Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
MACROCYCLE COMPOUNDS USEFUL AS KRAS INHIBITORS
Document Type and Number:
WIPO Patent Application WO/2024/008834
Kind Code:
A1
Abstract:
The present invention relates to compounds of formula (I), wherein R1 to R6, A1 and A2 are as described herein, and their pharmaceutically acceptable salt thereof, and compositions including the compounds and methods of using the compounds as KRAS inhibitors.

Inventors:
CHEN JIANGUO (CN)
LIU HAIXIA (CN)
SHEN HONG (CN)
ZHANG WEIXING (CN)
ZHAO DAN (CN)
ZHU WEI (CN)
Application Number:
PCT/EP2023/068641
Publication Date:
January 11, 2024
Filing Date:
July 06, 2023
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
HOFFMANN LA ROCHE (US)
HOFFMANN LA ROCHE (US)
International Classes:
A61P35/00; A61K31/4995; C07D513/22; C07D515/22
Domestic Patent References:
WO2023133543A12023-07-13
WO2023025832A12023-03-02
WO2022235870A12022-11-10
WO2022235864A12022-11-10
WO2023015559A12023-02-16
WO2022060836A12022-03-24
WO2021091982A12021-05-14
WO2021091967A12021-05-14
WO2021091956A12021-05-14
WO2022060836A12022-03-24
Other References:
ANSEL, HOWARD C ET AL.: "Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems", 2004, LIPPINCOTT, WILLIAMS & WILKINS
GENNARO, ALFONSO R ET AL.: "Remington: The Science and Practice of Pharmacy", 2000, LIPPINCOTT, WILLIAMS & WILKINS
ROWE, RAYMOND C: "Handbook of Pharmaceutical Excipients", 2005, PHARMACEUTICAL PRESS
Attorney, Agent or Firm:
JOCHNOWITZ, Dr. Evan (CH)
Download PDF:
Claims:
CLAIMS 1. A compound of formula (I), , wherein R1 is 3-oxabicyclo[3.1.0]hexanyl, C1-6alkylC3-7cycloalkyl, cyanoC3-7cycloalkyl or haloC1- 6alkylC3-7cycloalkyl; R2 is H or halogen; R3 is H or halogen; R4 is C1-6alkyl or haloC1-6alkyl; R5 is C1-6alkoxyC1-6alkyl; R6 is morpholinyl, (haloC1-6alkyl)piperazinyl or C1-6alkylpiperazinyl; A1 is thiazolylene; A2 is C1-6alkylene; with the proviso that R2 and R3 are not H simultaneously; or a pharmaceutically acceptable salt thereof. 2. A compound of formula (Ia),

, wherein R1 is 3-oxabicyclo[3.1.0]hexanyl, C1-6alkylC3-7cycloalkyl, cyanoC3-7cycloalkyl or haloC1- 6alkylC3-7cycloalkyl; R2 is H or halogen; R3 is H or halogen; R4 is C1-6alkyl or haloC1-6alkyl; R5 is C1-6alkoxyC1-6alkyl; R6 is morpholinyl, (haloC1-6alkyl)piperazinyl or C1-6alkylpiperazinyl; A1 is thiazolylene; A2 is C1-6alkylene; with the proviso that R2 and R3 are not H simultaneously; or a pharmaceutically acceptable salt thereof. 3. A compound according to claim 1 or 2, wherein R1 is C1-6alkylC3-7cycloalkyl or haloC1- 6alkylC3-7cycloalkyl. 4. A compound according to any one of claims 1-3, wherein R1 is methylcyclopropyl or (difluoromethyl)cyclopropyl. 5. A compound according to any one of claims 1-4, wherein R2 is H or fluoro. 6. A compound according to any one of claims 1-5, wherein R3 is H or fluoro.

7. A compound according to any one of claims 1-6, wherein R4 is ethyl or 2,2,2-trifluoroethyl. 8. A compound according to any one of claims 1-7, wherein R5 is 1-methoxyethyl. 9. A compound according to any one of claims 1-8, wherein R6 is morpholinyl or C1- 6alkylpiperazinyl. 10. A compound according to any one of claims 1-9, wherein R6 is morpholinyl or 4- methylpiperazin-1-yl. 11. A compound according to any one of claims 1-10, wherein A1 is , wherein bond “a” connects to indole ring. 12. A compound according to any one of claims 1-11, wherein A2 is dimethylmethylene. 13. A compound according to claim 1 or 2, wherein R1 is C1-6alkylC3-7cycloalkyl or haloC1-6alkylC3-7cycloalkyl; R2 is H or halogen; R3 is H or halogen; R4 is C1-6alkyl or haloC1-6alkyl; R5 is C1-6alkoxyC1-6alkyl; R6 is morpholinyl or C1-6alkylpiperazinyl; A1 is , wherein bond “a” connects to indole ring; A2 is C1-6alkylene; with the proviso that R2 and R3 are not H simultaneously; or a pharmaceutically acceptable salt thereof. 14. A compound according to claim 13, wherein R1 is 2-methylcyclopropyl or 2-(difluoromethyl)cyclopropyl; R2 is H or fluoro; R3 is H or fluoro; R4 is ethyl or 2,2,2-trifluoroethyl; R5 is (1S)-1-methoxyethyl; R6 is morpholinyl or 4-methylpiperazin-1-yl; A1 is , wherein bond “a” connects to indole ring; A2 is dimethylmethylene; with the proviso that R2 and R3 are not H simultaneously; or a pharmaceutically acceptable salt thereof. 15. A compound selected from: (1S,2S)-N-[(7S,13S)-21-ethyl-24-fluoro-(20M)-20-[2-[(1S)-1-methoxyethyl]-5-(4- methylpiperazin-1-yl)-3-pyridyl]-17,17-dimethyl-8,14-dioxo-15-oxa-4-thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaen-7-yl]-2- methyl-cyclopropanecarboxamide; (1R,5S)-N-[(7S,13S)-21-ethyl-24-fluoro-(20M)-20-[2-[(1S)-1-methoxyethyl]-5-(4- methylpiperazin-1-yl)-3-pyridyl]-17,17-dimethyl-8,14-dioxo-15-oxa-4-thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaen-7-yl]-3- oxabicyclo[3.1.0]hexane-6-carboxamide; (1S,2S)-2-(difluoromethyl)-N-[(7S,13S)-21-ethyl-24-fluoro-(20M)-20-[2-[(1S)-1- methoxyethyl]-5-(4-methylpiperazin-1-yl)-3-pyridyl]-17,17-dimethyl-8,14-dioxo-15-oxa-4-thia- 2,5 9,13 22,26 9,21,27,28-tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaen- 7-yl]cyclopropanecarboxamide; (1S,2S)-N-[(7S,13S)-24-fluoro-(20M)-20-[2-[(1S)-1-methoxyethyl]-5-(4-methylpiperazin- 1-yl)-3-pyridyl]-17,17-dimethyl-8,14-dioxo-21-(2,2,2-trifluoroethyl)-15-oxa-4-thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaen-7-yl]-2- methyl-cyclopropanecarboxamide; (1S,2S)-2-cyano-N-[(7S,13S)-21-ethyl-24-fluoro-(20M)-20-[2-[(1S)-1-methoxyethyl]-5-(4- methylpiperazin-1-yl)-3-pyridyl]-17,17-dimethyl-8,14-dioxo-15-oxa-4-thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaen-7- yl]cyclopropanecarboxamide; (1R,2R)-2-cyano-N-[(7S,13S)-21-ethyl-24-fluoro-(20M)-20-[2-[(1S)-1-methoxyethyl]-5-(4- methylpiperazin-1-yl)-3-pyridyl]-17,17-dimethyl-8,14-dioxo-15-oxa-4-thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaen-7- yl]cyclopropanecarboxamide; (1S,2S)-N-[(7S,13S)-21-ethyl-24-fluoro-(20M)-20-[2-[(1S)-1-methoxyethyl]-5-[4-(2,2,2- trifluoroethyl)piperazin-1-yl]-3-pyridyl]-17,17-dimethyl-8,14-dioxo-15-oxa-4-thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaen-7-yl]-2- methyl-cyclopropanecarboxamide; (1S,2S)-N-[(7S,13S)-21-ethyl-24-fluoro-(20M)-20-[2-[(1S)-1-methoxyethyl]-5-morpholino- 3-pyridyl]-17,17-dimethyl-8,14-dioxo-15-oxa-4-thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaen-7-yl]-2- methyl-cyclopropanecarboxamide; (1S,2S)-N-[(7S,13S)-24-fluoro-(20M)-20-[2-[(1S)-1-methoxyethyl]-5-[4-(2,2,2- trifluoroethyl)piperazin-1-yl]-3-pyridyl]-17,17-dimethyl-8,14-dioxo-21-(2,2,2-trifluoroethyl)-15- 2,5 9,13 22,26 oxa-4-thia-9,21,27,28-tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa- 1(25),2,5(28),19,22(26),23-hexaen-7-yl]-2-methyl-cyclopropanecarboxamide; (1S,2S)-N-[(7S,13S)-25-fluoro-(20M)-20-[2-[(1S)-1-methoxyethyl]-5-(4-methylpiperazin- 1-yl)-3-pyridyl]-17,17-dimethyl-8,14-dioxo-21-(2,2,2-trifluoroethyl)-15-oxa-4-thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaen-7-yl]-2- methyl-cyclopropanecarboxamide; (1S,2S)-2-(difluoromethyl)-N-[(7S,13S)-21-ethyl-24-fluoro-(20M)-20-[2-[(1S)-1- methoxyethyl]-5-morpholino-3-pyridyl]-17,17-dimethyl-8,14-dioxo-15-oxa-4-thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaen-7- yl]cyclopropanecarboxamide; and (1S,2S)-N-[(7S,13S)-24-fluoro-(20M)-20-[2-[(1S)-1-methoxyethyl]-5-morpholino-3- pyridyl]-17,17-dimethyl-8,14-dioxo-21-(2,2,2-trifluoroethyl)-15-oxa-4-thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaen-7-yl]-2- methyl-cyclopropanecarboxamide; or a pharmaceutically acceptable salt thereof.

16. A process for the preparation of a compound according to any one of claims 1 to 15 comprising any of the following steps: a) coupling reaction between compound of formula (II), the presence of a coupling reagent and a base to form the compound of formula (I); wherein R1, R2, R3, R4 , R5, R6, R7, A1 and A2 are defined as in any one of claims 1 to 14; the coupling reagent is T3P, HATU, PyBOP or EDCI/HOBt; the base is TEA, DIEPA or DMAP. 17. A compound or pharmaceutically acceptable salt according to any one of claims 1 to 15 for use as therapeutically active substance. 18. A pharmaceutical composition comprising a compound in accordance with any one of claims 1 to 15 and a pharmaceutically acceptable excipient. 19. The use of a compound according to any one of claims 1 to 15 for treating a KRAS G12C protein-related disease. 20. The use of a compound according to any one of claims 1 to 15 for treating a KRAS G12C, G12D and G12V protein-related disease. 21. The use of a compound according to any one of claims 1 to 15 for inhibiting RAS interaction with downstream effectors, wherein the downstream effectors are RAF and PI3K. 22. The use of a compound according to any one of claims 1 to 15 for inhibiting the propagating oncogenic MAPK and PI3K signaling.

23. The use of a compound according to any one of claims 1 to 15 for the treatment or prophylaxis of KRAS mutation driven cancers, wherein the cancer is selected from pancreatic cancer, colorectal cancer, lung cancer, esophageal cancer, gallbladder cancer, melanoma ovarian cancer and endometrial cancer. 24. The use of a compound according to any one of claims 1 to 15 for the treatment or prophylaxis of KRAS mutation driven cancers, wherein the cancer is selected from pancreatic adenocarcinoma, colorectal cancer and non-small cell lung cancer. 25. A compound or pharmaceutically acceptable salt according to any one of claims 1 to 15 for the treatment or prophylaxis of KRAS mutation driven cancers, wherein the cancer is selected from pancreatic adenocarcinoma, colorectal cancer and non-small cell lung cancer. 26. The use of a compound according to any one of claims 1 to 15 for the preparation of a medicament for the treatment or prophylaxis of KRAS mutation driven cancers, wherein the cancer is selected from pancreatic adenocarcinoma, colorectal cancer and non-small cell lung cancer. 27. A method for the treatment or prophylaxis of KRAS mutation driven cancers, wherein the cancer is selected from pancreatic adenocarcinoma, colorectal cancer and non-small cell lung cancer, which method comprises administering a therapeutically effective amount of a compound as defined in any one of claims 1 to 15. 28. A compound or pharmaceutically acceptable salt according to any one of claims 1 to 15, when manufactured according to a process of claim 16. 29. The invention as hereinbefore described.

Description:
Macrocycle compounds for the treatment of cancer The present invention relates to organic compounds useful for therapy and/or prophylaxis in a mammal, and in particular to inhibition of KRAS mutant useful for treating cancers. FIELD OF THE INVENTION RAS is one of the most well-known proto-oncogenes. Approximately 30% of human cancers contain mutations in three most notable members, KRAS, HRAS, and NRAS, making them the most prevalent oncogenic drivers. KRAS mutations are generally associated with poor prognosis especially in colorectal cancer, pancreatic cancer, lung cancers. As the most frequently mutated RAS isoform, KRAS has been intensively studied in the past years. Among the most commonly occurring KRAS alleles (including G12D, G12V, G12C, G13D, G12R, G12A, G12S, Q61H, etc), G12C, G12D, G12V represent more than half of all K-RAS-driven cancers across colorectal cancer (CRC), pancreatic ductal adenocarcinoma (PDAC), lung adenocarcinoma (LUAD). Of note, KRAS wild-type amplifications are also found in around 7% of all KRAS- altered cancers (ovarian, esophagogastric, uterine), ranking among the top alterations. All RAS proteins belong to a protein family of small GTPases that hydrolyze GTP to GDP. KRAS is structurally divided into an effector binding lobe followed by the allosteric lobe and a carboxy-terminal region that is responsible for membrane anchoring. The effector lobe comprises the P-loop, switch I, and switch II regions. The switch I/II loops play a critical role in KRAS downstream signaling through mediating protein–protein interactions with effector proteins that include RAF in the mitogen-activated protein kinase (MAPK) pathway or PI3K in the phosphatidylinositol 3‑kinase (PI3K)/protein kinase B (AKT) pathway. KRAS protein switches between an inactive to an active form via binding to GTP and GDP, respectively. Under physiological conditions, the transition between these two states is regulated by guanine nucleotide exchange factors (GEFs), such as Son Of Sevenless Homolog 1 (SOS1), or GTPase-activating proteins (GAPs) that involve catalyzing the exchange of GDP for GTP, potentiating intrinsic GTPase activity or accelerating RAS-mediated GTP hydrolysis. In response to extracellular stimuli, the inactive RAS-GDP is converted to active RAS-GTP which directly binds to RAF RAS binding domains (RAF RBD ), recruiting RAF kinase family from cytoplasm to membranes, where they dimerize and become active. The activated RAF subsequently carries out a chain of phosphorylation reactions to its downstream Mitogen- activated protein kinase (MEK) and extracellular signal-regulated kinase (ERK), and propagates the growth signal. Of the RAF family of protein kinases (three known isoforms ARAF, BRAF, CRAF/RAF1), BRAF is most frequently mutated and remains the most potent activator of MEK. Despite that individual RAS and RAF family members revealed distinct binding preferences, all RAFs possess the conserved RBD for forward transmission of MAPK singnaling, frequently used for characterize KRAS inhibition (e.g. KRAS-BRAF RBD herein). For KRAS, mutations at positions 12, 13, 61, and 146 lead to a shift toward the active KRAS form through impairing nucleotide hydrolysis or activating nucleotide exchange, leading to hyper-activation of the MAPK pathway that results in tumorigenesis. Despite its well-recognized importance in cancer malignancy, continuous efforts in the past failed to develop approved therapies for KRAS mutant cancer until recently, the first selective drug AMG510 has fast approval as second line treatment in KRAS G12C driven non-small cell lung cancer (NSCLC). Nevertheless, the clinical acquired resistance to KRAS G12C inhibitors emerge rigorously with disease progresses after around 6 month of treatment. All of the mutations converge to reactivate RAS–MAPK signaling, with secondary RAS mutants at oncogenic hotspots (e.g. G12/G13/Q61) and within the switch II pocket (e.g. H95, R68, and Y96) have been observed; moreover, over 85% of all KRAS-mutated or wild-type amplified driven cancers still lack novel agents. Altogether, both the myriad of escape mechanism and various oncogenic alleles, highlight the urgent medical need for additional KRAS therapies. As such, we invented oral compounds that target and inhibit KRAS alleles for the treatment of KRAS mutant driven cancers. SUMMARY OF THE INVENTION The present invention relates to novel compounds of formula (I), wherein R 1 is 3-oxabicyclo[3.1.0]hexanyl, C 1-6 alkylC 3-7 cycloalkyl, cyanoC 3-7 cycloalkyl or haloC 1- 6 alkylC 3-7 cycloalkyl; R 2 is H or halogen; R 3 is H or halogen; R 4 is C 1-6 alkyl or haloC 1-6 alkyl; R 5 is C 1-6 alkoxyC 1-6 alkyl; R 6 is morpholinyl, (haloC 1-6 alkyl)piperazinyl or C 1-6 alkylpiperazinyl; A 1 is thiazolylene; A 2 is C 1-6 alkylene; with the proviso that R 2 and R 3 are not H simultaneously; or a pharmaceutically acceptable salt thereof. The invention also relates to their manufacture, medicaments based on a compound in accordance with the invention and their production as well as the use of compounds of formula (I) or (Ia) thereof as inhibitor of KRAS. The compounds of formula (I) or (Ia) show good KRAS inhibition for G12C and G12V. In another embodiment, the compounds of this invention showed superior cancer cell inhibition and human hepatocyte stability. In addition, the compounds of formula (I) or (Ia) also show good or improved cytotoxicity and solubility profiles. Furthermore, the compound of current invention had good pharmacokinetic properties comparing with the reference compounds. DETAILED DESCRIPTION OF THE INVENTION DEFINITIONS The term “C 1-6 alkyl” denotes a saturated, linear or branched chain alkyl group containing 1 to 6, particularly 1 to 4 carbon atoms, for example methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl and the like. Particular “C 1-6 alkyl” groups are methyl, ethyl and n-propyl. The term “C 1-6 alkoxy” denotes C 1-6 alkyl-O-. The term “C 1-6 alkylene” denotes a linear or branched saturated divalent hydrocarbon group of 1 to 6 carbon atoms or a divalent branched saturated divalent hydrocarbon group of 3 to 6 carbon atoms. Examples of C 1-6 alkylene groups include methylene, ethylene, propylene, 2- methylpropylene, butylene, 2-ethylbutylene, pentylene, hexylene. The term “halogen” and “halo” are used interchangeably herein and denote fluoro, chloro, bromo, or iodo. The term “haloC 1-6 alkyl” denotes a C 1-6 alkyl group wherein at least one of the hydrogen atoms of the C 1-6 alkyl group has been replaced by same or different halogen atoms, particularly fluoro atoms. Examples of haloalkyl include monofluoro-, difluoro- or trifluoro-methyl, -ethyl or -propyl, for example 3,3,3-trifluoropropyl, 2-fluoroethyl, 2,2,2-trifluoroethyl, fluoromethyl, or trifluoromethyl. The term “C 3-7 cycloalkyl” denotes a monovalent saturated monocyclic or bicyclic hydrocarbon group of 3 to 7 ring carbon atoms. Bicyclic means consisting of two saturated carbocycles having one or more carbon atoms in common. Examples for monocyclic cycloalkyl are cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl or cycloheptyl. Examples for bicyclic cycloalkyl are bicyclo[1.1.0]butyl, bicyclo[2.2.1]heptanyl, bicyclo[1.1.1]pentanyl, or bicyclo[2.2.2]octanyl. The term “thiazolylene” denotes a divalent thiazolyl group. The term “oxo” denotes a divalent oxygen atom =O. The term “dimethylmethylene” denotes . The term “protecting group” denotes the group which selectively blocks a reactive site in a multifunctional compound such that a chemical reaction can be carried out selectively at another unprotected reactive site in the meaning conventionally associated with it in synthetic chemistry. Protecting groups can be removed at the appropriate point. Exemplary protecting groups are amino-protecting groups, carboxy-protecting groups or hydroxy-protecting groups. The skilled of the art would understand that the following structures of compounds of formula (I) and (I’) are equal especially for the chiral centers: The term “pharmaceutically acceptable salts” denotes salts which are not biologically or otherwise undesirable. Pharmaceutically acceptable salts include both acid and base addition salts. The term “pharmaceutically acceptable acid addition salt” denotes those pharmaceutically acceptable salts formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, carbonic acid, phosphoric acid, and organic acids selected from aliphatic, cycloaliphatic, aromatic, araliphatic, heterocyclic, carboxylic, and sulfonic classes of organic acids such as formic acid, acetic acid, propionic acid, glycolic acid, gluconic acid, lactic acid, pyruvic acid, oxalic acid, malic acid, maleic acid, maloneic acid, succinic acid, fumaric acid, tartaric acid, citric acid, aspartic acid, ascorbic acid, glutamic acid, anthranilic acid, benzoic acid, cinnamic acid, mandelic acid, embonic acid, phenylacetic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, and salicyclic acid. The term “pharmaceutically acceptable base addition salt” denotes those pharmaceutically acceptable salts formed with an organic or inorganic base. Examples of acceptable inorganic bases include sodium, potassium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, and aluminum salts. Salts derived from pharmaceutically acceptable organic nontoxic bases includes salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, 2-diethylaminoethanol, trimethamine, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine, purines, piperizine, piperidine, N-ethylpiperidine, and polyamine resins. The term “A pharmaceutically active metabolite” denotes a pharmacologically active product produced through metabolism in the body of a specified compound or salt thereof. After entry into the body, most drugs are substrates for chemical reactions that may change their physical properties and biologic effects. These metabolic conversions, which usually affect the polarity of the compounds of the invention, alter the way in which drugs are distributed in and excreted from the body. However, in some cases, metabolism of a drug is required for therapeutic effect. The term “therapeutically effective amount” denotes an amount of a compound or molecule of the present invention that, when administered to a subject, (i) treats or prevents the particular disease, condition or disorder, (ii) attenuates, ameliorates or eliminates one or more symptoms of the particular disease, condition, or disorder, or (iii) prevents or delays the onset of one or more symptoms of the particular disease, condition or disorder described herein. The therapeutically effective amount will vary depending on the compound, the disease state being treated, the severity of the disease treated, the age and relative health of the subject, the route and form of administration, the judgement of the attending medical or veterinary practitioner, and other factors. The term “pharmaceutical composition” denotes a mixture or solution comprising a therapeutically effective amount of an active pharmaceutical ingredient together with pharmaceutically acceptable excipients to be administered to a mammal, e.g., a human in need thereof. The terms “pharmaceutically acceptable excipient”, “pharmaceutically acceptable carrier” and “therapeutically inert excipient” can be used interchangeably and denote any pharmaceutically acceptable ingredient in a pharmaceutical composition having no therapeutic activity and being non-toxic to the subject administered, such as disintegrators, binders, fillers, solvents, buffers, tonicity agents, stabilizers, antioxidants, surfactants, carriers, diluents or lubricants used in formulating pharmaceutical products. INHIBITOR OF KRAS The present invention relates to (i) a compound of formula (I), wherein R 1 is 3-oxabicyclo[3.1.0]hexanyl, C 1-6 alkylC 3-7 cycloalkyl, cyanoC 3-7 cycloalkyl or haloC 1- 6 alkylC 3-7 cycloalkyl; R 2 is H or halogen; R 3 is H or halogen; R 4 is C 1-6 alkyl or haloC 1-6 alkyl; R 5 is C 1-6 alkoxyC 1-6 alkyl; R 6 is morpholinyl, (haloC 1-6 alkyl)piperazinyl or C 1-6 alkylpiperazinyl; A 1 is thiazolylene; A 2 is C 1-6 alkylene; with the proviso that R 2 and R 3 are not H simultaneously; or a pharmaceutically acceptable salt thereof. Another embodiment of present invention is (ii) a compound of formula (Ia), wherein R 1 is 3-oxabicyclo[3.1.0]hexanyl, C 1-6 alkylC 3-7 cycloalkyl, cyanoC 3-7 cycloalkyl or haloC 1- 6 alkylC 3-7 cycloalkyl; R 2 is H or halogen; R 3 is H or halogen; R 4 is C 1-6 alkyl or haloC 1-6 alkyl; R 5 is C 1-6 alkoxyC 1-6 alkyl; R 6 is morpholinyl, (haloC 1-6 alkyl)piperazinyl or C 1-6 alkylpiperazinyl; A 1 is thiazolylene; A 2 is C 1-6 alkylene; with the proviso that R 2 and R 3 are not H simultaneously; or a pharmaceutically acceptable salt thereof. A further embodiment of present invention is (iii) a compound of formula (I) or (Ia) according to (i) or (ii), or a pharmaceutically acceptable salt thereof, wherein R 1 is C 1-6 alkylC 3- 7cycloalkyl or haloC 1-6 alkylC 3-7 cycloalkyl. A further embodiment of present invention is (iv) a compound of formula (I) or (Ia), according to any one of (i) to (iii), or a pharmaceutically acceptable salt thereof, wherein R 1 is methylcyclopropyl or (difluoromethyl)cyclopropyl. A further embodiment of present invention is (v) a compound of formula (I) or (Ia) according to any one of (i) to (iv), wherein R 2 is H or fluoro. A further embodiment of present invention is (vi) a compound of formula (I) or (Ia), or a pharmaceutically acceptable salt thereof, according to any one of (i) to (v), wherein R 3 is H or fluoro. A further embodiment of present invention is (vii) a compound of formula (I) or (Ia), or a pharmaceutically acceptable salt thereof, according to any one of (i) to (vi), wherein R 4 is ethyl or 2,2,2-trifluoroethyl. A further embodiment of present invention is (viii) a compound of formula (I) or (Ia), or a pharmaceutically acceptable salt thereof, according to any one of (i) to (vii), wherein R 5 is 1- methoxyethyl. A further embodiment of present invention is (ix) a compound of formula (I) or (Ia), or a pharmaceutically acceptable salt thereof, according to any one of (i) to (viii), wherein R 6 is morpholinyl or C 1-6 alkylpiperazinyl. A further embodiment of present invention is (x) a compound of formula (I) or (Ia), or a pharmaceutically acceptable salt thereof, according to any one of (i) to (ix), wherein R 6 is morpholinyl or 4-methylpiperazin-1-yl. A further embodiment of present invention is (xi) a compound of formula (I) or (Ia), or a pharmaceutically acceptable salt thereof, according to any one of (i) to (x), wherein A 1 is , wherein bond “a” connects to indole ring. A further embodiment of present invention is (xii) a compound of formula (I) or (Ia), or a pharmaceutically acceptable salt thereof, according to any one of (i) to (xi), wherein A 2 is dimethylmethylene. Another embodiment of present invention is (xiii) a compound of formula (I) or (Ia), according to (i) or (ii), wherein R 1 is C 1-6 alkylC 3-7 cycloalkyl or haloC 1-6 alkylC 3-7 cycloalkyl; R 2 is H or halogen; R 3 is H or halogen; R 4 is C 1-6 alkyl or haloC 1-6 alkyl; R 5 is C 1-6 alkoxyC 1-6 alkyl; R 6 is morpholinyl or C 1-6 alkylpiperazinyl; A 1 is , wherein bond “a” connects to indole ring; A 2 is C 1-6 alkylene; with the proviso that R 2 and R 3 are not H simultaneously; or a pharmaceutically acceptable salt thereof. Another embodiment of present invention is (xiv) a compound of formula (I) or (Ia) , according to (xiii), wherein R 1 is 2-methylcyclopropyl or 2-(difluoromethyl)cyclopropyl; R 2 is H or fluoro; R 3 is H or fluoro; R 4 is ethyl or 2,2,2-trifluoroethyl; R 5 is (1S)-1-methoxyethyl; R 6 is morpholinyl or 4-methylpiperazin-1-yl; A 1 is , wherein bond “a” connects to indole ring; A 2 is dimethylmethylene; with the proviso that R 2 and R 3 are not H simultaneously; or a pharmaceutically acceptable salt thereof. Another embodiment of present invention is (xv) a compound of formula (I) or (Ia) selected from the following: (1S,2S)-N-[(7S,13S)-21-ethyl-24-fluoro-(20M)-20-[2-[(1S)-1-m ethoxyethyl]-5-(4- methylpiperazin-1-yl)-3-pyridyl]-17,17-dimethyl-8,14-dioxo-1 5-oxa-4-thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaen-7-yl]-2- methyl-cyclopropanecarboxamide; (1R,5S)-N-[(7S,13S)-21-ethyl-24-fluoro-(20M)-20-[2-[(1S)-1-m ethoxyethyl]-5-(4- methylpiperazin-1-yl)-3-pyridyl]-17,17-dimethyl-8,14-dioxo-1 5-oxa-4-thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaen-7-yl]-3- oxabicyclo[3.1.0]hexane-6-carboxamide; (1S,2S)-2-(difluoromethyl)-N-[(7S,13S)-21-ethyl-24-fluoro-(2 0M)-20-[2-[(1S)-1- methoxyethyl]-5-(4-methylpiperazin-1-yl)-3-pyridyl]-17,17-di methyl-8,14-dioxo-15-oxa-4-thia- 2,5 9,13 22,26 9,21,27,28-tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaen- 7-yl]cyclopropanecarboxamide; (1S,2S)-N-[(7S,13S)-24-fluoro-(20M)-20-[2-[(1S)-1-methoxyeth yl]-5-(4-methylpiperazin- 1-yl)-3-pyridyl]-17,17-dimethyl-8,14-dioxo-21-(2,2,2-trifluo roethyl)-15-oxa-4-thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaen-7-yl]-2- methyl-cyclopropanecarboxamide; (1S,2S)-2-cyano-N-[(7S,13S)-21-ethyl-24-fluoro-(20M)-20-[2-[ (1S)-1-methoxyethyl]-5-(4- methylpiperazin-1-yl)-3-pyridyl]-17,17-dimethyl-8,14-dioxo-1 5-oxa-4-thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaen-7- yl]cyclopropanecarboxamide; (1R,2R)-2-cyano-N-[(7S,13S)-21-ethyl-24-fluoro-(20M)-20-[2-[ (1S)-1-methoxyethyl]-5-(4- methylpiperazin-1-yl)-3-pyridyl]-17,17-dimethyl-8,14-dioxo-1 5-oxa-4-thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaen-7- yl]cyclopropanecarboxamide; (1S,2S)-N-[(7S,13S)-21-ethyl-24-fluoro-(20M)-20-[2-[(1S)-1-m ethoxyethyl]-5-[4-(2,2,2- trifluoroethyl)piperazin-1-yl]-3-pyridyl]-17,17-dimethyl-8,1 4-dioxo-15-oxa-4-thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaen-7-yl]-2- methyl-cyclopropanecarboxamide; (1S,2S)-N-[(7S,13S)-21-ethyl-24-fluoro-(20M)-20-[2-[(1S)-1-m ethoxyethyl]-5-morpholino- 3-pyridyl]-17,17-dimethyl-8,14-dioxo-15-oxa-4-thia-9,21,27,2 8- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaen-7-yl]-2- methyl-cyclopropanecarboxamide; (1S,2S)-N-[(7S,13S)-24-fluoro-(20M)-20-[2-[(1S)-1-methoxyeth yl]-5-[4-(2,2,2- trifluoroethyl)piperazin-1-yl]-3-pyridyl]-17,17-dimethyl-8,1 4-dioxo-21-(2,2,2-trifluoroethyl)-15- 2,5 9,13 22,26 oxa-4-thia-9,21,27,28-tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa- 1(25),2,5(28),19,22(26),23-hexaen-7-yl]-2-methyl-cyclopropan ecarboxamide; (1S,2S)-N-[(7S,13S)-25-fluoro-(20M)-20-[2-[(1S)-1-methoxyeth yl]-5-(4-methylpiperazin- 1-yl)-3-pyridyl]-17,17-dimethyl-8,14-dioxo-21-(2,2,2-trifluo roethyl)-15-oxa-4-thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaen-7-yl]-2- methyl-cyclopropanecarboxamide; (1S,2S)-2-(difluoromethyl)-N-[(7S,13S)-21-ethyl-24-fluoro-(2 0M)-20-[2-[(1S)-1- methoxyethyl]-5-morpholino-3-pyridyl]-17,17-dimethyl-8,14-di oxo-15-oxa-4-thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaen-7- yl]cyclopropanecarboxamide; and (1S,2S)-N-[(7S,13S)-24-fluoro-(20M)-20-[2-[(1S)-1-methoxyeth yl]-5-morpholino-3- pyridyl]-17,17-dimethyl-8,14-dioxo-21-(2,2,2-trifluoroethyl) -15-oxa-4-thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaen-7-yl]-2- methyl-cyclopropanecarboxamide; or a pharmaceutically acceptable salt thereof. Another embodiment of present invention is related to (xvi) a process for the preparation of a compound according to any one of (i) to (xv) comprising the following step: a) coupling reaction between compound of formula (II), the presence of a coupling reagent and a base to form the compound of formula (I); wherein R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , A 1 and A 2 are defined as in any one of (i) to (xiv); the coupling reagent is T3P, HATU, PyBOP or EDCI/HOBt; the base is TEA, DIEPA or DMAP. Another embodiment of present invention is (xvii) a compound or pharmaceutically acceptable salt according to any one of (i) to (xv) for use as therapeutically active substance. Another embodiment of present invention is (xviii) a pharmaceutical composition comprising a compound in accordance with any one of (i) to (xv) and a pharmaceutically acceptable excipient. Another embodiment of present invention is (xix) the use of a compound according to any one of (i) to (xv) for treating a KRAS G12C protein-related disease. Another embodiment of present invention is (xx) the use of a compound according to any one of (i) to (xv) for treating a KRAS G12C, G12D and G12V protein-related disease. Another embodiment of present invention is (xxi) the use of a compound according to any one of (i) to (xv) for inhibiting RAS interaction with downstream effectors, wherein the downstream effectors are RAF and PI3K. Another embodiment of present invention is (xxii) the use of a compound according to any one of (i) to (xv) for inhibiting the propagating oncogenic MAPK and PI3K signaling. Another embodiment of present invention is (xxiii) the use of a compound according to any one of (i) to (xv) for the treatment or prophylaxis of KRAS mutation driven cancers, wherein the cancer is selected from pancreatic cancer, colorectal cancer, lung cancer, esophageal cancer, gallbladder cancer, melanoma ovarian cancer and endometrial cancer. Another embodiment of present invention is (xxiv) the use of a compound according to any one of (i) to (xv) for the treatment or prophylaxis of KRAS mutation driven cancers, wherein the cancer is selected from pancreatic adenocarcinoma, colorectal cancer and non-small cell lung cancer. Another embodiment of present invention is (xxv) a compound or pharmaceutically acceptable salt according to any one of (i) to (xv) for the treatment or prophylaxis of KRAS mutation driven cancers, wherein the cancer is selected from pancreatic adenocarcinoma, colorectal cancer and non-small cell lung cancer. Another embodiment of present invention is (xxvi) the use of a compound according to any one of (i) to (xv) for the preparation of a medicament for the treatment or prophylaxis of KRAS mutation driven cancers, wherein the cancer is selected from pancreatic adenocarcinoma, colorectal cancer and non-small cell lung cancer. Another embodiment of present invention is (xxvii) a method for the treatment or prophylaxis of KRAS mutation driven cancers, wherein the cancer is selected from pancreatic adenocarcinoma, colorectal cancer and non-small cell lung cancer, which method comprises administering a therapeutically effective amount of a compound as defined in any one of (i) to (xv). Another embodiment of present invention is (xxviii) a compound or pharmaceutically acceptable salt according to any one of (i) to (xv), when manufactured according to a process of (xvi). PHARMACEUTICAL COMPOSITIONS AND ADMINISTRATION Another embodiment provides pharmaceutical compositions or medicaments containing the compounds of the invention and a therapeutically inert carrier, diluent or excipient, as well as methods of using the compounds of the invention to prepare such compositions and medicaments. In one example, compounds of formula (I) may be formulated by mixing at ambient temperature at the appropriate pH, and at the desired degree of purity, with physiologically acceptable carriers, i.e., carriers that are non-toxic to recipients at the dosages and concentrations employed into a galenical administration form. The pH of the formulation depends mainly on the particular use and the concentration of compound, but preferably ranges anywhere from about 3 to about 8. In one example, a compound of formula (I) is formulated in an acetate buffer, at pH 5. In another embodiment, the compounds of formula (I) are sterile. The compound may be stored, for example, as a solid or amorphous composition, as a lyophilized formulation or as an aqueous solution. Compositions are formulated, dosed, and administered in a fashion consistent with good medical practice. Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners. The “effective amount” of the compound to be administered will be governed by such considerations, and is the minimum amount necessary to inhibit mutant RAS (e.g. KRAS G12C) interaction with RAF, blocking the oncogenic MAPK signaling. For example, such amount may be below the amount that is toxic to normal cells, or the mammal as a whole. In one example, the pharmaceutically effective amount of the compound of the invention administered parenterally per dose will be in the range of about 0.1 to 1000 mg/kg, alternatively about 0.1 to 1000 mg/kg of patient body weight per day, with the typical initial range of compound used being 0.3 to 15 mg/kg/day. In another embodiment, oral unit dosage forms, such as tablets and capsules, preferably contain from about 1 to about 1000 mg of the compound of the invention. The compounds of the invention may be administered by any suitable means, including oral, topical (including buccal and sublingual), rectal, vaginal, transdermal, parenteral, subcutaneous, intraperitoneal, intrapulmonary, intradermal, intrathecal and epidural and intranasal, and, if desired for local treatment, intralesional administration. Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration. The compounds of the present invention may be administered in any convenient administrative form, e.g., tablets, powders, capsules, solutions, dispersions, suspensions, syrups, sprays, suppositories, gels, emulsions, patches, etc. Such compositions may contain components conventional in pharmaceutical preparations, e.g., diluents, carriers, pH modifiers, sweeteners, bulking agents, and further active agents. A typical formulation is prepared by mixing a compound of the present invention and a carrier or excipient. Suitable carriers and excipients are well known to those skilled in the art and are described in detail in, e.g., Ansel, Howard C., et al., Ansel’s Pharmaceutical Dosage Forms and Drug Delivery Systems. Philadelphia: Lippincott, Williams & Wilkins, 2004; Gennaro, Alfonso R., et al. Remington: The Science and Practice of Pharmacy. Philadelphia: Lippincott, Williams & Wilkins, 2000; and Rowe, Raymond C. Handbook of Pharmaceutical Excipients. Chicago, Pharmaceutical Press, 2005. The formulations may also include one or more buffers, stabilizing agents, surfactants, wetting agents, lubricating agents, emulsifiers, suspending agents, preservatives, antioxidants, opaquing agents, glidants, processing aids, colorants, sweeteners, perfuming agents, flavoring agents, diluents and other known additives to provide an elegant presentation of the drug (i.e., a compound of the present invention or pharmaceutical composition thereof) or aid in the manufacturing of the pharmaceutical product (i.e., medicament). An example of a suitable oral dosage form is a tablet containing about 1 to 1000 mg of the compound of the invention compounded with about 1 to 1000 mg anhydrous lactose, about 1 to 1000 mg sodium croscarmellose, about 1 to 1000 mg polyvinylpyrrolidone (PVP) K30, and about 1 to 1000 mg magnesium stearate. The powdered ingredients are first mixed together and then mixed with a solution of the PVP. The resulting composition can be dried, granulated, mixed with the magnesium stearate and compressed to tablet form using conventional equipment. An example of an aerosol formulation can be prepared by dissolving the compound, for example 5 to 400mg, of the invention in a suitable buffer solution, e.g. a phosphate buffer, adding a tonicifier, e.g. a salt such sodium chloride, if desired. The solution may be filtered, e.g., using a 0.2 micron filter, to remove impurities and contaminants. An embodiment, therefore, includes a pharmaceutical composition comprising a compound of formula (I), or a stereoisomer or pharmaceutically acceptable salt thereof. In a further embodiment includes a pharmaceutical composition comprising a compound of formula (I), or a stereoisomer or pharmaceutically acceptable salt thereof, together with a pharmaceutically acceptable carrier or excipient. Another embodiment includes a pharmaceutical composition comprising a compound of formula (I) for use in the treatment of mutant KRAS-driven cancers. Another embodiment includes a pharmaceutical composition comprising a compound of Formula (I) for use in the treatment of mutant KRAS-driven cancers. The following composition A and B illustrate typical compositions of the present invention, but serve merely as representative thereof. Composition A A compound of the present invention can be used in a manner known per se as the active ingredient for the production of tablets of the following composition: Per tablet Active ingredient 200 mg Microcrystalline cellulose 155 mg Corn starch 25 mg Talc 25 mg Hydroxypropylmethylcellulose 20 mg 425 mg Composition B A compound of the present invention can be used in a manner known per se as the active ingredient for the production of capsules of the following composition: Per capsule Active ingredient 100.0 mg Corn starch 20.0 mg Lactose 95.0 mg Talc 4.5 mg Magnesium stearate 0.5 mg 220.0 mg INDICATIONS AND METHODS OF TREATMENT The compounds of the invention induce a new binding pocket in KRAS by driving formation of a high affinity tri-complex between KRAS protein and the widely expressed cyclophilin A (CYPA), which inhibit KRAS interaction with downstream effectors, such as RAF and PI3K. Accordingly, the compounds of the invention are useful for inhibiting the propagating oncogenic MAPK and PI3K signaling, reducing cell proliferation, in particular cancer cells. Compounds of the invention are useful for termination of RAS signaling in cells that express RAS mutant, e.g. KRAS mutation driven pancreatic cancer, colorectal cancer, lung cancer, esophageal cancer, gallbladder cancer, melanoma ovarian cancer, endometrial cancer, etc. Alternatively, compounds of the invention are useful for termination of RAS signaling in malignant solid tumor where the oncogenic role of KRAS mutation is reinforced by dysregulation or mutation of effector pathways as MAPK, PI3K-AKT-mTOR (Mammalian target of rapamycin) driven signaling, for targeted therapy in pancreatic adenocarcinoma, colorectal cancer, non-small cell lung cancer, etc. Another embodiment includes a method of treating or preventing cancer in a mammal in need of such treatment, wherein the method comprises administering to said mammal a therapeutically effective amount of a compound of formula (I), a stereoisomer, tautomer, prodrug or pharmaceutically acceptable salt thereof. SYNTHESIS The compounds of the present invention can be prepared by any conventional means. Suitable processes for synthesizing these compounds as well as their starting materials are provided in the schemes below and in the examples. All substituents, in particular, R 1 to R 6 , A 1 and A 2 are as defined above unless otherwise indicated. Furthermore, and unless explicitly otherwise stated, all reactions, reaction conditions, abbreviations and symbols have the meanings well known to a person of ordinary skill in organic chemistry. General synthetic routes for preparing the compound of formula (I) are shown below. Scheme 1 Compound of formula II was synthesized according to the procedure described in Intermediate A to I. Compound of formula (I) can be obtained by a coupling reaction between acid (III) and compound of formula (II) with coupling reagent(s), such as T3P, HATU, PyBOP and EDCI/HOBt, in the presence of a base, such as TEA, DIEPA and DMAP. Compounds of this invention can be obtained as mixtures of diastereomers or enantiomers, which can be separated by methods well known in the art, e.g. (chiral) HPLC or SFC. In another embodiment, compound of formula (I) can be obtained according to above scheme by using corresponding chiral starting materials. This invention also relates to a process for the preparation of a compound of formula (I) comprising following step: a) coupling reaction between compound of formula (II), the presence of a coupling reagent and a base to form the compound of formula (I); wherein in step a) the coupling reagent can be, for example, T 3 P, HATU, PyBOP or EDCI/HOBt; the base can be, for example, TEA, DIEPA or DMAP. A compound of formula (I) or (Ia) when manufactured according to the above process is also an object of the invention. EXAMPLES The invention will be more fully understood by reference to the following examples. They should not, however, be construed as limiting the scope of the invention. ABBREVIATIONS The invention will be more fully understood by reference to the following examples. They should not, however, be construed as limiting the scope of the invention. Abbreviations used herein are as follows: ACN acetonitrile aq. Aqueous Boc-N-Me-Val-OH N-(tert-Butoxycarbonyl)-N-methyl-L-valine (Boc) 2 O Di-tert-butyldicarbonate (R)-binap (R)-(+)-2,2′-Bis(diphenylphosphino)-1,1′-binaphthyl CDCl 3 : deuterated chloroform CD 3 OD: deuterated methanol COMU (1-Cyano-2-ethoxy-2-oxoethylidenaminooxy)dimethylamino- morpholino-carbenium hexafluorophosphate DIEPA: N, N-diethylpropylamine DIBAL-H Diisobutylaluminium hydride DMAP: 4-Dimethylaminopyridine DMF: dimethyl formamide DMP 1,1,1-Tris(acetyloxy)-1,1-dihydro-1,2-benziodoxol-3-(1H)-one DMSO: dimethyl sulfoxide EDCI: N-Ethyl-N′-(3-dimethylaminopropyl)carbodiimide hydrochloride EtOAc or EA: ethyl acetate FRET fluorescence resonance energy transfer HATU: (1-[Bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5- b]pyridinium 3-oxid hexafluorophosphate) hr(s): hour(s) HPLC: high performance liquid chromatography HOBt: N-hydroxybenzotriazole H-VAL-OTBU HCl (S)-tert-Butyl 2-amino-3-methylbutanoate hydrochloride [Ir(OMe)(COD)] 2 (1,5-Cyclooctadiene)(methoxy)iridium(I) dimer LDA Lithium diisopropylamide MS: (ESI): mass spectroscopy (electron spray ionization) min(s) minute(s) MTBE Methyl tert-butyl ether NMM N-Methylmorpholine NMR: nuclear magnetic resonance NMO 4-Methylmorpholine N-oxide obsd. Observed Pd(dppf)Cl2 [1,1′-Bis(diphenylphosphino)ferrocene]dichloropalladium(II ) Pd(dtbpf)Cl 2 [1,1′-Bis(di-tert-butylphosphino)ferrocene]dichloropalladi um(II) prep-HPLC preparative high performance liquid chromatography PyBOP: benzotriazol-1-yloxytripyrrolidinophosphonium hexafluorophosphate RT or rt: room temperature sat. saturated SFC supercritical fluid chromatography TEA: triethylamine TFA: trifluoroacetic acid THF: tetrahydrofuran TEA: trimethylamine TMEDA Tetramethylethylenediamine TMSCF 3 Trifluoromethyltrimethylsilane T 3 P: propylphosphonic anhydride GENERAL EXPERIMENTAL CONDITIONS Intermediates and final compounds were purified by flash chromatography using one of the following instruments: i) Biotage SP1 system and the Quad 12/25 Cartridge module. ii) ISCO combi-flash chromatography instrument. Silica gel brand and pore size: i) KP-SIL 60 Å, particle size: 40-60 µm; ii) CAS registry NO: Silica Gel: 63231-67-4, particle size: 47-60 micron silica gel; iii) ZCX from Qingdao Haiyang Chemical Co., Ltd, pore: 200-300 or 300-400. Intermediates and final compounds were purified by preparative HPLC on reversed phase column using XBridge TM Prep-C18 (5 µm, OBDTM 30 × 100 mm) column, SunFire TM Prep-C18 (5 µm, OBD TM 30 × 100 mm) column, Phenomenex Synergi-C18 (10 µm, 25 × 150 mm) or Phenomenex Gemini-C18 (10 µm, 25 × 150 mm). Waters AutoP purification System (Sample Manager 2767, Pump 2525, Detector: Micromass ZQ and UV 2487, solvent system: acetonitrile and 0.1% ammonium hydroxide in water; acetonitrile and 0.1% FA in water or acetonitrile and 0.1% TFA in water). Or Gilson-281 purification System (Pump 322, Detector: UV 156, solvent system: acetonitrile and 0.05% ammonium hydroxide in water; acetonitrile and 0.225% FA in water; acetonitrile and 0.05% HCl in water; acetonitrile and 0.075% TFA in water; or acetonitrile and water). For SFC chiral separation, intermediates were separated by chiral column (Daicel chiralpak IC, 5 µm, 30 × 250 mm), AS (10 µm, 30 × 250 mm) or AD (10 µm, 30 × 250 mm) using Mettler Toledo Multigram III system SFC, Waters 80Q preparative SFC or Thar 80 preparative SFC, solvent system: CO 2 and IPA (0.5% TEA in IPA) or CO 2 and MeOH (0.1% NH 3 ∙H 2 O in MeOH), back pressure 100bar, detection UV@ 254 or 220 nm. LC/MS spectra of compounds were obtained using a LC/MS (Waters TM Alliance 2795- Micromass ZQ, Shimadzu Alliance 2020-Micromass ZQ or Agilent Alliance 6110-Micromass ZQ), LC/MS conditions were as follows (running time 3 or 1.5 mins): Acidic condition I: A: 0.1% TFA in H2O; B: 0.1% TFA in acetonitrile; Acidic condition II: A: 0.0375% TFA in H 2 O; B: 0.01875% TFA in acetonitrile; Basic condition I: A: 0.1% NH 3 ·H 2 O in H 2 O; B: acetonitrile; Basic condition II: A: 0.025% NH3·H2O in H2O; B: acetonitrile; Neutral condition: A: H2O; B: acetonitrile. Mass spectra (MS): generally only ions which indicate the parent mass are reported, and unless otherwise stated the mass ion quoted is the positive mass ion (MH) + . NMR Spectra were obtained using Bruker Avance 400 MHz or 500MHz. The microwave assisted reactions were carried out in a Biotage Initiator Sixty microwave synthesizer. All reactions involving air-sensitive reagents were performed under an argon or nitrogen atmosphere. Reagents were used as received from commercial suppliers without further purification unless otherwise noted. PREPARATIVE EXAMPLES Preparation of Intermediate Intermediate A 1-[6-[(1S)-1-methoxyethyl]-5-(4,4,5,5-tetramethyl-1,3,2-diox aborolan-2-yl)-3-pyridyl]-4- methyl-piperazine The title intermediate A was prepared according to the following scheme: Intermediate A Step 1: Preparation of 3-bromo-2-[(1S)-1-methoxyethyl]-5-(4,4,5,5-tetramethyl-1,3,2 - dioxaborolan-2-yl)pyridine (compound A2) To a solution of 3-bromo-2-[(1S)-1-methoxyethyl]pyridine (compound A1, 2.0 g, 9.26 mmol) and bis(pinacolato)diboron (3.5 g, 13.9 mmol) in THF (30 mL) were added 4,4'-di-tert- butyl-2,2'-bipyridin (372.7 mg, 1.39 mmol) and [Ir(OMe)(COD)] 2 (306.3 mg, 0.460 mmol). The mixture was stirred at 75 °C for 16 hours under N2 protection. The mixture was filtrated and the filtrate was concentrated in vacuo. The residue was purified by silica gel chromatography (EA/PE: 0-20%) to afford 3-bromo-2-[(1S)-1-methoxyethyl]-5-(4,4,5,5-tetramethyl-1,3,2 - dioxaborolan-2-yl)pyridine (compound A2, 2.4 g) as yellow oil. 1 H NMR (400 MHz, CDCl3) δ ppm 8.91 (d, J = 1.4 Hz, 1 H), 8.21 (d, J = 1.4 Hz, 1 H), 4.95 (q, J = 6.5 Hz, 1 H), 3.30 (s, 3 H), 1.49 (d, J = 6.5 Hz, 3 H), 1.35 (s, 12 H). Step 2: Preparation of 3-bromo-5-iodo-2-[(1S)-1-methoxyethyl]pyridine (compound A3) To a solution of 3-bromo-2-[(1S)-1-methoxyethyl]-5-(4,4,5,5-tetramethyl-1,3,2 - dioxaborolan-2-yl)pyridine (compound A2, 2.5 g, 7.3 mmol) in ACN (40 mL) was added N- iodosuccinimide (4.1 g, 18.27 mmol). The mixture was stirred at 90 °C for 40 hrs under N2 protection. The reaction was quenched with saturated solution of Na2SO3 (40 mL) and the reaction mixture was extracted with EtOAc (30 mL, twice). The combined organic layer was washed with brine (50 mL), filtered and the filtrate was concentrated under vacuum. The residue was purified by silica gel chromatography (EA/PE: 0-20%) to afford 3-bromo-5-iodo-2-[(1S)-1- methoxyethyl]pyridine (compound A3, 660 mg) as yellow oil. MS calc’d 342 (MH + ), measured 341.8 (MH + ). Step 3: Preparation of benzyl 4-[5-bromo-6-[(1S)-1-methoxyethyl]-3- pyridyl]piperazine-1-carboxylate (compound A5) To a solution of 3-bromo-5-iodo-2-[(1S)-1-methoxyethyl]pyridine (compound A3, 660 mg, 1.9 mmol) and 1-Cbz-piperazine (compound A4, 425.1 mg, 1.9 mmol) in toluene (10 mL) were added cesium carbonate (1.6 g, 4.83 mmol), (R)-BINAP (60.1 mg, 0.1 mmol) and palladium (II) acetate (43.3 mg, 0.19 mmol). The mixture was stirred at 100 °C for 12 hours under N2 protection. The mixture was filtered and the filtrate was concentrated under vacuum. The residue was purified by silica gel chromatography (EA/PE: 0-50%) to afford benzyl 4-[5-bromo-6-[(1S)- 1-methoxyethyl]-3-pyridyl]piperazine-1-carboxylate (compound A5, 740 mg) as a yellow solid. MS calc’d 434.1 (MH + ), measured 434.1 (MH + ). Step 4: Preparation of 1-[6-[(1S)-1-methoxyethyl]-5-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)-3-pyridyl]-4-methyl-piperazine (Intermediate A) To a solution of benzyl 4-[5-bromo-6-[(1S)-1-methoxyethyl]-3-pyridyl]piperazine-1- carboxylate (compound A5, 740 mg, 1.7 mmol) and bis(pinacolato)diboron (519.2 mg, 2.04 mmol) in toluene (12 mL) were added KOAc (418.0 mg, 4.26 mmol) and Pd(dppf)Cl2 (124.7 mg, 0.170 mmol). The reaction mixture was stirred at 90 °C for 12 hrs under N2 protection. The mixture was filtered and the filtrate was concentrated in vacuo. The residue was purified by silica gel column to afford 1-[6-[(1S)-1-methoxyethyl]-5-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)-3-pyridyl]-4-methyl-piperazine (Intermediate A, 470 mg) as a brown solid. MS calc’d 482.3 (MH + ), measured 482.2 (MH + ). Intermediate B Methyl (3S)-1-[(2S)-3-(4-bromothiazol-2-yl)-2-(tert-butoxycarbonyla mino)- propanoyl]hexahydropyridazine-3-carboxylate The intermediate B was prepared according to the following scheme: B8 In Step 1: Preparation of (4-bromothiazol-2-yl)methanol (compound B2) To a solution of 4-bromothiazole-2-carboxaldehyde (compound B1 6¸.0 g, 31.25 mmol) in methanol (70 mL) was added sodium borohydride (1.7 g, 46.87 mmol) at 0 °C. The mixture was stirred at 25 °C for 1 hour. The reaction was quenched with water (300 mL) at 0 °C and the reaction mixture was extracted by ethyl acetate (200 mL, three times). The combined organic phase was washed with brine (150 mL, twice), dried over anhydrous sodium sulfate, filtered and the filtrate was concentrated under vacuum to afford (4-bromothiazol-2-yl)methanol (compound B2, 6g) as colorless oil. Step 2: Preparation of 4-bromo-2-(bromomethyl)thiazole (compound B3) To a solution of (4-bromothiazol-2-yl)methanol (compound B2, 6.0 g, 30.92 mmol) in DCM (80 mL) was added CBr4 (15.4 g, 46.38 mmol) and triphenylphosphine (12.1 g, 46.38 mmol) at 0 °C. After being stirred at 25 °C for 1 hour, the mixture was filtered and the filtrate was concentrated under vacuum. The residue was purified by silica gel column, eluted with ethyl acetate in petroleum ether (0~10%) to afford (4-bromothiazol-2-yl)methanol (compound B3, 6.0 g) as yellow oil. MS calc’d 255.9 (MH + ), measured 255.9 (MH + ). Step 3: Preparation of 4-bromo-2-[[(2S,5R)-5-isopropyl-3,6-dimethoxy-2,5- dihydropyrazin-2-yl]methyl]thiazole (compound B5) To a mixture of (R)-2,5-dihydro-3,6-dimethoxy-2-isopropylpyrazine (compound B4, 4.3 g, 23.45 mmol) in THF (60 mL) was added n-butyllithium (10 mL, 25.22 mmol, 2.5 M) at -78 °C slowly. After addition, the mixture was stirred for 0.5 hour at -78 °C.4-bromo-2- (bromomethyl)thiazole (compound B3, 5.4 g, 21.02 mmol) was added into above mixture at - 78 °C which was stirred for another 1 hour. The reaction was quenched with saturated solution of NH4Cl (100 mL) and the reaction mixture was extracted with EtOAc (100 mL, twice). The combined organic layer was washed with brine (150 mL), dried over anhydrous sodium sulfate, filtered and the filtrate was concentrated under vacuum. The residue was purified by reversed- phase chromatography to afford 4-bromo-2-[[(2S,5R)-5-isopropyl-3,6-dimethoxy-2,5- dihydropyrazin-2-yl]methyl]thiazole (compound B5, 3.6 g) as yellow oil. MS calc’d 360 (MH + ), measured 359.9 (MH + ). Step 4: Preparation of methyl (2S)-2-amino-3-(4-bromothiazol-2-yl)propanoate (compound B6) To a solution of 4-bromo-2-[[(2S,5R)-5-isopropyl-3,6-dimethoxy-2,5-dihydropyr azin-2- yl]methyl]thiazole (compound B5, 3.6 g, 10 mmol) in ACN (20 mL) was added hydrochloric acid (66.6 mL, 0.3 M). The mixture was stirred at 25 °C for 2 hours. The mixture was basified by saturated solution of NaHCO3 until pH=8. The mixture was extracted with EtOAc (80 mL, six times). The combined organic layer was dried over anhydrous sodium sulfate, filtered and the filtrate was concentrated under vacuum to afford methyl (2S)-2-amino-3-(4-bromothiazol-2- yl)propanoate (compound B6, 3.1 g) as yellow oil. MS calc’d 264.9 (MH + ), measured 264.9 (MH + ). Step 5: Preparation of methyl (2S)-3-(4-bromothiazol-2-yl)-2-(tert- butoxycarbonylamino)propanoate (compound B7) To a solution of methyl (2S)-2-amino-3-(4-bromothiazol-2-yl)propanoate (compound B6, 3.1 g, 11.69 mmol) in DCM (40 mL) were added triethylamine (2.9 g, 29.23 mmol) and (Boc) 2 O (3.8 g, 17.54 mmol). After being stirred at 30 °C for 12 hours, the mixture was concentrated under vacuum. The residue was purified by silica gel column, eluted with ethyl acetate in petroleum ether (0~30%) to afford methyl (2S)-3-(4-bromothiazol-2-yl)-2-(tert- butoxycarbonylamino)propanoate (compound B7, 3.2 g) as yellow oil. MS calc’d 387(MNa + ), measured 386.9 (MNa + ). Step 6: Preparation of (2S)-3-(4-bromothiazol-2-yl)-2-(tert-butoxycarbonylamino)- propanoic acid (compound B8) To a solution of methyl (2S)-3-(4-bromothiazol-2-yl)-2-(tert- butoxycarbonylamino)propanoate (compound B7, 3.2 g, 8.76 mmol) in THF (30 mL), methanol (2 mL) and water (10 mL) was added lithium hydroxide (0.4 mL, 43.81 mmol). After being stirred at 25 °C for 1 hour, the reaction mixture was acidified by 1 M solution of HCl until pH=5. The mixture was extracted with EtOAc (40 mL, twice). The combined organic layer was washed with brine (100 mL), dried over anhydrous sodium sulfate, filtered and the filtrate was concentrated under vacuum to afford (2S)-3-(4-bromothiazol-2-yl)-2-(tert- butoxycarbonylamino)propanoic acid (compound B8, 3.1 g) as yellow oil. MS calc’d 373(MNa + ), measured 372.9 (MNa + ). Step 7: Preparation of methyl (3S)-1-[(2S)-3-(4-bromothiazol-2-yl)-2-(tert- butoxycarbonylamino)propanoyl]hexahydropyridazine-3-carboxyl ate (Intermediate B) To a solution of (2S)-3-(4-bromothiazol-2-yl)-2-(tert-butoxycarbonylamino)pro panoic acid (compound B8, 3.1 g, 8.83 mmol) in DCM (50 mL) was added methyl (3S)- hexahydropyridazine-3-carboxylate;hydrochloride (compound B9, 2.4 g, 13.24 mmol), EDCI (3.4 g, 17.65 mmol), 1-Hydroxybenzotriazole (238.5 mg, 1.77 mmol) and NMM (9.92 mL, 88.26 mmol) at 0 °C. After being stirred at 25 °C for 1 hour, the reaction mixture was diluted with water (60 mL) and extracted with EtOAc (60 mL, three times). The combined organic layer was washed with brine (100 mL), dried over anhydrous sodium sulfate, filtered and the filtrate was concentrated under vacuum. The residue was purified by silica gel column and eluted with ethyl acetate in petroleum ether (10~30%) to afford methyl (3S)-1-[(2S)-3-(4-bromothiazol-2-yl)-2- (tert-butoxycarbonylamino)propanoyl]hexahydropyridazine-3-ca rboxylate (intermediate B, 2.4 g). MS calc’d 477(MH + ), measured 476.9 (MH + ). Intermediate C (7S,13S)-7-amino-24-fluoro-(20M)-20-[2-[(1S)-1-methoxyethyl] -5-(4-methylpiperazin-1-yl)- 3-pyridyl]-17,17-dimethyl-21-(2,2,2-trifluoroethyl)-15-oxa-4 -thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaene-8,14- dione The title intermediate C was prepared according to the following scheme:

Step 1: Preparation of 1-(5-bromo-6-fluoro-1H-indol-3-yl)-3-((tert-butyldiphenylsil yl) oxy)-2,2-dimethylpropan-1-one (compound C3) To a mixture of 3-((tert-butyldiphenylsilyl)oxy)-2,2-dimethylpropanoyl chloride (compound C1, 35.0 g, 116.8 mmol) in DCM (400 mL) at 0 °C was added a solution of SnCl 4 (97.2 mL, 121.5 mmol) slowly. After being stirred at - 40 °C for 0.5 hour, 5-bromo-6-fluoro-1H- indole (compound C2, 25.0 g, 116.8 mmol) in DCM (200 mL) was added dropwise to the mixture which was stirred at - 40 °C for 15 min. After the reaction was completed, it was quenched with sat.NaHCO3 aq. (800 mL), and the reaction mixture was extracted with EtOAc (900 mL, twice). The combined organic layer was washed with brine (700 mL), dried over Na 2 SO 4 , filtered and concentrated in vacuo. The residue was triturated with the solution (100 mL, Petroleum ether: Ethyl acetate = 8:1) and filtered. The filter cake was dried in vacuo to afford 1- (5-bromo-6-fluoro-1H-indol-3-yl)-3-((tertbutyldiphenylsilyl) oxy)-2,2-dimethylpropan-1-one (compound C3, 50.0 g) as a yellow solid. MS calc’d 552.1 (MH + ), measured 552.1 (MH + ). Step 2: Preparation of [3-(5-bromo-6-fluoro-1H-indol-3-yl)-2,2-dimethyl-propoxy]- tert-butyl-diphenyl-silane (compound C4) To a mixture of 1-(5-bromo-6-fluoro-1H-indol-3-yl)-3-((tertbutyldiphenylsily l)oxy)-2,2- dimethylpropan-1-one (compound C3, 50.0 g, 90.49 mmol) in THF (600 mL) was added LiBH 4 (48.4 mL, 193.49 mmol, 4 M in THF) dropwise at 0 °C. The mixture was stirred at 70 °C for 24 hrs under nitrogen atmosphere. After the reaction was completed, it was quenched by addition of water (600 mL) at 0 °C slowly and the reaction mixture was extracted with EtOAc (600 mL, twice). The combined organic layer was washed with brine (600 mL), dried over Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica column chromatography (EtOAc in PE = 20% ~ 33%) to afford [3-(5-bromo-6-fluoro-1H-indol-3-yl)-2,2-dimethyl- propoxy]-tert-butyl-diphenyl-silane (compound C4, 46.0 g) as a white solid. MS calc’d 538.1 (MH + ), measured 538.2 (MH + ). Step 3: Preparation of [3-(5-bromo-6-fluoro-2-iodo-1H-indol-3-yl)-2,2-dimethyl- propoxy]-tert-butyl-diphenyl-silane (compound C5) To a mixture of [3-(5-bromo-6-fluoro-1H-indol-3-yl)-2,2-dimethyl-propoxy]-te rt-butyl- diphenyl-silane (compound C4, 35.4 g, 65.73 mmol) and iodine (18.4 g, 72.3 mmol) in THF (400 mL) was added silver trifluoromethanesulfonate (20.3 g, 78.88 mmol) at 0 °C. The mixture was stirred at 0 °C for 10 min. After the reaction was completed, it was quenched by sat. Na 2 SO 3 aq. (400 mL) and EtOAc (400 mL) and the reaction mixture was filtered. The organic layer was washed with brine (100 mL), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica column chromatography (EtOAc in PE = 0% ~ 2.5%) to afford [3-(5- bromo-6-fluoro-2-iodo-1H-indol-3-yl)-2,2-dimethyl-propoxy]-t ert-butyl-diphenyl-silane (compound C5, 43.0 g) as a yellow solid. MS calc’d 664.0 (MH + ), measured 664.1 (MH + ). Step 4: Preparation of benzyl 4-[5-[5-bromo-3-[3-[tert-butyl(diphenyl)silyl]oxy-2,2- dimethyl-propyl]-6-fluoro-1H-indol-2-yl]-6-[(1S)-1-methoxyet hyl]-3-pyridyl]piperazine-1- carboxylate (compound C6) To a mixture of [3-(5-bromo-6-fluoro-2-iodo-1H-indol-3-yl)-2,2-dimethyl-prop oxy]-tert- butyl-diphenyl-silane (compound C5, 16.7 g, 25.13 mmol) and benzyl 4-[6-[(1S)-1- methoxyethyl]-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl )-3-pyridyl]piperazine-1- carboxylate (Intermediate A, 16.7 g, 34.69 mmol) in a mixed solution of 1,4-dioxane (270 mL)/Toluene (90 mL) /water (90 mL) were added potassium phosphate (15.7 g, 73.92 mmol) and Pd(dppf)Cl2 (920 mg, 1.26 mmol). The mixture was stirred at 70 °C for 12 hrs under nitrogen atmosphere. After the reaction was completed, the mixture was filtered and concentrated in vacuo. The residue was purified by silica column chromatography (EtOAc in PE = 20% ~ 50%) to afford 4-[5-[5-bromo-3-[3-[tert-butyl(diphenyl)silyl]oxy-2,2-dimeth yl-propyl]- 6-fluoro-1H-indol-2-yl]-6-[(1S)-1-methoxyethyl]-3-pyridyl]pi perazine-1-carboxylate (compound C6, 19.5 g) as a white solid. MS calc’d 891.3 (MH + ), measured 891.3 (MH + ). Step 5: Preparation of benzyl 4-[(5M)-5-[5-bromo-3-[3-[tert-butyl(diphenyl)silyl]oxy- 2,2-dimethyl-propyl]-6-fluoro-1-(2,2,2-trifluoroethyl)indol- 2-yl]-6-[(1S)-1-methoxyethyl]-3- pyridyl]piperazine-1-carboxylate(compound C7) To a solution of 4-[5-[5-bromo-3-[3-[tert-butyl(diphenyl)silyl]oxy-2,2-dimeth yl-propyl]-6- fluoro-1H-indol-2-yl]-6-[(1S)-1-methoxyethyl]-3-pyridyl]pipe razine-1-carboxylate (compound C6, 14.5 g, 16.26 mmol) and Cs 2 CO 3 (15.9 g, 48.77 mmol) in DMF (200 mL) was added 2,2,2- trifluoroethyl trifluoromethanesulfonate (37.7 g, 162.56 mmol) dropwise at 0 °C, and the mixture was stirred at 20 °C for 12 hrs. After the reaction was completed, EtOAc (70 mL) and water (100 mL) were added and the layers were separated. The aqueous phase was extracted with EtOAc (70 mL, twice). Combined organic layer was washed with brine (100 mL, four times), dried over Na2SO4, filtered, and concentrated under vacuum to give a residue. The residue was purified by silica column chromatography to afford benzyl 4-[(5M)-5-[5-bromo-3-[3-[tert- butyl(diphenyl)silyl]oxy-2,2-dimethyl-propyl]-6-fluoro-1-(2, 2,2-trifluoroethyl)indol-2-yl]-6- [(1S)-1-methoxyethyl]-3-pyridyl]piperazine-1-carboxylate (compound C7, 8.0 g, PEAK 1, faster eluted) as yellow oil. MS calc’d 973.3 (MH + ), measured 973.2 (MH + ). Step 6: Preparation of benzyl 4-[(5M)-5-[5-bromo-6-fluoro-3-(3-hydroxy-2,2- dimethyl-propyl)-1-(2,2,2-trifluoroethyl)indol-2-yl]-6-[(1S) -1-methoxyethyl]-3- pyridyl]piperazine-1-carboxylate (compound C8) To a solution of benzyl 4-[(5M)-5-[5-bromo-3-[3-[tert-butyl(diphenyl)silyl]oxy-2,2- dimethyl-propyl]-6-fluoro-1-(2,2,2-trifluoroethyl)indol-2-yl ]-6-[(1S)-1-methoxyethyl]-3- pyridyl]piperazine-1-carboxylate (compound C7, 10.5 g, 10.78 mmol) in DMF (130 mL) was added cesium fluoride (8.2 g, 53.9 mmol) and the mixture was stirred at 60 °C for 24 hrs. After the reaction was completed, EtOAc (100 mL) and water (100 mL) were added and the layers were separated. The aqueous phase was extracted with EtOAc (100 mL, twice). The combined organic layer was washed with brine (80 mL, three times), dried over Na2SO4, filtered, and concentrated under vacuum to give a residue. The residue was purified by silica column chromatography (EtOAc in PE = 25% ~ 66%) to afford benzyl 4-[(5M)-5-[5-bromo-6-fluoro-3- (3-hydroxy-2,2-dimethyl-propyl)-1-(2,2,2-trifluoroethyl)indo l-2-yl]-6-[(1S)-1-methoxyethyl]-3- pyridyl]piperazine-1-carboxylate (compound C8, 6.5 g) as a yellow solid. MS calc’d 735.2 (MH + ), measured 735.1 (MH + ). Step 7: Preparation of benzyl 4-[(5M)-5-[6-fluoro-3-(3-hydroxy-2,2-dimethyl-propyl)- 5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1-(2,2,2-tri fluoroethyl)indol-2-yl]-6-[(1S)-1- methoxyethyl]-3-pyridyl]piperazine-1-carboxylate (compound C9) To a solution of benzyl 4-[(5M)-5-[5-bromo-6-fluoro-3-(3-hydroxy-2,2-dimethyl-propyl )- 1-(2,2,2-trifluoroethyl)indol-2-yl]-6-[(1S)-1-methoxyethyl]- 3-pyridyl]piperazine-1-carboxylate (compound C8, 5.4 g) , bis(pinacolato)diboron (2.8 g, 11.01 mmol) and potassium acetate (1.2 mL, 18.35 mmol) in toluene (70 mL) was added Pd(dppf)Cl 2 (537.1 mg, 0.73 mmol). The mixture was degassed and purged with nitrogen atmosphere for three times and the mixture was stirred at 90 °C for 12 hrs. After the reaction was completed, the mixture was cooled to room temperature. The reaction mixture was filtered and the filtrate was concentrated in vacuo to give a residue. The residue was purified by silica column chromatography (EtOAc in PE = 25% ~ 66%) to afford benzyl 4-[(5M)-5-[6-fluoro-3-(3-hydroxy-2,2-dimethyl-propyl)-5-(4,4 ,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)-1-(2,2,2-trifluoroethyl )indol-2-yl]-6-[(1S)-1- methoxyethyl]-3-pyridyl]piperazine-1-carboxylate (compound C9, 5.2 g) as yellow oil. MS calc’d 783.3 (MH + ), measured 783.3 (MH + ). Step 8: Preparation of methyl (3S)-1-[(2S)-3-[4-[(2M)-2-[5-(4- benzyloxycarbonylpiperazin-1-yl)-2-[(1S)-1-methoxyethyl]-3-p yridyl]-6-fluoro-3-(3- hydroxy-2,2-dimethyl-propyl)-1-(2,2,2-trifluoroethyl)indol-5 -yl]thiazol-2-yl]-2-(tert- butoxycarbonylamino)-propanoyl]hexahydropyridazine-3-carboxy late (compound C10) To a mixture of methyl (3S)-1-[(2S)-3-(4-bromothiazol-2-yl)-2-(tert- butoxycarbonylamino)propanoyl]hexahydropyridazine-3-carboxyl ate (intermediate B, 2.7 g, 5.69 mmol), benzyl 4-[(5M)-5-[6-fluoro-3-(3-hydroxy-2,2-dimethyl-propyl)-5-(4,4 ,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)-1-(2,2,2-trifluoroethyl )indol-2-yl]-6-[(1S)-1- methoxyethyl]-3-pyridyl]piperazine-1-carboxylate (compound C9, 4.9 g, 6.32 mmol) in toluene (60 mL)/1,4-dioxane (20 mL) / water (20 mL) were added K 3 PO 4 (3.4 g, 15.81 mmol) and Pd(dtbpf)Cl 2 (412.2 mg, 0.63 mmol) under nitrogen atmosphere. The mixture was stirred at 70 °C for 12 hrs. After the reaction was completed, the mixture was concentrated in vacuo to give a residue. The residue was purified by silica column (EtOAc in PE = 10% ~ 75%) to afford methyl (3S)-1-[(2S)-3-[4-[(2M)-2-[5-(4-benzyloxycarbonylpiperazin-1 -yl)-2-[(1S)-1-methoxyethyl]-3- pyridyl]-6-fluoro-3-(3-hydroxy-2,2-dimethyl-propyl)-1-(2,2,2 -trifluoroethyl)indol-5-yl]thiazol-2- yl]-2-(tert-butoxycarbonylamino)-propanoyl]hexahydropyridazi ne-3-carboxylate (compound C10, 3.6 g) as a brown solid. MS calc’d 1053.4 (MH + ), measured 1053.3 (MH + ). Step 9: Preparation of (3S)-1-[(2S)-3-[4-[(2M)-2-[5-(4-benzyloxycarbonylpiperazin-1 - yl)-2-[(1S)-1-methoxyethyl]-3-pyridyl]-6-fluoro-3-(3-hydroxy -2,2-dimethyl-propyl)-1-(2,2,2- trifluoroethyl)indol-5-yl]thiazol-2-yl]-2-(tert-butoxycarbon ylamino)propanoyl]hexahy- dropyridazine-3-carboxylic acid (compound C11) To a solution of methyl (3S)-1-[(2S)-3-[4-[(2M)-2-[5-(4-benzyloxycarbonylpiperazin-1 - yl)-2-[(1S)-1-methoxyethyl]-3-pyridyl]-6-fluoro-3-(3-hydroxy -2,2-dimethyl-propyl)-1-(2,2,2- trifluoroethyl)indol-5-yl]thiazol-2-yl]-2-(tert-butoxycarbon ylamino)-propanoyl]- hexahydropyridazine-3-carboxylate (compound C10, 3.6 g, 3.42 mmol) in DCE (50 mL) was added trimethylstannanol (2.4 g, 13.67 mmol) and the mixture was stirred at 60 °C for 12 hrs. After the reaction was completed, EtOAc (80 mL) and water (60 mL) were added and the layers were separated. The aqueous phase was extracted with EtOAc (80 mL, twice). The combined organic layer was washed with brine (100 mL), dried over Na2SO4, filtered, and concentrated under vacuum to give (3S)-1-[(2S)-3-[4-[(2M)-2-[5-(4-benzyloxycarbonylpiperazin-1 -yl)-2- [(1S)-1-methoxyethyl]-3-pyridyl]-6-fluoro-3-(3-hydroxy-2,2-d imethyl-propyl)-1-(2,2,2- trifluoroethyl)indol-5-yl]thiazol-2-yl]-2-(tert-butoxycarbon ylamino)propanoyl]hexahy- dropyridazine-3-carboxylic acid (compound C11, 4.3 g) as a brown solid. MS calc’d 1039.4 (MH + ), measured 1039.2 (MH + ). Step 10: Preparation of benzyl 4-[5-[(7S,13S)-7-(tert-butoxycarbonylamino)-24- fluoro-17,17-dimethyl-8,14-dioxo-21-(2,2,2-trifluoroethyl)-1 5-oxa-4-thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaen-(20M)- 20-yl]-6-[(1S)-1-methoxyethyl]-3-pyridyl]piperazine-1-carbox ylate (compound C12) To a mixture of (3S)-1-[(2S)-3-[4-[(2M)-2-[5-(4-benzyloxycarbonylpiperazin-1 -yl)-2- [(1S)-1-methoxyethyl]-3-pyridyl]-6-fluoro-3-(3-hydroxy-2,2-d imethyl-propyl)-1-(2,2,2- trifluoroethyl)indol-5-yl]thiazol-2-yl]-2-(tert-butoxycarbon ylamino)propanoyl]hexahy- dropyridazine-3-carboxylic acid (compound C11, 4.3 g, 4.14 mmol) in DCM (430 mL) was added DIEA (14.4 mL, 82.76 mmol), EDCI (11.9 g, 62.07 mmol) and 1-hydroxybenzotriazole (1.4 g, 10.35 mmol) at 0 °C. The mixture was stirred at 15 °C for 12 hrs. After the reaction was completed, the mixture was concentrated in vacuo, then diluted with water (80 mL), extracted with EtOAc (80 mL, twice). The combined organic layer was washed with brine (80 mL), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica column chromatography (EtOAc in PE = 25% ~ 66%) to afford benzyl 4-[5-[(7S,13S)-7-(tert- butoxycarbonylamino)-24-fluoro-17,17-dimethyl-8,14-dioxo-21- (2,2,2-trifluoroethyl)-15-oxa-4- 2,5 9,13 22,26 thia-9,21,27,28-tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23- hexaen-(20M)-20-yl]-6-[(1S)-1-methoxyethyl]-3-pyridyl]pipera zine-1-carboxylate (compound C12, 3.1 g) as yellow gum. MS calc’d 1021.4 (MH+), measured 1021.2 (MH+). Step 11: Preparation of tert-butyl N-[(7S,13S)-24-fluoro-(20M)-20-[2-[(1S)-1- methoxyethyl]-5-(4-methylpiperazin-1-yl)-3-pyridyl]-17,17-di methyl-8,14-dioxo-21-(2,2,2- 2,5 9,13 22,26 trifluoroethyl)-15-oxa-4-thia-9,21,27,28-tetrazapentacyclo[1 7.5.2.1 .1 .0 ]octacosa- 1(25),2,5(28),19,22(26),23-hexaen-7-yl]carbamate (compound C13) To a mixture of benzyl 4-[5-[(7S,13S)-7-(tert-butoxycarbonylamino)-24-fluoro-17,17- dimethyl-8,14-dioxo-21-(2,2,2-trifluoroethyl)-15-oxa-4-thia- 9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaen-(20M)-20- yl]-6-[(1S)-1-methoxyethyl]-3-pyridyl]piperazine-1-carboxyla te (compound C12, 3.1 g, 3.04 mmol) and formaldehyde aqueous (775.0 mg, 9.55 mmol) in methanol (150 mL) was added Pd(OH) 2 on activated carbon (2.79 g, 3.97 mmol). The mixture was degassed and purged with H 2 three times. The mixture was hydrogenated at 30 °C for 18 hrs. After the reaction was completed, the mixture was filtered and the filtrate was concentrated in vacuo to afford tert-butyl N- [(7S,13S)-24-fluoro-(20M)-20-[2-[(1S)-1-methoxyethyl]-5-(4-m ethylpiperazin-1-yl)-3-pyridyl]- 17,17-dimethyl-8,14-dioxo-21-(2,2,2-trifluoroethyl)-15-oxa-4 -thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaen-7- yl]carbamate (compound C13, 2.6 g) as a brown solid. MS calc’d 901.3 (MH + ), measured 901.3 (MH + ). Step 12: Preparation of (7S,13S)-7-amino-24-fluoro-(20M)-20-[2-[(1S)-1- methoxyethyl]-5-(4-methylpiperazin-1-yl)-3-pyridyl]-17,17-di methyl-21-(2,2,2- 2,5 9,13 22,26 trifluoroethyl)-15-oxa-4-thia-9,21,27,28-tetrazapentacyclo[1 7.5.2.1 .1 .0 ]octacosa- 1(25),2,5(28),19,22(26),23-hexaene-8,14-dione (intermediate C) To a mixture of tert-butyl N-[(7S,13S)-24-fluoro-(20M)-20-[2-[(1S)-1-methoxyethyl]-5-(4 - methylpiperazin-1-yl)-3-pyridyl]-17,17-dimethyl-8,14-dioxo-2 1-(2,2,2-trifluoroethyl)-15-oxa-4- 2,5 9,13 22,26 thia-9,21,27,28-tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23- hexaen-7-yl]carbamate (compound C13, 2.6 g, 2.89 mmol) in DCM (18 mL) was added TFA (14.0 mL, 181.72 mmol). The mixture was stirred at 15 °C for 0.5 h. After the reaction was completed, the mixture was concentrated in vacuo and diluted with sat. NaHCO 3 (30 mL), extracted with EtOAc (30 mL, three times). The combined organic layer was washed with brine (50 mL), dried over Na2SO4, filtered and concentrated in vacuo to afford (7S,13S)-7-amino-24- fluoro-(20M)-20-[2-[(1S)-1-methoxyethyl]-5-(4-methylpiperazi n-1-yl)-3-pyridyl]-17,17- dimethyl-21-(2,2,2-trifluoroethyl)-15-oxa-4-thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaene-8,14-dione (intermediate C, 2.0 g) as a yellow solid, which was used directly in the next step. MS calc’d 801.3 (MH + ), measured 801.2 (MH + ) Intermediate D

(7S,13S)-7-amino-21-ethyl-24-fluoro-(20M)-20-[2-[(1S)-1-m ethoxyethyl]-5-(4- methylpiperazin-1-yl)-3-pyridyl]-17,17-dimethyl-15-oxa-4-thi a-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaene-8,14- dione The title compound was prepared in analogy to the preparation of Intermediate C by using iodoethane instead of 2,2,2-trifluoroethyl trifluoromethanesulfonate. Intermediate E (7S,13S)-7-amino-24-fluoro-(20M)-20-[2-[(1S)-1-methoxyethyl] -5-[4-(2,2,2- trifluoroethyl)piperazin-1-yl]-3-pyridyl]-17,17-dimethyl-21- (2,2,2-trifluoroethyl)-15-oxa-4- 2,5 9,13 22,26 thia-9,21,27,28-tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23- hexaene-8,14-dione The compound was prepared according to the following scheme: E10 Intermediate E Step 1: Preparation of 1-[5-bromo-6-[(1S)-1-methoxyethyl]-3-pyridyl]-4-(2,2,2- trifluoroethyl)piperazine (compound E2). To a mixture of 3-bromo-5-iodo-2-[(1S)-1-methoxyethyl]pyridine (compound A3, 2.03 g, 5.95 mmol) and 1-(2,2,2-trifluoroethyl)piperazine (compound E1, 1.0 g, 5.95 mmol) in toluene (15 mL) were added Cs2CO3 (4.85 g, 14.88 mmol), (R)-binap (92.6 mg, 0.15 mmol) and Pd(OAc) 2 (66.8 mg, 0.3 mmol). The reaction mixture was degassed and purged with nitrogen for 3 times and the mixture was stirred at 100 °C for 12 hrs under nitrogen atmosphere. After being cooled to room temperature, the reaction mixture was filtered and the filtrate was concentrated in vacuo to give a residue. The residue was purified by column chromatography to 1-[5-bromo-6- [(1S)-1-methoxyethyl]-3-pyridyl]-4-(2,2,2-trifluoroethyl)pip erazine (compound E2, 2.0 g) as yellow oil. MS calc’d 382.2 (MH + ), measured 382.1 (MH + ) Step 2: 1-[6-[(1S)-1-methoxyethyl]-5-(4,4,5,5-tetramethyl-1,3,2-diox aborolan-2-yl)-3- pyridyl]-4-(2,2,2-trifluoroethyl)piperazine (compound E3). To a solution of 1-[5-bromo-6-[(1S)-1-methoxyethyl]-3-pyridyl]-4-(2,2,2- trifluoroethyl)piperazine (compound E2, 3.2 g, 8.37 mmol), bis(pinacolato)diboron (3.19 g, 12.56 mmol) and KOAc (2.1 g, 20.93 mmol) in toluene (50 mL) was added Pd(dppf)Cl2 (306.3 mg, 0.42 mmol). The mixture was degassed and purged with nitrogen for 3 times and the mixture was stirred at 90 °C for 12 hrs under nitrogen atmosphere. After being cooled to the room temperature, the reaction mixture was filtered, the filtrate was concentrated in vacuo to give a residue, which was purified by reversed phase column to afford 1-[6-[(1S)-1-methoxyethyl]-5- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3-pyridyl]-4-( 2,2,2-trifluoroethyl)piperazine (compound E3, 1.9 g) as a yellow gum. MS calc’d 430.2 (MH + ), measured 348.4 (M-C6H10+H + ). Step 3: Preparation of [3-[5-bromo-6-fluoro-2-[2-[(1S)-1-methoxyethyl]-5-[4-(2,2,2- trifluoroethyl)piperazin-1-yl]-3-pyridyl]-1H-indol-3-yl]-2,2 -dimethyl-propoxy]-tert-butyl- diphenyl-silane (compound E4). To a solution of 1-[6-[(1S)-1-methoxyethyl]-5-(4,4,5,5-tetramethyl-1,3,2-diox aborolan-2- yl)-3-pyridyl]-4-(2,2,2-trifluoroethyl)piperazine (compound E3, 1.9 g, 4.41 mmol), [3-(5-bromo- 6-fluoro-2-iodo-1H-indol-3-yl)-2,2-dimethyl-propoxy]-tert-bu tyl-diphenyl-silane (compound C5, 2.1 g, 3.15 mmol) in 1,4-dioxane (24 mL), water (8 mL) and toluene (8 mL) was added K3PO4 (2.1 g, 9.5 mmol) and Pd(dppf)Cl 2 (231 mg, 0.37 mmol). The mixture was degassed by bubbling nitrogen for 2 min, and the reaction mixture was stirred at 70 °C for 12 hrs. After being cooled to room temperature, the reaction mixture was filtered. The filtrate was concentrated in vacuo to give a residue. The residue was purified by column chromatography (EtOAc in PE : 30% - 60%) to afford [3-[5-bromo-6-fluoro-2-[2-[(1S)-1-methoxyethyl]-5-[4-(2,2,2- trifluoroethyl)piperazin- 1-yl]-3-pyridyl]-1H-indol-3-yl]-2,2-dimethyl-propoxy]-tert-b utyl-diphenyl-silane (compound E4, 960.0 mg) as a yellow gum. MS calc’d 839.3 (MH + ), measured 839.3 (MH + ) Step 4: Preparation of [3-[5-bromo-6-fluoro-(2M)-2-[2-[(1S)-1-methoxyethyl]-5-[4- (2,2,2-trifluoroethyl)piperazin-1-yl]-3-pyridyl]-1-(2,2,2-tr ifluoroethyl)indol-3-yl]-2,2- dimethyl-propoxy]-tert-butyl-diphenyl-silane (compound E5). To a solution of [3-[5-bromo-6-fluoro-2-[2-[(1S)-1-methoxyethyl]-5-[4-(2,2,2- trifluoroethyl)piperazin-1-yl]-3-pyridyl]-1H-indol-3-yl]-2,2 -dimethyl-propoxy]-tert-butyl- diphenyl-silane (compound E4, 1 g, 1.14 mmol) in DMF (35 mL) was added Cs2CO3 (1.1 g, 3.44 mmol) and 2,2,2-trifluoroethyl trifluoromethanesulfonate (2.7 g, 11.63 mmol) at 0 °C. After being stirred at 20 °C for 15 hrs, the reaction mixture was poured into water (100 mL), and extracted with EtOAc (50 mL, three times). The combined organic was washed with brine (50 mL, three times), dried over Na2SO4, filtered and concentrated under vacuum to give a residue which was purified by column chromatography (EtOAc in PE: 30% - 40%) to afford [3-[5- bromo-6-fluoro-(2M)-2-[2-[(1S)-1-methoxyethyl]-5-[4-(2,2,2-t rifluoroethyl)piperazin-1-yl]-3- pyridyl]-1-(2,2,2-trifluoroethyl)indol-3-yl]-2,2-dimethyl-pr opoxy]-tert-butyl-diphenyl-silane (compound E5, 640.0 mg, faster eluted) as a white solid. MS calc’d 921.3 (MH + ), measured 921.4 (MH + ). Step 5: Preparation of 3-[5-bromo-6-fluoro-(2M)-2-[2-[(1S)-1-methoxyethyl]-5-[4- (2,2,2-trifluoroethyl)piperazin-1-yl]-3-pyridyl]-1-(2,2,2-tr ifluoroethyl)indol-3-yl]-2,2- dimethyl-propan-1-ol (compound E6). To a solution of [3-[5-bromo-6-fluoro-(2M)-2-[2-[(1S)-1-methoxyethyl]-5-[4-(2 ,2,2- trifluoroethyl)piperazin-1-yl]-3-pyridyl]-1-(2,2,2-trifluoro ethyl)indol-3-yl]-2,2-dimethyl- propoxy]-tert-butyl-diphenyl-silane (compound E5, 640.0 mg, 0.69 mmol) in DMF (7 mL) was added cesium fluoride (421.8 mg, 2.78 mmol). The mixture was stirred at 60 °C for 16 hrs. After being cooled to room temperature, the reaction mixture was filtered and the filtrate was concentrated in vacuo to give a residue. The residue was purified by column chromatography (EtOAc in PE : 30% - 60%) to afford 3-[5-bromo-6-fluoro-(2M)-2-[2-[(1S)-1-methoxyethyl]-5- [4-(2,2,2-trifluoroethyl)piperazin-1-yl]-3-pyridyl]-1-(2,2,2 -trifluoroethyl)indol-3-yl]-2,2- dimethyl-propan-1-ol (compound E6, 360.0 mg) as yellow oil. MS calc’d 683.2 (MH + ), measured 683.1 (MH + ). Step 6: Preparation of 3-[5-bromo-6-fluoro-(2M)-2-[2-[(1S)-1-methoxyethyl]-5-[4- (2,2,2-trifluoroethyl)piperazin-1-yl]-3-pyridyl]-1-(2,2,2-tr ifluoroethyl)indol-3-yl]-2,2- dimethyl-propan-1-ol (compound E7). To a solution of 3-[5-bromo-6-fluoro-(2M)-2-[2-[(1S)-1-methoxyethyl]-5-[4-(2, 2,2- trifluoroethyl)piperazin-1-yl]-3-pyridyl]-1-(2,2,2-trifluoro ethyl)indol-3-yl]-2,2-dimethyl-propan- 1-ol (compound E6, 360.0 mg, 0.53 mmol), bis(pinacolato)diboron (200.6 mg, 0.79 mmol) in toluene (6 mL) was added potassium acetate (0.08 mL, 1.32 mmol) and Pd(dppf)Cl2 (40 mg, 0.1 mmol). The reaction mixture was degassed by bubbling nitrogen for 5 min then stirred at 80 °C for 15 hrs. After being cooled to room temperature, the reaction mixture was filtered and the filtrate was concentrated in vacuo to give a residue. The residue was purified by column chromatography (EtOAc in PE : 30% - 50%) to afford 3-[5-bromo-6-fluoro-(2M)-2-[2-[(1S)-1- methoxyethyl]-5-[4-(2,2,2-trifluoroethyl)piperazin-1-yl]-3-p yridyl]-1-(2,2,2-trifluoroethyl)indol- 3-yl]-2,2-dimethyl-propan-1-ol (compound E7, 300.0 mg) as yellow gum. MS calc’d 731.4 (MH + ), measured 731.4 (MH + ). Step 7: Preparation of methyl (3S)-1-[(2S)-2-(tert-butoxycarbonylamino)-3-[4-[6- fluoro-3-(3-hydroxy-2,2-dimethyl-propyl)-(2M)-2-[2-[(1S)-1-m ethoxyethyl]-5-[4-(2,2,2- trifluoroethyl)piperazin-1-yl]-3-pyridyl]-1-(2,2,2-trifluoro ethyl)indol-5-yl]thiazol-2- yl]propanoyl]hexahydropyridazine-3-carboxylate (compound E8). To a mixture of 3-[5-bromo-6-fluoro-(2M)-2-[2-[(1S)-1-methoxyethyl]-5-[4-(2, 2,2- trifluoroethyl)piperazin-1-yl]-3-pyridyl]-1-(2,2,2-trifluoro ethyl)indol-3-yl]-2,2-dimethyl-propan- 1-ol (compound E7, 0.3 g, 0.41 mmol) and methyl (3S)-1-[(2S)-3-(4-bromothiazol-2-yl)-2-(tert- butoxycarbonylamino)propanoyl]hexahydropyridazine-3-carboxyl ate (intermediate B, 196.7 mg, 0.41 mmol) in toluene (3 mL), 1,4-dioxane (1 mL) and water (1 mL) were added K3PO4 (221.3 mg, 1.04 mmol) and Pd(dtbpf)Cl 2 (27.05 mg, 0.04 mmol). The mixture was stirred at 70 °C for 12 hrs under nitrogen atmosphere. After being cooled to room temperature, the reaction mixture was filtered and the filtrate was concentrated in vacuo to give a residue. The residue was purified by column chromatography (EtOAc in PE : 60% - 80%) to afford methyl (3S)-1-[(2S)-2-(tert- butoxycarbonylamino)-3-[4-[6-fluoro-3-(3-hydroxy-2,2-dimethy l-propyl)-(2M)-2-[2-[(1S)-1- methoxyethyl]-5-[4-(2,2,2-trifluoroethyl)piperazin-1-yl]-3-p yridyl]-1-(2,2,2-trifluoroethyl)indol- 5-yl]thiazol-2-yl]propanoyl]hexahydropyridazine-3-carboxylat e (compound E8, 200.0 mg) as yellow gum. MS calc’d 1001.4 (MH + ), measured 1001.4 (MH + ). Step 8: Preparation of (3S)-1-[(2S)-2-(tert-butoxycarbonylamino)-3-[4-[6-fluoro-3-( 3- hydroxy-2,2-dimethyl-propyl)-(2M)-2-[2-[(1S)-1-methoxyethyl] -5-[4-(2,2,2- trifluoroethyl)piperazin-1-yl]-3-pyridyl]-1-(2,2,2-trifluoro ethyl)indol-5-yl]thiazol-2- yl]propanoyl]hexahydropyridazine-3-carboxylic acid (compound E9). To a mixture of methyl (3S)-1-[(2S)-2-(tert-butoxycarbonylamino)-3-[4-[6-fluoro-3-( 3- hydroxy-2,2-dimethyl-propyl)-(2M)-2-[2-[(1S)-1-methoxyethyl] -5-[4-(2,2,2- trifluoroethyl)piperazin-1-yl]-3-pyridyl]-1-(2,2,2-trifluoro ethyl)indol-5-yl]thiazol-2- yl]propanoyl]hexahydropyridazine-3-carboxylate (compound E8, 200.0 mg, 0.2 mmol) in DCE (5 mL) was added Me3SnOH (200.0 mg, 1.11 mmol). The mixture was stirred at 60 °C for 12 hrs. The reaction mixture was concentrated under vacuum to give a residue. EtOAc (10 mL) and water (10 mL) were added to the residue and the layers were separated. The aqueous phase was extracted with EtOAc (15 mL, twice). The combined organic layer was washed with brine (20 mL), dried over Na 2 SO 4 , filtered, and concentrated under vacuum to afford (3S)-1-[(2S)-2-(tert- butoxycarbonylamino)-3-[4-[6-fluoro-3-(3-hydroxy-2,2-dimethy l-propyl)-(2M)-2-[2-[(1S)-1- methoxyethyl]-5-[4-(2,2,2-trifluoroethyl)piperazin-1-yl]-3-p yridyl]-1-(2,2,2-trifluoroethyl)indol- 5-yl]thiazol-2-yl]propanoyl]hexahydropyridazine-3-carboxylic acid (compound E9, 188.0 mg) as a brown solid. MS calc’d 987.4 (MH + ), measured 987.4 (MH + ). Step 9: Preparation of tert-butyl N-[(7S,13S)-24-fluoro-(20M)-20-[2-[(1S)-1- methoxyethyl]-5-[4-(2,2,2-trifluoroethyl)piperazin-1-yl]-3-p yridyl]-17,17-dimethyl-8,14- dioxo-21-(2,2,2-trifluoroethyl)-15-oxa-4-thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaen-7- yl]carbamate (compound E10). To a mixture of (3S)-1-[(2S)-2-(tert-butoxycarbonylamino)-3-[4-[6-fluoro-3-( 3-hydroxy- 2,2-dimethyl-propyl)-(2M)-2-[2-[(1S)-1-methoxyethyl]-5-[4-(2 ,2,2-trifluoroethyl)piperazin-1- yl]-3-pyridyl]-1-(2,2,2-trifluoroethyl)indol-5-yl]thiazol-2- yl]propanoyl]hexahydropyridazine-3- carboxylic acid (compound E9, 188.0 mg, 0.19 mmol) in DCM (20 mL) were added DIEA (0.7 mL, 3.81 mmol), EDCI (550.0 mg, 2.87 mmol) and HOBt (65.0 mg, 0.48 mmol) at 0 °C. After being stirred at 20 °C for 12 hrs, the reaction mixture was poured into water (20 mL) and extracted with EtOAc (20 mL, three times). The combined organic layer was washed with brine (30 mL), dried over Na2SO4, filtered and concentrated under vacuum to give a residue which was purified by column chromatography (EtOAc in PE : 50% - 70%) to afford tert-butyl N-[(7S,13S)- 24-fluoro-(20M)-20-[2-[(1S)-1-methoxyethyl]-5-[4-(2,2,2-trif luoroethyl)piperazin-1-yl]-3- pyridyl]-17,17-dimethyl-8,14-dioxo-21-(2,2,2-trifluoroethyl) -15-oxa-4-thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaen-7- yl]carbamate (compound E10, 110.0 mg) as a yellow solid. MS calc’d 969.4 (MH+), measured 969.5 (MH + ). Step 10: Preparation of (7S,13S)-7-amino-24-fluoro-(20M)-20-[2-[(1S)-1- methoxyethyl]-5-[4-(2,2,2-trifluoroethyl)piperazin-1-yl]-3-p yridyl]-17,17-dimethyl-21- 2,5 9,13 22,26 (2,2,2-trifluoroethyl)-15-oxa-4-thia-9,21,27,28-tetrazapenta cyclo[17.5.2.1 .1 .0 ]- octacosa-1(25),2,5(28),19,22(26),23-hexaene-8,14-dione (Intermediate E). To a solution of tert-butyl N-[(7S,13S)-24-fluoro-(20M)-20-[2-[(1S)-1-methoxyethyl]-5-[4 - (2,2,2-trifluoroethyl)piperazin-1-yl]-3-pyridyl]-17,17-dimet hyl-8,14-dioxo-21-(2,2,2- 2,5 9,13 22,26 trifluoroethyl)-15-oxa-4-thia-9,21,27,28-tetrazapentacyclo[1 7.5.2.1 .1 .0 ]octacosa- 1(25),2,5(28),19,22(26),23-hexaen-7-yl]carbamate (compound E10, 110.0 mg, 0.11 mmol) in DCM (1 mL) was added TFA (1.0 mL, 12.98 mmol). The mixture was stirred at 20 °C for 1 h. After the reaction was completed, the reaction mixture was concentrated under vacuum to give a residue. Sat. NaHCO3 aq. (20 mL) was added and the mixture was extracted with EtOAc (15 mL, three times). The combined organic layer was washed with brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to afford (7S,13S)-7-amino-24-fluoro-(20M)- 20-[2-[(1S)-1-methoxyethyl]-5-[4-(2,2,2-trifluoroethyl)piper azin-1-yl]-3-pyridyl]-17,17- dimethyl-21-(2,2,2-trifluoroethyl)-15-oxa-4-thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]-octacosa-1(25),2,5(28),19,22(26),23-hexaene-8,14- dione (Intermediate E, 98.0 mg) as a yellow solid. MS calc’d 869.4 (MH + ), measured 869.2 (MH + ). Intermediate F (7S,13S)-7-amino-21-ethyl-24-fluoro-(20M)-20-[2-[(1S)-1-meth oxyethyl]-5-[4-(2,2,2- trifluoroethyl)piperazin-1-yl]-3-pyridyl]-17,17-dimethyl-15- oxa-4-thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaene-8,14- dione The title compound was prepared in analogy to the preparation of Intermediate E by using iodoethane instead of 2,2,2-trifluoroethyl trifluoromethanesulfonate. Intermediate G (7S,13S)-7-amino-21-ethyl-24-fluoro-(20M)-20-[2-[(1S)-1-meth oxyethyl]-5-morpholino-3- pyridyl]-17,17-dimethyl-15-oxa-4-thia-9,21,27,28-tetrazapent acyclo- 2,5 9,13 22,26 [17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaene-8,14-dione The title compound was prepared in analogy to the preparation of Intermediate E by using by using iodoethane and morpholine instead of 2,2,2-trifluoroethyl trifluoromethanesulfonate and 1-(2,2,2-trifluoroethyl)piperazine (compound E1). Intermediate H (7S,13S)-7-amino-25-fluoro-(20M)-20-[2-[(1S)-1-methoxyethyl] -5-(4-methylpiperazin-1-yl)- 3-pyridyl]-17,17-dimethyl-21-(2,2,2-trifluoroethyl)-15-oxa-4 -thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaene-8,14- dione The title compound was prepared in analogy to the preparation of Intermediate C by using 5-bromo-4-fluoro-1H-indole instead of 5-bromo-6-fluoro-1H-indole (compound C2). Intermediate I (7S,13S)-7-amino-24-fluoro-(20M)-20-[2-[(1S)-1-methoxyethyl] -5-morpholino-3-pyridyl]- 17,17-dimethyl-21-(2,2,2-trifluoroethyl)-15-oxa-4-thia-9,21, 27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaene-8,14- dione

The title compound was prepared in analogy to the preparation of Intermediate E by using by using morpholine instead of 1-(2,2,2-trifluoroethyl)piperazine (compound E1). Example 1 (1S,2S)-N-[(7S,13S)-21-ethyl-24-fluoro-(20M)-20-[2-[(1S)-1-m ethoxyethyl]-5-(4- methylpiperazin-1-yl)-3-pyridyl]-17,17-dimethyl-8,14-dioxo-1 5-oxa-4-thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaen-7-yl]-2- methyl-cyclopropanecarboxamide To a solution of (1S,2S)-2-methylcyclopropanecarboxylic acid (compound 1a, 64.3 mg, 0.64 mmol) in DMF (2 mL) were added DIEA (0.3 mL, 1.61 mmol), HATU (366.5 mg, 0.96 mmol) and (7S,13S)-7-amino-21-ethyl-24-fluoro-(20M)-20-[2-[(1S)-1-meth oxyethyl]-5-(4- methylpiperazin-1-yl)-3-pyridyl]-17,17-dimethyl-15-oxa-4-thi a-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaene-8,14-dione (intermediate D, 240.0 mg, 0.32 mmol) at 0 °C. After being stirred at 20 °C for 16 hrs, the reaction mixture was poured into ice water (10 mL) and extracted with EA (20 mL, three times). The combined organic layer was washed with brine (20 mL), dried over Na2SO4 and concentrated under vacuum to give a residue. The resulting residue was purified by silica gel chromatography to afford (1S,2S)-N-[(7S,13S)-21-ethyl-24-fluoro-(20M)-20-[2-[(1S)-1- methoxyethyl]-5-(4-methylpiperazin-1-yl)-3-pyridyl]-17,17-di methyl-8,14-dioxo-15-oxa-4-thia- 2,5 9,13 22,26 9,21,27,28-tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaen- 7-yl]-2-methyl-cyclopropanecarboxamide (Example 1, 156.41 mg) as a white solid. MS calc’d 829.5 (MH + ), measured 829.5 (MH + ). 1 H NMR (400 MHz, METHANOL-d 4 ) δ = 8.64 (d, J = 7.6 Hz, 1H), 8.58 (d, J = 8.4 Hz, 1H), 8.42 (d, J = 2.9 Hz, 1H), 7.63 (d, J = 2.4 Hz, 1H), 7.34 (d, J = 2.8 Hz, 1H), 7.31 (d, J = 12.7 Hz, 1H), 5.79 - 5.73 (m, 1H), 4.44 (br d, J = 11.4 Hz, 1H), 4.28 - 4.13 (m, 4H), 3.77 - 3.68 (m, 2H), 3.41 - 3.37 (m, 4H), 3.28 - 3.13 (m, 2H), 3.02 (br d, J = 13.6 Hz, 1H), 2.77 - 2.74 (m, 4H), 2.63 (br d, J = 14.3 Hz, 1H), 2.44 (s, 3H), 2.21 - 2.13 (m, 1H), 1.97 (s, 2H), 1.68 - 1.58 (m, 1H), 1.51 - 1.47 (m, 1H), 1.42 (d, J = 6.2 Hz, 2H), 1.37 (d, J = 6.6 Hz, 4H), 1.13 (d, J = 6.0 Hz, 3H), 1.08 (br dd, J = 4.2, 8.6 Hz, 1H), 0.98 (t, J = 7.0 Hz, 3H), 0.95 - 0.89 (m, 4H), 0.67 - 0.62 (m, 1H), 0.50 (s, 3H) ppm. Example 2 (1R,5S)-N-[(7S,13S)-21-ethyl-24-fluoro-(20M)-20-[2-[(1S)-1-m ethoxyethyl]-5-(4- methylpiperazin-1-yl)-3-pyridyl]-17,17-dimethyl-8,14-dioxo-1 5-oxa-4-thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaen-7-yl]-3- oxabicyclo[3.1.0]hexane-6-carboxamide The title compound was prepared in analogy to the preparation of Example 1 by using (1S,5R)-3-oxabicyclo[3.1.0]hexane-6-carboxylic acid instead of (1S,2S)-2- methylcyclopropanecarboxylic acid (compound 1a). Example 2 (10.2 mg) was obtained as a white solid. MS calc’d 857.4 (MH + ), measured 857.1 (MH + ). 1 H NMR (400 MHz, ACETONITRILE-d 3 ) δ = 8.61 (d, J = 7.6 Hz, 1H), 8.41 (d, J = 2.8 Hz, 1H), 7.62 (d, J = 2.5 Hz, 1H), 7.33 - 7.28 (m, 1H), 7.18 (d, J = 2.8 Hz, 1H), 7.04 (br d, J = 9.5 Hz, 1H), 5.68 (br t, J = 8.8 Hz, 1H), 4.41 - 4.30 (m, 2H), 4.20 - 4.05 (m, 4H), 3.88 - 3.83 (m, 2H), 3.71 - 3.64 (m, 4H), 3.37 (br d, J = 14.9 Hz, 1H), 3.28 - 3.24 (m, 4H), 3.22 - 3.17 (m, 3H), 3.13 (br dd, J = 9.1, 15.0 Hz, 1H), 2.91 (br d, J = 14.9 Hz, 1H), 2.70 (dt, J = 2.7, 12.9 Hz, 1H), 2.58 (br d, J = 14.1 Hz, 1H), 2.50 (br t, J = 4.8 Hz, 4H), 2.30 - 2.22 (m, 4H), 1.78 - 1.72 (m, 1H), 1.57 - 1.50 (m, 2H), 1.36 (d, J = 6.1 Hz, 3H), 1.27 (br s, 1H), 0.96 (br t, J = 7.1 Hz, 3H), 0.88 (s, 3H), 0.47 (s, 3H) ppm. Example 3 (1S,2S)-2-(difluoromethyl)-N-[(7S,13S)-21-ethyl-24-fluoro-(2 0M)-20-[2-[(1S)-1- methoxyethyl]-5-(4-methylpiperazin-1-yl)-3-pyridyl]-17,17-di methyl-8,14-dioxo-15-oxa-4- 2,5 9,13 22,26 thia-9,21,27,28-tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23- hexaen-7-yl]cyclopropanecarboxamide The title compound was prepared in analogy to the preparation of Example 1 by using (1S,2S)-2-(difluoromethyl)cyclopropanecarboxylic acid instead of (1S,2S)-2- methylcyclopropanecarboxylic acid (compound 1a). Example 3 (31.4 mg) was obtained as a yellow solid. MS calc’d 865.4 (MH + ), measured 865.1 (MH + ). 1 H NMR (400 MHz, CHLOROFORM-d) δ = 8.76 (br d, J = 2.6 Hz, 1H), 8.66 (d, J = 7.6 Hz, 1H), 7.63 (d, J = 2.1 Hz, 1H), 7.30 (br d, J = 2.4 Hz, 1H), 7.12 (d, J = 12.3 Hz, 1H), 6.78 (br d, J = 9.9 Hz, 1H), 6.00 - 5.92 (m, 1H), 4.66 - 4.53 (m, 1H), 4.36 (q, J = 6.2 Hz, 1H), 4.31 - 4.23 (m, 1H), 4.19 - 4.04 (m, 2H), 3.89 - 3.86 (m, 1H), 3.78 - 3.66 (m, 4H), 3.47 - 3.44 (m, 1H), 3.41 (s, 3H), 3.24 - 3.05 (m, 6H), 2.93 (s, 3H), 2.89 - 2.80 (m, 1H), 2.78 - 2.70 (m, 1H), 2.53 - 2.39 (m, 1H), 2.29 - 2.18 (m, 1H), 2.08 - 1.66 (m, 5H), 1.53 - 1.45 (m, 3H), 1.19 - 1.14 (m, 2H), 1.37 - 1.08 (m, 2H), 1.02 - 0.87 (m, 6H), 0.50 (s, 3H) ppm. Example 4 (1S,2S)-N-[(7S,13S)-24-fluoro-(20M)-20-[2-[(1S)-1-methoxyeth yl]-5-(4-methylpiperazin-1- yl)-3-pyridyl]-17,17-dimethyl-8,14-dioxo-21-(2,2,2-trifluoro ethyl)-15-oxa-4-thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaen-7-yl]-2- methyl-cyclopropanecarboxamide The title compound was prepared in analogy to the preparation of Example 1 by using (7S,13S)-7-amino-24-fluoro-(20M)-20-[2-[(1S)-1-methoxyethyl] -5-(4-methylpiperazin-1-yl)-3- pyridyl]-17,17-dimethyl-21-(2,2,2-trifluoroethyl)-15-oxa-4-t hia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaene-8,14-dione (Intermediate C) instead of (7S,13S)-7-amino-21-ethyl-24-fluoro-(20M)-20-[2-[(1S)-1- methoxyethyl]-5-(4-methylpiperazin-1-yl)-3-pyridyl]-17,17-di methyl-15-oxa-4-thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaene-8,14-dione (Intermediate D). Example 4 (15.7 mg) was obtained as a white solid. MS calc’d 883.5 (MH + ), measured 883.1 (MH + ). 1 H NMR (400MHz, Methanol-d4) δ = 8.67 (d, J = 7.6Hz, 1H), 8.50 (d, J = 2.8Hz, 1H), 7.70 (d, J = 2.0Hz, 1H), 7.54 - 7.49 (m, 1H), 7.49 - 7.45 (m, 1H), 5.71 (d, J = 9.2Hz, 1H), 5.20 - 5.14 (m, 1H), 4.47 - 4.41 (m, 1H), 4.24 - 4.18 (m, 2H), 4.09 - 3.94 (m, 2H), 3.80 - 3.61 (, 5H), 3.49 - 3.43 (m, 2H), 3.35 (s, 3H), 3.17 - 3.12 (m, 2H), 3.00 (s, 3H), 2.83 - 2.76 (m, 1H), 2.57 (d, J = 14.4Hz, 1H), 2.23 - 2.16 (m, 1H), 1.99 - 1.93 (m, 1H), 1.84 - 1.77 (m, 1H), 1.68 - 1.61 (m, 1H), 1.52 - 1.48 (m, 1H), 1.47 - 1.41 (m, 4H), 1.27 - 1.22 (m, 1H), 1.13 (d, J = 6.0Hz, 3H), 1.11 - 1.05 (m, 2H), 0.96 (s, 3H), 0.90 - 0.77 (m, 1H), 0.68 - 0.62 (m, 1H), 0.44 (s, 3H). Example 5 and Example 6 (1S,2S)-2-cyano-N-[(7S,13S)-21-ethyl-24-fluoro-(20M)-20-[2-[ (1S)-1-methoxyethyl]-5-(4- methylpiperazin-1-yl)-3-pyridyl]-17,17-dimethyl-8,14-dioxo-1 5-oxa-4-thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaen-7- yl]cyclopropanecarboxamide and (1R,2R)-2-cyano-N-[(7S,13S)-21-ethyl-24-fluoro-(20M)- 20-[2-[(1S)-1-methoxyethyl]-5-(4-methylpiperazin-1-yl)-3-pyr idyl]-17,17-dimethyl-8,14- 2,5 9,13 22,26 dioxo-15-oxa-4-thia-9,21,27,28-tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa- 1(25),2,5(28),19,22(26),23-hexaen-7-yl]cyclopropanecarboxami de The compounds were prepared according to the following scheme:

Step 1: Preparation of (1S,2S)-2-cyano-N-[(7S,13S)-21-ethyl-24-fluoro-(20M)-20-[2- [(1S)-1-methoxyethyl]-5-(4-methylpiperazin-1-yl)-3-pyridyl]- 17,17-dimethyl-8,14-dioxo-15- 2,5 9,13 22,26 oxa-4-thia-9,21,27,28-tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa- 1(25),2,5(28),19,22(26),23-hexaen-7-yl]cyclopropanecarboxami de and (1R,2R)-2-cyano-N- [(7S,13S)-21-ethyl-24-fluoro-(20M)-20-[2-[(1S)-1-methoxyethy l]-5-(4-methylpiperazin-1-yl)- 3-pyridyl]-17,17-dimethyl-8,14-dioxo-15-oxa-4-thia-9,21,27,2 8- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaen-7- yl]cyclopropanecarboxamide To a solution of trans-2-Cyanocyclopropanecarboxylic acid (compound 5a, 11.2 mg, 0.1 mmol) in DMF (0.5 mL) were added DIEA (0.1 mL, 0.33 mmol), HATU (76.4 mg, 0.2 mmol) and (7S,13S)-7-amino-21-ethyl-24-fluoro-(20M)-20-[2-[(1S)-1-meth oxyethyl]-5-(4- methylpiperazin-1-yl)-3-pyridyl]-17,17-dimethyl-15-oxa-4-thi a-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaene-8,14-dione (intermediate D, 50.0 mg, 0.07 mmol) at 0 °C. After being stirred at 20 °C for 16 hrs, the reaction mixture was prepared by prep-HPLC and SFC to afford Example 5 (13.4 mg, faster eluted) as a white solid and Example 6 (9.9 mg, slower eluted) as a white solid. Example 5: MS calc’d 840.4 (MH + ), measured 840.1 (MH + ). 1 H NMR (400 MHz, CHLOROFORM-d) δ = 8.63 (d, J = 7.3 Hz, 1H), 8.49 (d, J = 2.8 Hz, 1H), 7.59 (d, J = 2.0 Hz, 1H), 7.12 - 7.07 (m, 2H), 6.88 (br d, J = 8.4 Hz, 1H), 5.92 (t, J = 8.9 Hz, 1H), 4.60 (br dd, J = 2.4, 12.5 Hz, 1H), 4.30 - 4.19 (m, 3H), 4.08 - 3.99 (m, 2H), 3.85 (br d, J = 11.1 Hz, 1H), 3.74 - 3.70 (m, 1H), 3.49 (br d, J = 3.4 Hz, 4H), 3.36 (s, 3H), 3.20 - 3.13 (m, 2H), 2.76 - 2.68 (m, 4H), 2.63 (s, 1H), 2.46 (br d, J = 14.2 Hz, 1H), 2.25 - 2.20 (m, 2H), 2.01 (s, 1H), 1.95 - 1.90 (m, 2H), 1.58 (td, J = 5.0, 9.8 Hz, 2H), 1.47 - 1.42 (m, 5H), 1.30 - 1.22 (m, 4H), 0.97 (t, J = 7.1 Hz, 3H), 0.91 (s, 2H), 0.47 (s, 3H) ppm. Example 6: MS calc’d 840.4 (MH + ), measured 840.1 (MH + ). 1 H NMR (400 MHz, CHLOROFORM-d) δ = 8.64 (d, J = 7.6 Hz, 1H), 8.51 (d, J = 2.9 Hz, 1H), 7.63 (d, J = 2.2 Hz, 1H), 7.13 - 7.07 (m, 2H), 6.88 (d, J = 9.5 Hz, 1H), 5.95 (t, J = 8.9 Hz, 1H), 4.65 - 4.57 (m, 1H), 4.32 - 4.21 (m, 3H), 4.12 - 3.96 (m, 2H), 3.86 (d, J = 10.8 Hz, 1H), 3.75 (d, J = 11.1 Hz, 1H), 3.48 (br d, J = 14.7 Hz, 1H), 3.37 (s, 3H), 3.34 - 3.29 (m, 4H), 3.19 (dd, J = 8.8, 15.0 Hz, 1H), 3.05 (br d, J = 14.2 Hz, 1H), 2.76 - 2.62 (m, 5H), 2.52 (br d, J = 14.3 Hz, 1H), 2.41 (s, 3H), 2.27 - 2.19 (m, 2H), 2.04 - 2.00 (m, 2H), 1.48 - 1.43 (m, 4H), 1.32 - 1.23 (m, 2H), 1.00 (t, J = 7.1 Hz, 3H), 0.92 (s, 4H), 0.49 (s, 3H) ppm. Example 7 (1S,2S)-N-[(7S,13S)-21-ethyl-24-fluoro-(20M)-20-[2-[(1S)-1-m ethoxyethyl]-5-[4-(2,2,2- trifluoroethyl)piperazin-1-yl]-3-pyridyl]-17,17-dimethyl-8,1 4-dioxo-15-oxa-4-thia- 2,5 9,13 22,26 9,21,27,28-tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23- hexaen-7-yl]-2-methyl-cyclopropanecarboxamide

The title compound was prepared in analogy to the preparation of Example 1 by using (7S,13S)-7-amino-21-ethyl-24-fluoro-(20M)-20-[2-[(1S)-1-meth oxyethyl]-5-[4-(2,2,2- trifluoroethyl)piperazin-1-yl]-3-pyridyl]-17,17-dimethyl-15- oxa-4-thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaene-8,14-dione (Intermediate F) instead of (7S,13S)-7-amino-21-ethyl-24-fluoro-(20M)-20-[2-[(1S)-1- methoxyethyl]-5-(4-methylpiperazin-1-yl)-3-pyridyl]-17,17-di methyl-15-oxa-4-thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaene-8,14-dione (Intermediate D). Example 7 (12.5 mg) was obtained as a yellow solid. MS calc’d 897.4 (MH + ), measured 897.6 (MH + ). 1 H NMR (400 MHz, Methanol-d 4 ) δ = 8.68 (d, J = 7.6 Hz, 1H), 8.38 (d, J = 2.8 Hz, 1H), 7.78 (d, J = 2.4 Hz, 1H), 7.65 (d, J = 2.4 Hz, 1H), 7.35 (d, J = 12.8 Hz, 1H), 5.81 (d, J = 8.8 Hz, 1H), 4.48 - 4.40 (m, 1H), 4.38 - 4.31 (m, 1H), 4.30 - 4.19 (m, 1H), 4.16 - 4.01 (m, 2H), 3.81 - 3.68 (m, 2H), 3.49 - 3.44 (m, 4H), 3.43 - 3.40 (m, 1H), 3.37 (s, 3H), 3.21 - 3.12 (m, 3H), 3.04 - 3.95 (m, 1H), 2.92 - 2.86 (m, 4H), 2.81 - 2.70 (m, 2H), 2.21 - 2.11 (m, 1H), 1.99 - 1.89 (m, 1H), 1.84 - 1.70 (m, 1H), 1.67 - 1.57 (m, 1H), 1.51 - 1.46 (m, 1H), 1.44 (d, J = 6.4 Hz, 3H), 1.25 - 1.19 (m, 1H), 1.15 - 1.10 (m, 3H), 1.09 - 1.05 (m, 1H), 1.04 - 1.00 (m, 3H), 0.94 (s, 3H), 0.66 - 0.61 (m, 1H), 0.59 (s, 3H) ppm. Example 8 (1S,2S)-N-[(7S,13S)-21-ethyl-24-fluoro-(20M)-20-[2-[(1S)-1-m ethoxyethyl]-5-morpholino-3- pyridyl]-17,17-dimethyl-8,14-dioxo-15-oxa-4-thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaen-7-yl]-2- methyl-cyclopropanecarboxamide

The title compound was prepared in analogy to the preparation of Example 1 by using (7S,13S)-7-amino-21-ethyl-24-fluoro-(20M)-20-[2-[(1S)-1-meth oxyethyl]-5-morpholino-3- pyridyl]-17,17-dimethyl-15-oxa-4-thia-9,21,27,28-tetrazapent acyclo- 2,5 9,13 22,26 [17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaene-8,14-dione (Intermediate G) instead of (7S,13S)-7-amino-21-ethyl-24-fluoro-(20M)-20-[2-[(1S)-1-meth oxyethyl]-5-(4- methylpiperazin-1-yl)-3-pyridyl]-17,17-dimethyl-15-oxa-4-thi a-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaene-8,14-dione (Intermediate D). Example 8 (135.4 mg) was obtained as a light yellow solid. MS calc’d 816.4 (MH + ), measured 816.1 (MH + ). 1 H NMR (400 MHz, Methanol-d 4 ) δ = 8.70 (d, J = 7.5 Hz, 1H), 8.37 (d, J = 2.7 Hz, 1H), 7.95 (br s, 1H), 7.65 (d, J = 2.3 Hz, 1H), 7.37 (d, J = 12.6 Hz, 1H), 5.82 (br d, J = 8.9 Hz, 1H), 4.47 - 4.38 (m, 2H), 4.31 - 4.23 (m, 1H), 4.15 - 4.10 (m, 1H), 4.05 - 3.98 (m, 1H), 3.88 (br t, J = 4.7 Hz, 4H), 3.79 (br d, J = 11.0 Hz, 1H), 3.71 (d, J = 10.6 Hz, 1H), 3.47 - 3.38 (m, 10H), 3.00 (br d, J = 15.0 Hz, 1H), 2.84 - 2.72 (m, 2H), 2.19 - 2.12 (m, 1H), 1.97 - 1.90 (m, 1H), 1.64 (br d, J = 2.7 Hz, 1H), 1.50 - 1.45 (m, 4H), 1.23 (br dd, J = 4.2, 9.5 Hz, 2H), 1.12 (d, J = 5.9 Hz, 3H), 1.05 (q, J = 6.8 Hz, 4H), 0.96 (s, 3H), 0.62 (s, 3H) ppm. Example 9 (1S,2S)-N-[(7S,13S)-24-fluoro-(20M)-20-[2-[(1S)-1-methoxyeth yl]-5-[4-(2,2,2- trifluoroethyl)piperazin-1-yl]-3-pyridyl]-17,17-dimethyl-8,1 4-dioxo-21-(2,2,2- 2,5 9,13 22,26 trifluoroethyl)-15-oxa-4-thia-9,21,27,28-tetrazapentacyclo[1 7.5.2.1 .1 .0 ]octacosa- 1(25),2,5(28),19,22(26),23-hexaen-7-yl]-2-methyl-cyclopropan ecarboxamide

The title compound was prepared in analogy to the preparation of Example 1 by using (7S,13S)-7-amino-24-fluoro-(20M)-20-[2-[(1S)-1-methoxyethyl] -5-[4-(2,2,2- trifluoroethyl)piperazin-1-yl]-3-pyridyl]-17,17-dimethyl-21- (2,2,2-trifluoroethyl)-15-oxa-4-thia- 2,5 9,13 22,26 9,21,27,28-tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaene- 8,14-dione (Intermediate E) instead of (7S,13S)-7-amino-21-ethyl-24-fluoro-(20M)-20-[2-[(1S)- 1-methoxyethyl]-5-(4-methylpiperazin-1-yl)-3-pyridyl]-17,17- dimethyl-15-oxa-4-thia- 2,5 9,13 22,26 9,21,27,28-tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaene- 8,14-dione (Intermediate D). Example 9 mg) was obtained as an off-white solid. MS calc’d 951.4 (MH + ), measured 951.2 (MH + ). 1 H NMR (400 MHz, Methanol-d4) δ = 8.69 (d, J = 7.6 Hz, 1H), 8.42 (d, J = 2.8 Hz, 1H), 7.69 (d, J = 2.4 Hz, 1H), 7.56 - 7.43 (m, 2H), 5.73 (d, J = 8.8 Hz, 1H), 5.24 - 5.08 (m, 1H), 4.83 - 4.78 (m, 1H), 4.49 - 4.39 (m, 1H), 4.26 - 4.15 (m, 2H), 3.82 - 3.65 (m, 2H), 3.48 - 3.38 (m, 5H), 3.36 (s, 3H), 3.30 - 3.25 (m, 1H), 3.20 - 3.09 (m, 3H), 2.88 (t, J = 4.8 Hz, 4H), 2.84 - 2.75 (m, 1H), 2.67 - 2.59 (m, 1H), 2.26 - 2.15 (m, 1H), 1.99 - 1.91 (m, 1H), 1.87 - 1.74 (m, 1H), 1.70 - 1.56 (m, 1H), 1.52 - 1.47 (m, 1H), 1.45 (d, J = 6.0 Hz, 3H), 1.28 - 1.19 (m, 1H), 1.15 - 1.10 (m, 3H), 1.10 - 1.05 (m, 1H), 0.97 (s, 3H), 0.71 - 0.59 (m, 1H), 0.49 (s, 3H) ppm. Example 10 (1S,2S)-N-[(7S,13S)-25-fluoro-(20M)-20-[2-[(1S)-1-methoxyeth yl]-5-(4-methylpiperazin-1- yl)-3-pyridyl]-17,17-dimethyl-8,14-dioxo-21-(2,2,2-trifluoro ethyl)-15-oxa-4-thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaen-7-yl]-2- methyl-cyclopropanecarboxamide

The title compound was prepared in analogy to the preparation of Example 1 by using (7S,13S)-7-amino-25-fluoro-(20M)-20-[2-[(1S)-1-methoxyethyl] -5-(4-methylpiperazin-1-yl)-3- pyridyl]-17,17-dimethyl-21-(2,2,2-trifluoroethyl)-15-oxa-4-t hia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaene-8,14-dione (intermediate H) instead of (7S,13S)-7-amino-21-ethyl-24-fluoro-(20M)-20-[2-[(1S)-1- methoxyethyl]-5-(4-methylpiperazin-1-yl)-3-pyridyl]-17,17-di methyl-15-oxa-4-thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaene-8,14-dione (Intermediate D). Example 10 (11.2 mg) was obtained as a white solid. MS calc’d 883.4 (MH + ), measured 883.5 (MH + ). 1 H NMR (400 MHz, Methanol-d4) δ = 8.50 (d, J = 2.8 Hz, 1H), 7.52 - 7.37 (m, 4H), 6.03 – 5.88 (m, 1H), 5.19 - 5.07 (m, 1H), 4.80 - 4.76 (m, 2H), 4.44 - 4.34 (m, 2H), 4.12 - 3.96 (m, 3H), 3.72 - 3.47 (m, 6H), 3.41 - 3.33 (m, 2H), 3.28 - 3.25 (m, 2H), 3.19 - 3.05 (m, 5H), 2.99 (s, 4H), 2.59 - 2.52 (m, 1H), 1.66 - 1.53 (m, 1H), 1.44 (d, J = 6.0 Hz, 3H), 1.39 - 1.29 (m, 2H), 1.26 - 1.17 (m, 2H), 1.10 - 1.05 (m, 3H), 1.05 - 1.01 (m, 1H), 0.80 (s, 3H), 0.65 - 0.52 (m, 3H) ppm. Example 11 (1S,2S)-2-(difluoromethyl)-N-[(7S,13S)-21-ethyl-24-fluoro-(2 0M)-20-[2-[(1S)-1- methoxyethyl]-5-morpholino-3-pyridyl]-17,17-dimethyl-8,14-di oxo-15-oxa-4-thia- 2,5 9,13 22,26 9,21,27,28-tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23- hexaen-7-yl]cyclopropanecarboxamide

The title compound was prepared in analogy to the preparation of Example 1 by using (7S,13S)-7-amino-21-ethyl-24-fluoro-(20M)-20-[2-[(1S)-1-meth oxyethyl]-5-morpholino-3- pyridyl]-17,17-dimethyl-15-oxa-4-thia-9,21,27,28-tetrazapent acyclo- 2,5 9,13 22,26 [17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaene-8,14-dione (Intermediate G) and (1S,2S)-2-(difluoromethyl)cyclopropanecarboxylic acid instead of (7S,13S)-7-amino-21- ethyl-24-fluoro-(20M)-20-[2-[(1S)-1-methoxyethyl]-5-(4-methy lpiperazin-1-yl)-3-pyridyl]- 2,5 9,13 22,26 17,17-dimethyl-15-oxa-4-thia-9,21,27,28-tetrazapentacyclo[17 .5.2.1 .1 .0 ]octacosa- 1(25),2,5(28),19,22(26),23-hexaene-8,14-dione (Intermediate D) and (1S,2S)-2- methylcyclopropanecarboxylic acid (compound 1a). Example 11 (33.2 mg) was obtained as a yellow solid. MS calc’d 852.4 (MH + ), measured 852.2 (MH + ). 1 H NMR (400 MHz, CHLOROFORM-d) δ = 8.91 (br s, 1H), 8.66 (d, J = 7.6 Hz, 1H), 7.62 (d, J = 2.2 Hz, 1H), 7.41 (s, 1H), 7.13 (s, 1H), 7.10 (s, 1H), 6.75 (br d, J = 8.9 Hz, 1H), 5.99 - 5.93 (m, 1H), 5.83 (d, J = 3.1 Hz, 1H), 4.60 (br d, J = 11.9 Hz, 1H), 4.38 (br d, J = 6.4 Hz, 1H), 4.21 (br d, J = 9.2 Hz, 1H), 4.13 - 4.06 (m, 2H), 3.87 (br d, J = 11.4 Hz, 2H), 3.79 - 3.65 (m, 2H), 3.45 (br d, J = 15.2 Hz, 1H), 3.41 (s, 2H), 3.39 - 3.35 (m, 4H), 3.17 (br dd, J = 8.6, 15.0 Hz, 2H), 3.10 (br d, J = 11.6 Hz, 1H), 2.80 - 2.63 (m, 2H), 2.50 (br d, J = 13.9 Hz, 1H), 2.20 (br d, J = 10.9 Hz, 1H), 1.99 (br d, J = 17.9 Hz, 1H), 1.87 - 1.78 (m, 2H), 1.67 - 1.59 (m, 1H), 1.53 (d, J = 6.4 Hz, 3H), 1.33 - 1.21 (m, 3H), 1.18 - 1.12 (m, 1H), 1.01 (br t, J = 7.0 Hz, 3H), 0.94 (s, 3H), 0.53 (s, 3H) ppm. Example 12 (1S,2S)-N-[(7S,13S)-24-fluoro-(20M)-20-[2-[(1S)-1-methoxyeth yl]-5-morpholino-3-pyridyl]- 17,17-dimethyl-8,14-dioxo-21-(2,2,2-trifluoroethyl)-15-oxa-4 -thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaen-7-yl]-2- methyl-cyclopropanecarboxamide The title compound was prepared in analogy to the preparation of Example 1 by using (7S,13S)-7-amino-24-fluoro-(20M)-20-[2-[(1S)-1-methoxyethyl] -5-morpholino-3-pyridyl]- 17,17-dimethyl-21-(2,2,2-trifluoroethyl)-15-oxa-4-thia-9,21, 27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaene-8,14-dione (intermediate I) instead of (7S,13S)-7-amino-21-ethyl-24-fluoro-(20M)-20-[2-[(1S)-1- methoxyethyl]-5-(4-methylpiperazin-1-yl)-3-pyridyl]-17,17-di methyl-15-oxa-4-thia-9,21,27,28- 2,5 9,13 22,26 tetrazapentacyclo[17.5.2.1 .1 .0 ]octacosa-1(25),2,5(28),19,22(26),23-hexaene-8,14-dione (Intermediate D). Example 12 (150 mg) was obtained as a white solid. MS calc’d 870.4 (MH + ), measured 870.5 (MH + ). 1 H NMR (400 MHz, Methanol-d4) δ = 8.65-8.63 (d, J = 7.6 Hz, 1H), 8.41 - 8.40 (d, J = 2.8 Hz,1H), 7.70 - 7.65 (d, J = 2.4 Hz, 1H), 7.50-7.40 (d, J = 12.4 Hz, 1H), 7.31 - 7.28 (d, J = 2.4 Hz, 1H), 5.76 - 5.66 (d, J = 8.0Hz,1H), 5.18 - 5.05 (m, 1H), 4.96 - 4.88 (m, 1H), 4.47 - 4.39 (m, 1H), 4.23 - 4.10 (m, 2H), 3.90-3.83 (t, J = 4.8 Hz, J = 9.6 Hz, 4H), 3.80- 3.75 (d, J = 10.8Hz, 1H), 3.71 - 3.63 (m, 1H), 3.49 - 3.42 (m, 1H), 3.30 - 3.29 (m, 3H), 3.28 - 3.21 (m, 4H), 3.14 - 3.07 (d, J = 14.4 Hz, 1H), 2.86 - 2.77 (m, 1H), 2.63-2.55 (d, J = 14.4 Hz, 1H), 2.23 - 2.14 (m, 1H), 1.98 - 1.89 (m, 1H), 1.84 - 1.72 (m, 1H), 1.65 - 1.56 (m, 1H), 1.52 - 1.46 (m, 1H),1.45- 1.40 (d, J = 6.4 Hz, 3H), 1.30 - 1.19 (m, 2H), 1.13 - 1.06 (m, 4H), 0.95 (s, 3H), 0.69 - 0.58 (m, 1H), 0.48 - 0.39 (s, 3H) ppm. BIOLOGICAL EXAMPLE Compound A122 (page 70 of Table.1a) from WO2022060836 was cited as reference compound for this invention. Example 13 Cell viability assay The purpose of this cellular assay was to determine the effects of test compounds on the proliferation of human cancer cell lines NCI-H358 (ATCC-CRL5807) cells, AGS (ATCC-CRL- 1739) cells, SW620 (ATCC-CCL-227) over a 3-day treatment period by quantifying the amount of NADPH present at endpoint using Cell Counting Kit-8. Cells were seeded at 5,000 cells/well (NCI-H358), 2,000 cells/well (AGS) 2,000 cells/well (SW620) in 96-well assay plates (Corning-3699) and incubated overnight. On the day of the assay, diluted compounds were then added in a final concentration of 0.5% DMSO. After 72 hrs incubation, a tenth of the volume of cell counting kit 8(Dnjindo-CK04) was added into each well. Read the signal (OD450 minus OD650) using EnVision after 2 hrs incubation. IC 50 was determined by fitting a 4-parameter sigmoidal concentration response model. Table 1. Activity of Examples and Compounds of present invention in KRAS Cell viability assay

Example 14 KRAS-BRAF with CYPA (500 nM) interaction assay In this example, TR-FRET was also used to measure the compound or compound-CYPA dependent disruption of the KRAS G12C-BRAF complex. This protocol was also used to measure disruption of KRAS G12D or KRAS G12V binding to BRAF by a compound of the invention, respectively. In assay buffer containing 25mM HEPES PH=7.4 (4-(2-hydroxyethyl)- 1-piperazineethanesulfonic acid, Thermo, 15630080), 0.002% Tween20, 0.1% BSA, 100mM NaCl, 5mM MgCl2, 10 µM GMPPNP (Guanosine 5′-[β,γ-imido]triphosphate trisodium salt hydrate, Sigma, G0635), tagless CYPA, GMPPNP loaded 6His-KRAS proteins, and GST- BRAF RBD were mixed in a well of a 384-well assay plate at final concentrations of 50 nM, 6.25 nM and 1nM, respectively. Compound was present in plate wells as a 16-point 3-fold dilution series starting at a final concentration of 10 µM and incubated for 3 hours. A mixture of MAb Anti-6His-XL665 (Cisbio, 61HISXLB) and Mab anti-GST-TB cryptate (Cisbio, 61GSTTLB)was then added at a final concentration of 6.67 nM and 0.21 nM, respectively, and the plate was incubated for an additional 1.5 hours. TR-FRET signal was read on a PHERstar FSX microplate reader (Ex320 nm, Em 665/615 nm). Compounds that facilitate disruption of the KRAS-BRAF complex were identified as those eliciting a decrease in the TR-FRET ratio relative to DMSO control wells. Table 2. Activity of Examples and Compounds of present invention in KRAS-BRAF with CYPA (500 nM) interaction assay Example 15 pERK inhibition assay This assay is to measure the ability of test compounds in inhibiting the phosphorylation of ERK, the downstream signaling of KRAS G12C in NCI-H358 cells, KRAS G12D in AGS cells, and KRAS G12V in SW620. NCI-H358 (ATCC-CRL5807) cells, AGS (ATCC-CRL-1739) cells, SW620 (ATCC-CCL-227) cells were all grown and maintained using RPMI-1640 medium (Thermo Fisher Scientific) with 10% fetal bovine serum and 1% penicillin/streptomycin. On the day prior to compound addition, cells were plated in tissue culture-treated 96 well plates (Corning-3699) at a density of 30,000 cell/well, 20,000 cell/well, 30,000 cell/well for NCI-H358, AGS and SW620 respectively, and allowed for attachment overnight. Diluted compounds were then added in a final concentration of 0.5% DMSO. After 4 hours of incubation, the medium was removed, 100 µL of 4% formaldehyde was added, and the assay plates were incubated at room temperature for 20 minutes. The plates were then washed once with phosphate buffered saline (PBS), and permeabilized with 100 µL of chilled methanol for 10 minutes. Non-specific antibody binding to the plates was blocked using 50 µL 1X BSA blocking buffer (Thermo-37520, 10-fold dilution by Phosphate-Buffered Saline Tween (PBST) for at least 1 hour at room temperature. The amount of phosphor-ERK was determined using an antibody specific for phosphorylated form of ERK. Primary antibody (pERK, CST-4370, Cell Signaling Technology) was diluted 1:300 in blocking buffer, with 50 µL aliquoted to each well, and incubated overnight at 4 ℃. Cells was washed five times for 5 minutes with PBST. Secondary antibody (HRP-linked anti-rabbit IgG, CST-7074, Cell Signaling Technology) was diluted 1:1000 in blocking buffer, and 50 µL was added to each well and incubated 1-2 hrs at room temperature. Cells was washed 5 times for 5 minutes with PBST, 100µL TMB ELISA substrate (abcam-ab171523) were added and gently shake for 20 minutes.50µL stop solution (abcam-ab171529) were added, and then read the signal (OD450) by EnVision. IC50 was determined by fitting a 4-parameter sigmoidal concentration response model. Table 3. Activity of Examples and Compounds of present invention in KRAS pERK inhibition assay