, m), 2.25-2.32 (2H, m), 2.52 (3H, s), 2.63-2.68 (2H, m), 2.78-2.83 (2H, m), 2.84-2.91 (2H, m), 3.72 (3H, s), 3.86 (3H, s), 4.03 (1H, s), 4.27-4.37(2H, m), 4.68 (2H, s), 6.46 (2H, d), 6.58 (1H, d), 7.43 (1H, d), 7.83 (1H, d), 7.89 (1H, s). Step 3. Preparation of methyl 2-(5-{2-[1-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.2]non-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylate (INT-B2-3). A mixture of methyl 2-[5-(2-{1-[(2-amino-5-bromophenyl)amino]-3-azabicyclo[3.2.2]non-3-yl}ethoxy)-1-methylpyrazol-4-yl]-6-methylpyridine-4-carboxylate (350 mg, 0.60 mmol, 1 eq) and BrCN (95 mg, 0.90 mmol, 1.5 eq) in ethanol (2 mL) was stirred at room temperature overnight under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by Prep-TLC using petroleum ether/ethyl acetate (1/3) to give 360 mg of methyl 2-(5-{2-[1-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.2]non-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylate (INT-B2-3) (98%) as a brown solid. LCMS: m/z (ESI), [M+H] +=610.15. 1H NMR (DMSO-d 6 , 400 MHz) δ 1.79-1.97 (7H, m), 2.51 (3H, s), 2.70-2.74 (2H, m), 2.79-2.87 (2H, m), 2.92-2.99 (2H, m), 3.18 (2H, s), 3.74 (3H, s), 3.84 (3H, s), 4.37 (2H, t), 6.05 (2H, s), 6.99-7.08 (2H, m), 7.41 (1H, d), 7.46 (1H, d), 7.81 (1H, d), 7.89 (1H, s). Step 4. Preparation of lithium 2-(5-{2-[1-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.2]non-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpicolinate (INT-B2-4). A mixture of methyl 2-(5-{2-[1-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.2]non-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylate (340 mg, 0.55 mmol, 1 eq) and LiOH (16 mg, 0.67 mmol, 1.2 eq) in ethanol (2 mL) was stirred at 80 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature and concentrated under reduced pressure to give 2-(5-{2-[1-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.2]non-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylic acid (INT-B2-4) as a white solid. The crude product was used directly in the next reaction without further purification. LCMS: m/z (ESI), [M+H] +=594.45. Step 5. Preparation of 5-bromo-15,21-dimethyl-23-oxa-2,9,11,16,20,21, 26-heptaazaheptacyclic [26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3, 8}.0^{18,22}]triacontriacontria-3,5,7,9,13,15,17(34), 18(22),19-nonan-12-one (INT-B2-5). 2-(5-{2-[1-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.2]non-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylic acid (330 mg crude, about 0.5 mmol, about 1 equivalent), N,N,N,N-Tetramethyl- O-(7-Azabenzotriazol-1-yl)urea hexafluorophosphate (417 mg, 1.10 mmol, 2 equivalents) and N,N- A mixture of diisopropylethylamine (213 mg, 1.65 mmol, 3 equiv.) in 1,4-dioxane (5 mL) was stirred at 60 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (50 mL), and the mixture was extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×150 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by trituration with petroleum ether (10 mL) to give 280 mg of 5-bromo-15,21-dimethyl-23-oxa-2,9,11,16,20,21,26-heptaazaheptacyclo[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]triacontriacontria-3,5,7,9,13,15,17(34), 18(22),19-nonan-12-one (INT-B2-5) (88%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=578.05. Step 6. Preparation of 15,21-dimethyl-5-(morpholin-4-yl)-23-oxo-2,9,11, 16,20,21,26-heptaazaheptacyclo[26.2.2.1^{1,26}.1^{13,17}.0^{2, 10}.0^{3,8}.0^{18,22}]triacontria-3,5,7,9,13,15,17(34), 18(22),19-nonan-12-one (Example B2). 5-Bromo-15,21-dimethyl-23-oxa-2,9,11,16,20,21,26-heptaazaheptacyclic [26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]triacontria-3,5,7,9,13,15,17(34),18(22),19-nonan-12-one (100 mg, 0.17 mmol, 1 equivalent), Brettphos Pd G 3To a stirred mixture of 1,4-dioxane (10 mL) and morpholine (47 mg, 0.05 mmol, 0.3 equiv) was added LiHMDS (2 mL, 2.00 mmol, 11.53 equiv) dropwise. The resulting mixture was stirred at 60 °C under nitrogen atmosphere for 1 h and cooled to room temperature. The reaction was heated with saturated NH4Cl(aq) (50 mL) was quenched and the mixture was washed with CH2Cl 2(3×50 mL) extraction. The combined organic layers were washed with brine (3×50 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was subjected to prep-HPLC using an XBridge Prep OBD C18 column with 10 mmol/L NH4HCO 3of water and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 14.4 mg of 15,21-dimethyl-5-(morpholin-4-yl)-23-oxa-2,9,11,16,20, 21,26-heptaazaheptacyclo[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontria-3,5,7,9,13,15,17(34),18(22),19-nonan-12-one (Example B2) as a yellow solid (14%). LCMS: m/z (ESI), [M+H] += 583.40. 1H NMR (DMSO-d 6 , 400 MHz) δ 1.88-1.99 (4H, m), 2.03-2.11 (3H, m), 2.54 (3H, s), 2.78-2.90 (4H, m), 3.04-3.10 (4H, m), 3.42-3.50 (4H, m), 3.75-3.80 (7H, m), 4.36-4.40 (2H, m), 6.95 (1H, dd), 7.35 (1H, s), 7.51 (1H, d), 7.61 (1H, d), 7.89 (1H, s), 8.76 (1H, s), 13.21 (1H, s). Example B3 15,21-Dimethyl-5-(4-methylpiperazine-1-yl)-23-oxo-2,9,11,16, 20,21,26-heptaazaheptacyclic[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]triacontetrazol-3,5,7,9,13,15,17(34),18(22), 19-nonan-12-one Step 1. Preparation of 15,21-dimethyl-5-(4-methylpiperazin-1-yl)-23-oxa-2, 9,11,16,20,21,26-heptaazaheptacyclo[26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontria-3,5,7,9,13,15,17(34), 18(22),19-nonan-12-one (Example B3). 5-Bromo-15,21-dimethyl-23-oxa-2,9,11,16,20,21,26-heptaazaheptacyclic [26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]triacontria-3,5,7,9,13,15,17(34),18(22),19-nonan-12-one (100 mg, 0.17 mmol, 1 equivalent), BrettPhos Pd G 3To a stirred mixture of 1,4-dioxane (10 mL) and 1-methylpiperazine (550 mg, 0.60 mmol, 3.5 equiv) was added LiHMDS (2 mL, 2.000 mmol, 11.53 equiv) dropwise. The resulting mixture was stirred at 60 °C under nitrogen atmosphere for 1 h and cooled to room temperature. The reaction was heated with saturated NH4Cl(aq) (50 mL) was quenched and the mixture was washed with CH2Cl 2(3×50 mL) extraction. The combined organic layers were washed with brine (3×50 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was subjected to prep-HPLC using an XBridge Shield RP18 OBD column with 10 mmol/L NH4HCO 3of water and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to obtain 13.7 mg of 15,21-dimethyl-5-(4-methylpiperazin-1-yl)-23-oxa-2,9,11,16,20,21,26-heptaazaheptacyclic [26.2.2.1^{1,26}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontria-3,5,7,9,13,15,17(34), 18(22),19-nonan-12-one (Example B3) (13%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=596.40. 1H NMR (DMSO-d 6 , 400 MHz) δ 1.84-2.01 (4H, m), 2.03-2.10 (3H, m), 2.24 (3H, s), 2.54 (4H, s), 2.79-2.84 (2H, m), 2.85-2.90 (2H, m), 3.10 (4H, t), 3.28-3.36 (3H, m), 3.42-3.50 (4H, m), 3.76 (3H, s), 4.37 (2H, d), 6.94 (1H, dd), 7.34 (1H, s), 7.49 (1H, d), 7.61 (1H, d), 7.89 (1H, s), 8.76 (1H, s), 13.20 (1H, s). Example B4 15,21-Dimethyl-23-oxa-2,9,11,16,20,21,26-heptaazahexacyclo[24.4.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontahedral-3,5,7,9,13(32),14,16,18(22),19-nonan-12-one Step 1. Preparation of tert-butyl 3-[(2-nitrophenyl)amino]azoloheptan-1-carboxylate (INT-B4-1). Add o-fluoronitrobenzene (5.00 g, 35.44 mmol, 1 equivalent), K 2CO 3A mixture of (9.79 g, 70.87 mmol, 2 eq.) and tert-butyl 3-aminoazacycloheptane-1-carboxylate (7.59 g, 35.44 mmol, 1 eq.) in acetonitrile (80 mL) was stirred at 80 °C for 3 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (200 mL), and the mixture was extracted with ethyl acetate (3×200 mL). The combined organic layers were washed with brine (3×200 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by silica gel chromatography using petroleum ether/ethyl acetate (21/1) to give 9 g of tert-butyl 3-[(2-nitrophenyl)amino]azanacycloheptane-1-carboxylate (INT-B4-1) (75%) as a reddish brown oil. LCMS: m/z (ESI), [M+H] +=336.15. 1H NMR (400 MHz, DMSO-d 6) δ 1.38 (9H, s), 1.49-1.65 (1H, m), 1.62-1.75 (3H, m), 1.75-1.82 (1H, m), 1.96-2.01 (1H, m), 3.37-3.50 (2H , m), 3.61-3.69 (1H, m), 3.83-4.03 (2H, m), 6.66-6.76 (1H, m), 7.21-7.25 (1H, m), 7.51-7.61 (1H, m), 8.01 -8.13 (1H, m), 8.20 (1H, d). Step 2. N- Preparation of (2-nitrophenyl) azacycloheptane-3-amine (INT-B4-2). To a stirred mixture of tert-butyl 3-[(2-nitrophenyl) amino] azacycloheptane-1-carboxylate (4.00 g, 11.93 mmol, 1 equiv.) in dichloromethane (12 mL) was slowly added 1,4-dioxane (60 mL) containing HCl at room temperature. The resulting mixture was stirred at room temperature for 1.5 h. The mixture was concentrated under reduced pressure. The residue was treated with saturated NaHCO 3(aqueous solution) (100 mL) and extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure to obtain 2.8 g of yellow oil.N-(2-Nitrophenyl)azacycloheptane-3-amine (INT-B4-2). This material was used in the next reaction without further purification. LCMS: m/z (ESI), [M+H] +=236.10. Step 3. 1-{2-[(tert-butyldimethylsilyl)oxy]ethyl}- NPreparation of -(2-nitrophenyl) azacycloheptyl-3-amine (INT-B4-3). Freshly prepared N-(2-nitrophenyl) azacycloheptane-3-amine (2.8 g, about 1 equivalent), (2-bromoethoxy)(tert-butyl)dimethylsilane (6.39 g, 26.69 mmol, 2 equivalents), K 2CO 3A mixture of (3.69 g, 26.69 mmol, 2 equiv.) and KI (4.43 g, 26.69 mmol, 2 equiv.) in DMF (80 mL) was stirred at 60 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (100 mL), and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by prep-TLC using petroleum ether/ethyl acetate (19/1) to give 5.2 g of 1-{2-[(tert-butyldimethylsilyl)oxy]ethyl}- N-(2-Nitrophenyl) azacycloheptane-3-amine (INT-B4-3) (99%). LCMS: m/z (ESI), [M+H] +=394.20. 1H NMR (400 MHz, DMSO-d 6) δ 0.03 (6H, s), 0.81 (9H, s), 1.45-1.63 (3H, m), 1.74-1.81 (3H, m), 2.57-2.77 (3H, m), 2.78-2.95 (3H, m), 3.29 (1H, t), 3.63-3.84 (3H, m), 6.65-6.71 (1H, m), 7.05 (1H, d), 7.51-7.58 (1H, m), 8.07-8.13 (1H, m), 8.74 (1H, d). Step 4. Preparation of 2-{3-[(2-nitrophenyl)amino]azanacycloheptan-1-yl}ethanol (INT-B4-4). To a stirred mixture of 1-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-N-(2-nitrophenyl)azanacycloheptyl-3-amine (3 g, 7.62 mmol, 1 equiv) in tetrahydrofuran (50 mL) was added tetrahydrofuran (12 mL) containing tetrabutylammonium fluoride at room temperature under nitrogen atmosphere. The resulting mixture was stirred at room temperature for 1 h. The reaction was quenched with water (100 mL), and the mixture was extracted with dichloromethane (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using petroleum ether/ethyl acetate (1/1) to give 1.5 g of 2-{3-[(2-nitrophenyl)amino]azanacycloheptan-1-yl}ethanol (INT-B4-4) (70%) as a yellow oil. LCMS: m/z (ESI), [M+H] +=280.10. Step 5. Preparation of 2-methyl-6-[1-methyl-5-(2-{3-[(2-nitrophenyl)amino]azanacyclohept-1-yl}ethoxy)pyrazol-4-yl]pyridine-4-carboxylic acid methyl ester (INT-B4-5). To a stirred mixture of 2-{3-[(2-nitrophenyl)amino]azanacycloheptan-1-yl}ethanol (1.5 g, 5.370 mmol, 1 eq) and triphenylphosphine (4.30 g, 16.38 mmol, 3.05 eq) in tetrahydrofuran (30 mL) was added methyl 2-(5-hydroxy-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylate (M1, 1.46 g, 5.91 mmol, 1.1 eq) and diisopropyl azodicarboxylate (2.71 g, 13.43 mmol, 2.5 eq) at 0 °C under nitrogen atmosphere. The resulting mixture was stirred at room temperature for 2 h. The reaction was quenched with water (300 mL) and the mixture was extracted with dichloromethane (3×300 mL). The combined organic layers were washed with brine (3×300 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by reverse phase flash chromatography using 10% to 50% acetonitrile/water elution to give 1.6 g of methyl 2-methyl-6-[1-methyl-5-(2-{3-[(2-nitrophenyl)amino]azanacyclohept-1-yl}ethoxy)pyrazol-4-yl]pyridine-4-carboxylate (INT-B4-5) (59%) as a yellow oil. LCMS: m/z (ESI), [M+H] +=509.25. 1H NMR (400 MHz, DMSO-d 6) δ 1.44-1.80 (6H, m), 2.48-2.54 (2H, m), 2.71-2.81 (1H, m), 2.85-2.92 (2H, m), 2.94-3.14 (3H, m), 3.70 (3H, s), 3.86 (3H, s), 3.92 (1H, s), 4.28-4.35 (2H, m), 6.63-6.68 (1H, m), 6.96-7.03 (1H, m), 7.38 (1H, d), 7.46-7.51 (1H, m), 7.80-7.83 (1H, m), 7.88 (1H, s), 8.00-8.05 (1H, m), 8.62 (1H, d). Step 6. Preparation of methyl 2-[5-(2-{3-[(2-aminophenyl)amino]azacyclohept-1-yl}ethoxy)-1-methylpyrazol-4-yl]-6-methylpyridine-4-carboxylate (INT-B4-6). To a stirred mixture of methyl 2-methyl-6-[1-methyl-5-(2-{3-[(2-nitrophenyl)amino]azacyclohept-1-yl}ethoxy)pyrazol-4-yl]pyridine-4-carboxylate (1.00 g, 1.97 mmol, 1 eq.) and ranyl nickel (340 mg, 3.93 mmol, 2 eq.) in MeOH (30 mL) was added NH 2NH 2.H 2O (295 mg, 5.90 mmol, 3 eq.). The resulting mixture was stirred at room temperature for 1 h. The mixture was filtered and the filter cake was washed with dichloromethane (3×30 mL). The resulting solution was concentrated under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (30/1) to give 500 mg of 2-[5-(2-{3-[(2-aminophenyl)amino]azanacyclohept-1-yl}ethoxy)-1-methylpyrazol-4-yl]-6-methylpyridine-4-carboxylic acid methyl ester (INT-B4-6) (53%) as a reddish brown oil. LCMS: m/z (ESI), [M+H] +=479.25. Step 7. Preparation of methyl 2-(5-{2-[3-(2-amino-1,3-benzodiazol-1-yl)azepine-1-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylate (INT-B4-7). A mixture of methyl 2-[5-(2-{3-[(2-aminophenyl)amino]azepine-1-yl}ethoxy)-1-methylpyrazol-4-yl]-6-methylpyridine-4-carboxylate (490 mg, 1.02 mmol, 1 eq.) and cyanogen bromide (108 mg, 1.02 mmol, 1 eq.) in dichloromethane (5 mL) was stirred at room temperature for 2 h under a nitrogen atmosphere. The mixture was concentrated under reduced pressure, and the residue was purified by Prep-TLC using dichloromethane/methanol (30/1) to give 490 mg of methyl 2-(5-{2-[3-(2-amino-1,3-benzodiazol-1-yl)azinecyclohept-1-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylate (INT-B4-7) (95%) as a yellow oil. LCMS: m/z (ESI), [M+H] +=504.25. 1H NMR (400 MHz, DMSO-d 6) δ 1.50-1.56 (1H, m), 1.61-1.70 (1H, m), 1.73-1.99 (2H, m), 2.52 (5H, s), 2.93-3.03 (1H, m), 3.17 (1H, s), 3.26-3.32 (3H, m), 3.70 (3H, s), 3.84 (3H, s), 4.28 (2H, s), 4.62 (1H, s), 6.96 (1H, s), 7.00 (1H, d), 7.20 (1H, d), 7.39 (1H, s), 7.44 (1H, d), 7.83-7.88 (1H, m), 7.91 (1H, s). Step 8. Preparation of 2-(5-{2-[3-(2-amino-1,3-benzodiazol-1-yl)azacyclohept-1-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylic acid (INT-B4-8). 2-(5-{2-[3-(2-amino-1,3-benzodiazol-1-yl)azacyclohept-1-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylic acid methyl ester (480 mg, 0.95 mmol, 1 eq.) and LiOH (45 mg, 1.91 mmol, 2 eq.) were dissolved in tetrahydrofuran (6 mL) and H 2O (1.5 mL) was stirred at room temperature for 1 h. The mixture was concentrated under reduced pressure, and the residue was purified by reverse-phase flash chromatography using 10% to 30% acetonitrile/water elution to give 125 mg of 2-(5-{2-[3-(2-amino-1,3-benzodiazol-1-yl)azinecyclohept-1-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylic acid (INT-B4-8) (27%) as a white solid. LCMS: m/z (ESI), [M+H] +=490.15. Step 9. Preparation of 15,21-dimethyl-23-oxa-2,9,11,16,20,21,26-heptaazahexacyclo[24.4.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontria-3,5,7,9,13(32),14,16,18(22),19-nonan-12-one (Example B4). 2-(5-{2-[3-(2-amino-1,3-benzodiazol-1-yl)azacyclohept-1-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylic acid (120 mg, 0.25 mmol, 1 equivalent), N,N-Diisopropylethylamine (95 mg, 0.74 mmol, 3 equiv.) and N,N,N,N-Tetramethyl- OA mixture of -(7-azabenzotriazol-1-yl)uronium hexafluorophosphate (140 mg, 0.37 mmol, 1.5 equiv.) in 1,4-dioxane (6 mL) was stirred at room temperature under nitrogen atmosphere for 1 h. The mixture was concentrated under reduced pressure. The residue was purified by Prep-HPLC using a HPLC-HPLC column containing 10 mmol/L NH4HCO 3The product was purified by using water and acetonitrile as the mobile phase. The fractions containing the desired compound were evaporated to give 60.9 mg of 15,21-dimethyl-23-oxa-2,9,11,16,20,21,26-heptaazahexacyclo[24.4.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontac-3,5,7,9,13(32),14,16,18(22),19-nonan-12-one (Example B4) as a white solid (52%). LCMS: m/z (ESI), [M+H] +=472.25. 1H NMR (DMSO-d 6, 400 MHz) δ 1.63-2.02 (6H, m), 2.57 (3H, s), 2.62-2.76 (1H, m), 2.79-2.85 (1H, m), 2.88-3.01 (3H, m), 3.10-3.17 (1H, m), 3.76 (3H, s), 4.04 (1H, br s), 4.38 (1H, br s), 5.15 (1H, br s), 7.19-7.30 (2H, m), 7.52-7.59 (2H, m), 7.68 (1H, d), 7.91 (1H, s), 8.62 (1H, s), 12.72 (1H, s). Example B6 11-Cyclopropyl-26-methyl-8-(2,2,2-trifluoroethyl)-11H,51H-11-oxa-4,8-diazo-5(2,1)-benzo[d]imidazole-2(2,4)-pyridine-1(4,5)-pyrazolocycloundecane-3-one Step 1. Preparation of methyl 2-chloro-6-(2-cyclopropyl-3-oxo-1-((2-(trimethylsilyl)ethoxy)methyl)-2,3-dihydro-1H-pyrazol-4-yl)isonicotinate (INT-B6-1). To a stirred mixture of 2-cyclopropyl-4-iodo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-3-one (2.3 g, 6.04 mmol, 1 eq.) and freshly prepared (6-chloro-4-(methoxycarbonyl)pyridin-2-yl)boronic acid (M1-3, 9.0 g crude material, about 30 mmol, about 5 eq.) in toluene (20 mL) and water (5 mL) was added Pd at room temperature under nitrogen atmosphere.2(dba) 3(1.1 g, 1.21 mmol, 0.2 equiv.), di(1-adamantyl)-n-butylphosphine (0.1 g, 1.21 mmol, 0.2 equiv.) and Cs 2CO 3(4.9 g, 15.12 mmol, 2.5 equiv.). The resulting mixture was stirred at 80 °C for 2 h and cooled to room temperature. The reaction was quenched with water (300 mL), and the mixture was extracted with ethyl acetate (3×200 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by silica gel column chromatography using petroleum ether/ethyl acetate (1/1) to give 1.8 g of methyl 2-chloro-6-(2-cyclopropyl-3-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-4-yl)pyridine-4-carboxylate (INT-B6-1) (70%) as a yellow oil. 1H NMR (DMSO-d 6, 400 MHz) δ -0.05 (9H, s), 0.78-0.83 (2H, m), 0.87-0.90 (4H, m), 3.30 (2H, d), 3.94-3.98 (3H, m), 2.05-2.10 (1H, m), 5.41 (2H, s), 7.55-7.51 (1H, m), 8.66-8.70 (2H, m). Step 2. Preparation of 2-(2-cyclopropyl-3-oxo-1-((2-(trimethylsilyl)ethoxy)methyl)-2,3-dihydro-1H-pyrazol-4-yl)-6-methylisonicotinate (INT-B6-2). PCy was added to a stirred mixture of methyl 2-chloro-6-(2-cyclopropyl-3-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-4-yl)pyridine-4-carboxylate (1.8 g, 4.24 mmol, 1 eq.) and trimethyl-1,3,5,2,4,6-trioxatriborinane (3.2 g, 25.47 mmol, 6.0 eq.) in 1,4-dioxane (25 mL) at room temperature under nitrogen atmosphere.3.HBF 4(0.5 g, 1.27 mmol, 0.3 eq), Pd(dppf)Cl 2CH 2Cl 2(0.6 g, 0.84 mmol, 0.2 eq.) and K 2CO 3(1.1 g, 8.49 mmol, 2.0 equiv.). The resulting mixture was stirred at 100 °C overnight and cooled to room temperature. The reaction was quenched with water (50 mL), and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×200 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column using petroleum ether/ethyl acetate (1/1) to give 1.3 g of methyl 2-(2-cyclopropyl-3-oxo-1-((2-(trimethylsilyl)ethoxy)methyl)-2,3-dihydro-1H-pyrazol-4-yl)-6-methylisonicotinate (INT-B6-2) (75%) as a brown solid. 1H NMR (DMSO-d 6, 400 MHz) δ -0.05 (9H, s), 0.80-0.84 (2H, m), 1.00-1.05 (4H, m), 2.53 (3H, s), 2.74-2.80 (1H, m), 3.45-3.50 (2H, m), 3.89 (3H, s), 5.38 (2H, s), 7.45 (1H, d), 8.55 (1H, d), 8.59 (1H, s). Step 3. Preparation of methyl 2-(1-cyclopropyl-5-hydroxypyrazol-4-yl)-6-methylpyridine-4-carboxylate (INT-B6-3). A mixture of methyl 2-(2-cyclopropyl-3-oxo-1-{[2-(trimethylsilyl)ethoxy]methyl}pyrazol-4-yl)-6-methylpyridine-4-carboxylate (1.3 g, 3.22 mmol, 1 eq.) and HCl in 1,4-dioxane (10 mL) was stirred at room temperature for 2 h under nitrogen atmosphere and concentrated under vacuum. The residue was washed with CH2Cl 2(20 mL) and saturated NaHCO 3(aqueous solution) (50 mL). The resulting mixture was extracted with dichloromethane (3×100 mL). The combined organic layers were washed with brine (3×50 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was loaded on a silica gel column and eluted with dichloromethane/methanol (10/1) to give 844 mg of methyl 2-(1-cyclopropyl-5-hydroxypyrazol-4-yl)-6-methylpyridine-4-carboxylate (INT-B6-3) (95%) as a brown solid. 1H NMR (DMSO-d 6, 400 MHz) δ 0.88-1.00 (4H, m), 1.24 (1H, s), 2.57 (3H, s), 3.91 (3H, s), 7.33 (1H, s), 7.91 (2H, s). Step 4. Preparation of 2-(1-cyclopropyl-5-(2-((2-((2-nitrophenyl)amino)ethyl)(2,2,2-trifluoroethyl)amino)ethoxy)-1H-pyrazol-4-yl)-6-methylisonicotinate (INT-B6-4). To a stirred mixture of methyl 2-(1-cyclopropyl-5-hydroxypyrazol-4-yl)-6-methylpyridine-4-carboxylate (142 mg, 0.52 mmol, 1.0 equiv) and triphenylphosphine (409 mg, 1.56 mmol, 3.0 equiv) in tetrahydrofuran (10 mL) was added diisopropyl azodicarboxylate (315 mg, 1.56 mmol, 3.0 equiv) and 2-({2-[(2-nitrophenyl)amino]ethyl}(2,2,2-trifluoroethyl)amino)ethanol (INT-A1-2, 400 mg, 1.30 mmol, 2.5 equiv) at 0 °C under nitrogen atmosphere. The resulting mixture was stirred for 2 h. The reaction was quenched with water (50 mL) and the mixture was extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×50 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. Pass the residue through a silica gel column using CH 2Cl 2/methanol (10/1) for purification to obtain 540 mg of 2-(1-cyclopropyl-5-(2-((2-((2-nitrophenyl)amino)ethyl)(2,2,2-trifluoroethyl)amino)ethoxy)-1H-pyrazol-4-yl)-6-methylisonicotinate (INT-B6-4) (57%) as a yellow oil. 1H NMR (DMSO-d 6, 400 MHz) δ 0.91-0.99 (2H, m), 1.02-1.11 (2H, m), 2.53 (3H, s), 3.01-3.07 (2H, m), 3.39-3.43 (4H, m), 3.51-3.67 (3H, m), 3.88 (3H, s), 4.32 (2H, t), 6.61-6.66 (1H, m), 7.02-7.08 (1H. m), 7.38-7.54 (2H, m), 7.83-7.78 (2H, m), 8.03-8.07 (1H, m), 8.22 (1H, t). Step 5. Preparation of methyl 2-{5-[2-({2-[(2-aminophenyl)amino]ethyl}(2,2,2-trifluoroethyl)amino)ethoxy]-1-cyclopropylpyrazol-4-yl}-6-methylpyridine-4-carboxylate (INT-B6-5). Methyl 2-{1-cyclopropyl-5-[2-({2-[(2-nitrophenyl)amino]ethyl}(2,2,2-trifluoroethyl)amino)ethoxy]pyrazol-4-yl}-6-methylpyridine-4-carboxylate (500 mg, 0.88 mmol, 1 eq.) and Zn (464 mg, 7.11 mmol, 8 eq.) were added to tetrahydrofuran (10 mL) and H 2Add NH O (2.5 mL) to the stirring mixture4Cl (475 mg, 8.89 mmol, 10 equiv). The mixture was stirred at the temperature for 2 h. The reaction was quenched with water (50 mL), and the mixture was extracted with dichloromethane (3×50 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. Pass the residue through a silica gel column using CH 2Cl 2/methanol (5/1) for purification to obtain 190 mg of 2-{5-[2-({2-[(2-aminophenyl)amino]ethyl}(2,2,2-trifluoroethyl)amino)ethoxy]-1-cyclopropylpyrazol-4-yl}-6-methylpyridine-4-carboxylic acid methyl ester (INT-B6-5) (40%) as a yellow oil. 1H NMR (DMSO-d 6, 400 MHz) δ 0.92-1.01 (2H, m), 1.03-1.07 (2H, m), 2.55 (3H, s), 3.00 (2H, t), 3.14 (2H, s), 3.48-3.52 (2H, m), 3.58-3.63 (1H, m), 3.89 (3H, s), 3.98-4.04 (1H, m), 4.29-4.31 (2H, m), 4.40 (2H, s), 6.37-6.49 (3H, m), 6.48-6.54 (1H, m), 7.49 (1H, d), 7.81-7.91 (2H, m). Step 6. Preparation of methyl 2-[5-(2-{[2-(2-amino-1,3-benzodiazol-1-yl)ethyl](2,2,2-trifluoroethyl)amino}ethoxy)-1-cyclopropylpyrazol-4-yl]-6-methylpyridine-4-carboxylate (INT-B6-6). A mixture of methyl 2-{5-[2-({2-[(2-aminophenyl)amino]ethyl}(2,2,2-trifluoroethyl)amino)ethoxy]-1-cyclopropylpyrazol-4-yl}-6-methylpyridine-4-carboxylate (170 mg, 0.31 mmol, 1 eq) and BrCN (37 mg, 0.35 mmol, 1.1 eq) in ethanol (5 mL) was stirred under nitrogen atmosphere at room temperature for 4 h. The mixture was concentrated in vacuo. The residue was subjected to Prep-TLC using CH 2Cl 2/methanol (10/1) for purification to obtain 125 mg of 2-[5-(2-{[2-(2-amino-1,3-benzodiazol-1-yl)ethyl](2,2,2-trifluoroethyl)amino}ethoxy)-1-cyclopropylpyrazol-4-yl]-6-methylpyridine-4-carboxylic acid methyl ester (INT-B6-6) (70%) as a brown oil. 1H NMR (DMSO-d 6, 400 MHz) δ 0.92-0.98 (2H, m), 1.03-1.07 (2H, m), 2.54 (3H, s), 2.97-3.07 (2H, m), 3.14-3.24 (2H, m), 3.50-3.64 (3H, m), 3.86 (3H, s), 4.08 (2H, t), 4.25 (2H, t), 6.47 (2H, s), 6.78-6.80 (1H, m), 6.87-6.91 (1H, m), 7.03-7.17 (2H, m), 7.47 (1H, d), 7.85 (2H, d). Step 7. Preparation of 2-[5-(2-{[2-(2-amino-1,3-benzodiazol-1-yl)ethyl](2,2,2-trifluoroethyl)amino}ethoxy)-1-cyclopropylpyrazol-4-yl]-6-methylpyridine-4-carboxylic acid (INT-B6-7). 2-[5-(2-{[2-(2-amino-1,3-benzodiazol-1-yl)ethyl](2,2,2-trifluoroethyl)amino}ethoxy)-1-cyclopropylpyrazol-4-yl]-6-methylpyridine-4-carboxylic acid methyl ester (120 mg, 0.22 mmol, 1 eq) and LiOH (10 mg, 0.43 mmol, 2 eq) were dissolved in tetrahydrofuran (4 mL) and H 2The mixture in 4% ethyl acetate (1 mL) was stirred at room temperature under nitrogen atmosphere for 1 h. The mixture was concentrated in vacuo. The residue was purified by reverse flash chromatography using water and acetonitrile as mobile phase. The fractions containing the desired compound were evaporated to dryness to give 90 mg of 2-[5-(2-{[2-(2-amino-1,3-benzodiazol-1-yl)ethyl](2,2,2-trifluoroethyl)amino}ethoxy)-1-cyclopropylpyrazol-4-yl]-6-methylpyridine-4-carboxylic acid (INT-B6-7) (76%) as a brown solid. LCMS: m/z (ESI), [M+H] += 544.30. Step 8. Preparation of 5-cyclopropyl-26-methyl-10-(2,2,2-trifluoroethyl)-7-oxa-4,5,10,13,20,22,27-heptaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octacosa-1(27),2(6),3,14,16,18,20,24(28),25-nonan-23-one (Example B6). 2-[5-(2-{[2-(2-amino-1,3-benzodiazol-1-yl)ethyl](2,2,2-trifluoroethyl)amino}ethoxy)-1-cyclopropylpyrazol-4-yl]-6-methylpyridine-4-carboxylic acid (80 mg, 0.14 mmol, 1 equivalent) and N,N,N,N-Tetramethyl- O-(7-Azabenzotriazol-1-yl)urea hexafluorophosphate (83 mg, 0.22 mmol, 1.5 equivalents) and 1,4-dioxane (3 mL) were added to the stirred mixture.N,N-Diisopropylethylamine (57 mg, 0.44 mmol, 3.0 equiv.). The reaction mixture was stirred at the temperature for 2 h and concentrated under vacuum. The residue was purified by Prep-HPLC using a HPLC-HPLC system containing 10 mmol/L NH4HCO 3and 0.1% NH 4OH water and acetonitrile as mobile phase for purification to obtain 40 mg of 5-cyclopropyl-26-methyl-10-(2,2,2-trifluoroethyl)-7-oxa-4,5,10,13,20, 22,27-heptaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octacosa-1(27),2(6),3,14,16,18,20,24(28),25-nonan-23-one (Example B6) (52%) as a white solid. LCMS: m/z (ESI), [M+H] +=526.20. 1H NMR (DMSO-d 6, 400 MHz) δ 0.98-1.08 (2H, m), 1.04-1.15 (2H, m), 2.48-2.54 (1H, m), 2.57 (3H, s), 3.14-3.23 (2H, m), 3.33-3.38 (1H, m), 3.56-3.73 (3H, m), 4.31 (2H, t), 4.34-4.42 (2H, m), 7.20-7.35 (2H, m), 7.51-7.59 (2H, m), 7.72-7.79 (1H, m), 7.85 (1H, s), 8.55 (1H, d), 12.73 (1H, s). Example B7A and Example B7B 15,21-Dimethyl-23,29-dioxa-2,9,11,16,20,21,26-heptaazahexacyclo[24.4.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontidine-3,5,7,9,13(32),14,16,18(22),19-nonan-12-one, isomer 1 (Example B7A) and isomer 2 (Example B7B) Step 1. Preparation of tert-butyl 6-[(2-nitrophenyl)amino]-1,4-oxazinecycloheptane-4-carboxylate (INT-B7-1). Add o-fluoronitrobenzene (1 g, 7.09 mmol, 1 equivalent), K 2CO 3A mixture of tert-butyl 6-amino-1,4-oxazinecycloheptane-4-carboxylate (1.69 g, 7.79 mmol, 1.1 equiv.) in acetonitrile (20 mL) was stirred at 100 °C for 12 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (200 mL), and the mixture was extracted with ethyl acetate (3×200 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column using petroleum ether/ethyl acetate (3/1) to give 1.9 g of tert-butyl 6-[(2-nitrophenyl)amino]-1,4-oxazolidinone-4-carboxylate (INT-B7-1) (79%) as a yellow oil. LCMS: m/z (ESI), [M+H] +=338.10. 1H NMR (DMSO-d 6, 400 MHz) δ 1.03 (6H, s), 1.33 (3H, s), 3.29 (2H, d), 3.58-3.79 (2H, m), 3.81-3.96 (2H, m), 4.02-4.13 (2H, m), 6.72 (1H, q), 7.17 (1H, d), 7.55 (1H, q), 8.05-8.13 (1H, m), 8.36-8.46 (1H, m). Step 2. N- Preparation of (2-nitrophenyl)-1,4-oxazacycloheptane-6-amine (INT-B7-2). A mixture of tert-butyl 6-[(2-nitrophenyl)amino]-1,4-oxazacycloheptane-4-carboxylate (2 g, 5.928 mmol, 1 eq.) in trifluoroacetic acid (10 mL) and dichloromethane (40 mL) was stirred at room temperature overnight under a nitrogen atmosphere. The mixture was concentrated under reduced pressure. The residue was treated with saturated NaHCO 3(aqueous solution) (100 mL) and extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×200 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure to obtain 1.4 g of yellow oil.N-(2-Nitrophenyl)-1,4-oxazolidinone-6-heptylamine (INT-B7-2) (99%). This material was used in the next reaction without further purification. LCMS: m/z (ESI), [M+H] +=238.10. Step 3. 4-{2-[(tert-butyldimethylsilyl)oxy]ethyl}- N-(2-nitrophenyl)-1,4-oxazolidinone-6-heptylamine (INT-B7-3).N-(2-nitrophenyl)-1,4-oxazinecycloheptyl-6-amine (1.4 g, 5.90 mmol, 1 equivalent), (2-bromoethoxy)(tert-butyl)dimethylsilane (2.12 g, 8.85 mmol, 1.5 equivalent) and K 2CO 3(2.45 g, 17.70 mmol, 3 equiv.) in acetonitrile (20 mL) was stirred at 60 °C overnight under nitrogen atmosphere and cooled to room temperature. The reaction was quenched with water (100 mL), and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×200 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column using petroleum ether/ethyl acetate (1/1) to give 1.8 g of 4-{2-[(tert-butyldimethylsilyl)oxy]ethyl}- N-(2-Nitrophenyl)-1,4-oxazolidinone-6-heptylamine (INT-B7-3) (77%). LCMS: m/z (ESI), [M+H] +=396.15. Step 4. Preparation of 2-{6-[(2-nitrophenyl)amino]-1,4-oxazolidinylcyclohept-4-yl}ethanol (INT-B7-4). 4-{2-[(tert-butyldimethylsilyl)oxy]ethyl}- NA mixture of -(2-nitrophenyl)-1,4-oxazolidinone cycloheptyl-6-amine (1.7 g, 4.29 mmol, 1 equiv) and tetrabutylammonium fluoride (2.25 g, 8.59 mmol, 2 equiv) in tetrahydrofuran (20 mL) was stirred at room temperature under nitrogen atmosphere for 2 h. The reaction was quenched with water (100 mL), and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×200 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. Pass the residue through C18 FLASH using 10 mmol/L NH4HCO 3and 0.1% NH 3 .H 2Purification was performed using water with 0.05% iodine and acetonitrile as the mobile phase. The fractions containing the desired compound were evaporated to dryness to give 1 g of 2-{6-[(2-nitrophenyl)amino]-1,4-oxazolidinylcyclohept-4-yl}ethanol (INT-B7-4) (82%) as an orange oil. LCMS: m/z (ESI), [M+H] +=282.10. 1H NMR (DMSO-d 6, 400 MHz) δ 2.64-2.71 (3H, m), 2.81-2.94 (2H, m), 3.03-3.18 (1H, m), 3.45-3.59 (2H, m), 3.59-3.75 (3H, m), 3.95-3.99 (1H, m), 4.04-4.13 (1H, m), 4.41 (1H, t), 6.66-7.00 (1H, m), 7.01-7.02 (1H, m), 7.49-7.53 (1H, m), 8.07-8.10 (1H, m), 8.79 (1H, d) Step 5. Preparation of methyl 2-methyl-6-[1-methyl-5-(2-{6-[(2-nitrophenyl)amino]-1,4-oxazolidin-4-yl}ethoxy)pyrazol-4-yl]pyridine-4-carboxylate (INT-B7-5). To a stirred mixture of 2-{6-[(2-nitrophenyl)amino]-1,4-oxazolidinecycloheptan-4-yl}ethanol (1.00 g, 3.55 mmol, 1.2 equiv) and triphenylphosphine (2.33 g, 8.89 mmol, 3 equiv) in tetrahydrofuran (20 mL) was added diisopropyl azodicarboxylate (1.80 g, 8.89 mmol, 3 equiv) and methyl 2-(5-hydroxy-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylate (0.73 g, 2.96 mmol, 1.00 equiv) in THF (10 mL) at 0°C under nitrogen atmosphere. The mixture was stirred at 0°C for 2 h. The reaction was quenched with water (100 mL), and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×200 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column using petroleum ether/ethyl acetate (1/1) to give 0.9 g of 2-methyl-6-[1-methyl-5-(2-{6-[(2-nitrophenyl)amino]-1,4-oxazolidin-4-yl}ethoxy)pyrazol-4-yl]pyridine-4-carboxylic acid methyl ester (INT-B7-5) (59%) as an orange oil. LCMS: m/z (ESI), [M+H] +=511.20. 1H NMR (DMSO-d 6, 400 MHz) δ 2.52 (3H, s), 2.91-2.95 (2H, m), 3.05 (1H, d), 3.10 (1H, q), 3.17 (2H, d), 3.67 (2H, d), 3.70 (3H, s), 3.87 (3H, s), 3.90-3.95 (1H, m), 4.05-4.12 (2H, m), 4.20-4.34 (2H, m), 6.64-6.68 (1H, m), 6.97 (1H, d), 7.42 (1H, d), 7.45-7.50 (1H, m), 7.81 (1H, d), 7.88 (1H, s), 8.02-8.05 (1H, m), 8.67 (1H, d). Step 6. Preparation of methyl 2-[5-(2-{6-[(2-aminophenyl)amino]-1,4-oxazacycloheptan-4-yl}ethoxy)-1-methylpyrazol-4-yl]-6-methylpyridine-4-carboxylate (INT-B7-6). To a stirred mixture of methyl 2-methyl-6-[1-methyl-5-(2-{6-[(2-nitrophenyl)amino]-1,4-oxazacycloheptan-4-yl}ethoxy)pyrazol-4-yl]pyridine-4-carboxylate (540 mg, 1.06 mmol, 1 eq.) and ranyl nickel (725 mg, 8.46 mmol, 8 eq.) in methanol (5 mL) at 0 °C was added hydrazine hydrate (79 mg, 1.59 mmol, 1.5 eq.). The mixture was stirred at 0 °C for 2 h under nitrogen atmosphere. The resulting mixture was filtered. The filter cake was washed with methanol (3×50 mL), and the resulting solution was concentrated under reduced pressure to give 480 mg of methyl 2-[5-(2-{6-[(2-aminophenyl)amino]-1,4-oxazolidin-4-yl}ethoxy)-1-methylpyrazol-4-yl]-6-methylpyridine-4-carboxylate (INT-B7-6) as a brown solid. This material was used in the next reaction without further purification. LCMS: m/z (ESI), [M+H] += 481.25. Step 7. Preparation of methyl 2-(5-{2-[6-(2-amino-1,3-benzodiazol-1-yl)-1,4-oxazacyclohept-4-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylate (INT-B7-7). A mixture of methyl 2-[5-(2-{6-[(2-aminophenyl)amino]-1,4-oxazacyclohept-4-yl}ethoxy)-1-methylpyrazol-4-yl]-6-methylpyridine-4-carboxylate (500 mg, 1.04 mmol, 1 eq.) and BrCN (132 mg, 1.25 mmol, 1.2 eq.) in dichloromethane (5 mL) was stirred at room temperature overnight under nitrogen atmosphere. The reaction was quenched with water (80 mL), and the mixture was extracted with ethyl acetate (3×60 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using petroleum ether/ethyl acetate (1/1) to give 400 mg of methyl 2-(5-{2-[6-(2-amino-1,3-benzodiazol-1-yl)-1,4-oxazolidin-4-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylate (INT-B7-7) (76%) as an orange oil. LCMS: m/z (ESI), [M+H] +=506.30. Step 8. Preparation of 2-(5-{2-[6-(2-amino-1,3-benzodiazol-1-yl)-1,4-oxazacyclohept-4-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylic acid (INT-B7-8). 2-(5-{2-[6-(2-amino-1,3-benzodiazol-1-yl)-1,4-oxazacyclohept-4-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylic acid methyl ester (200 mg, 0.39 mmol, 1 equivalent) and LiOH.H 2A mixture of O (25 mg, 0.59 mmol, 1.5 equiv.) in tetrahydrofuran (4 mL) and water (1 mL) was stirred at room temperature for 2 h under a nitrogen atmosphere and concentrated under reduced pressure. The residue was passed through a C18 FLASH using a 10 mmol/L NH4HCO 3and 0.1% NH 3 .H 2Purification was performed using water with 0.04% iodine and acetonitrile as the mobile phase. The fractions containing the desired compound were evaporated to dryness to give 180 mg of 2-(5-{2-[6-(2-amino-1,3-benzodiazol-1-yl)-1,4-oxazol-4-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylic acid (INT-B7-8) (92%) as a pink solid. LCMS: m/z (ESI), [M+H] +=492.30. Step 9. 15,21-Dimethyl-23,29-dioxa-2,9,11,16,20,21, 26-heptaazahexacyclic [24.4.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18, 22}]32-3,5,7,9,13(3 2),14,1Preparation of 6,18(22),19-nonan-12-one (Example B7). 2-(5-{2-[6-(2-amino-1,3-benzodiazol-1-yl)-1,4-oxazolidinone-4-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylic acid (160 mg, 0.33 mmol, 1 equivalent), HATU (186 mg, 0.49 mmol, 1.5 equivalents) and N,N- A mixture of diisopropylethylamine (126 mg, 0.98 mmol, 3 equiv.) in 1,4-dioxane (5 mL) was stirred at room temperature overnight under nitrogen atmosphere. The reaction was quenched with water (30 mL), and the mixture was extracted with ethyl acetate (3×20 mL). The combined organic layers were washed with brine (3×30 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by trituration with methanol (50 mL) to give 8.3 mg of 15,21-dimethyl-23,29-dioxa-2,9,11,16,20,21,26-heptaazahexacyclo[24.4.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontac-3,5,7,9,13(32),14,16,18(22),19-nonan-12-one (Example B7) as a white solid (5%). LCMS: m/z (ESI), [M+H] += 474.15. 1H NMR (DMSO-d 6, 400 MHz) δ 2.57 (3H, s), 2.82-2.91 (3H, m), 2.93-3.01 (1H, m), 3.24-3.44 (2H, m), 3.75 (3H, s), 3.72-3.78 (1H, m), 3.96 (1H, br s), 4.03-4.26 (2H, m), 4.59 (2H, d), 5.45 (1H, br s), 7.20-7.33 (2H, m), 7.52-7.58 (2H, m), 7.69 (1H, d), 7.92 (1H, s), 8.61 (1H, s), 12.67 (1H, s). Step 10. Preparation of 15,21-dimethyl-23,29-dioxa-2,9,11,16,20,21, 26-heptaazahexacyclo[24.4.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18, 22}]triacontria-3,5,7,9,13(32),14,16,18(22),19-nonan-12-one, Isomer 1 (Example B7A) and Isomer 2 (Example B7B). A mixture of 15,21-dimethyl-23,29-dioxa-2,9,11,16,20,21,26-heptaazahexacyclo[24.4.1.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontac-3,5,7,9,13(32),14,16,18(22),19-nonan-12-one (Example B7, 80 mg) was chromatographed by Prep-chiral-HPLC using a CHIRALPAK IG column (2*25 cm, 5 μm) with hexane and EtOH/CH containing 0.2% DEA.2Cl 2(1/1) was used as the mobile phase to separate 32 mg of isomer 1 (Example B7A, 40%) and 34 mg of isomer 2 (Example B7B, 42%) as white solids. Isomer 1: LCMS: m/z (ESI), [M+H] +=474.15. 1H NMR (DMSO-d 6, 400 MHz) δ 2.57 (3H, s), 2.82-2.91 (3H, m), 2.93-3.01 (1H, m), 3.24-3.44 (2H, m), 3.75 (3H, s), 3.72-3.78 (1H, m), 3.96 (1H, br s), 4.03-4.26 (2H, m), 4.59 (2H, d), 5.45 (1H, br s), 7.20-7.33 (2H, m), 7.52-7.58 (2H, m), 7.69 (1H, d), 7.92 (1H, s), 8.61 (1H, s), 12.67 (1H, s). Chiral-HPLC, Rt=2.016 min. Isomer 2: LCMS: m/z (ESI), [M+H] +=474.15. 1H NMR (DMSO-d 6, 400 MHz) δ 2.57 (3H, s), 2.82-2.91 (3H, m), 2.93-3.01 (1H, m), 3.24-3.44 (2H, m), 3.75 (3H, s), 3.72-3.78 (1H, m), 3.96 (1H, br s), 4.03-4.26 (2H, m), 4.59 (2H, d), 5.45 (1H, br s), 7.20-7.33 (2H, m), 7.52-7.58 (2H, m), 7.69 (1H, d), 7.92 (1H, s), 8.61 (1H, s), 12.67 (1H, s). Chiral-HPLC, Rt = 2.626 minutes. Example B8A and Example B8B 15,21-Dimethyl-23-oxa-2,9,11,16,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontahedral-3,5,7,9,13,15,17(33),18(22), 19-nonan-12-one, isomer 1 (Example B8A) and isomer 2 (Example B8B) Step 1. Preparation of methyl 2-methyl-6-[1-methyl-5-(2-{1-[(2-nitrophenyl)amino]-3-azabicyclo[3.2.1]oct-3-yl}ethoxy)pyrazol-4-yl]pyridine-4-carboxylate (INT-B8-1). To a stirred mixture of 2-{1-[(2-nitrophenyl)amino]-3-azabicyclo[3.2.1]octan-3-yl}ethanol (INT-A61-4, 380 mg, 1.30 mmol, 1 eq.) and triphenylphosphine (685 mg, 2.61 mmol, 2 eq.) in tetrahydrofuran (10 mL) at 0 °C under nitrogen atmosphere was added methyl 2-(5-hydroxy-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylate (M1, 323 mg, 1.30 mmol, 1 eq.) and diisopropyl azodicarboxylate (528 mg, 2.61 mmol, 2 eq.). The resulting mixture was stirred at 0 °C for 2 h. The reaction was quenched with water (30 mL), and the mixture was washed with CH2Cl 2(3×50 mL) extraction. The combined organic layers were washed with brine (3×75 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by prep-TLC using petroleum ether/ethyl acetate (1/1) to give 460 mg of methyl 2-methyl-6-[1-methyl-5-(2-{1-[(2-nitrophenyl)amino]-3-azabicyclo[3.2.1]octan-3-yl}ethoxy)pyrazol-4-yl]pyridine-4-carboxylate (INT-B8-1) (67%) as a white solid. LCMS: m/z (ESI), [M+H] +=521.15. Step 2. Preparation of methyl 2-[5-(2-{1-[(2-aminophenyl)amino]-3-azabicyclo[3.2.1]oct-3-yl}ethoxy)-1-methylpyrazol-4-yl]-6-methylpyridine-4-carboxylate (INT-B8-2). To a stirred mixture of methyl 2-methyl-6-[1-methyl-5-(2-{1-[(2-nitrophenyl)amino]-3-azabicyclo[3.2.1]oct-3-yl}ethoxy)pyrazol-4-yl]pyridine-4-carboxylate (500 mg, 0.96 mmol, 1 eq.) and ranyl nickel (80 mg) in methanol (10 mL) was added hydrazine hydrate (92 mg, 2.88 mmol, 3 eq.) at room temperature under nitrogen atmosphere. The resulting mixture was stirred at room temperature for 1 h. The resulting mixture was filtered and the filter cake was washed with methanol (3×20 mL). The resulting solution was concentrated under reduced pressure. The mixture was purified by prep-TLC using dichloromethane/methanol (20/1) to give 400 mg of 2-[5-(2-{1-[(2-aminophenyl)amino]-3-azabicyclo[3.2.1]octan-3-yl}ethoxy)-1-methylpyrazol-4-yl]-6-methylpyridine-4-carboxylic acid methyl ester (INT-B8-2) (85%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=491.15. Step 3. Preparation of methyl 2-(5-{2-[1-(2-amino-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylate (INT-B8-3). To a stirred mixture of methyl 2-[5-(2-{1-[(2-aminophenyl)amino]-3-azabicyclo[3.2.1]oct-3-yl}ethoxy)-1-methylpyrazol-4-yl]-6-methylpyridine-4-carboxylate (455 mg, 0.93 mmol, 1 eq.) in ethanol (20 mL) was added BrCN (334 mg, 3.15 mmol, 3.4 eq.) at room temperature under nitrogen atmosphere. The resulting mixture was stirred at room temperature for 1 h and concentrated under reduced pressure. The residue was purified by prep-TLC using dichloromethane/methanol (20/1) to give 350 mg of methyl 2-(5-{2-[1-(2-amino-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]octan-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylate (INT-B8-3) (73%) as a brown solid. LCMS: m/z (ESI), [M+H] +=516.15. Step 4. Preparation of methyl 2-(5-{2-[1-(2-amino-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylate, isomer 1 (INT-B8-3A) and isomer 2 (INT-B8-3B). A racemic mixture of methyl 2-(5-{2-[1-(2-amino-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]octan-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylate (350 mg) was purified by Prep-chiral-HPLC using a CHIRALPAK IG chiral column (2×25 cm, 5 μm) using methyl tert-butyl ether and ethanol/CH 2Cl 2(1/1) was used as the mobile phase to separate 90 mg of isomer 1 (INT-B8-3A, 25%) and 80 mg of isomer 2 (INT-B8-3B, 22%) as white solids. Chiral-HPLC: Rt (isomer 1) = 1.312 min, Rt (isomer 2) = 2.047 min. Step 5. Preparation of 2-(5-{2-[1-(2-amino-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylic acid (INT-B8-4A). To a stirred mixture of methyl 2-(5-{2-[1-(2-amino-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylate (INT-B8-3A, 85 mg, 0.16 mmol, 1 equiv.) in tetrahydrofuran (8 mL) at room temperature under nitrogen atmosphere was added LiOH .H 2O (16 mg, 0.66 mmol, 4 equiv) of H 2O (2 mL). The resulting mixture was stirred at 60 °C for 30 minutes, cooled to room temperature, and concentrated under reduced pressure. The residue was purified by reverse flash chromatography using a C18 silica gel column using water and acetonitrile as mobile phases. The fractions containing the desired compound were evaporated to dryness to give 80 mg of 2-(5-{2-[1-(2-amino-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]octan-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylic acid (INT-B8-4A) (96%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=502.20. Step 6. Preparation of 15,21-dimethyl-23-oxa-2,9,11,16,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]tritriaconta-3,5,7,9,13,15,17(33),18(22), 19-nonan-12-one, isomer 1 (Example B8A). N, N-diisopropylethylamine (58 mg, 0.45 mmol, 3 equivalents), 2-(5-{2-[1-(2-amino-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylic acid (INT-B8-4A, 75 mg, 0.15 mmol, 1 equivalent) and N, N, N, N-Tetramethyl- OA mixture of -(7-azabenzotriazol-1-yl)urea hexafluorophosphate (85 mg, 0.22 mmol, 1.5 equiv.) in 1,4-dioxane (7 mL) was stirred at 60 °C for 1 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (20 mL), and the mixture was washed with CH2Cl 2(3×30 mL) extraction. The combined organic layers were washed with brine (3×30 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC and eluted with dichloromethane/methanol (25/1) to give 57.4 mg of 15,21-dimethyl-23-oxa-2,9,11,16,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontac-3,5,7,9,13,15,17(33), 18(22),19-nonan-12-one (isomer 1, Example B8A) (79%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=484.30. 1H NMR (DMSO-d 6, 400 MHz) δ 1.59-1.80 (3H, m), 2.02 (1H, d), 2.38-2.50 (3H, m), 2.56 (3H, s), 2.63-2.82 (3H, m), 3.22-3.45 (2H, m), 3.74 (3H, s), 3.77-3.83 (1H, m), 4.56 (1H, t), 4.71 (1H, d), 7.16-7.25 (2H, m), 7.55-7.63 (2H, m), 7.73-7.84 (1H, m), 7.94 (1H, s), 8.44 (1H, s), 12.87 (1H, s). SFC-HPLC, Rt = 1.282 min. Step 7. Preparation of 2-(5-{2-[1-(2-amino-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylic acid (INT-B8-4B). To a stirred mixture of methyl 2-(5-{2-[1-(2-amino-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylate (INT-B8-3B, 75 mg, 0.15 mmol, 1 equiv.) in tetrahydrofuran (8 mL) at room temperature under nitrogen atmosphere was added LiOH .H 2O (14 mg, 0.58 mmol, 4 equiv) of H 2O (2 mL). The resulting mixture was stirred at 60 °C for 30 min. The mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by reverse flash chromatography using a C18 silica gel column using water and acetonitrile as mobile phases. The fractions containing the desired compound were evaporated to dryness to give 70 mg of 2-(5-{2-[1-(2-amino-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]octan-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylic acid (INT-B8-4B) (96%) as an off-white solid. LCMS: m/z (ESI), [M+H] +=502.20. Step 8. Preparation of 15,21-dimethyl-23-oxa-2,9,11,16,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]tritriaconta-3,5,7,9,13,15,17(33),18(22), 19-nonan-12-one, isomer 2 (Example B8B). N, N-diisopropylethylamine (50 mg, 0.39 mmol, 3 equivalents), 2-(5-{2-[1-(2-amino-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylic acid (INT-B8-4B, 65 mg, 0.13 mmol, 1 equivalent) and N, N, N, N-Tetramethyl- OA mixture of -(7-azabenzotriazol-1-yl)urea hexafluorophosphate (74 mg, 0.20 mmol, 1.5 equiv) in 1,4-dioxane (6 mL) was stirred at 60 °C for 1 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (20 mL), and the mixture was washed with CH2Cl 2(3×30 mL) extraction. The combined organic layers were washed with brine (3×30 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC and eluted with dichloromethane/methanol (25/1) to give 46.5 mg of 15,21-dimethyl-23-oxa-2,9,11,16,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontac-3,5,7,9,13,15,17(33), 18(22),19-nonan-12-one, isomer 2 (Example B8B) (74%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=484.25. 1H NMR (DMSO-d 6, 400 MHz) δ 1.59-1.80 (3H, m), 2.02 (1H, d), 2.38-2.50 (3H, m), 2.56 (3H, s), 2.63-2.82 (3H, m), 3.22-3.45 (2H, m), 3.74 (3H, s), 3.77-3.83 (1H, m), 4.56 (1H, t), 4.71 (1H, d), 7.16-7.25 (2H, m), 7.55-7.63 (2H, m), 7.73-7.84 (1H, m), 7.94 (1H, s), 8.44 (1H, s), 12.87 (1H, s). SFC-HPLC, Rt = 1.060 minutes. Example B9A and Example B9B 15,21-Dimethyl-5-(4-methylpiperazin-1-yl)-23-oxa-2,9,11,16, 20,21,26-heptaazaheptacyclic [24.4.1.1^{1,28}.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]tritriacontahedral-3,5,7,9,13,15,17(33),18(22), 19-nonan-12-one, isomer 1 (Example B9A) and isomer 2 (Example B9B). Step 1. Preparation of methyl 2-[5-(2-{1-[(5-bromo-2-nitrophenyl)amino]-3-azabicyclo[3.2.1]oct-3-yl}ethoxy)-1-methylpyrazol-4-yl]-6-methylpyridine-4-carboxylate (INT-B9-1). To a stirred mixture of methyl 2-(5-hydroxy-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylate (M1, 1.40 g, 5.67 mmol, 1.05 equiv) and triphenylphosphine (3.97 g, 15.12 mmol, 2.8 equiv) in tetrahydrofuran (30 mL) at 0 °C under nitrogen atmosphere was added diisopropyl azodicarboxylate (2.73 g, 13.50 mmol, 2.5 equiv) followed by 2-{1-[(5-bromo-2-nitrophenyl)amino]-3-azabicyclo[3.2.1]octan-3-yl}ethanol (M4-11, 2 g, 5.40 mmol, 1 equiv). After stirring at 0 °C for 3 h, the reaction was quenched with water (400 mL), and the mixture was extracted with ethyl acetate (3×300 mL). The combined organic layers were washed with brine (3×300 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by silica gel column eluting with dichloromethane/methanol (30/1) to give 2 g of 2-[5-(2-{1-[(5-bromo-2-nitrophenyl)amino]-3-azabicyclo[3.2.1]octan-3-yl}ethoxy)-1-methylpyrazol-4-yl]-6-methylpyridine-4-carboxylic acid methyl ester (INT-B9-1) (61%) as a yellow oil. LCMS: m/z (ESI), [M+H] +=601.00. Step 2. Preparation of methyl 2-[5-(2-{1-[(2-amino-5-bromophenyl)amino]-3-azabicyclo[3.2.1]oct-3-yl}ethoxy)-1-methylpyrazol-4-yl]-6-methylpyridine-4-carboxylate (INT-B9-2). To a stirred mixture of methyl 2-[5-(2-{1-[(5-bromo-2-nitrophenyl)amino]-3-azabicyclo[3.2.1]oct-3-yl}ethoxy)-1-methylpyrazol-4-yl]-6-methylpyridine-4-carboxylate (2 g, 3.33 mmol, 1 eq.) in methanol (60 mL) at room temperature, add ranyl nickel (457 mg, 5.33 mmol, 1.6 eq.) and NH 2NH 2 .H 2O (835 mg, 16.68 mmol, 5 equiv.). The resulting mixture was stirred at room temperature for 1 h. The reaction was quenched with water (300 mL), and the mixture was extracted with ethyl acetate (3×200 mL). The combined organic layers were washed with brine (3×200 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by silica gel chromatography using dichloromethane/methanol (20/1) to give 1.34 g of methyl 2-[5-(2-{1-[(2-amino-5-bromophenyl)amino]-3-azabicyclo[3.2.1]oct-3-yl}ethoxy)-1-methylpyrazol-4-yl]-6-methylpyridine-4-carboxylate (INT-B9-2) (70%) as a reddish brown solid. LCMS: m/z (ESI), [M+H] += 571.05. Step 3. Preparation of methyl 2-(5-{2-[1-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylate (INT-B9-3). To a stirred mixture of methyl 2-[5-(2-{1-[(2-amino-5-bromophenyl)amino]-3-azabicyclo[3.2.1]oct-3-yl}ethoxy)-1-methylpyrazol-4-yl]-6-methylpyridine-4-carboxylate (500 mg, 0.87 mmol, 1 eq.) in ethanol (8 mL) was added BrCN (139 mg, 1.31 mmol, 1.5 eq.) at room temperature. The resulting mixture was stirred at room temperature for 2 h. The reaction was treated with saturated NaHCO3(100 mL) and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by silica gel chromatography using dichloromethane/methanol (15/1) to give 400 mg of methyl 2-(5-{2-[1-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylate (INT-B9-3) (76.63%) as a reddish brown oil. LCMS: m/z (ESI), [M+H] +=596.10. Step 4. Preparation of methyl 2-(5-{2-[1-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylate, isomer 1 (INT-B9-3A) and isomer 2 (INT-B9-3B). The racemic mixture of methyl 2-(5-{2-[1-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylate (400 mg) was subjected to Prep-chiral-HPLC using a CHIRAL ART cellulose-SB column (5*25 cm, 5 μm) using CO 2and CH 3OH/CH 2Cl 2(2/1) was used as the mobile phase to separate to obtain 150 mg of isomer 1 (INT-B9-3A, 42%) and 150 mg of isomer 2 (INT-B9-3B, 42%) as white solids. SFC-HPLC, Rt (isomer 1) = 1.760 min, Rt (isomer 2) = 1.889 min. Step 5. Preparation of 2-(5-{2-1-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylic acid (isomer 1, INT-B9-4A). To a stirred solution of methyl 2-(5-{2-[1-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]octan-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylate (490 mg, 0.82 mmol, 1 eq.) in THF (8 mL) at room temperature was added LiOH (78 mg, 3.29 mmol, 4 eq.) in H 2O (2 mL). The resulting mixture was stirred at room temperature for 3 h. The mixture was concentrated under reduced pressure, and the residue was purified by reverse flash chromatography using water and acetonitrile as mobile phases to give 410 mg of 2-(5-{2-[1-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]octan-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylic acid (isomer 1, INT-B9-4A) (85%) as a white solid. LCMS: m/z (ESI), [M+H] +=580.05. Step 6. Preparation of 5-bromo-15,21-dimethyl-23-oxa-2,9,11,16,20,21, 26-heptaazaheptacyclic [24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3, 8}.0^{18,22}]triacontria-3,5,7,9,13,15,17(33),18(22), 19-nonan-12-one (isomer 1, INT-B9-5A). To a stirred solution of 2-(5-{2-[1-(2-amino-6-bromo-1,3-benzodiazol-1-yl)-3-azabicyclo[3.2.1]oct-3-yl]ethoxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylic acid (390 mg, 0.67 mmol, 1 eq.) in dioxane (10 mL) was added at room temperature N,N-Diisopropylethylamine (260 mg, 2.01 mmol, 3 equiv.) and N,N,N,N-Tetramethyl- O-(7-Azabenzotriazole-1-yl)uronium hexafluorophosphate (383 mg, 1.01 mmol, 1.5 equiv). The reaction mixture was stirred at 50 °C for 2 h under a nitrogen atmosphere. After cooling to room temperature, the reaction was quenched with water (100 mL), and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The crude product was purified by prep-TLC using dichloromethane/methanol (25/1) to give 350 mg of 5-bromo-15,21-dimethyl-23-oxa-2,9,11,16,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontac-3,5,7,9,13,15,17(33),18(22), 19-nonan-12-one (isomer 1, INT-B9-5A) (92%) as a light yellow solid. LCMS: m/z (ESI), [M+H] +=564.05. Step 7. Preparation of 15,21-dimethyl-5-(4-methylpiperazin-1-yl)-23-oxa-2,9,11,16,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontahedral-3,5,7,9,13,15,17(33), 18(22), 19-nonan-12-one (isomer 1, Example B9A). 5-Bromo-15,21-dimethyl-23-oxa-2,9,11,16,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]tritriacontria-3,5,7,9,13,15,17(33), 18(22),19-nonan-12-one (130 mg, 0.23 mmol, 1 eq.), 1-methylpiperazine (34.7 mg, 0.35 mmol, 1.5 eq.) and BrettPhos Pd G 3To a stirred mixture of 2-nitropropene (62.8 mg, 0.07 mmol, 0.3 equiv) in dioxane (5 mL) was added LiHMDS (2.3 mL, 2.31 mmol, 10 equiv) dropwise. The reaction mixture was stirred at 60 °C under nitrogen atmosphere for 1 h. After cooling to room temperature, the reaction was heated with saturated NH4Cl (30 mL) was quenched, and the mixture was extracted with dichloromethane (3×20 mL). The combined organic layers were washed with brine (3×30 mL) and purified by anhydrous Na 2SO 4The residue was purified by prep-TLC eluting with dichloromethane/methanol (18/1) to give 41.6 mg of 15,21-dimethyl-5-(4-methylpiperazin-1-yl)-23-oxa-2,9,11,16,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontria-3,5,7,9,13,15,17(33),18(22),19-nonan-12-one, Isomer 1 (Example B9A) as a white solid (30%). LCMS: m/z (ESI), [M+H] +=582.40. 1H NMR (DMSO-d 6, 400 MHz) δ 1.52-1.82 (3H, m), 2.03 (1H, d), 2.24 (3H, s), 2.40-2.60 (8H, m), 2.55 (3H, s), 2.64-2.79 (2H, m), 3.12 (4H, q), 3.20-3.30 (1H, m), 3.37-3.45 (1H, m), 3.74 (3H, s), 3.77 (1H, br s), 4.56 (1H, t), 4.70 (1H, d), 6.93 (1H, dd), 7.19 (1H, d), 7.42 (1H, d), 7.57 (1H, d), 7.94 (1H, s), 8.42 (1H, s), 12.69 (1H, s). SFC-HPLC, Rt=3.968 minutes. Using INT-B9-3B as the starting material, the same reaction sequence described above for the synthesis of Example B9A gave 48.4 mg of 15,21-dimethyl-5-(4-methylpiperazin-1-yl)-23-oxa-2,9,11,16,20, 21,26-heptaazaheptacyclic [24.4.1.1^{1,28}.1^{13,17}.0^{2,10}. 0^{3,8}.0^{18,22}]tritriacontac-3,5,7,9,13,15,17(33), 18(22),19-nonan-12-one, isomer 2 (Example B9B) as a white solid. LCMS: m/z (ESI), [M+H] +=582.40. 1H NMR (DMSO-d 6, 400 MHz) δ 1.52-1.82 (3H, m), 2.03 (1H, d), 2.24 (3H, s), 2.40-2.60 (8H, m), 2.55 (3H, s), 2.64-2.79 (2H, m), 3.12 (4H, q), 3.20-3.30 (1H, m), 3.37-3.45 (1H, m), 3.74 (3H, s), 3.77 (1H, br s), 4.56 (1H, t), 4.70 (1H, d), 6.93 (1H, dd), 7.19 (1H, d), 7.42 (1H, d), 7.57 (1H, d), 7.94 (1H, s), 8.42 (1H, s), 12.69 (1H, s). SFC-HPLC, Rt = 3.745 minutes. Example B10A and Example B10B 15,21-Dimethyl-5-(morpholin-4-yl)-23-oxa-2,9,11,16,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}. 0^{18,22}]tritriacontahedral-3,5,7,9,13,15,17(33),18(22),19-nonan-12-one, isomer 1 (Example B10A) and isomer 2 (Example B10B) The title compound was prepared using the same reaction sequence described for the preparation of Examples B9A and B9B. Example B10A: LCMS: m/z (ESI), [M+H]+=569.40. 1H NMR (DMSO-d 6, 400 MHz) δ 1.56-1.81 (3H, m), 2.03 (1H, d), 2.37-2.48 (3H, m), 2.55 (3H, s), 2.57-2.79 (3H, m), 3.04-3.18 (4H, m), 3.19-3.29 (2H, m), 3.37-3.47 (1H, m), 3.74 (3H, s), 3.75-3.83 (4H, m), 4.56 (1H, t), 4.71 (1H, d), 6.94 (1H, dd), 7.21 (1H, d), 7.45 (1H, d), 7.57 (1H, d), 7.94 (1H, s), 8.43 (1H, s), 12.71 (1H, s). SFC-HPLC, Rt=1.549 min Example B10B: LCMS: m/z (ESI), [M+H]+=569.25. 1H NMR (DMSO-d 6, 400 MHz) δ 1.56-1.81 (3H, m), 2.03 (1H, d), 2.37-2.48 (3H, m), 2.55 (3H, s), 2.57-2.79 (3H, m), 3.04-3.18 (4H, m), 3.19-3.29 (2H, m), 3.37-3.47 (1H, m), 3.74 (3H, s), 3.75-3.83 (4H, m), 4.56 (1H, t), 4.71 (1H, d), 6.94 (1H, dd), 7.21 (1H, d), 7.45 (1H, d), 7.57 (1H, d), 7.94 (1H, s), 8.43 (1H, s), 12.71 (1H, s). SFC-HPLC, Rt=1.639 min Example B11 (12 R)-5,12,27-trimethyl-8-oxa-5,14,21,23,28-pentazapentacyclic [23.3.1.0^{2,7}.0^{14,22}.0^{15,20}]twenty-nine-1(29),2,6,15, 17,19,21,25,27-nonene-4,24-dione Step 1. (4 RPreparation of )-4-methyl-5-[(2-nitrophenyl)amino]pentan-1-ol (INT-B11-1). o-Fluoronitrobenzene (1.32 g, 9.386 mmol, 1.1 equivalents), (4 R)-5-amino-4-methylpentan-1-ol (1 g, 8.533 mmol, 1.00 equiv) and K 2CO 3(3.54 g, 25.599 mmol, 3 equiv.) in acetonitrile (20 mL) was stirred at 60 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (800 mL), and the mixture was extracted with dichloromethane (3×600 mL). The combined organic layers were washed with brine (3×500 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue is purified by silica gel chromatography using petroleum ether/ethyl acetate (1/1) to give 1.6 g (4 R)-4-methyl-5-[(2-nitrophenyl)amino]pentan-1-ol (INT-B11-1) (78%). LCMS: m/z (ESI), [M+H] +=239.05. Step 2. Preparation of 2-(benzyloxy)-5-[(2-methoxyethoxy)methoxy]pyridine (INT-B11-2). 6-(benzyloxy)pyridine-3-ol (5 g, 24.84 mmol, 1 equivalent) was added to N,N- To the stirred mixture in dimethylformamide was added sodium hydroxide (60% in oil, 4 g, 12.42 mmol, 2 eq). The mixture was stirred for 15 min before the addition of 1-(chloromethoxy)-2-methoxyethane (3.1 g, 24.84 mmol, 1 eq). The mixture was stirred at 0 °C for 2 h. The reaction was quenched with water (50 mL) and the mixture was extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×50 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column eluted with petroleum ether/ethyl acetate (3/1) to give 6 g of 2-(benzyloxy)-5-[(2-methoxyethoxy)methoxy]pyridine (INT-B11-2) (75%) as a light yellow oil. LCMS: m/z (ESI), [M+H] +=290.10. 1H NMR (DMSO- d 6, 400 MHz) δ 3.22 (3H, s), 3.36-3.51 (2H, m), 3.65-3.80 (2H, m), 5.21 (2H, s), 5.29 (2H, s), 6.85 (1H, d), 7.28-7.42 (3H, m), 7.35-7.50 (2H, m), 7.50 (1H, d), 7.93 (1H, d). Step 3. Preparation of 2-(benzyloxy)-4-iodo-5-[(2-methoxyethoxy)methoxy]pyridine (INT-B11-3). To a stirred mixture of 2-(benzyloxy)-5-[(2-methoxyethoxy)methoxy]pyridine (5.8 g, 20.04 mmol, 1 equiv) in tetrahydrofuran (5 mL) at -78 °C under nitrogen atmosphere was added n-butyl lithium solution (2.5 M in tetrahydrofuran, 17 mL, 2 mmol). The mixture was stirred at -78 °C for 30 minutes. I was then added dropwise.2(8.1 g, 32.07 mmol, 1.60 equiv) in 10 mL tetrahydrofuran, and the mixture was stirred at -78 °C under nitrogen atmosphere for 1 h. The reaction was treated with saturated NH4Cl (100 mL) was quenched, and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column eluted with petroleum ether/ethyl acetate (3/1) to give 5.5 g of 2-(benzyloxy)-4-iodo-5-[(2-methoxyethoxy)methoxy]pyridine (INT-B11-3) (59%) as a yellow oil. LCMS: m/z (ESI), [M+H] +=415.95. 1H NMR (CDCl 3, 400 MHz) δ 3.42 (3H, s), 3.52-3.70 (2H, m), 3.85-4.02 (2H, m), 5.27 (2H, s), 5.33 (2H, s), 7.35-7.46 (6H, m), 7.94 (1H, s) Step 4. Preparation of 2'-(benzyloxy)-6-chloro-5'-[(2-methoxyethoxy)methoxy]-[2,4'-bipyridine]-4-carboxylic acid methyl ester (INT-B11-4). 2-Chloro-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine-4-carboxylic acid methyl ester (M1-1, 4 g crude product), 2-(benzyloxy)-4-iodo-5-[(2-methoxyethoxy)methoxy]pyridine (4.1 g, 10.08 mmol, 1 equivalent), Pd 2(dba) 3(0.92 g, 1.00 mmol, 0.1 eq.), bis(adamantan-1-yl)(butyl)phosphine (0.36 g, 1.00 mmol, 0.1 eq.) and Cs 2CO 3(9.8 g, 30.24 mmol, 3.00 equiv.) in toluene/H 2O (40 mL/10 mL) was stirred at 80 °C for 16 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (100 mL), and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column using petroleum ether/ethyl acetate (1/1) to give 4.8 g of methyl 2'-(benzyloxy)-6-chloro-5'-[(2-methoxyethoxy)methoxy]-[2,4'-bipyridine]-4-carboxylate (INT-B11-4) (93%) as a brown solid. LCMS: m/z (ESI), [M+H] +=459.05. Step 5. Preparation of 2'-(benzyloxy)-5'-[(2-methoxyethoxy)methoxy]-6-methyl-[2,4'-bipyridine]-4-carboxylic acid methyl ester (INT-B11-5). 2'-(benzyloxy)-6-chloro-5'-[(2-methoxyethoxy)methoxy]-[2,4'-bipyridine]-4-carboxylic acid methyl ester (2.8 g, 6.10 mmol, 1 equivalent), Pd(dppf)Cl 2(0.89 g, 1.22 mmol, 0.2 equivalent), K 2CO 3(1.6 g, 12.20 mmol, 2 equivalents), PCy 3.HBF 4A mixture of (0.67 g, 1.83 mmol, 0.3 equiv) and trimethyl-1,3,5,2,4,6-trioxatriborinane (2.07 g, 16.47 mmol, 2.7 equiv) in 1,4-dioxane (28 mL) was stirred at 100 °C for 3 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (200 mL), and the mixture was extracted with ethyl acetate (3×200 mL). The combined organic layers were washed with brine (3×200 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column using petroleum ether/ethyl acetate (1/1) to give 1.8 g of 2'-(benzyloxy)-5'-[(2-methoxyethoxy)methoxy]-6-methyl-[2,4'-bipyridine]-4-carboxylic acid methyl ester (INT-B11-5) (60%) as a yellow solid. LCMS: m/z (ESI), [M+H] += 439.15. Step 6. Preparation of methyl 2'-hydroxy-5'-[(2-methoxyethoxy)methoxy]-6-methyl-[2,4'-bipyridine]-4-carboxylate (INT-B11-6). A mixture of methyl 2'-(benzyloxy)-5'-[(2-methoxyethoxy)methoxy]-6-methyl-[2,4'-bipyridine]-4-carboxylate (1.8 g, 4.105 mmol, 1 eq.) and Pd/C (10%, 500 mg) in methanol (10 mL) was stirred under a hydrogen atmosphere for 1 h. The resulting mixture was filtered, and the filter cake was washed with methanol (150 mL). The solution was concentrated under reduced pressure. The residue was purified on a silica gel column using dichloromethane/methanol (10/1) to give 1.3 g of 2'-hydroxy-5'-[(2-methoxyethoxy)methoxy]-6-methyl-[2,4'-bipyridine]-4-carboxylic acid methyl ester (INT-B11-6) (82%) as a yellow solid. LCMS: m/z (ESI), [M+H] += 349.05. Step 7. Preparation of 5'-[(2-methoxyethoxy)methoxy]-1',6-dimethyl-2'-oxo-[2,4'-bipyridine]-4-carboxylic acid methyl ester (INT-B11-7) Methyl 2'-hydroxy-5'-[(2-methoxyethoxy)methoxy]-6-methyl-[2,4'-bipyridine]-4-carboxylic acid methyl ester (1.1 g, 3.15 mmol, 1 equivalent), K 2CO 3(1.3 g, 9.47 mmol, 3 equiv.) and CH 3A mixture of I (672 mg, 4.73 mmol, 1.5 equiv.) in acetone (10 mL) was stirred at 60 °C for 12 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (50 mL), and the mixture was extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. Pass the residue through C18 FLASH using 10 mmol/L NH4HCO 3and 0.1% NH 3 .H 2O and acetonitrile as the mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 1 g of 5'-[(2-methoxyethoxy)methoxy]-1',6-dimethyl-2'-oxo-[2,4'-bipyridine]-4-carboxylic acid methyl ester (INT-B11-7) (83%) as a light yellow oil. LCMS: m/z (ESI), [M+H] +=363.10. 1H NMR (CDCl 3, 400 MHz) δ 2.66 (3H, s), 3.41 (3H, s), 3.58 (5H, d), 3.71-3.96 (2H, m), 3.96 (3H, s), 5.02 (2H, s), 6.92 (1H, s), 7.45 (1H, s), 7.73 (1H, d), 7.99 (1H, d) Step 8. Preparation of 5'-hydroxy-1',6-dimethyl-2'-oxo-[2,4'-bipyridine]-4-carboxylic acid methyl ester (INT-B11-8). A solution of 5'-[(2-methoxyethoxy)methoxy]-1',6-dimethyl-2'-oxo-[2,4'-bipyridine]-4-carboxylic acid methyl ester (370 mg, 1.02 mmol, 1 equiv) and HCl (10 mL, 4 M in MeOH) was stirred at room temperature under nitrogen atmosphere for 1.5 h. The mixture was concentrated under reduced pressure. The residue was treated with dichloromethane (30 mL) and NaHCO 3The solution was worked up and filtered. The filtrate was concentrated under reduced pressure to give 260 mg of 5'-hydroxy-1',6-dimethyl-2'-oxo-[2,4'-bipyridine]-4-carboxylic acid methyl ester (INT-B11-8) as a yellow solid. This material was used in the next reaction without further purification. LCMS: m/z (ESI), [M+H] +=275.05. Step 9. 1',6-dimethyl-5'-{[(4 RPreparation of methyl 5-[(2-nitrophenyl)amino]pentyl]oxy}-2'-oxo-[2,4'-bipyridine]-4-carboxylate (INT-B11-9). 5'-Hydroxy-1',6-dimethyl-2'-oxo-[2,4'-bipyridine]-4-carboxylate (282 mg, 1.03 mmol, 1 equivalent), (4 RA mixture of 2-(tributyl-1^[5]-phosphinilidene)acetonitrile (1.00 g, 4.14 mmol, 4.00 equiv)-4-methyl-5-[(2-nitrophenyl)amino]pentan-1-ol (247 mg, 1.03 mmol, 1 equiv) and 2-(tributyl-1^[5]-phosphinilidene)acetonitrile (1.00 g, 4.14 mmol, 4.00 equiv) in toluene (10 mL) was stirred at 100 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (100 mL), and the mixture was extracted with dichloromethane (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column using petroleum ether/ethyl acetate (1/1) to give 400 mg of 1',6-dimethyl-5'-{[(4R)-4-methyl-5-[(2-nitrophenyl)amino]pentyl]oxy}-2'-oxo-[2,4'-bipyridine]-4-carboxylic acid methyl ester (INT-B11-9) (77%) as a yellow oil. LCMS: m/z (ESI), [M+H] +=495.10. Step 10. Preparation of 5'-{[(4R)-5-[(2-aminophenyl)amino]-4-methylpentyl]oxy}-1',6-dimethyl-2'-oxo-[2,4'-bipyridine]-4-carboxylic acid methyl ester (INT-B11-10). To a stirred mixture of 1',6-dimethyl-5'-{[(4R)-4-methyl-5-[(2-nitrophenyl)amino]pentyl]oxy}-2'-oxo-[2,4'-bipyridine]-4-carboxylic acid methyl ester (400 mg, 0.80 mmol, 1 eq.) and ranyl nickel (34 mg, 0.40 mmol, 0.50 eq.) in methanol (10 mL) was added hydrazine hydrate (77 mg, 2.42 mmol, 3.00 eq.) at 0°C. The mixture was stirred at 0 °C for 2 h under nitrogen atmosphere. The mixture was filtered and the filter cake was washed with methanol (3×10 mL). The resulting solution was concentrated under reduced pressure. The residue was purified on prep-TLC using petroleum ether/ethyl acetate (1/1) to give 350 mg of 5'-{[(4 R)-5-[(2-aminophenyl)amino]-4-methylpentyl]oxy}-1',6-dimethyl-2'-oxo-[2,4'-bipyridine]-4-carboxylic acid methyl ester (INT-B11-10) (93%). LCMS: m/z (ESI), [M+H] +=465.20. Step 11. 5'-{[(4 RPreparation of methyl 4-[(2-amino-1,3-benzodiazol-1-yl)methyl]pentyl]oxy}-1',6-dimethyl-2'-oxo-[2,4'-bipyridine]-4-carboxylate (INT-B11-11). 5'-{[(4 RA mixture of methyl 5-[(2-aminophenyl)amino]-4-methylpentyl]oxy}-1',6-dimethyl-2'-oxo-[2,4'-bipyridine]-4-carboxylate (400 mg, 0.86 mmol, 1 equiv) and BrCN (182 mg, 1.722 mmol, 2.00 equiv) in ethanol (10 mL) was stirred at room temperature overnight under nitrogen atmosphere. The reaction was quenched with water (50 mL), and the mixture was extracted with ethyl acetate (3×50 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on prep-TLC using dichloromethane/methanol (20/1) to give 350 mg of 5'-{[(4 R)-4-[(2-amino-1,3-benzodiazol-1-yl)methyl]pentyl]oxy}-1',6-dimethyl-2'-oxo-[2,4'-bipyridine]-4-carboxylic acid methyl ester (INT-B11-11) (83%). LCMS: m/z (ESI), [M+H] +=490.10. Step 12. 5'-{[(4 R)-4-[(2-amino-1,3-benzodiazol-1-yl)methyl]pentyl]oxy}-1',6-dimethyl-2'-oxo-[2,4'-bipyridine]-4-carboxylic acid (INT-B11-12). 5'-{[(4 R)-4-[(2-amino-1,3-benzodiazol-1-yl)methyl]pentyl]oxy}-1',6-dimethyl-2'-oxo-[2,4'-bipyridine]-4-carboxylic acid methyl ester (300 mg, 0.61 mmol, 1 equivalent) and LiOH .H 2A mixture of O (44 mg, 1.83 mmol, 3 equiv.) in tetrahydrofuran/water (4 mL/1 mL) was stirred at room temperature under a nitrogen atmosphere for 2 h. The resulting mixture was concentrated under reduced pressure. The residue was subjected to C18 FLASH chromatography using a HPLC-MS/MS system containing 0.1% NH3 .H 2O and acetonitrile water as the mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 200 mg of 5'-{[(4 R)-4-[(2-amino-1,3-benzodiazol-1-yl)methyl]pentyl]oxy}-1',6-dimethyl-2'-oxo-[2,4'-bipyridine]-4-carboxylic acid (INT-B11-12) (68%). LCMS: m/z (ESI), [M+H] += 476.10. Step 13. (12 R)-5,12,27-trimethyl-8-oxa-5,14,21,23,28-pentazapentacyclo[23.3.1.0^{2,7}.0^{14,22}.0^{15,20}]twenty-nine-1(29),2,6,15,17,19,21,25,27-nonene-4,24-dione (Example B11). 5'-{[(4 R)-4-[(2-amino-1,3-benzodiazol-1-yl)methyl]pentyl]oxy}-1',6-dimethyl-2'-oxo-[2,4'-bipyridine]-4-carboxylic acid (400 mg, 0.84 mmol, 1 equivalent), N,N,N,N-Tetramethyl- O-(7-Azabenzotriazol-1-yl)urea hexafluorophosphate (479 mg, 1.26 mmol, 1.5 equivalents) and N, N- A mixture of diisopropylethylamine (217 mg, 1.68 mmol, 2 equiv.) in 1,4-dioxane (10 mL) was stirred at room temperature overnight under nitrogen atmosphere. The reaction was quenched with water (100 mL), and the mixture was extracted with dichloromethane (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was subjected to prep-HPLC using an XBridge Shield RP18 OBD column with 10 mmol/L NH4HCO 3and 0.1% NH 3 .H 2O water and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 186 mg (12 R)-5,12,27-trimethyl-8-oxa-5,14,21,23,28-pentazapentacyclic [23.3.1.0^{2,7}.0^{14,22}.0^{15,20}]twenty-nine-1(29),2,6,15,17,19,21,25,27-nonene-4,24-dione (Example B11) (48%). LCMS: m/z (ESI), [M+H] +=458.25. 1H NMR (DMSO-d 6, 400 MHz) δ 0.93 (3H, d), 1.47-1.60 (1H, m), 1.66-1.80 (1H, m), 1.96-2.20 (2H, m), 2.25-2.37 (1H, m), 2.63 (3H, s), 3.48 (3H, s), 3.93-4.08 (3H, m), 4.18 (1H, dd), 7.12 (1H, s), 7.19-7.34 (2H, m), 7.53-7.61 (3H, m), 7.83 (1H, d), 8.92 (1H, d), 12.80 (1H, s). Example B12 (11 R)-18-[2-(dimethylamino)ethyl]-5,11,30-trimethyl-7-oxo-4,5,13,18,24,26,31-heptaazahexacyclic[26.3.1.0^{2,6}.0^{13,25}. 0^{14,23}.0^{15,20}]triacontac-1(31),2(6),3,14,20,22,24, 28(32),29-nonan-27-one Step 1. 2,2,2-trifluoro-1-(5-{[(2 RPreparation of 2,2,2-trifluoro-1-(5-fluoro-6-nitro-3,4-dihydro-1H-isoquinolin-2-yl)ethanone (INT-A102-3, 15 g, 51.34 mmol, 1 equivalent), N,N-diisopropylethylamine (19.9 g, 154.01 mmol, 3 equivalents) and (4 RA mixture of 5-amino-4-methylpentan-1-ol (M3-6, 6016 mg, 51.34 mmol, 1 equivalent) in acetonitrile (150 mL) was stirred at 80 °C for 2 h. The mixture was cooled to room temperature. The reaction was quenched with water (500 mL), and the mixture was extracted with dichloromethane (3×500 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by eluting with petroleum ether/ethyl acetate (1/1) on a silica gel column to give 10 g of 2,2,2-trifluoro-1-(5-{[(2R)-5-hydroxy-2-methylpentyl]amino}-6-nitro-3,4-dihydro-1H-isoquinolin-2-yl)ethanone (INT-B12-1) (50%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=390.10. Step 2. (4 R)-4-methyl-5-[(6-nitro-1,2,3,4-tetrahydroisoquinolin-5-yl)amino]pentan-1-ol (INT-B12-2). 2,2,2-trifluoro-1-(5-{[(2 R)-5-hydroxy-2-methylpentyl]amino}-6-nitro-3,4-dihydro-1H-isoquinolin-2-yl)ethanone (10 g, 25.68 mmol, 1 equivalent) and K 2CO 3(10.6 g, 77.05 mmol, 3 equiv.) in methanol (80 mL) and H 2O (40 mL) was stirred at room temperature for 5 h. The reaction was treated with water (500 mL), and the mixture was extracted with dichloromethane (3×500 mL). The combined organic layers were washed with brine (3×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column using petroleum ether/ethyl acetate (1/1) to obtain 7 g (4 R)-4-methyl-5-[(6-nitro-1,2,3,4-tetrahydroisoquinolin-5-yl)amino]pentan-1-ol (INT-B12-2) (93%). LCMS: m/z (ESI), [M+H] +=294.15. 1H NMR (DMSO-d 6,400 MHz) δ 0.83-0.88 (3 H, m), 1.07-1.11 (1 H, m), 1.22-1.51 (3 H, m), 1.57-1.64 (1 H, m), 2.59 (2 H, t), 2.88-2.92 (3 H, m), 3.10-3.20 (1 H, m), 3.30-3.33 (2 H, m), 3.87 (2H, s), 4.37 (1 H, s), 6.56 (1 H, d), 6.69 (1 H, t), 7.26-7.37 (1 H, m), 7.70 (1 H, d). Step 3. 5-{[(2 RPreparation of tert-butyl 5-hydroxy-2-methylpentyl]amino}-6-nitro-3,4-dihydro-1H-isoquinoline-2-carboxylate (INT-B12-3). (4 RA mixture of )-4-methyl-5-[(6-nitro-1,2,3,4-tetrahydroisoquinolin-5-yl)amino]pentan-1-ol (8 g, 27.27 mmol, 1 equiv), trimethylamine (5.5 g, 54.54 mmol, 2 equiv) and di-tert-butyl dicarbonate (7.1 g, 32.72 mmol, 1.2 equiv) in dichloromethane (100 mL) was stirred at room temperature for 2 h. The reaction was quenched with water (300 mL), and the mixture was extracted with dichloromethane (3×300 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column using petroleum ether/ethyl acetate (1/1) to give 6.5 g of 5-{[(2 R)-5-hydroxy-2-methylpentyl]amino}-6-nitro-3,4-dihydro-1H-isoquinoline-2-carboxylic acid tert-butyl ester (INT-B12-3) (60%). LCMS: m/z (ESI), [M+H] +=394.20. 1H NMR (DMSO-d 6400 MHz) δ 0.84 (3H, d), 1.03-1.17 (1H, m), 1.22-1.45 (2H, m), 1.45 (9H, s), 1.61 (1H, q), 2.75 (2H, t), 2.93-2.97 (1H, m), 3.08-3.12 (1H, m), 3.30-3.33 (2H, m), 3.51 (2H, t), 4.36 (1H, t), 4.52 (2H, s), 6.73 (1H, d), 6.83 (1H, s), 7.80 (1H, d) Step 4. 5-{[(2 RPreparation of tert-butyl 5-({4-[4-(methoxycarbonyl)-6-methylpyridin-2-yl]-2-methylpyrazol-3-yl}oxy)-2-methylpentyl]amino}-6-nitro-3,4-dihydro-1H-isoquinoline-2-carboxylate (INT-B12-4). In a nitrogen atmosphere at 0 ° C, 5-{[(2 RTo a stirred mixture of tert-butyl 2-(5-hydroxy-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylate (1 g, 2.54 mmol, 1 eq.) and triphenylphosphine (2 g, 7.62 mmol, 3 eq.) in tetrahydrofuran (20 mL) were added methyl 2-(5-hydroxy-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylate (M1, 691 mg, 2.79 mmol, 1.1 eq.) and diisopropyl azodicarboxylate (1.5 g, 7.62 mmol, 3 eq.). The mixture was stirred at room temperature for 2 h. The reaction was quenched with water (100 mL), and the mixture was extracted with dichloromethane (3×80 mL). The combined organic layers were washed with brine (3×80 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column using petroleum ether/ethyl acetate (2/3) to obtain 900 mg of 5-{[(2 R)-5-({4-[4-(methoxycarbonyl)-6-methylpyridin-2-yl]-2-methylpyrazol-3-yl}oxy)-2-methylpentyl]amino}-6-nitro-3,4-dihydro-1H-isoquinoline-2-carboxylic acid tert-butyl ester (INT-B12-4) (57%). LCMS: m/z (ESI), [M+H] +=623.30. Step 5. 6-amino-5-{[(2 R)-5-({4-[4-(methoxycarbonyl)-6-methylpyridin-2-yl]-2-methylpyrazol-3-yl}oxy)-2-methylpentyl]amino}-3,4-dihydro-1H-isoquinoline-2-carboxylic acid tert-butyl ester (INT-B12-5). 5-{[(2 RA mixture of tert-butyl 5-({4-[4-(methoxycarbonyl)-6-methylpyridin-2-yl]-2-methylpyrazol-3-yl}oxy)-2-methylpentyl]amino}-6-nitro-3,4-dihydro-1H-isoquinoline-2-carboxylate (500 mg, 0.80 mmol, 1 eq.) and Pd/C (10%, 30 mg) in methanol (10 mL) was stirred under hydrogen atmosphere for 1 h. The resulting mixture was filtered, and the filter cake was washed with methanol (3×50 mL). The resulting solution was concentrated under reduced pressure. The residue was purified by prep-TLC using dichloromethane/methanol (10/1) to give 360 mg of 6-amino-5-{[(2 R)-5-({4-[4-(methoxycarbonyl)-6-methylpyridin-2-yl]-2-methylpyrazol-3-yl}oxy)-2-methylpentyl]amino}-3,4-dihydro-1H-isoquinoline-2-carboxylic acid tert-butyl ester (INT-B12-5) (76%). LCMS: m/z (ESI), [M+H] +=593.40. Step 6. 2-(5-{[(4 RPreparation of methyl pyridine-4-carboxylate (INT-B12-6). 6-amino-5-{[(2 RA mixture of tert-butyl 5-({4-[4-(methoxycarbonyl)-6-methylpyridin-2-yl]-2-methylpyrazol-3-yl}oxy)-2-methylpentyl]amino}-3,4-dihydro-1H-isoquinoline-2-carboxylate (350 mg, 0.59 mmol, 1 equiv) and BrCN (75 mg, 0.71 mmol, 1.2 equiv) in dichloromethane (4 mL) was stirred at room temperature under nitrogen atmosphere for 2 h. The reaction was quenched with water (60 mL), and the mixture was extracted with dichloromethane (3×60 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by prep-TLC using dichloromethane/methanol (11/1) to give 300 mg of 2-(5-{[(4 R)-4-{[2-amino-7-(tert-butoxycarbonyl)-6H,8H,9H-imidazo[4,5-f]isoquinolin-1-yl]methyl}pentyl]oxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylic acid methyl ester (INT-B12-6) (82%). LCMS: m/z (ESI), [M+H] +=618.35. Step 7. 2-(5-{[(4 RPreparation of 2-(5-{[(4 R)-4-{[2-amino-7-(tert-butoxycarbonyl)-6H,8H,9H-imidazo[4,5-f]isoquinolin-1-yl]methyl}pentyl]oxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylic acid methyl ester (290 mg, 0.47 mmol, 1 eq.) and LiOH (30 mg, 0.70 mmol, 1.5 eq.) in tetrahydrofuran/H 2The mixture in 400 mL/1 mL was stirred at room temperature for 2 h. The mixture was concentrated in vacuo. The residue was purified by C18 flash chromatography using water and acetonitrile as mobile phase. The fractions containing the desired compound were evaporated to dryness to give 900 mg of 2-(5-{[(4 R)-4-{[2-amino-7-(tert-butoxycarbonyl)-6H,8H,9H-imidazo[4,5-f]isoquinolin-1-yl]methyl}pentyl]oxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylic acid (INT-B12-7) (71%). LCMS: m/z (ESI), [M+H] +=604.30. Step 8. (11 RPreparation of tert-butyl ester (INT-B12-8) of 5,11,30-trimethyl-27-oxo-7-oxa-4,5,13, 18,24,26,31-heptaazahexacyclo[26.3.1.0^{2,6}.0^{13,25}.0^{14, 23}.0^{15,20}]tri-dodeca-1(31),2(6),3,14,20,22,24,28(32),29-nonene-18-carboxylate.R)-4-{[2-amino-7-(tert-butoxycarbonyl)-6H,8H,9H-imidazo[4,5-f]isoquinolin-1-yl]methyl}pentyl]oxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylic acid (200 mg, 0.33 mmol, 1 equivalent), N,N-Diisopropylethylamine (128 mg, 0.99 mmol, 3 equiv.) and N,N,N,N-Tetramethyl- OA mixture of -(7-azabenzotriazol-1-yl)urea hexafluorophosphate (252 mg, 0.66 mmol, 2 equiv.) in dioxane (5 mL) was stirred at room temperature under nitrogen atmosphere for 2 h. The reaction was quenched with water (60 mL), and the mixture was extracted with dichloromethane (3×60 mL). The combined organic layers were washed with brine (3×40 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by prep-TLC eluting with dichloromethane/methanol (15/1) to give 160 mg (11 R)-5,11,30-trimethyl-27-dioxo-7-dioxa-4,5,13, 18,24,26,31-heptaazahexacyclic [26.3.1.0^{2,6}.0^{13,25}.0^{14, 23}.0^{15,20}]triacontidine-1(31),2(6),3,14,20,22,24,28(32),29-nonene-18-carboxylic acid tert-butyl ester (INT-B12-8) (82%). LCMS: m/z (ESI), [M+H] +=586.30. Step 9. Preparation of (11R)-5,11,30-trimethyl-7-oxa-4,5,13,18,24,26, 31-heptaazahexacyclo[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15, 20}]triacontac-1(31),2(6),3,14,20,22,24,28(32),29-nonan-27-one (INT-B12-9). At room temperature, (11 RTo a stirred mixture of tert-butyl 5,11,30-trimethyl-27-oxo-7-oxa-4,5,13,18,24,26,31-heptaazahexacyclo[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]triacontria-1(31),2(6),3,14,20,22,24,28(32),29-nonene-18-carboxylate (120 mg, 0.21 mmol, 1 equiv) in 1,4-dioxane (0.5 mL) was added HCl solution (3 mL, 4 M in 1,4-dioxane). The mixture was stirred at room temperature for 2 h. The resulting mixture was treated with saturated NaHCO3(aqueous solution) (100 mL) and extracted with dichloromethane (3×100 mL). The combined organic layers were washed with brine (3×70 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure to give 99 mg (11 R)-5,11,30-trimethyl-7-oxa-4,5,13,18,24,26,31-heptaazahexacyclo[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]triacontac-1(31),2(6),3,14,20,22,24,28(32),29-nonan-27-one (INT-B12-9) (99%). This material was used in the next step without purification. LCMS: m/z (ESI), [M+H] +=486.30. Step 10. (11 R)-18-[2-(dimethylamino)ethyl]-5,11,30-trimethyl-7-oxa-4,5,13,18,24,26,31-heptaazahexacyclic [26.3.1.0^{2,6}. 0^{13,25}.0^{14,23}.0^{15,20}]triacontac-1(31),2(6),3,14, 20,22,24,28(32),29-nonan-27-one (Example B12). At room temperature, to (11 R)-5,11,30-trimethyl-7-oxa-4,5,13,18,24,26,31-heptaazahexacyclo[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]triacontac-1(31),2(6),3,14,20,22,24,28(32),29-nonan-27-one (99 mg, 0.20 mmol, 1 equivalent) and trimethylamine (62 mg, 0.61 mmol, 3 equivalents) in N,N-To the stirred mixture in dimethylformamide (5 mL), (2-bromoethyl)dimethylamine (56 mg, 0.37 mmol, 1.8 equiv) and KI (10 mg, 0.06 mmol, 0.3 equiv) were added. The resulting mixture was stirred at 80 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (50 mL), and the mixture was extracted with dichloromethane (3×60 mL). The combined organic layers were washed with brine (3×40 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by prep-TLC using dichloromethane/methanol (20/1). The product was purified by prep-HPLC using an XBridge Prep OBD C18 column containing 10 mmol/L NH4HCO 3The product was further purified using water and acetonitrile as mobile phase. The fractions containing the desired compound were evaporated to dryness to give 9.3 mg (11 R)-18-[2-(dimethylamino)ethyl]-5,11,30-trimethyl-7-oxa-4,5,13,18,24,26,31-heptaazahexacyclo[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]triacontac-1(31),2(6),3,14,20,22,24,28(32),29-nonan-27-one (Example B12) (8%). LCMS: m/z (ESI), [M+H] +=557.30. 1H NMR (DMSO- d 6, 400 MHz) δ 0.85 (3H, d), 1.41-1.53 (1H, m), 1.77-1.92 (1H, m), 1.95-2.08 (1H, m), 2.19 (6H, s), 2.21-2.29 (1H, m), 2.49-2.61 (5H, m), 2.72-2.86 (2H, m), 2.88-3.02 (1H, m), 3.10-3.27 (2H, m), 3.60-3.70 (2H, m), 3.73 (3H, s), 3.96-4.02 (1H, m), 4.22-4.28 (4H, m), 6.95 (1H, d), 7.39 (1H, d), 7.57 (1H, s), 7.93 (1H, s), 8.36 (1H, d), 12.75 (1H, s). Example B13 ( R)-11,26,7-trimethyl-57-(oxacyclobutane-3-yl)-56,57,58,59-tetrahydro-11H,51H-11-oxa-4-aza-5(2,1)-imidazo[4,5-f]isoquinoline-2(2,4)-pyridine-1(4,5)-pyrazolocycloundecane-3-one Step 1. (11 R)-11,26,7-trimethyl-57-(oxacyclobutane-3-yl)-56, 57,58,59-tetrahydro-11H,51H-11-oxa-4-aza-5(2,1)-imidazo[4,5-f]isoquinoline-2(2,4)-pyridine-1(4,5)-pyrazolocycloundecane-3-one (Example B13). (11 RA mixture of 5,11,30-trimethyl-7-oxa-4,5,13,18, 24,26,31-heptaazahexacyclo[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]triacontria-1(31),2(6),3,14,20,22,24,28(32),29-nonan-27-one (INT-B12-9, 40 mg, 0.08 mmol, 1 eq) in dichloromethane (2 mL) was treated with 3-oxacyclobutanone (12 mg, 0.16 mmol, 2 eq) and trimethylamine (17 mg, 0.16 mmol, 2 eq) under nitrogen atmosphere at room temperature for 0.5 min. h, followed by addition of NaBH(AcO) at 0°C 3(52 mg, 0.25 mmol, 3 equiv.). The resulting mixture was stirred at room temperature overnight under nitrogen atmosphere. The reaction was quenched with water (20 mL), and the mixture was extracted with dichloromethane (3×20 mL). The combined organic layers were washed with brine (3×20 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was subjected to prep-HPLC using an XBridge Shield RP18 OBD column with 10 mmol/L NH4HCO 3and 0.1% NH 3 .H 2O water and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 10.5 mg (11 R)-5,11,30-trimethyl-18-(oxacyclobutane-3-yl)-7-oxa-4,5,13,18,24,26,31-heptaazahexacyclo[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]triacontac-1(31),2(6),3,14,20,22,24,28(32),29-nonan-27-one (Example B13) (22%). LCMS: m/z (ESI), [M+H] +=542.20. 1H NMR (DMSO- d 6, 400 MHz) δ 0.86 (3H, d), 1.38-1.53 (1H, m), 1.79-1.93 (1H, m), 1.96-2.06 (1H, m), 2.16-2.28 (1H, m), 2.58 (3H, s), 2.58-2.73 (3H, m), 2.90-3.02 (1H, m), 3.15-3.26 (1H, m), 3.45-3.64 (3H, m), 3.73 (3H, s), 3.97-4.01 (1H, m), 4.14-4.38 (3H, m), 4.56 (2H, t), 4.65 (2H, t), 6.96 (1H, d), 7.41 (1H, d), 7.58 (1H, s), 7.93 (1H, s), 8.39 (1H, s), 12.78 (1H, s). Example B14 30-Methyl-11-oxa-8,9,17,24,26,31-hexaazahexacyclo[26.3.1.0^{2,10}.0^{3,8}.0^{17,25}.0^{18,23}]triacontac-1(31),2,4,6,9,18,20,22,24,28(32),29-undecane-27-one Step 1. Preparation of 1-(5-bromopentyl)-1,3-benzodiazol-2-amine (INT-B14-1). 2-aminobenzimidazole (4 g, 30.04 mmol, 1 equivalent), K 2CO 3A mixture of (12.46 g, 90.12 mmol, 3 equiv.) and 1,5-dibromopentane (27.63 g, 120.16 mmol, 4 equiv.) in acetonitrile (80 mL) was stirred at 60 °C for 5 h. The mixture was cooled to room temperature. The reaction was quenched with water (500 mL), and the mixture was extracted with ethyl acetate (3×500 mL). The combined organic layers were washed with brine (3×500 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column using dichloromethane/methanol (10/1) to give 4 g of 1-(5-bromopentyl)-1,3-benzodiazol-2-amine (INT-B14-1) (47%) as a brown solid. LCMS: m/z (ESI), [M+H] +=283.95. Step 2. Preparation of 1-[5-({3-bromopyrazolo[1,5-a]pyridin-2-yl}oxy)pentyl]-1,3-benzodiazol-2-amine (INT-B14-2). 1-(5-bromopentyl)-1,3-benzodiazol-2-amine (3.97 g, 14.08 mmol, 1.5 equivalents), 3-bromopyrazolo[1,5-a]pyridin-2-ol (2 g, 9.38 mmol, 1.00 equivalents) and K 2CO 3(2.59 g, 18.77 mmol, 2 equiv.) in acetonitrile (80 mL) was stirred at 60 °C for 2 h. The mixture was cooled to room temperature. The reaction was quenched with water (500 mL), and the mixture was extracted with ethyl acetate (3×500 mL). The combined organic layers were washed with brine (3×500 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue is purified by C18 FLASH chromatography using water and acetonitrile as mobile phase. The fractions containing the desired compound are evaporated to dryness to give 2.3 g of 1-[5-({3-bromopyrazolo[1,5-a]pyridin-2-yl}oxy)pentyl]-1,3-benzodiazol-2-amine (INT-B14-2) (39%) as a brown solid. LCMS: m/z (ESI), [M+H] +=414.20. 1H NMR (DMSO-d 6 , 400 MHz) δ 1.40-1.52 (2H, m), 1.63.175 (2H, m), 1.76-1.88 (2H, m), 3.92-4.03 (2H, m), 4.30 (2H, t), 6.37 (2H, s), 6.79-6.87 (2H, m), 6.88-6.94 (1H, m), 7.12 (2H, t), 7.26-7.39 (2H, m),8.54 (1H, d). Step 3. Preparation of 6-chloro-4-(methoxycarbonyl)pyridin-2-ylboronic acid) (M1-1). A mixture of bis(pinacol)diboron (1631.71 mg, 6.426 mmol, 1.5 eq) in methyl tert-butyl ether (20 mL) was warmed to 80 °C and stirred under nitrogen atmosphere for 0.5 h, then cooled to room temperature. To this mixture was added bis(1,5-cyclooctadiene)di-μ-methoxydiiridium(I) (283.95 mg, 0.428 mmol, 0.1 eq) and 4-tert-butyl-2-(4-tert-butylpyridin-2-yl)pyridine (229.95 mg, 0.857 mmol, 0.2 eq). The mixture was stirred at 25 °C for 0.5 h, then methyl 2-chloropyridine-4-carboxylate (735 mg, 4.284 mmol, 1 eq) was added. The reaction mixture was further stirred at 80°C for 16 h. The mixture was cooled to room temperature and concentrated under reduced pressure to give 850 mg (crude) of 6-chloro-4-(methoxycarbonyl)pyridin-2-ylboronic acid (M1-1) as a brown oil. This material was used in the next reaction without further purification. LCMS: m/z (ESI), [M+H] +=216.05. Step 4. 2-(2-{[5-(2-amino-1,3-benzodiazol-1-yl)pentyl]oxy}pyrazolo[1,5-a]pyridin-3-yl)-6-chloropyridine-4-carboxylic acid methyl ester (INT-B14-3). Freshly prepared 6-chloro-4-(methoxycarbonyl)pyridin-2-ylboronic acid (850 mg crude, about 2.5 equivalents), 1-[5-({3-bromopyrazolo[1,5-a]pyridin-2-yl}oxy)pentyl]-1,3-benzodiazol-2-amine (654 mg, 1.58 mmol, 1.00 equivalents), Cs 2CO 3(1543 mg, 4.73 mmol, 3 equivalents), Pd 2(dba) 3(144 mg, 0.15 mmol, 0.1 eq.) and bis(adamantan-1-yl)(butyl)phosphine (57 mg, 0.16 mmol, 0.1 eq.) in H 2A mixture of 20% O (4 mL) and toluene (20 mL) was stirred at 80 °C under nitrogen atmosphere for 2 h. The mixture was cooled to room temperature. The reaction was quenched with water (200 mL), and the mixture was extracted with ethyl acetate (3×200 mL). The combined organic layers were washed with brine (3×200 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column using petroleum ether/ethyl acetate (1/3) to give 250 mg of methyl 2-(2-{[5-(2-amino-1,3-benzodiazol-1-yl)pentyl]oxy}pyrazolo[1,5-a]pyridin-3-yl)-6-chloropyridine-4-carboxylate (INT-B14-3) (31%) as a brown solid. LCMS: m/z (ESI), [M+H] +=505.15 Step 5. Preparation of 2-(2-{[5-(2-amino-1,3-benzodiazol-1-yl)pentyl]oxy}pyrazolo[1,5-a]pyridin-3-yl)-6-methylpyridine-4-carboxylic acid methyl ester (INT-B14-4). 2-(2-{[5-(2-amino-1,3-benzodiazol-1-yl)pentyl]oxy}pyrazolo[1,5-a]pyridin-3-yl)-6-chloropyridine-4-carboxylic acid methyl ester (140 mg, 0.27 mmol, 1 equivalent), trimethyl-1,3,5,2,4,6-trioxatriboborohexane (69 mg, 0.55 mmol, 2 equivalents), Pd(dppf)Cl 2 .CH 2Cl 2(3 mg, 0.01 mmol, 0.2 equivalent), K 2CO 3(76 mg, 0.55 mmol, 2 eq.) and PCy 3HBF 4A mixture of (30 mg, 0.08 mmol, 0.3 equiv) in 1,4-dioxane (10 mL) was stirred at 100 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (200 mL), and the mixture was extracted with ethyl acetate (3×200 mL). The combined organic layers were washed with brine (3×200 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on Prep-TLC using dichloromethane/methanol (20/1) to give 80 mg of methyl 2-(2-{[5-(2-amino-1,3-benzodiazol-1-yl)pentyl]oxy}pyrazolo[1,5-a]pyridin-3-yl)-6-methylpyridine-4-carboxylate (INT-B14-4) (59%) as a brown solid. LCMS: m/z (ESI), [M+H] +=485.25. Step 6. Preparation of 2-(2-{[5-(2-amino-1,3-benzodiazol-1-yl)pentyl]oxy}pyrazolo[1,5-a]pyridin-3-yl)-6-methylpyridine-4-carboxylic acid (INT-B14-5). 2-(2-{[5-(2-amino-1,3-benzodiazol-1-yl)pentyl]oxy}pyrazolo[1,5-a]pyridin-3-yl)-6-methylpyridine-4-carboxylic acid methyl ester (70 mg, 0.14 mmol, 1 equivalent) and LiOH .H 2A mixture of O (18 mg, 0.43 mmol, 3 equiv.) in tetrahydrofuran (5 mL) and water (1.25 mL) was stirred at room temperature under a nitrogen atmosphere for 2 h. The resulting mixture was concentrated under reduced pressure. The residue was purified by C18 FLASH using water and acetonitrile as mobile phases. The fractions containing the desired compound were evaporated to dryness to give 30 mg of 2-(2-{[5-(2-amino-1,3-benzodiazol-1-yl)pentyl]oxy}pyrazolo[1,5-a]pyridin-3-yl)-6-methylpyridine-4-carboxylic acid (INT-B14-5) (44%) as a white solid. LCMS: m/z (ESI), [M+H] +=471.30. Step 7. Preparation of 30-methyl-11-oxa-8,9,17,24,26,31-hexaazahexacyclo[26.3.1.0^{2,10}.0^{3,8}.0^{17,25}.0^{18,23}]triacontidine-1(31),2,4,6,9,18,20,22,24,28(32),29-undecane-27-one (Example B14). 2-(2-{[5-(2-amino-1,3-benzodiazol-1-yl)pentyl]oxy}pyrazolo[1,5-a]pyridin-3-yl)-6-methylpyridine-4-carboxylic acid (25 mg, 0.05 mmol, 1 equivalent), N,N-Diisopropylethylamine (20 mg, 0.15 mmol, 3 equivalents) and N,N,N,N-Tetramethyl- OA mixture of -(7-azabenzotriazol-1-yl)uronium hexafluorophosphate (30 mg, 0.08 mmol, 1.5 equiv.) in 1,4-dioxane (10 mL) was stirred at room temperature for 2 h under a nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by prep-HPLC using an XBridge Prep OBD C18 column containing 10 mmol/L NH4HCO 3and 0.1% NH 3 .H 2O was used for purification with water and acetonitrile as the mobile phase. The fractions containing the desired compound were evaporated to dryness to give 9.4 mg of 30-methyl-11-oxa-8,9,17,24,26,31-hexaazahexacyclo[26.3.1.0^{2,10}.0^{3,8}.0^{17,25}.0^{18,23}]triacontac-1(31),2,4,6,9,18,20,22,24,28(32),29-undecane-27-one (Example B14) (39%) as a white solid. LCMS: m/z (ESI), [M+H] +=453.20. 1H NMR (DMSO-d 6 , 400 MHz) δ 1.91-2.12 (6H, m), 2.64 (3H, s), 4.23 (2H, t), 4.48 (2H, t), 6.91 (1H, t), 7.20-7.33 (2H, m), 7.42 (1H, t), 7.49 (1H, s), 7.55 (2H, d), 8.57-8.67 (2H, m), 9.04 (1H, s), 12.66 (1H, s). Example B15 30-Chloro-11-oxo-8,9,17,24,26,31-hexaazahexacyclo[26.3.1.0^{2,10}.0^{3,8}.0^{17,25}. 0^{18,23}]triacontidine-1(31),2,4,6,9,18,20,22,24,28(32),29-undecane-27-one Step 1. Preparation of 2-(2-{[5-(2-amino-1,3-benzodiazol-1-yl)pentyl]oxy}pyrazolo[1,5-a]pyridin-3-yl)-6-chloropyridine-4-carboxylic acid (INT-B15-1). 2-(2-{[5-(2-amino-1,3-benzodiazol-1-yl)pentyl]oxy}pyrazolo[1,5-a]pyridin-3-yl)-6-chloropyridine-4-carboxylic acid methyl ester (330 mg, 0.65 mmol, 1 equivalent) and LiOH .H2O (41 mg, 0.98 mmol, 1.5 equiv.) in tetrahydrofuran (10 mL) and H 2The mixture in 20% O (2.5 mL) was stirred at room temperature under nitrogen atmosphere for 2 h. The resulting mixture was concentrated under reduced pressure. The residue was subjected to prep-HPLC using an XBridge Shield RP18 OBD column with 10 mmol/L NH4HCO 3of water and acetonitrile as mobile phase. The fractions containing the desired compound were evaporated to dryness to give 134 mg of 2-(2-{[5-(2-amino-1,3-benzodiazol-1-yl)pentyl]oxy}pyrazolo[1,5-a]pyridin-3-yl)-6-chloropyridine-4-carboxylic acid (INT-B15-1) (41%) as a brown solid. LCMS: m/z (ESI), [M+H] +=491.15. Step 2. Preparation of 30-chloro-11-oxa-8,9,17,24,26,31-hexaazahexacyclo[26.3.1.0^{2,10}.0^{3,8}.0^{17,25}.0^{18,23}]triacontidine-1(31),2,4,6,9,18,20,22,24,28(32),29-undecane-27-one (Example B15). 2-(2-{[5-(2-amino-1,3-benzodiazol-1-yl)pentyl]oxy}pyrazolo[1,5-a]pyridin-3-yl)-6-chloropyridine-4-carboxylic acid (120 mg, 0.24 mmol, 1 equivalent), N,N-Diisopropylethylamine (94 mg, 0.73 mmol, 3 equiv.) and N,N,N,N-Tetramethyl- OA mixture of -(7-azabenzotriazol-1-yl)uronium hexafluorophosphate (139 mg, 0.36 mmol, 1.5 equiv) in 1,4-dioxane (10 mL) was stirred at room temperature for 2 h under nitrogen atmosphere. The reaction was quenched with water, and the mixture was extracted with ethyl acetate. The combined organic layers were dried and concentrated under reduced pressure. The residue was purified by prep-HPLC using an XBridge Prep OBD C18 column containing 10 mmol/L NH4HCO 3The mixture was purified by using water and acetonitrile as the mobile phase. The fractions containing the desired compound were evaporated to dryness to obtain 3.9 mg of 30-chloro-11-oxa-8,9,17,24,26,31-hexaazahexacyclo[26.3.1.0^{2,10}.0^{3,8}.0^{17,25}.0^{18,23}]triacontidine-1(31),2,4,6,9,18,20,22,25, 28(32),29-undecane-27-one (Example B15) (3%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=473.10. 1H NMR (DMSO-d 6 , 400 MHz) δ 1.93-2.13 (6H, m), 4.24 (2H, t), 4.49 (2H, t), 6.96-7.02 (1H, m), 7.22-7.35 (2H, m), 7.48-7.61 (4H, m), 8.44 (1H, d), 8.67 (1H, d), 9.17 (1H, d), 12.76 (1H, s). Example B16 5,30-dimethyl-7-oxa-4,5,13,18,24,26,31-heptaazahexacyclo[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]triacontac-1(31),2(6),3,14,20,22,24,28(32),29-nonan-27-one Step 1. Preparation of tert-butyl 5-((5-((4-(4-(methoxycarbonyl)-6-methylpyridin-2-yl)-1-methyl-1H-pyrazol-5-yl)oxy)pentyl)amino)-6-nitro-3,4-dihydroisoquinoline-2(1H)-carboxylate (INT-B16-1). To a stirred mixture of tert-butyl 5-[(5-hydroxypentyl)amino]-6-nitro-3,4-dihydro-1H-isoquinoline-2-carboxylate (1.6 g, 0.79 mmol, 1 eq), triphenylphosphine (3 g, 2.37 mmol, 3 eq) and methyl 2-(5-hydroxy-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylate (1 g, 0.79 mmol, 1 eq) in tetrahydrofuran (50 mL) was added diisopropyl azodicarboxylate (2.5 g, 1.19 mmol, 3 eq) under nitrogen atmosphere at 0°C. The resulting mixture was stirred at room temperature under nitrogen atmosphere for 3 h. The reaction was quenched with water (50 mL) and the mixture was extracted with dichloromethane (3×100 mL). The combined organic layers were washed with brine (3×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by silica gel chromatography using petroleum ether/ethyl acetate (1/1) to give 900 mg of 5-{[5-({4-[4-(methoxycarbonyl)-6-methylpyridin-2-yl]-2-methylpyrazol-3-yl}oxy)pentyl]amino}-6-nitro-3,4-dihydro-1H-isoquinoline-2-carboxylic acid tert-butyl ester (INT-B16-1) (36%) as a yellow oil. LCMS: m/z (ESI), [M+H] +=609.20. Step 2. Preparation of tert-butyl 6-amino-5-((5-((4-(4-(methoxycarbonyl)-6-methylpyridin-2-yl)-1-methyl-1H-pyrazol-5-yl)oxy)pentyl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (INT-B16-2). A mixture of tert-butyl 5-{[5-({4-[4-(methoxycarbonyl)-6-methylpyridin-2-yl]-2-methylpyrazol-3-yl}oxy)pentyl]amino}-6-nitro-3,4-dihydro-1H-isoquinoline-2-carboxylate (900 mg, 1.48 mmol, 1 eq) and Pd/C (15.73 mg, 0.148 mmol, 0.1 eq) in methanol (10 mL) was stirred under hydrogen atmosphere at room temperature for 1 h. The resulting mixture was filtered, and the filter cake was washed with methanol (3×10 mL). The resulting solution was concentrated under reduced pressure to give 570 mg of tert-butyl 6-amino-5-((5-((4-(4-(methoxycarbonyl)-6-methylpyridin-2-yl)-1-methyl-1H-pyrazol-5-yl)oxy)pentyl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (INT-B16-2) (67%) as a yellow oil. The crude product mixture was used directly in the next step without further purification. LCMS: m/z (ESI), [M+H] +=579.25. Step 3. Preparation of tert-butyl 2-amino-1-(5-((4-(4-(methoxycarbonyl)-6-methylpyridin-2-yl)-1-methyl-1H-pyrazol-5-yl)oxy)pentyl)-1,6,8,9-tetrahydro-7H-imidazo[4,5-f]isoquinoline-7-carboxylate (INT-B16-3). A mixture of tert-butyl 6-amino-5-{[5-({4-[4-(methoxycarbonyl)-6-methylpyridin-2-yl]-2-methylpyrazol-3-yl}oxy)pentyl]amino}-3,4-dihydro-1H-isoquinoline-2-carboxylate (570 mg, 0.99 mmol, 1 eq) and BrCN (115 mg, 1.08 mmol, 1.1 eq) in methanol (20 mL) was stirred at room temperature under nitrogen atmosphere for 1 h. The reaction was quenched with water (20 mL), and the mixture was extracted with dichloromethane (3×50 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by silica gel chromatography using dichloromethane/methanol (20/1) to give 500 mg of methyl 2-[5-({5-[2-amino-7-(tert-butoxycarbonyl)-6H,8H,9H-imidazo[4,5-f]isoquinolin-1-yl]pentyl}oxy)-1-methylpyrazol-4-yl]-6-methylpyridine-4-carboxylate (INT-B16-3) (84%) as an off-white solid. LCMS: m/z (ESI), [M+H] +=604.20. Step 4. Preparation of 2-(5-((5-(2-amino-7-(tert-butoxycarbonyl)-6,7,8,9-tetrahydro-1H-imidazo[4,5-f]isoquinolin-1-yl)pentyl)oxy)-1-methyl-1H-pyrazol-4-yl)-6-methylisonicotinic acid (INT-B16-4). 2-[5-({5-[2-amino-7-(tert-butoxycarbonyl)-6H,8H,9H-imidazo[4,5-f]isoquinolin-1-yl]pentyl}oxy)-1-methylpyrazol-4-yl]-6-methylpyridine-4-carboxylic acid methyl ester (500 mg, 0.828 mmol, 1 equivalent) and LiOH .H 2O (59.50 mg, 2.484 mmol, 3 equiv.) in MeOH (10 mL) and H 2The mixture in 4% O (5 mL) was stirred at room temperature under nitrogen atmosphere for 1 h and concentrated under reduced pressure. The residue was purified by prep-HPLC using an XBridge Shield RP18 OBD column with 10 mmol/L NH4HCO 3and 0.1% NH 3 .H 2O water and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 150 mg of 2-[5-({5-[2-amino-7-(tert-butoxycarbonyl)-6H,8H,9H-imidazo[4,5-f]isoquinolin-1-yl]pentyl}oxy)-1-methylpyrazol-4-yl]pyridine-4-carboxylic acid (INT-B16-4) (31%) as a white solid. LCMS: m/z (ESI), [M+H] +=590.25. Step 5. Preparation of tert-butyl 5,30-dimethyl-18-(tert-butoxycarbonyl)-7-oxa-4,5,13,18,24,26,31-heptaazahexacyclo[26.3.1.0^{2,6}. 0^{13,25}.0^{14,23}.0^{15,20}]triacontac-1(31),2(6),3,14,20, 22,24,28(32),29-nonan-27-one (INT-B16-5). 2-[5-({5-[2-amino-7-(tert-butoxycarbonyl)-6H,8H,9H-imidazo[4,5-f]isoquinolin-1-yl]pentyl}oxy)-1-methylpyrazol-4-yl]-6-methylpyridine-4-carboxylic acid (500 mg, 0.85 mmol, 1 equivalent), N,N-Diisopropylethylamine (219 mg, 1.7 mmol, 2 equivalents) and N,N,N,N-Tetramethyl- OA mixture of -(7-azabenzotriazol-1-yl)urea hexafluorophosphate (644.8 mg, 1.7 mmol, 2 equiv.) in dioxane (20 mL) was stirred at room temperature under nitrogen atmosphere for 2 h. The reaction was quenched with water (10 mL), and the mixture was extracted with dichloromethane (3×10 mL). The combined organic layers were washed with brine (3×10 mL), purified by anhydrous Na 2SO 4The residue was purified by silica gel column chromatography using dichloromethane/methanol (10/1) to give 200 mg of 5,30-dimethyl-18-(tert-butoxycarbonyl)-7-oxa-4,5,13,18,24,26,31-heptaazahexacyclo[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]triacontac-1(31),2(6),3,14,20,22,24, 28(32),29-nonan-27-one (INT-B16-5) (41%) as a brown solid. LCMS: m/z (ESI), [M+H] +=572.30. 1H NMR (CDCl 3, 400 MHz) δ 1.64 (17H, s), 1.95 (2H, d), 2.10-2.15 (2H, m), 2.68 (3H, s), 3.24 (2H, s), 3.80 (5H, d), 4.20-4.29 (2H, m), 4.38-4.47 (2H, m), 4.72 (2H, s), 7.03 (1H, d), 7.22 (1H, d), 7.69 (1H, s), 8.21 (1H, s), 8.49 (1H, s), 12.00 (1H, s). Step 6. Preparation of 5,30-dimethyl-7-oxa-4,5,13,18,24,26,31-heptaazahexacyclo[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]triacontac-1(31),2(6),3,14,20,22,24,28(32),29-nonan-27-one (Example B16). 1 1,2 6-Dimethyl-3-oxo-5- 6,5 7,5 8,5 9-Tetrahydrogen-1 1H,5 1H-11-oxa-4-aza-5(2,1)-imidazo[4,5-f]isoquinoline-2(2,4)-pyridine-1(4,5)-pyrazole cycloundecane-5 7A mixture of tert-butyl formate (30 mg, 0.090 mmol, 1 eq.) and HCl in 1,4-dioxane (5 mL) was stirred at room temperature under nitrogen atmosphere for 1 h. The mixture was washed with saturated NaHCO3(aqueous solution) was adjusted to pH 7, and the mixture was extracted with ethyl acetate (3×10 mL). The combined organic layers were washed with brine (3×10 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was subjected to prep-HPLC using an XBridge Shield RP18 OBD column with 10 mmol/L NH4HCO 3and 0.1% NH 3 .H 2O was used for purification with water and acetonitrile as the mobile phase. The fractions containing the desired compound were evaporated to dryness to give 1.5 mg of 5,30-dimethyl-7-oxa-4,5,13,18,24,26,31-heptaazahexacyclo[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]triacontac-1(31),2(6),3,14,20,22,24,28(32),29-nonan-27-one (Example B16) (0.37%) as a white solid. LCMS: m/z (ESI), [M+H] +=472.35. 1H NMR (DMSO, 400 MHz) δ 1.74-1.82 (2H, m), 1.96-2.10 (4H, m), 2.55 (3H, s), 3.02-3.16 (4H, m), 3.72 (3H, s), 3.95 (2H, s), 4.21 (2H, t), 4.35 (1H, d), 6.91 (1H, d), 7.33 (1H, d), 7.56 (1H, s), 7.93 (1H, s), 8.40 (1H, s). Example B17 5,30-Dimethyl-18-(oxacyclobutane-3-yl)-7-oxa-4,5,13,18,24,26, 31-heptaazahexacyclo[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15, 20}]triacontac-1(31),2(6),3,14,20,22,24,28(32),29-nonan-27-one Step 1. Preparation of 5,30-dimethyl-18-(oxacyclobutan-3-yl)-7-oxa-4,5,13,18,24,26,31-heptaazahexacyclo[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]triacontac-1(31),2(6),3,14,20,22,24, 28(32),29-nonan-27-one (Example B17). A stirred mixture of (25E)-5,30-dimethyl-7-oxa-4,5,13,18,24,26,31-heptaazahexacyclo[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]triacontria-1(31),2(6),3,14,20,22,25,28(32),29-nonan-27-one (Example B16, 30 mg, 0.06 mmol, 1 eq), 3-oxacyclobutanone (9.17 mg, 0.12 mmol, 2 eq) and trimethylamine (12.88 mg, 0.12 mmol, 2 eq) in dichloromethane (5 mL) was stirred under nitrogen atmosphere at room temperature for 30 minutes. Add NaBH(AcO) to the above mixture at room temperature 3(40.45 mg, 0.18 mmol, 3 equiv.). The resulting mixture was stirred at room temperature overnight under a nitrogen atmosphere. The reaction was quenched with water (10 mL), and the mixture was extracted with dichloromethane (3×10 mL). The combined organic layers were washed with brine (3×10 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was subjected to prep-HPLC using an XBridge Shield RP18 OBD column with 10 mmol/L NH4HCO 3and 0.1% NH 3.H 2O was used as the mobile phase for purification with water and acetonitrile. The fractions containing the desired compound were evaporated to dryness to give 5.8 mg of 5,30-dimethyl-18-(oxacyclobutane-3-yl)-7-oxa-4,5,13,18,24,26,31-heptaazahexacyclo[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]triacontac-1(31),2(6),3,14,20,22,24,28(32),29-nonan-27-one (Example B17) (17%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=528.20. 1H NMR (DMSO- d 6, 400 MHz) δ 1.71-1.85 (2H, m), 1.96-2.13 (4H, m), 2.56 (3H, s), 2.59-2.70 (2H, m), 3.22-3.30 (2H, m), 3.54 (2H, s), 3.57-3.65 (1H, m), 3.73 (3H, s), 4.21 (1H, t), 4.33 (2H, t), 4.55 (2H, t), 4.65 (2H, t), 6.96 (1H, d), 7.46 (1H, d), 7.57 (1H, s), 7.76 (1H, d), 7.98 (1H, s), 8.40 (1H, s). Example B18 5,30-Dimethyl-17-(oxacyclobutane-3-yl)-7-oxa-4,5,13,17,24,26, 31-heptaazahexacyclo[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}. 0^{15,20}]triacontac-1(31),2(6),3,14,20,22,24,28(32),29-nonan-27-one Step 1. Preparation of 2-(2-bromo-5-fluorophenyl)ethylamine (INT-B18-1). 2-(2-bromo-5-fluorophenyl)acetonitrile (10 g, 46.72 mmol, 1 equivalent) and BH 3-Me 2A mixture of S (10.65 g, 140.16 mmol, 3 equiv.) in tetrahydrofuran (100 mL) was stirred at 80 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (800 mL) at 0 °C, and the mixture was extracted with dichloromethane (3×200 mL). The combined organic layers were washed with brine (3×200 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue is purified by silica gel chromatography using dichloromethane/methanol (10/1) to give 8 g of 2-(2-bromo-5-fluorophenyl)ethylamine (Example INT-B18-1) (79%) as an off-white solid. LCMS: m/z (ESI), [M+H] += 220.10. Step 2. N-[2-(2-bromo-5-fluorophenyl)ethyl]-2,2,2-trifluoroacetamide (INT-B18-2) Preparation. A mixture of 2-(2-bromo-5-fluorophenyl)ethylamine (5 g, 23 mmol, 1 eq.), trifluoroacetic anhydride (5.3 g, 25 mmol, 1.1 eq.) and trimethylamine (4.64 g, 45.86 mmol, 2 eq.) in dichloromethane (100 mL) was stirred at 0 °C for 2 h under a nitrogen atmosphere. The reaction was quenched with water (200 mL), and the mixture was extracted with dichloromethane (3×200 mL). The combined organic layers were washed with brine (3×200 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by silica gel column chromatography eluting with petroleum ether/ethyl acetate (10/1) to obtain 4 g of white solid.N-[2-(2-bromo-5-fluorophenyl)ethyl]-2,2,2-trifluoroacetamide (INT-B18-2) (56%). 1H NMR (DMSO-d 6, 400 MHz) δ 2.90-2.96 (2H, m), 3.45-3.50 (2H, m), 7.04-7.26 (2H, m), 7.65 (1H, d), 9.55 (1H, s). Step 3. Preparation of 1-(5-bromo-8-fluoro-3,4-dihydro-1H-isoquinolin-2-yl)-2,2,2-trifluoroethanone (INT-B18-3). N-[2-(2-bromo-5-fluorophenyl)ethyl]-2,2,2-trifluoroacetamide (6.8 g, 21.65 mmol, 1 equivalent) and paraformaldehyde (28.6 g, 64.95 mmol, 3 equivalents) in H 2SO 4A mixture of (25 ml) and AcOH (30 ml) was stirred at 0 °C overnight under nitrogen atmosphere. The reaction was quenched with water (200 mL) at 0 °C and adjusted to pH 7 with NaOH. The mixture was extracted with dichloromethane (3×200 mL). The combined organic layers were washed with brine (3×200 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by silica gel chromatography eluting with petroleum ether/ethyl acetate (1/1) to give 5 g of 1-(5-bromo-8-fluoro-3,4-dihydro-1H-isoquinolin-2-yl)-2,2,2-trifluoroethanone (71%) as an off-white solid. 1H NMR (DMSO-d 6, 400 MHz) δ 2.85-2.92 (2H, m), 3.85-3.95 (2H, m), 4.77 (2H, d), 7.17 (1H, d), 7.63 (1H, d). Step 4. Preparation of 1-(5-bromo-8-fluoro-7-nitro-3,4-dihydro-1H-isoquinolin-2-yl)-2,2,2-trifluoroethanone (INT-B18-4). 1-(5-bromo-8-fluoro-3,4-dihydro-1H-isoquinolin-2-yl)-2,2,2-trifluoroethanone (6.46 g, 19.81 mmol, 1 equivalent) and KNO 3(2 g, 19.81 mmol, 1 equivalent) in H 2SO 4The mixture in (20 mL) was stirred at 0 °C for 0.5 h under nitrogen atmosphere. The reaction was quenched with water (200 mL) and adjusted to pH 9 with NaOH. The mixture was extracted with dichloromethane (3×200 mL). The combined organic layers were washed with brine (3×200 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by silica gel column chromatography using petroleum ether/ethyl acetate (3/1) to give 775 mg of 1-(5-bromo-8-fluoro-7-nitro-3,4-dihydro-1H-isoquinolin-2-yl)-2,2,2-trifluoroethanone (INT-B18-4) (12%) as a brownish yellow solid. 1H NMR (DMSO-d 6, 400 MHz) δ 2.95-3.02 (2H, m), 3.81-3.96 (2H, m), 4.75-4.90 (2H, m), 8.07 (1H, t). Step 5. Preparation of 5-[(5-bromo-7-nitro-1,2,3,4-tetrahydroisoquinolin-8-yl)amino]pentan-1-ol (INT-B18-5). 1-(5-bromo-8-fluoro-7-nitro-3,4-dihydro-1H-isoquinolin-2-yl)-2,2,2-trifluoroethanone (4.6 g, 12.4 mmol, 1 equivalent), K 2CO 3A mixture of (5.14 g, 37.2 mmol, 3 equiv.) and 5-aminopentanol (1.53 g, 14.88 mmol, 1.2 equiv.) in acetonitrile (50 mL) was stirred at 60 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature. The reaction was quenched with water (100 mL), and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by silica gel column chromatography eluting with dichloromethane/methanol (10/1) to give 600 mg of 5-[(5-bromo-7-nitro-1,2,3,4-tetrahydroisoquinolin-8-yl)amino]pentan-1-ol (INT-B18-5) (14%) as a yellow oil. LCMS: m/z (ESI), [M+H] +=360.00. Step 6. Preparation of tert-butyl 5-bromo-8-((5-hydroxypentyl)amino)-7-nitro-3,4-dihydroisoquinoline-2(1H)-carboxylate (INT-B18-6). A mixture of 5-[(5-bromo-7-nitro-1,2,3,4-tetrahydroisoquinolin-8-yl)amino]pentan-1-ol (650 mg, 1.81 mmol, 1 eq.), di-tert-butyl dicarbonate (475 mg, 2.18 mmol, 1.2 eq.) and trimethylamine (367 mg, 3.63 mmol, 2 eq.) in dichloromethane (20 mL) was stirred at room temperature overnight under a nitrogen atmosphere. The reaction mixture was concentrated under reduced pressure. The residue was purified by C18 FLASH using water and acetonitrile as mobile phase. The fractions containing the desired compound were evaporated to dryness to give 450 mg of tert-butyl 5-bromo-8-((5-hydroxypentyl)amino)-7-nitro-3,4-dihydroisoquinoline-2(1H)-carboxylate (INT-B18-6) (54%) as a yellow oil. LCMS: m/z (ESI), [M+H] +=460.25. Step 7. Preparation of tert-butyl 5-bromo-8-{[5-({4-[4-(methoxycarbonyl)-6-methylpyridin-2-yl]-2-methylpyrazol-3-yl}oxy)pentyl]amino}-7-nitro-3,4-dihydro-1H-isoquinoline-2-carboxylate (INT-B18-7). To a stirred mixture of tert-butyl 5-bromo-8-((5-hydroxypentyl)amino)-7-nitro-3,4-dihydroisoquinoline-2(1H)-carboxylate (450 mg, 0.98 mmol, 1 eq.), triphenylphosphine (772 mg, 2.95 mmol, 3 eq.) in tetrahydrofuran (20 mL) under nitrogen atmosphere at 0° C. was added methyl 2-(5-hydroxy-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylate (267 mg, 1.08 mmol, 1.1 eq.) and diisopropyl azodicarboxylate (595 mg, 2.95 mmol, 3 eq.). The reaction mixture was stirred at 60° C. for 3 h under nitrogen atmosphere and cooled to room temperature. The reaction was quenched with water (100 mL), and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue is purified by C18 FLASH using water and acetonitrile as mobile phase. The fractions containing the desired compound are evaporated to dryness to give 350 mg of 5-bromo-8-{[5-({4-[4-(methoxycarbonyl)-6-methylpyridin-2-yl]-2-methylpyrazol-3-yl}oxy)pentyl]amino}-7-nitro-3,4-dihydro-1H-isoquinoline-2-carboxylic acid tert-butyl ester (INT-B18-7) (52%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=689.05. Step 8. Preparation of tert-butyl 7-amino-8-((5-((4-(4-(methoxycarbonyl)-6-methylpyridin-2-yl)-1-methyl-1H-pyrazol-5-yl)oxy)pentyl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (INT-B18-8). A mixture of tert-butyl 5-bromo-8-{[5-({4-[4-(methoxycarbonyl)-6-methylpyridin-2-yl]-2-methylpyrazol-3-yl}oxy)pentyl]amino}-7-nitro-3,4-dihydro-1H-isoquinoline-2-carboxylate (330 mg, 0.48 mmol, 1 eq) and Pd/C (5.1 mg, 0.048 mmol, 0.1 eq) in methanol (20 mL) was stirred under hydrogen atmosphere at room temperature for 1 h. The resulting mixture was filtered and the filter cake was washed with methanol (3×30 mL). The resulting solution was concentrated under reduced pressure to give 172 mg of tert-butyl 7-amino-8-((5-((4-(4-(methoxycarbonyl)-6-methylpyridin-2-yl)-1-methyl-1H-pyrazol-5-yl)oxy)pentyl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (INT-B18-8) as a yellow oil. This material was used in the next reaction without further purification. LCMS: m/z (ESI), [M+H] +=579.25. Step 9. Preparation of 2-[5-({5-[2-amino-5-bromo-8-(tert-butoxycarbonyl)-6H,7H,9H-imidazo[4,5-h]isoquinolin-1-yl]pentyl}oxy)-1-methylpyrazol-4-yl]-6-methylpyridine-4-carboxylic acid methyl ester (INT-B18-9). A mixture of tert-butyl 7-amino-8-((5-((4-(4-(methoxycarbonyl)-6-methylpyridin-2-yl)-1-methyl-1H-pyrazol-5-yl)oxy)pentyl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (135 mg, about 0.21 mmol, about 1 equivalent) and BrCN (33 mg, 0.31 mmol, 1.5 equivalents) in dichloromethane (5 mL) was stirred at room temperature overnight under a nitrogen atmosphere. The reaction was quenched with water (20 mL), and the mixture was extracted with dichloromethane (3×50 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by silica gel column chromatography and eluted with dichloromethane/methanol (10/1) to give 90 mg of 2-[5-({5-[2-amino-5-bromo-8-(tert-butoxycarbonyl)-6H,7H,9H-imidazo[4,5-h]isoquinolin-1-yl]pentyl}oxy)-1-methylpyrazol-4-yl]-6-methylpyridine-4-carboxylic acid methyl ester (INT-B18-9) (64%) as a light yellow oil. LCMS: m/z (ESI), [M+H] +=604.25. Step 10. Preparation of 2-[5-({5-[2-amino-8-(tert-butoxycarbonyl)-6H,7H,9H-imidazo[4,5-h]isoquinolin-1-yl]pentyl}oxy)-1-methylpyrazol-4-yl]-6-methylpyridine-4-carboxylic acid (INT-B18-10). 2-[5-({5-[2-amino-5-bromo-8-(tert-butoxycarbonyl)-6H,7H,9H-imidazo[4,5-h]isoquinolin-1-yl]pentyl}oxy)-1-methylpyrazol-4-yl]-6-methylpyridine-4-carboxylate (90 mg, 0.15 mmol, 1 equivalent) and LiOH .H 2O (19 mg, 0.45 mmol, 3 equiv.) in MeOH (2 ml) and H 2The mixture in 400 μl (0.5 ml) was stirred at room temperature under nitrogen atmosphere for 2 h. The reaction mixture was concentrated under reduced pressure. The residue was purified by C18 FLASH using water and acetonitrile as mobile phase. The fractions containing the desired compound were evaporated to dryness to give 50 mg of 2-[5-({5-[2-amino-8-(tert-butoxycarbonyl)-6H,7H,9H-imidazo[4,5-h]isoquinolin-1-yl]pentyl}oxy)-1-methylpyrazol-4-yl]-6-methylpyridine-4-carboxylic acid (INT-B18-10) (57%) as a white solid. LCMS: m/z (ESI), [M+H] +=590.50. Step 11. Preparation of tert-butyl 5,30-dimethyl-27-oxo-7-oxa-4,5,13,17,24,26, 31-heptaazahexacyclo[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15, 20}]triacontidine-1(31),2(6),3,14,20,22,24,28(32),29-nonene-17-carboxylate (INT-B18-11). 2-[5-({5-[2-amino-8-(tert-butoxycarbonyl)-6H,7H,9H-imidazo[4,5-h]isoquinolin-1-yl]pentyl}oxy)-1-methylpyrazol-4-yl]-6-methylpyridine-4-carboxylic acid (35 mg, 0.05 mmol, 1 equivalent), N,N-Diisopropylethylamine (19.26 mg, 0.15 mmol, 3 equivalents) and N,N,N,N-Tetramethyl- OA mixture of -(7-azabenzotriazol-1-yl)uronium hexafluorophosphate (58.03 mg, 0.15 mmol, 3 equiv.) in dioxane (2 mL) was stirred at room temperature under nitrogen atmosphere for 2 h. The reaction was quenched with water (10 mL), and the mixture was extracted with dichloromethane (3×10 mL). The combined organic layers were washed with brine (3×10 mL), purified by anhydrous Na 2SO 4The residue was purified by silica gel column chromatography and eluted with dichloromethane/methanol (10/1) to give 30 mg of 5,30-dimethyl-27-oxo-7-oxa-4,5,13,17,24, 26,31-heptaazahexacyclo[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]triacontac-1(31),2(6),3,14,20,22,24,28(32),29-nonene-17-carboxylic acid tert-butyl ester (INT-B18-11) (93%) as a white solid. LCMS: m/z (ESI), [M+H] +=670.55. Step 12. Preparation of 5,30-dimethyl-7-oxa-4,5,13,17,24,26,31-heptaazahexacyclo[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]triacontac-1(31),2(6),3,14,20,22,24,28(32),29-nonan-27-one (INT-B18-12). A mixture of tert-butyl 5,30-dimethyl-27-oxo-7-oxa-4,5,13, 17,24,26,31-heptaazahexacyclo[26.3.1.0^{2,6}.0^{13,25}.0^{14, 23}.0^{15,20}]triacontria-1(31),2(6),3,14,20,22,24,28(32),29-nonene-17-carboxylate (30 mg, 0.05 mmol, 1 eq.) in trifluoroacetic acid (1 mL) and dichloromethane (2 mL) was stirred at room temperature for 30 minutes. The resulting mixture was concentrated under reduced pressure. The residue was subjected to prep-HPLC using an XBridge Shield RP18 OBD column and a column containing 10 mmol/L NH4HCO 3and 0.1% NH 3 .H 2O was used for purification with water and acetonitrile as mobile phase. The fractions containing the desired compound were evaporated to dryness to give 2 mg of 5,30-dimethyl-7-oxa-4,5,13,17,24,26,31-heptaazahexacyclo[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]triacontac-1(31),2(6),3,14,20,22,24,28(32),29-nonan-27-one (INT-B18-12) (8%) as a white solid. LCMS: m/z (ESI), [M+H] +=472.15. 1H NMR (DMSO-d 6, 400 MHz) δ 1.79 (2H, s), 2.04 (4H, s), 2.56 (3H, s), 2.80 (2H, d), 2.97 (2H, t), 3.73 (3H, s), 4.20-4.29 (4H, m), 4.32 (2H, s), 6.95 (1H, d), 7.34 (1H, d), 7.57 (1H, d), 7.92 (1H, s), 8.39 (1H, s). Step 13. Preparation of 5,30-dimethyl-17-(oxacyclobutan-3-yl)-7-oxa-4,5, 13,17,24,26,31-heptaazahexacyclo[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]triacontac-1(31),2(6),3,14,20,22,24, 28(32),29-nonan-27-one (Example B18). A mixture of 5,30-dimethyl-7-oxa-4,5,13,17,24,26,31-heptaazahexacyclo[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]triacontria-1(31),2(6),3,14,20, 22,24,28(32),29-nonan-27-one (7 mg, 0.015 mmol, 1 eq), 3-oxacyclobutanone (1.28 mg, 0.018 mmol, 1.2 eq) and trimethylamine (3 mg, 0.030 mmol, 2 eq) in dichloromethane (2 mL) was stirred under nitrogen atmosphere at room temperature for 30 min. Sodium acetoxyborohydride (9.44 mg, 0.045 mmol, 3 equiv.) was added to the above mixture in portions at room temperature. The resulting mixture was stirred at room temperature overnight under a nitrogen atmosphere. The reaction was quenched with water (10 mL), and the mixture was extracted with dichloromethane (3×10 mL). The combined organic layers were washed with brine (3×10 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The resulting mixture was concentrated under reduced pressure. The residue was passed through prep-HPLC using an XBridge Shield RP18 OBD column, using a HPLC-MS/HPLC-MS/HPLC-MS containing 10 mmol/L NH4HCO 3and 0.1% NH 3 .H 2O was used for purification with water and acetonitrile as the mobile phase. The fractions containing the desired compound were evaporated to dryness to give 1.8 mg of 5,30-dimethyl-17-(oxacyclobutane-3-yl)-7-oxa-4,5,13,17,24,26,31-heptaazahexacyclo[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]triacontac-1(31),2(6),3,14,20,22,24,28(32),29-nonan-27-one (Example B18) (23%) as a white solid. LCMS: m/z (ESI), [M+H] +=528.20. 1H NMR (DMSO-d 6, 400 MHz) δ 1.66-1.80 (2H, m), 1.86-2.11 (4H, m), 2.56 (3H, s), 2.87-2.98 (4H, m), 3.17 (2H, d), 3.73 (3H, s), 3.75-3.81 (1H, m), 4.04 (4H, s), 4.16-4.28 (4H, m), 4.61 (2H, t), 4.70 (2H, t), 7.01 (1H, d), 7.38 (1H, d), 7.57 (1H, s), 7.93 (1H, s), 8.40 (1H, s). Example B20 5,16,26-trimethyl-7-oxa-4,5,13,16,20,22,27-heptaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octadecene-1(27),2(6),3, 14(19),17,20,24(28),25-octaene-15,23-dione Step 1. Preparation of 4-methoxy-1-methyl-3-nitropyridin-2-one (INT-B20-1). 4-Hydroxy-3-nitro-1H-pyridin-2-one (10 g, 64.0 mmol, 1 equivalent), iodomethane (36.4 g, 256.2 mmol, 4 equivalents) and K 2CO 3(35.4 g, 256.2 mmol, 4 equivalents) in N,N-The mixture in dimethylformamide (100 mL) was stirred at 60 °C overnight under nitrogen atmosphere and cooled to room temperature. The reaction was quenched with water (500 mL), and the mixture was extracted with dichloromethane (3×500 mL). The combined organic layers were washed with brine (3×500 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue is purified by C18 FLASH using water and acetonitrile as mobile phase. The fractions containing the desired compound are evaporated to dryness to give 1600 mg of 4-methoxy-1-methyl-3-nitropyridin-2-one (INT-B20-1) (13%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=185.15. 1H NMR (DMSO-d 6, 400 MHz) δ 3.48 (3H, s), 3.96 (3H, s), 6.50 (1H, d), 8.03 (1H, d). Step 2. Preparation of 4-amino-1-methyl-3-nitropyridine-2-one (INT-B20-2). 4-Methoxy-1-methyl-3-nitropyridine-2-one (800 mg, 4.34 mmol, 1 equivalent) and NH 3The mixture in methanol (20 mL) was stirred at room temperature for 4 h under nitrogen atmosphere. The precipitated solid was collected by filtration and washed with methanol (3×40 mL) to obtain 630 mg of 4-amino-1-methyl-3-nitropyridin-2-one (INT-B20-2) (85%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=170.05. 1H NMR (DMSO-d 6, 400 MHz) δ 3.28 (3H, s), 5.87 (1H, d), 7.55 (1H, d), 8.03 (2H, s). Step 3. Preparation of 3,4-diamino-1-methylpyridin-2-one (INT-B20-3). A mixture of 4-amino-1-methyl-3-nitropyridin-2-one (1 g, 5.91 mmol, 1 eq.) and Pd/C (0.31 g, 2.91 mmol, 0.49 eq.) in methanol (30 mL) was stirred at room temperature for 4 h under hydrogen atmosphere. The resulting mixture was filtered, and the filter cake was washed with methanol (3×40 mL). The organic solution was concentrated under reduced pressure to give 900 mg of 3,4-diamino-1-methylpyridin-2-one (INT-B20-3) as a black solid. This material was used in the next reaction without further purification. LCMS: m/z (ESI), [M+H] += 140.10. Step 4. Preparation of 2-amino-5-methyl-3H-imidazo[4,5-c]pyridin-4-one (INT-B20-4). A mixture of 3,4-diamino-1-methylpyridin-2-one (INT-B20-3, 800 mg, 5.74 mmol, 1 eq.) and BrCN (730.71 mg, 6.89 mmol, 1.20 eq.) in ethanol (15 mL) was stirred at room temperature for 3 h under nitrogen atmosphere. The resulting mixture was concentrated under vacuum. The crude product mixture was used directly in the next step without further purification. LCMS: m/z (ESI), [M+H] +=165.15. Step 5. Preparation of 2-amino-3-(5-bromopentyl)-5-methylimidazo[4,5-c]pyridin-4-one (INT-B20-5). 2-amino-5-methyl-3H-imidazo[4,5-c]pyridin-4-one (INT-B20-4, 0.8 g, 4.87 mmol, 1 equivalent), 1,5-dibromopentane (5.6 g, 24.36 mmol, 5 equivalents) and K 2CO 3A mixture of (1.68 g, 12.13 mmol, 2.49 equiv) in acetonitrile (15 mL) was stirred at 60 °C for 2 h under nitrogen atmosphere and cooled to room temperature. The reaction was quenched with water (100 mL), and the mixture was extracted with ethyl acetate (3×60 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using ethyl acetate to give 230 mg of 2-amino-3-(5-bromopentyl)-5-methylimidazo[4,5-c]pyridin-4-one (INT-B20-5) (15%) as a white solid. LCMS: m/z (ESI), [M+H] +=313.10. 1H NMR (DMSO-d 6, 400 MHz) δ 1.38-1.52 (2H, m), 1.53-1.65 (2H, m), 1.74-1.85 (2H, m), 3.07-3.20 (2H, m), 3.28 (3H, s), 3.48-3.56 (2H, m), 5.76 (1H, d), 6.39 (2H, s), 7.34 (1H, d). Step 6. Preparation of 2-{5-[(5-{2-amino-5-methyl-4-oxoimidazolo[4,5-c]pyridin-3-yl}pentyl)oxy]-1-methylpyrazol-4-yl}-6-methylpyridine-4-carboxylic acid methyl ester (INT-B20-6). 2-Amino-3-(5-bromopentyl)-5-methylimidazo[4,5-c]pyridin-4-one (350 mg, 1.11 mmol, 1 equivalent), 2-(5-hydroxy-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylic acid methyl ester (M1, 303 mg, 1.23 mmol, 1.10 equivalent) and K 2CO 3(386 mg, 2.79 mmol, 2.5 equiv) in acetonitrile (5 mL) was stirred at 60 °C for 4 h under nitrogen atmosphere and then cooled to room temperature. The reaction was quenched with water (100 mL), and the mixture was extracted with dichloromethane (3×60 mL). The combined organic layers were washed with brine (3×50 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by Prep-TLC using dichloromethane/methanol (15/1) to give 160 mg of 2-{5-[(5-{2-amino-5-methyl-4-oxoimidazolo[4,5-c]pyridin-3-yl}pentyl)oxy]-1-methylpyrazol-4-yl}-6-methylpyridine-4-carboxylic acid methyl ester (INT-B20-6) (29%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=480.15. Step 7. Preparation of 2-{5-[(5-{2-amino-5-methyl-4-oxoimidazolo[4,5-c]pyridin-3-yl}pentyl)oxy]-1-methylpyrazol-4-yl}-6-methylpyridine-4-carboxylic acid (INT-B20-7). 2-{5-[(5-{2-amino-5-methyl-4-oxoimidazolo[4,5-c]pyridin-3-yl}pentyl)oxy]-1-methylpyrazol-4-yl}-6-methylpyridine-4-carboxylic acid methyl ester (160 mg, 0.33 mmol, 1 equivalent) and LiOH .H 2O (28 mg, 0.66 mmol, 2 equiv.) in THF (1 mL) and H 2O (0.25 mL) was stirred at room temperature for 2 h. The resulting mixture was concentrated under reduced pressure. The residue was purified by reverse flash chromatography using acetonitrile and water to give 130 mg of 2-{5-[(5-{2-amino-5-methyl-4-oxoimidazolo[4,5-c]pyridin-3-yl}pentyl)oxy]-1-methylpyrazol-4-yl}-6-methylpyridine-4-carboxylic acid (INT-B20-7) (83%) as a white solid. LCMS: m/z (ESI), [M+H] +=466.10. Step 8. Preparation of 5,16,26-trimethyl-7-oxa-4,5,13,16,20,22,27-heptaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octadecene-1(27),2(6),3,14(19),17,20,24(28),25-octaene-15,23-dione (Example B20). 2-{5-[(5-{2-amino-5-methyl-4-oxoimidazolo[4,5-c]pyridin-3-yl}pentyl)oxy]-1-methylpyrazol-4-yl}-6-methylpyridine-4-carboxylic acid (105 mg, 0.22 mmol, 1 equivalent), N,N,N,N-Tetramethyl-O-(7-azabenzotriazol-1-yl)urea hexafluorophosphate (128 mg, 0.33 mmol, 1.5 equivalents) and N,N- A mixture of diisopropylethylamine (87 mg, 0.67 mmol, 3 equiv.) in 1,4-dioxane (3 mL) was stirred at room temperature overnight under nitrogen atmosphere. The reaction was quenched with water (60 mL), and the mixture was extracted with dichloromethane (3×40 mL). The combined organic layers were washed with brine (3×30 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by trituration with DMF/DMSO (5 mL) to give 15 mg of 5,16,26-trimethyl-7-oxa-4,5,13,16,20,22,27-heptaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octacosa-1(27),2(6),3,14(19),17,20,24(28),25-octaene-15,23-dione (Example B20) as a yellow solid (13%). LCMS: m/z (ESI), [M+H] +=448.10. 1H NMR (DMSO-d 6, 400 MHz) δ 1.66-1.80 (2H, m), 1.86-2.11 (4H, m), 2.55 (3H, s), 3.53 (3H, s), 3.73 (3H, s), 4.40 (2H, t), 4.49 (2H, t), 6.58 (1H, d), 7.55 (1H, s), 7.60 (1H, d), 7.92 (1H, s), 8.38 (1H, s), 12.87 (1H, s). Example B21 5,16,26-trimethyl-7-oxa-4,5,13,16,17,20,22,27-octaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octadecene-1(27),2(6),3, 14(19),17,20,24(28),25-octaene-15,23-dione Step 1. Preparation of 4-hydroxy-2-methyl-5-nitrooxazin-3-one (INT-B21-1). 4,5-dichloro-2-methyloxazin-3-one (4.0 g, 22.34 mmol, 1 equivalent) and NaNO 2(6.1 g, 89.38 mmol, 4.0 equivalents) in N,N- A mixture in dimethylformamide (20 mL) was stirred at 90 °C for 24 h under nitrogen atmosphere. The mixture was cooled to room temperature and concentrated under reduced pressure. The residue was redissolved in warm 4 M HCl (20 mL) and cooled to room temperature. The solid was filtered and triturated with ether (100 mL) to give 1.0 g of 4-hydroxy-2-methyl-5-nitrooxazin-3-one (INT-B21-1) (26%) as a yellow solid. 1H NMR (DMSO-d 6, 400 MHz) δ 3.56 (3H, s), 8.20 (1H, s). Step 2. Preparation of 4-amino-2-methyl-5-nitrooxazin-3-one (INT-B21-2). 4-Hydroxy-2-methyl-5-nitrooxazin-3-one (2.5 g, 14.61 mmol, 1 equivalent) and NH 3The mixture in methanol (60 mL) was stirred at 60 °C for 20 h under nitrogen atmosphere. The mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by C18 FLASH using water and acetonitrile as mobile phase. The fractions containing the desired compound were evaporated to dryness to obtain 800 mg of 4-amino-2-methyl-5-nitrooxazin-3-one (INT-B21-2) (32%) as a yellow solid. 1H NMR (400 MHz, DMSO-d 6) δ 3.60 (s, 3H), 8.26 (s, 1H), 8.83 (s, 2H). Step 3. Preparation of 4,5-diamino-2-methyloxazin-3-one (INT-B21-3). A mixture of 4-amino-2-methyl-5-nitrooxazin-3-one (340 mg, 2.00 mmol, 1 eq.) and Pd/C (106 mg, 1.00 mmol, 0.5 eq.) in methanol (15 mL) was stirred at room temperature for 3 h under a hydrogen atmosphere. The resulting mixture was filtered, and the filter cake was washed with methanol (50 mL). The solution was concentrated under reduced pressure to give 200 mg of 4,5-diamino-2-methyloxazin-3-one (INT-B21-3) (71%) as a light yellow solid. This material was used in the next reaction without purification. LCMS: m/z (ESI), [M+H] += 141.00. Step 4. Preparation of 2-amino-5-methyl-3H-imidazo[4,5-d]oxazin-4-one (INT-B21-4). A mixture of 4,5-diamino-2-methyloxazin-3-one (160 mg, 1.14 mmol, 1 eq.) and cyanogen bromide (181 mg, 1.71 mmol, 1.5 eq.) in ethanol (5 mL) was stirred at room temperature for 16 h under nitrogen atmosphere. The mixture was concentrated under reduced pressure, and the residue was purified on Prep-TLC using dichloromethane/methanol (10/1) to give 160 mg of 2-amino-5-methyl-3H-imidazo[4,5-d]oxazin-4-one (INT-B21-4) (59%) as a yellow solid. LCMS: m/z (ESI), [M+H] += 166.00. Step 5. Preparation of 2-amino-3-(5-bromopentyl)-5-methylimidazo[4,5-d]oxazin-4-one (INT-B21-5). 2-amino-5-methyl-3H-imidazo[4,5-d]oxazin-4-one (100 mg, 0.60 mmol, 1 eq.), 1,5-dibromopentane (409 mg, 1.78 mmol, 3.0 eq.) and K 2CO 3A mixture of (246.02 mg, 1.78 mmol, 3.0 equiv) in acetonitrile (10 mL) was stirred at 60 °C for 18 h under nitrogen atmosphere and cooled to room temperature. The reaction was quenched with water (30 mL), and the mixture was extracted with ethyl acetate (3×30 mL). The combined organic layers were washed with brine (3×30 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue is purified by C18FLASH using water and acetonitrile as mobile phase. The fractions containing the desired compound are evaporated to dryness to give 70 mg of 2-amino-3-(5-bromopentyl)-5-methylimidazo[4,5-d]oxazin-4-one (INT-B21-5) (37%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=315.95. Step 6. Preparation of 2-{5-[(5-{2-amino-6-methyl-7-oxoimidazo[4,5-d]oxazin-1-yl}pentyl)oxy]-1-methylpyrazol-4-yl}-6-methylpyridine-4-carboxylic acid methyl ester (INT-B21-6). 2-amino-3-(5-bromopentyl)-5-methylimidazo[4,5-d]oxazin-4-one (55 mg, 0.17 mmol, 1 equivalent), 2-(5-hydroxy-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylic acid methyl ester (M1, 65 mg, 0.26 mmol, 1.5 equivalents) and K 2CO 3A mixture of (72 mg, 0.52 mmol, 3.0 equiv) in acetonitrile (2.0 mL) was stirred at 60 °C for 3 h under nitrogen atmosphere and then cooled to room temperature. The reaction was quenched with water (30 mL), and the mixture was extracted with ethyl acetate (3×30 mL). The combined organic layers were washed with brine (3×30 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by C18 FLASH using water and acetonitrile as mobile phase. The fractions containing the desired compound were evaporated to dryness to give 25 mg of 2-{5-[(5-{2-amino-6-methyl-7-oxoimidazolo[4,5-d]oxazin-1-yl}pentyl)oxy]-1-methylpyrazol-4-yl}-6-methylpyridine-4-carboxylic acid methyl ester (INT-B21-6) (29%) as a light yellow solid. LCMS: m/z (ESI), [M+H] +=481.20. Step 7. Preparation of 2-{5-[(5-{2-amino-6-methyl-7-oxoimidazolo[4,5-d]oxazin-1-yl}pentyl)oxy]-1-methylpyrazol-4-yl}-6-methylpyridine-4-carboxylic acid (INT-B21-7). Methyl 2-{5-[(5-{2-amino-6-methyl-7-oxoimidazolo[4,5-d]oxazin-1-yl}pentyl)oxy]-1-methylpyrazol-4-yl}-6-methylpyridine-4-carboxylate (25 mg, 0.05 mmol, 1 equivalent) and LiOH .H 2O (4.4 mg, 0.10 mmol, 2.0 equiv.) in methanol (0.5 mL) and H 2The mixture in 4% ethyl acetate (1.0 mL) was stirred at 20 °C for 2 h under nitrogen atmosphere. The mixture was concentrated under reduced pressure. The residue was purified by C18 FLASH using water and acetonitrile as mobile phase. The fractions containing the desired compound were evaporated to dryness to give 15 mg of 2-{5-[(5-{2-amino-6-methyl-7-oxoimidazolo[4,5-d]oxazin-1-yl}pentyl)oxy]-1-methylpyrazol-4-yl}-6-methylpyridine-4-carboxylic acid (INT-B21-7) (61%) as a light yellow solid. LCMS: m/z (ESI), [M+H] +=467.15. Step 8. Preparation of 5,16,26-trimethyl-7-oxa-4,5,13,16,17,20,22,27-octaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octadecene-1(27),2(6),3,14(19),17,20,24(28),25-octaene-15,23-dione (Example B21). 2-{5-[(5-{2-amino-6-methyl-7-oxoimidazolo[4,5-d]oxazin-1-yl}pentyl)oxy]-1-methylpyrazol-4-yl}-6-methylpyridine-4-carboxylic acid (15 mg, 0.03 mmol, 1 equivalent), N,N,N,N-Tetramethyl- O-(7-Azabenzotriazol-1-yl)urea hexafluorophosphate (24 mg, 0.06 mmol, 2.0 equiv) and N,NA mixture of 1-diisopropylethylamine (12 mg, 0.10 mmol, 3.0 equiv.) in 1,4-dioxane (1.0 mL) was stirred at room temperature under nitrogen atmosphere for 18 h. The mixture was concentrated under reduced pressure. The residue was treated with dimethyl sulfoxide (2 mL). The solid was collected by filtration and washed with methanol (3×5 mL) to give 14 mg of 5,16,26-trimethyl-7-oxa-4,5,13,16,17,20,22,27-octaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octadecano-1(27),2(6),3,14(19),17,20, 24(28),25-octaene-15,23-dione (Example B21) (99%) as a white solid. LCMS: m/z (ESI), [M+H] +=449.15. 1H NMR (CDCl 3, 400 MHz) δ 1.72-1.82 (2H, m), 2.00-2.14 (4H, m), 2.56 (3H, s), 3.73 (3H, s), 3.76 (3H, s), 4.20 (2H, t), 4.47 (2H, t), 7.56 (1H, s), 7.92 (1H, s), 8.22 (1H, s), 8.37 (1H, s). Example B22 16-(2-Hydroxyethyl)-5,26-dimethyl-7-oxa-4,5,13,16,20,22,27-heptaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octadecene-1(27),2(6),3,14(19),17,20,24(28),25-octaene-15,23-dione Step 1. Preparation of 4-(2-{[(tert-butyldimethylsilyl)oxy]amino}ethoxy)-1-(2-{[(tert-butyldimethylsilyl)oxy]amino}ethyl)-3-nitropyridin-2-one (INT-B22-1). 4-Hydroxy-3-nitro-1H-pyridin-2-one (7 g, 44.84 mmol, 1 equivalent), cesium carbonate (43.8 g, 134.53 mmol, 3 equivalents) and (2-bromoethoxy)(tert-butyl)dimethylsilane (42.9 g, 179.38 mmol, 4 equivalents) were added to N,N-The mixture in dimethylformamide (100 mL) was stirred at 60 °C for 12 h. After cooling to room temperature, the reaction was treated with water (600 mL), and the mixture was extracted with ethyl acetate (3×500 mL). The combined organic layers were washed with brine (3×500 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by silica gel column chromatography using petroleum ether/ethyl acetate (2/1) to give 8.6 g of 4-(2-{[(tert-butyldimethylsilyl)oxy]amino}ethoxy)-1-(2-{[(tert-butyldimethylsilyl)oxy]amino}ethyl)-3-nitropyridin-2-one (INT-B22-1) (37%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=473.30. 1H NMR (400 MHz, DMSO-d 6) δ 0-0.19 (12H, m), 0.69-0.97 (18H, m), 3.75-3.96 (4H, m), 3.98-4.15 (2H, m), 4.20-4.46 (2H, m), 6.47-6.61 (1H, m), 7.81-7.96 (1H, m). Step 2. Preparation of 4-amino-1-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-3-nitropyridine-2-one (INT-B22-2). 4-{2-[(tert-butyldimethylsilyl)oxy]ethoxy}-1-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-3-nitropyridin-2-one (8.6 g, 18.01 mmol, 1 equivalent) and NH 3The mixture in methanol (60 mL) was stirred at 60 °C for 12 h. After cooling to room temperature, the mixture was concentrated under reduced pressure to give 5.6 g of 4-amino-1-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-3-nitropyridin-2-one (INT-B22-2) (96%) as a yellow solid. This material was used in the next reaction without further purification. LCMS: m/z (ESI), [M+H] +=314.10. 1H NMR (400 MHz, DMSO-d 6) δ 0.05 (6H, s), 0.82 (9H, s), 3.72-3.78 (2H, m), 3.82-3.88 (2H, m), 5.84-5.89 (1H, m), 7.45 (1H, d), 8.04 (2H, s). Step 3. Preparation of 3,4-diamino-1-{2-[(tert-butyldimethylsilyl)oxy]ethyl}pyridin-2-one (INT-B22-3). To a stirred mixture of 4-amino-1-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-3-nitropyridin-2-one (4.4 g, 13.89 mmol, 1 eq.) and ranyl nickel (1.10 g, 12.79 mmol, 0.92 eq.) in methanol (200 mL) at 0 °C under nitrogen atmosphere was added hydrazine hydrate (4.26 g, 83.39 mmol, 6 eq.). The resulting mixture was stirred at room temperature for 4 h and filtered. The filter cake was washed with methanol (3×100 mL). The solution was concentrated under reduced pressure. The residue was purified by silica gel column chromatography using petroleum dichloromethane/methanol (10/1) to give 3.7 g of 3,4-diamino-1-{2-[(tert-butyldimethylsilyl)oxy]ethyl}pyridin-2-one (INT-B22-3) (93%) as a gray powder. LCMS: m/z (ESI), [M+H] +=284.15. 1H NMR (400 MHz, DMSO-d 6) δ 0.07 (6H, s), 0.82 (9H, s), 3.74 (2H, t), 3.80-3.89 (4H, m), 5.11 (2H, s), 5.73 (1H, d), 6.79(1H, d). Step 4. Preparation of 2-amino-5-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-3H-imidazo[4,5-c]pyridin-4-one (INT-B22-4). To a stirred mixture of 3,4-diamino-1-{2-[(tert-butyldimethylsilyl)oxy]ethyl}pyridin-2-one (3.7 g, 13.05 mmol, 1 eq) in ethanol (60 mL) was added cyanogen bromide (2.1 g, 19.58 mmol, 1.5 eq) at room temperature under nitrogen atmosphere. The resulting mixture was stirred at room temperature for 4 h and concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with dichloromethane/methanol (7/1) to give 6.6 g of 2-amino-5-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-3H-imidazo[4,5-c]pyridin-4-one (INT-B22-4) (95%) as a brown solid. LCMS: m/z (ESI), [M+H] +=309.25. 1H NMR (400 MHz, DMSO-d 6) δ 0.14 (6H, s), 0.71 (9H, s), 3.95 (2H, t), 4.36(2H, t), 6.73 (1H, d), 7.88 (1H, d), 8.03(2H, s). Step 5. Preparation of 2-amino-3-(5-bromopentyl)-5-{2-[(tert-butyldimethylsilyl)oxy]ethyl}imidazo[4,5-c]pyridin-4-one (INT-B22-5). 2-amino-5-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-3H-imidazo[4,5-c]pyridin-4-one (6.6 g, 21.40 mmol, 1 eq.) and K 2CO 3To a stirred mixture of 2-amino-3-(5-bromopentyl)-5-{2-[(tert-butyldimethylsilyl)oxy]ethyl}imidazo[4,5-c]pyridin-4-one (INT-B22-5) (21%) was added 1,5-dibromo-pentane (5.9 g, 42.79 mmol, 2 eq.) in acetonitrile (100 mL). The resulting mixture was stirred at 60 °C for 4 h. After cooling to room temperature, the mixture was filtered and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with dichloromethane/methanol (10/1) to give 2.2 g of 2-amino-3-(5-bromopentyl)-5-{2-[(tert-butyldimethylsilyl)oxy]ethyl}imidazo[4,5-c]pyridin-4-one (INT-B22-5) (21%) as a brown solid. LCMS: m/z (ESI), [M+H] +=457.20. 1H NMR (400 MHz, DMSO-d 6) δ 0.12 (6H, s), 0.80 (9H, s), 1.31-1.43 (2H,m), 1.62-1.68 (2H, m), 1.78-1.84 (2H, m), 3.51 (2H, t), 3.80 (2H, t), 3.99 (2H,t), 4.19 (2H, t), 6.22 (1H, d), 6.50 (2H, s), 7.16 (1H, d). Step 6. Preparation of methyl 2-(5-{[5-(2-amino-5-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-4-oxoimidazolo[4,5-c]pyridin-3-yl)pentyl]oxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylate (INT-B22-6). 2-amino-3-(5-bromopentyl)-5-{2-[(tert-butyldimethylsilyl)oxy]ethyl}imidazo[4,5-c]pyridin-4-one (2.2 g, 4.81 mmol, 1 equivalent) and K 2CO 3To a stirred mixture of 2-(5-hydroxy-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylic acid methyl ester (1.19 g, 4.81 mmol, 1 equiv.) (1.3 g, 9.62 mmol, 2 equiv.) in acetonitrile (20 mL) was added acetonitrile (5 mL). The resulting mixture was stirred at 60 °C for 12 h. After cooling to room temperature, the reaction was quenched with water (100 mL), and the mixture was extracted with ethyl acetate (3×100 mL). The combined organic layers were washed with brine (3×100 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified on a silica gel column eluting with dichloromethane/methanol (10/1) to give 2.5 g of methyl 2-(5-{[5-(2-amino-5-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-4-oxoimidazolo[4,5-c]pyridin-3-yl)pentyl]oxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylate (INT-B22-6) (80%) as a brown solid. LCMS: m/z (ESI), [M+H] +=610.30. Step 7. Preparation of 2-(5-{[5-(2-amino-5-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-4-oxoimidazolo[4,5-c]pyridin-3-yl)pentyl]oxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylic acid (INT-B22-7). Under nitrogen atmosphere at room temperature, methyl 2-(5-{[5-(2-amino-5-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-4-oxoimidazolo[4,5-c]pyridin-3-yl)pentyl]oxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylate (2.5 g, 4.00 mmol, 1 equivalent) was added to tetrahydrofuran (20 mL) and H 2Lithium hydroxide (336 mg, 8.01 mmol, 2 eq.) was added to the stirred mixture in 4% O (5 mL). The reaction mixture was stirred for 3 h. The resulting mixture was concentrated under reduced pressure and purified by reverse flash chromatography using 10% to 50% acetonitrile/water elution to give 2.0 g of 2-(5-{[5-(2-amino-5-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-4-oxoimidazolo[4,5-c]pyridin-3-yl)pentyl]oxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylic acid (INT-B22-7) (75%) as a brown solid. LCMS: m/z (ESI), [M+H] +=610.30. Step 8. Preparation of 16-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-5,26-dimethyl-7-oxa-4,5,13,16,20,22,27-heptaazapentacyclic[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octaconic-1(27),2(6),3, 14(19),17,20,24(28),25-octaene-15,23-dione (INT-B22-8). 2-(5-{[5-(2-amino-5-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-4-oxoimidazolo[4,5-c]pyridin-3-yl)pentyl]oxy}-1-methylpyrazol-4-yl)-6-methylpyridine-4-carboxylic acid (20 mg, 0.03 mmol, 1 equivalent) and N,N,N,N-Tetramethyl- O-(7-Azabenzotriazol-1-yl)urea hexafluorophosphate (18.71 mg, 0.05 mmol, 1.5 equivalents) was added to the stirred mixture in dioxane (4 mL) N,N-Diisopropylethylamine (12.72 mg, 0.10 mmol, 3 equiv.). After stirring at room temperature for 12 h, the reaction was quenched with water (100 mL), and the mixture was extracted with dichloromethane (2×80 mL). The combined organic layers were washed with brine (3×100 mL) and purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by prep-TLC using dichloromethane/methanol (10/1) to give 8 mg of 16-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-5,26-dimethyl-7-oxa-4,5,13,16,20,22,27-heptaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octacosa-1(27),2(6),3, 14(19),17,20,24(28),25-octaene-15,23-dione (INT-B22-8) (39%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=592.35. 1H NMR (400 MHz, CD 3OD) δ 0.09 (6H, s), 0.83 (9H, s), 1.51-1.63 (2H, m), 1.76-1.91 (4H, m), 2.56 (3H, d), 3.72 (3H, s), 3.89-3.96 (2H, m), 4.09-4.16 (4H, m), 4.25-4.35 (2H, m), 6.39 (1H, d), 7.25 (1H, d),7.51 (1H, d), 7.84 (1H, s), 7.88-7.93 (1H, m). Step 9. 16-(2-Hydroxyethyl)-5,26-dimethyl-7-oxo-4,5,13,16,20,22,27-heptaazapentacyclic [22.3.1.0^{2,6}.0^{13,21}. 0^{14,19}]octacosa-1(27),2(6),3,14(19),17,20,24(28),25-octaene-15,23-dione (Example B22). 16-{2-[(tert-butyldimethylsilyl)oxy]ethyl}-5,26-dimethyl-7-oxa-4,5,13,16,20,22,27-heptaazapentacyclo[22.3.1.0^{2,6}.0^{13,21}.0^{14,19}]octacosa-1(27),2(6),3,14(19),17,20,24(28),25-octaene-15,23-dione (40 mg, 0.068 mmol, 1 equivalent) and tetrabutylammonium fluoride (35.35 A mixture of 2-nitro-1-(2-nitro-4-yl)-6-nitropropene (6-nitropropene) (2.0 mg, 0.136 mmol, 2 eq.) in tetrahydrofuran (2 mL) was stirred at room temperature for 4 h. The resulting mixture was concentrated under reduced pressure. The residue was purified by reverse phase flash chromatography using 10% to 100% MeOH/water to give 7.7 mg of 16-(2-hydroxyethyl)-5,26-dimethyl-7-oxa-4,5,13,16,20,22,27-heptaazapentacyclo[22.3.1.0^{2,6}. 0^{13,21}.0^{14,19}]octacos-1(27),2(6),3,14(19),17,20, 24(28),25-octaene-15,23-dione (Example B22) as a yellow solid (21%). LCMS: m/z (ESI), [M+H] +=478.15. 1H NMR (CD 3OD, 400 MHz) δ 1.85-1.93 (2H, m), 2.11-2.23 (4H, m), 2.89 (3H, s), 3.87 (3H, s), 3.88-3.94 (2H, m), 4.22 (2H, t), 4.41 (2H, t), 4.60-4.69 (2H, m), 6.69 (1H, d), 7.59 (1H, d), 8.10 (1H, s), 8.36 (1H, s), 8.95 (1H, s). Example B23 18-[2-(dimethylamino)ethyl]-5,30-dimethyl-7-oxa-4,5,13, 18,24,26,31-heptaazahexacyclo[26.3.1.0^{2,6}.0^{13,25}.0^{14, 23}.0^{15,20}]triacontac-1(31),2(6),3,14,20,22,24,28(32),29-nonan-27-one Step 1. 18-[2-(dimethylamino)ethyl]-5,30-dimethyl-7-oxa-4,5,13,18,24,26,31-heptaazahexacyclo[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]triacontac-1(31),2(6),3,14,20,22,24, 28(32),29-nonan-27-one (Example B23). 5,30-dimethyl-7-oxa-4,5,13,18,24,26,31-heptaazahexacyclo[26.3.1.0^{2,6}.0^{13,25}. 0^{14,23}.0^{15,20}] 32-1(31),2(6),3,14,20,22,24, 28(32),29-nonan-27-one (Example B16, 50 mg, 0.10 mmol, 1 equivalent), (2-bromoethyl)dimethylamine (32 mg, 0.21 mmol, 2 equivalents), trimethylamine (21 mg, 0.21 mmol, 2 equivalents) and NaI (3.1 mg, 0.02 mmol, 0.2 equivalents) in N,N-dimethylformamide (2 mL) was stirred at 60 °C for 6 h. The mixture was cooled to room temperature. The reaction was quenched with water (20 mL), and the mixture was extracted with ethyl acetate (3×20 mL). The combined organic layers were washed with brine (3×20 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was subjected to prep-HPLC using an XBridge Prep OBD C18 column with 10 mmol/L NH4HCO 3and 0.1% NH 3 .H 2O water and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 5.4 mg of 18-[2-(dimethylamino)ethyl]-5,30-dimethyl-7-oxa-4,5,13,18,24,26,31-heptaazahexacyclo[26.3.1.0^{2,6}.0^{13,25}.0^{14,23}.0^{15,20}]triacontac-1(31),2(6),3,14,20,22,24,28(32),29-nonan-27-one (Example B23) as a white solid (8%). LCMS: m/z (ESI), [M+H] +=543.40. 1H NMR (DMSO- d 6, 400 MHz) δ 1.79-1.83 (2H, m), 2.02-2.06 (4H, m), 2.21 (6H, s), 2.56 (3H, s), 2.54-2.64 (2H, m), 2.77-2.81 (2H, m), 3.15-3.44 (4H, m), 3.67 (2H, s), 3.73 (3H, s), 4.22 (2H, t), 4.35 (2H, t), 6.94 (1H, d), 7.35 (1H, d), 7.57 (1H, s), 7.93 (1H, s), 8.41 (1H, s), 12.66 (1H, s). Example B27 5-[4-(dimethylamino)piperidin-1-yl]-15,21-dimethyl-23-oxo-2,9, 11,16,20,21,26-heptaazaheptacyclic[26.2.2.1^{1,26}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]triacontetrazol-3,5,7,9,13,15,17(34), 18(22),19-nonan-12-one Under nitrogen atmosphere at room temperature, 5-bromo-15,21-dimethyl-23-oxa-2,9,11,16,20,21,26-heptaazaheptacyclic [26.2.2.1^{1,26}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]triacontria-3,5,7,9,13,15,17(34), 18(22),19-nonan-12-one (INT-B2-5, 70 mg, 0.12 mmol, 1 equivalent) was added. N,N-dimethylpiperidin-4-amine (93 mg, 0.73 mmol, 6 equiv) and BrettPhos Pd G 3To a stirred mixture of 2-nitropropene (33 mg, 0.04 mmol, 0.3 eq.) and 1,4-dioxane (6 mL) was added LiHMDS (1.5 mL, 1.45 mmol, 12 eq.) dropwise. The mixture was stirred at 60 °C under nitrogen atmosphere for 2 h and cooled to room temperature. The reaction was heated with saturated NH4Cl(aq) (20 mL) was quenched at room temperature, and the mixture was washed with CH2Cl 2(3×50 mL) extraction. The combined organic layers were purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was purified by prep-TLC using dichloromethane/methanol (10:1). The product was purified by prep-HPLC using an XBridge Prep OBD C18 column containing 0.1% NH4HCO 3of water and acetonitrile. The fractions containing the desired compound were evaporated to dryness to give 20.4 mg of 5-[4-(dimethylamino)piperidin-1-yl]-15,21-dimethyl-23-oxa-2,9,11,16,20,21,26-heptaazaheptacyclo[26.2.2.1^{1,26}.1^{13, 17}.0^{2,10}.0^{3,8}.0^{18,22}]triacontria-3,5,7,9,13,15, 17(34),18(22), 19-nonan-12-one (Example B27) (26%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=624.30 1H NMR (DMSO-d 6, 400 MHz) δ 1.47-1.60 (2H, m), 1.88-2.04 (6H, m), 2.06-2.15 (3H, m), 2.21-2.28 (7H, m), 2.54 (3H, s), 2.61-2.71 (2H, m), 2.81 (2H, d), 2.86 (2H, d), 3.46 (4H, d), 3.56 (2H, d), 3.76 (3H, s), 4.38 (2H, t), 6.94 (1H, dd), 7.36 (1H, d), 7.48 (1H, d), 7.61 (1H, d), 7.89 (1H, s), 8.76 (1H, s), 13.20 (1H, s). Examples B28-B39 (Table 3) were prepared using the same reaction conditions described above for the synthesis of Example B27 from INT-B2-5. Example B40A and Example B40B 5-[4-(dimethylamino)piperidin-1-yl]-15,21-dimethyl-23-oxa-2,9, 11,16,20,21,26-heptaazaheptacyclic[24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]tritriacontah-3,5,7,9,13,15,17(33), 18(22),19-nonan-12-one (isomer 1, Example B40A) Under nitrogen atmosphere at room temperature, 5-bromo-15,21-dimethyl-23-oxa-2,9,11,16,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]tritriacontria-3,5,7,9,13,15,17(33), 18(22),19-nonan-12-one (isomer 1, INT-B9-5A, 80 mg, 0.142 mmol, 1 equivalent) was added.N,N-dimethylpiperidin-4-amine (54.71 mg, 0.426 mmol, 3 equiv) and BrettPhos Pd G 3To a stirred mixture of 2-nitropropene (38.68 mg, 0.043 mmol, 0.3 equiv) and 1,4-dioxane (8 mL) was added LiHMDS (1.420 mmol, 10 equiv) dropwise. The mixture was stirred at 60 °C for 1 h and then cooled to room temperature. The reaction was heated with saturated NH4Cl(aqueous solution) (10 mL) was quenched, and the mixture was extracted with dichloromethane (3×20 mL). The combined organic layers were washed with brine (3×30 mL), purified by anhydrous Na 2SO 4Dry and concentrate under reduced pressure. The residue was subjected to prep-HPLC using an XBridge Shield RP18 OBD column with 10 mmol/L NH4HCO 3and 0.1% NH 3.H 2O water and acetonitrile as mobile phase for purification. The fractions containing the desired compound were evaporated to dryness to give 33.2 mg of 5-[4-(dimethylamino)piperidin-1-yl]-15,21-dimethyl-23-oxa-2,9,11, 16,20,21,26-heptaazaheptacyclo[24.4.1.1^{1,28}.1^{13,17}.0^{2, 10}.0^{3,8}.0^{18,22}]tritriacontac-3,5,7,9,13,15,17(33), 18(22),19-nonan-12-one (isomer 1, Example B40A) (37%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=610.35. 1H NMR (DMSO- d 6, 400 MHz) δ 1.65-1.78 (6H, m), 1.87-1.93 (2H, m), 1.96-2.12 (1H, d), 2.25 (6H, s), 2.45-2.49 (2H, m), 2.51-2.55 (4H, m), 2.65-2.69 (4H, m), 3.20-3.26 (2H, m), 3.35-3.45 (1H, m), 3.61-3.68 (2H, m), 3.74 (4H, s), 4.56 (1H, t), 4.70 (1H, d), 6.90-7.02 (1H, m), 7.21 (1H, d), 7.42 (1H, d), 7.56 (1H, s), 7.94 (1H, s), 8.42 (1H, s), 12.69 (1H, s). SFC-HPLC, Rt = 8.811 minutes. 5-[4-(Dimethylamino)piperidin-1-yl]-15,21-dimethyl-23-oxo-2,9,11,16,20,21,26-heptaazaheptacyclic[24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]tritriacontahedral-3,5,7,9,13,15,17(33), 18(22), 19-nonan-12-one (isomer 2, Example B40B) Under nitrogen atmosphere at room temperature, 5-bromo-15,21-dimethyl-23-oxa-2,9,11,16,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,28}.1^{13,17}. 0^{2,10}.0^{3,8}.0^{18,22}]tritriacontria-3,5,7,9,13,15,17(33), 18(22),19-nonan-12-one (isomer 2, INT-B9-5B, 80 mg, 0.14 mmol, 1 equivalent) and N,N-dimethylpiperidin-4-amine (109 mg, 0.85 mmol, 6 equiv) and BrettPhos Pd G 3(38 mg, 0.04 mmol, 0.3 equiv) in 1,4-dioxane was added LiHMDS (3 mL, 3.00 mmol, 21.0 equiv) dropwise. The mixture was stirred at 60 °C for 1.5 h under nitrogen atmosphere. The mixture was cooled to room temperature and then quenched with saturated ammonium chloride (aqueous solution) (100 mL) at room temperature. The mixture was extracted with dichloromethane (3×150 mL). The combined organic layers were dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by prep-TLC eluting with dichloromethane/methanol (10/1). The product was purified by prep-HPLC using an XBridge Prep OBD C18 column (30*150 mm, 5μm) and water (10 mmol/L NH4HCO 3and 0.05% NH 3H 2O) Mobile phase A and acetonitrile mobile phase B were further purified to obtain 29.7 mg of 5-[4-(dimethylamino)piperidin-1-yl]-15,21-dimethyl-23-oxa-2,9,11,16,20,21,26-heptaazaheptacyclic [24.4.1.1^{1,28}.1^{13,17}.0^{2,10}.0^{3,8}.0^{18,22}]tritriacontac-3,5,7,9,13,15,17(33),18(22),19-nonan-12-one (isomer 2, Example B40B) (34%) as a yellow solid. LCMS: m/z (ESI), [M+H] +=610.30. 1H NMR (DMSO-d 6, 400 MHz) δ 1.51-1.75 (5H, m), 1.87-1.93 (2H, m), 2.03-2.12 (2H, m), 2.26 (6H, s), 2.45-2.49 (2H, m), 2.51-2.55 (4H, m), 2.72 (4H, d), 3.20-3.26 (2H, m), 3.35-3.45 (1H, m), 3.61-3.68 (2H, m), 3.74 (4H, s), 4.52-4.60 (1H, m), 4.70 (1H, d), 6.93 (1H, d), 7.21 (1H, s), 7.42 (1H, d), 7.57 (1H, s), 7.94 (1H, s), 8.42 (1H, s), 12.70 (1H, s). SFC-HPLC, Rt=7.034, ee=100%. Rt1=3.894, Rt2=4.686. Example B41A, Example B42A, Example B43A, Example B44A, Example B44A1, Example B44A2, Example B45A, Example B46A, Example B47A, Example B48A, Example B49A, Example B50A, Example B51A, Example B52A, Example B53A, Example B54A, Example B55A, Example B56A, Example B57A, Example B58A, Example B59A (Table 4) were prepared using similar reaction conditions described above for the synthesis of Example B40A from INT-9B-5A. Example B41B, Example B42B, Example B43B, Example B44B, Example B44B1, Example B44B2, Example B45B, Example B46B, Example B47B, Example B48B, Example B49B, Example B50B, Example B51B, Example B52B, Example B53B, Example B54B, Example B55B, Example B56B, Example B57B, Example B58B, Example B59B (Table 4) were prepared using similar reaction conditions described above for the synthesis of Example B40B from INT-9B-5B. Biological Examples Exemplary compounds disclosed herein have been characterized in one or more of the following biological assays. Biological Example 1: Inhibition of EGFR WT and mutant enzyme activity Recombinant EGFR wild-type, L858R, L858R/T790M, Exon19Del, Exon19Del/T790M, EGFR[T790M/C797S/L858R], and EGFR[T790M/C797S/d746-750] were purchased from Carna Bio. EGFR(d746-750 C797S) and EGFR(C797S L858R) were purchased from Signalchem. The inhibitory potency of compounds against these enzymes was assessed using a homogeneous time-resolved fluorescence method (HTRF, CisBio. Catalog No. 62TK0PEJ). The inhibitory effect of compounds was determined by measuring the enzymatic activity of EGFR enzyme phosphorylating 1 μM TK substrate-biotin in the presence of 1 mM 5'-adenosine triphosphate (ATP) and different concentrations of test compounds. The enzyme reaction buffer contained 5 mM MgCl2, 1 mM MnCl2, 0.01% CHAPS, 1 mM dithiothreitol (DTT), 0.5% dimethyl sulfoxide (DMSO) and 1X supplemental enzyme buffer. WT and mutant EGFR enzymes (Synogencon) were added to the plate containing inhibitors, and the kinase reaction was subsequently initiated by adding ATP and peptide substrate. After incubation for 1 h at room temperature, the reaction was terminated by adding a detection reagent mixture containing EDTA. Fluorescence was measured at 615 nm and 665 nm, respectively, with an excitation wavelength at 320 nm. The calculated 665 nm/615 nm signal ratio is proportional to the kinase activity. The compound concentration that produces 50% inhibition of the corresponding kinase (IC 50). Table 5 lists the data obtained through these measurements. Biological Example 2: Cell Proliferation Assay Ba/F3 cells expressing EGFR mutations were seeded at 5000 cells/well in RPMI1640 medium with 10% FBS in 384-well plates. PC-9 were seeded at 500 cells/well in RPMI1640 medium with 10% FBS in 384-well plates. After overnight incubation, the assay plates were dosed with a series of concentrations of compounds. In addition to dosing the assay plates, the day 0 plates were treated using the CellTiter-Glo® Luminescent Cell Viability Assay (Promega) to measure the number of viable cells (G0). The assay plates were further incubated for 72 hours, and the number of viable cells (G3) was measured. The proliferation percentage was calculated as follows: Proliferation% = 100 × (G3 value of sample well - G0 value) / (G3 value of DMSO control - G0 value). XLFit software was used to further calculate the 50% proliferation inhibition (GI) in the best fitting curve.50) concentrations of compounds in the presence of . Table 6 lists the data obtained from these assays. N/A = Not Available. The foregoing description is considered to be merely illustrative of the principles of the present disclosure. Further, since many modifications and variations will be apparent to those skilled in the art, it is not intended to limit the present invention to the exact construction and process shown as described above. Therefore, all suitable modifications and equivalents may be considered to fall within the scope of the present invention as defined by the appended claims.

Claims (61)

一種式(I)的化合物 (I) 或其藥學上可接受的鹽,其中: 環A選自由以下組成的組:環烷基、雜環基、芳基和雜芳基; 環B選自由以下組成的組:環烷基、雜環基、芳基和雜芳基; L 1選自由以下組成的組:鍵、烷基、烯基、炔基、雜烷基、雜烯基和雜炔基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基和所述雜炔基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基和氨基; L 2選自由以下組成的組:鍵、N(R A)、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個R B取代; 其中R A選自由以下組成的組:氫、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基、烷基和鹵代烷基; 其中每個R B獨立地選自由以下組成的組:羥基、鹵素、氰基、氨基、烷基、烯基、炔基、雜烷基、雜烯基、環烷基、雜環基、芳基和雜芳基; L 3選自由以下組成的組:鍵、烷基、烯基、炔基、雜烷基、雜烯基和雜炔基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基和所述雜炔基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基和氨基; L 4選自O、S或N(R C); 其中R C選自由以下組成的組:氫、烷基、烯基、炔基、鹵代烷基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基; 每個R 1獨立地選自由以下組成的組:羥基、鹵素、氰基、氨基、-N(R D) 2、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個R E取代; 其中每個R D獨立地選自由以下組成的組:氫、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、  -N(R F) 2或-OR G; 其中每個R E獨立地選自由以下組成的組:氫、羥基、鹵素、氰基、氨基、-N(R F) 2、-烷基-N(R F) 2、-C(O)OR G、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基和烷基; 其中R F和R G中的每一個獨立地選自由以下組成的組:烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基; 每個R 2獨立地選自由以下組成的組:氫、羥基、鹵素、氰基、氨基、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基、雜環基、芳基和雜芳基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基、所述雜環基、所述芳基和所述雜芳基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基、烷基和鹵代烷基; R 3選自由以下組成的組:氫、烷基、烯基、炔基、雜烷基、雜烯基、雜炔基、環烷基和雜環基,其中所述烷基、所述烯基、所述炔基、所述雜烷基、所述雜烯基、所述雜炔基、所述環烷基和所述雜環基任選地被一個或多個獨立地選自以下的基團取代:羥基、鹵素、氰基、氨基、烷基和鹵代烷基; m是0至5的整數;並且 n是0至4的整數。 A compound of formula (I) (I) or a pharmaceutically acceptable salt thereof, wherein: Ring A is selected from the group consisting of cycloalkyl, heterocyclic, aryl and heteroaryl; Ring B is selected from the group consisting of cycloalkyl, heterocyclic, aryl and heteroaryl; L1 is selected from the group consisting of a bond, an alkyl, an alkenyl, an alkynyl, a heteroalkyl, a heteroalkenyl and a heteroalkynyl, wherein the alkyl, the alkenyl, the alkynyl, the heteroalkyl, the heteroalkenyl and the heteroalkynyl are optionally substituted with one or more groups independently selected from the group consisting of a hydroxyl, a halogen, a cyano and an amino; L2 is selected from the group consisting of a bond, N(R A ), alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more RB ; wherein R A is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the group consisting of hydroxyl, halogen, cyano, amino, alkyl and halogenated alkyl; wherein each RB is independently selected from the group consisting of hydroxyl, halogen, cyano, amino, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, cycloalkyl, heterocyclo, aryl and heteroaryl; L wherein L is selected from the group consisting of: a bond, an alkyl, an alkenyl, an alkynyl, a heteroalkyl, a heteroalkenyl, and a heteroalkynyl, wherein the alkyl, the alkenyl, the alkynyl, the heteroalkyl, the heteroalkenyl, and the heteroalkynyl are optionally substituted with one or more groups independently selected from the group consisting of: a hydroxyl, a halogen, a cyano, and an amino group; L is selected from O, S, or N(R C ); wherein R C is selected from the group consisting of: hydrogen, an alkyl, an alkenyl, an alkynyl, a halogenated alkyl, a heteroalkyl, a heteroalkenyl, a heteroalkynyl, a cycloalkyl, a heterocyclo, an aryl, and a heteroaryl group; each R 1 is independently selected from the group consisting of: a hydroxyl, a halogen, a cyano, an amino group, -N(R D ) 2 , alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more R E ; wherein each R D is independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the group consisting of hydroxyl, halogen, cyano, -N( RF ) 2 or -ORG ; wherein each RE is independently selected from the group consisting of hydrogen, hydroxyl, halogen, cyano, amino, -N( RF ) 2 , -alkyl-N( RF ) 2 , -C(O) ORG , alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the following: hydroxyl, halogen, cyano, amino and alkyl; wherein each of RF and RG is independently selected from the group consisting of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl; each R R is independently selected from the group consisting of hydrogen, hydroxyl, halogen, cyano, amino, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclo, aryl and heteroaryl are optionally substituted with one or more groups independently selected from the group consisting of hydroxyl, halogen, cyano, amino, alkyl and halogenated alkyl; 3 is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl and heterocyclo, wherein the alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl and heterocyclo are optionally substituted with one or more groups independently selected from the group consisting of hydroxy, halogen, cyano, amino, alkyl and halogenated alkyl; m is an integer from 0 to 5; and n is an integer from 0 to 4. 如請求項1所述的化合物或其藥學上可接受的鹽,其中環A是芳基或雜芳基。The compound or pharmaceutically acceptable salt thereof as described in claim 1, wherein Ring A is an aryl group or a heteroaryl group. 如請求項2所述的化合物或其藥學上可接受的鹽,其中環A選自由以下組成的組:呋喃基、苯硫基、吡咯基、苯基、吡啶基、吡喃基、嘧啶基、噠嗪基、吡嗪基和四氫異喹啉基。The compound or a pharmaceutically acceptable salt thereof as described in claim 2, wherein Ring A is selected from the group consisting of furanyl, phenylthio, pyrrolyl, phenyl, pyridyl, pyranyl, pyrimidinyl, oxazinyl, pyrazinyl and tetrahydroisoquinolinyl. 如請求項3所述的化合物或其藥學上可接受的鹽,其中環A選自由以下組成的組: The compound or a pharmaceutically acceptable salt thereof as described in claim 3, wherein Ring A is selected from the group consisting of: , , , , , , , , , and . 如請求項1所述的化合物或其藥學上可接受的鹽,其中環B是芳基或雜芳基。The compound or pharmaceutically acceptable salt thereof as described in claim 1, wherein Ring B is an aryl group or a heteroaryl group. 如請求項5所述的化合物或其藥學上可接受的鹽,其中環B是苯基、吡啶基或吡唑基。The compound or pharmaceutically acceptable salt thereof as described in claim 5, wherein Ring B is phenyl, pyridyl or pyrazolyl. 如請求項6所述的化合物或其藥學上可接受的鹽,其中環B選自由以下組成的組: The compound or a pharmaceutically acceptable salt thereof as described in claim 6, wherein Ring B is selected from the group consisting of: , , , , and . 如請求項1所述的化合物或其藥學上可接受的鹽,其中L 1是鍵。 The compound or pharmaceutically acceptable salt thereof as described in claim 1, wherein L 1 is a bond. 如請求項8所述的化合物或其藥學上可接受的鹽,其中L 1是烷基。 The compound or pharmaceutically acceptable salt thereof as described in claim 8, wherein L 1 is alkyl. 如請求項9所述的化合物或其藥學上可接受的鹽,其中L 1選自由以下組成的組: ,其中L 1的*端與L 2連接。 The compound or a pharmaceutically acceptable salt thereof as described in claim 9, wherein L1 is selected from the group consisting of: , , , , , , and , where the * end of L1 is connected to L2 . 如請求項1所述的化合物或其藥學上可接受的鹽,其中L 2是鍵。 The compound or pharmaceutically acceptable salt thereof as described in claim 1, wherein L2 is a bond. 如請求項1所述的化合物或其藥學上可接受的鹽,其中L 2是N(R A),並且R A選自烷基或雜環基,其中所述烷基或所述雜環基任選地被一個或多個鹵素或烷基取代。 The compound or pharmaceutically acceptable salt thereof as described in claim 1, wherein L 2 is N( RA ), and RA is selected from alkyl or heterocyclic groups, wherein the alkyl or heterocyclic group is optionally substituted with one or more halogens or alkyl groups. 如請求項12所述的化合物或其藥學上可接受的鹽,其中R A是乙基、二氟乙基、三氟乙基或氧雜環丁烷基。 The compound or pharmaceutically acceptable salt thereof as described in claim 12, wherein RA is ethyl, difluoroethyl, trifluoroethyl or oxacyclobutane. 如請求項1所述的化合物或其藥學上可接受的鹽,其中L 2是任選地被一個或多個R B取代的環烷基。 The compound or pharmaceutically acceptable salt thereof as described in claim 1, wherein L 2 is a cycloalkyl group optionally substituted by one or more RBs . 如請求項14所述的化合物或其藥學上可接受的鹽,其中L 2,其任選地被一個或多個R B取代。 The compound or pharmaceutically acceptable salt thereof as described in claim 14, wherein L2 is , which is optionally replaced by one or more RBs . 如請求項1所述的化合物或其藥學上可接受的鹽,其中L 2是任選地被一個或多個R B取代的雜環基。 The compound or pharmaceutically acceptable salt thereof as described in claim 1, wherein L2 is a heterocyclic group optionally substituted by one or more RB . 如請求項16所述的化合物或其藥學上可接受的鹽,其中所述雜環基選自由以下組成的組: , 其中的每一個任選地被一個或多個R B取代,其中L 2的*端與L 3連接。 The compound or a pharmaceutically acceptable salt thereof as described in claim 16, wherein the heterocyclic group is selected from the group consisting of: , , , , and , each of which is optionally replaced by one or more RB , wherein the * end of L2 is connected to L3 . 如請求項16或17所述的化合物或其藥學上可接受的鹽,其中R B是烷基。 The compound or pharmaceutically acceptable salt thereof as described in claim 16 or 17, wherein RB is alkyl. 如請求項19所述的化合物或其藥學上可接受的鹽,其中R B是甲基。 The compound or pharmaceutically acceptable salt thereof as described in claim 19, wherein RB is methyl. 如請求項1所述的化合物或其藥學上可接受的鹽,其中L 2選自由以下組成的組: ,其中L 2的*端與L 3連接。 The compound or a pharmaceutically acceptable salt thereof as described in claim 1, wherein L2 is selected from the group consisting of: , , , , , , , , , , , and , where the * end of L2 is connected to L3 . 如請求項1所述的化合物或其藥學上可接受的鹽,其中L 3是烷基。 The compound or pharmaceutically acceptable salt thereof as described in claim 1, wherein L 3 is alkyl. 如請求項21所述的化合物或其藥學上可接受的鹽,其中L 3是乙基。 The compound or pharmaceutically acceptable salt thereof as described in claim 21, wherein L 3 is ethyl. 如請求項1所述的化合物或其藥學上可接受的鹽,其中L 4是O或NH。 The compound or pharmaceutically acceptable salt thereof as described in claim 1, wherein L 4 is O or NH. 如請求項1所述的化合物或其藥學上可接受的鹽,其中L 1是鍵,並且L 2是鍵或任選地被一個或多個R B取代的雜環基。 The compound or pharmaceutically acceptable salt thereof as described in claim 1, wherein L 1 is a bond, and L 2 is a bond or a heterocyclic group optionally substituted by one or more RBs . 如請求項1所述的化合物或其藥學上可接受的鹽,其中L 1是烷基,並且L 2是鍵、N(R A)或任選地被一個或多個R B取代的環烷基。 The compound or pharmaceutically acceptable salt thereof as described in claim 1, wherein L 1 is alkyl, and L 2 is a bond, N( RA ) or cycloalkyl optionally substituted by one or more RBs . 如請求項1所述的化合物或其藥學上可接受的鹽,其中R 1是羥基,並且m是1。 The compound or pharmaceutically acceptable salt thereof as described in claim 1, wherein R 1 is hydroxyl and m is 1. 如請求項1所述的化合物或其藥學上可接受的鹽,其中R 1是鹵素,並且m是1。 The compound or pharmaceutically acceptable salt thereof as described in claim 1, wherein R 1 is halogen and m is 1. 如請求項27所述的化合物或其藥學上可接受的鹽,其中R 1是溴或氟。 The compound or pharmaceutically acceptable salt thereof as described in claim 27, wherein R 1 is bromine or fluorine. 如請求項1所述的化合物或其藥學上可接受的鹽,其中R 1是-N(R D) 2,並且m是1。 The compound or a pharmaceutically acceptable salt thereof as described in claim 1, wherein R 1 is -N( RD ) 2 and m is 1. 如請求項29所述的化合物或其藥學上可接受的鹽,其中每個R D獨立地是氫或任選地被一個或多個獨立地選自-N(R F) 2或-OR G的基團取代的烷基。 The compound or pharmaceutically acceptable salt thereof as described in claim 29, wherein each RD is independently hydrogen or alkyl optionally substituted with one or more groups independently selected from -N( RF ) 2 or -ORG . 如請求項30所述的化合物或其藥學上可接受的鹽,其中每個R D獨立地是氫、甲基、甲氧基乙基、N,N-二甲基氨基乙基、羥乙基或N,N-二甲基氨基丙基。 The compound or pharmaceutically acceptable salt thereof as described in claim 30, wherein each RD is independently hydrogen, methyl, methoxyethyl, N,N-dimethylaminoethyl, hydroxyethyl or N,N-dimethylaminopropyl. 如請求項1所述的化合物或其藥學上可接受的鹽,其中R 1是任選地被一個或多個R E取代的雜環基,並且m是1。 The compound or pharmaceutically acceptable salt thereof as described in claim 1, wherein R 1 is a heterocyclic group optionally substituted by one or more RE , and m is 1. 如請求項32所述的化合物或其藥學上可接受的鹽,其中所述雜環基選自由以下組成的組: , 其中的每一個任選地被一個或多個R E取代。 The compound or a pharmaceutically acceptable salt thereof as described in claim 32, wherein the heterocyclic group is selected from the group consisting of: , , , , , , , , , , , , , , , , , and , each of which is optionally replaced by one or more RE . 如請求項32所述的化合物或其藥學上可接受的鹽,其中R E是鹵素。 The compound or pharmaceutically acceptable salt thereof as described in claim 32, wherein RE is a halogen. 如請求項33所述的化合物或其藥學上可接受的鹽,其中R E是F。 A compound or a pharmaceutically acceptable salt thereof as described in claim 33, wherein RE is F. 如請求項32所述的化合物或其藥學上可接受的鹽,其中每個R E獨立地選自鹵素、-N(R F) 2、-烷基-N(R F) 2、-C(O)OR G或任選地被一個或多個鹵素取代的烷基。 The compound or pharmaceutically acceptable salt thereof as described in claim 32, wherein each RE is independently selected from halogen, -N( RF ) 2 , -alkyl-N( RF ) 2 , -C(O) ORG , or alkyl optionally substituted with one or more halogens. 如請求項36所述的化合物或其藥學上可接受的鹽,其中R F和R G中的每一個獨立地是烷基。 A compound or a pharmaceutically acceptable salt thereof as described in claim 36, wherein each of RF and RG is independently an alkyl group. 如請求項36所述的化合物或其藥學上可接受的鹽,其中R F和R G中的每一個獨立地是C 1-3烷基。 A compound or a pharmaceutically acceptable salt thereof as described in claim 36, wherein each of RF and RG is independently a C1-3 alkyl group. 如請求項36所述的化合物或其藥學上可接受的鹽,其中每個R E獨立地選自由以下組成的組:F、 -N(CH 3) 2、-C 1-3烷基-N(CH 3) 2、-C(O)O(叔丁基)、甲基、乙基或三氟乙基。 The compound or pharmaceutically acceptable salt thereof as described in claim 36, wherein each RE is independently selected from the group consisting of F, -N( CH3 ) 2 , -C1-3alkyl -N( CH3 ) 2 , -C(O)O(tert-butyl), methyl, ethyl or trifluoroethyl. 如請求項32所述的化合物或其藥學上可接受的鹽,其中R E是環烷基或雜環基,其中所述環烷基和所述雜環基任選地被一個或多個烷基或鹵素取代。 The compound or pharmaceutically acceptable salt thereof as described in claim 32, wherein RE is cycloalkyl or heterocyclic, wherein the cycloalkyl and the heterocyclic are optionally substituted with one or more alkyl or halogen. 如請求項40所述的化合物或其藥學上可接受的鹽,其中R E選自環丙基、嗎啉基、哌嗪基、氧雜環丁烯基或氮雜環丁烷基,其中的每一個任選地被一個或多個烷基或鹵素取代。 A compound or a pharmaceutically acceptable salt thereof as described in claim 40, wherein RE is selected from cyclopropyl, morpholinyl, piperazinyl, oxacyclobutenyl or azacyclobutanyl, each of which is optionally substituted with one or more alkyl or halogen groups. 如請求項1所述的化合物或其藥學上可接受的鹽,其中m是2,R 1中的一個是鹵素,並且另一個R 1是任選地被一個或多個R E取代的雜環基。 The compound or pharmaceutically acceptable salt thereof as described in claim 1, wherein m is 2, one of R 1 is halogen, and the other R 1 is a heterocyclic group optionally substituted by one or more RE . 如請求項42所述的化合物或其藥學上可接受的鹽,其中所述雜環基選自由以下組成的組: , 其中的每一個任選地被一個或多個R E取代。 The compound or a pharmaceutically acceptable salt thereof as described in claim 42, wherein the heterocyclic group is selected from the group consisting of: , , , , , , , , , , , , , , , , , and , each of which is optionally replaced by one or more RE . 如請求項43所述的化合物或其藥學上可接受的鹽,其中每個R E獨立地選自鹵素、-N(R F) 2、-烷基-N(R F) 2、-C(O)OR G或任選地被一個或多個鹵素取代的烷基。 The compound or pharmaceutically acceptable salt thereof as described in claim 43, wherein each RE is independently selected from halogen, -N( RF ) 2 , -alkyl-N( RF ) 2 , -C(O) ORG , or alkyl optionally substituted with one or more halogens. 如請求項1所述的化合物或其藥學上可接受的鹽,其中R 1選自由以下組成的組: The compound or a pharmaceutically acceptable salt thereof as described in claim 1, wherein R1 is selected from the group consisting of: 如請求項1所述的化合物或其藥學上可接受的鹽,其中R 2是鹵素,並且n是1或2。 The compound or pharmaceutically acceptable salt thereof as described in claim 1, wherein R2 is a halogen and n is 1 or 2. 如請求項46所述的化合物或其藥學上可接受的鹽,其中R 2是F。 A compound as described in claim 46 or a pharmaceutically acceptable salt thereof, wherein R 2 is F. 如請求項1所述的化合物或其藥學上可接受的鹽,其中R 2是烷基或環烷基,其中所述烷基和環烷基任選地被一個或多個鹵素取代。 The compound or pharmaceutically acceptable salt thereof as described in claim 1, wherein R 2 is alkyl or cycloalkyl, wherein the alkyl and cycloalkyl are optionally substituted with one or more halogens. 如請求項48所述的化合物或其藥學上可接受的鹽,其中R 2是甲基、乙基、環丙基或三氟乙基。 The compound or pharmaceutically acceptable salt thereof as described in claim 48, wherein R 2 is methyl, ethyl, cyclopropyl or trifluoroethyl. 如請求項1所述的化合物或其藥學上可接受的鹽,其中R 3是烷基或環烷基。 The compound or pharmaceutically acceptable salt thereof as described in claim 1, wherein R 3 is alkyl or cycloalkyl. 如請求項50所述的化合物或其藥學上可接受的鹽,其中R 3是甲基、乙基或環丙基。 The compound or pharmaceutically acceptable salt thereof as described in claim 50, wherein R 3 is methyl, ethyl or cyclopropyl. 如請求項1所述的化合物或其藥學上可接受的鹽,其中m是0、1或2。A compound or a pharmaceutically acceptable salt thereof as described in claim 1, wherein m is 0, 1 or 2. 如請求項1所述的化合物或其藥學上可接受的鹽,其中n是0或1。The compound or pharmaceutically acceptable salt thereof as described in claim 1, wherein n is 0 or 1. 如請求項1所述的化合物或其藥學上可接受的鹽,其中所述化合物選自由以下組成的組: The compound or a pharmaceutically acceptable salt thereof as described in claim 1, wherein the compound is selected from the group consisting of: and . 一種藥物組合物,其包含如請求項1至54中任一項所述的化合物或其藥學上可接受的鹽以及藥學上可接受的載體。A pharmaceutical composition comprising a compound as described in any one of claims 1 to 54 or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier. 一種抑制有需要的受試者中EGFR活性的方法,所述方法包括向所述受試者施用有效量的如請求項1至54中任一項所述的化合物或其藥學上可接受的鹽或如請求項55所述的藥物組合物。A method for inhibiting EGFR activity in a subject in need thereof, comprising administering to the subject an effective amount of a compound or a pharmaceutically acceptable salt thereof as described in any one of claims 1 to 54 or a pharmaceutical composition as described in claim 55. 一種治療有需要的受試者中EGFR相關病症的方法,所述方法包括向所述受試者施用有效量的如請求項1至54中任一項所述的化合物或其藥學上可接受的鹽或向所述受試者施用如請求項55所述的藥物組合物。A method for treating an EGFR-related disorder in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound or a pharmaceutically acceptable salt thereof as described in any one of claims 1 to 54 or administering to the subject a pharmaceutical composition as described in claim 55. 如請求項57所述的方法,其中所述EGFR相關病症是自身免疫性疾病或癌症。The method of claim 57, wherein the EGFR-related disorder is an autoimmune disease or cancer. 如請求項58所述的方法,其中所述癌症選自由以下組成的組:肺癌、腦癌、結直腸癌、膀胱癌、尿路上皮癌、乳腺癌、前列腺癌、卵巢癌、頭頸癌、胰腺癌、胃癌和間皮瘤,包括轉移(特別是腦轉移)等。A method as described in claim 58, wherein the cancer is selected from the group consisting of lung cancer, brain cancer, colorectal cancer, bladder cancer, urothelial cancer, breast cancer, prostate cancer, ovarian cancer, head and neck cancer, pancreatic cancer, gastric cancer and mesothelioma, including metastases (especially brain metastases), etc. 如請求項57至59中任一項所述的方法,其中所述化合物與一種或多種另外的治療劑同時、單獨或順序施用。A method as described in any one of claims 57 to 59, wherein the compound is administered simultaneously, separately or sequentially with one or more additional therapeutic agents. 如請求項60所述的方法,其中所述一種或多種另外的治療劑選自由以下組成的組:EGFR TKI、EGFR抗體、MEK抑制劑、c-MET抑制劑、有絲分裂激酶抑制劑、免疫治療劑、抗血管生成劑、凋亡誘導劑、mTOR抑制劑、組蛋白去乙醯化酶抑制劑、IL6抑制劑和JAK抑制劑。A method as described in claim 60, wherein the one or more additional therapeutic agents are selected from the group consisting of: EGFR TKI, EGFR antibody, MEK inhibitor, c-MET inhibitor, mitotic kinase inhibitor, immunotherapy, anti-angiogenic agent, apoptosis inducer, mTOR inhibitor, histone deacetylase inhibitor, IL6 inhibitor and JAK inhibitor.
TW112129923A 2022-09-02 2023-08-09 Egfr inhibitors and uses thereof TW202416963A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
WOPCT/CN2022/116914 2022-09-02
WOPCT/CN2023/107228 2023-07-13

Publications (1)

Publication Number Publication Date
TW202416963A true TW202416963A (en) 2024-05-01

Family

ID=

Similar Documents

Publication Publication Date Title
TWI704152B (en) Naphthyridine compounds as jak kinase inhibitors
JP6948659B1 (en) Pyridadinyl thiaazole carboxamide compound
AU2017250369A1 (en) Amine-substituted aryl or heteroaryl compounds as EHMT1 and EHMT2 inhibitors
JP2023528903A (en) KRAS G12C protein inhibitors and uses thereof
CN112778276A (en) Compound as SHP2 inhibitor and application thereof
MX2013005603A (en) Cyclobutyl substituted pyrrolopyridine and pyrrolopyrimidine derivatives as jak inhibitors.
TW201350476A (en) Compounds and compositions for modulating EGFR activity
US11292791B2 (en) Tetrahydro-imidazo quinoline compositions as CBP/P300 inhibitors
TW201838981A (en) Pyrimidinyl-pyridyloxy-naphthyl compounds and methods of treating ire1-related diseases and disorders
AU2018201317A1 (en) Methyl-and trifluoromethyl-substituted pyrrolopyridine modulators of RORc2 and methods of use thereof
EP2976338B1 (en) N-(2-cyano heterocyclyl)pyrazolo pyridones as janus kinase inhibitors
IL259862A (en) Inhibitors of bruton's tyrosine kinase and methods of their use
KR20230021121A (en) N-phenylaminocarbonyl pyridino-, pyrimidino and benzo-tropanes as modulators of GPR65
EP4284365A1 (en) Cdk2 inhibitors and methods of using the same
TW202128699A (en) Therapeutic compounds
TW202220999A (en) Indazoles as hematopoietic progenitor kinase 1 (hpk1) inhibitors and methods of using same
JP7282743B2 (en) heterocyclic compound
TW202334167A (en) Fused tetracyclic quinazoline derivatives as inhibitors of erbb2
CA3116141C (en) Cycloalkane-1,3-diamine derivative
TWI652265B (en) Azaindole derivatives
TW202416963A (en) Egfr inhibitors and uses thereof
CN115667218A (en) Kinase inhibitors
WO2024046221A1 (en) Egfr inhibitors and uses thereof
TW202321239A (en) Substituted pyrazine-2-carboxamides as hpk1 inhibitors for the treatment of cancer
CA3213823A1 (en) Selective modulators of ataxia telangiectasia mutated (atm) kinase and uses thereof