WO2025214350A1 - Macrocyclic compounds as ras inhibitors for the treatment of cancer - Google Patents
Macrocyclic compounds as ras inhibitors for the treatment of cancerInfo
- Publication number
- WO2025214350A1 WO2025214350A1 PCT/CN2025/087799 CN2025087799W WO2025214350A1 WO 2025214350 A1 WO2025214350 A1 WO 2025214350A1 CN 2025087799 W CN2025087799 W CN 2025087799W WO 2025214350 A1 WO2025214350 A1 WO 2025214350A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- optionally substituted
- membered
- alkyl
- solvate
- heterocycloalkyl
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/495—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
- A61K31/50—Pyridazines; Hydrogenated pyridazines
- A61K31/504—Pyridazines; Hydrogenated pyridazines forming part of bridged ring systems
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/535—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
- A61K31/5375—1,4-Oxazines, e.g. morpholine
- A61K31/5377—1,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/54—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
- A61K31/541—Non-condensed thiazines containing further heterocyclic rings
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D498/00—Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
- C07D498/22—Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains four or more hetero rings
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D513/00—Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
- C07D513/22—Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains four or more hetero rings
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D519/00—Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00
Definitions
- the present invention belongs to the field of medicine, and specifically, relates to macrocyclic com-pounds with stereoisomers, pharmaceutically acceptable salts, solvates, eutectic or deuterated compounds, and their used in the treatment of RAS protein-mediated related diseases
- the RAS protein (including KRAS4A, KRAS4B, HRAS, and NRAS) is a small, membrane-bound guanine nucleotide-binding protein that acts as a molecular switch by cycling between an active GTP-bound state and an inactive GDP-bound state.
- RAS binds to GEF family proteins such as SOS1, its conformation changes, reducing its affinity for GDP and causing its dissociation from GDP and binding to GTP in the high concentration in the environment, thus becoming active.
- RAS receptor tyrosine kinases
- EGFR epidermal growth factor receptor
- RAS mutations occur in approximately 19%of all cancers and play an important role in tumor occur-rence and progression.
- KRAS is the most commonly mutated subtype, followed by NRAS and HRAS.
- KRAS mutations commonly occur in pancreatic ductal adenocarcinoma, lung adenocarcinoma, and colorectal adenocarcinoma, while NRAS mutations have a relatively higher frequency in hematologic malignancies such as chronic myeloid leukemia and acute myeloid leukemia, as well as in malignant mel-anoma, thyroid cancer, and laryngeal cancer.
- HRAS mutations are less common in human cancers, but have a relatively higher mutation rate in head and neck squamous cell carcinoma, bladder cancer, salivary gland carcinoma, and oral cancer.
- the RAS molecular glue can simultaneously bind to highly activated RAS (RAS-ON) proteins and a ubiquitously expressed protein in cells, forming a tricomplex to occupy binding sites for RAS interacting proteins such as SOS1 and RAF1, keeping RAS protein in a locked state alone, unable to activate down-stream signaling pathways, thereby blocking RAS signaling pathway transmission and inhibiting tumor growth. Therefore, there is an urgent clinical need to develop an inhibitor of RAS molecular glue type with good activity and high druggability.
- RAS-ON highly activated RAS
- Ras inhibitors as molecular glues.
- the approach described herein entails for-mation of a high affinity three-component complex between a synthetic ligand and two intracellular pro-teins which do not interact under normal physiological conditions:
- the target of interest e.g. Ras
- a widely expressed cytosolic chaperone in cells e.g. cyclophilin A
- the Ras molecular glues described herein induce a new binding pocket in Ras by forming a three-component complex with high-affinity between the Ras protein and the cytosolic chaperone, cyclo-philin A.
- the inventors believe that one way the inhibitory effect on Ras is effected by compounds of the invention and the three-component complexes they form is by steric occlu-sion of the interaction site between Ras and downstream effector molecules, such as RAF and PI3K, which are required for propagating the oncogenic signal.
- the present invention provides a compound, or a stereoisomer thereof, a pharma-ceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt thereof, wherein the compound is a compound of formula (I) ;
- n 0, 1, 2, or 3;
- A is optionally substituted 5 to 6-membered heterocycloalkylene, optionally substituted 5 to 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene;
- B is selected from optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 3 to 6-membered heterocycloalkenylene, optionally substituted 4 to 11-membered bicyclic cycloalkylene, or optionally substituted 4 to 11-membered bicyclic heterocycloalkylene;
- G is optionally substituted C 1-6 alkylene
- R 8 and R 9 are each independently selected from hydrogen, deuterium, halogen, hydroxy, cyano, op-tionally substituted C 1-3 alkyl, or R 8 and R 9 combine with the atoms to which they are attached to form an optionally substituted C 3-6 cycloalkyl or a carbonyl;
- R 10 and R 11 are each independently selected from hydrogen, deuterium, halogen, hydroxy, cyano, op-tionally substituted C 1-3 alkyl, or R 10 and R 11 combine with the atoms to which they are attached to form an optionally substituted C 3-6 cycloalkyl or a carbonyl;
- R 12 and R 13 are each independently selected from hydrogen, deuterium, halogen, hydroxy, optionally substituted C 1-3 alkyl, -O (C 1-6 alkyl) , -S (C 1-6 alkyl) or -N (C 1-6 alkyl) (C 1-6 alkyl) ; furthermore, the C 1-6 alkyl may be further optionally substituted;
- R A is optionally substituted C 3-6 cycloalkyl, optionally substituted C 3-6 heterocycloalkyl (or 4 to 7-membered heterocycloalkyl) , optionally substituted 5 to 8-membered aryl, optionally substituted 5 to 8-membered heteroaryl, optionally substituted 8 to 10-membered fused bicyclic aryl or optionally substi-tuted 8 to 10-membered fused bicyclic heteroaryl;
- R B is selected from hydrogen, C 1-6 alkyl, C 1-6 alkoxyl, C 1-6 haloalkyl, C 1-6 haloalkoxyl, optionally sub-stituted C 3-6 cycloalkyl, or optionally substituted C 3-6 heterocycloalkyl (or 4 to 7-membered heterocycloal-kyl) ; or provided that when n exceeds 1, any two of R B combine with the atoms to which they are attached to form a 3 to 6-membered ring, wherein the ring can be optionally substituted with halogen, hydroxy, and C 1-3 alkyl;
- X 1 is N or C
- X 2 is N or -CR a -;
- X 3 is N or -CR b -;
- X 4 is N or -CR c -;
- X 5 is N or C
- X 6 is S, O, N, or -CH-;
- R a , R b , and R c are each independently hydrogen, halogen, cyano, C 1-3 alkyl, C 1-3 alkoxyl, C 1-3 haloal-kyl, C 1-3 haloalkoxyl, C 3-6 cycloalkyl, or C 3-6 heterocycloalkyl (or 4 to 7-membered heterocycloalkyl) ;
- R 3 is absent, or selected from hydrogen, C 1-6 alkyl, or C 1-6 haloalkyl, provided that when X 6 is O or S, R 3 is absent;
- E is a bond, or selected from N, or -CR d -, wherein R d is selected from hydrogen, halogen, hydroxy, cyano, carboxyl, optionally substituted C 1-6 alkyl, optionally substituted C 1-6 alkoxyl, C 1-6 aminoalkyl, C 1-6 hydroxyalkyl, or -NR e R f ; wherein R e and R f are independently selected from hydrogen, or optionally sub-stituted C 1-6 alkyl;
- R 4 is hydrogen, optionally substituted C 1-6 alkyl, optionally substituted C 1-6 heteroalkyl, optionally substituted C 3-10 cycloalkyl, optionally substituted C 3-10 cycloalkenyl, optionally substituted C 3-10 heterocy-cloalkyl (or 4 to 12-membered heterocycloalkyl) , optionally substituted 6 to 10-membered aryl, optionally substituted 5 to 10-membered heteroaryl, or
- R 5 is hydrogen, C 1-6 alkyl or C 3-6 cycloalkyl, furthermore, the C 1-6 alkyl or C 3-6 cycloalkyl may be fur-ther optionally substituted;
- R 4 and R 5 combine with the atoms to which they are attached to form a ring, wherein the ring is se-lected from optionally substituted 3 to 10-membered cycloalkyl, optionally substituted 3 to 10-membered heterocycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 4 to 11-membered bicyclic cycloalkyl, optionally substituted 4 to 11-membered bicyclic heterocycloalkyl;
- L is absent, or selected from -CH 2 -, -C (O) -, -CHR g -or -C (R g ) 2 -, wherein R g is optionally substituted C 1-6 alkyl;
- R 6 is hydrogen, or optionally substituted C 1-6 alkyl
- R 7 is optionally substituted C 1-6 alkyl, optionally substituted C 1-6 heteroalkyl, optionally substituted C 3-10 cycloalkyl, optionally substituted C 3-10 heterocycloalkyl (or 3 to 10-membered heterocycloalkyl) , op-tionally substituted 6 to 10-membered aryl, optionally substituted 5 to 10-membered heteroaryl;
- R 6 and R 7 combine with the atoms to which they are attached to form a ring, wherein the ring is selected from optionally substituted 3 to 10-membered cycloalkyl, optionally substituted 3 to 10-membered heterocycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 4 to 11-membered bicyclic cycloalkyl, optionally substituted 4 to 11-membered bicyclic heterocycloalkyl.
- the present invention provides a pharmaceutical composition
- a pharmaceutical composition comprising the compound according to the first aspect the stereoisomer thereof, the pharmaceutically acceptable salt thereof, the metabolite thereof, the prodrug thereof, the solvate thereof or the solvate of the pharmaceuti-cally acceptable salt thereof, and a pharmaceutically acceptable carrier.
- the present invention provides a use of the compound according to the first aspect the stereoisomer thereof, the pharmaceutically acceptable salt thereof, the metabolite thereof, the prodrug thereof, the solvate thereof or the solvate of the pharmaceutically acceptable salt thereof in the preparation of a medicament used for the treatment of cancer.
- Fig. 1 The reference compound and the representative Compound A significantly inhibited the interac-tion of RAF1 with active RAS Mutant protein or RAS WT protein.
- Fig. 2 The reference compound and the representative Compound A significantly inhibited the cell viability of KRAS mutant cells.
- TGI tumor growth inhibition ratio
- TGI tumor growth inhibition ratio
- TGI tumor growth inhibition ratio
- TGI tumor growth inhibition ratio
- Fig. 7 and Fig. 8 Bioluminescence signal changes tumor growth inhibition ratio (TGI) after adminis-trating vehicle or test articles to female BALB/c nude mice bearing NCI-H1373-luc intracranial tumors. Data endpoints represent mean ⁇ SEM.
- the present invention provides a compound of formula (I) :
- R 8 , R 9 , R 10 , R 11 , R 12 and R 13 is H.
- the compound of formula (I) is a compound of formula (I-1)
- X 1 , X 2 , X 3 , X 4 , X 5 , X 6 , A, B, E, G, R A , R B , R 3 , R 4 , R 5 and n are as defined in formula I.
- B is optionally substituted
- B is optionally substituted
- R A is In some embodiments, R 1 is selected from the group con-sisting of hydrogen, C 1-6 alkyl, C 1-6 alkoxyl, C 1-6 haloalkyl, C 1-6 haloalkoxyl, C 1-6 hydroxyalkyl, C 1-6 amino-alkyl, optionally substituted C 3-6 cycloalkyl, optionally substituted C 3-6 heterocycloalkyl, optionally substi-tuted 5 to 8-membered aryl, optionally substituted 5 to 8-membered heteroaryl, optionally substituted 8 to 10-membered fused bicyclic aryl, optionally substituted 8 to 10-membered fused bicyclic heteroaryl, op-tionally substituted -C 1-2 alkylene-C 3-6 cycloalkyl, optionally substituted -C 1-2 alkylene-C 3-6 heterocycloalkyl, optionally substituted -C 1-2 alkylene-5 to 8-membered aryl,
- R 1 is optionally substituted -C 1-2 alkylene-C 3-6 cycloalkyl, optionally substitut-ed -C 1-2 alkylene-C 3-6 heterocycloalkyl, optionally substituted -C 1-2 alkylene-5 to 8-membered aryl, option-ally substituted -C 1-2 alkylene-5 to 8-membered heteroaryl, optionally substituted -C 1-2 alkylene-8 to 10-membered fused bicyclic aryl, and optionally substituted -C 1-2 alkylene-8 to 10-membered fused bicy-clic heteroaryl; preferably, is optionally substituted -C 1-2 alkylene-C 3-6 cycloalkyl, optionally substituted -C 1-2 alkylene-C 3-6 heterocycloalkyl; more preferably, optionally substituted -methylene-C 3-6 heterocycloal-kyl.
- C 3-6 heterocycloalkyl is selected from heterocycloalkyl for R 1’ as defined in formula VI, VII or VIII.
- the substituent is selected from the group consisting of C 1-6 alkyl, C 1-6 alkoxyl, C 1-6 haloalkyl, C 1-6 haloalkoxyl, or C 1-6 hydroxyalkyl (preferably, the C 1-6 alkyl) is further substituted
- the substituent is selected from the group consisting of C 1-6 alkyl, C 1-6 alkoxyl, C 1-6 haloalkyl, C 1-6 haloalkoxyl, C 1-6 hydroxyalkyl, C 1-6 aminoalkyl, optionally substituted C 3-6 cycloalkyl, optionally substituted C 3-6 heterocycloalkyl, optionally substituted 5 to 8-membered aryl, optionally substituted 5 to 8-membered heteroaryl, optionally substituted 8 to 10-membered fused bicyclic aryl, optionally substituted 8 to 10-membered fused bicyclic heteroaryl; pref-erably, the substituent is selected from the group consisting of optionally substituted
- substituents selected from the group consisting of deuterium, halogen, C 1-6 alkyl, C 1-6 hydroxyalkyl, C 1-6 deuteroalkyl, C 3-6 cycloalkyl, C 3-6 heterocyclo
- optionally substituted means unsubstituted or being substituted with one or more (e.g., 1, 2 or 3) substituents selected from the group consisting of deuterium, halogen, C 1-6 alkyl, C 1-6 hydroxyalkyl, and C 1-6 deuteroalkyl.
- R 1 is - (CH 2 ) -R 1’ , and R 1’ is as defined in formula VI, VII, or VIII.
- R M and R N are each independently selected from hydrogen, halogen, hydroxy, cyano, carboxyl, C 1-6 alkyl, C 1-6 alkoxyl, C 1-6 haloalkyl, C 1-6 haloalkoxyl, C 1-6 hydroxyalkyl, C 1-6 aminoalkyl, optionally substituted C 3-6 cycloalkyl, optionally substituted C 3-6 heterocycloalkyl; furthermore, the C 1-6 alkyl, C 1-6 alkoxyl, C 1-6 haloalkyl, C 1-6 haloalkoxyl, C 1-6 hydroxyalkyl may be further optionally substituted.
- R M and R N are each independently selected from hydrogen, and C 1-6 alkyl.
- Y is -CH-or N; preferably, Y is N.
- R 2 is optionally substituted C 1-6 alkoxy, or optionally substituted C 1-6 alkyl; preferably, R 2 is optionally substituted C 1-6 alkyl.
- optionally substituted means unsubstituted or being substituted with one or more (e.g., 1, 2 or 3) substituents selected from the group consisting of deuterium, C 1-6 alkoxy, C 1-6 deu-teroalkoxy, and C 1-6 haloalkoxy.
- B is R A is optionally substituted phenyl or optionally substituted 6-membered heteroaryl; in some embodiments, the compound of formula (I) is a compound of formula (II) :
- A is optionally substituted 5 to 6-membered heterocycloalkylene, optionally substituted 5 to 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene;
- G is optionally substituted C 1-6 alkylene
- Y is -CH-or N
- R 8 and R 9 are each independently selected from hydrogen, deuterium, halogen, hydroxy, cyano, op-tionally substituted C 1-3 alkyl, or R 8 and R 9 combine with the atoms to which they are attached to form an optionally substituted C 3-6 cycloalkyl or a carbonyl;
- R 10 and R 11 are each independently selected from hydrogen, deuterium, halogen, hydroxy, cyano, op-tionally substituted C 1-3 alkyl, or R 10 and R 11 combine with the atoms to which they are attached to form an optionally substituted C 3-6 cycloalkyl or a carbonyl;
- R 12 and R 13 are each independently selected from hydrogen, deuterium, halogen, hydroxy, optionally substituted C 1-3 alkyl, -O (C 1-6 alkyl) , -S (C 1-6 alkyl) or -N (C 1-6 alkyl) (C 1-6 alkyl) ; furthermore, the C 1-6 alkyl may be further optionally substituted;
- Y is -CH-or N;
- X 1 is N or C
- X 2 is N or -CR a -;
- X 3 is N or -CR b -;
- X 4 is N or -CR c -;
- X 5 is N or C
- X 6 is S, O, N, or -CH-;
- R a , R b , and R c are each independently hydrogen, halogen, cyano, C 1-3 alkyl, C 1-3 alkoxyl, C 1-3 haloal-kyl, C 1-3 haloalkoxyl, C 3-6 cycloalkyl, or C 3-6 heterocycloalkyl (or 3 to 6-membered heterocycloalkyl) ;
- R 1 is selected from hydrogen, C 1-6 alkyl, C 1-6 alkoxyl, C 1-6 haloalkyl, C 1-6 haloalkoxyl, C 1-6 hydroxy-alkyl, C 1-6 aminoalkyl, optionally substituted C 3-6 cycloalkyl, optionally substituted 3 to 6-membered het-erocycloalkyl, optionally substituted 5 to 8-membered aryl, optionally substituted 5 to 8-membered het-eroaryl, optionally substituted 8 to 10-membered fused bicyclic aryl or optionally substituted 8 to 10-membered fused bicyclic heteroaryl; furthermore, the C 1-6 alkyl, C 1-6 alkoxyl, C 1-6 haloalkyl, C 1-6 haloalkoxyl, and C 1-6 hydroxyalkyl may be further optionally substituted;
- R 2 is optionally substituted C 1-6 alkoxy, or optionally substituted C 1-6 alkyl
- R M and R N are each independently selected from hydrogen, halogen, hydroxy, cyano, carboxyl, C 1-6 alkyl, C 1-6 alkoxyl, C 1-6 haloalkyl, C 1-6 haloalkoxyl, C 1-6 hydroxyalkyl, C 1-6 aminoalkyl, optionally substi-tuted C 3-6 cycloalkyl, optionally substituted C 3-6 heterocycloalkyl (or 4 to 7-membered heterocycloalkyl) ; furthermore, the C 1-6 alkyl, C 1-6 alkoxyl, C 1-6 haloalkyl, C 1-6 haloalkoxyl, and C 1-6 hydroxyalkyl may be fur-ther optionally substituted;
- R 3 is absent, or selected from hydrogen, C 1-6 alkyl, or C 1-6 haloalkyl, provided that when X 6 is O or S, R 3 is absent;
- E is a bond, or selected from N, or -CR d -, wherein R d is selected from hydrogen, halogen, hydroxy, cyano, carboxyl, optionally substituted C 1-6 alkyl, optionally substituted C 1-6 alkoxyl, C 1-6 aminoalkyl, C 1-6 hydroxyalkyl, or -NR e R f ; wherein R e and R f are each independently selected from hydrogen, or optionally substituted C 1-6 alkyl;
- R 4 is hydrogen, optionally substituted C 1-6 alkyl, optionally substituted C 1-6 heteroalkyl, optionally substituted C 3-10 cycloalkyl, optionally substituted C 3-10 cycloalkenyl, optionally substituted C 3-10 heterocy-cloalkyl (or 4 to 12-membered heterocycloalkyl) , optionally substituted 6 to 10-membered aryl, optionally substituted 5 to 10-membered heteroaryl, or
- R 5 is hydrogen, C 1-6 alkyl or C 3-6 cycloalkyl, furthermore, the C 1-6 alkyl or C 3-6 cycloalkyl may be fur-ther optionally substituted;
- R 4 and R 5 combine with the atoms to which they are attached to form a ring, wherein the ring is se-lected from optionally substituted 3 to 10-membered cycloalkyl, optionally substituted 3 to 10-membered heterocycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 4 to 11-membered bicyclic cycloalkyl, optionally substituted 4 to 11-membered bicyclic heterocycloalkyl.
- L is absent, or selected from -CH 2 -, -C (O) -, -CHR g -or -C (R g ) 2 -, wherein R g is optionally substituted C 1-6 alkyl;
- R 6 is hydrogen, or optionally substituted C 1-6 alkyl
- R 7 is optionally substituted C 1-6 alkyl, optionally substituted C 1-6 heteroalkyl, optionally substituted C 3-10 cycloalkyl, optionally substituted C 3-10 heterocycloalkyl (or 4 to 12-membered heterocycloalkyl) , op-tionally substituted 6 to 10-membered aryl, optionally substituted 5 to 10-membered heteroaryl;
- R 6 and R 7 combine with the atoms to which they are attached to form a ring, wherein the ring is selected from optionally substituted 3 to 10-membered cycloalkyl, optionally substituted 3 to 10-membered heterocycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 4 to 11-membered bicyclic cycloalkyl, optionally substituted 4 to 11-membered bicyclic heterocycloalkyl.
- X 1 is C
- X 2 is -CR a - (preferably -CH-)
- X 3 is -CR b - (preferably -CH-)
- X 4 is -CR c - (preferably -CH-)
- X 5 is -C-.
- R a , R b , and R c are each independently selected from the group consisting of hydrogen, halogen, cyano, C 1-3 alkyl, C 1-3 alkoxyl, C 1-3 haloalkyl, and C 1-3 haloalkoxyl; preferably, R a , R b , and R c are each independently hydrogen, or C 1-3 alkyl.
- X 1 , X 2 , X 3 , X 4 and X 5 are -CH-, R M and R N are hy-drogen.
- R 8 , R 9 , R 10 , R 11 , R 12 and R 13 are each independently selected from hydrogen, deuterium, halogen, hydroxy, cyano, optionally substituted C 1-3 alkyl; preferably, R 8 , R 9 , R 10 , R 11 , R 12 and R 13 are each independently hydrogen.
- the compound of formula (II) is a compound of formula (II-1)
- X 1 , X 2 , X 3 , X 4 , X 5 , X 6 , A, B, E, G, Y, R M , R N , R 1 , R 3 , R 4 , R 5 and n are as defined in formula II.
- the compound of formula (II) is a compound of formula (III) :
- A is optionally substituted 5 to 6-membered heterocycloalkylene, optionally substituted 5 to 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene;
- G is optionally substituted C 1-6 alkylene
- Y is -CH-or N
- X 6 is S, O, N, or -CH-;
- R a and R b are each independently hydrogen, halogen, cyano, C 1-3 alkyl, C 1-3 alkoxyl, C 1-3 haloalkyl, C 1-3 haloalkoxyl, C 3-6 cycloalkyl, or C 3-6 heterocycloalkyl (or 4 to 7-membered heterocycloalkyl) ;
- R 1 is selected from hydrogen, C 1-6 alkyl, C 1-6 alkoxyl, C 1-6 haloalkyl, C 1-6 haloalkoxyl, C 1-6 hydroxy-alkyl, C 1-6 aminoalkyl, optionally substituted C 3-6 cycloalkyl, optionally substituted C 3-6 heterocycloalkyl (or 4 to 7-membered heterocycloalkyl) , optionally substituted 5 to 8-membered aryl, optionally substituted 5 to 8-membered heteroaryl, optionally substituted 8 to 10-membered fused bicyclic aryl or optionally sub-stituted 8 to 10-membered fused bicyclic heteroaryl; furthermore, the C 1-6 alkyl, C 1-6 alkoxyl, C 1-6 haloalkyl, C 1-6 haloalkoxyl, and C 1-6 hydroxyalkyl may be further optionally substituted;
- R 2 is optionally substituted C 1-6 alkoxy, or optionally substituted C 1-6 alkyl
- R 3 is absent, or selected from hydrogen, C 1-6 alkyl, or C 1-6 haloalkyl, provided that when X 6 is O or S, R 3 is absent;
- E is a bond, or selected from N, or -CR d -, wherein R d is selected from hydrogen, halogen, hydroxy, cyano, carboxyl, optionally substituted C 1-6 alkyl, optionally substituted C 1-6 alkoxyl, C 1-6 aminoalkyl, C 1-6 hydroxyalkyl, or -NR e R f ; wherein R e and R f are independently selected from hydrogen, or optionally sub-stituted C 1-6 alkyl;
- R 4 is hydrogen, optionally substituted C 1-6 alkyl, optionally substituted C 1-6 heteroalkyl, optionally substituted C 3-10 cycloalkyl, optionally substituted C 3-10 cycloalkenyl, optionally substituted C 3-10 heterocy-cloalkyl (or 4 to 12-membered heterocycloalkyl) , optionally substituted 6 to 10-membered aryl, optionally substituted 5 to 10-membered heteroaryl, or
- R 5 is hydrogen, C 1-6 alkyl or C 3-6 cycloalkyl, furthermore, the C 1-6 alkyl or C 3-6 cycloalkyl may be fur-ther optionally substituted;
- R 4 and R 5 combine with the atoms to which they are attached to form a ring, wherein the ring is se-lected from optionally substituted 3 to 10-membered cycloalkyl, optionally substituted 3 to 10-membered heterocycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 4 to 11-membered bicyclic cycloalkyl, optionally substituted 4 to 11-membered bicyclic heterocycloalkyl;
- L is absent, or selected from -CH 2 -, -C (O) -, -CHR g -or -C (R g ) 2 -, wherein R g is optionally substituted C 1-6 alkyl;
- R 6 is hydrogen, or optionally substituted C 1-6 alkyl
- R 7 is optionally substituted C 1-6 alkyl, optionally substituted C 1-6 heteroalkyl, optionally substituted C 3-10 cycloalkyl, optionally substituted C 3-10 heterocycloalkyl (or 3 to 10-membered heterocycloalkyl) , op-tionally substituted 6 to 10-membered aryl, optionally substituted 5 to 10-membered heteroaryl;
- R 6 and R 7 combine with the atoms to which they are attached to form a ring, wherein the ring is selected from optionally substituted 3 to 10-membered cycloalkyl, optionally substituted 3 to 10-membered heterocycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 4 to 11-membered bicyclic cycloalkyl, optionally substituted 4 to 11-membered bicyclic heterocycloalkyl.
- the heteroatom is independently selected from one or more of N, O and S, and the number of the heteroatom is independently 1 to 4.
- the compound of formula (III) is a compound of for-mula (IVa) or formula (IVb) :
- A is optionally substituted 5 to 6-membered heterocycloalkylene, optionally substituted 5 to 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene;
- G is optionally substituted C 1-6 alkylene
- Y is -CH-or N
- X 6 is S, O, N, or -CH-;
- R a and R b are each independently hydrogen, halogen, cyano, C 1-3 alkyl, C 1-3 alkoxyl, C 1-3 haloalkyl, C 1-3 haloalkoxyl, C 3-6 cycloalkyl, C 3-6 heterocycloalkyl (or 4 to 7-membered heterocycloalkyl) ;
- R 1 is selected from hydrogen, C 1-6 alkyl, C 1-6 alkoxyl, C 1-6 haloalkyl, C 1-6 haloalkoxyl, C 1-6 hydroxy-alkyl, C 1-6 aminoalkyl, optionally substituted C 3-6 cycloalkyl, optionally substituted C 3-6 heterocycloalkyl (or 4 to 7-membered heterocycloalkyl) , optionally substituted 5 to 8-membered aryl, optionally substituted 5 to 8-membered heteroaryl, optionally substituted 8 to 10-membered fused bicyclic aryl or optionally sub-stituted 8 to 10-membered fused bicyclic heteroaryl; furthermore, the C 1-6 alkyl, C 1-6 alkoxyl, C 1-6 haloalkyl, C 1-6 haloalkoxyl, C 1-6 hydroxyalkyl may be further optionally substituted;
- R 1 is optionally substituted C 1-6 alkyl
- the C 1-6 alkyl is further substituted, the substituent is selected from optionally sub-stituted C 3-6 cycloalkyl, optionally substituted C 3-6 heterocycloalkyl, optionally substituted 5 to 8-membered aryl, optionally substituted 5 to 8-membered heteroaryl, optionally substituted 8 to 10-membered fused bicyclic aryl, optionally substituted 8 to 10-membered fused bicyclic heteroaryl;
- the substituent is optionally substituted C 3-6 heterocycloalkyl
- R 1 is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
- R 1 is
- R 1 is
- R 2 is optionally substituted C 1-6 alkoxy, or optionally substituted C 1-6 alkyl
- R 3 is absent, or selected from hydrogen, C 1-6 alkyl, or C 1-6 haloalkyl, provided that when X 6 is O or S, R 3 is absent;
- E is a bond, or selected from N, or -CR d -, wherein R d is selected from hydrogen, halogen, hydroxy, cyano, carboxyl, optionally substituted C 1-6 alkyl, optionally substituted C 1-6 alkoxyl, C 1-6 aminoalkyl, C 1-6 hydroxyalkyl, or -NR e R f ; wherein R e and R f are independently selected from hydrogen, or optionally sub-stituted C 1-6 alkyl;
- R 4 is hydrogen, optionally substituted C 1-6 alkyl, optionally substituted C 1-6 heteroalkyl, optionally substituted C 3-10 cycloalkyl, optionally substituted C 3-10 cycloalkenyl, optionally substituted C 3-10 heterocy-cloalkyl (or 4 to 12-membered heterocycloalkyl) , optionally substituted 6 to 10-membered aryl, optionally substituted 5 to 10-membered heteroaryl, or
- R 5 is hydrogen, C 1-6 alkyl or C 3-6 cycloalkyl, furthermore, the C 1-6 alkyl or C 3-6 cycloalkyl may be fur-ther optionally substituted;
- R 4 and R 5 combine with the atoms to which they are attached to form a ring, wherein the ring is se-lected from optionally substituted 3 to 10-membered cycloalkyl, optionally substituted 3 to 10-membered heterocycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 4 to 11-membered bicyclic cycloalkyl, optionally substituted 4 to 11-membered bicyclic heterocycloalkyl;
- the ring is selected from optionally substituted 3 to 10-membered cycloalkyl, optionally substituted 3 to 10-membered heterocycloalkyl;
- the ring is selected from optionally substituted 3 to 10-membered cycloalkyl
- L is absent, or selected from -CH 2 -, -C (O) -, -CHR g -or -C (R g ) 2 -, wherein R g is optionally substituted C 1-6 alkyl;
- R 6 is hydrogen, or optionally substituted C 1-6 alkyl
- R 7 is optionally substituted C 1-6 alkyl, optionally substituted C 1-6 heteroalkyl, optionally substituted C 3-10 cycloalkyl, optionally substituted C 3-10 heterocycloalkyl, optionally substituted 6 to 10-membered aryl, optionally substituted 5 to 10-membered heteroaryl;
- R 6 and R 7 combine with the atoms to which they are attached to form a ring, wherein the ring is selected from optionally substituted 3 to 10-membered cycloalkyl, optionally substituted 3 to 10-membered heterocycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 4 to 11-membered bicyclic cycloalkyl, optionally substituted 4 to 11-membered bicyclic heterocycloalkyl.
- the heteroatom is independently selected from one or more of N, O and S, and the number of the heteroatom is independently 1 to 4.
- R 4 and R 5 combine with the atoms to which they are attached to form a ring, wherein the ring is optionally substituted 3 to 10-membered cycloalkyl; preferably, is optionally substituted 3 to 6-membered cycloalkyl such as optionally substituted cyclopropyl, optionally substituted cyclobutyl, optionally substituted cyclopentyl or optionally substituted cyclohexyl, more preferably, optionally substi-tuted 3-membered cycloalkyl i.e., optionally substituted cyclopropyl.
- the 5 to 6-membered heterocycloalkylene contains one or two heteroatoms; preferably the heteroatom is each independently selected from N, O and S.
- A is optionally substituted 5 to 6-membered heterocy-cloalkylene, the heteroatom is independently selected from one or two of N, O and S, and the number of the heteroatom is independently 1 to 2. In some embodiments, A is
- A is optionally substituted 5 to 6-membered arylene
- A is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
- the 5 to 6-membered heteroarylene contains one, two or three (preferably one or two) heteroatoms; preferably the heteroatom is each independently selected from N, O and S.
- A is optionally substituted 5 to 6-membered het-eroarylene
- the heteroatom is independently selected from one or two of N, O and S, and the number of the heteroatom is independently 1 to 3.
- A is optionally substituted 5-membered heteroarylene
- the heteroatom is independently selected from N, O and S, and the number of the heteroatom is inde-pendently 1 to 2; preferably, one of the heteroatom is N, and the other, if present, is O or S.
- A is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
- A is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
- B is selected from the group consisting of optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 3 to 6-membered heterocycloal-kenylene, optionally substituted 4 to 11-membered bicyclic alkylene, and optionally substituted 4 to 11-membered bicyclic heteroalkylene.
- B is
- G is optionally substituted C 1-6 alkylene; preferabaly, G is optionally substituted -C (C 1-3 alkyl) 2 -.
- optionally substituted means unsubstitued or being substituted with one or more deuterium.
- G is N
- Y is N.
- X 6 is N.
- R 1 is C 1-6 alkyl, C 1-6 alkoxyl, C 1-6 haloalkyl, C 1-6 haloalkoxyl, C 1-6 hydroxyalkyl, C 1-6 aminoalkyl, optionally substituted C 3-6 cycloalkyl, or optionally substi-tuted C 3-6 heterocycloalkyl (or 3 to 6-membered heterocycloalkyl) .
- R 1 is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
- R 1 is C 1-6 alkyl and the C 1-6 alkyl is further substituted with optionally substituted C 3-6 cycloalkyl, optionally substituted C 3-6 heterocycloalkyl.
- R 1 is C 1 alkyl; and the C 1 alkyl is further substituted with optionally substituted C 3-6 heterocycloalkyl, wherein the heterocycloalkyl is saturated and the hetero-cycloalkyl is attached to the rest of the molecule via the N atom on the ring.
- R 1 is C 1 alkyl; and the C 1 alkyl is further substituted with optionally substituted C 3-6 heterocycloalkyl, wherein the heterocycloalkyl is saturated and the hetero-cycloalkyl is attached to the rest of the molecule via the N atom on the ring. and the C 3-6 heterocycloalkyl is 5 to 7 menbered heterocycloalkyl and has l to 2 heteroatoms.
- R 1 is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
- R 1 is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
- R 1 is optionally substituted 4 to 5-membered het-eroalkyl and 4 to 5-membered heteroaryl.
- R 1 is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
- R 1 is optionally substituted 5 to 8-membered aryl, op-tionally substituted 5 to 8-membered heteroaryl, optionally substituted 8 to 10-membered fused bicyclic aryl or optionally substituted 8 to 10-membered fused bicyclic heteroaryl, the heteroatom is independently selected from one or two of N, O and S, and the number of the heteroatom is independently 1 to 4.
- R 1 is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
- R 2 is optionally substituted C 1-6 alkyl or optionally substituted C 1-6 alkoxy.
- R 2 is preferably,
- R 3 is C 1-6 alkyl. In some embodiments, R 3 is C 1-6 deu-terated alkyl. In some embodiments, R 3 is
- R 3 is C 1-6 haloalky.
- R 3 is
- E is a bond, or selected from N, or -CR d -, wherein R d is selected from hydrogen, halogen, hydroxy, cyano, carboxyl, optionally substituted C 1-6 alkyl, optionally substituted C 1-6 alkoxyl, C 1-6 aminoalkyl, C 1-6 hydroxyalkyl, or -NR e R f ; wherein R e and R f are independent-ly selected from hydrogen, or optionally substituted C 1-6 alkyl; preferably, E is a bond, or selected from N, or -CR d -, wherein R d is selected from hydrogen, halogen, hydroxy or C 1-6 alkyl.
- R d is halogen; In some embodiments, R d is -F, -Cl, -Br or -I; preferably, -F.
- R d is C 1-6 alkyl
- R d is
- R d is C 1-6 haloalkyl; In some embodiments, R d is -CF 3 .
- R d is C 1-6 alkoxyl
- R d is
- R d is C 1-6 aminoalkyl; In some embodiments, R d is
- R e or R f is C 1-6 alkyl; In some embodiments, R e or R f is independently selected from and preferably the other is hydrogen.
- L is selected from -CH 2 -, -C (O) -, -CHR g -or -C (R g ) 2 -, wherein R g is optionally substituted C 1-6 alkyl.
- R 4 is hydrogen, optionally substituted C 1-6 alkyl, optionally substituted C 1-6 heteroalkyl, optionally substituted C 3-10 cycloalkyl, optionally substituted C 3-10 heterocycloalkyl (or 4 to 12-membered heterocycloalkyl) , optionally substituted 6 to 10-membered aryl, optionally substituted 5 to 10-membered heteroaryl, or
- R 4 is hydrogen, optionally substituted C 1-6 alkyl, or
- R 6 is hydrogen, or optionally substituted C 1-6 alkyl; preferably, hydrogen, or C 1-6 alkyl.
- R 7 is optionally substituted C 1-6 alkyl, optionally substituted C 1-6 heteroalkyl, optionally substituted C 3-10 cycloalkyl, optionally substituted C 3-10 heterocycloalkyl, optionally substituted 6 to 10-membered aryl, optionally substituted 5 to 10-membered heteroaryl; preferably, R 7 is C 1-6 alkyl, op-tionally substituted C 3-10 cycloalkyl.
- R 4 is hydrogen, optionally substituted C 1-6 alkyl, op-tionally substituted C 1-6 heteroalkyl, optionally substituted C 3-10 cycloalkyl, optionally substituted C 3-10 het-erocycloalkyl (or 4 to 12-membered heterocycloalkyl) , optionally substituted 6 to 10-membered aryl, op-tionally substituted 5 to 10-membered heteroaryl, or wherein R 6 is hydrogen, or optionally sub-stituted C 1-6 alkyl; R 7 is optionally substituted C 1-6 alkyl, optionally substituted C 1-6 heteroalkyl, optionally substituted C 3-10 cycloalkyl, optionally substituted C 3-10 heterocycloalkyl, optionally substituted 6 to 10-membered aryl, optionally substituted 5 to 10-membered heteroaryl.
- optionally substituted means unsubstituted or being substituted by one or more substituents selected from the group consisting of hydroxyl, cyano, halogen, and C 1-6 alkyl.
- R 4 is hydrogen, C 1-6 alkyl, 3 to 6-membered heterocycloalkyl, 3 to 6-membered cycloalkyl, 6 to 10-membered aryl, 5 to 10-membered heteroaryl, or wherein R 6 is hydrogen, or C 1-6 alkyl; R 7 is C 1-6 alkyl, 3-to 10-membered heterocycloalkyl; wherein the 3 to 10-membered heterocycloalkyl is optionally substituted by C 1-6 alkyl, 6 to 10-membered aryl, -CO-O-C 3-6 cycloalkyl or -CO-O-C 1-6 alkyl; the 3 to 6-membered cycloalkyl is optionally substituted by hydroxyl,
- C 3-10 heterocycloalkyl (or 4 to 12-membered heterocycloalkyl) contains 1 or 2 heteroatoms selected from the group consisting of N, O and S.
- R 4 is optionally substituted C 1-6 alkyl, optionally sub-stituted C 1-6 heteroalkyl, optionally substituted C 3-10 cycloalkyl, optionally substituted C 3-10 heterocycloalkyl, the heteroatom is N, O or S, and the number of the heteroatom is independently 1 to 2.
- R 4 is
- R 4 is optionally substituted 6 to 10-membered aryl, optionally substituted 5 to 10-membered heteroaryl.
- R 4 is
- R 5 is optionally substituted C 1-6 alkyl. In some em-bodiments, R 5 is
- R 5 is optionally substituted C 3-6 cycloalkyl. In some embodiments, R 5 is
- the het-erocycloalkyl or the bicyclic heterocycloalkyl contains 1, 2 or 3 heteroatom selected from the group con-sisting of N, O and S.
- R 4 and R 5 combine with the atoms to which they are attached to form a ring, wherein the ring is selected from optionally substituted 3 to 10-membered cyclo-alkyl, optionally substituted 3 to 10-membered heterocycloalkyl, optionally substituted 4 to 11-membered bicyclic cycloalkyl, optionally substituted 4 to 11-membered bicyclic heterocycloalkyl, the heteroatom is N, O or S, and the number of the heteroatom is independently 1 to 3,
- the ring is
- R 4 and R 5 combine with the atoms to which they are attached to form a ring, wherein the ring is optionally substituted 3 to 10-membered cycloalkyl; preferably, the ring is optionally substituted 3 to 4-membered cycloalkyl; more preferably, the ring is cy optionally substituted clopropyl; preferably, wherein optionally substituted means unsubstituted or being substituted with one or more sub-stituents selected from the group consisting of C 1-4 alkyl, C 1-4 deuteroalkyl, cyano, C 1-4 hydroxyalkyl, and C 1-4 haloalkyl.
- R 6 is optionally substituted C 1-6 alkyl; In some em-bodiments, R 6 is
- R 7 is optionally substituted C 3-10 cycloalkyl, or optionally substituted C 3-10 het-erocycloalkyl.
- substituents for C 3-10 heterocycloalkyl are selected from the group con-sisting of halogen, C 1-6 alkyl, 6 to 10-membered aryl, -CO-O-C 3-6 cycloalkyl, -CO-O-C 1-6 alkyl, -CO-C 3-6 cycloalkyl, -CO-C 1-6 alkyl, -CO-C 1-6 alkenyl, and -CO-C 1-6 alkynyl, and/or substituents for C 3-10 cycloalkyl are selected from the group consisting of hydroxyl, cyano, halogen, or C 1-6 alkyl.
- substituents for C 3-10 heterocycloalkyl are selected from the group con-sisting of C 1-6 alkyl, 6 to 10-membered aryl, -CO-O-C 3-6 cycloalkyl and -CO-O-C 1-6 alkyl, and/or substitu-ents for C 3-10 cycloalkyl are selected from the group consisting of hydroxyl, cyano, halogen, or C 1-6 alkyl.
- C 3-10 heterocycloalkyl contains 1 to 3 heteroatoms selected from the group consisting of N, O and S.
- R 7 is optionally substituted C 3-10 cycloalkyl, optionally substituted C 3-10 heter-ocycloalkyl; preferably, the C 3-10 heterocycloalkyl optionally substituted by C 1-6 alkyl, C 6-10 aryl , -CO-O-C 3-6 cycloalkyl or -CO-O-C 1-6 alkyl; the C 3-10 cycloalkyl optionally substituted by hydroxyl, cyano, halogen, or C 1-6 alkyl; in C 3-10 heterocycloalkyl, the heteroatom is independently selected from one or more of N, O and S, and the number of the heteroatom is independently 1 to 3.
- R 7 is C 1-6 alkyl.
- R 7 is
- R 7 is optionally substituted 3 to 6-membered heterocycloalkyl.
- the 3 to 6-membered heterocycloalkyl contains 1 or 2 heteroatom se-lected from N, O or S; preferably, is
- R 7 is 3 to 6-membered heterocycloalkyl, preferably, the heteroatom is N, O or S, and the number of the heteroatom is independently 1 to 2. In some embodi-ments, R 7 is
- R 7 is optionally substituted C 3-6 cycloalkyl. In some embodiments, in R 7 , the C 3-6 cycloalkyl is
- R 7 is C 3-6 cycloalkyl
- R 7 is
- R 7 is optionally substituted C 6-10 aryl. In some embodiments, in R 7 , the C 6-10 aryl is
- R 7 is C 6-10 aryl.
- R 7 is
- R 7 is optionally substituted C 1-6 alkyl or optionally substituted 3 to 6-membered heterocycloalkyl.
- substituents for the 3 to 6-membered heterocycloalkyl is selected from -C (O) R i or -NHC (O) R i ; wherein R i is selected from the group consisting of C 1-6 alkyl, -C 2-6 alkenyl, C 2-6 al-kynyl; wherein the alkyl, alkenyl, or alkynyl is unsubtituted or one or more (such as 1, 2, 3 or 4) of H atom in the group being substituted with substituents selected from the group consisting of D, halogen, -CN, -OH, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 deuteroalkyl, C 1-6 hydroxyalkyl, C 1-6 aminoalkyl, alkylamino.
- substituents for the 3 to 6-membered heterocycloalkyl is selected from 6 to 10-membered aryl or -CO-O-C 1-6 alkyl.
- the 3-to 6-membered heterocycloalkyl contains 1 or 2 heteroatoms se-lected from N and O.
- R 7 is C 1-6 alkyl or 3 to 6-membered heterocycloalkyl; the 3 to 6-membered heterocycloalkyl optionally substituted by 6 to 10-membered aryl or -CO-O-C 1-6 alkyl; in 3 to 6-membered heterocycloalkyl, the heteroatom is independently selected from one or two of N and O, and the number of the heteroatom is independently 1 to 2.
- R 7 is
- L, R 6 and R 7 combine with the atoms to which they are attached to form a ring, wherein the ring is selected from optionally substituted 3 to 10-membered cy-cloalkyl, optionally substituted 3 to 10-membered heterocycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 4 to 11-membered bicyclic cycloalkyl, optionally substi-tuted 4 to 11-membered bicyclic heterocycloalkyl.
- substituents for the ring formed by L, R 6 and R 7 combined with the atoms to which they are attached is -C (O) R i ; wherein R i is selected from the group consisting of C 1-6 alkyl, -C 2-6 alkenyl, C 2-6 alkynyl, C 3-10 cycloalkyl, and 3 to 10-membered heterocycloalky; wherein the alkyl, alkenyl, alkynyl, cycloalkyl, or heterocycloalky is unsubtituted or one or more (such as 1, 2, 3 or 4) of H atom in the group being substituted with substituents selected from the group consisting of D, halogen, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 deuteroalkyl, C 1-6 hydroxyalkyl, C 1-6 aminoalkyl, alkylamino, and C 3-6 cycloalkyl.
- the “halogen” is F, Cl, Br or I; preferably, F.
- the compound of formula (IVa) is a compound of formula (V) :
- X 1 , X 2 , X 3 , X 4 , X 5 , X 6 , A, B, E, G, L, Y, R a , R b , R c , R A , R B , R M , R N , R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 and n are each independently corresponding groups in specific compound shown below or compounds of examples 1 to 30.
- the present invention also has some schemes derived from any combination of the above-mentioned variables.
- the present invention also provides, but is not limited to, the following compounds, and stereoisomers thereof or their pharmacologically acceptable salts:
- the atoms may exhibit their natural isotopic abundances, or one or more of the atoms may be artificially enriched in a particular isotope having the same atomic num-ber, but an atomic mass or mass number different from the atomic mass or mass number predominantly found in nature.
- the present disclosure as described and claimed herein is meant to include all suitable isotopic variations of the compounds of Formulas above and embodiments thereof.
- different isotopic forms of hydrogen (H) include protium (1H) and deuterium (2H, also denoted herein as D) .
- Pro-tium is the predominant hydrogen isotope found in nature.
- Enriching for deuterium may afford certain therapeutic advantages, such as increasing in vivo half-life or reducing dosage requirements or may pro-vide a compound useful as a standard for characterization of biological samples.
- Isotopically-enriched compounds can be prepared without undue experimentation by conventional techniques well known to those skilled in the art or by processes analogous to those described in the Schemes and Examples herein using appropriate isotopically-enriched reagents and/or intermediates.
- the compound of formula (V) is a compound of for-mula (VI) :
- R 1’ of formula (VI) is selected from:
- the compound of formula (IVa) is a compound of formula (VII) :
- R 1’ of formula (VII) is selected from:
- the compound of formula (IVa) is a compound of formula (VIII) :
- R 1’ of formula (VIII) is selected from:
- the compound of formula (I) is Compound A as defined below.
- the present invention provides a Compound A, a pharmaceutically acceptable salt thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt, wherein, the Compound A is selected from any one of the following compounds in Table 1:
- the present invention also provides a pharmaceutical composition
- a pharmaceutical composition comprising a compound, wherein a pharmaceutically acceptable salt thereof, a solvate thereof or a solvate of the phar-maceutically acceptable salt as defined above, and a pharmaceutically acceptable carrier, preferably, the compound is the Compound A as defined above.
- the present invention also provides a use of a compound, wherein a pharmaceuti-cally acceptable salt thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt, or the pharmaceutical composition as defined above in the preparation of a medicament used for the treatment of cancer, preferably, the compound is the Compound A as defined above.
- the cancer is a Ras-driven cancer.
- the cancer comprises a Ras mutation.
- Ras mutation is a position 12, 13 or 61.
- Ras mutation is at position 12.
- Ras mutation is selected from the group consisting of G12C, G12D, G12V, G12A, G12R, G12S, G13C, G13D, Q61H, Q61R and Q61L, or a com-bination thereof.
- Ras mutation is a position selected from the group consisting of G12D, G12V, and G12R, or a combination thereof.
- the cancer is pancreatic cancer, appendiceal cancer, small bowel cancer, colorectal cancer, ampullary cancer, non-small cell lung cancer, cervical cancer, lung cancer, endometrial cancer, acute myeloid leukemia, gastrointestinal neuroendocrine tumor, uterine endometrioid carcinoma, oesophagogastric cancer, bladder cancer, ovarian cancer, melanoma, multiple myeloma, thyroid gland adenocarcinoma, a myelodysplastic syndrome, or squamous cell lung carcinoma.
- cancer is pancreatic cancer, lung cancer, or colorectal cancer.
- Ras protein is KRAS.
- the compound can be used in combination with an additional anti-cancer ther-apy.
- the additional anti-cancer therapy is an EGFR inhibitor, a second Ras inhibitor, a SHP2 inhibitor, a SOS1 inhibitor, a Raf inhibitor, a MEK inhibitor, an ERK inhibitor, a Pl3K inhibitor, a PTEN inhibitor, an AKT inhibitor, an mTORC1 inhibitor, a BRAF inhibitor, a PD-L1 inhibitor, a PD-1 in-hibitor, a CDK4/6 inhibitor, a HER2 inhibitor, or a combination thereof.
- the present invention also provides a method for treating cancer in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of the com-pound or a pharmaceutically acceptable salt thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt as defined above, or the pharmaceutical composition as defined above, preferably the com-pound is the Compound A as defined above.
- the cancer is a Ras-driven cancer.
- the cancer comprises a Ras mutation.
- Ras mutation is a position 12, 13 or 61.
- Ras mutation is at position 12.
- the Ras mutation is at a position selected from the group consisting of G12C, G12D, G12V, G12A, G12R, G12S, G13C, G13D, Q61H, Q61R and Q61L, or a combination thereof.
- Ras mutation is a position selected from the group consisting of G12D, G12V, and G12R, or a combination thereof.
- the cancer is pancreatic cancer, appendiceal cancer, small bowel cancer, colorectal cancer, ampullary cancer, non-small cell lung cancer, cervical cancer, lung cancer, endometrial cancer, acute myeloid leukemia, gastrointestinal neuroendocrine tumour, uterine endometrioid carcinoma, oesophagogastric cancer, bladder cancer, ovarian cancer, melanoma, multiple myeloma, thyroid gland adenocarcinoma, a myelodysplastic syndrome, or squamous cell lung carcinoma.
- cancer is pancreatic cancer, lung cancer, or colorectal cancer.
- Ras protein is KRAS.
- the method is provided of treating a Ras protein-related disorder in a subject in need thereof.
- the method further comprises administering an additional anti-cancer therapy.
- the additional anti-cancer therapy is an EGFR inhibitor, a second Ras inhibitor, a SHP2 inhibitor, a SOS1 inhibitor, a Raf inhibitor, a MEK inhibitor, an ERK inhibitor, a Pl3K inhibitor, a PTEN inhibitor, an AKT inhibitor, an mTORC1 inhibitor, a BRAF inhibitor, a PD-L1 inhibitor, a PD-1 in-hibitor, a CDK4/6 inhibitor, a HER2 inhibitor, or a combination thereof.
- Embodiment 1 The compound of any one of the preceding embodiments, or a stereoisomer thereof, a pharmaceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt thereof, wherein the compound is a compound of formula (I) ;
- n 0, 1, 2, or 3;
- A is optionally substituted 5 to 6-membered heterocycloalkylene, optionally substituted 5 to 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene;
- B is selected from optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 3 to 6-membered heterocycloalkenylene, optionally substituted 4 to 11-membered bicyclic cycloalkylene, or optionally substituted 4 to 11-membered bicyclic heterocycloalkylene;
- G is optionally substituted C 1-6 alkylene
- R 8 and R 9 are each independently selected from hydrogen, deuterium, halogen, hydroxy, cyano, op-tionally substituted C 1-3 alkyl, or R 8 and R 9 combine with the atoms to which they are attached to form an optionally substituted C 3-6 cycloalkyl or a carbonyl;
- R 10 and R 11 are each independently selected from hydrogen, deuterium, halogen, hydroxy, cyano, op-tionally substituted C 1-3 alkyl, or R 10 and R 11 combine with the atoms to which they are attached to form an optionally substituted C 3-6 cycloalkyl or a carbonyl;
- R 12 and R 13 are each independently selected from hydrogen, deuterium, halogen, hydroxy, optionally substituted C 1-3 alkyl, -O (C 1-6 alkyl) , -S (C 1-6 alkyl) or -N (C 1-6 alkyl) (C 1-6 alkyl) ; furthermore, the C 1-6 alkyl may be further optionally substituted;
- R A is optionally substituted C 3-6 cycloalkyl, optionally substituted C 3-6 heterocycloalkyl (or 4 to 7-membered heterocycloalkyl) , optionally substituted 5 to 8-membered aryl, optionally substituted 5 to 8-membered heteroaryl, optionally substituted 8 to 10-membered fused bicyclic aryl or optionally substi-tuted 8 to 10-membered fused bicyclic heteroaryl;
- R B is selected from hydrogen, C 1-6 alkyl, C 1-6 alkoxyl, C 1-6 haloalkyl, C 1-6 haloalkoxyl, optionally sub-stituted C 3-6 cycloalkyl, or optionally substituted C 3-6 heterocycloalkyl (or 4 to 7-membered heterocycloal-kyl) ; or provided that when n exceeds 1, any two of R B combine with the atoms to which they are attached to form a 3 to 6-membered ring, wherein the ring can be optionally substituted with halogen, hydroxy, and C 1-3 alkyl;
- X 1 is N or C
- X 2 is N or -CR a -;
- X 3 is N or -CR b -;
- X 4 is N or -CR c -;
- X 5 is N or C
- X 6 is S, O, N, or -CH-;
- R a , R b , and R c are each independently hydrogen, halogen, cyano, C 1-3 alkyl, C 1-3 alkoxyl, C 1-3 haloal-kyl, C 1-3 haloalkoxyl, C 3-6 cycloalkyl, or C 3-6 heterocycloalkyl (or 4 to 7-membered heterocycloalkyl) ;
- R 3 is absent, or selected from hydrogen, C 1-6 alkyl, or C 1-6 haloalkyl, provided that when X 6 is O or S, R 3 is absent;
- E is a bond, or selected from N, or -CR d -, wherein R d is selected from hydrogen, halogen, hydroxy, cyano, carboxyl, optionally substituted C 1-6 alkyl, optionally substituted C 1-6 alkoxyl, C 1-6 aminoalkyl, C 1-6 hydroxyalkyl, or -NR e R f ; wherein R e and R f are independently selected from hydrogen, or optionally sub-stituted C 1-6 alkyl;
- R 4 is hydrogen, optionally substituted C 1-6 alkyl, optionally substituted C 1-6 heteroalkyl, optionally substituted C 3-10 cycloalkyl, optionally substituted C 3-10 cycloalkenyl, optionally substituted C 3-10 heterocy-cloalkyl (or 4 to 12-membered heterocycloalkyl) , optionally substituted 6 to 10-membered aryl, optionally substituted 5 to 10-membered heteroaryl, or
- R 5 is hydrogen, C 1-6 alkyl or C 3-6 cycloalkyl, furthermore, the C 1-6 alkyl or C 3-6 cycloalkyl may be fur-ther optionally substituted;
- R 4 and R 5 combine with the atoms to which they are attached to form a ring, wherein the ring is se-lected from optionally substituted 3 to 10-membered cycloalkyl, optionally substituted 3 to 10-membered heterocycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 4 to 11-membered bicyclic cycloalkyl, optionally substituted 4 to 11-membered bicyclic heterocycloalkyl;
- L is absent, or selected from -CH 2 -, -C (O) -, -CHR g -or -C (R g ) 2 -, wherein R g is optionally substituted C 1-6 alkyl;
- R 6 is hydrogen, or optionally substituted C 1-6 alkyl
- R 7 is optionally substituted C 1-6 alkyl, optionally substituted C 1-6 heteroalkyl, optionally substituted C 3-10 cycloalkyl, optionally substituted C 3-10 heterocycloalkyl (or 3 to 10-membered heterocycloalkyl) , op-tionally substituted 6 to 10-membered aryl, optionally substituted 5 to 10-membered heteroaryl;
- R 6 and R 7 combine with the atoms to which they are attached to form a ring, wherein the ring is selected from optionally substituted 3 to 10-membered cycloalkyl, optionally substituted 3 to 10-membered heterocycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 4 to 11-membered bicyclic cycloalkyl, optionally substituted 4 to 11-membered bicyclic heterocycloalkyl.
- Embodiment 2 The compound of embodiment 1, or a stereoisomer thereof, a pharmaceutically ac-ceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharma-ceutically acceptable salt thereof, wherein the compound of formula (I) is a compound of formula (I-1)
- X 1 , X 2 , X 3 , X 4 , X 5 , X 6 , A, B, E, G, R A , R B , R 3 , R 4 , R 5 and n are as defined in embodiment 1.
- Embodiment 3 The compound of any one of embodiments 1 to 2, or a stereoisomer thereof, a phar-maceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt thereof, wherein
- R 1 is selected from the group consisting of hydrogen, C 1-6 alkyl, C 1-6 alkoxyl, C 1-6 haloalkyl, C 1-6 haloalkoxyl, C 1-6 hydroxyalkyl, C 1-6 aminoalkyl, optionally substituted C 3-6 cycloalkyl, optionally substi-tuted C 3-6 heterocycloalkyl, optionally substituted 5 to 8-membered aryl, optionally substituted 5 to 8-membered heteroaryl, optionally substituted 8 to 10-membered fused bicyclic aryl, optionally substituted 8 to 10-membered fused bicyclic heteroaryl, optionally substituted -C 1-2 alkylene-C 3-6 cycloalkyl, optionally substituted -C 1-2 alkylene-C 3-6 heterocycloalkyl, optionally substituted -C 1-2 alkylene-5 to 8-membered aryl, optionally substituted -C 1-2 alkylene-5 to 8-membered hetero
- the substituent is selected from the group consisting of C 1-6 alkyl, C 1-6 alkoxyl, C 1-6 haloalkyl, C 1-6 haloalkoxyl, C 1-6 hydroxyalkyl, C 1-6 aminoalkyl, optionally substituted C 3-6 cycloalkyl, optionally sub-stituted C 3-6 heterocycloalkyl, optionally substituted 5 to 8-membered aryl, optionally substituted 5 to 8-membered heteroaryl, optionally substituted 8 to 10-membered fused bicyclic aryl, optionally substituted 8 to 10-membered fused bicyclic heteroaryl; preferably, the substituent is selected from the group consist-ing of optionally substituted C 3-6 cycloalkyl, optionally substituted C
- Y is -CH-or N
- R 2 is optionally substituted C 1-6 alkoxy, or optionally substituted C 1-6 alkyl; preferably, R 2 is optionally substituted C 1-6 alkyl; and
- R M and R N are each independently selected from hydrogen, halogen, hydroxy, cyano, carboxyl, C 1-6 alkyl, C 1-6 alkoxyl, C 1-6 haloalkyl, C 1-6 haloalkoxyl, C 1-6 hydroxyalkyl, C 1-6 aminoalkyl, optionally substi-tuted C 3-6 cycloalkyl, optionally substituted C 3-6 heterocycloalkyl; furthermore, the C 1-6 alkyl, C 1-6 alkoxyl, C 1-6 haloalkyl, C 1-6 haloalkoxyl, C 1-6 hydroxyalkyl may be further optionally substituted.
- R M and R N are each independently selected from hydrogen, and C 1-6 alkyl.
- Embodiment 4 The compound of embodiment 3, or a stereoisomer thereof, a pharmaceutically ac-ceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharma-ceutically acceptable salt thereof, wherein
- R 1 is optionally substituted C 1-6 alkyl
- the C 1-6 alkyl is further substituted, and the substituent is selected from optionally substi-tuted C 3-6 cycloalkyl, optionally substituted C 3-6 heterocycloalkyl, optionally substituted 5 to 8-membered aryl, optionally substituted 5 to 8-membered heteroaryl, optionally substituted 8 to 10-membered fused bicyclic aryl, optionally substituted 8 to 10-membered fused bicyclic heteroaryl.
- Embodiment 5 The compound of any one of embodiments 3 to 4, or a stereoisomer thereof, a phar-maceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt thereof, wherein
- R 1 is optionally substituted C 1-6 alkyl
- the C 1-6 alkyl is further substituted, and the substituent is optionally substituted C 3-6 heterocy-cloalkyl.
- Embodiment 6 The compound of any one of embodiments 3 to 4, or a stereoisomer thereof, a phar-maceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt thereof, wherein R 1 is C 1 alkyl; and the C 1 alkyl is further substitut-ed with optionally substituted C 3-6 heterocycloalkyl, wherein the heterocycloalkyl is saturated and the het-erocycloalkyl is attached to the rest of the molecule via the N atom on the ring.
- Embodiment 7 The compound of any one of embodiments 3 to 4, or a stereoisomer thereof, a phar-maceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt thereof, wherein R 1 is C 1 alkyl; and the C 1 alkyl is further substitut-ed with optionally substituted C 3-6 heterocycloalkyl, wherein the heterocycloalkyl is saturated and the het-erocycloalkyl is attached to the rest of the molecule via the N atom on the ring. and the C 3-6 heterocycloal-kyl is 5 to 7 menbered heterocycloalkyl and has l to 2 heteroatoms.
- Embodiment 8 The compound of any one of embodiments 3 to 7, or a stereoisomer thereof, a phar-maceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt thereof, wherein
- R 1 is selected from the group consisting of
- Embodiment 9 The compound according to any one of embodiments 3 to 8, or a stereoisomer thereof, a pharmaceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt thereof, wherein R 1 is C 1 alkyl substituted by C 6 heterocy-cloalkyl, optionally wherein the C 6 heterocycloalkyl is piperazine or morpholine.
- Embodiment 10 The compound of any one of embodiments 1 to 9, or a stereoisomer thereof, a phar-maceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt thereof, wherein R 4 and R 5 combine with the atoms to which they are attached to form a ring, wherein the ring is optionally substituted 3 to 10-membered cycloalkyl; pref-erably, is optionally substituted 3 to 6-membered cycloalkyl; more preferably, optionally substituted cyclo-propyl.
- Embodiment 11 The compound of any one of embodiments 1 to 9, or a stereoisomer thereof, a phar-maceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt thereof, wherein
- Embodiment 12 The compound of any one of embodiments 1 to 11, or a stereoisomer thereof, a pharmaceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt thereof, wherein
- A is optionally substituted 5 to 6-membered heteroarylene, the heteroatom is independently selected from one or two of N, O and S, and the number of the heteroatom is independently 1 to 3
- A is optionally substituted 5-membered heteroarylene, the heteroatom is independently se-lected from N, O and S, and the number of the heteroatom is independently 1 to 2; preferably, one of the heteroatom is N, and the other, if present, is O or S;
- A is selected from the group consisting of
- Embodiment 13 The compound of any one of embodiments 1 to 12, or a stereoisomer thereof, a pharmaceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt thereof, wherein
- X 1 is C
- X 2 is -CR a -
- X 3 is -CR b
- X 4 is -CR c -
- X 5 is -C-.
- Embodiment 14 The compound of any one of embodiments 1 to 13, or the stereoisomer thereof, the pharmaceutically acceptable salt thereof, the metabolite thereof, the prodrug thereof, the solvate thereof or the solvate of the pharmaceutically acceptable salt thereof, wherein, the compound is selected from any one of the compounds in Table 1.
- Embodiment 15 The compound of any one of embodiments 1 to 14, or the stereoisomer thereof, the pharmaceutically acceptable salt thereof, the metabolite thereof, the prodrug thereof, the solvate thereof or the solvate of the pharmaceutically acceptable salt thereof, wherein, the compound is selected from com-pounds of Examples 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 27, 28, 29, 30, 31, 32, 33, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 84, 86, 87, 88, and 90 in Table 1.
- Embodiment 16 A pharmaceutical composition comprising: the compound of any one of embodi-ments 1 to 15, or the stereoisomer thereof, the pharmaceutically acceptable salt thereof, the metabolite thereof, the prodrug thereof, the solvate thereof or the solvate of the pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
- Embodiment 17 A use of the compound of any one of embodiments 1 to 15, or the stereoisomer thereof, the pharmaceutically acceptable salt thereof, the metabolite thereof, the prodrug thereof, the solv-ate thereof or the solvate of the pharmaceutically acceptable salt thereof, or the pharmaceutical composi-tion according to embodiment 16, or the pharmaceutical composition comprising the same in the prepara-tion of a medicament used for the treatment of cancer.
- Embodiment 18 The use of the compound of embodiment 17, wherein the cancer is selected from the group consisting of small cell lung cancer (SCLC) , colorectal cancer (CRC) , pancreatic cancer, and chol-angiocarcinoma, pancreatic cancer, appendiceal cancer, small bowel cancer, colorectal cancer, ampullary cancer, non-small cell lung cancer, cervical cancer, lung cancer, endometrial cancer, acute myeloid leuke-mia, gastrointestinal neuroendocrine tumor, uterine endometrioid carcinoma, oesophagogastric cancer, bladder cancer, ovarian cancer, melanoma, multiple myeloma, thyroid gland adenocarcinoma, a myelodys-plastic syndrome, and squamous cell lung carcinoma, or combinations thereof.
- SCLC small cell lung cancer
- CRC colorectal cancer
- pancreatic cancer pancreatic cancer
- chol-angiocarcinoma pancreatic cancer
- Embodiment 19 The use according to any one of embodiments 17 to 18, wherein the cancer is se-lected from the group consisting of non-small cell lung cancer, small cell lung cancer, colorectal cancer, pancreatic cancer, and cholangiocarcinoma.
- Embodiment 20 The compound according to any one of embodiments 1 to 15, or the stereoisomer thereof, the pharmaceutically acceptable salt thereof, the metabolite thereof, the prodrug thereof, the solv-ate thereof or the solvate of the pharmaceutically acceptable salt thereof, or the pharmaceutical composi-tion comprising the same for use in treating cancer.
- Embodiment 21 The compound for use according to embodiment 20, wherein the cancer is as defined in embodiment 18.
- Embodiment 22 The compound for use according to any one of embodiments 20 to 21, wherein the cancer is as defined in embodiment 19.
- Embodiment 23 A method for treating cancer in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of the compound of any one of the preced-ing embodiments, or the stereoisomer thereof, the pharmaceutically acceptable salt thereof, the metabolite thereof, the prodrug thereof, the solvate thereof or the solvate of the pharmaceutically acceptable salt thereof, or the pharmaceutical composition according to embodiment 16, or the pharmaceutical composi-tion comprising the same.
- Embodiment 24 The method according to embodiment 23, wherein the cancer is as defined in em-bodiment 18.
- Embodiment 25 The method according to any one of embodiments 23 to 24, wherein the cancer is as defined in embodiment 19.
- the term “pharmaceutically acceptable salts” refers to salts prepared from pharmaceu-tically acceptable non-toxic bases or acids.
- the compound of the present invention is acidic, its cor-responding salt can be conveniently prepared from pharmaceutically acceptable non-toxic bases, including inorganic bases and organic bases.
- the compound of the present invention is basic, its corresponding salt can be conveniently prepared from pharmaceutically acceptable non-toxic acids, including inorganic and organic acids.
- solvate refers to solvent addition forms that contain either stoichiometric or non-stoichiometric amounts of solvent.
- excipient that is useful in preparing a pharmaceutical composition that is generally safe, nontoxic and neither biologically nor otherwise undesirable, and includes carrier that is acceptable for veterinary use as well as human pharmaceutical use.
- a “pharmaceutically acceptable carrier” as used herein includes both one and more than one such car-rier.
- the term “pharmaceutically acceptable carrier” also encompasses “pharmaceutically acceptable ex-cipient” and “pharmaceutically acceptable diluent” .
- the particular carrier used in the pharmaceutical compositions of the present disclosure will depend upon the means and purpose for which the compounds of the present disclosure are being applied.
- the term “optionally substituted” refers to unsubtituted or being substituted with one or more (such as 1, 2, 3 or 4) substituents. Suitable substituents for each group can be found herein. Unless otherwise specified, exemplary substituents can be selected from the group consisting of D, halogen, -CN, -NO 2 , R i , -C 1-4 alkylene-R i , -C 2-4 alkenylene-R i , -C 2-4 alkynylene-R i , -OR i , -OC (O) R i , -C (O) R i , -CO 2 R i , -CONR i R ii , -OC (O) NNR i R ii , -NR ii C (O) R i , -NR i -C (O) NR ii R ii , -NR i -C (O) NR
- an optionally substituted group can be further substituted means in addition to being substituted by the above-mentioned substituents for “optionally substituted” , one or more hydrogen atoms in the group may also be substituted by other substituents specified (such as substituents for further substi-tution) .
- alkyl refers to a saturated aliphatic hydrocarbon group including straight chain and branched chain groups having the number of carbon atoms designated (i.e., C 1-20 means 1 to 20 carbon atoms) .
- an alkyl group is an alkyl having 1 to 12 i.e., C 1-12 alkyl. Sometimes preferably 1 to 6 i.e., C 1-6 alkyl, sometimes more preferably 1 to 4, carbon atoms i.e., C 1-4 alkyl.
- Representative exam-ples include, but are not limited to methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1, 1-dimethyl propyl, 1, 2-dimethyl propyl, 2, 2-dimethyl propyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1, 1, 2-trimethylpropyl, 1, 1-dimethylbutyl, 1, 2-dimethylbutyl, 2, 2-dimethylbutyl, 1, 3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2, 3-dimethylbutyl, n-heptyl, 2-methylhexyl, 3-methylhexyl, 4-methylhexyl, 5-methylhex
- an alkyl group is a lower alkyl having 1 to 6 carbon atoms.
- Representative examples include, but are not lim-ited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1, 1-dimethylpropyl, 1, 2-dimethylpropyl, 2, 2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1, 1, 2-trimethylpropyl, 1, 1-dimethylbutyl, 1.2-dimethylbutyl, 2, 2-dimethylbutyl, 1, 3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2, 3-dimethylbutyl, etc.
- the alkyl group can be substituted or unsubstituted.
- the substit-uent group (s) can be substituted at any available connection point, preferably the substituent group (s) is one or more substituents independently selected from the group consisting of alkyl, halogen, alkoxy, alkenyl, alkynyl, alkylsulfo, alkylamino, thiol, hydroxy, nitro, cyano, amino, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxyl, heterocylic, cycloalkylthio, heterocylic alkylthio and oxo group.
- alkylene refers to a saturated linear or branched aliphatic hydrocarbon group, wherein having 2 residues derived by removing two hydrogen atoms from the same carbon atom of the parent al-kane or two different carbon atoms.
- the straight or branched chain group containing 1 to 20 carbon atoms preferably has 1 to 12 carbon atoms, more preferably 1 to 6 carbon atoms.
- Non-limiting examples of al-kylene groups include, but are not limited to, methylene (-CH 2 -) , 1, 1-ethylene (-CH (CH 3 ) -) , 1, 2-ethylene (-CH 2 CH 2 ) -, 1, 1-propylene (-CH (CH 2 CH 3 ) -) , 1, 2-propylene (-CH 2 CH (CH 3 ) -) , 1, 3-propylene (-CH 2 CH 2 CH 2 -) , 1, 4-butylidene (-CH 2 CH 2 CH 2 CH 2 -) etc.
- the alkylene group can be substituted or unsub-stituted.
- the substituent group (s) is preferably one or more, sometimes preferably one to five, sometimes more preferably one to three, group (s) independently selected from the group consisting of selected from alkyl, alkenyl, alkynyl, alkoxy, alkylsulfo, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloalkoxyl, heterocylic alkoxyl, cycloalkylthio and het-erocylic alkylthio.
- alkenyl refers to an alkyl defined as above that has at least two carbon at-oms and at least one carbon-carbon double bond, for example, vinyl, 1-propenyl, 2-propenyl, 1-2-, or 3-butenyl, etc., preferably C 2-20 alkenyl, more preferably C 2-12 alkenyl, and most preferably C 2-6 alkenyl.
- the alkenyl group can be substituted or unsubstituted.
- the substituent group (s) is pref-erably one or more, sometimes preferably one to five, sometimes more preferably one to three, group (s) independently selected from the group consisting of alkyl, halogen, alkoxy, alkenyl, alkynyl, alkylsulfo, alkylamino, thiol, hydroxy, nitro, cyano, amino, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloal-koxyl, heterocylic, cycloalkylthio, heterocylic alkylthio and oxo group.
- alkenylene refers to an alkylene defined as above that has at least two carbon atomsand at least one carbon-carbon double bond, preferably C 2-20 alkenylene, morepreferably C 2-12 alkenylene, and most preferably C 2-6 alkenylene.
- the alkenylene group can be sub-stituted or unsubstituted.
- the substituent group (s) is preferably one or more, sometimes preferably one to five, sometimes more preferably one to three, group (s) independently selected from the group consisting of selected from alkyl, alkenyl, alkynyl, alkoxy, alkylsulfo, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloalkoxyl, heterocylic alkoxyl, cycloalkylthio and heterocylic alkylthio.
- alkynyl refers to an alkyl defined as above that has at least two carbon at-oms and at least one carbon-carbon triple bond, for example, ethynyl, 1-propynyl, 2-propynyl, 1-2-, or 3-butynyl etc., preferably C 2-20 alkynyl, more preferably C 2-12 alkynyl, and most preferably C 2-6 alkynyl.
- the alkynyl group can be substituted or unsubstituted.
- the substituent group (s) is pref-erably one or more, sometimes preferably one to five, sometimes more preferably one to three, group (s) independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkylsulfo, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloalkoxyl, hetero-cylic alkoxyl, cycloalkylthio and heterocylic alkylthio.
- aryl refers to a 6 to 14-membered (or C 6-14 ) all-carbon monocyclic ring or a polycyclic fused ring (a "fused" ring system means that each ring in the system shares an adjacent pair of carbon atoms with another ring in the system) group, and has a completely conjugated pi-electron system.
- aryl is 6 to 10-membered (or C 6-10 ) , such as phenyl and naphthyl, most preferably phenyl.
- the aryl can be fused to the ring of heteroaryl, heterocyclyl or cycloalkyl, wherein the ring bound to parent structure is aryl. Representative examples include, but are not limited to, the following substituents:
- the aryl group can be substituted or unsubstituted.
- the substituent group (s) is pref-erably one or more, sometimes preferably one to five, sometimes more preferably one to three, substituents independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkylsulfo, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloalkoxyl, hetero-cylicalkoxyl, cycloalkylthio and heterocylic alkylthio.
- arylene is a divalent group which is connected to the rest of the molecule through two sin-gle bonds, and the rest definitions are the same as the term “aryl” .
- heteroaryl refers to an aryl system having 1 to 4 heteroatoms selected from the group consisting of O, S and N as ring atoms and having 5 to 14 ring atoms (referred as 5 to 14-membered) .
- a heteroaryl is 5 to 10-membered, more preferably 5 or 6-membered, for exam-ple, thiadiazolyl, pyrazolyl, oxazolyl, oxadiazolyl, imidazolyl, triazolyl, thiazolyl, thiazolylfuryl, thienyl, pyridyl, pyrrolyl, N-alkyl pyrrolyl, pyrimidinyl, pyrazinyl, imidazolyl, tetrazolyl, and the like.
- the het-eroaryl can be fused with the ring of an aryl, heterocyclyl or cycloalkyl, wherein the ring bound to parent structure is heteroaryl.
- Representative examples include, but are not limited to, the following substituents:
- the heteroaryl group can be substituted or unsubstituted.
- the substituent group (s) is preferably one or more, sometimes preferably one to five, sometimes more preferably one to three, sub-stituents independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkylsulfo, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloal-koxyl, heterocylic alkoxyl, cycloalkylthio, heterocylic alkylthio and -NR e R f .
- heteroarylene is a divalent group which is connected to the rest of the molecule through two single bonds, and the rest definitions are the same as the term “heteroaryl” .
- bicyclic is intended to include spiro, fused-ring or bridged-ring.
- spiro refers to two rings that shares one ring atom (e.g., carbon) .
- fused refers to two rings that share two adjacent ring atoms with one an-other.
- bridged refers to two rings that share three adjacent ring atoms with one another.
- cycloalkyl refers to a saturated and/or partially unsaturated monocyclic or polycyclic hydrocarbon group having 3 to 20 carbon atoms (refered as C 3-20 or 3 to 20 membered) , prefera-bly 3 to 12 carbon atoms (refered as C 3-12 or 3 to 12 membered) , more preferably 3 to 10 carbon atoms (refered as C 3-10 or 3 to 10 membered) , and most preferably 3 to 8 carbon atoms (refered as C 3-8 or 3 to 8 membered) or 3 to 6 carbon atoms (refered as C 3-6 or 3 to 6 membered) .
- cycloalkyl is monocyclic cycloalkyl.
- monocyclic cycloalkyls include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cy-cloheptyl, cycloheptatrienyl, cyclooctyl, etc.
- cycloalkyl is polycyclic cycloalkyl (such as bicyclic cycloalkyl) .
- cycloalkyl is a spiro cycloalkyl, a fused cycloalkyl or a bridged cycloalkyl.
- Polycyclic cycloalkyl includes a cycloalkyl having a spiro ring, fused ring or bridged ring.
- the cycloalkyl can be fused to the ring of an aryl, heteroaryl or heterocyclic alkyl, wherein the ring bound to the parent structure is cycloalkyl.
- Representative examples include, but are not limited to inda-nylacetic, tetrahydronaphthalene, benzocycloheptyl and so on.
- the cycloalkyl is optionally substituted or unsubstituted.
- the substituent group (s) is preferably one or more, sometimes preferably one to five, sometimes more preferably one to three, substituents independently selected from the group consisting of alkyl, halogen, alkoxy, alkenyl, alkynyl, alkylsulfo, alkylamino, thiol, hydroxy, nitro, cyano, amino, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloalkoxyl, heterocylic, cyeloalkylthio, hetero-cylic alkylthio and oxo group.
- cycloalkylene is a divalent group which is connected to the rest of the molecule through two single bonds, and the rest definitions are the same as the term “cycloalkyl” .
- “Spiro Cycloalkyl” refers to a 5 to 20-membered polycyclic group with rings connected through one common carbon atom (called a spiro atom) , wherein one or more rings can contain one or more double bonds, but none of the rings has a completely conjugated pi-electron system.
- a spiro cycloalkyl is 6 to 14-membered, and more preferably 7 to 10-membered.
- a spiro cycloalkyl is divided into mono-spiro cycloalkyl, di-spiro cycloalkyl, or poly-spiro cycloal-kyl, and preferably refers to a mono-spiro cycloalkyl or di-spiro cycloalkyl, more preferably 4-membered/4-membered, 4-membered/5-membered, 4-membered/6-membered, 5-membered/5-membered, or 5-membered/6-membered mono-spiro cycloalkyl.
- Representative examples of spiro cycloalkyl include, but are not limited to the following substituents:
- “Fused Cycloalkyl” refers to a 5 to 20-membered polycyclic hydrocarbon group, wherein each ring in the system shares an adjacent pair of carbon atoms with another ring, wherein one or more rings can con-tain one or more double bonds, but none of the rings has a completely conjugated pi-electron system.
- a fused cycloalkyl group is 6 to 14-membered, more preferably 7 to 10-membered.
- fused cycloalkyl is divided into bicyclic, tricyclic, tetracyclic or polycyclic fused cycloalkyl, and preferably refers to a bicyclic or tricyclic fused cycloalkyl, more preferably 5-membered/5-membered, or 5-membered/6-membered bicyclic fused cycloalkyl.
- Representative exam-ples of fused cycloalkyls include, but are not limited to, the following substituents:
- “Bridged Cycloalkyl” refers to a 5 to 20-membered polycyclic hydrocarbon group. wherein every two rings in the system share two disconnected carbon atoms. The rings can have one or more double bonds, but have no completely conjugated pi-electron system.
- a bridged cycloalkyl is 6 to 14-membered, and more preferably 7 to 10-membered.
- bridged cycloalkyl is divided into bicyclic, tricyclic, tetracyclic or polycyclic bridged cycloalkyl, and pref-erably refers to a bicyclic, tricyclic or tetracyclic bridged cycloalkyl, more preferably a bicyclic or tricyclic bridged cycloalkyl.
- Representative examples of bridged cycloalkyls include, but are not limited to, the fol-lowing substituents:
- heterocyclyl or “heterocycloalkyl” refers to a 3 to 20-membered saturated and/or partially unsaturated monocyclic or polycyclic hydrocarbon group having one or more, sometimes preferably one to five, sometimes more preferably one to three carbon ring atoms being replaced with het-eroatoms selected from the group consisting of N, O, and S (O) m (wherein m is 0, l, or 2) as ring atoms, but excluding -O-O-, -O-S-or -S-S-in the ring, the remaining ring atoms being C.
- heterocyclyl is a 3 to 12-membered ring having 1 to 4 heteroatoms (also referred as for example 3-12 membered heterocy-clyl or heterocycloalkyl, or C 2-11 heterocyclyl or heterocycloalkyl) ; more preferably a 4 to 12-membered ring having l to 3 heteroatoms (also referred as for example 4-12 membered heterocyclyl or heterocycloal-kyl, or C 3-10 heterocyclyl or heterocycloalkyl) ; more preferably a 3 to 10-membered ring having 1 to 3 het-eroatoms (also referred as for example 3-10 membered heterocyclyl or heterocycloalkyl, or C 2-9 heterocy-clyl or heterocycloalkyl) ; more preferably a 4 to 8-membered ring having l to 3 heteroatoms (i.e., 4-8 membered heterocyclyl or heterocycloalkyl, or C 2-7 heteroatoms (
- heterocycloalkyl/heterocyclyl is monocyclic heterocycloal-kyl/heterocyclyl.
- Representative examples of monocyclic heterocyclyls or heterocycloalkyl include, but arenot limited to, oxetanyl, azabutyl, pyrrolidyl, piperidyl, piperazinyl, morpholinyl, sulfo-morpholinyl, homopiperazinyl, and so on.
- heterocycloalkyl/heterocyclyl is polycyclic heterocy-cloalkyl/heterocyclyl (such as bicyclic heterocycloalkyl/heterocyclyl) .
- heterocyclo-alkyl/heterocyclyl is a spiro heterocycloalkyl/heterocyclyl, a fused heterocycloalkyl/heterocyclyl or a bridged heterocycloalkyl/heterocyclyl.
- Polycyclic heterocyclyl or heterocycloalkyl includes the heterocy-clyl having a spiro ring, fused ring or bridged ring.
- the “heterocyclyl” or “heterocycloalkyl” can be at-tached to the rest of the molecule via the N or C atom on the ring.
- the “heterocyclyl” or “heterocycloalkyl” is attached to the rest of the molecule via the N atom on the ring. In some embodi-ments, specifically in the definition of R 1 , the “heterocyclyl” or “heterocycloalkyl” is attached to the rest of the molecule via the C atom on the ring. In some embodiments, the “heterocyclyl” or “heterocycloalkyl” has a structure of wherein is a heteroatom or a carbon atom. Examples of such heterocyclyl groups are When the heter-ocyclyl has substituents, the substituents may be attached to any atom in the ring, provided that a stable chemical structure results.
- heterocycloalkylene is a divalent group which is connected to the rest of the molecule by two single bonds, and the rest definitions are the same as the term “heterocycloalkyl” .
- “Spiro heterocyclyl” refers to a 5 to 20-membered polycyclic heterocyclyl with rings connected through one common carbon atom (called a spiro atom) , wherein said rings have one or more, sometimes preferably one to five, sometimes more preferably oneto three, heteroatoms selected from the group con-sisting of N, O, and S (O) m . (wherein m is 0, 1 or 2) as ring atoms, the remaining ring atoms being C, wherein one or more rings can contain one or more double bonds, but none of the rings has a completely conjugated pi-electron system.
- a spiro heterocyclyl is 6 to 14-membered, and more preferably 7 to 10-membered.
- spiro heterocyclyl is divided into mono-spiro heterocyclyl, di-spiro heterocyclyl, or poly-spiro heterocyclyl, and preferably refers to mono-spiro heterocyclyl or di-spiro heterocyclyl, more preferably 4-membered/4-membered, 4-membered/5-membered, 4-membered/6-membered, 5-membered/5-membered, or 5-membered/6-membered mono-spiro heterocyclyl.
- Representative examples of spiro heterocyclyl include, but are not limited to the following substituents:
- “Fused Heterocyclyl” refers to a 5 to 20-membered polycyclic heterocyclyl group, wherein each ring in the system shares an adjacent pair of carbon atoms with the other ring, wherein one or more rings can contain one or more double bonds, but none of the rings has a completely conjugated pi-electron system, and wherein said rings have one or more, sometimes preferably one to five, sometimes more preferably one to three, heteroatoms selected from the group consisting of N, O, and S (O) p , (wherein p is 0, 1, or 2) as ring atoms, the remaining ring atoms being C.
- a fused heterocyclyl is 6 to 14-membered, and more preferably 7 to 10-membered.
- fused heterocyclyl is di-vided into bicyclic, tricyclic, tetracyclic or polycyclic fused heterocyclyl, preferably refers to bicyclic or tricyclic fused heterocyclyl, more preferably 5-membered/5-membered, or 5-membered/6-membered bicy-clic fused heterocyclyl.
- fused heterocyclyl include, but are not limited to, the following substituents:
- the ring of said heterocyclyl can be fused to the ring of an aryl, heteroaryl orcycloal-kyl, wherein the ringbound to the parent structure is heterocyclyl.
- Representative examples include, but are not limited to the following substituents:
- the heterocyclyl is optionally substituted or unsubstituted.
- the substituent group (s) is preferably one or more, sometimes preferably one to five, sometimes more preferably one to three, group (s) independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkylsulfo, alkylamino, halogenthiol, hydroxy, nitro, cyano, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloal-koxyl, heterocylic alkoxyl, cycloalkylthio, heterocylic alkylthio.
- “Bridged Heterocyclyl” refers to a 5 to l4-membered polycyclic heterocyclic alkyl group, wherein every two rings in the system share two disconnected atoms, the rings can have one or more double bonds, but have no completely conjugated pi-electron system, and the rings have one or more heteroatoms select-ed from the group consisting of N, O, and S (O) m (wherein m is 0, 1, or 2) as ring atoms, the remaining ring atoms being C.
- a bridged heterocyclyl is 6 to 14-membered, and more preferably 7 to 10-membered.
- bridged heterocyclyl is divided into bicyclic, tricyclic, tetracyclic or polycyclic bridged heterocyclyl, and preferably refers to bicyclic, tricyclic or tetra-cyclic bridged heterocyclyl, more preferably bicyclic or tricyclic bridged heterocyclyl.
- Representative ex-amples of bridged heterocyclyl include, but are not limited to, the following substituents:
- C 1-6 haloalkoxyl refers to an alkoxyl group in which one or more hydrogen atoms are replaced by a halogen, for example, -OCF 3 .
- haloalkyl refers to an alkyl group in which one or more hydrogen atoms are replaced by a halogen, for example, -CF 3 .
- C 1-6 alkoxyC 1-6 alkyl refers to a C 1-6 alkyl group in which one or more hy-drogen atoms are replaced by C 1-6 alkoxy or a C 1-6 alkoxy group in which one or more hydrogen atoms are replaced by C 1-6 alkyl.
- alkoxy refers to a straight or branched alkoxy group containing the speci-fied number of carbon atoms.
- C 1-6 alkoxy means a straight or branched alkoxy group con-taining at least 1, and at most 6, carbon atoms.
- alkoxy as used herein include, but not lim-ited to, methoxy, ethoxy, prop-1-oxy, pro-2-oxy, pentoxy, hexyloxy, and the like.
- aminoalkyl refers to an alkyl moiety substituted by one or more amino moieties. Such as -CH 2 (NH 2 ) .
- alkylamino refers to an amino group substituted by one or more alkyl such as -NH (C 1-6 alkyl) or -N (C 1-6 alkyl) 2 .
- alkylamino include, but not limited to, -NH (CH 3 ) , -N (CH 3 ) 2 .
- halogen refers to fluorine (F) , chlorine (Cl) , bromine (Br) or io-dine (I) .
- the term “Bond” refers to a covalent bond using a sign of “-” . In some embodi-ments, represents a double bond or single bond.
- the compounds of the present invention can be prepared by any conventional means. Suitable pro-cesses for synthesizing these compounds as well as their starting materials are provided in the schemes be-low and in the examples. All substituents 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.
- Scheme 1 illustrates the preparation of compound of formula (II) .
- the coupling of compounds of formula (1) can be obtained through standard Suzuki coupling conditions (for example Pd (dppf) Cl 2 and K 2 CO 3 ) to provide compound of formula (2) .
- Compound of formula (2) was coupled with compound of formula (3) under Suzuki coupling conditions (for example Pd (dppf) Cl 2 and K 2 CO 3 ) to give compound of formula (4) , which was converted to compound of formula (5) in the presence of iodization reagents, such I 2 or NIS.
- Deprotection of compound of formula (5) to provide compound of formula (6) such as hydroly-sis of the ester with LiOH ⁇ OH, or with TBAF deprotecting silicon protective group on hydroxyl group.
- Compound of formula (8) was attained by a routine coupling condition between compound of formula (6) and amine of formula (7) with coupling reagents, such as HATU, EDCI/HOBt or PyBOP etc, in the pres-ence of an organic base, such as Et 3 N, DIPEA or pyridine, which was hydrolyzed to provide acid of for-mula (9) under a base, such as LiOH ⁇ OH.
- an organic base such as Et 3 N, DIPEA or pyridine
- Compound of formula (II) can be obtained by a coupling reaction between acid of formula (16) and compound of for-mula (15) with coupling reagents, such as HATU, EDCI/HOBy, PyBOP or COMU, in the presence of a base, such as TEA, DIPEA, pyridine or 2, 6-lutidine.
- coupling reagents such as HATU, EDCI/HOBy, PyBOP or COMU
- reaction mixture was diluted with NaHCO 3 solution (100 mL) , then the resulting mixture was extract-ed with DCM (100 mL x 4) , the combined organic phase was washed with saturated NaCl (50 mL) , then died over with Na 2 SO 4 , following with concentration under reduced pressure to obtain crude one, which was purified by silica gel column eluting with petroleum ether/EtOAc (from 0%to 20%) to afford 3- (5-bromo-1H-indol-3-yl) -2, 2-dimethylpropyl acetate (82.0 g, 79%) as a white solid.
- reaction solution was diluted with H 2 O and extracted with DCM, the combined organic phase was washed with H 2 O dried over Na 2 SO 4 , following with concentration under reduced pressure to obtain crude product which was purified by silica gel column (eluting with EA/PE from 0%to 20%) to give (S) -4-benzyl-3- (2- (3-oxocyclobutyl) acetyl) oxazolidin-2-one (68 g 58.3 %) as a yellow oil.
- the reac-tion mixture was stirred at -78°C for 30 mins, then 1, 3-Dimethyl-3, 4, 5, 6-tetrahydro-2 (1H) -pyrimidinone (310 mL, 2565.0 mmol) was added dropwise at -78°C.
- the reaction mixture was stirred at r.t. for 14 h. Af-ter completion, the reaction solution was quenched with water (200 mL) , then LiOH ⁇ H 2 O (10.8 g, 256.5 mmol) was added and the reaction solution was stirred at r.t. for 2 h.
- the resulting mixture was extracted with EtOAc, washed with water and brine, then dried over Na 2 SO 4 , following with concentration under reduced pressure to obtain crude product (S) -2, 3-bis (tert-butoxycarbonyl) -2, 3-diazabicyclo [3.1.1] heptane-4-carboxylic acid, which was used directly for the next step.
- reaction solution was concentration under reduced pressure to obtain crude product which was purified by silica gel column (eluting with EA/PE from 0%to 23%) to give 2, 3-di-tert-butyl 4-methyl (S) -2, 3-diazabicyclo [3.1.1] heptane-2, 3, 4-tricarboxylate (5.6 g, 18.45 %in two-steps) as a light yellow oil.
- reaction solution was concentrated under reduced pressure to remove 1, 4-dioxane, the re-sidual was diluted with H 2 O and extracted with EtOAc (500 mL x 3) , the combined organic phase was washed with saturated NaCl (500 mL) , then dried over Na 2 SO 4 , following with concentration under re-duced pressure to obtain crude product which was purified by silica gel column eluting with EtOAc/PE from 0%to 27%to afford 2, 2-dimethyl-3- (5- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -1H-indol-3-yl) propyl acetate (18.7 g, 81.4 %) as a light yellow solid.
- Step 7 Preparation of tert-butyl ( (6 4 S, 4S, Z) -1 2 -iodo-10, 10-dimethyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
- Step 8 Preparation of tert-butyl ( (6 4 S, 4S, Z) -10, 10-dimethyl-5, 7-dioxo-1 2 - (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
- reaction mixture was diluted with EtOAc (500 mL) , washed with saturated NH 4 Cl (300 x 3 mL) , then dried over Na 2 SO 4 , following with concentration under reduced pressure to obtain crude product which was purified by silica gel column (eluting with EtOAc/PE from 0%to 25%) to give (S) -1- (3-bromopyridin-2-yl) ethan-1-ol (28.80 g, 95.0 %) as a yellow oil.
- the raw materials and reagents used in the following embodiments are com-mercially available or may be prepared by known methods.
- Step 1 Synthesis of tert-butyl (S) -2-ethynylpyrrolidine-1-carboxylate
- reaction solution was concentrated under reduced pressure to remove MeOH, the residual was diluted with H 2 O and extracted with EtOAc, the combined organic phase was dried over Na 2 SO 4 , following with concentration under reduced pressure to obtain crude product which was purified by silica gel column (eluting with EA/PE from 0%to 20%) to give tert-butyl (S) -2-ethynylpyrrolidine-1-carboxylate (1.7 g, 86.7 %) as a colourless oil.
- Step 2 Synthesis of tert-butyl (S) -2- ( (5-bromo-6- ( (S) -1-methoxyethyl) pyridin-3-yl) ethynyl) pyrrolidine-1-carboxylate
- Step 3 Synthesis of 3-bromo-2- ( (S) -1-methoxyethyl) -5- ( ( (S) -pyrrolidin-2-yl) ethynyl) pyridine
- Step 4 Synthesis of 3-bromo-2- ( (S) -1-methoxyethyl) -5- ( ( (S) -1-methylpyrrolidin-2-yl) ethynyl) pyridine
- reaction mixture was diluted with EtOAc (50 mL) , washed with brine (30 mL) , then dried over Na 2 SO 4 , following with concen-tration under reduced pressure to obtain crude product tert-butyl (S) -3-ethynylpyrrolidine-1-carboxylate (980 mg) as a yellow oil.
- Step 1 Synthesis of 1-methyl-4- ( (trimethylsilyl) ethynyl) -1H-pyrazole
- reaction mixture was concen-trated to give the crude product which was purified by silica gel column (eluting with EtOAc/PE from 0%to 50%) to give 1-methyl-4- ( (trimethylsilyl) ethynyl) -1H-pyrazole (500 mg, 58.4%) as a brown solid.
- Step 3 Synthesis of (S) -3-bromo-2- (1-methoxyethyl) -5- ( (1-methyl-1H-pyrazol-4-yl) ethynyl) pyridine
- Step 1 Synthesis of (S) -3- ( (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) ethynyl) imidazo [1, 2-b] pyridazine
- Step 1 Synthesis of (S) -3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) -N, N-dimethylprop-2-yn-1-amine (2)
- Step 1 Synthesis of 3-bromo-5- (3- ( (R) -3-fluoropyrrolidin-1-yl) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridine
- Step 1 Preparation of (S) -3-bromo-2- (1-methoxyethyl) -5- (3- (piperidin-1-yl) prop-1-yn-1-yl) pyridine
- Step 1 Preparation of (S) -1- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) -4-methylpiperazine
- Step 1 Synthesis of (S) -1- (4- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) piperazin-1-yl) ethan-1-one
- Step 1 Synthesis of ( (R) -4- (3- (5-bromo-6- ( (S) -1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) morpholin-3-yl) methanol
- Step 1 Preparation of ( (S) -4- (3- (5-bromo-6- ( (S) -1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) morpholin-3-yl) methanol
- Step 1 Preparation of tert-butyl (S) -4- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) piperazine-1-carboxylate
- Step 2 Preparation of (S) -1- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) piperazine
- Step 3 Preparation of (S) -1- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) -4- (methylsulfonyl) piperazine
- Step 1 Synthesis of (S) -5-bromo-6- (1-methoxyethyl) pyridin-3-ol
- Step 2 Synthesis of tert-butyl ( (6 4 S, 4S, Z) -1 2 - (5-hydroxy-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
- Step 3 Synthesis of 5- ( (6 4 S, 4S, Z) -4- ( (tert-butoxycarbonyl) amino) -10, 10-dimethyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiaz ola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-1 2 -yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl trifluoromethanesulfonate
- Step 4 Synthesis of tert-butyl ( (6 4 S, 4S, Z) -1 2 - (2- ( (S) -1-methoxyethyl) -5- ( (trimethylsilyl) ethynyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
- Step 5 Synthesis of tert-butyl ( (6 4 S, 4S, Z) -1 1 -ethyl-1 2 - (5-ethynyl-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
- Step 6 Synthesis of (6 4 S, 4S, Z) -4-amino-1 1 -ethyl-1 2 - (5-ethynyl-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione To a stirred solution of tert-butyl ( (6 4 S, 4S, Z) -1 1 -ethyl-1 2 - (5-ethynyl-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4
- Step 7 Synthesis of (1S, 2S) -N- ( (64S, 4S, Z) -11-ethyl-12- (5-ethynyl-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2-methylcyclopropane-1-carboxamide
- Step 1 Preparation of tert-butyl ( (6 4 S, 4S, Z) -1 2 - (5- (3-hydroxy-3-methylbut-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
- Step 2 Preparation of tert-butyl ( (6 4 S, 4S, Z) -1 1 -ethyl-1 2 - (5- (3-hydroxy-3-methylbut-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dime-thyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
- Step 3 Preparation of (6 4 S, 4S, Z) -4-amino-1 1 -ethyl-1 2 - (5- (3-hydroxy-3-methylbut-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione
- Step 4 Preparation of (1r, 2R, 3S) -N- ( (6 4 S, 4S, Z) -1 1 -ethyl-1 2 - (5- (3-hydroxy-3-methylbut-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
- Step 1 Synthesis of tert-butyl ( (6 4 S, 4S, Z) -1 2 - (2- ( (S) -1-methoxyethyl) -5- ( ( (S) -1-methylpyrrolidin-2-yl) ethynyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
- Step 2 Synthesis of tert-butyl ( (6 4 S, 4S, Z) -1 1 -ethyl-1 2 - (2- ( (S) -1-methoxyethyl) -5- ( ( (S) -1-methylpyrrolidin-2-yl) ethynyl) pyridin-3-yl) -10, 10 -dimethyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
- Step 3 Synthesis of (6 4 S, 4S, Z) -4-amino-1 1 -ethyl-1 2 - (2- ( (S) -1-methoxyethyl) -5- ( ( (S) -1-methylpyrrolidin-2-yl) ethynyl) pyridin-3-yl) -10, 10-dimethyl-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacyclou ndecaphane-5, 7-dione
- Step 4 Synthesis of (1r, 2R, 3S) -N- ( (6 4 S, 4S, Z) -1 1 -ethyl-1 2 - (2- ( (S) -1-methoxyethyl) -5- ( ( (S) -1-methylpyrrolidin-2-yl) ethynyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
- Step 1 Preparation of (6 4 S, 4S, Z) -4-amino-10, 10-dimethyl-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione
- Step 2 Preparation of (1r, 2R, 3S) -N- ( (6 4 S, 4S, Z) -10, 10-dimethyl-5, 7-dioxo-1 2 - (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
- Step 3 Preparation of tert-butyl (3S) -3- ( (5- ( (6 4 S, 4S, Z) -4- ( (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxamido) -10, 10-dimethyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-1 2 -yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl) ethynyl) pyrrolidine-1-carboxylate
- Step 4 Preparation of tert-butyl (3S) -3- ( (5- ( (6 4 S, 4S, Z) -4- ( (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxamido) -1 1 -ethyl-10, 10-dimethyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-1 2 -yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl) ethynyl) pyrrolidine-1-carboxylate
- Step 5 Preparation of (1r, 2R, 3S) -N- ( (6 4 S, 4S, Z) -1 1 -ethyl-1 2 - (2- ( (S) -1-methoxyethyl) -5- ( ( (S) -pyrrolidin-3-yl) ethynyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptan acycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
- Step 6 Preparation of (1r, 2R, 3S) -N- ( (6 4 S, 4S, Z) -1 1 -ethyl-1 2 - (2- ( (S) -1-methoxyethyl) -5- ( ( (S) -1-methylpyrrolidin-3-yl) ethynyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1 ] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
- Step 1 Synthesis of (1S, 2S) -N- ( (6 4 S, 4S, Z) -1 2 - (2- ( (S) -1-methoxyethyl) -5- ( (1-methyl-1H-pyrazol-4-yl) ethynyl) pyridin-3-yl) -10, 10 -dimethyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2-methylcyclopropane-1-carboxamide
- Step 2 Synthesis of (1S, 2S) -N- ( (6 4 S, 4S, Z) -1 1 -ethyl-1 2 - (2- ( (S) -1-methoxyethyl) -5- ( (1-methyl-1H-pyrazol-4-yl) ethynyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2-methylcyclopropane-1-carboxamide
- Step 1 Synthesis of (6 4 S, 4S, Z) -4-amino-10, 10-dimethyl-1 2 - (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione
- the oaranic layer was separated, dried over anhydrous Na 2 SO 4 and concentrated to give (6 4 S, 4S, Z) -4-amino-10, 10-dimethyl-1 2 - (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione (777 mg) as a grey solid.
- Step 2 Synthesis of (1S, 2S) -N- ( (6 4 S, 4S, Z) -10, 10-dimethyl-5, 7-dioxo-1 2 - (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2-methyl cyclopropane-1-carboxamide
- Step 3 Synthesis of (1S, 2S) -N- ( (6 4 S, 4S, Z) -1 2 - (5- (imidazo [1, 2-b] pyridazin-3-ylethynyl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacy cloundecaphane-4-yl) -2-methylcyclopropane-1-carboxamide
- Step 4 Synthesis of (1S, 2S) -N- ( (6 4 S, 4S, Z) -1 1 -ethyl-1 2 - (5- (imidazo [1, 2-b] pyridazin-3-ylethynyl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2-methylcyclopropane-1-carboxamide
- Step 1 Synthesis of (1r, 2R, 3S) -N- ( (6 4 S, 4S, Z) -1 2 - (5- (3- (dimethylamino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 1 0-dimethyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
- Step 2 Synthesis of (1r, 2R, 3S) -N- ( (6 4 S, 4S, Z) -1 2 - ( (R) -5- (3- (dimethylamino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) -3, 4-dihydropyridin-3-yl) -1 1 -ethyl-10, 10-dimethyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
- Step 1 Preparation of (1r, 2R, 3S) -N- ( (6 4 S, 4S, Z) -1 2 - (2- ( (S) -1-methoxyethyl) -5- (3- (piperidin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 1 0-dimethyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
- Step 2 Preparation of (1r, 2R, 3S) -N- ( (6 4 S, 4S, Z) -1 1 -ethyl-1 2 - (2- ( (S) -1-methoxyethyl) -5- (3- (piperidin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
- Step 1 Preparation of tert-butyl ( (6 4 S, 4S, Z) -1 2 - (2- ( (S) -1-methoxyethyl) -5- (3- (4-methylpiperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dime-thyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
- Step 2 Preparation of tert-butyl ( (6 4 S, 4S, Z) -1 1 -ethyl-1 2 - (2- ( (S) -1-methoxyethyl) -5- (3- (4-methylpiperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
- Step 3 Preparation of (6 4 S, 4S, Z) -4-amino-1 1 -ethyl-1 2 - (2- ( (S) -1-methoxyethyl) -5- (3- (4-methylpiperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione
- Step 4 Preparation of (1r, 2R, 3S) -N- ( (6 4 S, 4S, Z) -1 1 -ethyl-1 2 - (2- ( (S) -1-methoxyethyl) -5- (3- (4-methylpiperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
- Step 1 Preparation of tert-butyl ( (6 4 S, 4S, Z) -1 2 - (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
- Step 2 Preparation of tert-butyl ( (6 4 S, 4S, Z) -1 1 -ethyl-1 2 - (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimeth yl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
- reaction was monitored by LCMS. After completion, the reaction mixture was diluted with EtOAc (100 mL) , washed with water (50 mL x 3) and saturated NaCl (50 x 2 mL) , then dried over Na 2 SO 4 , following with concen-tration under reduced pressure to obtain crude product which was purified by pre-HPLC (eluting with CH 3 CN/H 2 O (0.1%NH 4 HCO 3 ) from 20%to 80%) to give tert-butyl ( (6 4 S, 4S, Z) -1 1 -ethyl-1 2 - (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimet hyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -in
- Step 3 Preparation of (6 4 S, 4S, Z) -4-amino-1 1 -ethyl-1 2 - (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10 -dimethyl-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione
- reaction mixture was concen-trated and lyophilized to give (6 4 S, 4S, Z) -4-amino-1 1 -ethyl-1 2 - (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundeca phane-5, 7-dione (110 mg) of its TFA salt as a yellow solid.
- Step 4 Preparation of (1r, 2R, 3S) -N- ( (6 4 S, 4S, Z) -1 1 -ethyl-1 2 - (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptan acycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
- the reaction mixture was concentrated to give the crude product.
- the crude product was purified by pre-HPLC (ACN/H 2 O (0.5 %NH 4 HCO 3 ) from 20%to 95%in 30 min) to give (1r, 2R, 3S) -N- ( (6 4 S, 4S, Z) -1 1 -ethyl-1 2 - (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl ) -10, 10-dimethyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide (85 mg, 61.7%) as a white solid.
- Step 1 Preparation of tert-butyl ( (6 4 S, 4S, Z) -1 2 - (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10 -dimethyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
- Step 2 tert-butyl ( (6 4 S, 4S, Z) -1 2 - (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -1 1 -ethyl-10, 10-dimethyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
- Step 3 (6 4 S, 4S, Z) -4-amino-1 2 - (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -1 1 -ethyl-10, 10-dimethyl-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] hepta nacycloundecaphane-5, 7-dione
- Step 4 (1S, 2S) -N- ( (6 4 S, 4S, Z) -1 2 - (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -1 1 -ethyl-10, 10-dimethyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2-methylcyclopropane-1-carboxamide
- Step 1 Synthesis of tert-butyl ( (6 4 S, 4S, Z) -1 2 - (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10 -dimethyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
- Step 2 Synthesis of tert-butyl ( (6 4 S, 4S, Z) -1 2 - (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -1 1 -ethyl-10, 10-dimethyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
- Step 3 Synthesis of (6 4 S, 4S, Z) -4-amino-1 2 - (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -1 1 -ethyl-10, 10-dimethyl-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione
- reaction mixture was concen-trated to give crude (6 4 S, 4S, Z) -4-amino-1 2 - (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -1 1 -ethyl-10, 10-dimethyl-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione which was used to next step without purification.
- Step 4 Synthesis of (1r, 2R, 3S) -N- ( (6 4 S, 4S, Z) -1 2 - ( (R) -5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) -3, 4-dihydropyridin-3-yl) -1 1 -ethyl-10, 10-dimethyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
- Step 1 Synthesis of tert-butyl ( (6 4 S, 4S, Z) -1 2 - (5- (3- ( (R) -3- (hydroxymethyl) morpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
- Step 2 Synthesis of tert-butyl ( (6 4 S, 4S, Z) -1 1 -ethyl-1 2 - (5- (3- ( (R) -3- (hydroxymethyl) morpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyrid-in-3-yl) -10, 10-dimethyl-5, 7-dioxo-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1 ] heptanacycloundecaphane-4-yl) carbamate
- Step 3 Synthesis of (6 4 S, 4S, Z) -4-amino-1 1 -ethyl-1 2 - (5- (3- ( (R) -3- (hydroxymethyl) morpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-1 1 H-8-oxa-6 2 , 6 3 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione
Landscapes
- Chemical & Material Sciences (AREA)
- Health & Medical Sciences (AREA)
- Organic Chemistry (AREA)
- Animal Behavior & Ethology (AREA)
- Life Sciences & Earth Sciences (AREA)
- Pharmacology & Pharmacy (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Medicinal Chemistry (AREA)
- Epidemiology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
Related to macrocyclic compounds and therapeutic uses thereof. Disclosed a compound of formula (I), or a pharmaceutically acceptable salt thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt, and their use for the preparation of a medicament of treatment of cancer. Provided herein are useful compounds for the inhibition of RAS mutant and the compound shows good inhibitory activity against RAS.
Description
The present invention belongs to the field of medicine, and specifically, relates to macrocyclic com-pounds with stereoisomers, pharmaceutically acceptable salts, solvates, eutectic or deuterated compounds, and their used in the treatment of RAS protein-mediated related diseases
The RAS protein (including KRAS4A, KRAS4B, HRAS, and NRAS) is a small, membrane-bound guanine nucleotide-binding protein that acts as a molecular switch by cycling between an active GTP-bound state and an inactive GDP-bound state. When RAS binds to GEF family proteins such as SOS1, its conformation changes, reducing its affinity for GDP and causing its dissociation from GDP and binding to GTP in the high concentration in the environment, thus becoming active. In normal cells, activation of receptor tyrosine kinases such as epidermal growth factor receptor (EGFR) promotes the exchange of GDP and GTP in RAS, activating RAS, which plays a key role in regulating cell survival, proliferation, and dif-ferentiation through multiple cascading reactions, including the PI3K-AKT-mTOR, RAF-MEK-ERK, and RALGDS-RAL pathways. Mutations in RAS are often driving forces for carcinogenesis and tumor devel-opment, as they disrupt the guanine exchange cycle and lock RAS in the activated state, continuously acti-vating downstream signaling pathways and leading to the development of tumors.
RAS mutations occur in approximately 19%of all cancers and play an important role in tumor occur-rence and progression. Among them, KRAS is the most commonly mutated subtype, followed by NRAS and HRAS. KRAS mutations commonly occur in pancreatic ductal adenocarcinoma, lung adenocarcinoma, and colorectal adenocarcinoma, while NRAS mutations have a relatively higher frequency in hematologic malignancies such as chronic myeloid leukemia and acute myeloid leukemia, as well as in malignant mel-anoma, thyroid cancer, and laryngeal cancer. HRAS mutations are less common in human cancers, but have a relatively higher mutation rate in head and neck squamous cell carcinoma, bladder cancer, salivary gland carcinoma, and oral cancer.
In May 2021, the FDA approved AMG510 of Amgen, making it the first approved KRAS-targeted drug of the world for the treatment of patients with locally advanced or advanced non-small cell lung can-cer harboring mutations in the KRAS G12C gene, this also marked a major breakthrough in the treatment of KRAS G12C mutations. In December 2022, MRTX849 of Mirati therapeutics was also successfully ap-proved for marketing in United States, which was the second marketed oral inhibitor in the world that spe-cifically optimizes the KRAS G12C mutant. The approval of these two drugs marked a successful break-through in KRAS, the most difficult druggable target in history, and has become a milestone achievement. However, in the current clinical research, mutations of various subtypes of RAS protein (KRAS, NRAS and HRAS) was reflected in pan-tumor species, and the treatment of a single mutation could not improve the objective remission rate of patients. At present, there are no highly effective inhibitors to meet various clinical needs, and the emergence of RAS molecular glue is expected to successfully solve this problem.
The RAS molecular glue can simultaneously bind to highly activated RAS (RAS-ON) proteins and a ubiquitously expressed protein in cells, forming a tricomplex to occupy binding sites for RAS interacting proteins such as SOS1 and RAF1, keeping RAS protein in a locked state alone, unable to activate down-stream signaling pathways, thereby blocking RAS signaling pathway transmission and inhibiting tumor growth. Therefore, there is an urgent clinical need to develop an inhibitor of RAS molecular glue type with good activity and high druggability.
Provided herein are Ras inhibitors as molecular glues. The approach described herein entails for-mation of a high affinity three-component complex between a synthetic ligand and two intracellular pro-teins which do not interact under normal physiological conditions: The target of interest (e.g. Ras) , and a widely expressed cytosolic chaperone in cells (e.g. cyclophilin A) . More specifically, in some embodiments, the Ras molecular glues described herein induce a new binding pocket in Ras by forming a three-component complex with high-affinity between the Ras protein and the cytosolic chaperone, cyclo-philin A. Without being bound by theory, the inventors believe that one way the inhibitory effect on Ras is effected by compounds of the invention and the three-component complexes they form is by steric occlu-sion of the interaction site between Ras and downstream effector molecules, such as RAF and PI3K, which are required for propagating the oncogenic signal.
In the first aspect, the present invention provides a compound, or a stereoisomer thereof, a pharma-ceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt thereof, wherein the compound is a compound of formula (I) ;
wherein
n is 0, 1, 2, or 3;
A is optionally substituted 5 to 6-membered heterocycloalkylene, optionally substituted 5 to 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene;
B is selected from optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 3 to 6-membered heterocycloalkenylene, optionally substituted 4 to 11-membered bicyclic cycloalkylene, or optionally substituted 4 to 11-membered bicyclic heterocycloalkylene;
G is optionally substituted C1-6 alkylene;
R8 and R9 are each independently selected from hydrogen, deuterium, halogen, hydroxy, cyano, op-tionally substituted C1-3 alkyl, or R8 and R9 combine with the atoms to which they are attached to form an optionally substituted C3-6 cycloalkyl or a carbonyl;
R10 and R11 are each independently selected from hydrogen, deuterium, halogen, hydroxy, cyano, op-tionally substituted C1-3 alkyl, or R10 and R11 combine with the atoms to which they are attached to form an optionally substituted C3-6 cycloalkyl or a carbonyl;
R12 and R13 are each independently selected from hydrogen, deuterium, halogen, hydroxy, optionally substituted C1-3 alkyl, -O (C1-6 alkyl) , -S (C1-6 alkyl) or -N (C1-6 alkyl) (C1-6 alkyl) ; furthermore, the C1-6 alkyl may be further optionally substituted;
RA is optionally substituted C3-6 cycloalkyl, optionally substituted C3-6 heterocycloalkyl (or 4 to 7-membered heterocycloalkyl) , optionally substituted 5 to 8-membered aryl, optionally substituted 5 to 8-membered heteroaryl, optionally substituted 8 to 10-membered fused bicyclic aryl or optionally substi-tuted 8 to 10-membered fused bicyclic heteroaryl;
RB is selected from hydrogen, C1-6 alkyl, C1-6 alkoxyl, C1-6 haloalkyl, C1-6 haloalkoxyl, optionally sub-stituted C3-6 cycloalkyl, or optionally substituted C3-6 heterocycloalkyl (or 4 to 7-membered heterocycloal-kyl) ; or provided that when n exceeds 1, any two of RB combine with the atoms to which they are attached to form a 3 to 6-membered ring, wherein the ring can be optionally substituted with halogen, hydroxy, and C1-3 alkyl;
X1 is N or C;
X2 is N or -CRa-;
X3 is N or -CRb-;
X4 is N or -CRc-;
X5 is N or C;
X6 is S, O, N, or -CH-;
Ra, Rb, and Rc are each independently hydrogen, halogen, cyano, C1-3 alkyl, C1-3 alkoxyl, C1-3 haloal-kyl, C1-3 haloalkoxyl, C3-6 cycloalkyl, or C3-6 heterocycloalkyl (or 4 to 7-membered heterocycloalkyl) ;
R3 is absent, or selected from hydrogen, C1-6 alkyl, or C1-6 haloalkyl, provided that when X6 is O or S, R3 is absent;
E is a bond, or selected from N, or -CRd-, wherein Rd is selected from hydrogen, halogen, hydroxy, cyano, carboxyl, optionally substituted C1-6 alkyl, optionally substituted C1-6 alkoxyl, C1-6 aminoalkyl, C1-6 hydroxyalkyl, or -NReRf; wherein Re and Rf are independently selected from hydrogen, or optionally sub-stituted C1-6 alkyl;
R4 is hydrogen, optionally substituted C1-6 alkyl, optionally substituted C1-6 heteroalkyl, optionally substituted C3-10 cycloalkyl, optionally substituted C3-10 cycloalkenyl, optionally substituted C3-10 heterocy-cloalkyl (or 4 to 12-membered heterocycloalkyl) , optionally substituted 6 to 10-membered aryl, optionally substituted 5 to 10-membered heteroaryl, or
R5 is hydrogen, C1-6 alkyl or C3-6 cycloalkyl, furthermore, the C1-6 alkyl or C3-6 cycloalkyl may be fur-ther optionally substituted;
or R4 and R5 combine with the atoms to which they are attached to form a ring, wherein the ring is se-lected from optionally substituted 3 to 10-membered cycloalkyl, optionally substituted 3 to 10-membered heterocycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 4 to 11-membered bicyclic cycloalkyl, optionally substituted 4 to 11-membered bicyclic heterocycloalkyl;
wherein
L is absent, or selected from -CH2-, -C (O) -, -CHRg-or -C (Rg) 2-, wherein Rg is optionally substituted C1-6 alkyl;
R6 is hydrogen, or optionally substituted C1-6 alkyl;
R7 is optionally substituted C1-6 alkyl, optionally substituted C1-6 heteroalkyl, optionally substituted C3-10 cycloalkyl, optionally substituted C3-10 heterocycloalkyl (or 3 to 10-membered heterocycloalkyl) , op-tionally substituted 6 to 10-membered aryl, optionally substituted 5 to 10-membered heteroaryl;
or L, R6 and R7 combine with the atoms to which they are attached to form a ring, wherein the ring is selected from optionally substituted 3 to 10-membered cycloalkyl, optionally substituted 3 to 10-membered heterocycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 4 to 11-membered bicyclic cycloalkyl, optionally substituted 4 to 11-membered bicyclic heterocycloalkyl.
In the second aspect, the present invention provides a pharmaceutical composition comprising the compound according to the first aspect the stereoisomer thereof, the pharmaceutically acceptable salt thereof, the metabolite thereof, the prodrug thereof, the solvate thereof or the solvate of the pharmaceuti-cally acceptable salt thereof, and a pharmaceutically acceptable carrier.
In the third aspect, the present invention provides a use of the compound according to the first aspect the stereoisomer thereof, the pharmaceutically acceptable salt thereof, the metabolite thereof, the prodrug thereof, the solvate thereof or the solvate of the pharmaceutically acceptable salt thereof in the preparation of a medicament used for the treatment of cancer.
DESCRIPTION OF THE DRAWINGS
Fig. 1 The reference compound and the representative Compound A significantly inhibited the interac-tion of RAF1 with active RASMutant protein or RASWT protein.
Fig. 2 The reference compound and the representative Compound A significantly inhibited the cell viability of KRAS mutant cells.
Fig. 3 Tumor volume changes and tumor growth inhibition ratio (TGI) after administrating vehicle or test articles to female Balb/c nude mice bearing HPAC PDAC tumors. Data endpoints represent mean ±SEM. p.o.: oral administration; QD: once a day; mpk: mg/kg.
Fig. 4 Tumor volume changes and tumor growth inhibition ratio (TGI) after administrating vehicle or test articles to female Balb/c nude mice bearing PK59 PDAC tumors. Data endpoints represent mean ±SEM. p.o.: oral administration; QD: once a day; mpk: mg/kg.
Fig. 5 Tumor volume changes and tumor growth inhibition ratio (TGI) after administrating vehicle or test articles to male Balb/c nude mice bearing HPAC PDAC tumors. Data endpoints represent mean ± SEM. p.o.: oral administration; QD: once a day; mpk: mg/kg.
Fig. 6 Tumor volume changes and tumor growth inhibition ratio (TGI) after administrating vehicle or test articles to male Balb/c nude mice bearing LU99 NSCLC tumors. Data endpoints represent mean ±SEM. p.o.: oral administration; QD: once a day; mpk: mg/kg.
Fig. 7 and Fig. 8 Bioluminescence signal changes tumor growth inhibition ratio (TGI) after adminis-trating vehicle or test articles to female BALB/c nude mice bearing NCI-H1373-luc intracranial tumors. Data endpoints represent mean ± SEM.
EMBODIMENTS FOR CARRYING OUT THE INVENTION
After extensive and in-depth research, the inventors have unexpectedly developed a kind of novel compounds that can effectively inhibit the interaction of RAF1 with active RASMutant protein or RASWT protein and proliferation of KRAS-related tumor cells. In addition, some compounds of the present inven-tion also showed significantly improved activity than the positive control compounds (such as compound Ref. 1) in animal model studies, even though the compounds of the present invention showed similar ac-tivities to in the positive control compounds in vitro studies. The present invention is completed on this basis.
In one aspect, the present invention provides a compound of formula (I) :
or a pharmaceutically acceptable salt thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt, wherein the variables are as described above.
In some embodiments, R8, R9, R10, R11, R12 and R13 is H.
In some embodiments, the compound of formula (I) is a compound of formula (I-1)
wherein X1, X2, X3, X4, X5, X6, A, B, E, G, RA, RB, R3, R4, R5 and n are as defined in formula I.
In some embodiments, B is optionally substituted
In some embodiments, B is optionally substituted
In some embodiments, or deuteratedpreferably,
In some embodiments, RA isIn some embodiments, R1 is selected from the group con-sisting of hydrogen, C1-6 alkyl, C1-6 alkoxyl, C1-6 haloalkyl, C1-6 haloalkoxyl, C1-6 hydroxyalkyl, C1-6 amino-alkyl, optionally substituted C3-6 cycloalkyl, optionally substituted C3-6 heterocycloalkyl, optionally substi-tuted 5 to 8-membered aryl, optionally substituted 5 to 8-membered heteroaryl, optionally substituted 8 to 10-membered fused bicyclic aryl, optionally substituted 8 to 10-membered fused bicyclic heteroaryl, op-tionally substituted -C1-2 alkylene-C3-6 cycloalkyl, optionally substituted -C1-2 alkylene-C3-6 heterocycloalkyl, optionally substituted -C1-2 alkylene-5 to 8-membered aryl, optionally substituted -C1-2 alkylene-5 to 8-membered heteroaryl, optionally substituted -C1-2 alkylene-8 to 10-membered fused bicyclic aryl, and optionally substituted -C1-2 alkylene-8 to 10-membered fused bicyclic heteroaryl (preferably, R1 is selected from the group consisting of hydrogen, C1-6 alkyl, C1-6 alkoxyl, C1-6 haloalkyl, C1-6 haloalkoxyl, C1-6 hy-droxyalkyl, C1-6 aminoalkyl, optionally substituted C3-6 cycloalkyl, optionally substituted C3-6 heterocyclo-alkyl, optionally substituted 5 to 8-membered aryl, optionally substituted 5 to 8-membered heteroaryl, op-tionally substituted 8 to 10-membered fused bicyclic aryl, and optionally substituted 8 to 10-membered fused bicyclic heteroaryl) ; furthermore, the C1-6 alkyl, C1-6 alkoxyl, C1-6 haloalkyl, C1-6 haloalkoxyl, C1-6 hy-droxyalkyl (preferably the C1-6 alkyl) may be further optionally substituted.
In some embodiments, R1 is optionally substituted -C1-2 alkylene-C3-6 cycloalkyl, optionally substitut-ed -C1-2 alkylene-C3-6 heterocycloalkyl, optionally substituted -C1-2 alkylene-5 to 8-membered aryl, option-ally substituted -C1-2 alkylene-5 to 8-membered heteroaryl, optionally substituted -C1-2 alkylene-8 to 10-membered fused bicyclic aryl, and optionally substituted -C1-2 alkylene-8 to 10-membered fused bicy-clic heteroaryl; preferably, is optionally substituted -C1-2 alkylene-C3-6 cycloalkyl, optionally substituted -C1-2 alkylene-C3-6 heterocycloalkyl; more preferably, optionally substituted -methylene-C3-6 heterocycloal-kyl.
In some embodiments, “C3-6 heterocycloalkyl” is selected from heterocycloalkyl for R1’ as defined in formula VI, VII or VIII.
In some embodiments, in R1, when the C1-6 alkyl, C1-6 alkoxyl, C1-6 haloalkyl, C1-6 haloalkoxyl, or C1-6 hydroxyalkyl (preferably, the C1-6 alkyl) is further substituted, the substituent is selected from the group consisting of C1-6 alkyl, C1-6 alkoxyl, C1-6 haloalkyl, C1-6 haloalkoxyl, C1-6 hydroxyalkyl, C1-6 aminoalkyl, optionally substituted C3-6 cycloalkyl, optionally substituted C3-6 heterocycloalkyl, optionally substituted 5 to 8-membered aryl, optionally substituted 5 to 8-membered heteroaryl, optionally substituted 8 to 10-membered fused bicyclic aryl, optionally substituted 8 to 10-membered fused bicyclic heteroaryl; pref-erably, the substituent is selected from the group consisting of optionally substituted C3-6 cycloalkyl, op-tionally substituted C3-6 heterocycloalkyl, optionally substituted 5 to 8-membered aryl, optionally substi-tuted 5 to 8-membered heteroaryl, optionally substituted 8 to 10-membered fused bicyclic aryl, optionally substituted 8 to 10-membered fused bicyclic heteroaryl; more preferably, the substituent is optionally sub-stituted C3-6 heterocycloalkyl.
In some embodiments, in R1, optionally substituted means unsubstituted or being substituted with one or more (e.g., 1, 2 or 3) substituents selected from the group consisting of deuterium, halogen, C1-6 alkyl, C1-6 hydroxyalkyl, C1-6 deuteroalkyl, C3-6 cycloalkyl, C3-6 heterocycloalkyl, -CONRiRii (e.g., -CON (C1-6 al-kyl) 2) , -S (O) 2Ri (e.g., -S (O) 2 (C1-6 alkyl) ) , and -S (O) (=N-Ri) Rii (e.g., ) .
In some embodiments, in R1, optionally substituted means unsubstituted or being substituted with one or more (e.g., 1, 2 or 3) substituents selected from the group consisting of deuterium, halogen, C1-6 alkyl, C1-6 hydroxyalkyl, and C1-6 deuteroalkyl.
In some embodiments, R1 is - (CH2) -R1’ , and R1’ is as defined in formula VI, VII, or VIII.
In some embodiments, RM and RN are each independently selected from hydrogen, halogen, hydroxy, cyano, carboxyl, C1-6 alkyl, C1-6 alkoxyl, C1-6 haloalkyl, C1-6 haloalkoxyl, C1-6 hydroxyalkyl, C1-6 aminoalkyl, optionally substituted C3-6 cycloalkyl, optionally substituted C3-6 heterocycloalkyl; furthermore, the C1-6 alkyl, C1-6 alkoxyl, C1-6 haloalkyl, C1-6 haloalkoxyl, C1-6 hydroxyalkyl may be further optionally substituted. In some embodiments, RM and RN are each independently selected from hydrogen, and C1-6 alkyl.
In some embodiments, Y is -CH-or N; preferably, Y is N.
In some embodiments, R2 is optionally substituted C1-6 alkoxy, or optionally substituted C1-6 alkyl; preferably, R2 is optionally substituted C1-6 alkyl.
In some embodiments, in R2, optionally substituted means unsubstituted or being substituted with one or more (e.g., 1, 2 or 3) substituents selected from the group consisting of deuterium, C1-6 alkoxy, C1-6 deu-teroalkoxy, and C1-6 haloalkoxy.
In some embodiments of the present disclosure, B isRA is optionally substituted phenyl or optionally substituted 6-membered heteroaryl; in some embodiments, the compound of formula (I) is a compound of formula (II) :
or a pharmaceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt thereof,
wherein
A is optionally substituted 5 to 6-membered heterocycloalkylene, optionally substituted 5 to 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene;
G is optionally substituted C1-6 alkylene;
Y is -CH-or N;
R8 and R9 are each independently selected from hydrogen, deuterium, halogen, hydroxy, cyano, op-tionally substituted C1-3 alkyl, or R8 and R9 combine with the atoms to which they are attached to form an optionally substituted C3-6 cycloalkyl or a carbonyl;
R10 and R11 are each independently selected from hydrogen, deuterium, halogen, hydroxy, cyano, op-tionally substituted C1-3 alkyl, or R10 and R11 combine with the atoms to which they are attached to form an optionally substituted C3-6 cycloalkyl or a carbonyl;
R12 and R13 are each independently selected from hydrogen, deuterium, halogen, hydroxy, optionally substituted C1-3 alkyl, -O (C1-6 alkyl) , -S (C1-6 alkyl) or -N (C1-6 alkyl) (C1-6 alkyl) ; furthermore, the C1-6 alkyl may be further optionally substituted; Y is -CH-or N;
X1 is N or C;
X2 is N or -CRa-;
X3 is N or -CRb-;
X4 is N or -CRc-;
X5 is N or C;
X6 is S, O, N, or -CH-;
Ra, Rb, and Rc are each independently hydrogen, halogen, cyano, C1-3 alkyl, C1-3 alkoxyl, C1-3 haloal-kyl, C1-3 haloalkoxyl, C3-6 cycloalkyl, or C3-6 heterocycloalkyl (or 3 to 6-membered heterocycloalkyl) ;
R1 is selected from hydrogen, C1-6 alkyl, C1-6 alkoxyl, C1-6 haloalkyl, C1-6 haloalkoxyl, C1-6 hydroxy-alkyl, C1-6 aminoalkyl, optionally substituted C3-6 cycloalkyl, optionally substituted 3 to 6-membered het-erocycloalkyl, optionally substituted 5 to 8-membered aryl, optionally substituted 5 to 8-membered het-eroaryl, optionally substituted 8 to 10-membered fused bicyclic aryl or optionally substituted 8 to 10-membered fused bicyclic heteroaryl; furthermore, the C1-6 alkyl, C1-6 alkoxyl, C1-6 haloalkyl, C1-6 haloalkoxyl, and C1-6 hydroxyalkyl may be further optionally substituted;
R2 is optionally substituted C1-6 alkoxy, or optionally substituted C1-6 alkyl;
RM and RN are each independently selected from hydrogen, halogen, hydroxy, cyano, carboxyl, C1-6 alkyl, C1-6 alkoxyl, C1-6 haloalkyl, C1-6 haloalkoxyl, C1-6 hydroxyalkyl, C1-6 aminoalkyl, optionally substi-tuted C3-6 cycloalkyl, optionally substituted C3-6 heterocycloalkyl (or 4 to 7-membered heterocycloalkyl) ; furthermore, the C1-6 alkyl, C1-6 alkoxyl, C1-6 haloalkyl, C1-6 haloalkoxyl, and C1-6 hydroxyalkyl may be fur-ther optionally substituted;
R3 is absent, or selected from hydrogen, C1-6 alkyl, or C1-6 haloalkyl, provided that when X6 is O or S, R3 is absent;
E is a bond, or selected from N, or -CRd-, wherein Rd is selected from hydrogen, halogen, hydroxy, cyano, carboxyl, optionally substituted C1-6 alkyl, optionally substituted C1-6 alkoxyl, C1-6 aminoalkyl, C1-6 hydroxyalkyl, or -NReRf; wherein Re and Rf are each independently selected from hydrogen, or optionally substituted C1-6 alkyl;
R4 is hydrogen, optionally substituted C1-6 alkyl, optionally substituted C1-6 heteroalkyl, optionally substituted C3-10 cycloalkyl, optionally substituted C3-10 cycloalkenyl, optionally substituted C3-10 heterocy-cloalkyl (or 4 to 12-membered heterocycloalkyl) , optionally substituted 6 to 10-membered aryl, optionally substituted 5 to 10-membered heteroaryl, or
R5 is hydrogen, C1-6 alkyl or C3-6 cycloalkyl, furthermore, the C1-6 alkyl or C3-6 cycloalkyl may be fur-ther optionally substituted;
or R4 and R5 combine with the atoms to which they are attached to form a ring, wherein the ring is se-lected from optionally substituted 3 to 10-membered cycloalkyl, optionally substituted 3 to 10-membered heterocycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 4 to 11-membered bicyclic cycloalkyl, optionally substituted 4 to 11-membered bicyclic heterocycloalkyl.
wherein
L is absent, or selected from -CH2-, -C (O) -, -CHRg-or -C (Rg) 2-, wherein Rg is optionally substituted C1-6 alkyl;
R6 is hydrogen, or optionally substituted C1-6 alkyl;
R7 is optionally substituted C1-6 alkyl, optionally substituted C1-6 heteroalkyl, optionally substituted C3-10 cycloalkyl, optionally substituted C3-10 heterocycloalkyl (or 4 to 12-membered heterocycloalkyl) , op-tionally substituted 6 to 10-membered aryl, optionally substituted 5 to 10-membered heteroaryl;
or L, R6 and R7 combine with the atoms to which they are attached to form a ring, wherein the ring is selected from optionally substituted 3 to 10-membered cycloalkyl, optionally substituted 3 to 10-membered heterocycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 4 to 11-membered bicyclic cycloalkyl, optionally substituted 4 to 11-membered bicyclic heterocycloalkyl.
In some embodiments, X1 is C, and/or X2 is -CRa- (preferably -CH-) , and/or X3 is -CRb- (preferably -CH-) , and/or X4 is -CRc- (preferably -CH-) , and/or X5 is -C-.
In some embodiments, Ra, Rb, and Rc are each independently selected from the group consisting of hydrogen, halogen, cyano, C1-3 alkyl, C1-3 alkoxyl, C1-3 haloalkyl, and C1-3 haloalkoxyl; preferably, Ra, Rb, and Rc are each independently hydrogen, or C1-3 alkyl.
In some embodiments of the present disclosure, X1, X2, X3, X4 and X5 are -CH-, RM and RN are hy-drogen.
In some embodiments of the present disclosure, R8, R9, R10, R11, R12 and R13 are each independently selected from hydrogen, deuterium, halogen, hydroxy, cyano, optionally substituted C1-3 alkyl; preferably, R8, R9, R10, R11, R12 and R13 are each independently hydrogen.
In some embodiments, the compound of formula (II) is a compound of formula (II-1)
wherein X1, X2, X3, X4, X5, X6, A, B, E, G, Y, RM, RN, R1, R3, R4, R5 and n are as defined in formula II.
In some embodiments, the compound of formula (II) is a compound of formula (III) :
or a pharmaceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt thereof,
wherein
A is optionally substituted 5 to 6-membered heterocycloalkylene, optionally substituted 5 to 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene;
G is optionally substituted C1-6 alkylene;
Y is -CH-or N;
X6 is S, O, N, or -CH-;
Ra and Rb are each independently hydrogen, halogen, cyano, C1-3 alkyl, C1-3 alkoxyl, C1-3 haloalkyl, C1-3 haloalkoxyl, C3-6 cycloalkyl, or C3-6 heterocycloalkyl (or 4 to 7-membered heterocycloalkyl) ;
R1 is selected from hydrogen, C1-6 alkyl, C1-6 alkoxyl, C1-6 haloalkyl, C1-6 haloalkoxyl, C1-6 hydroxy-alkyl, C1-6 aminoalkyl, optionally substituted C3-6 cycloalkyl, optionally substituted C3-6 heterocycloalkyl (or 4 to 7-membered heterocycloalkyl) , optionally substituted 5 to 8-membered aryl, optionally substituted 5 to 8-membered heteroaryl, optionally substituted 8 to 10-membered fused bicyclic aryl or optionally sub-stituted 8 to 10-membered fused bicyclic heteroaryl; furthermore, the C1-6 alkyl, C1-6 alkoxyl, C1-6 haloalkyl, C1-6 haloalkoxyl, and C1-6 hydroxyalkyl may be further optionally substituted;
R2 is optionally substituted C1-6 alkoxy, or optionally substituted C1-6 alkyl;
R3 is absent, or selected from hydrogen, C1-6 alkyl, or C1-6 haloalkyl, provided that when X6 is O or S, R3 is absent;
E is a bond, or selected from N, or -CRd-, wherein Rd is selected from hydrogen, halogen, hydroxy, cyano, carboxyl, optionally substituted C1-6 alkyl, optionally substituted C1-6 alkoxyl, C1-6 aminoalkyl, C1-6 hydroxyalkyl, or -NReRf; wherein Re and Rf are independently selected from hydrogen, or optionally sub-stituted C1-6 alkyl;
R4 is hydrogen, optionally substituted C1-6 alkyl, optionally substituted C1-6 heteroalkyl, optionally substituted C3-10 cycloalkyl, optionally substituted C3-10 cycloalkenyl, optionally substituted C3-10 heterocy-cloalkyl (or 4 to 12-membered heterocycloalkyl) , optionally substituted 6 to 10-membered aryl, optionally substituted 5 to 10-membered heteroaryl, or
R5 is hydrogen, C1-6 alkyl or C3-6 cycloalkyl, furthermore, the C1-6 alkyl or C3-6 cycloalkyl may be fur-ther optionally substituted;
or R4 and R5 combine with the atoms to which they are attached to form a ring, wherein the ring is se-lected from optionally substituted 3 to 10-membered cycloalkyl, optionally substituted 3 to 10-membered heterocycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 4 to 11-membered bicyclic cycloalkyl, optionally substituted 4 to 11-membered bicyclic heterocycloalkyl;
wherein
L is absent, or selected from -CH2-, -C (O) -, -CHRg-or -C (Rg) 2-, wherein Rg is optionally substituted C1-6 alkyl;
R6 is hydrogen, or optionally substituted C1-6 alkyl;
R7 is optionally substituted C1-6 alkyl, optionally substituted C1-6 heteroalkyl, optionally substituted C3-10 cycloalkyl, optionally substituted C3-10 heterocycloalkyl (or 3 to 10-membered heterocycloalkyl) , op-tionally substituted 6 to 10-membered aryl, optionally substituted 5 to 10-membered heteroaryl;
or L, R6 and R7 combine with the atoms to which they are attached to form a ring, wherein the ring is selected from optionally substituted 3 to 10-membered cycloalkyl, optionally substituted 3 to 10-membered heterocycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 4 to 11-membered bicyclic cycloalkyl, optionally substituted 4 to 11-membered bicyclic heterocycloalkyl.
Furthermore, in the 5 to 6-membered heterocycloalkylene, 5 to 6-membered heteroarylene, C3-5 het-erocycloalkyl (or 4 to 6-membered heterocycloalkyl) , C3-6 heterocycloalkyl (or 4 to 7-membered heterocy-cloalkyl) , 5 to 6-membered heteroaryl, 8 to 10-membered fused bicyclic heteroaryl, C1-6 heteroalkyl, C3-10 heterocycloalkyl, 5 to 10-membered heteroaryl, 3 to 10-membered heterocycloalkyl or 4 to 11-membered bicyclic heterocycloalkyl, the heteroatom is independently selected from one or more of N, O and S, and the number of the heteroatom is independently 1 to 4.
In some embodiments of the present disclosure, the compound of formula (III) is a compound of for-mula (IVa) or formula (IVb) :
or a pharmaceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt thereof,
wherein
A is optionally substituted 5 to 6-membered heterocycloalkylene, optionally substituted 5 to 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene;
G is optionally substituted C1-6 alkylene;
Y is -CH-or N;
X6 is S, O, N, or -CH-;
Ra and Rb are each independently hydrogen, halogen, cyano, C1-3 alkyl, C1-3 alkoxyl, C1-3 haloalkyl, C1-3 haloalkoxyl, C3-6 cycloalkyl, C3-6 heterocycloalkyl (or 4 to 7-membered heterocycloalkyl) ;
R1 is selected from hydrogen, C1-6 alkyl, C1-6 alkoxyl, C1-6 haloalkyl, C1-6 haloalkoxyl, C1-6 hydroxy-alkyl, C1-6 aminoalkyl, optionally substituted C3-6 cycloalkyl, optionally substituted C3-6 heterocycloalkyl (or 4 to 7-membered heterocycloalkyl) , optionally substituted 5 to 8-membered aryl, optionally substituted 5 to 8-membered heteroaryl, optionally substituted 8 to 10-membered fused bicyclic aryl or optionally sub-stituted 8 to 10-membered fused bicyclic heteroaryl; furthermore, the C1-6 alkyl, C1-6 alkoxyl, C1-6 haloalkyl, C1-6 haloalkoxyl, C1-6 hydroxyalkyl may be further optionally substituted;
In some embodiments, R1 is optionally substituted C1-6 alkyl;
preferably, in R1, the C1-6 alkyl is further substituted, the substituent is selected from optionally sub-stituted C3-6 cycloalkyl, optionally substituted C3-6 heterocycloalkyl, optionally substituted 5 to 8-membered aryl, optionally substituted 5 to 8-membered heteroaryl, optionally substituted 8 to 10-membered fused bicyclic aryl, optionally substituted 8 to 10-membered fused bicyclic heteroaryl;
more preferably, the substituent is optionally substituted C3-6 heterocycloalkyl;
In some embodiments, R1 is
preferably, R1 is
more preferably, R1 is
R2 is optionally substituted C1-6 alkoxy, or optionally substituted C1-6 alkyl;
R3 is absent, or selected from hydrogen, C1-6 alkyl, or C1-6 haloalkyl, provided that when X6 is O or S, R3 is absent;
E is a bond, or selected from N, or -CRd-, wherein Rd is selected from hydrogen, halogen, hydroxy, cyano, carboxyl, optionally substituted C1-6 alkyl, optionally substituted C1-6 alkoxyl, C1-6 aminoalkyl, C1-6 hydroxyalkyl, or -NReRf; wherein Re and Rf are independently selected from hydrogen, or optionally sub-stituted C1-6 alkyl;
R4 is hydrogen, optionally substituted C1-6 alkyl, optionally substituted C1-6 heteroalkyl, optionally substituted C3-10 cycloalkyl, optionally substituted C3-10 cycloalkenyl, optionally substituted C3-10 heterocy-cloalkyl (or 4 to 12-membered heterocycloalkyl) , optionally substituted 6 to 10-membered aryl, optionally substituted 5 to 10-membered heteroaryl, or
R5 is hydrogen, C1-6 alkyl or C3-6 cycloalkyl, furthermore, the C1-6 alkyl or C3-6 cycloalkyl may be fur-ther optionally substituted;
or R4 and R5 combine with the atoms to which they are attached to form a ring, wherein the ring is se-lected from optionally substituted 3 to 10-membered cycloalkyl, optionally substituted 3 to 10-membered heterocycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 4 to 11-membered bicyclic cycloalkyl, optionally substituted 4 to 11-membered bicyclic heterocycloalkyl;
preferably, the ring is selected from optionally substituted 3 to 10-membered cycloalkyl, optionally substituted 3 to 10-membered heterocycloalkyl;
more preferably, the ring is selected from optionally substituted 3 to 10-membered cycloalkyl;
In some embodiments of the present disclosure, is
preferably, is
more preferably, is
wherein
L is absent, or selected from -CH2-, -C (O) -, -CHRg-or -C (Rg) 2-, wherein Rg is optionally substituted C1-6 alkyl;
R6 is hydrogen, or optionally substituted C1-6 alkyl;
R7 is optionally substituted C1-6 alkyl, optionally substituted C1-6 heteroalkyl, optionally substituted C3-10 cycloalkyl, optionally substituted C3-10 heterocycloalkyl, optionally substituted 6 to 10-membered aryl, optionally substituted 5 to 10-membered heteroaryl;
or L, R6 and R7 combine with the atoms to which they are attached to form a ring, wherein the ring is selected from optionally substituted 3 to 10-membered cycloalkyl, optionally substituted 3 to 10-membered heterocycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 4 to 11-membered bicyclic cycloalkyl, optionally substituted 4 to 11-membered bicyclic heterocycloalkyl.
Furthermore, in the 5 to 6-membered heterocycloalkylene, 5 to 6-membered heteroarylene, C3-5 het-erocycloalkyl, C3-6 heterocycloalkyl (or 3 to 6-membered heterocycloalkyl) , 5 to 6-membered heteroaryl, 8 to 10-membered fused bicyclic heteroaryl, C1-6 heteroalkyl, C3-10 heterocycloalkyl (or 3 to 10-membered heterocycyoalkyl) , 5 to 10-membered heteroaryl, 3 to 10-membered heterocycloalkyl or 4 to 11-membered bicyclic heteroalkyl, the heteroatom is independently selected from one or more of N, O and S, and the number of the heteroatom is independently 1 to 4.
In some embodiments, R4 and R5 combine with the atoms to which they are attached to form a ring, wherein the ring is optionally substituted 3 to 10-membered cycloalkyl; preferably, is optionally substituted 3 to 6-membered cycloalkyl such as optionally substituted cyclopropyl, optionally substituted cyclobutyl, optionally substituted cyclopentyl or optionally substituted cyclohexyl, more preferably, optionally substi-tuted 3-membered cycloalkyl i.e., optionally substituted cyclopropyl.
In some embodiments of the present disclosure, the 5 to 6-membered heterocycloalkylene contains one or two heteroatoms; preferably the heteroatom is each independently selected from N, O and S.
In some embodiments of the present disclosure, A is optionally substituted 5 to 6-membered heterocy-cloalkylene, the heteroatom is independently selected from one or two of N, O and S, and the number of the heteroatom is independently 1 to 2. In some embodiments, A is
In some embodiments of the present disclosure, A is optionally substituted 5 to 6-membered arylene;
In some embodiments, A is
In some embodiments of the present disclosure, the 5 to 6-membered heteroarylene contains one, two or three (preferably one or two) heteroatoms; preferably the heteroatom is each independently selected from N, O and S.
In some embodiments of the present disclosure, A is optionally substituted 5 to 6-membered het-eroarylene, the heteroatom is independently selected from one or two of N, O and S, and the number of the heteroatom is independently 1 to 3.
In some embodiments of the present disclosure, A is optionally substituted 5-membered heteroarylene, the heteroatom is independently selected from N, O and S, and the number of the heteroatom is inde-pendently 1 to 2; preferably, one of the heteroatom is N, and the other, if present, is O or S.
In some embodiments, A is
In some embodiments, A is
In some embodiments of the present disclosure, B is selected from the group consisting of optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 3 to 6-membered heterocycloal-kenylene, optionally substituted 4 to 11-membered bicyclic alkylene, and optionally substituted 4 to 11-membered bicyclic heteroalkylene.
In some embodiments, B is
In some embodiments of the present disclosure, G is optionally substituted C1-6 alkylene; preferabaly, G is optionally substituted -C (C1-3alkyl) 2-.
In some embodiments, in G, optionally substituted means unsubstitued or being substituted with one or more deuterium.
In some embodiments, G is
In some embodiments of the present disclosure, Y is N.
In some embodiments of the present disclosure, X6 is N.
In some embodiments of the present disclosure, R1 is C1-6 alkyl, C1-6 alkoxyl, C1-6 haloalkyl, C1-6 haloalkoxyl, C1-6 hydroxyalkyl, C1-6 aminoalkyl, optionally substituted C3-6 cycloalkyl, or optionally substi-tuted C3-6 heterocycloalkyl (or 3 to 6-membered heterocycloalkyl) .
In some embodiments, R1 is
In some embodiments of the present disclosure, R1 is C1-6 alkyl and the C1-6 alkyl is further substituted with optionally substituted C3-6 cycloalkyl, optionally substituted C3-6 heterocycloalkyl.
In some embodiments of the present disclosure, R1 is C1 alkyl; and the C1 alkyl is further substituted with optionally substituted C3-6 heterocycloalkyl, wherein the heterocycloalkyl is saturated and the hetero-cycloalkyl is attached to the rest of the molecule via the N atom on the ring.
In some embodiments of the present disclosure, R1 is C1 alkyl; and the C1 alkyl is further substituted with optionally substituted C3-6 heterocycloalkyl, wherein the heterocycloalkyl is saturated and the hetero-cycloalkyl is attached to the rest of the molecule via the N atom on the ring. and the C3-6 heterocycloalkyl is 5 to 7 menbered heterocycloalkyl and has l to 2 heteroatoms.
In some embodiments, R1 is
In some embodiments, R1 is
In some embodiments of the present disclosure, R1 is optionally substituted 4 to 5-membered het-eroalkyl and 4 to 5-membered heteroaryl.
In some embodiments, R1 is
In some embodiments of the present disclosure, R1 is optionally substituted 5 to 8-membered aryl, op-tionally substituted 5 to 8-membered heteroaryl, optionally substituted 8 to 10-membered fused bicyclic aryl or optionally substituted 8 to 10-membered fused bicyclic heteroaryl, the heteroatom is independently selected from one or two of N, O and S, and the number of the heteroatom is independently 1 to 4.
In some embodiments, R1 is
In some embodiments of the present disclosure, R2 is optionally substituted C1-6 alkyl or optionally substituted C1-6 alkoxy.
In some embodiments, R2 ispreferably,
In some embodiments of the present disclosure, R3 is C1-6 alkyl. In some embodiments, R3 is C1-6 deu-terated alkyl. In some embodiments, R3 is
In some embodiments of the present disclosure, R3 is C1-6 haloalky.
In some embodiments, R3 is
In some embodiments of the present disclosure, E is a bond, or selected from N, or -CRd-, wherein Rd is selected from hydrogen, halogen, hydroxy, cyano, carboxyl, optionally substituted C1-6 alkyl, optionally substituted C1-6 alkoxyl, C1-6 aminoalkyl, C1-6 hydroxyalkyl, or -NReRf; wherein Re and Rf are independent-ly selected from hydrogen, or optionally substituted C1-6 alkyl; preferably, E is a bond, or selected from N, or -CRd-, wherein Rd is selected from hydrogen, halogen, hydroxy or C1-6 alkyl.
In some embodiments of the present disclosure, Rd is halogen; In some embodiments, Rd is -F, -Cl, -Br or -I; preferably, -F.
In some embodiments of the present disclosure, Rd is C1-6 alkyl;
In some embodiments, Rd is
In some embodiments of the present disclosure, Rd is C1-6 haloalkyl; In some embodiments, Rd is -CF3.
In some embodiments of the present disclosure, Rd is C1-6 alkoxyl;
In some embodiments, Rd is
In some embodiments of the present disclosure, Rd is C1-6 aminoalkyl; In some embodiments, Rd is
In some embodiments of the present disclosure, Re or Rf is C1-6 alkyl; In some embodiments, Re or Rf is independently selected fromand preferably the other is hydrogen.
In some embodiments, L is selected from -CH2-, -C (O) -, -CHRg-or -C (Rg) 2-, wherein Rg is optionally substituted C1-6 alkyl.
In some embodiments, R4 is hydrogen, optionally substituted C1-6 alkyl, optionally substituted C1-6 heteroalkyl, optionally substituted C3-10 cycloalkyl, optionally substituted C3-10 heterocycloalkyl (or 4 to 12-membered heterocycloalkyl) , optionally substituted 6 to 10-membered aryl, optionally substituted 5 to 10-membered heteroaryl, or
preferably, R4 is hydrogen, optionally substituted C1-6 alkyl, or
In some embodiments, R6 is hydrogen, or optionally substituted C1-6 alkyl; preferably, hydrogen, or C1-6 alkyl.
In some embodiments, R7 is optionally substituted C1-6 alkyl, optionally substituted C1-6 heteroalkyl, optionally substituted C3-10 cycloalkyl, optionally substituted C3-10 heterocycloalkyl, optionally substituted 6 to 10-membered aryl, optionally substituted 5 to 10-membered heteroaryl; preferably, R7 is C1-6 alkyl, op-tionally substituted C3-10 cycloalkyl.
In some embodiments of the present disclosure, R4 is hydrogen, optionally substituted C1-6 alkyl, op-tionally substituted C1-6 heteroalkyl, optionally substituted C3-10 cycloalkyl, optionally substituted C3-10 het-erocycloalkyl (or 4 to 12-membered heterocycloalkyl) , optionally substituted 6 to 10-membered aryl, op-tionally substituted 5 to 10-membered heteroaryl, orwherein R6 is hydrogen, or optionally sub-stituted C1-6 alkyl; R7 is optionally substituted C1-6 alkyl, optionally substituted C1-6 heteroalkyl, optionally substituted C3-10 cycloalkyl, optionally substituted C3-10 heterocycloalkyl, optionally substituted 6 to 10-membered aryl, optionally substituted 5 to 10-membered heteroaryl.
In some embodiments, in R4, optionally substituted means unsubstituted or being substituted by one or more substituents selected from the group consisting of hydroxyl, cyano, halogen, and C1-6 alkyl. In some embodiments of the present disclosure, R4 is hydrogen, C1-6 alkyl, 3 to 6-membered heterocycloalkyl, 3 to 6-membered cycloalkyl, 6 to 10-membered aryl, 5 to 10-membered heteroaryl, orwherein R6 is hydrogen, or C1-6 alkyl; R7 is C1-6 alkyl, 3-to 10-membered heterocycloalkyl; wherein the 3 to 10-membered heterocycloalkyl is optionally substituted by C1-6 alkyl, 6 to 10-membered aryl, -CO-O-C3-6 cycloalkyl or -CO-O-C1-6 alkyl; the 3 to 6-membered cycloalkyl is optionally substituted by hydroxyl, cy-ano, halogen, or C1-6 alkyl; in 3 to 10-membered heterocycloalkyl, the heteroatom is independently selected from one or more of N, O and S, and the number of the heteroatom is independently 1 to 3; more prefera-bly, R4 is hydrogen, C1-6 alkyl, orwherein R6 is hydrogen, or C1-6 alkyl; R7 is C1-6 alkyl, 3 to 10-membered heterocycloalkyl.
In some embodiments, in R4, C3-10 heterocycloalkyl (or 4 to 12-membered heterocycloalkyl) contains 1 or 2 heteroatoms selected from the group consisting of N, O and S.
In some embodiments of the present disclosure, R4 is optionally substituted C1-6 alkyl, optionally sub-stituted C1-6 heteroalkyl, optionally substituted C3-10 cycloalkyl, optionally substituted C3-10 heterocycloalkyl, the heteroatom is N, O or S, and the number of the heteroatom is independently 1 to 2. In some embodi-ments, R4 is
In some embodiments of the present disclosure, R4 is optionally substituted 6 to 10-membered aryl, optionally substituted 5 to 10-membered heteroaryl.
In some embodiments, R4 is
In some embodiments of the present disclosure, R5 is optionally substituted C1-6 alkyl. In some em-bodiments, R5 is
In some embodiments of the present disclosure, R5 is optionally substituted C3-6 cycloalkyl. In some embodiments, R5 is
In some embodiments, in the ring formed by R4, R5 and the atoms to which they are attached, the het-erocycloalkyl or the bicyclic heterocycloalkyl contains 1, 2 or 3 heteroatom selected from the group con-sisting of N, O and S.
In some embodiments of the present disclosure, R4 and R5 combine with the atoms to which they are attached to form a ring, wherein the ring is selected from optionally substituted 3 to 10-membered cyclo-alkyl, optionally substituted 3 to 10-membered heterocycloalkyl, optionally substituted 4 to 11-membered bicyclic cycloalkyl, optionally substituted 4 to 11-membered bicyclic heterocycloalkyl, the heteroatom is N, O or S, and the number of the heteroatom is independently 1 to 3,
preferably, the ring is
In some embodiments, R4 and R5 combine with the atoms to which they are attached to form a ring, wherein the ring is optionally substituted 3 to 10-membered cycloalkyl; preferably, the ring is optionally substituted 3 to 4-membered cycloalkyl; more preferably, the ring is cy optionally substituted clopropyl; preferably, wherein optionally substituted means unsubstituted or being substituted with one or more sub-stituents selected from the group consisting of C1-4 alkyl, C1-4 deuteroalkyl, cyano, C1-4 hydroxyalkyl, and C1-4 haloalkyl.
In some embodiments of the present disclosure, R6 is optionally substituted C1-6 alkyl; In some em-bodiments, R6 is
In some embodiments, R7 is optionally substituted C3-10 cycloalkyl, or optionally substituted C3-10 het-erocycloalkyl.
In some embodiments, in R7, substituents for C3-10 heterocycloalkyl are selected from the group con-sisting of halogen, C1-6 alkyl, 6 to 10-membered aryl, -CO-O-C3-6 cycloalkyl, -CO-O-C1-6 alkyl, -CO-C3-6 cycloalkyl, -CO-C1-6 alkyl, -CO-C1-6 alkenyl, and -CO-C1-6 alkynyl, and/or substituents for C3-10 cycloalkyl are selected from the group consisting of hydroxyl, cyano, halogen, or C1-6 alkyl.
In some embodiments, in R7, substituents for C3-10 heterocycloalkyl are selected from the group con-sisting of C1-6 alkyl, 6 to 10-membered aryl, -CO-O-C3-6 cycloalkyl and -CO-O-C1-6 alkyl, and/or substitu-ents for C3-10 cycloalkyl are selected from the group consisting of hydroxyl, cyano, halogen, or C1-6 alkyl.
In some embodiments, in R7, C3-10 heterocycloalkyl contains 1 to 3 heteroatoms selected from the group consisting of N, O and S.
In some embodiments, R7 is optionally substituted C3-10 cycloalkyl, optionally substituted C3-10 heter-ocycloalkyl; preferably, the C3-10 heterocycloalkyl optionally substituted by C1-6 alkyl, C6-10 aryl , -CO-O-C3-6 cycloalkyl or -CO-O-C1-6 alkyl; the C3-10 cycloalkyl optionally substituted by hydroxyl, cyano, halogen, or C1-6 alkyl; in C3-10 heterocycloalkyl, the heteroatom is independently selected from one or more of N, O and S, and the number of the heteroatom is independently 1 to 3.
In some embodiments of the present disclosure, R7 is C1-6 alkyl.
In some embodiments, R7 is
In some embodiments, R7 is optionally substituted 3 to 6-membered heterocycloalkyl.
In some embodiments, in R7, the 3 to 6-membered heterocycloalkyl contains 1 or 2 heteroatom se-lected from N, O or S; preferably, is
In some embodiments of the present disclosure, R7 is 3 to 6-membered heterocycloalkyl, preferably, the heteroatom is N, O or S, and the number of the heteroatom is independently 1 to 2. In some embodi-ments, R7 is
In some embodiments, R7 is optionally substituted C3-6 cycloalkyl. In some embodiments, in R7, the C3-6 cycloalkyl is
In some embodiments of the present disclosure, R7 is C3-6 cycloalkyl;
In some embodiments, R7 is
In some embodiments, R7 is optionally substituted C6-10 aryl. In some embodiments, in R7, the C6-10 aryl is
In some embodiments of the present disclosure, R7 is C6-10 aryl.
In some embodiments, R7 is
In some embodiments, R7 is optionally substituted C1-6 alkyl or optionally substituted 3 to 6-membered heterocycloalkyl.
In some embodiments, in R7, substituents for the 3 to 6-membered heterocycloalkyl is selected from -C (O) Ri or -NHC (O) Ri; wherein Ri is selected from the group consisting of C1-6 alkyl, -C2-6alkenyl, C2-6 al-kynyl; wherein the alkyl, alkenyl, or alkynyl is unsubtituted or one or more (such as 1, 2, 3 or 4) of H atom in the group being substituted with substituents selected from the group consisting of D, halogen, -CN, -OH, C1-6 alkyl, C1-6 haloalkyl, C1-6 deuteroalkyl, C1-6 hydroxyalkyl, C1-6 aminoalkyl, alkylamino.
In some embodiments, in R7, substituents for the 3 to 6-membered heterocycloalkyl is selected from 6 to 10-membered aryl or -CO-O-C1-6 alkyl.
In some embodiments, in R7, the 3-to 6-membered heterocycloalkyl contains 1 or 2 heteroatoms se-lected from N and O.
In some embodiments of the present disclosure, R7 is C1-6 alkyl or 3 to 6-membered heterocycloalkyl; the 3 to 6-membered heterocycloalkyl optionally substituted by 6 to 10-membered aryl or -CO-O-C1-6 alkyl; in 3 to 6-membered heterocycloalkyl, the heteroatom is independently selected from one or two of N and O, and the number of the heteroatom is independently 1 to 2.
In some embodiments, R7 is
In some embodiments of the present disclosure, L, R6 and R7 combine with the atoms to which they are attached to form a ring, wherein the ring is selected from optionally substituted 3 to 10-membered cy-cloalkyl, optionally substituted 3 to 10-membered heterocycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 4 to 11-membered bicyclic cycloalkyl, optionally substi-tuted 4 to 11-membered bicyclic heterocycloalkyl.
In some embodiments, substituents for the ring formed by L, R6 and R7 combined with the atoms to which they are attached is -C (O) Ri; wherein Ri is selected from the group consisting of C1-6 alkyl, -C2-6alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, and 3 to 10-membered heterocycloalky; wherein the alkyl, alkenyl, alkynyl, cycloalkyl, or heterocycloalky is unsubtituted or one or more (such as 1, 2, 3 or 4) of H atom in the group being substituted with substituents selected from the group consisting of D, halogen, C1-6 alkyl, C1-6 haloalkyl, C1-6 deuteroalkyl, C1-6 hydroxyalkyl, C1-6 aminoalkyl, alkylamino, and C3-6 cycloalkyl.
In some embodiments, is
In some embodiments of the present disclosure, is
In some embodiments of the present disclosure, the “halogen” is F, Cl, Br or I; preferably, F.
In some embodiments of the present disclosure, the compound of formula (IVa) is a compound of formula (V) :
or a pharmaceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt thereof, wherein the variables are as described above.
In some embodiments, X1, X2, X3, X4, X5, X6, A, B, E, G, L, Y, Ra, Rb, Rc, RA, RB, RM, RN, R1, R2, R3, R4, R5, R6, R7 and n are each independently corresponding groups in specific compound shown below or compounds of examples 1 to 30.
The present invention also has some schemes derived from any combination of the above-mentioned variables.
In another aspect, the present invention also provides, but is not limited to, the following compounds, and stereoisomers thereof or their pharmacologically acceptable salts:
In some compounds of Formulas above, the atoms may exhibit their natural isotopic abundances, or one or more of the atoms may be artificially enriched in a particular isotope having the same atomic num-ber, but an atomic mass or mass number different from the atomic mass or mass number predominantly found in nature. The present disclosure as described and claimed herein is meant to include all suitable isotopic variations of the compounds of Formulas above and embodiments thereof. For example, different isotopic forms of hydrogen (H) include protium (1H) and deuterium (2H, also denoted herein as D) . Pro-tium is the predominant hydrogen isotope found in nature. Enriching for deuterium may afford certain therapeutic advantages, such as increasing in vivo half-life or reducing dosage requirements or may pro-vide a compound useful as a standard for characterization of biological samples. Isotopically-enriched compounds can be prepared without undue experimentation by conventional techniques well known to those skilled in the art or by processes analogous to those described in the Schemes and Examples herein using appropriate isotopically-enriched reagents and/or intermediates.
In some embodiments of the present disclosure, the compound of formula (V) is a compound of for-mula (VI) :
or a pharmaceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt thereof.
In some embodiments, R1’ of formula (VI) is selected from:
In some embodiments of the present disclosure, the compound of formula (IVa) is a compound of formula (VII) :
In some embodiments, R1’ of formula (VII) is selected from:
In some embodiments of the present disclosure, the compound of formula (IVa) is a compound of formula (VIII) :
In some embodiments, R1’ of formula (VIII) is selected from:
In some embodiments, the compound of formula (I) is Compound A as defined below.
It is to be understood that the foregoing description of preferred embodiments is intended to be purely illustrative of the principles of the invention, rather than exhaustive thereof, and that changes and varia-tions will be apparent to those skilled in the art, and that the present invention is not intended to be limited other than expressly set forth in the following claims.
The present invention provides a Compound A, a pharmaceutically acceptable salt thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt, wherein, the Compound A is selected from any one of the following compounds in Table 1:
Table 1. Certain Compounds of the Present invention
In another aspect, the present invention also provides a pharmaceutical composition comprising a compound, wherein a pharmaceutically acceptable salt thereof, a solvate thereof or a solvate of the phar-maceutically acceptable salt as defined above, and a pharmaceutically acceptable carrier, preferably, the compound is the Compound A as defined above.
In another aspect, the present invention also provides a use of a compound, wherein a pharmaceuti-cally acceptable salt thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt, or the pharmaceutical composition as defined above in the preparation of a medicament used for the treatment of cancer, preferably, the compound is the Compound A as defined above.
In some embodiments of the present disclosure, wherein the cancer is a Ras-driven cancer.
In some embodiments of the present disclosure, wherein the cancer comprises a Ras mutation.
In some embodiments of the present disclosure, wherein the Ras mutation is a position 12, 13 or 61.
In some embodiments of the present disclosure, wherein the Ras mutation is at position 12.
In some embodiments of the present disclosure, wherein the Ras mutation is selected from the group consisting of G12C, G12D, G12V, G12A, G12R, G12S, G13C, G13D, Q61H, Q61R and Q61L, or a com-bination thereof.
In some embodiments of the present disclosure, wherein the Ras mutation is a position selected from the group consisting of G12D, G12V, and G12R, or a combination thereof.
In some embodiments of the present disclosure, wherein the cancer is pancreatic cancer, appendiceal cancer, small bowel cancer, colorectal cancer, ampullary cancer, non-small cell lung cancer, cervical cancer, lung cancer, endometrial cancer, acute myeloid leukemia, gastrointestinal neuroendocrine tumor, uterine endometrioid carcinoma, oesophagogastric cancer, bladder cancer, ovarian cancer, melanoma, multiple myeloma, thyroid gland adenocarcinoma, a myelodysplastic syndrome, or squamous cell lung carcinoma.
In some embodiments of the present disclosure, wherein the cancer is pancreatic cancer, lung cancer, or colorectal cancer.
In some embodiments of the present disclosure, wherein the Ras protein is KRAS.
In some embodiments, the compound can be used in combination with an additional anti-cancer ther-apy.
In some embodiments, the additional anti-cancer therapy is an EGFR inhibitor, a second Ras inhibitor, a SHP2 inhibitor, a SOS1 inhibitor, a Raf inhibitor, a MEK inhibitor, an ERK inhibitor, a Pl3K inhibitor, a PTEN inhibitor, an AKT inhibitor, an mTORC1 inhibitor, a BRAF inhibitor, a PD-L1 inhibitor, a PD-1 in-hibitor, a CDK4/6 inhibitor, a HER2 inhibitor, or a combination thereof.
In another aspect, the present invention also provides a method for treating cancer in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of the com-pound or a pharmaceutically acceptable salt thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt as defined above, or the pharmaceutical composition as defined above, preferably the com-pound is the Compound A as defined above.
In some embodiments of the present disclosure, wherein the cancer is a Ras-driven cancer.
In some embodiments of the present disclosure, wherein the cancer comprises a Ras mutation.
In some embodiments of the present disclosure, wherein the Ras mutation is a position 12, 13 or 61.
In some embodiments of the present disclosure, wherein the Ras mutation is at position 12.
In some embodiments of the present disclosure, wherein the Ras mutation is at a position selected from the group consisting of G12C, G12D, G12V, G12A, G12R, G12S, G13C, G13D, Q61H, Q61R and Q61L, or a combination thereof.
In some embodiments of the present disclosure, wherein the Ras mutation is a position selected from the group consisting of G12D, G12V, and G12R, or a combination thereof.
In some embodiments of the present disclosure, wherein the cancer is pancreatic cancer, appendiceal cancer, small bowel cancer, colorectal cancer, ampullary cancer, non-small cell lung cancer, cervical cancer, lung cancer, endometrial cancer, acute myeloid leukemia, gastrointestinal neuroendocrine tumour, uterine endometrioid carcinoma, oesophagogastric cancer, bladder cancer, ovarian cancer, melanoma, multiple myeloma, thyroid gland adenocarcinoma, a myelodysplastic syndrome, or squamous cell lung carcinoma.
In some embodiments of the present disclosure, wherein the cancer is pancreatic cancer, lung cancer, or colorectal cancer.
In some embodiments of the present disclosure, wherein the Ras protein is KRAS.
In some embodiments, the method is provided of treating a Ras protein-related disorder in a subject in need thereof.
In some embodiments, the method further comprises administering an additional anti-cancer therapy.
In some embodiments, the additional anti-cancer therapy is an EGFR inhibitor, a second Ras inhibitor, a SHP2 inhibitor, a SOS1 inhibitor, a Raf inhibitor, a MEK inhibitor, an ERK inhibitor, a Pl3K inhibitor, a PTEN inhibitor, an AKT inhibitor, an mTORC1 inhibitor, a BRAF inhibitor, a PD-L1 inhibitor, a PD-1 in-hibitor, a CDK4/6 inhibitor, a HER2 inhibitor, or a combination thereof.
It should be understood that each of the above technical features of the invention and each technical feature specifically described below (such as in Examples) can be combined with each other within the scope of the present invention so as to constitute new or preferred technical solutions.
EXEMPLARY EMBODIMENTS
Embodiment 1. The compound of any one of the preceding embodiments, or a stereoisomer thereof, a pharmaceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt thereof, wherein the compound is a compound of formula (I) ;
wherein
n is 0, 1, 2, or 3;
A is optionally substituted 5 to 6-membered heterocycloalkylene, optionally substituted 5 to 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene;
B is selected from optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 3 to 6-membered heterocycloalkenylene, optionally substituted 4 to 11-membered bicyclic cycloalkylene, or optionally substituted 4 to 11-membered bicyclic heterocycloalkylene;
G is optionally substituted C1-6 alkylene;
R8 and R9 are each independently selected from hydrogen, deuterium, halogen, hydroxy, cyano, op-tionally substituted C1-3 alkyl, or R8 and R9 combine with the atoms to which they are attached to form an optionally substituted C3-6 cycloalkyl or a carbonyl;
R10 and R11 are each independently selected from hydrogen, deuterium, halogen, hydroxy, cyano, op-tionally substituted C1-3 alkyl, or R10 and R11 combine with the atoms to which they are attached to form an optionally substituted C3-6 cycloalkyl or a carbonyl;
R12 and R13 are each independently selected from hydrogen, deuterium, halogen, hydroxy, optionally substituted C1-3 alkyl, -O (C1-6 alkyl) , -S (C1-6 alkyl) or -N (C1-6 alkyl) (C1-6 alkyl) ; furthermore, the C1-6 alkyl may be further optionally substituted;
RA is optionally substituted C3-6 cycloalkyl, optionally substituted C3-6 heterocycloalkyl (or 4 to 7-membered heterocycloalkyl) , optionally substituted 5 to 8-membered aryl, optionally substituted 5 to 8-membered heteroaryl, optionally substituted 8 to 10-membered fused bicyclic aryl or optionally substi-tuted 8 to 10-membered fused bicyclic heteroaryl;
RB is selected from hydrogen, C1-6 alkyl, C1-6 alkoxyl, C1-6 haloalkyl, C1-6 haloalkoxyl, optionally sub-stituted C3-6 cycloalkyl, or optionally substituted C3-6 heterocycloalkyl (or 4 to 7-membered heterocycloal-kyl) ; or provided that when n exceeds 1, any two of RB combine with the atoms to which they are attached to form a 3 to 6-membered ring, wherein the ring can be optionally substituted with halogen, hydroxy, and C1-3 alkyl;
X1 is N or C;
X2 is N or -CRa-;
X3 is N or -CRb-;
X4 is N or -CRc-;
X5 is N or C;
X6 is S, O, N, or -CH-;
Ra, Rb, and Rc are each independently hydrogen, halogen, cyano, C1-3 alkyl, C1-3 alkoxyl, C1-3 haloal-kyl, C1-3 haloalkoxyl, C3-6 cycloalkyl, or C3-6 heterocycloalkyl (or 4 to 7-membered heterocycloalkyl) ;
R3 is absent, or selected from hydrogen, C1-6 alkyl, or C1-6 haloalkyl, provided that when X6 is O or S, R3 is absent;
E is a bond, or selected from N, or -CRd-, wherein Rd is selected from hydrogen, halogen, hydroxy, cyano, carboxyl, optionally substituted C1-6 alkyl, optionally substituted C1-6 alkoxyl, C1-6 aminoalkyl, C1-6 hydroxyalkyl, or -NReRf; wherein Re and Rf are independently selected from hydrogen, or optionally sub-stituted C1-6 alkyl;
R4 is hydrogen, optionally substituted C1-6 alkyl, optionally substituted C1-6 heteroalkyl, optionally substituted C3-10 cycloalkyl, optionally substituted C3-10 cycloalkenyl, optionally substituted C3-10 heterocy-cloalkyl (or 4 to 12-membered heterocycloalkyl) , optionally substituted 6 to 10-membered aryl, optionally substituted 5 to 10-membered heteroaryl, or
R5 is hydrogen, C1-6 alkyl or C3-6 cycloalkyl, furthermore, the C1-6 alkyl or C3-6 cycloalkyl may be fur-ther optionally substituted;
or R4 and R5 combine with the atoms to which they are attached to form a ring, wherein the ring is se-lected from optionally substituted 3 to 10-membered cycloalkyl, optionally substituted 3 to 10-membered heterocycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 4 to 11-membered bicyclic cycloalkyl, optionally substituted 4 to 11-membered bicyclic heterocycloalkyl;
wherein
L is absent, or selected from -CH2-, -C (O) -, -CHRg-or -C (Rg) 2-, wherein Rg is optionally substituted C1-6 alkyl;
R6 is hydrogen, or optionally substituted C1-6 alkyl;
R7 is optionally substituted C1-6 alkyl, optionally substituted C1-6 heteroalkyl, optionally substituted C3-10 cycloalkyl, optionally substituted C3-10 heterocycloalkyl (or 3 to 10-membered heterocycloalkyl) , op-tionally substituted 6 to 10-membered aryl, optionally substituted 5 to 10-membered heteroaryl;
or L, R6 and R7 combine with the atoms to which they are attached to form a ring, wherein the ring is selected from optionally substituted 3 to 10-membered cycloalkyl, optionally substituted 3 to 10-membered heterocycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 4 to 11-membered bicyclic cycloalkyl, optionally substituted 4 to 11-membered bicyclic heterocycloalkyl.
Embodiment 2. The compound of embodiment 1, or a stereoisomer thereof, a pharmaceutically ac-ceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharma-ceutically acceptable salt thereof, wherein the compound of formula (I) is a compound of formula (I-1)
wherein X1, X2, X3, X4, X5, X6, A, B, E, G, RA, RB, R3, R4, R5 and n are as defined in embodiment 1.
Embodiment 3. The compound of any one of embodiments 1 to 2, or a stereoisomer thereof, a phar-maceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt thereof, wherein
B is optionally substitutedand
RA is
wherein
R1 is selected from the group consisting of hydrogen, C1-6 alkyl, C1-6 alkoxyl, C1-6 haloalkyl, C1-6 haloalkoxyl, C1-6 hydroxyalkyl, C1-6 aminoalkyl, optionally substituted C3-6 cycloalkyl, optionally substi-tuted C3-6 heterocycloalkyl, optionally substituted 5 to 8-membered aryl, optionally substituted 5 to 8-membered heteroaryl, optionally substituted 8 to 10-membered fused bicyclic aryl, optionally substituted 8 to 10-membered fused bicyclic heteroaryl, optionally substituted -C1-2 alkylene-C3-6 cycloalkyl, optionally substituted -C1-2 alkylene-C3-6 heterocycloalkyl, optionally substituted -C1-2 alkylene-5 to 8-membered aryl, optionally substituted -C1-2 alkylene-5 to 8-membered heteroaryl, optionally substituted -C1-2 alkylene-8 to 10-membered fused bicyclic aryl, and optionally substituted -C1-2 alkylene-8 to 10-membered fused bicy-clic heteroaryl; furthermore, the C1-6 alkyl, C1-6 alkoxyl, C1-6 haloalkyl, C1-6 haloalkoxyl, C1-6 hydroxyalkyl may be further optionally substituted;
in R1, when the C1-6 alkyl, C1-6 alkoxyl, C1-6 haloalkyl, C1-6 haloalkoxyl, or C1-6 hydroxyalkyl is further substituted, the substituent is selected from the group consisting of C1-6 alkyl, C1-6 alkoxyl, C1-6 haloalkyl, C1-6 haloalkoxyl, C1-6 hydroxyalkyl, C1-6 aminoalkyl, optionally substituted C3-6 cycloalkyl, optionally sub-stituted C3-6 heterocycloalkyl, optionally substituted 5 to 8-membered aryl, optionally substituted 5 to 8-membered heteroaryl, optionally substituted 8 to 10-membered fused bicyclic aryl, optionally substituted 8 to 10-membered fused bicyclic heteroaryl; preferably, the substituent is selected from the group consist-ing of optionally substituted C3-6 cycloalkyl, optionally substituted C3-6 heterocycloalkyl, optionally substi-tuted 5 to 8-membered aryl, optionally substituted 5 to 8-membered heteroaryl, optionally substituted 8 to 10-membered fused bicyclic aryl, optionally substituted 8 to 10-membered fused bicyclic heteroaryl; more preferably, the substituent is optionally substituted C3-6 heterocycloalkyl;
Y is -CH-or N;
R2 is optionally substituted C1-6 alkoxy, or optionally substituted C1-6 alkyl; preferably, R2 is optionally substituted C1-6 alkyl; and
RM and RN are each independently selected from hydrogen, halogen, hydroxy, cyano, carboxyl, C1-6 alkyl, C1-6 alkoxyl, C1-6 haloalkyl, C1-6 haloalkoxyl, C1-6 hydroxyalkyl, C1-6 aminoalkyl, optionally substi-tuted C3-6 cycloalkyl, optionally substituted C3-6 heterocycloalkyl; furthermore, the C1-6 alkyl, C1-6 alkoxyl, C1-6 haloalkyl, C1-6 haloalkoxyl, C1-6 hydroxyalkyl may be further optionally substituted. In some embodi-ments, RM and RN are each independently selected from hydrogen, and C1-6 alkyl.
Embodiment 4. The compound of embodiment 3, or a stereoisomer thereof, a pharmaceutically ac-ceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharma-ceutically acceptable salt thereof, wherein
R1 is optionally substituted C1-6 alkyl;
and, in R1, the C1-6 alkyl is further substituted, and the substituent is selected from optionally substi-tuted C3-6 cycloalkyl, optionally substituted C3-6 heterocycloalkyl, optionally substituted 5 to 8-membered aryl, optionally substituted 5 to 8-membered heteroaryl, optionally substituted 8 to 10-membered fused bicyclic aryl, optionally substituted 8 to 10-membered fused bicyclic heteroaryl.
Embodiment 5. The compound of any one of embodiments 3 to 4, or a stereoisomer thereof, a phar-maceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt thereof, wherein
R1 is optionally substituted C1-6 alkyl;
in R1, the C1-6 alkyl is further substituted, and the substituent is optionally substituted C3-6 heterocy-cloalkyl.
Embodiment 6. The compound of any one of embodiments 3 to 4, or a stereoisomer thereof, a phar-maceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt thereof, wherein R1 is C1 alkyl; and the C1 alkyl is further substitut-ed with optionally substituted C3-6 heterocycloalkyl, wherein the heterocycloalkyl is saturated and the het-erocycloalkyl is attached to the rest of the molecule via the N atom on the ring.
Embodiment 7. The compound of any one of embodiments 3 to 4, or a stereoisomer thereof, a phar-maceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt thereof, wherein R1 is C1 alkyl; and the C1 alkyl is further substitut-ed with optionally substituted C3-6 heterocycloalkyl, wherein the heterocycloalkyl is saturated and the het-erocycloalkyl is attached to the rest of the molecule via the N atom on the ring. and the C3-6 heterocycloal-kyl is 5 to 7 menbered heterocycloalkyl and has l to 2 heteroatoms.
Embodiment 8. The compound of any one of embodiments 3 to 7, or a stereoisomer thereof, a phar-maceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt thereof, wherein
R1 is selected from the group consisting of
Embodiment 9. The compound according to any one of embodiments 3 to 8, or a stereoisomer thereof, a pharmaceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt thereof, wherein R1 is C1 alkyl substituted by C6 heterocy-cloalkyl, optionally wherein the C6 heterocycloalkyl is piperazine or morpholine.
Embodiment 10. The compound of any one of embodiments 1 to 9, or a stereoisomer thereof, a phar-maceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt thereof, wherein R4 and R5 combine with the atoms to which they are attached to form a ring, wherein the ring is optionally substituted 3 to 10-membered cycloalkyl; pref-erably, is optionally substituted 3 to 6-membered cycloalkyl; more preferably, optionally substituted cyclo-propyl.
Embodiment 11. The compound of any one of embodiments 1 to 9, or a stereoisomer thereof, a phar-maceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt thereof, wherein
is selected from the group consisting of:
is
Embodiment 12. The compound of any one of embodiments 1 to 11, or a stereoisomer thereof, a pharmaceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt thereof, wherein
A is optionally substituted 5 to 6-membered heteroarylene, the heteroatom is independently selected from one or two of N, O and S, and the number of the heteroatom is independently 1 to 3
preferably, A is optionally substituted 5-membered heteroarylene, the heteroatom is independently se-lected from N, O and S, and the number of the heteroatom is independently 1 to 2; preferably, one of the heteroatom is N, and the other, if present, is O or S;
more preferably, A is selected from the group consisting of
Embodiment 13. The compound of any one of embodiments 1 to 12, or a stereoisomer thereof, a pharmaceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt thereof, wherein
X1 is C, and/or X2 is -CRa-, and/or X3 is -CRb, and/or X4 is -CRc-, and/or X5 is -C-.
Embodiment 14. The compound of any one of embodiments 1 to 13, or the stereoisomer thereof, the pharmaceutically acceptable salt thereof, the metabolite thereof, the prodrug thereof, the solvate thereof or the solvate of the pharmaceutically acceptable salt thereof, wherein, the compound is selected from any one of the compounds in Table 1.
Embodiment 15. The compound of any one of embodiments 1 to 14, or the stereoisomer thereof, the pharmaceutically acceptable salt thereof, the metabolite thereof, the prodrug thereof, the solvate thereof or the solvate of the pharmaceutically acceptable salt thereof, wherein, the compound is selected from com-pounds of Examples 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 27, 28, 29, 30, 31, 32, 33, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 84, 86, 87, 88, and 90 in Table 1.
Embodiment 16. A pharmaceutical composition comprising: the compound of any one of embodi-ments 1 to 15, or the stereoisomer thereof, the pharmaceutically acceptable salt thereof, the metabolite thereof, the prodrug thereof, the solvate thereof or the solvate of the pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
Embodiment 17. A use of the compound of any one of embodiments 1 to 15, or the stereoisomer thereof, the pharmaceutically acceptable salt thereof, the metabolite thereof, the prodrug thereof, the solv-ate thereof or the solvate of the pharmaceutically acceptable salt thereof, or the pharmaceutical composi-tion according to embodiment 16, or the pharmaceutical composition comprising the same in the prepara-tion of a medicament used for the treatment of cancer.
Embodiment 18. The use of the compound of embodiment 17, wherein the cancer is selected from the group consisting of small cell lung cancer (SCLC) , colorectal cancer (CRC) , pancreatic cancer, and chol-angiocarcinoma, pancreatic cancer, appendiceal cancer, small bowel cancer, colorectal cancer, ampullary cancer, non-small cell lung cancer, cervical cancer, lung cancer, endometrial cancer, acute myeloid leuke-mia, gastrointestinal neuroendocrine tumor, uterine endometrioid carcinoma, oesophagogastric cancer, bladder cancer, ovarian cancer, melanoma, multiple myeloma, thyroid gland adenocarcinoma, a myelodys-plastic syndrome, and squamous cell lung carcinoma, or combinations thereof.
Embodiment 19. The use according to any one of embodiments 17 to 18, wherein the cancer is se-lected from the group consisting of non-small cell lung cancer, small cell lung cancer, colorectal cancer, pancreatic cancer, and cholangiocarcinoma.
Embodiment 20. The compound according to any one of embodiments 1 to 15, or the stereoisomer thereof, the pharmaceutically acceptable salt thereof, the metabolite thereof, the prodrug thereof, the solv-ate thereof or the solvate of the pharmaceutically acceptable salt thereof, or the pharmaceutical composi-tion comprising the same for use in treating cancer.
Embodiment 21. The compound for use according to embodiment 20, wherein the cancer is as defined in embodiment 18.
Embodiment 22. The compound for use according to any one of embodiments 20 to 21, wherein the cancer is as defined in embodiment 19.
Embodiment 23. A method for treating cancer in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of the compound of any one of the preced-ing embodiments, or the stereoisomer thereof, the pharmaceutically acceptable salt thereof, the metabolite thereof, the prodrug thereof, the solvate thereof or the solvate of the pharmaceutically acceptable salt thereof, or the pharmaceutical composition according to embodiment 16, or the pharmaceutical composi-tion comprising the same.
Embodiment 24. The method according to embodiment 23, wherein the cancer is as defined in em-bodiment 18.
Embodiment 25. The method according to any one of embodiments 23 to 24, wherein the cancer is as defined in embodiment 19.
TERM DEFINITION
Given below are definitions of terms used in this application. Any term not defined herein takes the normal meaning as the skilled person would understand the term.
As used herein, the term “pharmaceutically acceptable salts” refers to salts prepared from pharmaceu-tically acceptable non-toxic bases or acids. When the compound of the present invention is acidic, its cor-responding salt can be conveniently prepared from pharmaceutically acceptable non-toxic bases, including inorganic bases and organic bases. When the compound of the present invention is basic, its corresponding salt can be conveniently prepared from pharmaceutically acceptable non-toxic acids, including inorganic and organic acids.
As used herein, the term “solvate” refers to solvent addition forms that contain either stoichiometric or non-stoichiometric amounts of solvent.
As used herein, excipient that is useful in preparing a pharmaceutical composition that is generally safe, nontoxic and neither biologically nor otherwise undesirable, and includes carrier that is acceptable for veterinary use as well as human pharmaceutical use.
A “pharmaceutically acceptable carrier” as used herein includes both one and more than one such car-rier. The term “pharmaceutically acceptable carrier” also encompasses “pharmaceutically acceptable ex-cipient” and “pharmaceutically acceptable diluent” . The particular carrier used in the pharmaceutical compositions of the present disclosure will depend upon the means and purpose for which the compounds of the present disclosure are being applied.
As used herein, unless otherwise specified, the term “optionally substituted” refers to unsubtituted or being substituted with one or more (such as 1, 2, 3 or 4) substituents. Suitable substituents for each group can be found herein. Unless otherwise specified, exemplary substituents can be selected from the group consisting of D, halogen, -CN, -NO2, Ri, -C1-4 alkylene-Ri, -C2-4 alkenylene-Ri, -C2-4 alkynylene-Ri, -ORi, -OC (O) Ri, -C (O) Ri, -CO2Ri, -CONRiRii, -OC (O) NNRiRii, -NRiiC (O) Ri, -NRi-C (O) NRiiRiii, -NRiiC (O) 2Ri, -NH-C (NH2) =NH, -NRiC (NH2) =NH, -NH-C (NH2) =NRi, -S (O) Ri, -S (O) 2Ri, -S (O) 2NRiRii, -S (O) (=N-Ri) Rii (e.g., ) , -NRiS (O) 2Rii; wherein Ri, Rii and Riii are each independently selected from the group consisting of H, C1-6 alkyl, -C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C3-10 cycloalkenyl, 3 to 10-membered heterocycloalkyl, 3 to 10-membered heterocycloalkenyl, C6-10 aryl, 5 to 10-membered het-eroaryl, -C1-4 alkylene-C3-10 cycloalkyl, -C1-4 alkylene-C3-10 cycloalkenyl, -C1-4 alkylene-3 to 10-membered heterocycloalkyl, -C1-4 alkylene-3 to 10-membered heterocycloalkenyl, -C1-4 alkylene-C6-10 aryl, and -C1-4 alkylene-5 to 10-membered heteroaryl; or Ri and Rii, Rii and Riii together with the atom to which they are attached to form C3-10 cycloalkenyl, 3 to 10-membered heterocycloalkyl, 3 to 10-membered heterocycloal-kenyl, C6-10 aryl, 5 to 10-membered heteroaryl; wherein the alkylene, alkenylene, alkynylene, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, heterocycloalkyl, heterocycloalkenyl, aryl and heteroaryl can be unsub-tituted or one or more (such as 1, 2, 3 or 4) or all of H atom in the group being substituted with substituents selected from the group consisting of D, halogen, -CN, -OH, C1-6 alkyl, C1-6 haloalkyl, C1-6 deuteroalkyl, C1-6 hydroxyalkyl, C1-6 aminoalkyl, amino, alkylamino, -C (O) C1-6 alkyl, -OC1-6 alkyl, -OC (O) C1-6 alkyl, -S (O) C1-6 alkyl, -S (O) 2C1-6 alkyl, -COOC1-6 alkyl, oxo (=O) , C1-6 alkylidene, C1-6 haloalkylidene, C1-6 deuteroalkylidene, C1-6 hydroxyalkylidene, C1-6 aminoalkylidene, C3-6 cycloalkyl optionally substituted with one or more of D, halogen, -CN, -OH, C1-6 alkyl, C1-6 haloalkyl, C1-6 deuteroalkyl, C1-6 hydroxyalkyl, and C1-6 aminoalkyl, 3 to 6-membered heterocycloalkyl optionally substituted with one or more of D, hal-ogen, -CN, -OH, C1-6 alkyl, C1-6 haloalkyl, C1-6 deuteroalkyl, C1-6 hydroxyalkyl, and C1-6 aminoalkyl, 3 to 6-membered heterocycloalkenyl optionally substituted with one or more of D, halogen, -CN, -OH, C1-6 al-kyl, C1-6 haloalkyl, C1-6 deuteroalkyl, C1-6 hydroxyalkyl, and C1-6 aminoalkyl, phenyl, 5 to 6-membered het-eroaryl optionally substituted with one or more of D, halogen, -CN, -OH, C1-6 alkyl, C1-6 haloalkyl, C1-6 deuteroalkyl, C1-6 hydroxyalkyl, and C1-6 aminoalkyl. In some embodiments, said “optionally substituted” means that the group is substituted by one or more substituents selected from the group consisting of deu-terium, halogen, amino, nitro, cyano, hydroxyl, oxo (=O) , thio (=S) , C1-C4 alkyl, C1-C4 haloalkyl, C1-C4 hydroxyalkyl, C1-C4 alkoxy, C1-C4 haloalkoxy, C2-C4 alkenyl, C2-C4 alkyne, C3-C6 cycloalkyl, 3 to 6-membered heterocycloalkyl.
In addition, if an optionally substituted group can be further substituted means in addition to being substituted by the above-mentioned substituents for “optionally substituted” , one or more hydrogen atoms in the group may also be substituted by other substituents specified (such as substituents for further substi-tution) .
As used herein, the term “alkyl” refers to a saturated aliphatic hydrocarbon group including straight chain and branched chain groups having the number of carbon atoms designated (i.e., C1-20 means 1 to 20 carbon atoms) . Preferably an alkyl group is an alkyl having 1 to 12 i.e., C1-12 alkyl. Sometimes preferably 1 to 6 i.e., C1-6 alkyl, sometimes more preferably 1 to 4, carbon atoms i.e., C1-4 alkyl. Representative exam-ples include, but are not limited to methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1, 1-dimethyl propyl, 1, 2-dimethyl propyl, 2, 2-dimethyl propyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1, 1, 2-trimethylpropyl, 1, 1-dimethylbutyl, 1, 2-dimethylbutyl, 2, 2-dimethylbutyl, 1, 3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2, 3-dimethylbutyl, n-heptyl, 2-methylhexyl, 3-methylhexyl, 4-methylhexyl, 5-methylhexyl, 2, 3-dimethylpentyl, 2, 4-dimethylpentyl, 2, 2-dimethylpentyl, 3, 3-dimethylpentyl, 2-ethylpentyl, 3-ethylpentyl, n-octyl, 2, 3-dimethylhexyl, 2, 4-dimethylhexyl, 2, 5-dimethylhexyl, 2, 2-dimethylhexyl, 3, 3-dimethylhexyl, 4.4-dimethylhexyl, 2-ethylhexyl, 3-ethylhexyl, 4-ethylhexyl, 2-methyl-2-ethylpentyl, 2-methy1-3-ethylpentyl, n-nonyl, 2-methy1-2-ethylhexyl, 2-methy1-3-ethylhexyl, 2, 2-diethylpentyl, n-decyl, 3, 3-diethylhexyl, 2, 2-diethylhexyl, and the isomers of branched chain thereof. More preferably an alkyl group is a lower alkyl having 1 to 6 carbon atoms. Representative examples include, but are not lim-ited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1, 1-dimethylpropyl, 1, 2-dimethylpropyl, 2, 2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1, 1, 2-trimethylpropyl, 1, 1-dimethylbutyl, 1.2-dimethylbutyl, 2, 2-dimethylbutyl, 1, 3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2, 3-dimethylbutyl, etc. The alkyl group can be substituted or unsubstituted. When substituted, the substit-uent group (s) can be substituted at any available connection point, preferably the substituent group (s) is one or more substituents independently selected from the group consisting of alkyl, halogen, alkoxy, alkenyl, alkynyl, alkylsulfo, alkylamino, thiol, hydroxy, nitro, cyano, amino, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxyl, heterocylic, cycloalkylthio, heterocylic alkylthio and oxo group.
The term “alkylene” refers to a saturated linear or branched aliphatic hydrocarbon group, wherein having 2 residues derived by removing two hydrogen atoms from the same carbon atom of the parent al-kane or two different carbon atoms. The straight or branched chain group containing 1 to 20 carbon atoms, preferably has 1 to 12 carbon atoms, more preferably 1 to 6 carbon atoms. Non-limiting examples of al-kylene groups include, but are not limited to, methylene (-CH2-) , 1, 1-ethylene (-CH (CH3) -) , 1, 2-ethylene (-CH2CH2) -, 1, 1-propylene (-CH (CH2CH3) -) , 1, 2-propylene (-CH2CH (CH3) -) , 1, 3-propylene (-CH2CH2CH2-) , 1, 4-butylidene (-CH2CH2CH2CH2-) etc. The alkylene group can be substituted or unsub-stituted. When substituted, the substituent group (s) is preferably one or more, sometimes preferably one to five, sometimes more preferably one to three, group (s) independently selected from the group consisting of selected from alkyl, alkenyl, alkynyl, alkoxy, alkylsulfo, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloalkoxyl, heterocylic alkoxyl, cycloalkylthio and het-erocylic alkylthio.
As used herein, the term “alkenyl” refers to an alkyl defined as above that has at least two carbon at-oms and at least one carbon-carbon double bond, for example, vinyl, 1-propenyl, 2-propenyl, 1-2-, or 3-butenyl, etc., preferably C2-20 alkenyl, more preferably C2-12 alkenyl, and most preferably C2-6 alkenyl. The alkenyl group can be substituted or unsubstituted. When substituted, the substituent group (s) is pref-erably one or more, sometimes preferably one to five, sometimes more preferably one to three, group (s) independently selected from the group consisting of alkyl, halogen, alkoxy, alkenyl, alkynyl, alkylsulfo, alkylamino, thiol, hydroxy, nitro, cyano, amino, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloal-koxyl, heterocylic, cycloalkylthio, heterocylic alkylthio and oxo group.
The term “alkenylene” refers to an alkylene defined as above that has at least two carbon atomsand at least one carbon-carbon double bond, preferably C2-20 alkenylene, morepreferably C2-12 alkenylene, and most preferably C2-6 alkenylene. Non-limiting examples of alkenylene groups include, but are not limited to, -CH=CH-, -CH=CHCH2, -CH=CHCH2CH2, -CH2CH=CHCH2-etc. The alkenylene group can be sub-stituted or unsubstituted. When substituted, the substituent group (s) is preferably one or more, sometimes preferably one to five, sometimes more preferably one to three, group (s) independently selected from the group consisting of selected from alkyl, alkenyl, alkynyl, alkoxy, alkylsulfo, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloalkoxyl, heterocylic alkoxyl, cycloalkylthio and heterocylic alkylthio.
As used herein, the term “alkynyl” refers to an alkyl defined as above that has at least two carbon at-oms and at least one carbon-carbon triple bond, for example, ethynyl, 1-propynyl, 2-propynyl, 1-2-, or 3-butynyl etc., preferably C2-20 alkynyl, more preferably C2-12 alkynyl, and most preferably C2-6 alkynyl. The alkynyl group can be substituted or unsubstituted. When substituted, the substituent group (s) is pref-erably one or more, sometimes preferably one to five, sometimes more preferably one to three, group (s) independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkylsulfo, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloalkoxyl, hetero-cylic alkoxyl, cycloalkylthio and heterocylic alkylthio.
As used herein, the term “aryl” refers to a 6 to 14-membered (or C6-14) all-carbon monocyclic ring or a polycyclic fused ring (a "fused" ring system means that each ring in the system shares an adjacent pair of carbon atoms with another ring in the system) group, and has a completely conjugated pi-electron system. Preferably aryl is 6 to 10-membered (or C6-10) , such as phenyl and naphthyl, most preferably phenyl. The aryl can be fused to the ring of heteroaryl, heterocyclyl or cycloalkyl, wherein the ring bound to parent structure is aryl. Representative examples include, but are not limited to, the following substituents:
The aryl group can be substituted or unsubstituted. When substituted, the substituent group (s) is pref-erably one or more, sometimes preferably one to five, sometimes more preferably one to three, substituents independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkylsulfo, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloalkoxyl, hetero-cylicalkoxyl, cycloalkylthio and heterocylic alkylthio.
The term “arylene” is a divalent group which is connected to the rest of the molecule through two sin-gle bonds, and the rest definitions are the same as the term “aryl” .
As used herein, the term “heteroaryl” refers to an aryl system having 1 to 4 heteroatoms selected from the group consisting of O, S and N as ring atoms and having 5 to 14 ring atoms (referred as 5 to 14-membered) . Preferably a heteroaryl is 5 to 10-membered, more preferably 5 or 6-membered, for exam-ple, thiadiazolyl, pyrazolyl, oxazolyl, oxadiazolyl, imidazolyl, triazolyl, thiazolyl, thiazolylfuryl, thienyl, pyridyl, pyrrolyl, N-alkyl pyrrolyl, pyrimidinyl, pyrazinyl, imidazolyl, tetrazolyl, and the like. The het-eroaryl can be fused with the ring of an aryl, heterocyclyl or cycloalkyl, wherein the ring bound to parent structure is heteroaryl. Representative examples include, but are not limited to, the following substituents:
The heteroaryl group can be substituted or unsubstituted. When substituted, the substituent group (s) is preferably one or more, sometimes preferably one to five, sometimes more preferably one to three, sub-stituents independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkylsulfo, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloal-koxyl, heterocylic alkoxyl, cycloalkylthio, heterocylic alkylthio and -NReRf.
The term “heteroarylene” is a divalent group which is connected to the rest of the molecule through two single bonds, and the rest definitions are the same as the term “heteroaryl” .
The term "bicyclic" is intended to include spiro, fused-ring or bridged-ring.
As used herein, the term “spiro” refers to two rings that shares one ring atom (e.g., carbon) .
As used herein, the term “fused” refers to two rings that share two adjacent ring atoms with one an-other.
As used herein, the term “bridged” refers to two rings that share three adjacent ring atoms with one another.
As used herein, the term “cycloalkyl” refers to a saturated and/or partially unsaturated monocyclic or polycyclic hydrocarbon group having 3 to 20 carbon atoms (refered as C3-20 or 3 to 20 membered) , prefera-bly 3 to 12 carbon atoms (refered as C3-12 or 3 to 12 membered) , more preferably 3 to 10 carbon atoms (refered as C3-10 or 3 to 10 membered) , and most preferably 3 to 8 carbon atoms (refered as C3-8 or 3 to 8 membered) or 3 to 6 carbon atoms (refered as C3-6 or 3 to 6 membered) . In some embodiments, cycloalkyl is monocyclic cycloalkyl. Representative examples of monocyclic cycloalkyls include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cy-cloheptyl, cycloheptatrienyl, cyclooctyl, etc. In some embodiments, cycloalkyl is polycyclic cycloalkyl (such as bicyclic cycloalkyl) . In some embodiments, cycloalkyl is a spiro cycloalkyl, a fused cycloalkyl or a bridged cycloalkyl. Polycyclic cycloalkyl includes a cycloalkyl having a spiro ring, fused ring or bridged ring. The cycloalkyl can be fused to the ring of an aryl, heteroaryl or heterocyclic alkyl, wherein the ring bound to the parent structure is cycloalkyl. Representative examples include, but are not limited to inda-nylacetic, tetrahydronaphthalene, benzocycloheptyl and so on. The cycloalkyl is optionally substituted or unsubstituted. When substituted, the substituent group (s) is preferably one or more, sometimes preferably one to five, sometimes more preferably one to three, substituents independently selected from the group consisting of alkyl, halogen, alkoxy, alkenyl, alkynyl, alkylsulfo, alkylamino, thiol, hydroxy, nitro, cyano, amino, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloalkoxyl, heterocylic, cyeloalkylthio, hetero-cylic alkylthio and oxo group.
The term “cycloalkylene” is a divalent group which is connected to the rest of the molecule through two single bonds, and the rest definitions are the same as the term “cycloalkyl” .
“Spiro Cycloalkyl” refers to a 5 to 20-membered polycyclic group with rings connected through one common carbon atom (called a spiro atom) , wherein one or more rings can contain one or more double bonds, but none of the rings has a completely conjugated pi-electron system. Preferably a spiro cycloalkyl is 6 to 14-membered, and more preferably 7 to 10-membered. According to the number of common spiro atoms, a spiro cycloalkyl is divided into mono-spiro cycloalkyl, di-spiro cycloalkyl, or poly-spiro cycloal-kyl, and preferably refers to a mono-spiro cycloalkyl or di-spiro cycloalkyl, more preferably 4-membered/4-membered, 4-membered/5-membered, 4-membered/6-membered, 5-membered/5-membered, or 5-membered/6-membered mono-spiro cycloalkyl. Representative examples of spiro cycloalkyl include, but are not limited to the following substituents:
“Fused Cycloalkyl” refers to a 5 to 20-membered polycyclic hydrocarbon group, wherein each ring in the system shares an adjacent pair of carbon atoms with another ring, wherein one or more rings can con-tain one or more double bonds, but none of the rings has a completely conjugated pi-electron system. Pref-erably, a fused cycloalkyl group is 6 to 14-membered, more preferably 7 to 10-membered. According to the number of membered rings, fused cycloalkyl is divided into bicyclic, tricyclic, tetracyclic or polycyclic fused cycloalkyl, and preferably refers to a bicyclic or tricyclic fused cycloalkyl, more preferably 5-membered/5-membered, or 5-membered/6-membered bicyclic fused cycloalkyl. Representative exam-ples of fused cycloalkyls include, but are not limited to, the following substituents:
“Bridged Cycloalkyl” refers to a 5 to 20-membered polycyclic hydrocarbon group. wherein every two rings in the system share two disconnected carbon atoms. The rings can have one or more double bonds, but have no completely conjugated pi-electron system. Preferably, a bridged cycloalkyl is 6 to 14-membered, and more preferably 7 to 10-membered. According to the number of membered rings, bridged cycloalkyl is divided into bicyclic, tricyclic, tetracyclic or polycyclic bridged cycloalkyl, and pref-erably refers to a bicyclic, tricyclic or tetracyclic bridged cycloalkyl, more preferably a bicyclic or tricyclic bridged cycloalkyl. Representative examples of bridged cycloalkyls include, but are not limited to, the fol-lowing substituents:
As used herein, the term “heterocyclyl” or “heterocycloalkyl” refers to a 3 to 20-membered saturated and/or partially unsaturated monocyclic or polycyclic hydrocarbon group having one or more, sometimes preferably one to five, sometimes more preferably one to three carbon ring atoms being replaced with het-eroatoms selected from the group consisting of N, O, and S (O) m (wherein m is 0, l, or 2) as ring atoms, but excluding -O-O-, -O-S-or -S-S-in the ring, the remaining ring atoms being C. Preferably, heterocyclyl is a 3 to 12-membered ring having 1 to 4 heteroatoms (also referred as for example 3-12 membered heterocy-clyl or heterocycloalkyl, or C2-11 heterocyclyl or heterocycloalkyl) ; more preferably a 4 to 12-membered ring having l to 3 heteroatoms (also referred as for example 4-12 membered heterocyclyl or heterocycloal-kyl, or C3-10 heterocyclyl or heterocycloalkyl) ; more preferably a 3 to 10-membered ring having 1 to 3 het-eroatoms (also referred as for example 3-10 membered heterocyclyl or heterocycloalkyl, or C2-9 heterocy-clyl or heterocycloalkyl) ; more preferably a 4 to 8-membered ring having l to 3 heteroatoms (i.e., 4-8 membered heterocyclyl or heterocycloalkyl, or C2-7 heterocyclyl or heterocycloalkyl) ; most preferably a 5 to 7-membered ring having l to 2 heteroatoms (also referred as for example 5-7 membered heterocyclyl or heterocycloalkyl, or C3-6 heterocyclyl or heterocycloalkyl) or a 5 to 6-membered ring having l to 2 het-eroatoms (also referred as for example 5-6 membered heterocyclyl or heterocycloalkyl, or C3-5 heterocyclyl or heterocycloalkyl) . In some embodiments, heterocycloalkyl/heterocyclyl is monocyclic heterocycloal-kyl/heterocyclyl. Representative examples of monocyclic heterocyclyls or heterocycloalkyl include, but arenot limited to, oxetanyl, azabutyl, pyrrolidyl, piperidyl, piperazinyl, morpholinyl, sulfo-morpholinyl, homopiperazinyl, and so on. In some embodiments, heterocycloalkyl/heterocyclyl is polycyclic heterocy-cloalkyl/heterocyclyl (such as bicyclic heterocycloalkyl/heterocyclyl) . In some embodiments, heterocyclo-alkyl/heterocyclyl is a spiro heterocycloalkyl/heterocyclyl, a fused heterocycloalkyl/heterocyclyl or a bridged heterocycloalkyl/heterocyclyl. Polycyclic heterocyclyl or heterocycloalkyl includes the heterocy-clyl having a spiro ring, fused ring or bridged ring. The “heterocyclyl” or “heterocycloalkyl” can be at-tached to the rest of the molecule via the N or C atom on the ring. In some embodiments, the “heterocyclyl” or “heterocycloalkyl” is attached to the rest of the molecule via the N atom on the ring. In some embodi-ments, specifically in the definition of R1, the “heterocyclyl” or “heterocycloalkyl” is attached to the rest of the molecule via the C atom on the ring. In some embodiments, the “heterocyclyl” or “heterocycloalkyl” has a structure ofwherein is a heteroatom or a carbon atom. Examples of such heterocyclyl groups areWhen the heter-ocyclyl has substituents, the substituents may be attached to any atom in the ring, provided that a stable chemical structure results.
The term “heterocycloalkylene” is a divalent group which is connected to the rest of the molecule by two single bonds, and the rest definitions are the same as the term “heterocycloalkyl” .
“Spiro heterocyclyl” refers to a 5 to 20-membered polycyclic heterocyclyl with rings connected through one common carbon atom (called a spiro atom) , wherein said rings have one or more, sometimes preferably one to five, sometimes more preferably oneto three, heteroatoms selected from the group con-sisting of N, O, and S (O) m. (wherein m is 0, 1 or 2) as ring atoms, the remaining ring atoms being C, wherein one or more rings can contain one or more double bonds, but none of the rings has a completely conjugated pi-electron system. Preferably a spiro heterocyclyl is 6 to 14-membered, and more preferably 7 to 10-membered. According to the number of common spiroatoms, spiro heterocyclyl is divided into mono-spiro heterocyclyl, di-spiro heterocyclyl, or poly-spiro heterocyclyl, and preferably refers to mono-spiro heterocyclyl or di-spiro heterocyclyl, more preferably 4-membered/4-membered, 4-membered/5-membered, 4-membered/6-membered, 5-membered/5-membered, or 5-membered/6-membered mono-spiro heterocyclyl. Representative examples of spiro heterocyclyl include, but are not limited to the following substituents:
“Fused Heterocyclyl" refers to a 5 to 20-membered polycyclic heterocyclyl group, wherein each ring in the system shares an adjacent pair of carbon atoms with the other ring, wherein one or more rings can contain one or more double bonds, but none of the rings has a completely conjugated pi-electron system, and wherein said rings have one or more, sometimes preferably one to five, sometimes more preferably one to three, heteroatoms selected from the group consisting of N, O, and S (O) p, (wherein p is 0, 1, or 2) as ring atoms, the remaining ring atoms being C. Preferably a fused heterocyclyl is 6 to 14-membered, and more preferably 7 to 10-membered. According to the number of membered rings, fused heterocyclyl is di-vided into bicyclic, tricyclic, tetracyclic or polycyclic fused heterocyclyl, preferably refers to bicyclic or tricyclic fused heterocyclyl, more preferably 5-membered/5-membered, or 5-membered/6-membered bicy-clic fused heterocyclyl. Representative examples of fused heterocyclyl include, but are not limited to, the following substituents:
As used herein, the ring of said heterocyclyl can be fused to the ring of an aryl, heteroaryl orcycloal-kyl, wherein the ringbound to the parent structure is heterocyclyl. Representative examples include, but are not limited to the following substituents:
The heterocyclyl is optionally substituted or unsubstituted. When substituted, the substituent group (s) is preferably one or more, sometimes preferably one to five, sometimes more preferably one to three, group (s) independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkylsulfo, alkylamino, halogenthiol, hydroxy, nitro, cyano, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloal-koxyl, heterocylic alkoxyl, cycloalkylthio, heterocylic alkylthio.
“Bridged Heterocyclyl” refers to a 5 to l4-membered polycyclic heterocyclic alkyl group, wherein every two rings in the system share two disconnected atoms, the rings can have one or more double bonds, but have no completely conjugated pi-electron system, and the rings have one or more heteroatoms select-ed from the group consisting of N, O, and S (O) m (wherein m is 0, 1, or 2) as ring atoms, the remaining ring atoms being C. Preferably a bridged heterocyclyl is 6 to 14-membered, and more preferably 7 to 10-membered. According to the number of membered rings, bridged heterocyclyl is divided into bicyclic, tricyclic, tetracyclic or polycyclic bridged heterocyclyl, and preferably refers to bicyclic, tricyclic or tetra-cyclic bridged heterocyclyl, more preferably bicyclic or tricyclic bridged heterocyclyl. Representative ex-amples of bridged heterocyclyl include, but are not limited to, the following substituents:
As used herein, the term “C1-6 haloalkoxyl” refers to an alkoxyl group in which one or more hydrogen atoms are replaced by a halogen, for example, -OCF3.
As used herein, the term “haloalkyl” refers to an alkyl group in which one or more hydrogen atoms are replaced by a halogen, for example, -CF3.
As used herein, the term “C1-6 alkoxyC1-6 alkyl” refers to a C1-6 alkyl group in which one or more hy-drogen atoms are replaced by C1-6 alkoxy or a C1-6 alkoxy group in which one or more hydrogen atoms are replaced by C1-6 alkyl.
As used herein, the term “alkoxy” refers to a straight or branched alkoxy group containing the speci-fied number of carbon atoms. For example, C1-6 alkoxy means a straight or branched alkoxy group con-taining at least 1, and at most 6, carbon atoms. Examples of “alkoxy” as used herein include, but not lim-ited to, methoxy, ethoxy, prop-1-oxy, pro-2-oxy, pentoxy, hexyloxy, and the like.
As used herein, the term “aminoalkyl” refers to an alkyl moiety substituted by one or more amino moieties. Such as -CH2 (NH2) .
As used herein, the term “alkylamino” refers to an amino group substituted by one or more alkyl such as -NH (C1-6 alkyl) or -N (C1-6 alkyl) 2. Examples of alkylamino include, but not limited to, -NH (CH3) , -N (CH3) 2.
As used herein, the term “halogen” or “halo” refers to fluorine (F) , chlorine (Cl) , bromine (Br) or io-dine (I) .
As used herein, the term "oxo" means that =O, oxygen atoms replace two hydrogens on the same carbon atom, that is, carbonyl groups replace methylene groups.
As used herein, the term “Bond” refers to a covalent bond using a sign of “-” . In some embodi-ments, represents a double bond or single bond.
DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENT
Methods of Synthesis
The compounds of the present invention can be prepared by any conventional means. Suitable pro-cesses for synthesizing these compounds as well as their starting materials are provided in the schemes be-low and in the examples. All substituents 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 procedure for preparing compounds as formula (II) are shown in Scheme 1:
Scheme 1
Scheme 1 illustrates the preparation of compound of formula (II) . At first the coupling of compounds of formula (1) can be obtained through standard Suzuki coupling conditions (for example Pd (dppf) Cl2 and K2CO3) to provide compound of formula (2) . Compound of formula (2) was coupled with compound of formula (3) under Suzuki coupling conditions (for example Pd (dppf) Cl2 and K2CO3) to give compound of formula (4) , which was converted to compound of formula (5) in the presence of iodization reagents, such I2 or NIS. Deprotection of compound of formula (5) to provide compound of formula (6) , such as hydroly-sis of the ester with LiOH·OH, or with TBAF deprotecting silicon protective group on hydroxyl group. Compound of formula (8) was attained by a routine coupling condition between compound of formula (6) and amine of formula (7) with coupling reagents, such as HATU, EDCI/HOBt or PyBOP etc, in the pres-ence of an organic base, such as Et3N, DIPEA or pyridine, which was hydrolyzed to provide acid of for-mula (9) under a base, such as LiOH·OH. The intramolecular coupling of compound of formula (9) with coupling reagents, such as EDCI/HOBt and HATU, to give compound of formula (10) . Then the coupling of compounds of formula (10) can be obtained through standard Suzuki coupling conditions (for example Pd (dppf) Cl2 and K2CO3) to provide compound of formula (11) . Compound of formula (11) was coupled with compound of formula (12) under Suzuki coupling conditions (for example Pd (dppf) Cl2 and K2CO3) to give compound of formula (13) , which was converted to compound of formula (14) with alkylate reagents, such iodomethane or iodoethane, in the presence of an inorganic base, such as Cs2CO3. Deprotection of compound (14) can afford compound of formula (15) in the presence of an acid, such as TFA. Compound of formula (II) can be obtained by a coupling reaction between acid of formula (16) and compound of for-mula (15) with coupling reagents, such as HATU, EDCI/HOBy, PyBOP or COMU, in the presence of a base, such as TEA, DIPEA, pyridine or 2, 6-lutidine.
Preparation of Intermediate
Intermediate A: 3- (5-bromo-1H-indol-3-yl) -2, 2-dimethylpropyl acetate
Step 1. Preparation of (5-bromo-1H-indol-3-yl) methanol
To a solution of 5-bromo-1H-indole-3-carbaldehyde (100.0 g, 0.45 mol) in premixed solvents THF/MeOH (1 L, V: V = 4: 1) was stirred at 0℃ for 5 mins, then NaBH4 (20.0g, 0.53 mol) was added into the above solution at room temperature. The resulting mixture was stirred at 0℃ for 2h. The reaction was monitored by LCMS. After completion, the reaction mixture was quenched with H2O (300 mL) dropwise and diluted with EtOAc (100 mL) , then the resulting mixture was extracted with EtOAc (100 mL x 4) . The organic layer was separated, dried over anhydrous Na2SO4 and concentrated to give the crude product (5-bromo-1H-indol-3-yl) methanol (107.5g, 98%) as a white solid. The crude product was proceeded to the next reaction without purification.
LCMS (ESI) calcd. for C9H8BrNO [M-H] + m/z 224.07, found: 224.10
Step 2. Preparation of methyl 3- (5-bromo-1H-indol-3-yl) -2, 2-dimethylpropanoate
To a solution of ( (1-methoxy-2-methylprop-1-en-1-yl) oxy) trimethylsilane (157.3 g, 0.90 mol) in dry THF (150 mL) under N2 atmosphere, the reaction mixture was stirred at -40℃ for 5 mins, then (5-bromo-1H-indol-3-yl) methanol (107.5 g, 0.48 mol) in dry THF (50 mL) was added under N2 atmos-phere, After 10 mins, TMSOTf (90.0 g, 0.40 mol) in dry THF (50 mL) was added into the above solution by dropwise at nitrogen atmosphere. The reaction mixture was stirred at this temperature for 2h. The reac-tion was monitored by LCMS. After completion, the reaction mixture was warmed to room temperature and diluted with NaHCO3 solution (100 mL) , then the resulting mixture was extracted with EtOAc (100 mL x 4) . The organic layer was separated, dried over anhydrous Na2SO4 and concentrated to give the crude product, which was further purified by silica gel chromatography eluting with petroleum ether/EtOAc (from 0%to 20%) to obtain methyl 3- (5-bromo-1H-indol-3-yl) -2, 2-dimethylpropanoate (98g, 70%) as a yellow solid.
LCMS (ESI) calcd. for C14H16BrNO2 [M+H] + m/z 310.19, found: 310.20
Step 3. Preparation of 3- (5-bromo-1H-indol-3-yl) -2, 2-dimethylpropan-1-ol
To a solution of methyl 3- (5-bromo-1H-indol-3-yl) -2, 2-dimethylpropanoate (98g, 0.32 mol) and LiBH4 (40g, 1.82 mol) was added in dry THF (300 mL) at room temperature, the reaction mixture was stirred at 80℃ for 12h. The reaction was monitored by LCMS. After completion, the reaction mixture was cooled to room temperature, quenched with NH4Cl solution (150 mL) and diluted with EtOAc (100 mL) , then the resulting mixture was extracted with EtOAc (100 mL x 4) . The organic layer was separated, dried over anhydrous Na2SO4 and concentrated to give the crude product 3- (5-bromo-1H-indol-3-yl) -2, 2-dimethylpropan-1-ol (91.3g, 99%) as a light-yellow solid. The crude prod-uct was proceeded to the next reaction without purification.
LCMS (ESI) calcd. for C13H16BrNO [M+H] + m/z 282.18, found: 282.40
Step 4. Preparation of 3- (5-bromo-1H-indol-3-yl) -2, 2-dimethylpropyl acetate
To a solution of 3- (5-bromo-1H-indol-3-yl) -2, 2-dimethylpropan-1-ol (91.3g, 0.31 mol) in dry DCM (150 mL) at room temperature under N2 atmosphere, then DIPEA (62.7g, 0.49 mol) , DMAP (4.0g, 0.03 mol) and AC2O (24.0 g, 0.31 mol) was added into the above solution under N2 atmosphere. The reaction mixture was stirred at room temperature for 3h. The reaction was monitored by LCMS. After completion, The reaction mixture was diluted with NaHCO3 solution (100 mL) , then the resulting mixture was extract-ed with DCM (100 mL x 4) , the combined organic phase was washed with saturated NaCl (50 mL) , then died over with Na2SO4, following with concentration under reduced pressure to obtain crude one, which was purified by silica gel column eluting with petroleum ether/EtOAc (from 0%to 20%) to afford 3- (5-bromo-1H-indol-3-yl) -2, 2-dimethylpropyl acetate (82.0 g, 79%) as a white solid.
LCMS (ESI) calcd. for C15H18BrNO2 [M+H] + m/z 324.22, found: 324.60
Intermediate B: methyl (S) -3- (4-bromothiazol-2-yl) -2- ( (tert-butoxycarbonyl) amino) propanoate
Step 1. Preparation of methyl (S) -3- (4-bromothiazol-2-yl) -2- ( (tert-butoxycarbonyl) amino) propanoate
To a solution of activated Zn dust (22 g, 334 mmol) and DMF (100 mL) was added to a 250 mL three-necked round-bottomed flask and purged with N2, then a solution of I2 (1.5 g, 6.08 mmol) in DMF (5 mL) was added to the above solution. The mixture was stirred for 10 min at rt, then a solution of methyl (R) -2- ( (tert-butoxycarbonyl) amino) -3-iodopropanoate (20 g, 60.79 mmol) in DMF (200 mL) was added dropwise over a period of 10 min. The mixture was heated at 35℃ and stirred for 2 h, then the reaction was cooled to rt. Transfer the liquid to another 500 mL three-necked round-bottomed flask and purged with N2, and a solution of Pd (PPh3) Cl2 (2.1 g, 3.04 mmol) and 2, 4-dibromothiazole 2 (17.7 g, 72.95 mmol) in DMF (100 mL) was added dropwise over a period of 10 min. The reaction was stirred at 50℃ for 16 h under N2 atmosphere, the reaction was monitored by LCMS. After completion, the reaction mixture was diluted with brine and extracted with EtOAc (2 x 500 mL) . The organic layers were combined, washed with saturated NaCl (2 x 500 mL) , dried over anhydrous Na2SO4 and concentrated to give the crude product. The crude product was purified by silica gel column chromatography purified by silica gel column eluting with EtOAc/PE from 0%to 25%to afford methyl (S) -3- (4-bromothiazol-2-yl) -2- ( (tert-butoxycarbonyl) amino) propanoate (10 g, 45.2 %) as a yellow solid.
LCMS (ESI) calcd. for C12H17BrN2O4S [M+H] + m/z 365.01, found: 367.3
Intermediate C: (S) -4- (methoxycarbonyl) -2, 3-diazabicyclo [3.1.1] heptan-2-ium 2, 2, 2-trifluoroacetate
Step 1. Preparation of 3-oxocyclobutane-1-carbonyl chloride
To a stirred solution of 3-oxocyclobutane-1-carboxylic acid (50 g, 438.60 mmol) in DCM (500 mL) and DMF (321 mg, 4.39 mmol) was added (COCl) 2 (83.51 g, 657.89 mmol) , the resulting mixture was stirred at rt for 3 h. The reaction mixture was concentrated under reduced pressure to give the crude prod-uct. The crude product was used to next step without purification.
LCMS (ESI) calcd. for C5H5ClO2 [M+H] + m/z 133.00, found: 133.4
Step 2. Preparation of 3- (2-diazoacetyl) cyclobutan-1-one
To a stirred solution of crude 3-oxocyclobutane-1-carbonyl chloride in THF (250 mL) and CH3CN (250 mL) was added TMSCHN2 (2M in hexanes, 285 mL, 570.18 mmol) dropwisely at 0℃. The resulting mixture was stirred at rt for 18 h. After completion, the reaction solution was concentrated under reduced pressure to give the residual, the residual was purified by silica gel column (eluting with EtOAc/PE from 0%to 50%) to afford 3- (2-diazoacetyl) cyclobutan-1-one (60.2 g, 99.5 %) as a yellow oil.
LCMS (ESI) calcd. for C6H6N2O2 [M+H] + m/z 139.04, found: 139.4
Step 3. Preparation of 2- (3-oxocyclobutyl) acetic acid
To a stirred solution of CF3COOAg (4.82 g, 21.81 mmol) in THF (300 mL) , H2O (30 mL) and TEA (181.91 mL, 1.31 mol) was added a solution of 3- (2-diazoacetyl) cyclobutan-1-one (60.20 g, 436.23 mmol) in THF (300 mL) and H2O (30 mL) at 0℃, the reaction mixture was stirred at rt for 18 h. After completion, the reaction solution was concentrated under reduced pressure to give the residual, the residual was diluted with H2O and acidified with HCl (2N) to pH = 2. The resuliting mixture was extracted with EtOAc (5*300 mL) . The organic layers were combined and dried over Na2SO4, following with concentration under re-duced pressure to afford crude 2- (3-oxocyclobutyl) acetic acid (55.8g, 99.9 %) as a brown oil.
LCMS (ESI) calcd. for C6H8O3 [M+H] + m/z 129.13, found: 129.4
Step 4. Preparation of (S) -4-benzyl-3- (2- (3-oxocyclobutyl) acetyl) oxazolidin-2-one
To a stirred solution of 2- (3-oxocyclobutyl) acetic acid (52.0 g, 406.3 mmol) , (S) -4-benzyloxazolidin-2-one (71.9 g, 406.3 mmol) , 4-Dimethylaminopyridine (5.0 g, 40.6 mmol) and tri-ethylamine (141.2 mL, 1015.6 mmol) in DCM (1 L) was added 2-Chloro-1-methylpyridinium iodide (135.0 g, 528.1 mmol) in portions at 0℃ . The reaction mixture was stirred at R. T. for 4 h. After comple-tion, the reaction solution was diluted with H2O and extracted with DCM, the combined organic phase was washed with H2O dried over Na2SO4, following with concentration under reduced pressure to obtain crude product which was purified by silica gel column (eluting with EA/PE from 0%to 20%) to give (S) -4-benzyl-3- (2- (3-oxocyclobutyl) acetyl) oxazolidin-2-one (68 g 58.3 %) as a yellow oil.
LCMS (ESI) calcd. for C16H17NO4 [M+H] + m/z 288.1, found: 288.5
Step 5. Preparation of (S) -4-benzyl-3- (2- (3-hydroxycyclobutyl) acetyl) oxazolidin-2-one
To a stirred solution of (S) -4-benzyl-3- (2- (3-oxocyclobutyl) acetyl) oxazolidin-2-one (45.0 g, 156.8 mmol) in THF (500 mL) was added AcOH (17.9 mL, 313.6 mmol) and NaBH4 (6.5 g, 172.5 mmol) in por-tions at 0℃. The reaction mixture was stirred at r.t. for 2 h. After completion, the reaction solution was di-luted with H2O then concentrated under reduced pressure to remove THF. The residual was extracted with EtOAc, the combined organic phase was dried over Na2SO4, following with concentration under reduced pressure to obtain (S) -4-benzyl-3- (2- (3-hydroxycyclobutyl) acetyl) oxazolidin-2-one (45 g, 99.3 %) as a light yellow oil.
LCMS (ESI) calcd. for C16H19NO4 [M+H] + m/z 290.1, found: 290.5
Step 6. Preparation of (S) -3- (2- (4-benzyl-2-oxooxazolidin-3-yl) -2-oxoethyl) cyclobutyl 4-methylbenzenesulfonate
To a stirred solution of (S) -4-benzyl-3- (2- (3-hydroxycyclobutyl) acetyl) oxazolidin-2-one (48.0 g, 156.8 mmol) and DIEA (43.3 g, 249.1 mmol) in DCM (500 mL) was added 4-Dimethylaminopyridine (16.2 g, 132.9 mmol) and Tosyl chloride (34.8 g, 182.7 mmol) at 0℃. The reaction mixture was stirred at r.t. for 14 h. After completion, the reaction solution was diluted with H2O and extracted with DCM, the combined organic phase was washed with H2O dried over Na2SO4, following with concentration under re-duced pressure to obtain crude product which was purified by silica gel column (eluting with EA/PE from 0%to 30%) to give (S) -3- (2- (4-benzyl-2-oxooxazolidin-3-yl) -2-oxoethyl) cyclobutyl 4-methylbenzenesulfonate (62.0 g, 84.3 %) as a yellow oil.
LCMS (ESI) calcd. for C23H25NO6S [M+H] + m/z 444.1, found: 444.6
Step 7. Preparation of (S) -4-benzyl-3- (2- (3-bromocyclobutyl) acetyl) oxazolidin-2-one
To a stirred solution of (S) -3- (2- (4-benzyl-2-oxooxazolidin-3-yl) -2-oxoethyl) cyclobutyl 4-methylbenzenesulfonate (62.0 g, 139.8 mmol) in N-Methyl-2-pyrrolidone (650 mL) was added Lithium bromide (24.3 g, 279.6 mmol) , the resulting mixture was stirred at 65℃ for 13 h. The reaction was moni-tored by LCMS. After completion, the reaction solution was diluted with H2O and extracted with EtOAc, the combined organic phase was washed with NaCl (aq. ) dried over Na2SO4, following with concentration under reduced pressure to obtain crude product which was purified by silica gel column (eluting with EA/PE from 0%to 20%) to give (S) -4-benzyl-3- (2- (3-bromocyclobutyl) acetyl) oxazolidin-2-one (42 g, 85.3 %) as a light yellow oil.
LCMS (ESI) calcd. for C16H18BrNO3 [M+H] + m/z 352.0, found: 352.5
Step 8. Preparation of (S) -2, 3-bis (tert-butoxycarbonyl) -2, 3-diazabicyclo [3.1.1] heptane-4-carboxylic acid
To a stirred solution of (S) -4-benzyl-3- (2- (3-bromocyclobutyl) acetyl) oxazolidin-2-one (30.0 g, 85.5 mmol) in THF (300 mL) was added LDA (2M in THF, 111.0 mmol) was added dropwise over a period of 15 minutes at -78℃ under N2 condition. The reaction mixture was stirred at -78℃ for 30 mins, then a solu-tion of Di-tert-Butyl azodicarboxylate (23.6 g, 102.6 mmol) in DCM (50 mL) was added rapidly. The reac-tion mixture was stirred at -78℃ for 30 mins, then 1, 3-Dimethyl-3, 4, 5, 6-tetrahydro-2 (1H) -pyrimidinone (310 mL, 2565.0 mmol) was added dropwise at -78℃. The reaction mixture was stirred at r.t. for 14 h. Af-ter completion, the reaction solution was quenched with water (200 mL) , then LiOH·H2O (10.8 g, 256.5 mmol) was added and the reaction solution was stirred at r.t. for 2 h. After completion, the reaction solution was diluted with brine (200 mL) and washed with tert-Butyl methyl ether 2 times, the aqueous phase was acidified to pH = 3-4 with 2 N HCl. The resulting mixture was extracted with EtOAc, washed with water and brine, then dried over Na2SO4, following with concentration under reduced pressure to obtain crude product (S) -2, 3-bis (tert-butoxycarbonyl) -2, 3-diazabicyclo [3.1.1] heptane-4-carboxylic acid, which was used directly for the next step.
LCMS (ESI) calcd. for C16H26N2O6 [M+H] + m/z 343.39, found: 343.5
Step 9. Preparation of 2, 3-di-tert-butyl 4-methyl (S) -2, 3-diazabicyclo [3.1.1] heptane-2, 3, 4-tricarboxylate
To a stirred solution of crude product (S) -2, 3-bis (tert-butoxycarbonyl) -2, 3-diazabicyclo [3.1.1] heptane-4-carboxylic acid (27.0 g, 78.9 mmol) in DMF (300 mL) was added K2CO3 (27.3 g, 197.4 mmol) and Iodomethane (10 mL, 157.8 mmol) at r.t.. The reaction mixture was stirred at r.t. for 2 h. After completion, the reaction solution was concentration under reduced pressure to obtain crude product which was purified by silica gel column (eluting with EA/PE from 0%to 23%) to give 2, 3-di-tert-butyl 4-methyl (S) -2, 3-diazabicyclo [3.1.1] heptane-2, 3, 4-tricarboxylate (5.6 g, 18.45 %in two-steps) as a light yellow oil.
LCMS (ESI) calcd. for C17H28N2O6 [M-H] -m/z 357.2, found: 357.6
Step 10. Preparation of (S) -4- (methoxycarbonyl) -2, 3-diazabicyclo [3.1.1] heptan-2-ium 2, 2, 2-trifluoroacetate
To a stirred solution of 2, 3-di-tert-butyl 4-methyl (S) -2, 3-diazabicyclo [3.1.1] heptane-2, 3, 4-tricarboxylate (7.3 g, 0.18 mmol) in DCM (100 mL) was added TFA (40 mL) , the resulting mixture was stirred at rt for 2 h. The reaction mixture was concentrated to give the crude product (S) -4- (methoxycarbonyl) -2, 3-diazabicyclo [3.1.1] heptan-2-ium 2, 2, 2-trifluoroacetate (7.2 g) as a yellow solid.
LCMS (ESI) calcd. for C15H23BO4 [M+H] + m/z 157.09, found: 157.4
Intermediate D: tert-butyl
( (63S, 4S, Z) -10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -61, 62, 63, 64, 65, 66-hexahy-
dro-11H-8-oxa-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (1, 3) -pyridazinacycloundecaphane-4-yl) carbamate
Step 1. Preparation of 2, 2-dimethyl-3- (5- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -1H-indol-3-yl) propyl acetate
To a stirred solution of 3- (5-bromo-1H-indol-3-yl) -2, 2-dimethylpropyl acetate (20 g, 61.92 mmol) in 1, 4-dioxane (400 mL) was added Pd (dppf) Cl2 (2.2 g, 3.10 mmol) and KOAc (18.4 g, 185.76 mmol) . The reaction was stirred at 90℃ for 16 h under N2 atmosphere, the reaction was monitored by LCMS. After completion, the reaction solution was concentrated under reduced pressure to remove 1, 4-dioxane, the re-sidual was diluted with H2O and extracted with EtOAc (500 mL x 3) , the combined organic phase was washed with saturated NaCl (500 mL) , then dried over Na2SO4, following with concentration under re-duced pressure to obtain crude product which was purified by silica gel column eluting with EtOAc/PE from 0%to 27%to afford 2, 2-dimethyl-3- (5- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -1H-indol-3-yl) propyl acetate (18.7 g, 81.4 %) as a light yellow solid.
LCMS (ESI) calcd. for C21H30BNO4 [M+H] + m/z 372.23, found: 372.5
Step 2. Preparation of methyl (S) -3- (4- (3- (3-acetoxy-2, 2-dimethylpropyl) -1H-indol-5-yl) thiazol-2-yl) -2- ( (tert-butoxycarbonyl) amino) propanoate
To a stirred solution of 2, 2-dimethyl-3- (5- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -1H-indol-3-yl) propyl acetate (18.7 g, 50.40 mmol) , methyl (S) -3- (4-bromothiazol-2-yl) -2- ( (tert-butoxycarbonyl) amino) propanoate (27.5 g, 75.61 mmol) and K3PO4 (26.7 g, 126.01 mmol) in toluene/1, 4-dioxane/H2O (300 mL, V: V: V = 4: 1: 1 ) was added Pd (dppf) Cl2 (1.6 g, 2.52 mmol) at rt. The reaction mixture was stirred at 70℃ under N2 for 16 h. After completion, the reaction solution was concentrated under reduced pressure to remove 1, 4-dioxane and toluene, the residual was diluted with H2O and extracted with EtOAc (500 mL x 3) , the combined or-ganic phase was washed with saturated NaCl (500 mL) , then dried over Na2SO4, following with concentra-tion under reduced pressure to obtain crude product which was purified by silica gel column eluting with EtOAc/PE from 0%to 36%to afford methyl (S) -3- (4- (3- (3-acetoxy-2, 2-dimethylpropyl) -1H-indol-5-yl) thiazol-2-yl) -2- ( (tert-butoxycarbonyl) amino) propanoate (20.7 g, 77.6 %) as a light yellow solid.
LCMS (ESI) calcd. for C27H35N3O6S [M+H] + m/z 530.22, found: 530.6
Step 3. Preparation of methyl (S) -3- (4- (3- (3-acetoxy-2, 2-dimethylpropyl) -2-iodo-1H-indol-5-yl) thiazol-2-yl) -2- ( (tert-butoxycarbonyl) amino) propanoate
To a stirred solution of methyl (S) -3- (4- (3- (3-acetoxy-2, 2-dimethylpropyl) -1H-indol-5-yl) thiazol-2-yl) -2- ( (tert-butoxycarbonyl) amino) propanoate (12.3 g, 23.25 mmol) and NaHCO3 (2.3 g, 27.90 mmol) in THF (150 mL) under ice-water bath was added AgSO3CF3 (7.2 g, 27.90 mmol) in THF (50 mL) and I2 (5.0 g, 19.76 mmol) in THF (50 mL) by dropwise to the reaction mixture. The reaction was stirred at -20℃ for 1 h, the reaction was monitored by LCMS. After completion, The reaction was quenched with saturated Na2S2O3 (200 mL) and was extracted with EtOAc (500 mL x 2) , the combined organic phase was washed with saturated NaCl (500 mL) , then dried over Na2SO4, following with concentration under reduced pressure to obtain crude product which was purified by silica gel column eluting with EtOAc/PE from 0%to 40%to afford methyl (S) -3- (4- (3- (3-acetoxy-2, 2-dimethylpropyl) -2-iodo-1H-indol-5-yl) thiazol-2-yl) -2- ( (tert-butoxycarbonyl) amino) propanoate (12.0 g, 78.8 %) as a light yellow solid.
LCMS (ESI) calcd. for C27H34IN3O6S [M+H] + m/z 656.12, found: 656.5
Step 4. Preparation of (S) -2- ( (tert-butoxycarbonyl) amino) -3- (4- (3- (3-hydroxy-2, 2-dimethylpropyl) -2-iodo-1H-indol-5-yl) thiazol-2-yl) propanoic acid
To a solution of methyl (S) -3- (4- (3- (3-acetoxy-2, 2-dimethylpropyl) -2-iodo-1H-indol-5-yl) thiazol-2-yl) -2- ( (tert-butoxycarbonyl) amino) propanoate (25 g, 38.17 mmol) in THF (500 mL) and H2O (100 mL) was added LiOH·H2O (4.6 g, 190.84 mmol) at 0-10℃, the resulting mixture was stirred at 0-10℃ for 16 h. The reaction was monitored by LCMS. After completion, the reaction mixture was acidified to pH = 3-4 with 2 N HCl. The resulting mixture was extracted with EtOAc (300 mL x 2) , the combined organic phase was washed with saturated NaCl (500 mL) , then dried over Na2SO4 and concentrated to give the (S) -2- ( (tert-butoxycarbonyl) amino) -3- (4- (3- (3-hydroxy-2, 2-dimethylpropyl) -2-iodo-1H-indol-5-yl) thiazol-2-yl) propanoic acid (22.8 g, 100 %) as a light yellow solid.
LCMS (ESI) calcd. for C24H30IN3O5S [M+H] + m/z 599.10, found: 600.6
Step 5. Preparation of (S) -2- ( (S) -2- ( (tert-butoxycarbonyl) amino) -3- (4- (3- (3-hydroxy-2, 2-dimethylpropyl) -2-iodo-1H-indol-5-yl) thiazol-2-yl) propanoyl) -2, 3-diazabicyclo [3.1.1] heptane-4-carboxylate
To a solution of (S) -2- ( (tert-butoxycarbonyl) amino) -3- (4- (3- (3-hydroxy-2, 2-dimethylpropyl) -2-iodo-1H-indol-5-yl) thiazol-2-yl) propanoic acid (1.10 g, 1.84 mmol) in DMF (15 mL) were added HATU (1.04 g, 2.75 mmol) and DIEA (1.30 mL, 7.35 mmol) at 0-10℃. The resulting mixture was stirred at 0-10℃ for 10 min. Then me-thyl (S) -2, 3-diazabicyclo [3.1.1] heptane-4-carboxylate (472 mg, 1.84 mmol) was added into the reaction mixture, the resulting mixture was stirred at rt for 2 h. The reaction was monitored by LCMS. After com-pletion, the reaction mixture was diluted with brine and extracted with EtOAc (2 x 400 mL) , the combined organic phase was washed with saturated NaCl (500 mL) , then dried over Na2SO4, following with concen-tration under reduced pressure to obtain crude product which was purified by silica gel column eluting with EtOAc/PE from 0%to 60%to afford methyl (S) -2- ( (S) -2- ( (tert-butoxycarbonyl) amino) -3- (4- (3- (3-hydroxy-2, 2-dimethylpropyl) -2-iodo-1H-indol-5-yl) thiazol-2-yl) propanoyl) -2, 3-diazabicyclo [3.1.1] heptane-4-carboxylate (1.23 g, 90.7 %) as a light yellow solid.
LCMS (ESI) calcd. for C31H40IN5O6S [M+H] + m/z 738.2, found: 738.6
Step 6. Preparation of (S) -2- ( (S) -2- ( (tert-butoxycarbonyl) amino) -3- (4- (3- (3-hydroxy-2, 2-dimethylpropyl) -2-iodo-1H-indol-5-yl) thiazol-2-yl) propanoyl) -2, 3-diazabicyclo [3.1.1] heptane-4-carboxylic acid
To a solution of methyl (S) -2- ( (S) -2- ( (tert-butoxycarbonyl) amino) -3- (4- (3- (3-hydroxy-2, 2-dimethylpropyl) -2-iodo-1H-indol-5-yl) thiazol-2-yl) propanoyl) -2, 3-diazabicyclo [3.1.1] heptane-4-carboxylate (1.23 g, 24.53 mmol) in THF (1.23 g, 24.53 mmol) and H2O (6 mL) was added LiOH·H2O (210 mg, 5.01 mmol) at 0-10℃, the resulting mixture was stirred at 0-10℃ for 2 h. The reaction was monitored by LCMS. After completion, the reaction mix-ture was acidified to pH = 3-4 with 2 N HCl. The resulting mixture was extracted with EtOAc (300 mL x 2) , the combined organic phase was washed with saturated NaCl (500 mL) , then dried over Na2SO4 and concentrated to give the (S) -2- ( (S) -2- ( (tert-butoxycarbonyl) amino) -3- (4- (3- (3-hydroxy-2, 2-dimethylpropyl) -2-iodo-1H-indol-5-yl) thiazol-2-yl) propanoyl) -2, 3-diazabicyclo [3.1.1] heptane-4-carboxylic acid (1.03 g, 85.3 %) as a light yellow solid.
LCMS (ESI) calcd. for C30H38IN5O6S [M+H] + m/z 724.1, found: 724.9
Step 7. Preparation of tert-butyl ( (64S, 4S, Z) -12-iodo-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
To a solution of TCFH (994 mg, 3.54 mmol) in ACN (80 mL) was added a solution of (S) -2- ( (S) -2- ( (tert-butoxycarbonyl) amino) -3- (4- (3- (3-hydroxy-2, 2-dimethylpropyl) -2-iodo-1H-indol-5-yl) thiazol-2-yl) propanoyl) -2, 3-diazabicyclo [3.1.1] heptane-4-carboxylic acid 22 (640 mg, 0.89 mmol) and NMI (582 mg, 7.08 mmol) in ACN (48 mL) , the reaction was gradually improved temperature to room temperature and stirred for 2 h. The reaction was monitored by LCMS. After completion, the reaction was concentrated under reduced pressure to give crude product which was purified by silica gel column (eluting with EtOAc/PE from 0%to 65%to afford tert-butyl ( (64S, 4S, Z) -12-iodo-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate 23 (199 mg, 31.7 %) as a light yellow solid.
LCMS (ESI) calcd. for C30H36IN5O5S [M+H] + m/z 706.2, found: 706.8
Step 8. Preparation of tert-butyl ( (64S, 4S, Z) -10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
To a stirred solution of tert-butyl ( (64S, 4S, Z) -12-iodo-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (790 mg, 1.12 mmol) , Pd2 (dba) 3 (154 mg, 0.17 mmol) , SPhos (154 mg, 0.17 mmol) and AcOK (495 mg, 5.04 mmol) in toluene was added B2Pin2 (1.08 g, 8.40 mmol) at 0℃ under N2 atmosphere. The reaction mixture was stirred at 60℃ under N2 for 3h. After completion, the reaction mixture was concentrated under reduced pressure to give the crude product which was purified by silica gel column (eluting with EtOAc/PE from 0%to 60%) to give crude tert-butyl ( (64S, 4S, Z) -10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (810 mg) as a pale yellow solid.
LCMS (ESI) calcd. for C36H48BN5O7S [M+H] + m/z 706.3, found: 707.0
Intermediate E: (S) -3-bromo-5-iodo-2- (1-methoxyethyl) pyridine
Step 1. Preparation of (S) -1- (3-bromopyridin-2-yl) ethan-1-ol
To a stirred solution of TEA (151.76 g, 1.50 mol) was added FA (13.81 g, 299.96 mmol) at 0℃. Then (S,S) -N- (p-Toluenesulfonyl) -1, 2-diphenylethanediamine (chloro) (p-cymene) ruthenium (II) (800 mg, 1.25 mmol) was added into the reaction mixture, the resulting mixture was stirred at 40℃ under N2 for 0.5 h. 1- (3-bromopyridin-2-yl) ethan-1-one (25.00 g, 124.98 mmol) was added into the reaction mixture, the re-sulitng mixture was stirred at 40℃ under N2 for 1 h. The reaction was monitored by LCMS. After comple-tion, the reaction mixture was diluted with EtOAc (500 mL) , washed with saturated NH4Cl (300 x 3 mL) , then dried over Na2SO4, following with concentration under reduced pressure to obtain crude product which was purified by silica gel column (eluting with EtOAc/PE from 0%to 25%) to give (S) -1- (3-bromopyridin-2-yl) ethan-1-ol (28.80 g, 95.0 %) as a yellow oil.
LCMS (ESI) calcd. for C7H6BrNO [M+H] + m/z 200.98, found: 202.1
Step 2. Preparation of (S) -3-bromo-2- (1-methoxyethyl) pyridine
To a stirred solution of (S) -1- (3-bromopyridin-2-yl) ethan-1-ol (23.80 g, 117.82 mmol) in THF (240 mL) was added NaH (60%in oil, 7.07 g, 176.73 mmol) portion wisely at 0℃, the resulting mixture was stirred at 0℃ for 1 h. CH3I (33.45 g, 235.64 mmol) was added into the reaction mixture at 0℃, the result-ing mixture was stirred at rt for 15 h. The reaction mixture was quenched with H2O (50 mL) at 0℃ and extracted with EtOAc (2*200 mL) . The organic layers were combined, washed with brine, dried over Na2SO4 and concentrated to give the crude product which was purified by silica gel column (eluting with EtOAc/PE from 0%to 25%) to give (S) -3-bromo-2- (1-methoxyethyl) pyridine (23.39 g, 91.9 %) as a light yellow oil.
LCMS (ESI) calcd. for C8H10BrNO [M+H] + m/z 214.99, found: 216.3
Step 3. Preparation of (S) -3-bromo-2- (1-methoxyethyl) -5- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyridine
To a stirred solution of (S) -3-bromo-2- (1-methoxyethyl) pyridine (12.00 g, 55.56 mmol) B2Pin2 (15.53 g, 61.11 mmol) in THF (180 mL) was added [Ir (COD) (OMe) ] 2 (737 mg, 1.11 mmol) and dtbpy (895 mg, 3.33 mmol) at rt , the resulting mixture was stirred at 75℃ under N2 for 23 h. The reaction was monitored by LCMS. After completion, the reaction mixture was concentrated to give the crude product which was purified by silica gel column (eluting with EtOAc/PE from 0%to 80%) to give crude (S) -3-bromo-2- (1-methoxyethyl) -5- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyridine (20.9 g) as an orange oil.
LCMS (ESI) calcd. for C14H21BBrNO3 [M+H] + m/z 341.08, found: 342.4
Step 4. Preparation of (S) - (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) boronic acid
To a stirred solution of crude (S) -3-bromo-2- (1-methoxyethyl) -5- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyridine (16.20 g, 47.51 mmol) in THF (150 mL) and H2O (150 mL) were added NH4OAc (14.60 g, 190.03 mmol ) and NaIO4 (40.60 g, 190.03 mmol) at 0℃, the resulting mixture was stirred at rt for 3h. The reaction was monitored by LCMS. After completion, the reaction mixture was extracted with EtOAc (2*300 mL) . The organic lay-ers were combined, washed with brine, dried over Na2SO4 and concentrated to give the crude product which was purified by silica gel column (eluting with EtOAc/PE from 0%to 80%) to give (S) - (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) boronic acid (10.98 g, 89.2%) as a yellow solid.
LCMS (ESI) calcd. for C8H11BBrNO3 [M+H] + m/z 259.00, found: 260.3
Step 5. Preparation of (S) -3-bromo-5-iodo-2- (1-methoxyethyl) pyridine
To a stirred solution of (S) - (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) boronic acid (3.00 g, 11.58 mmol) in CH3CN (45 mL) was added NIS (3.13 g, 13.90 mmol) at rt, the resulting mixture was stirred at 75℃ for 20h. The reaction was monitored by LCMS. After completion, the reaction mixture was quenched with sat. aq. Na2SO3 and concentrated to give the residue. The residue was extracted with EtOAc (2*30 mL) . The organic layers were combined and washed with brine, then dried over Na2SO4 and concentrated to give the crude product which was purified by silica gel column (eluting with EtOAc/PE from 0%to 20%) to give (S) -3-bromo-5-iodo-2- (1-methoxyethyl) pyridine (2.65 g, 67.1%) as a brown solid.
LCMS (ESI) calcd. for C8H9BrINO3 [M+H] + m/z 340.89, found: 342.3
Preparation of compounds
Unless otherwise defined, all technical and scientific terms in the present invention have the same meanings as generally understood by a person skilled in the art to which the invention belongs. In accord-ance with the general technical knowledge and customary means in the art, under the premise of not de-parting from the above-mentioned basic technical ideas of the present invention, other forms of modifica-tion, replacement or change can also be made.
Unless otherwise stated, the raw materials and reagents used in the following embodiments are com-mercially available or may be prepared by known methods.
Intermediate of Example 3:
Synthesis of (S) -4- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) -2-methylbut-3-yn-2-ol
Step 1. Preparation of (S) -4- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) -2-methylbut-3-yn-2-ol
A mixture of (S) -3-bromo-5-iodo-2- (1-methoxyethyl) pyridine (200 mg, 0.59 mmol) , 2-methylbut-3-yn-2-ol (99 mg, 1.17 mmol) , K2CO3 (243 mg, 1.76 mmol) , Pd (PPh3) 2Cl2 (41 mg, 0.059 mmol) and CuI (11 mg, 0.059 mmol) in THF (4.5 mL) was stirred at 65℃ under N2 for 23 h. The reaction was monitored by LCMS. After completion, the reaction mixture was concentrated to give the crude prod-uct which was purified by silica gel column (eluting with EtOAc/PE from 0%to 40%) to give (S) -4- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) -2-methylbut-3-yn-2-ol (160 mg, 91.3%) as a brown solid.
LCMS (ESI) calcd. for C13H16BrNO2 [M+H] + m/z 297.04, found: 298.4
Intermediate of Example 4:
Synthesis of 3-bromo-2- ( (S) -1-methoxyethyl) -5- ( ( (S) -1-methylpyrrolidin-2-yl) ethynyl) pyridine
Step 1: Synthesis of tert-butyl (S) -2-ethynylpyrrolidine-1-carboxylate
To a stirred solution of tert-butyl (S) -2-formylpyrrolidine-1-carboxylate (2.0 g, 10.05 mmol) in MeOH (40 mL) was added K2CO3 (3.5 g, 25.13 mmol) and Dimethyl (1-Diazo-2-oxopropyl) phosphonate (2.3 g, 12.06 mmol) at r.t.. The reaction mixture was stirred at r.t. for overnight. After completion, the reaction solution was concentrated under reduced pressure to remove MeOH, the residual was diluted with H2O and extracted with EtOAc, the combined organic phase was dried over Na2SO4, following with concentration under reduced pressure to obtain crude product which was purified by silica gel column (eluting with EA/PE from 0%to 20%) to give tert-butyl (S) -2-ethynylpyrrolidine-1-carboxylate (1.7 g, 86.7 %) as a colourless oil.
LCMS (ESI) calcd. for C11H17NO2 [M+H] + m/z 195.1, found: 196.3.
Step 2: Synthesis of tert-butyl (S) -2- ( (5-bromo-6- ( (S) -1-methoxyethyl) pyridin-3-yl) ethynyl) pyrrolidine-1-carboxylate
To a stirred solution of tert-butyl (S) -2-ethynylpyrrolidine-1-carboxylate (1.7 g, 8.72 mmol) , (S) -3-bromo-5-iodo-2- (1-methoxyethyl) pyridine (2.5 g, 7.26 mmol) , Pd (PPh3) 2Cl2 (509 mg, 0.73 mmol) and CuI (277 mg, 1.45 mmol) in 1, 4-dioxane (40 mL) was added TEA (4 mL, 29.04 mmol) at r.t.. The re-action mixture was stirred at 35℃ under N2 for 8 h. After completion, the reaction solution was concen-trated under reduced pressure to remove 1, 4-dioxane, the residual was diluted with H2O and extracted with EtOAc, the combined organic phase was dried over Na2SO4, following with concentration under reduced pressure to obtain crude product which was purified by silica gel column (eluting with EA/PE from 0%to 30%) to give tert-butyl (S) -2- ( (5-bromo-6- ( (S) -1-methoxyethyl) pyridin-3-yl) ethynyl) pyrrolidine-1-carboxylate (2.8 g, 93.6 %) as a light yellow oil.
LCMS (ESI) calcd. for C19H25BrN2O3 [M+H] + m/z 409.1, found: 310.9.
Step 3: Synthesis of 3-bromo-2- ( (S) -1-methoxyethyl) -5- ( ( (S) -pyrrolidin-2-yl) ethynyl) pyridine
To a stirred solution of tert-butyl (S) -2- ( (5-bromo-6- ( (S) -1-methoxyethyl) pyridin-3-yl) ethynyl) pyrrolidine-1-carboxylate (2.8 g, 6.84 mmol) in dichloromethane (30 mL) was added TFA (10 mL) slowly at r.t., the resulting mixture was stirred at rt for 3 h. The reaction mixture was concentrated to give the crude product 3-bromo-2- ( (S) -1-methoxyethyl) -5- ( ( (S) -pyrrolidin-2-yl) ethynyl) pyridine (2.8 g, crude) as a yellow oil.
LCMS (ESI) calcd. for C14H17BrN2O [M+H] + m/z 309.2, found: 310.6.
Step 4: Synthesis of 3-bromo-2- ( (S) -1-methoxyethyl) -5- ( ( (S) -1-methylpyrrolidin-2-yl) ethynyl) pyridine
To a stirred solution of (S) -4- (benzyloxy) -1-bromo-2- (1-methoxyethyl) benzene (2.12 g, 6.84 mmol) in DCM/MeOH (55 mL, V: V = 5: 1 ) was added Formaldehyde (2.9 g, 37%in water , 35.9 mmol) and AcOH (0.5 mL) at r.t.. The reaction mixture was stirred at r.t. for 5 h, then NaBH3CN (734 mg, 11.7 mmol) was added. The reaction mixture was stirred at r.t. for 2 h. After completion, the reaction solution was di-luted with H2O and extracted with DCM, the combined organic phase was dried over Na2SO4, following with concentration under reduced pressure to obtain crude product which was purified by silica gel col-umn (eluting with DCM/MeOH from 0%to 10%) to give 3-bromo-2- ( (S) -1-methoxyethyl) -5- ( ( (S) -1-methylpyrrolidin-2-yl) ethynyl) pyridine (2.13 g, 96.1 %) as a light yellow oil.
LCMS (ESI) calcd. for C15H19BrN2O [M+H] + m/z 323.1, found: 323.4.
Intermediate of Example 5:
Synthesis of tert-butyl
(S) -3- ( (5-bromo-6- ( (S) -1-methoxyethyl) pyridin-3-yl) ethynyl) pyrrolidine-1-carboxylate
Step 1. Preparation of tert-butyl (S) -3-ethynylpyrrolidine-1-carboxylate
To a stirred solution of tert-butyl (R) -3-formylpyrrolidine-1-carboxylate (1 g, 5.0 mmol) and K2CO3 (1.4 g, 10.0 mmol) in MeOH (15 mL) was added dimethyl (1-diazo-2-oxopropyl) phosphonate (1.2 g, 6.0 mmol) at RT. The reaction mixture was stirred at RT for 2 h. After completion, the reaction mixture was diluted with EtOAc (50 mL) , washed with brine (30 mL) , then dried over Na2SO4, following with concen-tration under reduced pressure to obtain crude product tert-butyl (S) -3-ethynylpyrrolidine-1-carboxylate (980 mg) as a yellow oil.
LCMS (ESI) calcd. for C11H17NO2 [M+H-56] + m/z 140.1, found: 140.4
Step 2. Preparation of tert-butyl (S) -3- ( (5-bromo-6- ( (S) -1-methoxyethyl) pyridin-3-yl) ethynyl) pyrrolidine-1-carboxylate
A mixture of tert-butyl (S) -3-ethynylpyrrolidine-1-carboxylate (980 mg, 5.0 mmol) , (S) -3-bromo-5-iodo-2- (1-methoxyethyl) pyridine (1.7 g, 5.0 mmol) , CuI (96 mg, 0.5 mmol) TEA (1.0 g, 10.0 mmol) and Pd (PPh3) 2Cl2 (352 mg, 0.5 mmol) in THF (20 mL) was stirred at 50℃ under N2 for 2 h. After completion, the reaction mixture was concentrated to give the crude product which was purified by silica gel column (eluting with EA/PE from 0%to 25%) to give tert-butyl (S) -3- ( (5-bromo-6- ( (S) -1-methoxyethyl) pyridin-3-yl) ethynyl) pyrrolidine-1-carboxylate (1.8 g, 90 %in 2 steps) as a brown oil.
LCMS (ESI) calcd. for C19H25BrN2O3 [M+H] + m/z 409.1, found: 409.6
Intermediate of Example 6:
Synthesis of (S) -3-bromo-2- (1-methoxyethyl) -5- ( (1-methyl-1H-pyrazol-4-yl) ethynyl) pyridine
Step 1: Synthesis of 1-methyl-4- ( (trimethylsilyl) ethynyl) -1H-pyrazole
A mixture of 4-iodo-1-methyl-1H-pyrazole (1 g, 4.81 mmol) , ethynyltrimethylsilane (708 mg, 7.21 mmol) , TEA (1.46 g, 14.42 mmol) , Pd (PPh3) 2Cl2 (337 mg, 0.48 mmol) and CuI (92 mg, 0.48 mmol) in THF (15 mL) was stirred at 50℃ under N2 for 1 h. After completion, the reaction mixture was concen-trated to give the crude product which was purified by silica gel column (eluting with EtOAc/PE from 0%to 50%) to give 1-methyl-4- ( (trimethylsilyl) ethynyl) -1H-pyrazole (500 mg, 58.4%) as a brown solid.
LCMS (ESI) calcd. for C9H14N2Si [M+H] + m/z 179.1, found: 179.4.
Step 2: Synthesis of 4-ethynyl-1-methyl-1H-pyrazole
To a stirred solution of 1-methyl-4- ( (trimethylsilyl) ethynyl) -1H-pyrazole (500 mg, 2.81 mmol) in THF (5mL) was added TBAF (1M in THF, 4.2 mL, 4.2 mmol) dropwisely at rt, the resulting mixture was stirred at rt for 1 h. After completion, the reaction mixture was used to next step without purification.
LCMS (ESI) calcd. for C6H6N2 [M+H] + m/z 106.1, found: 107.4.
Step 3: Synthesis of (S) -3-bromo-2- (1-methoxyethyl) -5- ( (1-methyl-1H-pyrazol-4-yl) ethynyl) pyridine
A mixture of (S) -3-bromo-5-iodo-2- (1-methoxyethyl) pyridine (735 mg, 2.16 mmol) , 4-ethynyl-1-methyl-1H-pyrazole 3 (300 mg, 2.80 mmol) , TEA (654 mg, 6.47 mmol) , Pd (PPh3) 2Cl2 (151 mg, 0.22 mmol) and CuI (41 mg, 0.22 mmol) in THF (6 mL) was stirred at 50℃ under N2 for 2 h. The re-action was monitored by LCMS. After completion, the reaction mixture was concentrated to give the crude product which was purified by silica gel column (eluting with EtOAc/PE from 0%to 50%) to give (S) -3-bromo-2- (1-methoxyethyl) -5- ( (1-methyl-1H-pyrazol-4-yl) ethynyl) pyridine (379 mg, 55.0%) as a yellow solid.
LCMS (ESI) calcd. for C13H16BrNO2 [M+H] + m/z 320.0, found: 320.5.
Intermediate of Example 8:
Synthesis of (S) -5- ( (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) ethynyl) benzo [d] thiazole
Synthesis of (S) -5- ( (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) ethynyl) benzo [d] thiazole
To a stirred solution of 5-ethynylbenzo [d] thiazole (100 mg, 0.63 mmol) , (S) -3-bromo-5-iodo-2- (1-methoxyethyl) pyridine (178 mg, 0.52 mmol) , Pd (PPh3) 2Cl2 (37 mg, 0.05 mmol) and CuI (20 mg, 0.10 mmol) in 1, 4-dioxane (3 mL) was added TEA (0.3 mL, 2.09 mmol) at r.t.. The re-action mixture was stirred at 50℃ under N2 for 4 h. After completion, the reaction solution was concen-trated under reduced pressure to remove 1, 4-dioxane, the residual was diluted with H2O and extracted with EtOAc , the combined organic phase was dried over Na2SO4, following with concentration under reduced pressure to obtain crude product which was purified by silica gel column (eluting with EA/PE from 0%to 95%) to give (S) -5- ( (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) ethynyl) benzo [d] thiazole (190 mg, 80.9%) as a light yellow solid.
LCMS (ESI) calcd. for C17H13BrN2OS [M+H] + m/z 373.0, found: 373.4.
Intermediate of Example 9:
Synthesis of (S) -3- ( (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) ethynyl) imidazo [1, 2-b] pyridazine
Step 1: Synthesis of (S) -3- ( (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) ethynyl) imidazo [1, 2-b] pyridazine
A mixture of (S) -3-bromo-5-iodo-2- (1-methoxyethyl) pyridine (200 mg, 0.59 mmol) , 3-ethynylimidazo [1, 2-b] pyridazine 2 (126 mg, 0.88 mmol) , CuI (11 mg, 0.059 mmol) , TEA (227 mg, 1.76 mmol) and Pd (PPh3) 2Cl2 (41 mg, 0.059 mmol) in DMF (3 mL) was stirred at 65℃ for 1 h under N2 at-mosphere, the reaction was monitored by LCMS. After completion, the mixture was filtered and the filtrate was concentrated, the residue was purified by silica gel column chromatography (eluting with EtOAc/PE from 0%to 100%) to obtain (S) -3- ( (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) ethynyl) imidazo [1, 2-b] pyridazine (200 mg, 95.2%) as a yellow oil.
LCMS (ESI) calcd. for C16H13BrN4O [M+H] + m/z 357.0, found 357.4.
The following intermediates in Table 2 were prepared using the method described above in step for the preparation of (S) -4- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) -2-methylbut-3-yn-2-ol or tert-butyl (S) -3- ( (5-bromo-6- ( (S) -1-methoxyethyl) pyridin-3-yl) ethynyl) pyrrolidine-1-carboxylate and utilizing the appropriate starting materials and modifications.
Table 2. Certain Intermediates containing alkynyl group of the Present invention
Intermediate of Example 10:
Synthesis of (S) -3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) -N, N-dimethylprop-2-yn-1-amine
Step 1: Synthesis of (S) -3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) -N, N-dimethylprop-2-yn-1-amine (2)
To a solution of (R) -morpholin-3-ylmethanol (58 mg, 0.71 mmol) in DCM (2 mL) were added DIEA (244 mg, 1.89 mmol) and a solution of (S) -3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl 4-methylbenzenesulfonate (200 mg, 0.47 mmol) in DCM (2 mL) at rt, the resulting mixture was stirred at rt for 15 h. The reaction was monitored by LCMS. After completion, the mixture was concentrated under reduced pressure to give the residual which was purified by silica gel column chromatography (eluting with MeOH/DCM from 0%to 10%) to give (S) -3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) -N, N-dimethylprop-2-yn-1-amine (73 mg, yield: 52.5%) as a brown oil.
LCMS (ESI) calcd. for C13H17BrN2O [M+H] + m/z 297.1, found 297.5.
Intermediate of Example 11:
Synthesis of
3-bromo-5- (3- ( (R) -3-fluoropyrrolidin-1-yl) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridine
Step 1: Synthesis of 3-bromo-5- (3- ( (R) -3-fluoropyrrolidin-1-yl) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridine
To a stirred solution of (S) -3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl 4-methylbenzenesulfonate (350 mg, 0.83 mmol) in DCM (10 mL) was added (R) -3-fluoropyrrolidine (81 mg, 0.91 mmol) and DIEA (170 mg, 1.32 mmol) at r.t., the resulting mixture was stirred at r.t. for 8 h. The reaction was monitored by LCMS. After completion, the reaction mixture was diluted with DCM/MeOH (30 mL, V: V = 10: 1) , washed with water (10 mL x 3) and saturated NaCl (10 x 2 mL) , then dried over Na2SO4, following with concentration under reduced pressure to obtain crude product which was purified by silica gel column (eluting with MeOH/DCM from 0%to 5%) to give 3-bromo-5- (3- ( (R) -3-fluoropyrrolidin-1-yl) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridine (230 mg, 81.5 %) as a lihgt yellow oil.
LCMS (ESI) calcd. for C15H18BrFN2O [M+H] + m/z 341.1, found: 341.53.
Intermediate of Example 12:
Synthesis of (S) -3-bromo-2- (1-methoxyethyl) -5- (3- (piperidin-1-yl) prop-1-yn-1-yl) pyridine
Step 1: Preparation of (S) -3-bromo-2- (1-methoxyethyl) -5- (3- (piperidin-1-yl) prop-1-yn-1-yl) pyridine
A mixture of piperidine (66.2 mg, 0.78 mmol) , (S) -3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl 4-methylbenzenesulfonate (300 mg, 0.71 mmol) in DCM (5 mL) was added DIEA (182.7 mg, 1.4 mmol) . The reaction mixture was stirred at rt for 3h. After completion, the mixture was was concentrated under reduced pressure. The residue was purified by FCC with EtOAc/PE = 0-30%to afford the (S) -3-bromo-2- (1-methoxyethyl) -5- (3- (piperidin-1-yl) prop-1-yn-1-yl) pyridine (206 mg, 86%) as a yellow oil.
LCMS (ESI) calcd. for C16H21BrN2O [M+H] + m/z 337.1, found 337.6
Intermediate of Example 13:
Synthesis of (S) -1- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) -4-methylpiperazine
Step 1: Preparation of (S) -1- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) -4-methylpiperazine
A mixture of 1-methylpiperazine (52 mg, 0.52 mmol) , (S) -3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl 4-methylbenzenesulfonate (200 mg, 0.47 mmol) in DCM (5 mL) was added DIEA (121.5 mg, 0.94 mmol) . The reaction mixture was stirred at rt for 3h. After completion, the mixture was was concentrated under reduced pressure. The residue was purified by FCC with MeOH/DCM = 0-6%to afford the (S) -1- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) -4-methylpiperazine (130 mg, 79%) as a yellow oil.
LCMS (ESI) calcd. for C16H22BrN3O [M+H] + m/z 352.1, found 352.6.
Intermediate of Example 14:
Synthesis of
(S) -1- (4- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) piperazin-1-yl) ethan-1-one
Step 1: Synthesis of (S) -1- (4- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) piperazin-1-yl) ethan-1-one
To a stirred solution of (S) -3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl 4-methylbenzenesulfonate (350 mg, 0.83 mmol) in DCM (10 mL) was added 1- (piperazin-1-yl) ethan-1-one (120 mg, 0.91 mmol) and DIEA (170 mg, 1.32 mmol) at r.t., the resulting mixture was stirred at r.t. for 8 h. The reaction was monitored by LCMS. After completion, the reaction mixture was diluted with DCM/MeOH (30 mL, V: V = 10: 1) , washed with water (10 mL x 3) and saturated NaCl (10 x 2 mL) , then dried over Na2SO4, following with concentration under reduced pressure to obtain crude product which was purified by silica gel column (eluting with MeOH/DCM from 0%to 5%) to give (S) -1- (4- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) piperazin-1-yl) ethan-1-one (190 mg, 60.4 %) as a lihgt yellow oil.
LCMS (ESI) calcd. for C17H22BrN3O2 [M+H] + m/z 380.1, found: 380.5.
Intermediate of Example 15:
Synthesis of (S) -4- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) morpholine
Step 1. Preparation of 3-bromo-2- ( (S) -1-methoxyethyl) -5- (3- ( (tetrahydro-2H-pyran-2-yl) oxy) prop-1-yn-1-yl) pyridine
A mixture of (S) -3-bromo-5-iodo-2- (1-methoxyethyl) pyridine (10.00 g, 29.24 mmol) , 2- (prop-2-yn-1-yloxy) tetrahydro-2H-pyran (4.92 g, 35.09 mmol) , CuI (0.45 g, 2.34 mmol) , TEA (8.88 g, 8.73 mmol) and Pd (PPh3) 2Cl2 (1.64 g, 2.34 mmol) in THF (120.00 mL) was stirred at 50℃ for 4h under N2 atmosphere, the reaction was monitored by LCMS. After completion, the mixture was filtered and the fil-trate was concentrated, the residue was purified by silica gel column chromatography (eluting with EtOAc/PE, from 0%to 3%in 20 min) to obtain 3-bromo-2- ( (S) -1-methoxyethyl) -5- (3- ( (tetrahydro-2H-pyran-2-yl) oxy) prop-1-yn-1-yl) pyridine (10.10 g, yield: 97.5%) as a yellow oil.
LCMS (ESI) calcd. for C16H20BrNO3 [M+H] + m/z 354.1, found 354.5.
Step 2. Preparation of (S) -3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-ol
To a solution of 3-bromo-2- ( (S) -1-methoxyethyl) -5- (3- ( (tetrahydro-2H-pyran-2-yl) oxy) prop-1-yn-1-yl) pyridine (10.10 g, 28.51 mmol) in MeOH (100.00 mL) was added TsOH (19.64 g, 114.05 mmol) at 0℃, the mixture was stirred at room temperature for 5 h under N2 atmosphere, the reaction was monitored by LCMS. After completion, the mixture was diluted with DCM (50.00 mL) and H2O (150.00 mL) and extracted with DCM (50 mL x 3) , the combined layers were washed with saturated NaCl (150.00 mL) , dried over Na2SO4, con-centrated to obtain (S) -3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-ol (7.10 g, yield: 92.2%) as a yellow oil.
LCMS (ESI) calcd. for C11H12BrNO2 [M+H] + m/z 270.0, found 270.3.
Step 3. Preparation of (S) -3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl 4-methylbenzenesulfonate
To a solution of (S) -3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-ol (7.10 g, 26.28 mmol) in THF (100.00 mL) were added TsCl (10.02 g, 52.57 mmol) and KOH (2.95 g, 52.57 mmol) at 0℃, the mixture was stirred at 0℃ for 2 h under N2 atmosphere, the reaction was monitored by LCMS. After com-pletion, the mixture was diluted with EtOAc (50.00 mL) and water (100.00 mL) , then extracted with EA (50.00 mL x 3) . The combined organic phase was washed with brine (100.00 mL) , dried over Na2SO4 and concentrated under reduced pressure. The residual was purified by silica gel column chromatography (eluting with EtOAc/PE, from 0%to 30%in 30 min) to obtain (S) -3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl 4-methylbenzenesulfonate (10.50 g, yield: 94.2%) as a brown solid.
LCMS (ESI) calcd. for C18H18BrNO4S [M+H] + m/z 424.0, found 424.8.
Step 4. Preparation of (S) -4- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) morpholine
To a solution of morpholine (358 mg, 4.11 mmol) in DCM (10 mL) were added DIEA (1.21 g, 9.34 mmol) and a solution of (S) -3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl 4-methylbenzenesulfonate (1.58 g, 3.74 mmol) in DCM (10 mL) at rt, the resulting mixture was stirred at rt for 16 h. The reaction was monitored by LCMS. After completion, the mixture was concentrated under re-duced pressureto give the residual which was purified by silica gel column chromatography (eluting with EtOAc/PE from 0%to 40%) to obtain (S) -4- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) morpholine (980 mg, yield: 77.5%) as a brown oil.
LCMS (ESI) calcd. for C15H19BrNO2 [M+H] + m/z 339.1, found 339.6
Intermediate of Example 17:
Synthesis of (S) -4- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) thiomorpholine 1, 1-dioxide
Step 1. Preparation of 3-bromo-2- ( (S) -1-methoxyethyl) -5- (3- ( (tetrahydro-2H-pyran-2-yl) oxy) prop-1-yn-1-yl) pyridine
A mixture of (S) -3-bromo-5-iodo-2- (1-methoxyethyl) pyridine (10.00 g, 29.24 mmol) , 2- (prop-2-yn-1-yloxy) tetrahydro-2H-pyran (4.92 g, 35.09 mmol) , CuI (0.45 g, 2.34 mmol) , TEA (8.88 g, 8.73 mmol) and Pd (PPh3) 2Cl2 (1.64 g, 2.34 mmol) in THF (120.00 mL) was stirred at 50℃ for 4h under N2 atmosphere, the reaction was monitored by LCMS. After completion, the mixture was filtered and the fil-trate was concentrated, the residue was purified by silica gel column chromatography (eluting with EtOAc/PE, from 0%to 3%in 20 min) to obtain 3-bromo-2- ( (S) -1-methoxyethyl) -5- (3- ( (tetrahydro-2H-pyran-2-yl) oxy) prop-1-yn-1-yl) pyridine 32 (10.10 g, yield: 97.5%) as a yellow oil.
LCMS (ESI) calcd. for C16H20BrNO3 [M+H] + m/z 354.1, found 354.5.
Step 2. Preparation of (S) -3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-ol
To a solution of 3-bromo-2- ( (S) -1-methoxyethyl) -5- (3- ( (tetrahydro-2H-pyran-2-yl) oxy) prop-1-yn-1-yl) pyridine (10.10 g, 28.51 mmol) in MeOH (100.00 mL) was added TsOH (19.64 g, 114.05 mmol) at 0℃, the mixture was stirred at room temperature for 5 h under N2 atmosphere, the reaction was monitored by LCMS. After completion, the mixture was diluted with DCM (50.00 mL) and H2O (150.00 mL) and extracted with DCM (50 mL x 3) , the combined layers were washed with saturated NaCl (150.00 mL) , dried over Na2SO4, con-centrated to obtain (S) -3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-ol (7.10 g, yield: 92.2%) as a yellow oil.
LCMS (ESI) calcd. for C11H12BrNO2 [M+H] + m/z 270.0, found 270.3.
Step 3. Preparation of (S) -3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl 4-methylbenzenesulfonate
To a solution of (S) -3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-ol (7.10 g, 26.28 mmol) in THF (100.00 mL) were added TsCl (10.02 g, 52.57 mmol) and KOH (2.95 g, 52.57 mmol) at 0℃, the mixture was stirred at 0℃ for 2 h under N2 atmosphere, the reaction was monitored by LCMS. After com-pletion, the mixture was diluted with EtOAc (50.00 mL) and water (100.00 mL) , then extracted with EA (50.00 mL x 3) . The combined organic phase was washed with brine (100.00 mL) , dried over Na2SO4 and concentrated under reduced pressure. The residual was purified by silica gel column chromatography (eluting with EtOAc/PE, from 0%to 30%in 30 min) to obtain (S) -3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl 4-methylbenzenesulfonate (10.50 g, yield: 94.2%) as a brown solid.
LCMS (ESI) calcd. for C18H18BrNO4S [M+H] + m/z 424.0, found 424.8.
Step 4. Preparation of (S) -4- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) thiomorpholine 1, 1-dioxide
To a solution of thiomorpholine 1, 1-dioxide (160 mg, 1.18 mmol) in DCM (2.5 mL) were added DIEA (229 mg, 1.77 mmol) and a solution of (S) -3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl 4-methylbenzenesulfonate (250 mg, 0.59 mmol) in DCM (2.5 mL) at rt, the resulting mixture was stirred at rt for 20 h. The reaction was monitored by LCMS. After completion, the mixture was concentrated under reduced pressure to give the residual which was purified by silica gel column chromatography (eluting with EtOAc/PE from 0%to 60%) to ob-tain (S) -4- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) thiomorpholine 1, 1-dioxide (186 mg, yield: 81.7%) as a brown oil. LCMS (ESI) calcd. for C15H19BrN2O3S [M+H] + m/z 387.0, found 387.5.
Intermediate of Example 19:
Synthesis of
( (R) -4- (3- (5-bromo-6- ( (S) -1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) morpholin-3-yl) methanol
Step 1: Synthesis of ( (R) -4- (3- (5-bromo-6- ( (S) -1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) morpholin-3-yl) methanol
To a solution of (R) -morpholin-3-ylmethanol (500 mg, 4.27 mmol) in DCM (10 mL) were added DIEA (1.10 g, 8.51 mmol) and a solution of (S) -3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl 4-methylbenzenesulfonate (1.80 g, 4.24 mmol) in DCM (15 mL) at rt, the resulting mixture was stirred at rt for 16 h. The reaction was monitored by LCMS. After completion, the mixture was concentrated under reduced pressure to give the residual which was purified by silica gel column chromatography (eluting with MeOH/DCM from 0%to 15%) to give ( (R) -4- (3- (5-bromo-6- ( (S) -1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) morpholin-3-yl) methanol (1.1 g, yield: 70.0%) as a brown oil.
LCMS (ESI) calcd. for C16H21BrN2O3 [M+H] + m/z 369.1, found 369.5.
Intermediate of Example 20:
Synthesis of
( (S) -4- (3- (5-bromo-6- ( (S) -1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) morpholin-3-yl) methanol
Step 1: Preparation of ( (S) -4- (3- (5-bromo-6- ( (S) -1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) morpholin-3-yl) methanol
A mixture of (S) -morpholin-3-ylmethanol (61 mg, 0.52 mmol) , (S) -3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl 4-methylbenzenesulfonate (200 mg, 0.47 mmol) in DCM (5 mL) was added DIEA (121.5 mg, 0.94 mmol) . The reaction mixture was stirred at rt for 3h. After completion, the mixture was was concentrated under reduced pressure. The residue was purified by FCC with EtOAc/PE = 0-30%to afford the ( (S) -4- (3- (5-bromo-6- ( (S) -1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) morpholin-3-yl) methanol (148 mg, 85%) as a yellow oil. LCMS (ESI) calcd. for C16H21BrN2O3 [M+H] + m/z 369.1, found 369.5
Intermediate of Example 31:
Synthesis of
(S) -1- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) -4- (methylsulfonyl) piperazine
Step 1: Preparation of tert-butyl (S) -4- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) piperazine-1-carboxylate
To a stirred solution of tert-butyl piperazine-1-carboxylate (145 mg, 0.778 mmol) in DCM (8 mL) was added (S) -3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl 4-methylbenzenesulfonate (300 g, 0.707 mmol) and DIEA (183 mg, 1.414 mmol) . The resulting mixture was stirred at rt for 4h. The reac-tion was concentrated under reduced pressure. The residue was purified by FCC with MeOH/DCM = 0-5%to afford the tert-butyl (S) -4- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) piperazine-1-carboxylate (200 mg, 65%) as yellow oil. LCMS (ESI) calcd. for C20H28BrN3O3 [M+H] + m/z 438.1, found 438.5
Step 2: Preparation of (S) -1- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) piperazine
To a stirred solution of tert-butyl (S) -4- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) piperazine-1-carboxylate (200 mg, 0.456 mmol) in DCM (5 mL) was added TFA (1 mL) . The resulting mixture was stirred at rt for 1h. The reaction was concentrated under reduced pressure to afford the crude (S) -1- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) piperazine (220 mg) as yellow oil. LCMS (ESI) calcd. for C15H20BrN3O [M+H] + m/z 338.1, found 338.5
Step 3: Preparation of (S) -1- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) -4- (methylsulfonyl) piperazine
To a stirred solution of (S) -1- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) piperazine (152 mg, 0.45 mmol) in DCM (5 mL) was added TEA (136.5 mg, 1.35 mmol) and MsCl (51.5 mg, 0.45 mmol) at 0℃. The resulting mixture was stirred at 0℃ for 0.5h. After completion, the reaction was quenched with H2O (10 mL) and extracted with EtOAc (10 mL*3) . The combined organic layer was dried over anhydrous Na2SO4 and concentrated under reduced pressure. The residue was was purified by FCC with MeOH/DCM = 0-5%to afford the (S) -1- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) -4- (methylsulfonyl) piperazine (170 mg, 90%) as a light yellow oil. LCMS (ESI) calcd. for C16H22BrN3O3S [M+H] + m/z 416.1, found 416.5 The following intermediates in Table 3 were prepared using the method described above in step for the preparation of (S) -4- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) morpholine and uti-lizing the appropriate starting materials and modifications.
Table 3. Certain Intermediates containing alkynyl group of the Present invention
Intermediate of Example 29:
Synthesis of N- (dimethylcarbamoyl) -N-methyl-L-valine
Step 1. Preparation of benzyl N- (dimethylcarbamoyl) -N-methyl-L-valinate
A mixture of benzyl (2S) -3-methy1-2- (methylamino) butanoate (500 mg, 2.26 mmol) and dimethyl-carbamyl chloride (1.215 g, 11.3 mmol) in THF (5 mL) , was added TEA (2.286 g, 22.59 mmol and DMAP (276.02 mg, 2.26 mmol) in portions under nitrogen atmosphere. The reaction mixture was stirred at 65℃ for 12 h under nitrogen atmosphere, then quenched with water (100 ml) and was extracted with EtOAc (50 mL x 3) . The combined organic phase was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by reverse phase chromatography to afford benzyl N- (dimethylcarbamoyl) -N-methyl-L-valinate (400 mg, 58.3%yield) as a colorless oil.
LCMS (ESI) calcd. for C16H25N2O3 [M+H] + m/z 293.2, found: 293.4
Step 2. Preparation of N- (dimethylcarbamoyl) -N-methyl-L-valine
A mixture of benzyl N- (dimethylcarbamoyl) -N-methyl-L-valinate (400 mg, 1.37 mmol) and palladi-um hydroxide on carbon (400 mg, 2.85 mmol) in MeOH (10 mL) was stirred for 4 h under hydrogen at-mosphere. The reaction mixture was filtered and the filter cake was washed with MeOH (100 mL x3) . The filtrate was concentrated under reduced pressure to afford N- (dimethylcarbamoyl) -N-methyl-L-valine (200 mg, crude) as a colorless oil.
LCMS (ESI) calcd. for C9H19N2O3 [M+H] + m/z 203.1, found: 203.1
The following compounds in Table 4 were prepared according to the representative procedure de-scribed above for the synthesis of N-methyl-N- ( (2R, 3R) -3-phenyltetrahydrofuran-2-carbonyl) -L-valine and utilizing the appropriate starting materials and modifications.
Table 4. Certain Peptides of the Present invention
SPECIFIC EMBODIMENTS
The present invention is described in detail below by embodiments, but does not imply any adverse restriction on the present invention. The compounds of the present invention can be prepared by a variety of synthesis methods well known to those skilled in the art, including the specific embodiments listed be-low, the embodiments formed by the combination of the invention with other chemical synthesis methods, and the equivalent substitution methods well known to those skilled in the art. Preferred embodiments in-clude, but are not limited to, embodiments of the present invention. To those skilled in the art, various changes and improvements to the specific embodiments of the present invention without departing from the spirit and scope of the invention will be obvious and shall also be regarded as the scope of protection of the present invention.
Each of the compounds set forth was prepared following one of the procedures set forth below. Unless otherwise specified, intermediates that do not mention the synthesis method in the following synthesis routes are obtained by purchase or customization
Example 1
(1S, 2S) -N- ( (64S, 4S, Z) -11-ethyl-12- (5-ethynyl-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-di-oxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2-methylcyclopropane-1-carboxamide
Method 1:
Substituting (S) -4- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) -2-methylbut-3-yn-2-ol with (S) -3-bromo-5-ethynyl-2- (1-methoxyethyl) pyridine in the Step 1 of Example 2 and Substituting (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxylic acid with (1S, 2S) -2-methylcyclopropane-1-carboxylic acid in the Step 4 of Example 2, the title compound was prepared by the same procedures as described for Example 2.
Method 2:
Step 1: Synthesis of (S) -5-bromo-6- (1-methoxyethyl) pyridin-3-ol
To a stirred solution of crude (S) -3-bromo-2- (1-methoxyethyl) -5- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyridine (1.00 g, 2.93 mmol) in THF (10 mL) and H2O (15 mL) were added NaOH (234 mg, 5.86 mmol ) and H2O2 (195 mg, 14.6 mmol) at rt. The resulting mixture was stirred at rt for 2 h. After completion, the reaction mixture was acified by 1M HCl aquous to pH = 4. The mixture was extracted with EtOAc (2*20 mL) . The organic lay-ers were combined, washed with brine, dried over Na2SO4 and concentrated to give the crude product which was purified by silica gel column (eluting with EtOAc/PE from 0%to 80%) to give (S) -5-bromo-6- (1-methoxyethyl) pyridin-3-ol (600 mg, 88%) as a white solid.
LCMS (ESI) calcd. for C8H10BrNO2 [M+H] + m/z 231.0, found: 232.3
Step 2: Synthesis of tert-butyl ( (64S, 4S, Z) -12- (5-hydroxy-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
A mixture of (S) -5-bromo-6- (1-methoxyethyl) pyridin-3-ol (329 mg, 1.42 mmol) , tert-butyl ( (64S, 4S, Z) -10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (500 mg, 0.709 mmol) , K2CO3 (386 mg, 2.84 mmol) and Pd (dppf) Cl2 (58 mg, 0.071 mmol) in dioxane (16 mL) and H2O (4 mL) was stirred at 85℃ under N2 for 19 h. After completion, the reaction mixture was concentrated to give the crude product which was purified by silica gel column (eluting with EtOAc 100%) to give tert-butyl ( (64S, 4S, Z) -12- (5-hydroxy-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (500 mg, 96%) as a brown solid.
LCMS (ESI) calcd. for C38H46N6O7S [M+H] + m/z 730.3, found: 731.8
Step 3: Synthesis of 5- ( (64S, 4S, Z) -4- ( (tert-butoxycarbonyl) amino) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiaz ola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl trifluoromethanesulfonate
To a solution of tert-butyl ( (64S, 4S, Z) -12- (5-hydroxy-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63 -diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (410 mg, 0.561 mmol) in DCM (6 mL) were added TEA (116 mg, 1.1 mmol) and N,N-Bis (trifluoromethanesulfonyl) aniline (228 g, 0.638 mmol) at 0℃, the mixture was stirred at 0℃ for 2 h under N2 atmosphere, the reaction was monitored by LCMS. After completion, the mixture was concen-trated under reduced pressure. The residual was purified by silica gel column chromatography (eluting with EtOAc/PE, from 0%to 50%in 20 min) to obtain 5- ( (64S, 4S, Z) -4- ( (tert-butoxycarbonyl) amino) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazo-la-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl trifluoromethanesulfonate (390 mg, yield: 80%) as a white solid.
LCMS (ESI) calcd. for C39H45F3N6O9S2 [M+H] + m/z 862.3, found 864.2.
Step 4: Synthesis of tert-butyl ( (64S, 4S, Z) -12- (2- ( (S) -1-methoxyethyl) -5- ( (trimethylsilyl) ethynyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11 H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
A mixture of 5- ( (64S, 4S, Z) -4- ( (tert-butoxycarbonyl) amino) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazo-la-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl trifluoromethanesulfonate (340 mg, 0.394 mmol) , CuI (7 mg, 0.039 mmol) , TEA (199 mg, 1.97 mmol) and Pd (PPh3) 4 (45 mg, 0.039 mmol) in THF (10 mL) was stirred at 75℃ for 4h under N2 atmosphere, the reac-tion was monitored by LCMS. After completion, the mixture was filtered and the filtrate was concentrated, the residue was purified by silica gel column chromatography (eluting with EtOAc/PE, from 0%to 50%in 10 min) to obtain tert-butyl ( (64S, 4S, Z) -12- (2- ( (S) -1-methoxyethyl) -5- ( (trimethylsilyl) ethynyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (170 mg, yield: 53%) as a yellow oil.
LCMS (ESI) calcd. for C43H54N6O6SSi [M+H] + m/z 810.4, found 812.3.
Step 5: Synthesis of tert-butyl ( (64S, 4S, Z) -11-ethyl-12- (5-ethynyl-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
To a stirred solution of tert-butyl ( (64S, 4S, Z) -12- (2- ( (S) -1-methoxyethyl) -5- ( (trimethylsilyl) ethynyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (120 mg, 0.148 mmol) in DMF (2 mL) were added Cs2CO3 (145 mg, 0.444 mmol) and Iodoethane (81 mg, 0.518 mmol) at 0℃, the resulting mixture was stirred at rt overnight. The reaction was monitored by LCMS. After completion, the reaction mixture was diluted with EtOAc (10 mL) , washed with water (10 mL) and saturated NaCl (5 mL) , then dried over Na2SO4, following with concentration under reduced pressure to obtain crude product which was purified by silica gel column chromatography (eluting with EtOAc/PE, from 0%to 50%in 10 min) to obtain tert-butyl ( (64S, 4S, Z) -11-ethyl-12- (5-ethynyl-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (100 mg, yield: 88%) as a yellow solid.
LCMS (ESI) calcd. for C42H50N6O6S [M+H] + m/z 766.4, found: 768.4
Step 6: Synthesis of (64S, 4S, Z) -4-amino-11-ethyl-12- (5-ethynyl-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione To a stirred solution of tert-butyl ( (64S, 4S, Z) -11-ethyl-12- (5-ethynyl-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (120 mg, 0.156 mmol) in dichloromethane (2 mL) was added TFA (1 mL) dropwisely at rt, the resulting mixture was stirred at rt for 1 h. The reaction mixture was concentrated. The residue was dissolved in EtOAc (15 mL) . The mixture was washed with sat. NaHCO3 aquous (10 mL) and brine (10 mL) , then dried over Na2SO4, following with concentration under reduced pressure to obtain (64S, 4S, Z) -4-amino-11-ethyl-12- (5-ethynyl-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione (100 mg, yield: 96%) as a white solid.
LCMS (ESI) calcd. for C37H42N6O4S [M+H] + m/z 666.3, found: 667.7
Step 7: Synthesis of (1S, 2S) -N- ( (64S, 4S, Z) -11-ethyl-12- (5-ethynyl-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2-methylcyclopropane-1-carboxamide
To a stirred solution of (64S, 4S, Z) -4-amino-11-ethyl-12- (5-ethynyl-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione (100 mg, 0.150 mmol) and (1S, 2S) -2-methylcyclopropane-1-carboxylic acid (18 mg, 0.18 mmol) and 1-methyl-1H-imidazole (37 mg, 0.45 mmol) in ACN (2 mL) was added TCFH (63 mg, 0.225 mmol) at rt, the resulting mixture was stirred at rt for 1 h. The reaction mixture was concentrated to give the crude product. The crude product was purified by pre-HPLC (ACN/H2O (0.5 %NH4HCO3) from 20%to 95%in 30 min) to give (1S, 2S) -N- ( (64S, 4S, Z) -11-ethyl-12- (5-ethynyl-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2-methylcyclopropane-1-carboxamide as atropisomer 1 (18 mg, 15%) as a white solid and (1S, 2S) -N- ( (64S, 4S, Z) -11-ethyl-12- (5-ethynyl-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2-methylcyclopropane-1-carboxamide as atropisomer 2 (i.e., compound of Example 1) (17 mg, 15%) as a white solid.
LCMS (ESI) calcd. for C42H48N6O5S [M+H] + m/z 748.3, found: 749.8.
atropisomer 1: 1H NMR (400 MHz, DMSO-d6) δ 8.84 (d, J = 1.8 Hz, 1H) , 8.51 (d, J = 8.9 Hz, 1H) , 8.41 (s, 1H) , 8.06 (d, J = 2.0 Hz, 1H) , 7.80 (s, 1H) , 7.72 (d, J = 8.9 Hz, 1H) , 7.52 (d, J = 8.6 Hz, 1H) , 5.89 (d, J =11.0 Hz, 1H) , 5.30 (t, J = 8.3 Hz, 1H) , 4.71 (d, J = 11.1 Hz, 1H) , 4.50 -4.44 (m, 2H) , 3.96-3.89 (m, 2H) , 3.81-3.76 (m, 1H) , 3.60-3.48 (m, 2H) , 3.24 (d, J = 14.8 Hz, 1H) , 3.16 -2.97 (m, 6H) , 2.65-2.63 (m, 1H) , 2.35 -2.28 (m, 2H) , 2.13 (t, J = 9.8 Hz, 1H) , 1.60 (t, J = 9.3 Hz, 1H) , 1.49 (s, 1H) , 1.18 (d, J = 6.2 Hz, 3H) , 1.11-1.06 (m, 7H) , 0.90 (s, 4H) , 0.56-0.55 (m, , 1H) , 0.45 (s, 3H) .
atropisomer 2 (i.e., compound of Example 1) : 1H NMR (400 MHz, DMSO-d6) δ 8.83 (d, J = 2.0 Hz, 1H) , 8.50 (d, J = 8.9 Hz, 1H) , 8.40 (s, 1H) , 7.91 (d, J = 2.0 Hz, 1H) , 7.80 (s, 1H) , 7.73 (d, J = 9.9 Hz, 1H) , 7.56 (d, J = 8.6 Hz, 1H) , 5.92 (d, J = 11.1 Hz, 1H) , 5.39-5.35 (m, 1H) , 4.65 (d, J = 11.2 Hz, 1H) , 4.52 (s, 1H) , 4.49 -4.44 (m, 1H) , 4.34 -4.23 (m, 2H) , 4.07-4.01 (m, 1H) , 3.57-3.49 (m, 2H) , 3.27 -3.08 (m, 6H) , 2.91 (d, J = 14.3 Hz, 1H) , 2.62-2.60 (m, 1H) , 2.47-2.42 (m, 1H) , 2.32-2.29 (m, 1H) , 2.13 (t, J = 9.9 Hz, 1H) , 1.54 (t, J = 9.5 Hz, 1H) , 1.49-1.47 (m, 1H) , 1.34 (d, J = 6.1 Hz, 3H) , 1.05 (s, 4H) , 0.91 -0.81 (m, 7H) , 0.55-0.54 (m, 2H) , 0.30 (s, 3H) .
Example 2
(1S, 2S) -N- ( (63S, 4S, Z) -11-ethyl-12- (5- (3-hydroxy-3-methylbut-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (1, 3) -pyridazinacycloundecaphane-4-yl) -2-methylcyclopropane-1-carboxamide
Step 1: Preparation of tert-butyl ( (64S, 4S, Z) -12- (5- (3-hydroxy-3-methylbut-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
A mixture of (S) -4- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) -2-methylbut-3-yn-2-ol (106 mg, 0.37 mmol) , tert-butyl ( (64S, 4S, Z) -10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (307 mg, 0.44 mmol) , K2CO3 (127 mg, 0.92 mmol) and Pd (dppf) Cl2 (27 mg, 0.037 mmol) in toluene (6 mL) , dioxane (2 mL) and H2O (2 mL) was stirred at 75℃ under N2 for 22 h. After completion, the reaction mixture was concentrated to give the crude product which was purified by silica gel column (eluting with MeOH/DCM from 0%to 100%) to give tert-butyl ( (64S, 4S, Z) -12- (5- (3-hydroxy-3-methylbut-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl -5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (159 mg, 54.8 %) as a brown solid.
LCMS (ESI) calcd. for C42H52N6O7S [M+H] + m/z 784.36, found: 776.5
Step 2: Preparation of tert-butyl ( (64S, 4S, Z) -11-ethyl-12- (5- (3-hydroxy-3-methylbut-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dime-thyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
To a stirred solution of tert-butyl ( (64S, 4S, Z) -12- (5- (3-hydroxy-3-methylbut-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl -5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (240 mg, 0.31 mmol) in DMF (3.5 mL) was added Cs2CO3 (200 mg, 0.61 mmol) and Iodoethane (96 mg, 0.61 mmol) at 0℃, the resulting mixture was stirred at rt for 3 h. The reaction was monitored by LCMS. After completion, the reaction mixture was diluted with EtOAc (100 mL) , washed with water (100 mL x 3) and saturated NaCl (100 x 2 mL) , then dried over Na2SO4, following with con-centration under reduced pressure to obtain crude product which was purified by silica gel column (eluting with MeOH/DCM from 0%to 5%) to give tert-butyl ( (64S, 4S, Z) -11-ethyl-12- (5- (3-hydroxy-3-methylbut-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dime-thyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate as two atropisomers (190 mg, 75.5 %) . Then the mixture was purified by prep-HPLC (Prep-HPLC method: column: Phenomenex Luna Cis75x30mmx3um; mobile phase: [wa-ter (FA) -ACN] ; B%: 1%-40%, 8min) to give the atropisomer 1 (84 mg, 34%) as a brown foam.
LCMS (ESI) calcd. for C44H56N6O7S [M+H] + m/z 812.39, found: 814.5
Step 3: Preparation of (64S, 4S, Z) -4-amino-11-ethyl-12- (5- (3-hydroxy-3-methylbut-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione
To a stirred solution of tert-butyl ( (64S, 4S, Z) -11-ethyl-12- (5- (3-hydroxy-3-methylbut-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dime-thyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (atropisomer 1) (84 mg, 0.10 mmol) in dichloromethane (4 mL) was added TFA (1 mL) dropwise at rt, the resulting mixture was stirred at rt for 1h. The reaction mixture was concentrated to give the residue. The residue was purified with reverse phase chromatography (eluting with ACN/H2O (0.5 %NH4HCO3) from 0%to 60%) to give (64S, 4S, Z) -4-amino-11-ethyl-12- (5- (3-hydroxy-3-methylbut-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl ) -10, 10-dimethyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione (62 mg, 93.6 %) as a yellow solid.
LCMS (ESI) calcd. for C39H48N6O5S [M+H] + m/z 712.34, found: 714.1
Step 4: Preparation of (1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (5- (3-hydroxy-3-methylbut-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
To a stirred solution of (63S, 4S, Z) -4-amino-11-ethyl-12- (5- (3-hydroxy-3-methylbut-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (1, 3) -pyridazinacyc loundecaphane-5, 7-dione (62 mg, 0.10 mmol) and (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxylic acid (11 mg, 0.11 mmol) and 2, 6-lutidine (11 mg, 0.44 mmol) in ACN (8 mL) was added COMU (56 mg, 0.13 mmol) at rt, the resulting mixture was stirred at rt for 2h. The reaction mixture was concentrated to give the crude product. The crude product was purified by HPLC (ACN/H2O (0.5 %NH4HCO3) from 20%to 95%in 30min) to give (1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (5- (3-hydroxy-3-methylbut-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide (40 mg, 64.5 %) as a white solid.
LCMS (ESI) calcd. for C44H54N6O6S [M+H] + m/z 794.38, found: 796.3. 1H NMR (400 MHz, DMSO-d6) δ 8.75 (d, J = 2.1 Hz, 1H) , 8.55 -8.48 (m, 2H) , 7.81 (s, 1H) , 7.78 -7.73 (m, 2H) , 7.58 (d, J =8.7 Hz, 1H) , 5.61 -5.51 (m, 2H) , 5.08 (d, J = 12.3 Hz, 1H) , 4.37 -4.14 (m, 4H) , 4.07 (dd, J = 14.7, 7.3 Hz, 1H) , 3.57 (s, 2H) , 3.34 (s, 1H) , 3.30 (s, 2H) , 3.25 (s, 3H) , 3.14 (dd, J = 14.5, 9.1 Hz, 1H) , 2.98 (d, J = 14.5 Hz, 1H) , 2.81 -2.70 (m, 1H) , 2.36 (d, J = 14.1 Hz, 1H) , 2.08 (d, J = 10.2 Hz, 1H) , 1.79 (s, 2H) , 1.49 (s, 7H) , 1.35 (d, J = 6.0 Hz, 3H) , 1.05 (d, J = 10.9 Hz, 4H) , 0.88 (dd, J = 17.3, 10.4 Hz, 6H) , 0.54 (d, J = 5.4 Hz, 1H) , 0.33 (s, 3H) .
Example 3
(1S, 2S) -N- ( (64S, 4S, Z) -11-ethyl-12- (5- ( (1-fluorocyclopropyl) ethynyl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2-methylcyclopropane-1-carboxamide
Substituting (S) -4- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) -2-methylbut-3-yn-2-ol with (S) -3-bromo-5- ( (1-fluorocyclopropyl) ethynyl) -2- (1-methoxyethyl) pyridine in the Step 1 of Example 2 and Substituting (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxylic acid with (1S, 2S) -2-methylcyclopropane-1-carboxylic acid in the Step 4 of Example 2, the title compound was pre-pared by the same procedures as described for Example 2. LCMS (ESI) : 807.4 [M+1] +. 1H NMR (500 MHz, CDCl3) δ 8.28 (d, J = 2.0 Hz, 2H) , 8.08 (d, J = 12.6 Hz, 2H) , 7.92 (d, J = 2.0 Hz, 2H) , 7.77 (d, J =2.4 Hz, 2H) , 7.69 (dd, J = 7.7, 2.4 Hz, 2H) , 7.59 (s, 2H) , 7.48 (d, J = 7.7 Hz, 2H) , 5.24 (q, J = 5.4 Hz, 2H) , 4.90 (d, J = 8.4 Hz, 2H) , 4.64 (q, J = 12.4 Hz, 2H) , 4.15 (q, J = 4.4 Hz, 4H) , 4.08 -3.99 (m, 4H) , 3.99 -3.89 (m, 4H) , 3.33 (s, 6H) , 3.31 -3.23 (m, 3H) , 3.22 (d, J = 3.5 Hz, 1H) , 3.06 (d, J = 2.6 Hz, 4H) , 2.72 (h, J = 7.9 Hz, 2H) , 2.23 (q, J = 4.9 Hz, 8H) , 2.13 -1.97 (m, 8H) , 1.94 (td, J = 10.2, 7.3 Hz, 2H) , 1.69 -1.60 (m, 8H) , 1.55 (ddd, J = 9.9, 8.8, 4.9 Hz, 2H) , 1.37 (t, J = 4.4 Hz, 6H) , 1.16 -1.04 (m, 14H) , 0.90 (d, J = 5.0 Hz, 6H) .
Example 4
(1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( ( (S) -1-methylpyrrolidin-2-yl) ethynyl) pyri-din-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
Method 1:
Substituting 3-bromo-2- ( (S) -1-methoxyethyl) -5- ( ( (S) -1-methylpyrrolidin-3-yl) ethynyl) pyridine with 3-bromo-2- ( (S) -1-methoxyethyl) -5- ( ( (S) -1-methylpyrrolidin-2-yl) ethynyl) pyridine in the Step 3 of Exam-ple 5, the title compound was prepared by the same procedures as described for Example 5.
Method 2:
Step 1: Synthesis of tert-butyl ( (64S, 4S, Z) -12- (2- ( (S) -1-methoxyethyl) -5- ( ( (S) -1-methylpyrrolidin-2-yl) ethynyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
A mixture of 3-bromo-2- ( (S) -1-methoxyethyl) -5- ( ( (S) -1-methylpyrrolidin-2-yl) ethynyl) pyridine (87 mg, 0.27 mmol) , tert-butyl ( (64S, 4S, Z) -10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (210 mg, 0.30 mmol) , K2CO3 (112 mg, 0.81 mmol) and Pd (dppf) Cl2 (30 mg, 0.041 mmol) in dioxane (3 mL) and H2O (0.7 mL) was stirred at 85℃ under N2 for 16 h. After completion, the reaction mixture was concen-trated to give the crude product which was purified by silica gel column (eluting with MeOH/DCM from 0%to 10%) to give tert-butyl ( (64S, 4S, Z) -12- (2- ( (S) -1-methoxyethyl) -5- ( ( (S) -1-methylpyrrolidin-2-yl) ethynyl) pyridin-3-yl) -10, 10-dimet hyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (150 mg, 67.7 %) as a yellow solid.
LCMS (ESI) calcd. for C45H55N7O6S [M+H] + m/z 822.4, found: 823.6.
Step 2: Synthesis of tert-butyl ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( ( (S) -1-methylpyrrolidin-2-yl) ethynyl) pyridin-3-yl) -10, 10 -dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
To a stirred solution of tert-butyl ( (64S, 4S, Z) -12- (2- ( (S) -1-methoxyethyl) -5- ( ( (S) -1-methylpyrrolidin-2-yl) ethynyl) pyridin-3-yl) -10, 10-dimet hyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (147 mg, 0.18 mmol) in DMF (2 mL) were added Cs2CO3 (105 mg, 0.32 mmol) and Iodoethane (50 mg, 0.32 mmol) at 0℃, the resulting mixture was stirred at rt for 3 h. The reaction was monitored by LCMS. After completion, the reaction mixture was diluted with EtOAc (20 mL) , washed with water (30 mL x 2) and saturated NaCl (50 mL) , then dried over Na2SO4, following with concentration under reduced pressure to obtain crude product which was purified by pre-TLC (eluting with DCM: MeOH = 15 : 1) to give tert-butyl ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( ( (S) -1-methylpyrrolidin-2-yl) ethynyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacy cloundecaphane-4-yl) carbamate (26 mg, 16.9%) as atropisomer 1 and tert-butyl ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( ( (S) -1-methylpyrrolidin-2-yl) ethynyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacy cloundecaphane-4-yl) carbamate (17 mg, 11.1%) as atropisomer 2 as pale yellow solid.
LCMS (ESI) calcd. for C47H59N7O6S [M+H] + m/z 850.4, found: 851.4.
Step 3: Synthesis of (64S, 4S, Z) -4-amino-11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( ( (S) -1-methylpyrrolidin-2-yl) ethynyl) pyridin-3-yl) -10, 10-dimethyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacyclou ndecaphane-5, 7-dione
To a stirred solution of tert-butyl ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( ( (S) -1-methylpyrrolidin-2-yl) ethynyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacy cloundecaphane-4-yl) carbamate as atropisomer 2 (17 mg, 0.020 mmol) in dichloromethane (1 mL) was added TFA (0.25 mL) dropwisely at rt, the resulting mixture was stirred at rt for 2 h. The reaction mixture was concentrated to give crude (64S, 4S, Z) -4-amino-11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( ( (S) -1-methylpyrrolidin-2-yl) ethynyl) pyridin-3 -yl) -10, 10-dimethyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacyclo undecaphane-5, 7-dione which was used to next step without purification.
LCMS (ESI) calcd. for C42H51N7O4S [M+H] + m/z 750.4, found: 750.9.
Step 4: Synthesis of (1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( ( (S) -1-methylpyrrolidin-2-yl) ethynyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
To a stirred solution of crude (64S, 4S, Z) -4-amino-11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( ( (S) -1-methylpyrrolidin-2-yl) ethynyl) pyridin-3-yl) -10, 10-dimethyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacyclo undecaphane-5, 7-dione (17 mg, 0.020 mmol) and (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxylic acid (2.2 mg, 0.020 mmol) and 2, 6-lutidine (13 mg, 0.12 mmol) in ACN (1 mL) was added COMU (13 mg, 0.030 mmol) at rt, the resulting mixture was stirred at rt for 2 h. The reaction mixture was concentrated to give the crude product. The crude product was purified by pre-HPLC (ACN/H2O (0.5 %NH4HCO3) from 20%to 95%in 30 min) to give (1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( ( (S) -1-methylpyrrolidin-2-yl) ethynyl) pyrid-in-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide (5.6 mg) as a white solid.
LCMS (ESI) : 846.4 [M+1] +. 1H NMR (400 MHz, DMSO) δ 8.76 (d, J = 2.1 Hz, 1H) , 8.42 -8.34 (m, 2H) , 7.83 -7.77 (m, 1H) , 7.73 (d, J = 8.7 Hz, 1H) , 7.56 (d, J = 8.5 Hz, 1H) , 5.90 (d, J = 11.0 Hz, 1H) , 5.36 (s, 1H) , 4.64 (d, J = 10.7 Hz, 1H) , 4.46 (d, J = 4.8 Hz, 1H) , 4.33 -4.20 (m, 2H) , 4.04 (dd, J = 14.4, 7.3 Hz, 1H) , 3.60 -3.46 (m, 2H) , 3.40 -3.32 (m, 1H) , 3.21 (s, 3H) , 3.12 (dd, J = 14.9, 7.3 Hz, 1H) , 2.91 (d, J =13.7 Hz, 1H) , 2.82 -2.72 (m, 1H) , 2.61 (d, J = 5.8 Hz, 1H) , 2.40 (d, J = 16.0 Hz, 2H) , 2.37 -2.27 (m, 4H) , 2.19 -2.07 (m, 2H) , 1.84 (ddd, J = 46.4, 30.9, 20.3 Hz, 4H) , 1.56 (d, J = 9.6 Hz, 1H) , 1.33 (t, J = 6.6 Hz, 3H) , 1.20 (d, J = 10.7 Hz, 4H) , 1.15 (d, J = 3.2 Hz, 2H) , 1.10 -1.00 (m, 4H) , 0.92 -0.77 (m, 5H) , 0.31 (s, 3H) .
Example 5
(1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( ( (S) -1-methylpyrrolidin-3-yl) ethynyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
Step 1: Preparation of (64S, 4S, Z) -4-amino-10, 10-dimethyl-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione
To a stirred solution of tert-butyl ( (64S, 4S, Z) -10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (1.5 g, 2.1 mmol) in dichloromethane (30 mL) was added TFA (3 mL) dropwise at rt, the resulting mixture was stirred at rt for 1 h. The reaction mixture was washed with sat. NaHCO3 (30 mL) , brine (30 mL) , then dried over Na2SO4, following with concentration under reduced pressure to obtain crude product (64S, 4S, Z) -4-amino-10, 10-dimethyl-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione (1.3 g) as a yel-low solid.
LCMS (ESI) calcd. for C31H40BN5O5S [M+H] + m/z 606.3, found: 607.4
Step 2: Preparation of (1r, 2R, 3S) -N- ( (64S, 4S, Z) -10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
To a stirred solution of (64S, 4S, Z) -4-amino-10, 10-dimethyl-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione (1.3 g, 2.1 mmol) and (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxylic acid (242 mg, 2.1 mmol) and NMI (436 mg, 5.3 mmol) in ACN/DCM (15 mL/15 mL) was added TCFH (896 mg, 3.2 mmol) at rt, the resulting mixture was stirred at rt for 30 min. The reaction mixture was concentrated to give the crude product. The crude product was purified by silica gel column (eluting with EtOAc/PE from 0%to 80%) to give (1r, 2R, 3S) -N- ( (64S, 4S, Z) -10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide (1.5 g, 99%in 2 steps) as a yellow solid.
LCMS (ESI) calcd. for C37H48BN5O6S [M+H] + m/z 702.3, found: 703.5
Step 3: Preparation of tert-butyl (3S) -3- ( (5- ( (64S, 4S, Z) -4- ( (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxamido) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl) ethynyl) pyrrolidine-1-carboxylate
A mixture of (1r, 2R, 3S) -N- ( (64S, 4S, Z) -10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide (1 g, 1.4 mmol) , tert-butyl (S) -3- ( (5-bromo-6- ( (S) -1-methoxyethyl) pyridin-3-yl) ethynyl) pyrrolidine-1-carboxylate (583 mg, 1.4 mmol) , K2CO3 (491 mg, 3.6 mmol) and Pd (dppf) Cl2 (209 mg, 0.3 mmol) in dioxane (40 mL) and H2O (8 mL) was stirred at 90℃ under N2 for 19 h. After completion, the reaction mixture was concentrated to give the crude product which was purified by silica gel column (eluting with EA/PE from 0%to 100%) to give tert-butyl (3S) -3- ( (5- ( (64S, 4S, Z) -4- ( (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxamido) -10, 10-dimethyl-5, 7-dioxo -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl) ethynyl) pyrrolidine-1-carboxylate (900 mg, 70 %) as a yellow solid.
LCMS (ESI) calcd. for C50H61N7O7S [M+H] + m/z 904.4, found: 904.9
Step 4: Preparation of tert-butyl (3S) -3- ( (5- ( (64S, 4S, Z) -4- ( (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxamido) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl) ethynyl) pyrrolidine-1-carboxylate
To a stirred solution of (1r, 2R, 3S) -N- ( (64S, 4S, Z) -14-amino-10, 10-dimethyl-5, 7-dioxo-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-6 (2, 4) -bicyclo [3.1.1] heptana-1 (1, 3) -benzenacyclododecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide (900 mg, 1.0 mmol) in DMF (9 mL) were added Cs2CO3 (649 mg, 2.0 mmol) and Iodoethane (311 mg, 2.0 mmol) at 0℃, the resulting mixture was stirred at rt for 3 h. The reaction was monitored by LCMS. After completion, the reaction mixture was diluted with EtOAc (100 mL) , washed with water (50 mL x 3) and saturated NaCl (50 x 2 mL) , then dried over Na2SO4, following with concentration under reduced pressure to obtain crude product which was purified by silica gel column (eluting with EA/PE from 0%to 100%) to give tert-butyl (3S) -3- ( (5- ( (64S, 4S, Z) -4- ( (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxamido) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl) ethynyl) pyrrolidine-1-carboxylate as two atropisomers (927 mg, 99%) . Then the mixture was purified by prep-HPLC (Prep-HPLC method: column: Phenomenex Luna Cis75x30mmx3um; mobile phase: [water (FA) -ACN] ; B%: 1%-40%, 8min) to give the atropisomer 1 (412 mg, 44%) as yellow solids.
LCMS (ESI) calcd. for C52H65N7O7S [M+H] + m/z 932.5, found: 933.6
Step 5: Preparation of (1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( ( (S) -pyrrolidin-3-yl) ethynyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptan acycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
To a stirred solution of tert-butyl (3S) -3- ( (5- ( (64S, 4S, Z) -4- ( (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxamido) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl) ethynyl) pyrrolidine-1-carboxylate (atropisomer 1) (412 mg, 1.0 mmol) in dichloromethane (10 mL) was added TFA (2.5 mL) dropwise at rt, the resulting mixture was stirred at rt for 1 h. The reaction mixture was washed with sat. NaHCO3 (30 mL) , brine (30 mL) , then dried over Na2SO4, following with concentration under reduced pressure to obtain crude product (1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( ( (S) -pyrrolidin-3-yl) ethynyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptan acycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide (276 mg, 75%) as a yellow solid.
LCMS (ESI) calcd. for C47H57N7O5S [M+H] + m/z 832.4, found: 832.9
Step 6: Preparation of (1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( ( (S) -1-methylpyrrolidin-3-yl) ethynyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1 ] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
To a stirred solution of (1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( ( (S) -pyrrolidin-3-yl) ethynyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptan acycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide (276 mg, 0.33 mmol) in MeOH (12 mL) were added formaldehyde (266 mg, 3.3 mmol, 37%in water) and NaBH3CN (62 mg, 0.97 mmol) at 0℃, the resulting mixture was stirred at rt for 1 h. The reaction was monitored by LCMS. After completion, the reaction mixture was diluted with EtOAc (100 mL) , washed with water (50 mL x 3) and saturated NaCl (50 x 2 mL) , then dried over Na2SO4, following with concentration under reduced pressure to obtain crude product which was purified by pre-HPLC (eluting with CH3CN/H2O (0.1%NH4HCO3) from 20%to 80%) to give (1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( ( (S) -1-methylpyrrolidin-3-yl) ethynyl) pyrid-in-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide (156 mg, 25%) as a white solid.
LCMS (ESI) calcd. for C48H59N7O5S [M+H] + m/z 846.4, found: 846.9.
1H NMR (400 MHz, DMSO-d6) δ 8.72 (s, 1H) , 8.40-8.37 (m, 2H) , 7.80 (s, 1H) , 7.76-7.72 (m, 2H) , 7.55 (d, J = 8.4 Hz, 1H) , 5.90 (d, J = 11.2 Hz, 1H) , 5.35 (m, 1H) , 4.64 (d, J = 11.2 Hz, 1H) , 4.47-4.46 (m, 1H) , 4.30 -4.22 (m, 2H) , 4.07-4.03 (m, 1H) , 3.57 -3.50 (m, 2H) , 3.22-3.20 (m, 5H) , 3.14-3.09 (m, 1H) , 2.93-2.89 (m, 1H) , 2.81 (t, J = 8.4 Hz, 1H) , 2.61-2.60 (m, 1H) , 2.52 -2.48 (m, 5H) , 2.45-2.37 (m, 2H) , 2.32-2.30 (m, 1H) , 2.23-2.11 (m, 4H) , 1.89-1.81 (m, 1H) , 1.57 -1.55 (m, 1H) , 1.30 (d, J = 6.0 Hz, 1H) , 1.21-1.14 (m, 1H) , 1.08-1.04 (m, 5H) , 0.86-0.83 (m, 5H) , 0.30 (s, 3H) .
Example 6
(1S, 2S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( (1-methyl-1H-pyrazol-4-yl) ethynyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2-methylcyclopropane-1-carboxamide
Method 1:
Substituting (S) -4- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) -2-methylbut-3-yn-2-ol with (S) -3-bromo-2- (1-methoxyethyl) -5- ( (1-methyl-1H-pyrazol-4-yl) ethynyl) pyridine in the Step 1 of Example 2 and Substituting (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxylic acid with (1S, 2S) -2-methylcyclopropane-1-carboxylic acid in the Step 4 of Example 2, the title compound was pre-pared by the same procedures as described for Example 2.
Method 2:
Step 1: Synthesis of (1S, 2S) -N- ( (64S, 4S, Z) -12- (2- ( (S) -1-methoxyethyl) -5- ( (1-methyl-1H-pyrazol-4-yl) ethynyl) pyridin-3-yl) -10, 10 -dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2-methylcyclopropane-1-carboxamide
A mixture of (S) -3-bromo-2- (1-methoxyethyl) -5- ( (1-methyl-1H-pyrazol-4-yl) ethynyl) pyridine (43 mg, 0.13 mmol) , (64S, 4S, Z) -4-amino-10, 10-dimethyl-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione (100 mg, 0.15 mmol) , K2CO3 (56 mg, 0.40 mmol) and Pd (dppf) Cl2 (15 mg, 0.020 mmol) in dioxane (2 mL) and H2O (0.5 mL) was stirred at 85℃ under N2 for 5 h. After completion, the reaction mixture was concentrated to give the crude product which was purified by silica gel column (eluting with MeOH/DCM from 0%to 10%) to give (1S, 2S) -N- ( (64S, 4S, Z) -12- (2- ( (S) -1-methoxyethyl) -5- ( (1-methyl-1H-pyrazol-4-yl) ethynyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacy cloundecaphane-4-yl) -2-methylcyclopropane-1-carboxamide (58 mg, 55.8 %) as a yellow solid.
LCMS (ESI) calcd. for C44H48N8O5S [M+H] + m/z 801.4, found: 801.8
Step 2: Synthesis of (1S, 2S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( (1-methyl-1H-pyrazol-4-yl) ethynyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2-methylcyclopropane-1-carboxamide
To a stirred solution of (1S, 2S) -N- ( (64S, 4S, Z) -12- (2- ( (S) -1-methoxyethyl) -5- ( (1-methyl-1H-pyrazol-4-yl) ethynyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacy cloundecaphane-4-yl) -2-methylcyclopropane-1-carboxamide (58 mg, 0.073 mmol) in DMF (1.5 mL) were added Cs2CO3 (47 mg, 0.15 mmol) and Iodoethane (23 mg, 0.15 mmol) at 0℃, the resulting mixture was stirred at rt for 22 h. The reaction was monitored by LCMS. After completion, the reaction mixture was diluted with EtOAc (20 mL) , washed with water (20 mL x 2) and saturated NaCl (20 mL) , then dried over Na2SO4, following with concentration under reduced pressure to obtain crude product which was purified by pre-HPLC (eluting with CH3CN/H2O (0.1%NH4HCO3) from 20%to 80%) to give (1S, 2S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( (1-methyl-1H-pyrazol-4-yl) ethynyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2-methylcyclopropane-1-carboxamide as atropisomer 1 (19 mg) and (1S, 2S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( (1-methyl-1H-pyrazol-4-yl) ethynyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2-methylcyclopropane-1-carboxamide as atropisomer 2 (i.e., compound of Example 6) (16 mg) as white solids.
LCMS (ESI) calcd. for C46H52N8O5S [M+H] + m/z 829.4, found: 829.9
atropisomer 1: 1H NMR (400 MHz, DMSO-d6) δ 8.83 (d, J = 1.8 Hz, 1H) , 8.52 (d, J = 9.0 Hz, 1H) , 8.41 (s, 1H) , 8.12 (s, 1H) , 8.01 (d, J = 1.7 Hz, 1H) , 7.80 (s, 1H) , 7.72 (d, J = 4.5 Hz, 1H) , 7.53 (d, J = 8.7 Hz, 1H) , 5.89 (d, J = 10.8 Hz, 1H) , 5.31 (t, J = 8.0 Hz, 1H) , 4.72 (d, J = 11.3 Hz, 1H) , 4.48 (d, J = 5.1 Hz, 1H) , 3.93 (dt, J = 12.6, 6.8 Hz, 2H) , 3.87 -3.74 (m, 3H) , 3.58 (d, J = 10.8 Hz, 1H) , 3.50 (d, J = 10.8 Hz, 1H) , 3.24 (d, J =14.9 Hz, 1H) , 3.16 -2.98 (m, 4H) , 2.64 (d, J = 5.9 Hz, 1H) , 2.34 (d, J = 13.9 Hz, 2H) , 2.18 -2.08 (m, 1H) , 1.60 (t, J = 9.3 Hz, 1H) , 1.49 (s, 1H) , 1.19 (d, J = 6.2 Hz, 3H) , 1.08 (dd, J = 14.2, 6.9 Hz, 5H) , 0.90 (s, 3H) , 0.56 (d, J = 5.3 Hz, 1H) , 0.48 (s, 2H) .
atropisomer 2 (i.e., compound of Example 6) : 1H NMR (400 MHz, DMSO-d6) δ 8.82 (d, J = 1.9 Hz, 1H) , 8.51 (d, J = 8.7 Hz, 1H) , 8.41 (s, 1H) , 8.11 (s, 1H) , 7.87 (d, J = 2.0 Hz, 1H) , 7.80 (s, 1H) , 7.73 (d, J = 9.9 Hz, 1H) , 7.57 (d, J = 8.6 Hz, 1H) , 5.92 (d, J = 11.1 Hz, 1H) , 5.36 (s, 1H) , 4.65 (d, J = 11.0 Hz, 1H) , 4.47 (d, J = 5.1 Hz, 1H) , 4.36 -4.21 (m, 2H) , 4.07 (dd, J = 15.0, 7.4 Hz, 1H) , 3.84 (s, 2H) , 3.54 (dd, J = 20.2, 10.6 Hz, 2H) , 3.28 (s, 2H) , 3.22 (s, 3H) , 3.11 (dd, J = 14.7, 7.3 Hz, 1H) , 2.93 (d, J = 14.4 Hz, 1H) , 2.61 (d, J = 5.6 Hz, 1H) , 2.43 (d, J = 17.5 Hz, 2H) , 2.35 -2.25 (m, 1H) , 2.14 (t, J = 9.6 Hz, 1H) , 1.57 (d, J = 9.4 Hz, 1H) , 1.48 (s, 1H) , 1.35 (d, J = 6.0 Hz, 2H) , 1.21 (s, 1H) , 1.05 (s, 3H) , 0.89 (q, J = 7.6 Hz, 5H) , 0.55 (d, J = 5.3 Hz, 1H) , 0.31 (s, 3H) .
Example 7
(1S, 2S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (pyrimidin-5-ylethynyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2-methylcyclopropane-1-carboxamide
Substituting (S) -4- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) -2-methylbut-3-yn-2-ol with (S) -5- ( (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) ethynyl) pyrimidine in the Step 1 of Example 2 and Sub-stituting (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxylic acid with (1S, 2S) -2-methylcyclopropane-1-carboxylic acid in the Step 4 of Example 2, the title compound was pre-pared by the same procedures as described for Example 2. LCMS (ESI) : 827.4 [M+1] +. 1H NMR (500 MHz, CDCl3) δ 9.15 (t, J = 1.7 Hz, 2H) , 8.79 (d, J = 1.8 Hz, 4H) , 8.31 (d, J = 2.0 Hz, 2H) , 8.08 (d, J = 12.6 Hz, 2H) , 8.04 (d, J = 2.0 Hz, 2H) , 7.77 (d, J = 2.4 Hz, 2H) , 7.69 (dd, J = 7.7, 2.4 Hz, 2H) , 7.59 (s, 2H) , 7.48 (d, J = 7.7 Hz, 2H) , 5.24 (q, J = 5.4 Hz, 2H) , 4.90 (d, J = 8.4 Hz, 2H) , 4.64 (q, J = 12.4 Hz, 2H) , 4.15 (q, J = 4.4 Hz, 4H) , 4.08 -3.99 (m, 4H) , 3.99 -3.89 (m, 4H) , 3.33 (s, 6H) , 3.31 -3.23 (m, 3H) , 3.22 (d, J = 3.5 Hz, 1H) , 3.06 (d, J = 2.6 Hz, 4H) , 2.72 (h, J = 7.9 Hz, 2H) , 2.13 -2.03 (m, 6H) , 2.03 -1.97 (m, 2H) , 1.94 (td, J = 10.2, 7.2 Hz, 2H) , 1.67 (d, J = 5.3 Hz, 6H) , 1.65 -1.60 (m, 1H) , 1.55 (ddd, J = 9.9, 8.8, 4.9 Hz, 2H) , 1.37 (t, J = 4.4 Hz, 6H) , 1.15 (s, 6H) , 1.10 (s, 8H) , 0.91 (d, J = 5.0 Hz, 6H) .
Example 8
(1S, 2S) -N- ( (64S, 4S, Z) -12- (5- (benzo [d] thiazol-5-ylethynyl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2-methylcyclopropane-1-carboxamide
Substituting (S) -4- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) -2-methylbut-3-yn-2-ol with (S) -5- ( (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) ethynyl) benzo [d] thiazole in the Step 1 of Example 2 and Substituting (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxylic acid with (1S, 2S) -2-methylcyclopropane-1-carboxylic acid in the Step 4 of Example 2, the title compound was pre-pared by the same procedures as described for Example 2. LCMS (ESI) : 882.3 [M+1] +. 1H NMR (500 MHz, CDCl3) δ 9.01 (s, 2H) , 8.39 (d, J = 2.0 Hz, 2H) , 8.11 (d, J = 12.5 Hz, 2H) , 7.97 (d, J = 2.0 Hz, 2H) , 7.83 (d, J = 1.7 Hz, 2H) , 7.77 (d, J = 2.2 Hz, 2H) , 7.72 -7.66 (m, 4H) , 7.63 -7.57 (m, 4H) , 7.48 (d, J = 7.7 Hz, 2H) , 5.24 (q, J = 5.4 Hz, 2H) , 4.89 (d, J = 8.6 Hz, 2H) , 4.64 (q, J = 12.4 Hz, 2H) , 4.15 (q, J = 4.4 Hz, 4H) , 4.08 -3.99 (m, 4H) , 3.99 -3.89 (m, 4H) , 3.33 (s, 6H) , 3.31 -3.23 (m, 3H) , 3.22 (d, J = 3.5 Hz, 1H) , 3.06 (d, J = 2.6 Hz, 4H) , 2.72 (h, J = 7.9 Hz, 2H) , 2.13 -1.89 (m, 10H) , 1.69 -1.60 (m, 8H) , 1.55 (ddd, J =9.9, 8.8, 4.9 Hz, 2H) , 1.37 (t, J = 4.4 Hz, 6H) , 1.12 (d, J = 24.9 Hz, 14H) , 0.91 (d, J = 5.0 Hz, 6H) .
Example 9
(1S, 2S) -N- ( (64S, 4S, Z) -11-ethyl-12- (5- (imidazo [1, 2-b] pyridazin-3-ylethynyl) -2- ( (S) -1-methoxyethyl) pyrid-in-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2-methylcyclopropane-1-carboxamide
Method 1:
Substituting (S) -4- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) -2-methylbut-3-yn-2-ol with (S) -3- ( (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) ethynyl) imidazo [1, 2-a] pyrimidine in the Step 1 of Ex-ample 2 and Substituting (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxylic acid with (1S, 2S) -2-methylcyclopropane-1-carboxylic acid in the Step 4 of Example 2, the title compound was pre-pared by the same procedures as described for Example 2.
Method 2:
Step 1: Synthesis of (64S, 4S, Z) -4-amino-10, 10-dimethyl-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione
To a stirred solution of tert-butyl ( (64S, 4S, Z) -10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (1.0 g, 0.12 mmol) in dichloromethane (10 mL) was added TFA (1 mL) dropwisely at rt, the resulting mixture was stirred at rt for 4 h. The reaction mixture was diluted with DCM and basified to pH = 7 with saturated aq. NaHCO3 solution. The oaranic layer was separated, dried over anhydrous Na2SO4 and concentrated to give (64S, 4S, Z) -4-amino-10, 10-dimethyl-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione (777 mg) as a grey solid.
LCMS (ESI) calcd. for C31H40BN5O5S [M+H] + m/z 606.3, found: 607.1.
Step 2: Synthesis of (1S, 2S) -N- ( (64S, 4S, Z) -10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2-methyl cyclopropane-1-carboxamide
To a stirred solution of (64S, 4S, Z) -4-amino-10, 10-dimethyl-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione 5 (777 mg, 1.28 mmol) and (1S, 2S) -2-methylcyclopropane-1-carboxylic acid (129 mg, 1.28 mmol) and NMI (316 mg, 3.85 mmol) in ACN (7 mL) and DCM (7 mL) was added TCFH (540 mg, 1.93 mmol) at rt, the resulting mixture was stirred at rt for 1 h. The reaction mixture was concentrated to give the crude product. The crude product was purified by silica gel column chromatography (eluting with EtOAc/PE from 0%to 60%) to give (1S, 2S) -N- ( (64S, 4S, Z) -10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2-methyl cyclopropane-1-carboxamide (1.15 g) as a pale yellow solid.
LCMS (ESI) calcd. for C36H46BN5O6S [M+H] + m/z 688.3, found: 689.4.
Step 3: Synthesis of (1S, 2S) -N- ( (64S, 4S, Z) -12- (5- (imidazo [1, 2-b] pyridazin-3-ylethynyl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacy cloundecaphane-4-yl) -2-methylcyclopropane-1-carboxamide
A mixture of (S) -3- ( (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) ethynyl) imidazo [1, 2-b] pyridazine (57 mg, 0.16 mmol) , (64S, 4S, Z) -4-amino-10, 10-dimethyl-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione (120 mg, 0.17 mmol) , K2CO3 (66 mg, 0.48 mmol) and Pd (dppf) Cl2 (18 mg, 0.024 mmol) in dioxane (2 mL) and H2O (0.5 mL) was stirred at 85℃ under N2 for 3 h. After completion, the reaction mixture was concentrated to give the crude product which was purified by silica gel column (eluting with MeOH/DCM from 0%to 10%) to give (1S, 2S) -N- ( (64S, 4S, Z) -12- (5- (imidazo [1, 2-b] pyridazin-3-ylethynyl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2-methylcyclopropane-1-carboxamide (93 mg, 79.0 %) as a brown solid.
LCMS (ESI) calcd. for C46H47N9O5S [M+H] + m/z 838.3, found: 838.8.
Step 4: Synthesis of (1S, 2S) -N- ( (64S, 4S, Z) -11-ethyl-12- (5- (imidazo [1, 2-b] pyridazin-3-ylethynyl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2-methylcyclopropane-1-carboxamide
To a stirred solution of (1S, 2S) -N- ( (64S, 4S, Z) -12- (5- (imidazo [1, 2-b] pyridazin-3-ylethynyl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -1 0, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2-methylcyclopropane-1-carboxamide (90 mg, 0.11 mmol) in DMF (2 mL) were added Cs2CO3 (70 mg, 0.22 mmol) and Iodoethane (34 mg, 0.22 mmol) at 0℃, the resulting mixture was stirred at rt for 2 h. The reaction was monitored by LCMS. After completion, the reaction mixture was di-luted with EtOAc (20 mL) , washed with water (50 mL x 2) and saturated NaCl (50 mL) , then dried over Na2SO4, following with concentration under reduced pressure to obtain crude product which was purified by pre-HPLC (eluting with CH3CN/H2O (0.1%NH4HCO3) from 20%to 75%) to give (1S, 2S) -N- ( (64S, 4S, Z) -11-ethyl-12- (5- (imidazo [1, 2-b] pyridazin-3-ylethynyl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2-methylcyclopropane-1-carboxamide as atropisomer 1 (33.5 mg) and (1S, 2S) -N- ( (64S, 4S, Z) -11-ethyl-12- (5- (imidazo [1, 2-b] pyridazin-3-ylethynyl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2-methylcyclopropane-1-carboxamide as atropisomer 2 (i.e., compound of Example 9) (23.0 mg) as white solids.
LCMS (ESI) calcd. for C48H51N9O5S [M+H] + m/z 866.4, found: 866.8.
atropisomer 1: 1H NMR (400 MHz, DMSO-d6) δ 8.96 (d, J = 2.0 Hz, 1H) , 8.73 -8.66 (m, 1H) , 8.51 (d, J = 8.9 Hz, 1H) , 8.42 (s, 1H) , 8.26 -8.22 (m, 1H) , 8.17 (d, J = 2.0 Hz, 1H) , 7.80 (s, 1H) , 7.73 (d, J = 8.6 Hz, 1H) , 7.54 (d, J = 8.7 Hz, 1H) , 7.38 (dd, J = 9.2, 4.5 Hz, 1H) , 5.88 (d, J = 11.1 Hz, 1H) , 5.31 (t, J = 8.0 Hz, 1H) , 4.72 (d, J = 11.2 Hz, 1H) , 4.48 (d, J = 5.0 Hz, 1H) , 4.03 -3.77 (m, 3H) , 3.59 (d, J = 10.8 Hz, 1H) , 3.51 (d, J = 10.9 Hz, 1H) , 3.24 (d, J = 14.7 Hz, 1H) , 3.19 -2.98 (m, 4H) , 2.70 -2.61 (m, 1H) , 2.34 (dd, J = 16.0, 10.2 Hz, 2H) , 2.18 -2.07 (m, 1H) , 1.60 (t, J = 9.3 Hz, 1H) , 1.49 (s, 1H) , 1.21 (d, J = 6.2 Hz, 3H) , 1.11 (t, J = 7.1 Hz, 2H) , 1.06 (s, 3H) , 0.91 (s, 3H) , 0.58 -0.44 (m, 3H) .
atropisomer 2 (i.e., compound of Example 9) : 1H NMR (400 MHz, DMSO-d6) δ 8.95 (d, J = 1.9 Hz, 1H) , 8.68 (d, J = 3.8 Hz, 1H) , 8.50 (d, J = 8.9 Hz, 1H) , 8.42 (s, 1H) , 8.24 (d, J = 6.6 Hz, 2H) , 8.01 (d, J = 1.7 Hz, 1H) , 7.79 (s, 1H) , 7.74 (d, J = 8.3 Hz, 1H) , 7.57 (d, J = 8.7 Hz, 1H) , 7.37 (dd, J = 9.2, 4.4 Hz, 1H) , 5.91 (d, J = 11.2 Hz, 1H) , 5.37 (s, 1H) , 4.65 (d, J = 11.2 Hz, 1H) , 4.47 (d, J = 4.7 Hz, 1H) , 4.37 -4.21 (m, 2H) , 4.08 (dd, J = 14.5, 7.2 Hz, 1H) , 3.55 (dd, J = 21.5, 10.6 Hz, 2H) , 3.24 (s, 3H) , 3.12 (dd, J = 14.9, 7.6 Hz, 1H) , 2.95 (d, J = 14.7 Hz, 1H) , 2.61 (d, J = 5.3 Hz, 1H) , 2.44 (d, J = 3.9 Hz, 1H) , 2.35 -2.24 (m, 1H) , 2.14 (t, J =9.6 Hz, 1H) , 1.55 (t, J = 9.3 Hz, 1H) , 1.48 (s, 1H) , 1.37 (d, J = 6.0 Hz, 3H) , 1.20 (s, 1H) , 1.03 (d, J = 19.4 Hz, 4H) , 0.89 (d, J = 9.7 Hz, 6H) , 0.54 (d, J = 4.9 Hz, 1H) , 0.34 (s, 3H) .
Example 10
(1r, 2R, 3S) -N- ( (64S, 4S, Z) -12- (5- (3- (dimethylamino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
Method 1:
Substituting (S) -4- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) morpholine with (S) -3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) -N, N-dimethylprop-2-yn-1-amine in the Step 1 of Exam-ple 16, the title compound was prepared by the same procedures as described for Example 16.
Method 2:
Step 1: Synthesis of (1r, 2R, 3S) -N- ( (64S, 4S, Z) -12- (5- (3- (dimethylamino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 1 0-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
A mixture of (S) -3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) -N, N-dimethylprop-2-yn-1-amine (70 mg, 0.24 mmol) , (1r, 2R, 3S) -N- ( (64S, 4S, Z) -10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide (191 mg, 0.27 mmol) , K2CO3 (98 mg, 0.71 mmol) and Pd (dppf) Cl2 (26 mg, 0.035 mmol) in dioxane (3 mL) and H2O (0.7 mL) was stirred at 85℃ under N2 for 3 h. After com-pletion, the reaction mixture was concentrated to give the crude product which was purified by silica gel column (eluting with MeOH/DCM from 0%to 10%) to give (1r, 2R, 3S) -N- ( (64S, 4S, Z) -12- (5- (3- (dimethylamino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide (150 mg, 79.0 %) as a brown solid.
LCMS (ESI) calcd. for C44H53N7O5S [M+H] + m/z 792.4, found: 792.9.
Step 2: Synthesis of (1r, 2R, 3S) -N- ( (64S, 4S, Z) -12- ( (R) -5- (3- (dimethylamino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) -3, 4-dihydropyridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
To a stirred solution of (1r, 2R, 3S) -N- ( (64S, 4S, Z) -12- (5- (3- (dimethylamino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide (150 mg, 0.19 mmol) in DMF (4 mL) were added Cs2CO3 (99 mg, 0.30 mmol) and Iodoethane (48 mg, 0.30 mmol) at 0℃, the resulting mixture was stirred at rt for 4 h. The reaction was monitored by LCMS. After completion, the reaction mixture was diluted with EtOAc (20 mL) , washed with water (100 mL x 2) and saturated NaCl (60 mL) , then dried over Na2SO4, following with concentration under reduced pressure to obtain crude product which was purified by pre-HPLC (eluting with CH3CN/H2O (0.1%NH4HCO3) from 20%to 85%) to give (1r, 2R, 3S) -N- ( (64S, 4S, Z) -12- ( (R) -5- (3- (dimethylamino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) -3, 4-dihydropyridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide as atropisomer 1 (25.5 mg) and (1r, 2R, 3S) -N- ( (64S, 4S, Z) -12- ( (R) -5- (3- (dimethylamino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) -3, 4-dihydropyridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide as atropisomer 2 (i.e., compound of Example 10) (18.3 mg) as white solids.
LCMS (ESI) calcd. for C46H59N7O5S [M+H] + m/z 822.4, found: 822.0.
atropisomer 1: 1H NMR (400 MHz, DMSO-d6) δ 8.80 (d, J = 2.0 Hz, 1H) , 8.39 (d, J = 10.0 Hz, 2H) , 7.98 (d, J = 2.0 Hz, 1H) , 7.80 (s, 1H) , 7.72 (d, J = 8.7 Hz, 1H) , 7.52 (d, J = 8.7 Hz, 1H) , 5.86 (d, J = 11.1 Hz, 1H) , 5.29 (t, J = 8.5 Hz, 1H) , 4.71 (d, J = 11.2 Hz, 1H) , 4.47 (d, J = 5.1 Hz, 1H) , 4.01 -3.85 (m, 2H) , 3.79 (dd, J = 14.6, 7.4 Hz, 1H) , 3.63 -3.45 (m, 3H) , 3.23 (d, J = 14.7 Hz, 2H) , 3.16 -2.96 (m, 4H) , 2.71 -2.57 (m, 1H) , 2.37 -2.20 (m, 6H) , 2.19 -2.07 (m, 1H) , 1.60 (t, J = 9.3 Hz, 1H) , 1.30 -1.11 (m, 5H) , 1.14 -0.99 (m, 6H) , 0.86 (d, J = 26.9 Hz, 2H) , 0.46 (s, 2H) .
atropisomer 2 (i.e., compound of Example 10) : 1H NMR (400 MHz, DMSO-d6) δ 8.79 (d, J = 2.0 Hz, 1H) , 8.44 -8.34 (m, 2H) , 7.85 (d, J = 1.9 Hz, 1H) , 7.80 (s, 1H) , 7.73 (d, J = 8.6 Hz, 1H) , 7.56 (d, J = 8.8 Hz, 1H) , 5.91 (d, J = 10.9 Hz, 1H) , 5.36 (s, 1H) , 4.63 (d, J = 11.3 Hz, 1H) , 4.46 (d, J = 5.0 Hz, 1H) , 4.27 (dt, J =12.2, 6.8 Hz, 2H) , 4.05 (dd, J = 14.6, 7.5 Hz, 1H) , 3.60 -3.47 (m, 3H) , 3.27 -3.05 (m, 5H) , 2.91 (d, J = 13.6 Hz, 1H) , 2.60 (d, J = 5.5 Hz, 1H) , 2.45 -2.38 (m, 1H) , 2.29 (d, J = 11.6 Hz, 5H) , 2.17 -2.09 (m, 1H) , 1.60 -1.48 (m, 1H) , 1.34 (d, J = 6.0 Hz, 2H) , 1.28 -1.10 (m, 4H) , 1.06 (dd, J = 10.2, 5.8 Hz, 4H) , 0.93 -0.77 (m, 4H) , 0.31 (s, 2H) .
Example 11
(1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (5- (3- ( (R) -3-fluoropyrrolidin-1-yl) prop-1-yn-1-yl) -2- ( (S) -1-methoxyeth-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
Substituting (S) -4- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) morpholine with 3-bromo-5- (3- ( (R) -3-fluoropyrrolidin-1-yl) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridine in the Step 1 of Example 16, the title compound was prepared by the same procedures as described for Example 16. LCMS (ESI) : 864.4 [M+1] +. 1H NMR (500 MHz, CDCl3) δ 8.39 (d, J = 2.0 Hz, 1H) , 8.20 (d, J = 13.0 Hz, 1H) , 8.02 (d, J = 2.0 Hz, 1H) , 7.77 (d, J = 2.4 Hz, 1H) , 7.69 (dd, J = 7.7, 2.4 Hz, 1H) , 7.59 (s, 1H) , 7.48 (d, J = 7.7 Hz, 1H) , 5.24 (q, J = 5.4 Hz, 1H) , 4.89 (d, J = 8.6 Hz, 1H) , 4.87 -4.80 (m, 1H) , 4.63 (dt, J = 13.0, 12.3 Hz, 1H) , 4.15 (q, J = 4.4 Hz, 2H) , 4.08 -3.99 (m, 2H) , 3.99 -3.89 (m, 2H) , 3.63 -3.57 (m, 1H) , 3.55 (s, 1H) , 3.52 -3.45 (m, 1H) , 3.33 (s, 3H) , 3.31 -3.18 (m, 3H) , 3.06 (d, J = 2.6 Hz, 2H) , 2.94 (t, J = 5.6 Hz, 2H) , 2.72 (h, J = 7.9 Hz, 1H) , 2.47 (t, J = 8.6 Hz, 1H) , 2.29 (q, J = 5.6 Hz, 1H) , 2.14 -1.97 (m, 5H) , 1.74 (dq, J = 8.6, 5.2 Hz, 2H) , 1.67 (d, J = 5.3 Hz, 3H) , 1.37 (t, J = 4.4 Hz, 3H) , 1.15 (s, 3H) , 1.10 (s, 3H) , 0.95 (d, J = 5.1 Hz, 6H) .
Example 12
(1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- (piperidin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
Method 1:
Substituting (S) -4- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) morpholine with (S) -3-bromo-2- (1-methoxyethyl) -5- (3- (piperidin-1-yl) prop-1-yn-1-yl) pyridine in the Step 1 of Example 16, the title compound was prepared by the same procedures as described for Example 16.
Method 2:
Step 1: Preparation of (1r, 2R, 3S) -N- ( (64S, 4S, Z) -12- (2- ( (S) -1-methoxyethyl) -5- (3- (piperidin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 1 0-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
A mixture of (S) -3-bromo-2- (1-methoxyethyl) -5- (3- (piperidin-1-yl) prop-1-yn-1-yl) pyridine (48 mg, 0.14 mmol) , (1r, 2R, 3S) -N- ( (64S, 4S, Z) -10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide (100 mg, 0.14 mmol) , K2CO3 (58 mg, 0.42 mmol) and Pd (dppf) Cl2 (22 mg, 0.03 mmol) in dioxane (5 mL) and H2O (1 mL) was stirred at 85℃ under N2 for 16h. After com-pletion, the reaction mixture was concentrated under reduced pressure. The residue was purified by FCC with MeOH/DCM = 0-6%to afford the (1r, 2R, 3S) -N- ( (64S, 4S, Z) -12- (2- ( (S) -1-methoxyethyl) -5- (3- (piperidin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacy cloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide (98 mg, 84%) as a yellow solid. LCMS (ESI) calcd. for C47H57N7O5S [M+H] + m/z 832.4, found: 832.9
Step 2: Preparation of (1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- (piperidin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
To a stirred solution of (1r, 2R, 3S) -N- ( (64S, 4S, Z) -12- (2- ( (S) -1-methoxyethyl) -5- (3- (piperidin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacy cloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide (98 mg, 0.117 mmol) in DMF (5 mL) were added Cs2CO3 (115.1 mg, 0.353 mmol) and Iodoethane (55.2 mg, 0.353 mmol) . The resulting mixture was stirred at rt for 0.5h. After completion, the reaction mixture was diluted with EtOAc (20 mL) and H2O (20 mL) . The mixture was extracted with EtOAc (10 mL*3) . The combined organic layer was washed with brine (20 mL*2) , dried over anhydrous Na2SO4 and concentration under reduced pressure. The residue was purified by pre-HPLC to give the (1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- (piperidin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] h eptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide as atropisomer 1 (38 mg) and (1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- (piperidin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide as atropisomer 2 (i.e., compound of Example 12 ) (28 mg) as white solids.
LCMS (ESI) calcd. for C49H61N7O5S [M+H] + m/z 860.4, found: 860.5.
atropisomer 1: 1H NMR (400 MHz, DMSO-d6) δ 8.81 (d, J = 1.8 Hz, 1H) , 8.42 (d, J = 8.8 Hz, 2H) , 7.99 (d, J = 1.9 Hz, 1H) , 7.82 (s, 1H) , 7.74 (d, J = 8.4 Hz, 1H) , 7.54 (d, J = 8.7 Hz, 1H) , 5.89 (d, J = 10.9 Hz, 1H) , 5.31 (t, J = 8.3 Hz, 1H) , 4.73 (d, J = 11.1 Hz, 1H) , 4.49 (d, J = 5.0 Hz, 1H) , 3.99 -3.89 (m, 2H) , 3.81 (dd, J = 14.8, 7.4 Hz, 1H) , 3.60 (d, J = 10.8 Hz, 1H) , 3.52 (d, J = 7.2 Hz, 3H) , 3.30 -2.98 (m, 7H) , 2.66 (dd, J = 11.4, 5.6 Hz, 1H) , 2.44 (s, 3H) , 2.39 -2.29 (m, 2H) , 2.15 (t, J = 9.8 Hz, 1H) , 1.67 -1.46 (m, 5H) , 1.43 -1.01 (m, 18H) , 0.92 (s, 3H) , 0.48 (s, 3H) .
atropisomer 2 (i.e., compound of Example 12 ) : 1H NMR (400 MHz, DMSO) δ 8.78 (d, J = 1.8 Hz, 1H) , 8.40 (d, J = 8.8 Hz, 2H) , 7.97 (d, J = 1.9 Hz, 1H) , 7.80 (s, 1H) , 7.72 (d, J = 8.4 Hz, 1H) , 7.52 (d, J =8.7 Hz, 1H) , 5.87 (d, J = 10.9 Hz, 1H) , 5.29 (t, J = 8.3 Hz, 1H) , 4.71 (d, J = 11.1 Hz, 1H) , 4.47 (d, J = 5.0 Hz, 1H) , 3.99 -3.86 (m, 2H) , 3.79 (dd, J = 14.8, 7.4 Hz, 1H) , 3.63 -3.42 (m, 4H) , 3.26 -2.98 (m, 6H) , 2.64 (d, J = 5.7 Hz, 1H) , 2.36 -2.27 (m, 2H) , 2.13 (t, J = 9.8 Hz, 1H) , 1.60 (t, J = 9.4 Hz, 1H) , 1.54 -1.47 (m, 3H) , 1.35 (s, 2H) , 1.29 -1.00 (m, 14H) , 0.90 (s, 3H) , 0.46 (s, 3H) .
Example 13
(1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- (4-methylpiperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
Method 1:
Substituting (S) -4- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) morpholine with (S) -1- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) -4-methylpiperazine in the Step 1 of Example 16, the title compound was prepared by the same procedures as described for Example 16.
Method 2:
Step 1: Preparation of tert-butyl ( (64S, 4S, Z) -12- (2- ( (S) -1-methoxyethyl) -5- (3- (4-methylpiperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dime-thyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
A mixture of (S) -1- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) -4-methylpiperazine (100 mg, 0.28 mmol) , tert-butyl ( (64S, 4S, Z) -10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (200 mg, 0.28 mmol) , K2CO3 (117.5 mg, 0.85 mmol) and Pd (dppf) Cl2 (41.5 mg, 0.06 mmol) in dioxane (6 mL) and H2O (1.5 mL) was stirred at 85℃ under N2 for 16h. After completion, the reaction mixture was concen-trated under reduced pressure. The residue was purified by FCC with MeOH/DCM = 0-10%to afford the tert-butyl ( (64S, 4S, Z) -12- (2- ( (S) -1-methoxyethyl) -5- (3- (4-methylpiperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dime-thyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (180 mg, 76%) as a yellow solid. LCMS (ESI) calcd. for C46H58N8O6S [M+H] +m/z 851.4, found: 852.1
Step 2: Preparation of tert-butyl ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- (4-methylpiperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
To a stirred solution of tert-butyl ( (64S, 4S, Z) -12- (2- ( (S) -1-methoxyethyl) -5- (3- (4-methylpiperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dime-thyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (180 mg, 0.21 mmol) in DMF (6 mL) were added Cs2CO3 (206.4 mg, 0.64 mmol) and Iodoethane (99 mg, 0.64 mmol) . The resulting mixture was stirred at rt for 0.5h. After completion, the reaction mixture was diluted with EtOAc (20 mL) and H2O (20 mL) . The mixture was extracted with EtOAc (10 mL*3) . The combined organic layer was washed with brine (20 mL*2) , dried over anhydrous Na2SO4 and concentration under reduced pressure. The residue was purified by FCC with MeOH/DCM =0-7%to afford the tert-butyl ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- (4-methylpiperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (115 mg, 62%) as yellow solid.
LCMS (ESI) calcd. for C48H62N8O6S [M+H] + m/z 879.5, found: 880.0
Step 3: Preparation of (64S, 4S, Z) -4-amino-11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- (4-methylpiperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione
To a stirred solution of tert-butyl ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- (4-methylpiperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptan acycloundecaphane-4-yl) carbamate (115 mg, 0.13 mmol) in DCM (5 mL) was added TFA (1 mL) . The resulting mixture was stirred at rt for 1 h. The reaction mixture was concentrated under reduced pressure to give
(64S, 4S, Z) -4-amino-11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- (4-methylpiperazin-1-yl) prop-1-yn-1-yl) pyri din-3-yl) -10, 10-dimethyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione 7 (186 mg) of its TFA salt as a yellow solid. LCMS (ESI) calcd. for C43H54N8O4S [M+H] + m/z 779.4, found: 779.9
Step 4: Preparation of (1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- (4-methylpiperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
To a stirred solution of (64S, 4S, Z) -4-amino-11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- (4-methylpiperazin-1-yl) prop-1-yn-1-yl) pyri din-3-yl) -10, 10-dimethyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione (101.3 mg, 0.13 mmol) in MeCN (2.5 mL) and DCM (2.5 mL) was added (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxylic acid (16.3 mg, 0.14 mmol) , TCFH (109.3 mg, 0.39 mmol) and NMI (106.7 mg, 1.3 mmol) . The resulting mixture was stirred at rt for 0.5h. The reaction mix-ture was concentrated under reduced pressure. The residue was purified by pre-HPLC to give the (1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- (4-methylpiperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicycl o[3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide as atropisomer 1 (38 mg) and (1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- (4-methylpiperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicycl o[3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide as atropisomer 2 (i.e., compound of Example 13 ) (22 mg) as white solids.
LCMS (ESI) calcd. for C49H62N8O5S [M+H] + m/z 875.5, found: 875.6
atropisomer 1: 1H NMR (400 MHz, DMSO-d6) δ 8.78 (d, J = 1.9 Hz, 1H) , 8.39 (d, J = 11.7 Hz, 2H) , 7.97 (d, J = 2.0 Hz, 1H) , 7.79 (s, 1H) , 7.71 (d, J = 7.5 Hz, 1H) , 7.52 (d, J = 8.6 Hz, 1H) , 5.86 (d, J = 11.1 Hz, 1H) , 5.29 (t, J = 7.9 Hz, 1H) , 4.71 (d, J = 11.1 Hz, 1H) , 4.47 (d, J = 5.1 Hz, 1H) , 3.99 -3.72 (m, 4H) , 3.60 -3.47 (m, 4H) , 3.27 -2.98 (m, 8H) , 2.64 (dd, J = 11.6, 5.9 Hz, 1H) , 2.52 (s, 2H) , 2.41 -2.22 (m, 6H) , 2.17 -2.09 (m, 4H) , 1.59 (t, J = 9.2 Hz, 1H) , 1.27 -1.13 (m, 7H) , 1.11 -1.03 (m, 8H) , 0.89 (s, 3H) , 0.45 (s, 3H) .
atropisomer 2 (i.e., compound of Example 13 ) : 1H NMR (400 MHz, DMSO) δ 8.78 (d, J = 1.9 Hz, 1H) , 8.39 (d, J = 11.7 Hz, 2H) , 7.97 (d, J = 2.0 Hz, 1H) , 7.79 (s, 1H) , 7.71 (d, J = 7.5 Hz, 1H) , 7.52 (d, J =8.6 Hz, 1H) , 5.86 (d, J = 11.1 Hz, 1H) , 5.29 (t, J = 7.9 Hz, 1H) , 4.71 (d, J = 11.1 Hz, 1H) , 4.47 (d, J = 5.1 Hz, 1H) , 3.99 -3.72 (m, 4H) , 3.60 -3.47 (m, 4H) , 3.27 -2.98 (m, 8H) , 2.60 (dd, J = 27.9, 22.1 Hz, 5H) , 2.41 -2.22 (m, 6H) , 2.17 -2.09 (m, 4H) , 1.59 (t, J = 9.2 Hz, 1H) , 1.29 -1.02 (m, 16H) , 0.89 (s, 3H) , 0.45 (s, 3H) .
Example 14
(1r, 2R, 3S) -N- ( (64S, 4S, Z) -12- (5- (3- (4-acetylpiperazin-1-yl) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
Substituting (S) -4- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) morpholine with (S) -1- (4- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) piperazin-1-yl) ethan-1-one in the Step 1 of Example 16, the title compound was prepared by the same procedures as described for Example 16. LCMS (ESI) : 903.5 [M+1] +. 1H NMR (500 MHz, CDCl3) δ 8.39 (d, J = 2.0 Hz, 1H) , 8.20 (d, J = 13.0 Hz, 1H) , 8.02 (d, J = 2.0 Hz, 1H) , 7.77 (d, J = 2.3 Hz, 1H) , 7.69 (dd, J = 7.7, 2.4 Hz, 1H) , 7.59 (s, 1H) , 7.48 (d, J = 7.7 Hz, 1H) , 5.24 (q, J = 5.4 Hz, 1H) , 4.89 (d, J = 8.6 Hz, 1H) , 4.63 (dt, J = 13.0, 12.3 Hz, 1H) , 4.15 (q, J = 4.4 Hz, 2H) , 4.08 -3.99 (m, 2H) , 3.99 -3.89 (m, 2H) , 3.60 -3.52 (m, 3H) , 3.52 -3.45 (m, 4H) , 3.33 (s, 3H) , 3.31 -3.23 (m, 2H) , 3.22 (d, J = 3.5 Hz, 1H) , 3.06 (d, J = 2.6 Hz, 2H) , 2.72 (h, J = 7.9 Hz, 1H) , 2.55 -2.50 (m, 4H) , 2.50 -2.43 (m, 2H) , 2.13 -1.97 (m, 7H) , 1.74 (dq, J = 8.6, 5.2 Hz, 2H) , 1.67 (d, J = 5.3 Hz, 3H) , 1.37 (t, J = 4.4 Hz, 3H) , 1.15 (s, 3H) , 1.10 (s, 3H) , 0.95 (d, J = 5.1 Hz, 6H) .
Example 15
(1S, 2S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2-methylcyclopropane-1-carboxamide
Substituting (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxylic acid with (1S, 2S) -2-methylcyclopropane-1-carboxylic acid in the Step 4 of Example 16, the title compound was prepared by the same procedures as described for Example 16. LCMS (ESI) : 848.4 [M+1] +. 1H NMR (500 MHz, CDCl3) δ 8.39 (d, J = 2.0 Hz, 1H) , 8.11 (d, J = 12.5 Hz, 1H) , 8.00 (d, J = 2.0 Hz, 1H) , 7.77 (d, J =2.3 Hz, 1H) , 7.69 (dd, J = 7.7, 2.4 Hz, 1H) , 7.59 (s, 1H) , 7.48 (d, J = 7.7 Hz, 1H) , 5.24 (q, J = 5.4 Hz, 1H) , 4.89 (d, J = 8.6 Hz, 1H) , 4.64 (q, J = 12.4 Hz, 1H) , 4.15 (q, J = 4.4 Hz, 2H) , 4.08 -3.99 (m, 2H) , 3.99 -3.89 (m, 2H) , 3.73 (dd, J = 6.5, 3.8 Hz, 2H) , 3.66 (dd, J = 6.5, 3.8 Hz, 2H) , 3.58 (d, J = 2.6 Hz, 2H) , 3.33 (s, 3H) , 3.31 -3.23 (m, 2H) , 3.22 (d, J = 3.5 Hz, 1H) , 3.06 (d, J = 2.6 Hz, 2H) , 2.78 -2.66 (m, 5H) , 2.13 -2.03 (m, 3H) , 2.03 -1.97 (m, 1H) , 1.94 (td, J = 10.2, 7.2 Hz, 1H) , 1.67 (d, J = 5.3 Hz, 3H) , 1.65 -1.60 (m, 1H) , 1.55 (ddd, J = 9.9, 8.8, 4.9 Hz, 1H) , 1.37 (t, J = 4.4 Hz, 3H) , 1.15 (s, 3H) , 1.10 (s, 4H) , 0.91 (d, J = 5.0 Hz, 3H) .
Example 16
Synthesis of
(1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
Step 1: Preparation of tert-butyl ( (64S, 4S, Z) -12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
A mixture of (S) -4- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) morpholine (253 mg, 0.74 mmol) , tert-butyl ( (64S, 4S, Z) -10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (525 mg, 0.74 mmol) , K2CO3 (309 mg, 2.24 mmol) and Pd (dppf) Cl2 (55 mg, 0.075 mmol) in dioxane (8 mL) and H2O (2 mL) was stirred at 85℃ under N2 for 19 h. After completion, the reaction mixture was concentrated to give the crude product which was purified by silica gel column (eluting with MeOH/DCM from 0%to 100%) to give tert-butyl ( (64S, 4S, Z) -12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-di-oxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (323 mg, 52.1 %) as a yellow solid.
LCMS (ESI) calcd. for C45H55N7O7S [M+H] + m/z 838.4, found: 839.5
Step 2: Preparation of tert-butyl ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimeth yl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
To a stirred solution of tert-butyl ( (64S, 4S, Z) -12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-di-oxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (323 mg, 0.39 mmol) in DMF (4.5 mL) were added Cs2CO3 (251 mg, 0.77 mmol) and Io-doethane (120 mg, 0.77 mmol) at 0℃, the resulting mixture was stirred at rt for 3 h. The reaction was monitored by LCMS. After completion, the reaction mixture was diluted with EtOAc (100 mL) , washed with water (50 mL x 3) and saturated NaCl (50 x 2 mL) , then dried over Na2SO4, following with concen-tration under reduced pressure to obtain crude product which was purified by pre-HPLC (eluting with CH3CN/H2O (0.1%NH4HCO3) from 20%to 80%) to give tert-butyl ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimet hyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate as atropisomer 1 (100 mg) and atropisomer 2 (140 mg) as white solids.
LCMS (ESI) calcd. for C47H59N7O7S [M+H] + m/z 866.4, found: 866.8. Step 3: Preparation of (64S, 4S, Z) -4-amino-11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10 -dimethyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione
To a stirred solution of tert-butyl ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimet hyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate as atropisomer 1 (100 mg, 0.12 mmol) in dichloromethane (4 mL) was added TFA (1 mL) dropwisely at rt, the resulting mixture was stirred at rt for 1 h. The reaction mixture was concen-trated and lyophilized to give (64S, 4S, Z) -4-amino-11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundeca phane-5, 7-dione (110 mg) of its TFA salt as a yellow solid.
LCMS (ESI) calcd. for C42H51N7O5S [M+H] + m/z 766.4, found: 766.9
Step 4: Preparation of (1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptan acycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
To a stirred solution of (64S, 4S, Z) -4-amino-11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundeca phane-5, 7-dione (125 mg, 0.16 mmol) and (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxylic acid (19 mg, 0.16 mmol) and 2, 6-lutidine (88 mg, 0.82 mmol) in ACN (5 mL) was added COMU (105 mg, 0.24 mmol) at rt, the resulting mixture was stirred at rt for 2 h. The reaction mixture was concentrated to give the crude product. The crude product was purified by pre-HPLC (ACN/H2O (0.5 %NH4HCO3) from 20%to 95%in 30 min) to give (1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl ) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide (85 mg, 61.7%) as a white solid.
LCMS (ESI) calcd. for C48H59N7O6S [M+H] + m/z 862.4, found: 862.9.
1H NMR (400 MHz, DMSO-d6) δ 8.82 (s, 1H) , 8.40 (d, J = 13.3 Hz, 2H) , 7.90 (s, 1H) , 7.81 (s, 1H) , 7.74 (d, J = 8.8 Hz, 1H) , 7.57 (d, J = 8.8 Hz, 1H) , 5.92 (d, J = 10.8 Hz, 1H) , 5.34 (d, J = 22.7 Hz, 2H) , 4.64 (d, J =11.2 Hz, 1H) , 4.48 (d, J = 4.9 Hz, 1H) , 4.37 -4.21 (m, 2H) , 4.06 (d, J = 7.6 Hz, 5H) , 3.75 -3.48 (m, 6H) , 3.22 (s, 3H) , 3.16 (s, 10H) , 3.00 -2.85 (m, 2H) , 2.62 (d, J = 5.7 Hz, 3H) , 2.41 (d, J = 15.0 Hz, 1H) , 2.32 (s, 1H) , 2.21 -2.09 (m, 1H) , 1.99 (d, J = 8.0 Hz, 1H) , 1.55 (s, 1H) , 1.35 (d, J = 6.1 Hz, 2H) , 1.22 (s, 4H) , 1.16 (s, 2H) , 1.07 (dd, J = 10.3, 5.8 Hz, 4H) , 0.87 (t, J = 9.6 Hz, 4H) , 0.32 (s, 3H) .
Example 17
(1S, 2S) -N- ( (64S, 4S, Z) -12- (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyrid-in-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2-methylcyclopropane-1-carboxamide
Method 1:
Substituting (S) -4- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) morpholine with (S) -4- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) thiomorpholine 1, 1-dioxide in the Step 1 of Example 16 and Substituting (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxylic acid with (1S, 2S) -2-methylcyclopropane-1-carboxylic acid in the Step 4 of Example 16, the title compound was prepared by the same procedures as described for Example 16.
Method 2:
Step 1: Preparation of tert-butyl ( (64S, 4S, Z) -12- (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10 -dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
A mixture of (S) -4- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) thiomorpholine 1, 1-dioxide (209 mg, 0.54 mmol) , tert-butyl ( (64S, 4S, Z) -10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (383 mg, 0.54 mmol) , K2CO3 (225 mg, 1.62 mmol) and Pd (dppf) Cl2 (78 mg, 0.11 mmol) in dioxane (10 mL) and H2O (2.5 mL) was stirred at 85℃ under N2 for 5 h. After completion, the reaction mixture was concentrat-ed to give the crude product which was purified by silica gel column (eluting with MeOH/DCM from 0%to 10%) to give tert-butyl ( (64S, 4S, Z) -12- (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacy cloundecaphane-4-yl) carbamate (312 mg, 65.1%) as a yellow solid.
LCMS (ESI) calcd. for C45H55N7O8S2 [M+H] + m/z 886.3, found: 886.9
Step 2: tert-butyl ( (64S, 4S, Z) -12- (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
To a stirred solution of tert-butyl ( (64S, 4S, Z) -12- (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacy cloundecaphane-4-yl) carbamate (310 mg, 0.35 mmol) in DMF (5 mL) were added Cs2CO3 (183 mg, 0.56 mmol) and Iodoethane (88 mg, 0.56 mmol) at 0℃, the resulting mixture was stirred at rt for 3 h. The reac-tion was monitored by LCMS. After completion, the reaction mixture was diluted with EtOAc (20 mL) , washed with water (50 mL x 3) and saturated NaCl (50 x 2 mL) , then dried over Na2SO4, following with concentration under reduced pressure to obtain crude product which was purified by column chromatog-raphy (eluting with EtOAc/PE from 0%to 100%) to give tert-butyl ( (64S, 4S, Z) -12- (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -11-e thyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate as atropisomer 1 (160 mg) and atropisomer 2 (156 mg) (Yield: 98.8%) as a yellow foam.
LCMS (ESI) calcd. for C47H59N7O8S2 [M+H] + m/z 914.4, found: 914.9
Step 3: (64S, 4S, Z) -4-amino-12- (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -11-ethyl-10, 10-dimethyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] hepta nacycloundecaphane-5, 7-dione
To a stirred solution of tert-butyl ( (64S, 4S, Z) -12- (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate as atropisomer 1 (160 mg, 0.18 mmol) in dichloromethane (3 mL) was added TFA (1 mL) dropwise at rt, the resulting mixture was stirred at rt for 1 h. The reaction mixture was concentrated and lyophilized to give (64S, 4S, Z) -4-amino-12- (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3 -yl) -11-ethyl-10, 10-dimethyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione (190 mg) of its TFA salt as a yellow solid.
LCMS (ESI) calcd. for C42H51N7O6S2 [M+H] + m/z 814.3, found: 814.8
Step 4: (1S, 2S) -N- ( (64S, 4S, Z) -12- (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2-methylcyclopropane-1-carboxamide
To a stirred solution of (64S, 4S, Z) -4-amino-12- (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3 -yl) -11-ethyl-10, 10-dimethyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] hept anacycloundecaphane-5, 7-dione (190 mg, 0.16 mmol) and (1S, 2S) -2-methylcyclopropane-1-carboxylic acid (18 mg, 0.16 mmol) and NMI (128 mg, 1.56 mmol) in ACN (2 mL) and DCM (2 mL) was added TCFH (131 mg, 0.47 mmol) at rt, the resulting mixture was stirred at rt for 2 h. The reaction mixture was concentrated to give the crude product. The crude product was purified by pre-TLC (with EA) to give (1S, 2S) -N- ( (64S, 4S, Z) -12- (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2-methylcyclopropane-1-carboxamide (23 mg, 16.1%) as a white solid.
LCMS (ESI) calcd. for C47H57N7O7S2 [M+H] + m/z 896.3, found: 896.4.
1H NMR (400 MHz, DMSO-d6) δ 8.78 (d, J = 2.0 Hz, 1H) , 8.49 (d, J = 8.8 Hz, 1H) , 8.40 (s, 1H) , 7.85 (d, J = 2.1 Hz, 1H) , 7.79 (s, 1H) , 7.73 (d, J = 7.4 Hz, 1H) , 7.56 (d, J = 8.6 Hz, 1H) , 5.91 (d, J = 11.1 Hz, 1H) , 5.37 (s, 1H) , 4.64 (d, J = 10.8 Hz, 1H) , 4.47 (d, J = 5.1 Hz, 1H) , 4.37 -4.20 (m, 2H) , 4.04 (dd, J =15.2, 7.3 Hz, 1H) , 3.73 (s, 2H) , 3.54 (dd, J = 20.9, 11.1 Hz, 2H) , 3.21 (s, 4H) , 3.11 (t, J = 10.5 Hz, 4H) , 3.03 (s, 3H) , 2.91 (d, J = 14.3 Hz, 1H) , 2.61 (d, J = 5.7 Hz, 1H) , 2.40 (d, J = 14.1 Hz, 1H) , 2.31 (s, 1H) , 2.18 -2.08 (m, 1H) , 1.55 (s, 1H) , 1.47 (s, 1H) , 1.34 (d, J = 6.0 Hz, 2H) , 1.21 (s, 1H) , 1.05 (s, 3H) , 0.86 (t, J = 6.9 Hz, 5H) , 0.54 (d, J = 5.4 Hz, 1H) , 0.31 (s, 3H) .
Example 18
(1r, 2R, 3S) -N- ( (64S, 4S, Z) -12- (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyri-din-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
Method 1:
Substituting (S) -4- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) morpholine with (S) -4- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) thiomorpholine 1, 1-dioxide in the Step 1 of Example 16, the title compound was prepared by the same procedures as described for Example 16.
Method 2:
Step 1: Synthesis of tert-butyl ( (64S, 4S, Z) -12- (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10 -dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
A mixture of (S) -4- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) thiomorpholine 1, 1-dioxide (186 mg, 0.48 mmol) , tert-butyl ( (64S, 4S, Z) -10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (341 mg, 0.48 mmol) , K2CO3 (200 mg, 1.45 mmol) and Pd (dppf) Cl2 (71 mg, 0.097 mmol) in dioxane (7.6 mL) and H2O (1.9 mL) was stirred at 85℃ under N2 for 15 h. After completion, the reaction mixture was concen-trated to give the crude product which was purified by silica gel column (eluting with MeOH/DCM from 0%to 10%) to give tert-butyl ( (64S, 4S, Z) -12- (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (312 mg, 73.5 %) as a brown solid.
LCMS (ESI) calcd. for C45H55N7O8S2 [M+H] + m/z 886.4, found: 886.9
Step 2: Synthesis of tert-butyl ( (64S, 4S, Z) -12- (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
To a stirred solution of tert-butyl ( (64S, 4S, Z) -12- (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacy cloundecaphane-4-yl) carbamate (310 mg, 0.35 mmol) in DMF (5 mL) were added Cs2CO3 (183 mg, 0.56 mmol) and Iodoethane (88 mg, 0.56 mmol) at 0℃, the resulting mixture was stirred at rt for 3 h. The reac-tion was monitored by LCMS. After completion, the reaction mixture was diluted with EtOAc (20 mL) , washed with water (50 mL x 2) and saturated NaCl (50 mL) , then dried over Na2SO4, following with con-centration under reduced pressure to obtain crude product which was purified by silica gel column (elut-ing with EtOAc/PE from 0%to 100%) to give tert-butyl ( (64S, 4S, Z) -12- (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (316 mg, 97.8%) as brown solid.
LCMS (ESI) calcd. for C47H59N7O8S2 [M+H] + m/z 914.4, found: 914.9.
Step 3: Synthesis of (64S, 4S, Z) -4-amino-12- (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -11-ethyl-10, 10-dimethyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione
To a stirred solution of tert-butyl ( (64S, 4S, Z) -12- (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] h eptanacycloundecaphane-4-yl) carbamate (316mg, 0.35mmol) in dichloromethane (3 mL) was added TFA (1 mL) dropwisely at rt, the resulting mixture was stirred at rt for 1 h. The reaction mixture was concen-trated to give crude (64S, 4S, Z) -4-amino-12- (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -11-ethyl-10, 10-dimethyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione which was used to next step without purification.
LCMS (ESI) calcd. for C42H51N7O6S2 [M+H] + m/z 814.3, found: 814.8
Step 4: Synthesis of (1r, 2R, 3S) -N- ( (64S, 4S, Z) -12- ( (R) -5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) -3, 4-dihydropyridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
To a stirred solution of crude (64S, 4S, Z) -4-amino-11-ethyl-12- (5- (3- ( (R) -3- (hydroxymethyl) morpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyeth-yl) pyridin-3-yl) -10, 10-dimethyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione (220 mg, 0.27 mmol) and (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxylic acid (24 mg, 0.21 mmol) and NMI (132 mg, 1.61 mmol) in ACN (3 mL) and DCM (3 mL) was added TCFH (136 mg, 0.49 mmol) at rt, the resulting mixture was stirred at rt for 1 h. The reaction mixture was concentrated to give the crude product. The crude product was purified by pre-TLC (eluting with 100%EtOAc) to give (1r, 2R, 3S) -N- ( (64S, 4S, Z) -12- ( (R) -5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl ) -3, 4-dihydropyridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide (34.3 mg) as a white solid.
LCMS (ESI) calcd. for C48H61N7O7S2 [M+H] + m/z 912.4, found: 911.2.
1H NMR (400 MHz, DMSO-d6) δ 8.79 (s, 1H) , 8.39 (d, J = 11.6 Hz, 2H) , 7.86 (s, 1H) , 7.80 (s, 1H) , 7.73 (d, J = 8.1 Hz, 1H) , 7.56 (d, J = 8.8 Hz, 1H) , 5.91 (d, J = 11.0 Hz, 1H) , 5.36 (s, 1H) , 4.64 (d, J = 11.0 Hz, 1H) , 4.46 (d, J = 4.6 Hz, 1H) , 4.35 -4.21 (m, 2H) , 4.05 (dd, J = 14.8, 6.9 Hz, 2H) , 3.73 (s, 2H) , 3.53 (dd, J = 20.6, 10.7 Hz, 2H) , 3.21 (s, 3H) , 3.13 (d, J = 4.9 Hz, 4H) , 3.03 (s, 3H) , 2.61 (d, J = 6.0 Hz, 1H) , 2.42 (s, 1H) , 2.30 (s, 1H) , 2.17 -2.09 (m, 1H) , 1.56 (d, J = 8.9 Hz, 1H) , 1.40 -1.28 (m, 3H) , 1.25 -1.10 (m, 5H) , 1.11 -0.99 (m, 5H) , 0.94 -0.80 (m, 5H) , 0.31 (s, 3H) .
Example 19
(1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (5- (3- ( (R) -3- (hydroxymethyl) morpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
Method 1:
Substituting (S) -4- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) morpholine with ( (R) -4- (3- (5-bromo-6- ( (S) -1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) morpholin-3-yl) methanol in the Step 1 of Example 16, the title compound was prepared by the same procedures as described for Example 16.
Method 2:
Step 1: Synthesis of tert-butyl ( (64S, 4S, Z) -12- (5- (3- ( (R) -3- (hydroxymethyl) morpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
A mixture of ( (R) -4- (3- (5-bromo-6- ( (S) -1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) morpholin-3-yl) methanol (71 mg, 0.19 mmol) , tert-butyl ( (64S, 4S, Z) -10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (150 mg, 0.21 mmol) , K2CO3 (80 mg, 0.58 mmol) and Pd (dppf) Cl2 (21 mg, 0.029 mmol) in dioxane (3 mL) and H2O (0.7 mL) was stirred at 85℃ under N2 for 16 h. After completion, the reaction mixture was concen-trated to give the crude product which was purified by silica gel column (eluting with MeOH/DCM from 0%to 10%) to give tert-butyl ( (64S, 4S, Z) -12- (5- (3- ( (R) -3- (hydroxymethyl) morpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (146 mg, 88.5 %) as a pale yellow solid.
LCMS (ESI) calcd. for C46H57N7O8S [M+H] + m/z 868.4, found: 868.9
Step 2: Synthesis of tert-butyl ( (64S, 4S, Z) -11-ethyl-12- (5- (3- ( (R) -3- (hydroxymethyl) morpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyrid-in-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1 ] heptanacycloundecaphane-4-yl) carbamate
To a stirred solution of tert-butyl ( (64S, 4S, Z) -12- (5- (3- ( (R) -3- (hydroxymethyl) morpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (915 mg, 1.05 mmol) in DMF (10 mL) were added Cs2CO3 (628 mg, 1.93 mmol) and Iodoethane (301 mg, 1.93 mmol) at 0℃, the resulting mixture was stirred at rt for 2 h. The reaction was monitored by LCMS. After completion, the reaction mixture was diluted with EtOAc (20 mL) , washed with water (100 mL x 2) and saturated NaCl (60 mL) , then dried over Na2SO4, following with concentration under reduced pressure to obtain crude product which was purified by pre-HPLC (eluting with CH3CN/H2O (0.1%NH4HCO3) from 20%to 85%) to give tert-butyl ( (64S, 4S, Z) -11-ethyl-12- (5- (3- ( (R) -3- (hydroxymethyl) morpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (190 mg, 20.0%) as white solid.
LCMS (ESI) calcd. for C47H59N7O6S [M+H] + m/z 896.4, found: 896.9
Step 3: Synthesis of (64S, 4S, Z) -4-amino-11-ethyl-12- (5- (3- ( (R) -3- (hydroxymethyl) morpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione
To a stirred solution of tert-butyl ( (64S, 4S, Z) -11-ethyl-12- (5- (3- ( (R) -3- (hydroxymethyl) morpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (190 mg, 0.21 mmol) in dichloromethane (3 mL) was added TFA (0.75 mL) dropwisely at rt, the resulting mixture was stirred at rt for 2 h. The reaction mixture was concentrated to give crude (64S, 4S, Z) -4-amino-11-ethyl-12- (5- (3- ( (R) -3- (hydroxymethyl) morpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyeth-yl) pyridin-3-yl) -10, 10-dimethyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione which was used to next step without purification.
LCMS (ESI) calcd. for C43H53N7O6S [M+H] + m/z 796.4, found: 796.9.
Step 4: Synthesis of (1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (5- (3- ( (R) -3- (hydroxymethyl) morpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyeth-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
To a stirred solution of crude (64S, 4S, Z) -4-amino-11-ethyl-12- (5- (3- ( (R) -3- (hydroxymethyl) morpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyeth-yl) pyridin-3-yl) -10, 10-dimethyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione (256 mg, 0.29 mmol) and (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxylic acid (32 mg, 0.29 mmol) and 2, 6-lutidine (181 mg, 1.68 mmol) in ACN (10 mL) was added COMU (181 mg, 0.42 mmol) at rt, the resulting mixture was stirred at rt for 1 h. The reaction mixture was concentrated to give the crude product. The crude product was purified by pre-HPLC (eluting with ACN/H2O (0.5 %NH4HCO3) from 20%to 95%in 30 min) to give (1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (5- (3- ( (R) -3- (hydroxymethyl) morpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyeth-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide (98 mg) as a white solid.
LCMS (ESI) calcd. for C49H61N7O7S [M+H] + m/z 892.4, found: 892.9.
1H NMR (400 MHz, DMSO-d6) δ 8.78 (d, J = 1.8 Hz, 1H) , 8.43 -8.35 (m, 2H) , 7.87 -7.76 (m, 2H) , 7.73 (d, J = 8.6 Hz, 1H) , 7.56 (d, J = 8.7 Hz, 1H) , 5.91 (d, J = 11.2 Hz, 1H) , 5.36 (s, 1H) , 4.68 -4.41 (m, 3H) , 4.35 -4.18 (m, 2H) , 4.05 (dd, J = 14.8, 7.2 Hz, 1H) , 3.86 -3.61 (m, 4H) , 3.62 -3.39 (m, 4H) , 3.33 (s, 1H) , 3.27 -3.16 (m, 4H) , 3.17 -3.03 (m, 2H) , 2.92 (d, J = 14.1 Hz, 1H) , 2.68 (d, J = 11.4 Hz, 1H) , 2.63 -2.56 (m, 2H) , 2.53 (d, J = 10.6 Hz, 1H) , 2.45 -2.35 (m, 2H) , 2.34 -2.25 (m, 1H) , 2.13 (t, J = 9.9 Hz, 1H) , 1.60 -1.49 (m, 1H) , 1.34 (d, J = 6.0 Hz, 3H) , 1.18 (d, J = 24.3 Hz, 4H) , 1.06 (dd, J = 10.2, 5.7 Hz, 5H) , 0.87 (d, J = 10.1 Hz, 5H) , 0.32 (s, 3H) .
LCMS (ESI) : 892.4 [M+1] +. 1H NMR (400 MHz, DMSO) δ 8.78 (d, J = 1.8 Hz, 1H) , 8.43 -8.35 (m, 2H) , 7.87 -7.76 (m, 2H) , 7.73 (d, J = 8.6 Hz, 1H) , 7.56 (d, J = 8.7 Hz, 1H) , 5.91 (d, J = 11.2 Hz, 1H) , 5.36 (s, 1H) , 4.68 -4.41 (m, 3H) , 4.35 -4.18 (m, 2H) , 4.05 (dd, J = 14.8, 7.2 Hz, 1H) , 3.86 -3.61 (m, 4H) , 3.62 -3.39 (m, 4H) , 3.33 (s, 1H) , 3.27 -3.16 (m, 4H) , 3.17 -3.03 (m, 2H) , 2.92 (d, J = 14.1 Hz, 1H) , 2.68 (d, J = 11.4 Hz, 1H) , 2.63 -2.56 (m, 2H) , 2.53 (d, J = 10.6 Hz, 1H) , 2.45 -2.35 (m, 2H) , 2.34 -2.25 (m, 1H) , 2.13 (t, J = 9.9 Hz, 1H) , 1.60 -1.49 (m, 1H) , 1.34 (d, J = 6.0 Hz, 3H) , 1.18 (d, J = 24.3 Hz, 4H) , 1.06 (dd, J = 10.2, 5.7 Hz, 5H) , 0.87 (d, J = 10.1 Hz, 5H) , 0.32 (s, 3H) .
Example 20
(1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (5- (3- ( (S) -3- (hydroxymethyl) morpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -in dola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
Method 1:
Substituting (S) -4- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) morpholine with ( (S) -4- (3- (5-bromo-6- ( (S) -1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) morpholin-3-yl) methanol in the Step 1 of Example 16, the title compound was prepared by the same procedures as described for Example 16.
Method 2:
Step 1: Preparation of (1r, 2R, 3S) -N- ( (64S, 4S, Z) -12- (5- (3- ( (S) -3- (hydroxymethyl) morpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
A mixture of ( (S) -4- (3- (5-bromo-6- ( (S) -1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) morpholin-3-yl) methanol (70.2 mg, 0.19 mmol) , (1r, 2R, 3S) -N- ( (64S, 4S, Z) -10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide (160 mg, 0.23 mmol) , K2CO3 (79 mg, 0.57 mmol) and Pd (dppf) Cl2 (27.8 mg, 0.04 mmol) in dioxane (8 mL) and H2O (1.6 mL) was stirred at 85℃ under N2 for 16h. After completion, the reaction mixture was concentrated under reduced pressure. The residue was purified by FCC with MeOH/DCM = 0-7%to afford the (1r, 2R, 3S) -N- ( (64S, 4S, Z) -12- (5- (3- ( (S) -3- (hydroxymethyl) morpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyet hyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide (145 mg, 88%) as a yellow solid.
LCMS (ESI) calcd. for C47H57N7O7S [M+H] + m/z 864.4, found: 865.1
Step 2: Preparation of (1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (5- (3- ( (S) -3- (hydroxymethyl) morpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyeth-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
To a stirred solution of (1r, 2R, 3S) -N- ( (64S, 4S, Z) -12- (5- (3- ( (S) -3- (hydroxymethyl) morpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyet hyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide (115 mg, 0.133 mmol) in DMF (5 mL) were added Cs2CO3 (129.8 mg, 0.4 mmol) and Iodoethane (62.3 mg, 0.4 mmol) . The resulting mixture was stirred at rt for 0.5h. After completion, the reaction mixture was diluted with EtOAc (20 mL) and H2O (20 mL) . The mixture was extracted with EtOAc (10 mL*3) . The combined or-ganic layer was washed with brine (20 mL*2) , dried over anhydrous Na2SO4 and concentration under re-duced pressure. The residue was purified by pre-HPLC to give the (1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (5- (3- ( (S) -3- (hydroxymethyl) morpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyeth-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide (38 mg) as white sol-id.
LCMS (ESI) calcd. for C49H61N7O7S [M+H] + m/z 892.4, found: 892.9.
1H NMR (500 MHz, CDCl3) δ 8.39 (d, J = 2.0 Hz, 1H) , 8.20 (d, J = 13.0 Hz, 1H) , 7.95 (d, J = 2.0 Hz, 1H) , 7.77 (d, J = 2.4 Hz, 1H) , 7.69 (dd, J = 7.7, 2.4 Hz, 1H) , 7.59 (s, 1H) , 7.48 (d, J = 7.7 Hz, 1H) , 5.24 (q, J = 5.4 Hz, 1H) , 4.89 (d, J = 8.6 Hz, 1H) , 4.63 (dt, J = 13.0, 12.3 Hz, 1H) , 4.15 (q, J = 4.4 Hz, 2H) , 4.08 -3.99 (m, 2H) , 3.99 -3.89 (m, 2H) , 3.84 -3.78 (m, 1H) , 3.78 -3.73 (m, 3H) , 3.73 -3.64 (m, 4H) , 3.59 (d, J = 8.2 Hz, 1H) , 3.33 (s, 3H) , 3.31 -3.23 (m, 1H) , 3.22 (d, J = 3.5 Hz, 1H) , 3.11 -3.01 (m, 3H) , 3.01 -2.93 (m, 2H) , 2.72 (h, J = 7.9 Hz, 1H) , 2.47 (t, J = 8.6 Hz, 1H) , 2.13 -2.03 (m, 3H) , 2.03 -1.97 (m, 1H) , 1.74 (dq, J = 8.6, 5.2 Hz, 2H) , 1.67 (d, J = 5.3 Hz, 3H) , 1.37 (t, J = 4.4 Hz, 3H) , 1.15 (s, 3H) , 1.10 (s, 3H) , 0.95 (d, J = 5.1 Hz, 6H) .
Example 21
(2S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (64S, 4S, Z) -12- (5-ethynyl-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11- (2, 2, 2-trifluoro-ethyl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -3-methylbutanamide
Substituting (S) -4- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) -2-methylbut-3-yn-2-ol with (S) -3-bromo-5-ethynyl-2- (1-methoxyethyl) pyridine in the Step 1 of Example 2, Substituting iodoethane with 1, 1, 1-trifluoro-2-iodoethane in the Step 2 of Example 2 and Substituting (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxylic acid with (S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -3-meth ylbutanoic acid in the Step 4 of Example 2, the title compound was prepared by the same procedures as described for Example 2. LCMS (ESI) : 1052.5 [M+1] +. 1H NMR (500 MHz, CDCl3) δ 8.49 (d, J = 2.0 Hz, 1H) , 8.25 (d, J = 12.5 Hz, 1H) , 8.03 (d, J = 2.0 Hz, 1H) , 7.86 (d, J = 2.4 Hz, 1H) , 7.64 (dd, J = 7.7, 2.4 Hz, 1H) , 7.59 (s, 1H) , 7.46 (d, J = 7.9 Hz, 1H) , 5.24 (q, J = 5.4 Hz, 1H) , 4.90 (d, J = 8.4 Hz, 1H) , 4.72 (d, J =2.7 Hz, 2H) , 4.57 (q, J = 12.4 Hz, 1H) , 4.08 -3.99 (m, 2H) , 3.99 -3.89 (m, 2H) , 3.85 (d, J = 6.2 Hz, 1H) , 3.64 -3.54 (m, 3H) , 3.46 (s, 1H) , 3.37 (d, J = 1.3 Hz, 2H) , 3.33 (s, 3H) , 3.31 -3.18 (m, 2H) , 3.01 (d, J =2.0 Hz, 2H) , 2.99 -2.92 (m, 1H) , 2.88 -2.80 (m, 3H) , 2.79 -2.72 (m, 1H) , 2.70 (d, J = 8.0 Hz, 1H) , 2.41 (t, J = 1.5 Hz, 3H) , 2.21 (dp, J = 6.5, 5.7 Hz, 1H) , 2.13 -2.08 (m, 1H) , 2.08 -2.04 (m, 2H) , 2.04 -1.98 (m, 2H) , 1.94 (t, J = 6.7 Hz, 1H) , 1.89 -1.81 (m, 3H) , 1.67 (d, J = 5.3 Hz, 3H) , 1.21 (dd, J = 7.9, 4.9 Hz, 2H) , 1.15 (s, 3H) , 1.10 (s, 3H) , 1.05 (dd, J = 7.6, 5.0 Hz, 2H) , 0.95 (d, J = 5.7 Hz, 3H) , 0.90 (d, J = 5.5 Hz, 3H) .
Example 22
(2S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (64S, 4S, Z) -12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11- (2, 2, 2-trifluoroethyl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -3-methylbutanamide
Substituting iodoethane with 1, 1, 1-trifluoro-2-iodoethane in the Step 2 of Example 16 and Substitut-ing (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxylic acid with (S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -3-meth ylbutanoic acid in the Step 4 of Example 16, the title compound was prepared by the same procedures as described for Example 16. LCMS (ESI) : 1151.6 [M+1] +. 1H NMR (500 MHz, CDCl3) δ 8.39 (d, J = 2.0 Hz, 1H) , 8.25 (d, J = 12.5 Hz, 1H) , 8.01 (d, J = 1.8 Hz, 1H) , 7.86 (d, J = 2.4 Hz, 1H) , 7.64 (dd, J = 7.7, 2.4 Hz, 1H) , 7.59 (s, 1H) , 7.46 (d, J = 7.9 Hz, 1H) , 5.24 (q, J = 5.4 Hz, 1H) , 4.90 (d, J = 8.4 Hz, 1H) , 4.72 (d, J =2.7 Hz, 2H) , 4.57 (q, J = 12.4 Hz, 1H) , 4.08 -3.99 (m, 2H) , 3.99 -3.89 (m, 2H) , 3.85 (d, J = 6.2 Hz, 1H) , 3.73 (dd, J = 6.5, 3.8 Hz, 2H) , 3.66 (dd, J = 6.5, 3.8 Hz, 2H) , 3.63 -3.54 (m, 5H) , 3.37 (d, J = 1.3 Hz, 2H) , 3.33 (s, 3H) , 3.31 -3.18 (m, 2H) , 3.01 (d, J = 2.0 Hz, 2H) , 3.00 -2.92 (m, 1H) , 2.88 -2.80 (m, 3H) , 2.79 -2.66 (m, 6H) , 2.41 (t, J = 1.5 Hz, 3H) , 2.21 (dp, J = 6.5, 5.7 Hz, 1H) , 2.13 -2.08 (m, 1H) , 2.08 -2.04 (m, 2H) , 2.04 -1.97 (m, 2H) , 1.94 (t, J = 6.7 Hz, 1H) , 1.89 -1.81 (m, 3H) , 1.67 (d, J = 5.3 Hz, 3H) , 1.21 (dd, J = 7.9, 4.9 Hz, 2H) , 1.15 (s, 3H) , 1.10 (s, 3H) , 1.05 (dd, J = 7.6, 5.0 Hz, 2H) , 0.95 (d, J = 5.7 Hz, 3H) , 0.90 (d, J = 5.5 Hz, 3H) .
Example 23
(2S) -2-cyclopentyl-2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (22S, 64S, 4S) -12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11- (2, 2, 2-trifluoroethyl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) acetamide
Substituting tert-butyl ( (64S, 4S, Z) -10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate with tert-butyl ( (22S, 64S, 4S) -10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate in the Step 1 of Example 16, Substituting iodoethane with 1, 1, 1-trifluoro-2-iodoethane in the Step 2 of Ex-ample 16 and Substituting (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxylic acid with (S) -2-cyclopentyl-2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) acetic acid in the Step 4 of Example 16, the title compound was prepared by the same procedures as described for Example 16. LCMS (ESI) : 1179.7 [M+1] +. 1H NMR (500 MHz, CDCl3) δ 8.54 (s, 1H) , 8.38 (d, J = 1.3 Hz, 1H) , 8.14 (s, 1H) , 8.06 (d, J = 1.3 Hz, 1H) , 7.56 (s, 1H) , 7.33 (s, 1H) , 7.13 (s, 1H) , 4.76 (s, 1H) , 4.57 -4.51 (m, 2H) , 4.46 (d, J = 12.5 Hz, 1H) , 4.32 (s, 1H) , 4.24 (s, 1H) , 4.15 (s, 1H) , 4.01 (d, J =13.7 Hz, 2H) , 3.89 (d, J = 12.8 Hz, 2H) , 3.84 -3.78 (m, 3H) , 3.77 (s, 1H) , 3.73 (d, J = 17.2 Hz, 3H) , 3.68 (s, 2H) , 3.63 (s, 2H) , 3.50 (d, J = 16.8 Hz, 2H) , 3.38 -3.24 (m, 7H) , 2.85 -2.75 (m, 3H) , 2.67 (s, 1H) , 2.62 -2.55 (m, 6H) , 2.52 (s, 1H) , 2.47 (d, J = 10.1 Hz, 4H) , 2.25 (s, 1H) , 2.20 (d, J = 10.3 Hz, 2H) , 1.98 (s, 1H) , 1.92 (d, J = 11.5 Hz, 2H) , 1.88 (s, 1H) , 1.85 -1.77 (m, 4H) , 1.75 (d, J = 12.3 Hz, 1H) , 1.72 -1.65 (m, 7H) , 1.59 (d, J = 13.0 Hz, 2H) , 1.44 (d, J = 13.0 Hz, 2H) , 1.37 (d, J = 4.9 Hz, 2H) , 1.23 (d, J = 4.9 Hz, 2H) , 1.16 (s, 3H) , 1.11 (s, 3H) .
Example 24
1- (4- (dimethylamino) -4-methylpent-2-ynoyl) -N- ( (2S) -1- ( ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) -4-fluoro-N-methylpiperidine-4-carboxamide
Substituting (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxylic acid with N- (1- (4- (dimethylamino) -4-methylpent-2-ynoyl) -4-fluoropiperidine-4-carbonyl) -N-methyl-L-valine in the Step 4 of Example 16, the title compound was prepared by the same procedures as described for Example 16.LCMS (ESI) : 1145.6 [M+1] +. 1H NMR (500 MHz, CDCl3) δ 8.39 (d, J = 2.0 Hz, 1H) , 8.25 (d, J = 12.6 Hz, 1H) , 8.00 (d, J = 2.0 Hz, 1H) , 7.77 (d, J = 2.4 Hz, 1H) , 7.69 (dd, J = 7.7, 2.4 Hz, 1H) , 7.59 (s, 1H) , 7.48 (d, J = 7.7 Hz, 1H) , 5.24 (q, J = 5.4 Hz, 1H) , 4.90 (d, J = 8.4 Hz, 1H) , 4.75 (d, J = 7.3 Hz, 1H) , 4.65 (q, J =12.4 Hz, 1H) , 4.15 (q, J = 4.4 Hz, 2H) , 4.08 -3.99 (m, 2H) , 3.99 -3.89 (m, 2H) , 3.76 -3.68 (m, 4H) , 3.68 -3.54 (m, 6H) , 3.33 (s, 3H) , 3.32 -3.18 (m, 2H) , 3.06 (d, J = 2.6 Hz, 2H) , 2.96 (s, 3H) , 2.78 -2.66 (m, 5H) , 2.47 (dt, J = 9.3, 6.5 Hz, 4H) , 2.35 (s, 6H) , 2.21 (dhept, J = 7.3, 5.1 Hz, 1H) , 2.13 -1.97 (m, 4H) , 1.67 (d, J = 5.3 Hz, 3H) , 1.43 -1.35 (m, 9H) , 1.15 (s, 3H) , 1.10 (s, 3H) , 0.79 (d, J = 5.1 Hz, 3H) , 0.75 (d, J = 5.0 Hz, 3H) .
Example 25
1- (4- (dimethylamino) -4-methylpent-2-ynoyl) -N- ( (2S) -1- ( ( (64S, 4S, Z) -11-ethyl-12- (5-ethynyl-2- ( (S) -1-methoxyeth-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicy-clo [3.1.1] heptanacycloundecaphane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) -4-fluoro-N-methylpiperidine-4-carboxamide
Substituting (S) -4- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) -2-methylbut-3-yn-2-ol with (S) -3-bromo-5-ethynyl-2- (1-methoxyethyl) pyridine in the Step 1 of Example 2 and Substituting (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxylic acid with N- (1- (4- (dimethylamino) -4-methylpent-2-ynoyl) -4-fluoropiperidine-4-carbonyl) -N-methyl-L-valine in the Step 4 of Example 2, the title compound was prepared by the same procedures as described for Example 2. LCMS (ESI) : 1046.5 [M+1] +. 1H NMR (500 MHz, CDCl3) δ 8.49 (d, J = 2.0 Hz, 1H) , 8.25 (d, J = 12.6 Hz, 1H) , 8.08 (d, J = 2.0 Hz, 1H) , 7.77 (d, J = 2.4 Hz, 1H) , 7.69 (dd, J = 7.7, 2.4 Hz, 1H) , 7.59 (s, 1H) , 7.48 (d, J = 7.7 Hz, 1H) , 5.24 (q, J = 5.4 Hz, 1H) , 4.90 (d, J = 8.4 Hz, 1H) , 4.75 (d, J = 7.3 Hz, 1H) , 4.65 (q, J =12.4 Hz, 1H) , 4.15 (q, J = 4.4 Hz, 2H) , 4.08 -3.99 (m, 2H) , 3.99 -3.89 (m, 2H) , 3.71 (dd, J = 9.3, 6.8 Hz, 2H) , 3.62 (dd, J = 9.4, 6.7 Hz, 2H) , 3.46 (s, 1H) , 3.33 (s, 3H) , 3.32 -3.18 (m, 2H) , 3.06 (d, J = 2.6 Hz, 2H) , 2.96 (s, 3H) , 2.72 (h, J = 7.9 Hz, 1H) , 2.47 (dt, J = 9.3, 6.5 Hz, 4H) , 2.35 (s, 6H) , 2.21 (dhept, J = 7.3, 5.1 Hz, 1H) , 2.13 -1.97 (m, 4H) , 1.67 (d, J = 5.3 Hz, 3H) , 1.43 -1.35 (m, 9H) , 1.15 (s, 3H) , 1.10 (s, 3H) , 0.79 (d, J = 5.1 Hz, 3H) , 0.75 (d, J = 5.0 Hz, 3H) .
Example 26
1- (4- (dimethylamino) -4-methylpent-2-ynoyl) -N- ( (2S) -1- ( ( (22S, 64S, 4S) -11-ethyl-12- (5-ethynyl-2- ( (S) -1-methoxyeth-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) -4-fluoro-N-methylpiperidine-4-carboxamide
Method 1:
Substituting (S) -4- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) morpholine with (S) -3-bromo-5-ethynyl-2- (1-methoxyethyl) pyridine in the Step 1 of Example 23 and Substituting (S) -2-cyclopentyl-2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) acetic acid with N- (1- (4- (dimethylamino) -4-methylpent-2-ynoyl) -4-fluoropiperidine-4-carbonyl) -N-methyl-L-valine in the Step 4 of Example 23, the title compound was prepared by the same procedures as described for Example 23.
Method 2:
Step 1: Synthesis of methyl tert-butyl ( (22S, 64S, 4S) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( (triethylsilyl) ethynyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-di-oxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
A mixture of 5- ( (22S, 64S, 4S) -4- ( (tert-butoxycarbonyl) amino) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyet hyl) pyridin-3-yl trifluoromethanesulfonate (190 mg, 0.21 mmol) , (triethylsilyl) acetylene (149 mg, 1.06 mmol) , CuI (8 mg, 0.04 mmol) and Pd (PPh3) 4 (49 mg, 0.04 mmol) in TEA/DMF (2 mL/10 mL) was stirred at 100℃ for 1 h under N2 atmosphere, the reaction was monitored by LCMS. After completion, the mixture was filtered and the filtrate was concentrated, the residue was purified by silica gel column eluting with EtOAc/PE from 0%to 100%to afford tert-butyl ( (22S, 64S, 4S) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( (triethylsilyl) ethynyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (170 mg, 90 %) as a light yellow solid.
LCMS (ESI) calcd. for C49H70N6O7Si [M+H] + m/z 883.5, found: 884.4.
Step 2: Synthesis of (22S, 64S, 4S) -4-amino-11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( (triethylsilyl) ethynyl) pyridin-3-yl) -10, 10-dimethyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione
To a stirred solution of tert-butyl ( (22S, 64S, 4S) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( (triethylsilyl) ethynyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (170 mg, 0.2 mmol) in dichloromethane (3 mL) was added TFA (1 mL) dropwise at rt, the resulting mixture was stirred at rt for 1 h. The reaction mixture was washed with sat. NaHCO3 (3 mL) , brine (3 mL) , then dried over Na2SO4, following with concentration under reduced pressure to obtain crude product (22S, 64S, 4S) -4-amino-11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( (triethylsilyl) ethynyl) pyridin-3-yl) -10, 10-dime-thyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione (150 mg, crude) as a yellow solid.
LCMS (ESI) calcd. for C44H62N6O5Si [M+H] + m/z 783.5, found: 784.5.
Step 3: Synthesis of tert-butyl 4- ( ( (2S) -1- ( ( (22S, 64S, 4S) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( (triethylsilyl) ethynyl) pyridin-3-yl) -10, 10-dime-thyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacyclou ndecaphane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) (methyl) carbamoyl) -4-fluoropiperidine-1-carboxylate
To a solution of N- (1- (tert-butoxycarbonyl) -4-fluoropiperidine-4-carbonyl) -N-methyl-L-valine (83 mg, 0.23 mmol) in DMF (3 mL) were added HATU (109 mg, 0.29 mmol) and DIEA (123 mg, 0.96 mmol) at 0-10℃. The resulting mixture was stirred at 0-10℃ for 10 min. Then (22S, 64S, 4S) -4-amino-11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( (triethylsilyl) ethynyl) pyridin-3-yl) -10, 10-dime-thyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione (150 mg, 0.20 mmol) was added into the reaction mixture, the resulting mixture was stirred at rt for 2 h. The reaction was monitored by LCMS. After completion, the reaction mixture was diluted with brine and extracted with EtOAc (2 x 30 mL) , the combined organic phase was washed with saturated NaCl (100 mL) , then dried over Na2SO4, following with concentration under reduced pressure to obtain crude product which was purified by silica gel column eluting with EtOAc/PE from 0%to 100%to afford tert-butyl 4- ( ( (2S) -1- ( ( (22S, 64S, 4S) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( (triethylsilyl) ethynyl) pyridin-3-yl) -10, 10-dime-thyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacyclou ndecaphane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) (methyl) carbamoyl) -4-fluoropiperidine-1-carboxylate (40 mg, 19 %) as a light yellow solid.
LCMS (ESI) calcd. for C61H89FN8O9Si [M+H] + m/z 1125.7, found: 1126.7.
Step 4: Synthesis of tert-butyl 4- ( ( (2S) -1- ( ( (22S, 64S, 4S) -11-ethyl-12- (5-ethynyl-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) (methyl) carbamoyl) -4-fluoropiperidine-1-carboxylate
To a solution of tert-butyl 4- ( ( (2S) -1- ( ( (22S, 64S, 4S) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( (triethylsilyl) ethynyl) pyridin-3-yl) -10, 10-dime-thyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) (methyl) carbamoyl) -4-fluoropiperidine-1-carboxylate (40 mg, 0.036 mmol) in THF (3 mL) was added TBAF (0.07 mL, 1M in THF, 0.07 mmol) at 0-10℃. The resulting mixture was stirred at rt for 2 h. The reaction was monitored by LCMS. After completion, the re-action mixture was diluted with brine and extracted with EtOAc (2 x 30 mL) , the combined organic phase was washed with saturated NaCl (100 mL) , then dried over Na2SO4, following with concentration under reduced pressure to obtain crude tert-butyl 4- ( ( (2S) -1- ( ( (22S, 64S, 4S) -11-ethyl-12- (5-ethynyl-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) (methyl) carbamoyl) -4-fluoropiperidine-1-carboxylate (36 mg, crude) as a light yellow solid.
LCMS (ESI) calcd. for C55H75FN8O9 [M+H] + m/z 1011.6, found: 1012.7.
Step 5: Synthesis of N- ( (2S) -1- ( ( (22S, 64S, 4S) -11-ethyl-12- (5-ethynyl-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) -4-fluoro-N-methylpiperidine-4-carboxamide
To a stirred solution of tert-butyl 4- ( ( (2S) -1- ( ( (22S, 64S, 4S) -11-ethyl-12- (5-ethynyl-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) (methyl) carbamoyl) -4-fluoropiperidine-1-carboxylate (36 mg, 0.036 mmol) in dichloromethane (3 mL) was added TFA (1 mL) dropwise at rt, the resulting mixture was stirred at rt for 1 h. The reaction mixture was washed with sat. NaHCO3 (3 mL) , brine (3 mL) , then dried over Na2SO4, following with concentration under reduced pressure to obtain crude product N- ( (2S) -1- ( ( (22S, 64S, 4S) -11-ethyl-12- (5-ethynyl-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) -4-fluoro-N-methylpiperidine-4-carboxamide (32 mg, crude) as a yellow solid.
LCMS (ESI) calcd. for C37H47F3N6O8S [M+H] + m/z 911.5, found: 912.1.
Step 6: Synthesis of 1- (4- (dimethylamino) -4-methylpent-2-ynoyl) -N- ( (2S) -1- ( ( (22S, 64S, 4S) -11-ethyl-12- (5-ethynyl-2- ( (S) -1-meth oxyeth-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicy-clo[3.1.1] heptanacycloundecaphane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) -4-fluoro-N-methylpiperidine-4-carboxamide
To a solution of 4- (dimethylamino) -4-methylpent-2-ynoic acid (9.2 mg, 0.06 mmol) in DMF (2 mL) were added HATU (23 mg, 0.06 mmol) and DIEA (25 mg, 0.20 mmol) at 0-10℃. The resulting mixture was stirred at 0-10℃ for 10 min. Then N- ( (2S) -1- ( ( (22S, 64S, 4S) -11-ethyl-12- (5-ethynyl-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) -4-fluoro-N-methylpiperidine-4-carboxamide (36 mg, 0.04 mmol) was added into the reaction mixture, the resulting mixture was stirred at rt for 2 h. The reaction was moni-tored by LCMS. After completion, the reaction mixture was diluted with brine and extracted with EtOAc (2 x 30 mL) , the combined organic phase was washed with saturated NaCl (100 mL) , then dried over Na2SO4, following with concentration under reduced pressure to obtain crude product which was purified by pre-HPLC (ACN/H2O (0.5 %NH4HCO3) from 20%to 95%in 30 min) to give 1- (4- (dimethylamino) -4-methylpent-2-ynoyl) -N- ( (2S) -1- ( ( (22S, 64S, 4S) -11-ethyl-12- (5-ethynyl-2- ( (S) -1-met hoxyeth-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicy-clo [3.1.1] heptanacycloundecaphane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) -4-fluoro-N-methylpiperidine-4-carboxamide (5 mg, 12%) as a white solid.
LCMS (ESI) calcd. for C61H79N11O7 [M+H] + m/z 1048.6, found: 1049.5.
1H NMR (500 MHz, CDCl3) δ 8.49 (d, J = 2.0 Hz, 1H) , 8.24 (d, J = 12.1 Hz, 1H) , 8.06 (d, J = 2.0 Hz, 1H) , 7.36 (d, J = 7.9 Hz, 1H) , 7.21 (d, J = 2.2 Hz, 1H) , 7.01 (dd, J = 7.9, 2.2 Hz, 1H) , 5.24 (q, J = 5.4 Hz, 1H) , 4.90 (d, J = 8.6 Hz, 1H) , 4.75 (d, J = 7.3 Hz, 1H) , 4.47 (q, J = 11.9 Hz, 1H) , 4.15 (q, J = 4.4 Hz, 2H) , 4.12 -4.06 (m, 1H) , 4.06 -3.99 (m, 2H) , 3.99 -3.89 (m, 2H) , 3.86 (dd, J = 7.5, 4.8 Hz, 1H) , 3.81 (dd, J =7.5, 4.8 Hz, 1H) , 3.74 -3.65 (m, 3H) , 3.65 -3.58 (m, 3H) , 3.55 (d, J = 8.6 Hz, 1H) , 3.48 -3.42 (m, 2H) , 3.33 (s, 3H) , 3.09 (s, 2H) , 2.96 (s, 3H) , 2.72 (h, J = 7.9 Hz, 1H) , 2.47 (dt, J = 9.3, 6.5 Hz, 4H) , 2.35 (s, 6H) , 2.25 -1.97 (m, 7H) , 1.67 (d, J = 5.3 Hz, 3H) , 1.43 -1.35 (m, 9H) , 1.15 (s, 3H) , 1.10 (s, 3H) , 0.79 (d, J =5.1 Hz, 3H) , 0.75 (d, J = 5.0 Hz, 3H) .
Example 27
(2S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -3-methyl-2- (1, 3, 3-trimethylureido) butanamide
Substituting (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxylic acid with N- (dimethylcarbamoyl) -N-methyl-L-valine in the Step 4 of Example 16, the title compound was prepared by the same procedures as described for Example 16. LCMS (ESI) : 950.5 [M+1] +. 1H NMR (500 MHz, CDCl3) δ 8.39 (d, J = 2.0 Hz, 1H) , 8.25 (d, J = 12.6 Hz, 1H) , 8.00 (d, J = 2.0 Hz, 1H) , 7.77 (d, J = 2.3 Hz, 1H) , 7.69 (dd, J = 7.7, 2.4 Hz, 1H) , 7.59 (s, 1H) , 7.48 (d, J = 7.7 Hz, 1H) , 5.24 (q, J = 5.4 Hz, 1H) , 4.89 (d, J = 8.6 Hz, 1H) , 4.70 -4.59 (m, 2H) , 4.15 (q, J = 4.4 Hz, 2H) , 4.08 -3.99 (m, 2H) , 3.99 -3.89 (m, 2H) , 3.73 (dd, J = 6.5, 3.8 Hz, 2H) , 3.66 (dd, J = 6.5, 3.8 Hz, 2H) , 3.58 (d, J = 2.6 Hz, 2H) , 3.33 (s, 3H) , 3.32 -3.18 (m, 2H) , 3.06 (d, J = 2.6 Hz, 2H) , 2.93 (d, J = 1.6 Hz, 9H) , 2.78 -2.66 (m, 5H) , 2.21 (dhept, J = 8.1, 5.1 Hz, 1H) , 2.13 -1.97 (m, 4H) , 1.67 (d, J = 5.3 Hz, 3H) , 1.37 (t, J = 4.4 Hz, 3H) , 1.15 (s, 3H) , 1.10 (s, 3H) , 0.79 (d, J = 5.1 Hz, 3H) , 0.75 (d, J = 5.1 Hz, 3H) .
Example 28
N- ( (2S) -1- ( ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundeca-phane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) -N-methylmorpholine-4-carboxamide
Substituting (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxylic acid with N-methyl-N- (morpholine-4-carbonyl) -L-valine in the Step 4 of Example 16, the title compound was pre-pared by the same procedures as described for Example 16. LCMS (ESI) : 992.5 [M+1] +. 1H NMR (500 MHz, CDCl3) δ 8.39 (d, J = 2.0 Hz, 1H) , 8.25 (d, J = 12.6 Hz, 1H) , 8.00 (d, J = 2.0 Hz, 1H) , 7.77 (d, J =2.3 Hz, 1H) , 7.69 (dd, J = 7.7, 2.4 Hz, 1H) , 7.59 (s, 1H) , 7.48 (d, J = 7.7 Hz, 1H) , 5.24 (q, J = 5.4 Hz, 1H) , 4.89 (d, J = 8.6 Hz, 1H) , 4.70 -4.59 (m, 2H) , 4.15 (q, J = 4.4 Hz, 2H) , 4.08 -3.99 (m, 2H) , 3.99 -3.89 (m, 2H) , 3.73 (dd, J = 6.5, 3.8 Hz, 2H) , 3.66 (dd, J = 6.5, 3.8 Hz, 2H) , 3.64 -3.58 (m, 2H) , 3.58 -3.51 (m, 8H) , 3.33 (s, 3H) , 3.31 -3.18 (m, 2H) , 3.06 (d, J = 2.6 Hz, 2H) , 2.93 (s, 3H) , 2.78 -2.66 (m, 5H) , 2.22 (dhept, J = 8.1, 5.1 Hz, 1H) , 2.13 -1.97 (m, 4H) , 1.67 (d, J = 5.3 Hz, 3H) , 1.37 (t, J = 4.4 Hz, 3H) , 1.15 (s, 3H) , 1.10 (s, 3H) , 0.79 (d, J = 5.1 Hz, 3H) , 0.75 (d, J = 5.0 Hz, 3H) .
Example 29
(2R, 3R) -N- ( (2S) -1- ( ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) -N-methyl-3-phenyltetrahydrofuran-2-carboxamide
Substituting (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxylic acid with N-methyl-N- ( (2R, 3R) -3-phenyltetrahydrofuran-2-carbonyl) -L-valine in the Step 4 of Example 16, the title compound was prepared by the same procedures as described for Example 16. LCMS (ESI) : 1053.5 [M+1] +. 1H NMR (500 MHz, CDCl3) δ 8.39 (d, J = 2.0 Hz, 1H) , 8.25 (d, J = 12.6 Hz, 1H) , 8.00 (d, J = 2.0 Hz, 1H) , 7.77 (d, J = 2.4 Hz, 1H) , 7.69 (dd, J = 7.7, 2.4 Hz, 1H) , 7.59 (s, 1H) , 7.48 (d, J = 7.7 Hz, 1H) , 7.33 -7.22 (m, 5H) , 5.24 (q, J = 5.4 Hz, 1H) , 4.90 (d, J = 8.4 Hz, 1H) , 4.79 (d, J = 8.1 Hz, 1H) , 4.75 (d, J = 7.3 Hz, 1H) , 4.65 (q, J = 12.4 Hz, 1H) , 4.15 (q, J = 4.4 Hz, 2H) , 4.08 -3.99 (m, 2H) , 3.99 -3.94 (m, 2H) , 3.94 -3.89 (m, 1H) , 3.89 -3.84 (m, 1H) , 3.79 -3.70 (m, 3H) , 3.66 (dd, J = 6.5, 3.8 Hz, 2H) , 3.58 (d, J = 2.6 Hz, 2H) , 3.33 (s, 3H) , 3.31 -3.18 (m, 2H) , 3.06 (d, J = 2.6 Hz, 2H) , 2.97 (s, 3H) , 2.78 -2.66 (m, 5H) , 2.30 (ddd, J = 7.5, 5.5, 4.8 Hz, 1H) , 2.26 -2.15 (m, 2H) , 2.13 -1.97 (m, 4H) , 1.67 (d, J = 5.3 Hz, 3H) , 1.37 (t, J = 4.4 Hz, 3H) , 1.15 (s, 3H) , 1.10 (s, 3H) , 0.79 (d, J = 5.1 Hz, 3H) , 0.74 (d, J = 5.0 Hz, 3H) .
Example 30
methyl (3S, 4R) -3- ( ( (2S) -1- ( ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) (methyl) carbamoyl) -4-methylpyrrolidine-1-carboxylate
Substituting (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxylic acid with N- ( (3S, 4R) -1- (methoxycarbonyl) -4-methylpyrrolidine-3-carbonyl) -N-methyl-L-valine in the Step 4 of Example 16, the title compound was prepared by the same procedures as described for Example 16. LCMS (ESI) : 1048.5 [M+1] +. 1H NMR (500 MHz, CDCl3) δ 8.39 (d, J = 2.0 Hz, 1H) , 8.30 (d, J = 12.6 Hz, 1H) , 8.00 (d, J = 2.0 Hz, 1H) , 7.77 (d, J = 2.3 Hz, 1H) , 7.69 (dd, J = 7.7, 2.4 Hz, 1H) , 7.59 (s, 1H) , 7.48 (d, J = 7.7 Hz, 1H) , 5.24 (q, J = 5.4 Hz, 1H) , 4.90 (d, J = 8.4 Hz, 1H) , 4.77 (d, J = 8.1 Hz, 1H) , 4.65 (q, J =12.4 Hz, 1H) , 4.15 (q, J = 4.4 Hz, 2H) , 4.08 -3.99 (m, 2H) , 3.99 -3.89 (m, 2H) , 3.79 -3.70 (m, 3H) , 3.69 -3.61 (m, 6H) , 3.61 -3.54 (m, 2H) , 3.54 -3.48 (m, 1H) , 3.40 (dd, J = 9.5, 6.4 Hz, 1H) , 3.33 (s, 3H) , 3.31 -3.21 (m, 2H) , 3.21 -3.12 (m, 1H) , 3.06 (d, J = 2.6 Hz, 2H) , 2.99 (s, 3H) , 2.78 -2.66 (m, 5H) , 2.54 (dt, J = 12.4, 6.3 Hz, 1H) , 2.20 (dhept, J = 8.1, 5.1 Hz, 1H) , 2.13 -1.97 (m, 4H) , 1.67 (d, J = 5.3 Hz, 3H) , 1.37 (t, J = 4.4 Hz, 3H) , 1.15 (s, 3H) , 1.10 (s, 3H) , 1.01 (d, J = 6.2 Hz, 3H) , 0.79 (d, J = 5.1 Hz, 3H) , 0.74 (d, J =5.0 Hz, 3H) .
Example 31
(1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- (4-methylpiperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
Step 1: Preparation of tert-butyl ( (64S, 4S, Z) -12- (2- ( (S) -1-methoxyethyl) -5- (3- (4- (methylsulfonyl) piperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptan acycloundecaphane-4-yl) carbamate
A mixture of (S) -1- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) -4- (methylsulfonyl) piperazine (118.1 mg, 0.28 mmol) , tert-butyl ( (64S, 4S, Z) -10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (200 mg, 0.28 mmol) , K2CO3 (117.5 mg, 0.85 mmol) and Pd (dppf) Cl2 (41.5 mg, 0.06 mmol) in dioxane (6 mL) and H2O (1.5 mL) was stirred at 85℃ under N2 for 4h. After completion, the reaction mixture was concentrated under reduced pressure. The residue was purified by FCC with MeOH/DCM = 0-5%to afford the tert-butyl ( (64S, 4S, Z) -12- (2- ( (S) -1-methoxyethyl) -5- (3- (4- (methylsulfonyl) piperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] hept anacycloundecaphane-4-yl) carbamate 7 (176 mg, 69%) as a yellow solid.
LCMS (ESI) calcd. for C46H58N8O8S2 [M+H] + m/z 915.4, found: 915.9
Step 2: Preparation of tert-butyl ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- (4- (methylsulfonyl) piperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
To a stirred solution of tert-butyl ( (64S, 4S, Z) -12- (2- ( (S) -1-methoxyethyl) -5- (3- (4- (methylsulfonyl) piperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] hept anacycloundecaphane-4-yl) carbamate (176 mg, 0.192 mmol) in DMF (5 mL) were added Cs2CO3 (187.7 mg, 0.577 mmol) and Iodoethane (90 mg, 0.577 mmol) . The resulting mixture was stirred at rt for 2h. After completion, the reaction mixture was diluted with EtOAc (5 mL) and H2O (10 mL) . The mixture was ex-tracted with EtOAc (10 mL*3) . The combined organic layer was washed with brine (10 mL*2) , dried over anhydrous Na2SO4 and concentration under reduced pressure. The residue was purified by FCC with MeOH/DCM = 0-5%to afford the tert-butyl ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- (4- (methylsulfonyl) piperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (131 mg, 72%) as white solid.
LCMS (ESI) calcd. for C48H62N8O8S2 [M+H] + m/z 943.4, found: 943.9
Step 3: Preparation of (64S, 4S, Z) -4-amino-11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- (4- (methylsulfonyl) piperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione
To a stirred solution of tert-butyl ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- (4- (methylsulfonyl) piperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (131 mg, 0.138 mmol) in DCM (5 mL) was added TFA (1 mL) . The resulting mixture was stirred at rt for 1 h. The reaction mixture was concentrated under reduced pressure to give (64S, 4S, Z) -4-amino-11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- (4- (methylsulfonyl) piperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione (162 mg) of its TFA salt as a yellow oil.
LCMS (ESI) calcd. for C43H54N8O6S2 [M+H] + m/z 843.4, found: 843.8
Step 4: Preparation of (1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- (4- (methylsulfonyl) piperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
To a stirred solution of (64S, 4S, Z) -4-amino-11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- (4- (methylsulfonyl) piperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione (116 mg, 0.138 mmol) in ACN (3 mL) and DCM (3 mL) was added (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxylic acid (17.4 mg, 0.152 mmol) , TCFH (116 mg, 0.414 mmol) and NMI (113 mg, 1.38 mmol) . The resulting mixture was stirred at rt for 0.5h. The reaction mix-ture was concentrated under reduced pressure. The residue was purified by pre-TLC with EtOAc to give the (1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- (4- (methylsulfonyl) piperazin-1-yl) prop -1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2 , 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide as atropiso-mer 1 (11 mg) and (1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- (4- (methylsulfonyl) piperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide as atropiso-mer 2 (i.e., compound of Example 31) (15 mg) as white solids.
LCMS (ESI) calcd. for C49H62N8O7S2 [M+H] + m/z 939.4, found: 939.9
atropisomer 1: 1H NMR (400 MHz, DMSO-d6) δ 8.82 -8.76 (m, 1H) , 8.40 (t, J = 10.7 Hz, 2H) , 7.87 (d, J = 2.1 Hz, 1H) , 7.80 (s, 1H) , 7.73 (d, J = 7.3 Hz, 1H) , 7.56 (d, J = 8.6 Hz, 1H) , 5.91 (d, J = 11.3 Hz, 1H) , 5.35 (s, 1H) , 4.64 (d, J = 10.9 Hz, 1H) , 4.46 (dd, J = 9.6, 4.2 Hz, 1H) , 4.37 -4.21 (m, 2H) , 4.05 (dd, J = 15.4, 7.9 Hz, 1H) , 3.67 -3.48 (m, 4H) , 3.27 -3.19 (m, 3H) , 3.17 -3.08 (m, 4H) , 2.86 (s, 2H) , 2.70 -2.56 (m, 4H) , 2.32 (ddd, J = 15.5, 12.2, 10.3 Hz, 2H) , 2.17 -2.10 (m, 1H) , 1.55 (s, 1H) , 1.37 -0.99 (m, 18H) , 0.94 -0.73 (m, 7H) , 0.31 (s, 2H) .
atropisomer 2: 1H NMR (400 MHz, DMSO-d6) δ 8.82 (d, J = 1.8 Hz, 1H) , 8.42 (d, J = 8.6 Hz, 2H) , 8.03 (d, J = 1.9 Hz, 1H) , 7.82 (s, 1H) , 7.74 (d, J = 7.9 Hz, 1H) , 7.54 (d, J = 8.7 Hz, 1H) , 5.89 (d, J = 11.1 Hz, 1H) , 5.31 (t, J = 8.1 Hz, 1H) , 4.73 (d, J = 11.0 Hz, 1H) , 4.49 (d, J = 5.0 Hz, 1H) , 3.99 -3.90 (m, 2H) , 3.82 (dd, J = 14.6, 7.2 Hz, 1H) , 3.70 -3.57 (m, 3H) , 3.51 (d, J = 10.6 Hz, 1H) , 3.25 (d, J = 14.6 Hz, 1H) , 3.19 -2.99 (m, 8H) , 2.88 (s, 2H) , 2.64 (s, 4H) , 2.40 -2.28 (m, 2H) , 2.15 (t, J = 9.6 Hz, 1H) , 1.62 (t, J = 9.4 Hz, 1H) , 1.31 -1.04 (m, 18H) , 0.94 -0.77 (m, 4H) , 0.48 (s, 3H) .
Example 32
(2S) -4- (3- (5- ( (64S, 4S, Z) -4- ( (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxamido) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundeca-phane-12-yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) -N, N-dimethylmorpholine-2-carboxamide
Step 1: tert-butyl (S) -2- (dimethylcarbamoyl) morpholine-4-carboxylate
To a stirred solution of (S) -4- (tert-butoxycarbonyl) morpholine-2-carboxylic acid (833 mg, 3.60 mmol) , dimethylamine hydrochloride (352 mg, 4.32 mmol) in DMF (10 mL) were added DIEA (1.4 g, 10.81 mmol) and HATU (2.1 g, 5.40 mmol) at rt , the resulting mixture was stirred at rt for 22 h. The reac-tion was monitored by LCMS. After completion, The reaction was diluted with H2O (100 mL) and ex-tracted with EtOAc (20 mL x 2) , the combined organic phase was washed with saturated NaCl (500 mL) , then dried over Na2SO4, following with concentration under reduced pressure to obtain crude product which was purified by silica gel column (eluting with CH3OH/DCM from 0%to 10%) to afford tert-butyl (S) -2- (dimethylcarbamoyl) morpholine-4-carboxylate (405 mg, 43.6 %) as a brown oil.
LCMS (ESI) calcd. for C12H22N2O4 [M+H] + m/z 259.2, found: 259.5.
Step 2: (S) -N, N-dimethylmorpholine-2-carboxamide
To a stirred solution of tert-butyl (S) -2- (dimethylcarbamoyl) morpholine-4-carboxylate (405 mg, 1.57 mmol) in DCM (6 mL) was TFA (2 mL) at rt , the resulting mixture was stirred at rt for 2 h. The reaction was monitored by LCMS. After completion, the reaction mixture was concentrated to give the crude (S) -N, N-dimethylmorpholine-2-carboxamide (880 mg) as a brown oil of its TFA salt.
LCMS (ESI) calcd. for C7H14N2O2 [M+H] + m/z 159.1, found: 159.3.
Step 3: (64S, 4S, Z) -4-amino-10, 10-dimethyl-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza -2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione
To a stirred solution of tert-butyl ( (64S, 4S, Z) -10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (4.3 g, 6.10 mmol) in DCM (40 mL) was added TFA (5 mL) at rt, the resulting mixture was stirred at rt for 1 h. The reaction was monitored by LCMS. After completion, the reaction mixture was diluted with DCM (50 mL) , washed with saturated NaHCO3 (100 x 2 mL) , then dried over Na2SO4, following with concentration under reduced pressure to obtain (64S, 4S, Z) -4-amino-10, 10-dimethyl-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione (3.4 g, 91.9 %) as a brown foam.
LCMS (ESI) calcd. for C31H40BN5O5S [M+H] + m/z 606.3, found: 607.0.
Step 4: (1r, 2R, 3S) -N- ( (64S, 4S, Z) -10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
To a stirred solution of (64S, 4S, Z) -4-amino-10, 10-dimethyl-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione (2.1 g, 3.42 mmol) and (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxylic acid (390 mg, 3.42 mmol) and NMI (702 mg, 8.55 mmol) in ACN (20 mL) and DCM (20 mL) was added TCFH (1.4 g, 5.13 mmol) at rt, the resulting mix-ture was stirred at rt for 1 h. The reaction mixture was concentrated to give the crude product. The crude product was purified by column chromatography (eluting with EtOAc/PE from 0%to 65%) to give (1r, 2R, 3S) -N- ( (64S, 4S, Z) -10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide (2.2 g, 93.4%) as a pale yellow foam.
LCMS (ESI) calcd. for C37H48BN5O6S [M+H] + m/z 702.3, found: 701.7.
Step 5: (1r, 2R, 3S) -N- ( (64S, 4S, Z) -12- (2- ( (S) -1-methoxyethyl) -5- (3- ( (tetrahydro-2H-pyran-2-yl) oxy) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
A mixture of 3-bromo-2- ( (S) -1-methoxyethyl) -5- (3- ( (tetrahydro-2H-pyran-2-yl) oxy) prop-1-yn-1-yl) pyridine (1.0 g, 2.83 mmol) , (1r, 2R, 3S) -N- ( (64S, 4S, Z) -10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide (2.1 g, 2.97 mmol) , K2CO3 (1.2 g, 8.50 mmol) and Pd (dppf) Cl2 (311 mg, 0.42 mmol) in dioxane (30 mL) and H2O (7.5 mL) was stirred at 85℃ under N2 for 6 h. After comple-tion, the reaction mixture was concentrated to give the crude product which was purified by silica gel col-umn (eluting with MeOH/DCM from 0%to 10%) to give (1r, 2R, 3S) -N- ( (64S, 4S, Z) -12- (2- ( (S) -1-methoxyethyl) -5- (3- ( (tetrahydro-2H-pyran-2-yl) oxy) prop-1-yn-1-yl) pyri-din-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1 .1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide (1.1 g, 46.7 %) as a yellow foam.
LCMS (ESI) calcd. for C47H56N6O7S [M+H] + m/z 849.4, found: 849.9.
Step 6: (1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- ( (tetrahydro-2H-pyran-2-yl) oxy) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
To a stirred solution of (1r, 2R, 3S) -N- ( (64S, 4S, Z) -12- (2- ( (S) -1-methoxyethyl) -5- (3- ( (tetrahydro-2H-pyran-2-yl) oxy) prop-1-yn-1-yl) pyri-din-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide (1.1 g, 1.30 mmol) in DMF (11 mL) were added Cs2CO3 (844 mg, 2.59 mmol) and Iodoethane (405 mg, 2.59 mmol) at 0℃, the result-ing mixture was stirred at rt for 2 h. The reaction was monitored by LCMS. After completion, the reaction mixture was diluted with EtOAc (25 mL) , washed with water (50 mL x 3) and saturated NaCl (50 x 2 mL) , then dried over Na2SO4, following with concentration under reduced pressure to obtain crude product which was purified by pre-HPLC (eluting with CH3CN/H2O (0.1%NH4HCO3) from 10%to 95%) to give (1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- ( (tetrahydro-2H-pyran-2-yl) oxy) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide as atropisomer 1 (171 mg) and atropisomer 2 (168 mg) (Yield: 29.8%) as a white solid.
LCMS (ESI) calcd. for C49H60N6O7S [M+H] + m/z 877.4, found: 877.9
Step 7: (1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (5- (3-hydroxyprop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
To a stirred solution of (1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- ( (tetrahydro-2H-pyran-2-yl) oxy) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide as atropisomer 1 (171 mg, 0.20 mmol) in MeOH (9 mL) was added TsOH (329 mg) at rt, the resulting mixture was stirred at rt for 16 h. The reaction mixture was concentrated and purified by silica gel column (eluting with EtOAc/PE from 0%to 100%) to give crude (1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (5- (3-hydroxyprop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide (183 mg) as a yellow foam.
LCMS (ESI) calcd. for C44H52N6O6S [M+H] + m/z 793.4, found: 793.9
Step 8: 3- (5- ( (64S, 4S, Z) -4- ( (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxamido) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl methanesulfonate
To a stirred solution of crude (1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (5- (3-hydroxyprop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -1 0, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide (183 mg, 0.23 mmol) in DCM (7.5 mL) were added DIEA (298 mg, 2.30 mmol) and Ms2O (241 mg, 1.38 mmol) at rt, the resulting mixture was stirred at rt for 1 h. The reaction mixture was diluted with DCM (15 mL) and washed with H2O (20 mL) and brine (20 mL) . The organic layer was separated and concentrated to give crude 3- (5- ( (64S, 4S, Z) -4- ( (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxamido) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl methanesulfonate (132 mg, Yield: 65.7%) as a pale yellow foam.
LCMS (ESI) calcd. for C45H54N6O8S2 [M+H] + m/z 871.3, found: 871.9
Step 9: (2S) -4- (3- (5- ( (64S, 4S, Z) -4- ( (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxamido) -11-ethyl-10, 10-dimethyl -5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundeca phane-12-yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) -N, N-dimethylmorpholine-2-carboxamide
To a stirred solution of (S) -N, N-dimethylmorpholine-2-carboxamide (132 mg, 0.23 mmol) in DCM (10 mL) were added DIEA (391 mg, 3.02 mmol) and the crude 3- (5- ( (64S, 4S, Z) -4- ( (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxamido) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl methanesulfonate (263 mg, 0.30 mmol) at rt, the resulting mixture was stirred at 40℃ for 16 h. The reaction mixture was concentrated to give the crude product. The crude product was purified by pre-HPLC (eluting with CH3CN/H2O (0.1%NH4HCO3) from 10%to 80%) to give (2S) -4- (3- (5- ( (64S, 4S, Z) -4- ( (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxamido) -11-ethyl-10, 10-dimethyl -5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) -N, N-dimethylmorpholine-2-carboxamide (18 mg, Yield: 64.3%) as a white solid.
LCMS (ESI) calcd. for C51H64N8O7S [M+H] + m/z 933.5, found: 934.0.
1H NMR (400 MHz, DMSO-d6) δ 8.80 (d, J = 1.9 Hz, 1H) , 8.41 (d, J = 9.0 Hz, 2H) , 7.90 -7.80 (m, 2H) , 7.75 (d, J = 8.5 Hz, 1H) , 7.58 (d, J = 8.6 Hz, 1H) , 5.94 (d, J = 11.1 Hz, 1H) , 5.38 (s, 1H) , 4.66 (d, J =10.9 Hz, 1H) , 4.48 (d, J = 5.0 Hz, 1H) , 4.36 -4.22 (m, 3H) , 4.07 (dd, J = 15.0, 7.5 Hz, 1H) , 3.87 (d, J =10.1 Hz, 1H) , 3.67 -3.49 (m, 4H) , 3.23 (s, 3H) , 3.19 -3.07 (m, 1H) , 3.01 (s, 3H) , 2.94 (d, J = 14.3 Hz, 1H) , 2.86 -2.71 (m, 4H) , 2.63 (d, J = 5.8 Hz, 1H) , 2.43 (dd, J = 20.4, 9.5 Hz, 3H) , 2.34 (dd, J = 11.2, 8.0 Hz, 2H) , 2.20 -2.10 (m, 1H) , 1.57 (s, 1H) , 1.36 (d, J = 6.0 Hz, 3H) , 1.28 -1.20 (m, 1H) , 1.17 (d, J = 4.1 Hz, 2H) , 1.08 (dd, J = 10.4, 5.7 Hz, 5H) , 0.87 (d, J = 7.1 Hz, 5H) , 0.33 (s, 3H) .
Example 33
(2S) -2- (7- ( (R) -aziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -3-methylbutanamide
Step 1: Synthesis of methyl trityl-D-serinate
To a mixture of methyl D-serinate hydrochloride (5.00 g, 32.14 mmol) and TEA (8.13 g, 80.34 mmol) in DCE (70 mL) was added TrtCl (8.96 g, 32.14 mmol) at room temperature, the mixture was stirred at 50℃under N2 atmosphere overnight. The reaction was monitored by LCMS, after completion, the mixture was concentrated and the residue was purified by silica gel column chromatography (eluting with EtOAc/PE, from 0%to 6%in 20 min) to obtain methyl trityl-D-serinate (11.20 g, 30.99 mmol, yield: 96.4 %) as a light yellow solid.
LCMS (ESI) calcd. for C23H23NO3 [M+Na] + m/z 384.2, found 384.5.
Step 2: Synthesis of methyl (R) -1-tritylaziridine-2-carboxylate
To a mixture of (1.00 g, 2.77 mmol) and TEA (0.84 g, 8.30 mmol) in DCE (10 mL) was added MsCl (0.38 g, 3.32 mmol) at 0℃, the mixture was stirred at r.t. for 4h and at 80℃ for 16 h under N2 atmosphere, the reaction was monitored by LCMS. After completion, the mixture was diluted with water (40 mL) and extracted with DCM (20 mL x 3) , the combined organic layer was washed with brine (40 mL) , dried over Na2SO4, concentrated and purified by flash chromatography column (eluting with EtOAc/PE from 0%to 2%in 10 min) to obtain methyl (R) -1-tritylaziridine-2-carboxylate (760 mg, 2.21 mmol, yield: 79.8 %) as a white solid.
LCMS (ESI) calcd. for C23H21NO2 [M+H] + m/z 344.2, found 344.6.
Step 3: Synthesis of (R) -1-tritylaziridine-2-carboxylic acid
Methyl (R) -1-tritylaziridine-2-carboxylate (500 mg, 1.46 mmol) was dissolved in a mixture of THF (15 mL) and H2O (2 mL) at 0℃, 1M NaOH (20 mL) was added, followed by 1M LiOH (10 mL) , the mix-ture was stirred for 2h at 0℃. The reaction was monitored by LCMS, after completion, the mixture was diluted with DCM (50 mL) and 15%aqueous citric acid (40 mL) , the organic phase was further washed with 15%citric acid solution (3 x 20 mL) and the combined aqueous phases were extracted with DCM (3 x 20 mL) , the combined organic phase was concentrated and the residue was purified by reverse phase col-umn (eluting with MeCN/H2O, from 5%to 34%in 35 min) to give (R) -1-tritylaziridine-2-carboxylic acid (380 mg, 1.15 mmol, yield: 78.8%) as a white solid.
LCMS (ESI) calcd. for C22H19NO2 [M-H] -m/z 328.1, found: 328.5.
Step 4: Synthesis of benzyl (R) -2-hydroxy-3-methylbutanoate
To a mixture of BnOH (3.84 g, 35.55 mmol) in toluene (50 mL) was added TsOH (0.58g, 3.39 mmol) at r.t., the mixture was stirred at 80℃ for 1h and then (R) -2-hydroxy-3-methylbutanoic acid (4.00 g, 33.86 mmol) was added, the reaction was stirred at 80℃ for 1 h under N2 atmosphere, the mixture was monitored by LCMS. After completion, the mixture was diluted with water (100 mL) and extracted with EA (50 mL x 3) .The combined organic phase was concentrated under reduced pressure and the the residue was purified by silica gel column chromatography (eluting with EtOAc/PE, from 0%to 5%in 20 min) to afford benzyl (R) -2-hydroxy-3-methylbutanoate (5.21 g, 25.02 mmol, yield: 73.9%) as a colorless oil.
LCMS (ESI) calcd. for C12H16O3 [M+H] + m/z 209.1, found: 209.3.
Step 5: Synthesis of benzyl (R) -3-methyl-2- ( ( (trifluoromethyl) sulfonyl) oxy) butanoate
To a mixture of benzyl (R) -2-hydroxy-3-methylbutanoate (5.21 g, 25.02 mmol) and 2, 6-lutidine (2.95g, 27.52 mmol) in DCM (100 mL) was added Tf2O (7.41 g, 26.27 mmol) slowly at 0℃, the mixture was stirred at 0℃ for 2 h under N2 atmosphere, the reaction was monitored by LCMS. After completion, the mixture was diluted with water (100 mL) and extracted with DCM (50 mL x 3) , the combined organic layer was washed with brine (100 mL) , dried over Na2SO4 and concentrated, the residue was purified by flash chromatography column (eluting with EtOAc/PE, from 0%to 2%in 10 min) to obtain benzyl (R) -3-methyl-2- ( ( (trifluoromethyl) sulfonyl) oxy) butanoate (6.70 g, 19.69 mmol, yield: 78.7 %) as a color-less oil.
LCMS (ESI) calcd. for C13H15F3O5S [M+NH4] + m/z 358.1, found: 358.4.
Step 6: Synthesis of tert-butyl 7- ( (S) -1- (benzyloxy) -3-methyl-1-oxobutan-2-yl) -2, 7-diazaspiro [4.4] nonane-2-carboxylate
A mixture of benzyl (R) -3-methyl-2- ( ( (trifluoromethyl) sulfonyl) oxy) butanoate (376 mg, 1.10 mmol) and tert-butyl 2, 7-diazaspiro [4.4] nonane-2-carboxylate (250 mg, 1.10 mmol) in THF (10 mL) was stirred at r.t. for 2h, the reaction was monitored by LCMS. After completion, the mixture was diluted with EtOAc (20) and water (40 mL) , then extracted with EtOAc (20 mL x 3) , the combined organic layer was washed with brine (40 mL) , dried over Na2SO4 and concentrated , the residual was purified by silica gel column chro-matography (eluting with EtOAc/PE, from 0%to 10%in 15 min) to obtain tert-butyl 7- ( (S) -1- (benzyloxy) -3-methyl-1-oxobutan-2-yl) -2, 7-diazaspiro [4.4] nonane-2-carboxylate (200 mg, 0.48 mmol, yield: 43.6%) as a colorless oil.
LCMS (ESI) calcd. for C24H36N2O4 [M+H] + m/z 417.3, found: 418.2.
Step 7: Synthesis of benzyl (2S) -3-methyl-2- (2, 7-diazaspiro [4.4] nonan-2-yl) butanoate hydrochloride
A mixture of tert-butyl 7- ( (S) -1- (benzyloxy) -3-methyl-1-oxobutan-2-yl) -2, 7-diazaspiro [4.4] nonane-2-carboxylate (200 mg, 0.48 mmol) in 4M HCl in dioxane (3 mL) was stirred at r.t. for 1h under N2 atmosphere, the reaction was moni-tored by LCMS. After completion, the mixture was concentrated to afford benzyl (2S) -3-methyl-2- (2, 7-diazaspiro [4.4] nonan-2-yl) butanoate hydrochloride (169 mg, 0.48 mmol, yield: 100.0%) as a white solid.
LCMS (ESI) calcd. for C19H28N2O2 [M+H] + m/z 317.2, found: 317.6.
Step 8: Synthesis of benzyl (2S) -3-methyl-2- (7- ( (R) -1-tritylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) butanoate
To a mixture of benzyl (2S) -3-methyl-2- (2, 7-diazaspiro [4.4] nonan-2-yl) butanoate hydrochloride (169 mg, 0.48 mmol) , DIEA (619 mg, 4.79 mmol) and (R) -1-tritylaziridine-2-carboxylic acid (175 mg, 0.48 mmol) in ACN (5 mL) was added HATU (273 mg, 0.72mmol) , the mixture was stirred at r.t. for 3 h under N2 atmosphere, the reaction was monitored by LCMS. After completion, the mixture was purified by reverse phase column (eluting with MeCN/H2O, from 5%to 88%in 35 min) to afford benzyl (2S) -3-methyl-2- (7- ( (R) -1-tritylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) butanoate (210 mg, 0.33 mmol, yield: 69.6%) as a light yellow solid.
LCMS (ESI) calcd. for C41H45N3O3 [M+H] + m/z 628.4, found: 629.6.
Step 9: Synthesis of (2S) -3-methyl-2- (7- ( (R) -1-tritylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) butanoic acid
A mixture of benzyl (2S) -3-methyl-2- (7- ( (R) -1-tritylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) butanoate (210 mg, 0.33 mmol) and 10%Pd/C (40 mg) in MeOH (5 mL) was stirred at room temperature for 16 h under H2 atmosphere, the reaction was monitored by LCMS. After completion, the mixture was filtered and the fil-trate was concentrated to afford (2S) -3-methyl-2- (7- ( (R) -1-tritylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) butanoic acid (120 mg, 0.22 mmol, yield: 66.7 %) as a white solid.
LCMS (ESI) calcd. for C34H39N3O3 [M+H] + m/z 538.3, found: 538.7.
Step 10: Synthesis of (2S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dime-thyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -3-methyl-2- (7- ( (R) -1-tritylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) butanamide
To a solution of (2S) -3-methyl-2- (7- ( (R) -1-tritylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) butanoic acid (16 mg, 0.03 mmol) in DMF (1 mL) were added HATU (16 mg, 0.04 mmol) and DIEA (19 mg, 0.14 mmol) at 0-10℃. The resulting mixture was stirred at RT for 10 min. Then (64S, 4S, Z) -4-amino-11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundeca phane-5, 7-dione (22 mg, 0.03 mmol) was added into the reaction mixture, the resulting mixture was stirred at rt for 2 h. The reaction was monitored by LCMS. After completion, the reaction mixture was diluted with brine and extracted with EtOAc (2 x 30 mL) , the combined organic phase was washed with saturated NaCl (50 mL) , then dried over Na2SO4, following with concentration under reduced pressure to obtain crude product which was purified by silica gel column eluting with EtOAc/PE from 0%to 100%to af-ford (2S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 1 0-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundeca-phane-4-yl) -3-methyl-2- (7- ( (R) -1-tritylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) butanamide (27 mg, 73 %) as a yellow solid.
LCMS (ESI) calcd. for C76H88N10O7S [M+H] + m/z 1285.7.
Step 11: Synthesis of (2S) -2- (7- ( (R) -aziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyeth-yl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -3-methylbutanamide
To a stirred solution of (2S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundeca-phane-4-yl) -3-methyl-2- (7- ( (R) -1-tritylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) butanamide (27 mg, 0.02 mmol) in dichloromethane (2 mL) was added TFA (0.5 mL) dropwisely at rt, the resulting mixture was stirred at rt for 1 h. The reaction mixture was washed with sat. NaHCO3 (10 mL) , brine (10 mL) , then dried over Na2SO4, following with concentration under reduced pressure, the residue was puri-fied by pre-HPLC (eluting with CH3CN/H2O (0.1%NH4HCO3) from 20%to 70%) to give (2S) -2- (7- ( (R) -aziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyeth-yl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -3-methylbutanamide (1.4 mg, 6.4 %) as a white solid.
LCMS (ESI) calcd. for C57H74N10O7S [M+H] + m/z 1043.6, found: 1045.2.
1H NMR (400 MHz, DMSO-d6) δ 8.78 (d, J = 1.9 Hz, 1H) , 8.40 (s, 1H) , 8.22 (m, 1H) , 7.83 (d, J =9.8 Hz, 2H) , 7.74 (d, J = 8.4 Hz, 1H) , 7.57 (d, J = 8.7 Hz, 1H) , 5.98 -5.88 (m, 1H) , 5.33 (s, 1H) , 4.73 (d, J = 10.4 Hz, 1H) , 4.49 (s, 1H) , 4.37 -4.21 (m, 2H) , 4.07 (m, 1H) , 3.75 (m, 1H) , 3.67 -3.45 (m, 7H) , 3.42 -3.30 (m, 8H) , 3.27 -3.15 (m, 4H) , 2.96 (d, J = 14.2 Hz, 1H) , 2.78 (m, 3H) , 2.61 (m, 3H) , 2.54 -2.50 (m, 3H) , 2.47 -2.43 (m, 2H) , 2.34 (dd, J = 20.4, 9.5 Hz, 2H) , 2.18 -2.09 (m, 1H) , 2.03 -1.54 (m, 7H) , 1.34 (d, J = 6.0 Hz, 3H) , 1.24 -1.12 (m, 2H) , 1.04 -0.74 (m, 9H) , 0.28 (s, 3H) .
Example 34
(2S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (22S, 64S, 4S) -11-ethyl-12- (5-ethynyl-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -3-methylbutanamide
Substituting N- (1- (tert-butoxycarbonyl) -4-fluoropiperidine-4-carbonyl) -N-methyl-L-valine with (S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -3-meth ylbutanoic acid in the Step 3 of Example 26, the title compound was prepared by the same procedures as described for Example 26.
Example 35
(2S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (22S, 64S, 4S) -12- (5-ethynyl-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11- (2, 2, 2-trifluoro-ethyl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -3-methylbutanamide
Substituting bromoethane with 2-bromo-1, 1, 1-trifluoroethane in the Step of Example 34, the title compound was prepared by the same procedures as described for Example 34.
Example 36
(2S) -2-cyclopentyl-2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (22S, 64S, 4S) -12- (5-ethynyl-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11- (2, 2, 2-trifluoroethyl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) acetamide
Substituting (S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -3-meth ylbutanoic acid with (S) -2-cyclopentyl-2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) acetic acid in the Step 3 of Example 34, the title compound was prepared by the same procedures as described for Example 34.
Example 37
(2S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (22S, 64S, 4S) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dime-thyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -3-methylbutanamide
Substituting triethyl (ethynyl) silane with 4- (prop-2-yn-1-yl) morpholine in the Step 1 and substituting N- (1- (tert-butoxycarbonyl) -4-fluoropiperidine-4-carbonyl) -N-methyl-L-valine with (S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -3-meth ylbutanoic acid in the Step 3 of Example 26, the title compound was prepared by the same procedures as described for Example 26.
Example 38
(2S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (22S, 64S, 4S) -12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11- (2, 2, 2-trifluoroethyl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -3-methylbutanamide
Substituting bromoethane with 2-bromo-1, 1, 1-trifluoroethane in the Step of Example 37, the title com-pound was prepared by the same procedures as described for Example 37. Example 39
1-acryloyl-N- ( (2S) -1- ( ( (63S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyri-din-3-yl) -10, 10-dimethyl-5, 7-dioxo-61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (1, 3) -pyridazinacycloundecaphane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) -4-fluoro-N-methylpiperidine-4-carboxamide
Step 1: Synthesis of benzyl N- (tert-butoxycarbonyl) -N-methyl-L-valinate
To a stirred solution of (S) -2, 3-bis (tert-butoxycarbonyl) -2, 3-diazabicyclo [3.1.1] heptane-4-carboxylic acid (2.31 g, 10.0 mmol) in DMF (20 mL) was added K2CO3 (2.72 g, 20.0 mmol) and (bromome-thyl) benzene (2.05 g, 12.0 mmol) at r.t.. The reaction mixture was stirred at r.t. for 3 h. After completion, the reaction mixture was diluted with brine and extracted with EtOAc (2 x 50 mL) . The organic layers were combined, washed with saturated NaCl (2 x 50 mL) , dried over anhydrous Na2SO4 and concentrated to give the crude product which was purified by silica gel column (eluting with EA/PE from 0%to 10%) to give benzyl N- (tert-butoxycarbonyl) -N-methyl-L-valinate (2) (2.9 g, 90%) as a colorless oil.
LCMS (ESI) calcd. for C18H27NO4 [M-H] + m/z 321.2, found: 322.6.
Step 2: Synthesis of benzyl methyl-L-valinate HCl salt
To a stirred solution of benzyl N- (tert-butoxycarbonyl) -N-methyl-L-valinate (2.90 g, 9.03 mmol) in DCM (10 mL) was added HCl/dioxane (15 mL, 4M) , the resulting mixture was stirred at rt for 2 h. The reaction mixture was concentrated to give the crude product benzyl methyl-L-valinate HCl salt (2.3 g, 99%) as a white solid.
LCMS (ESI) calcd. for C13H19NO2 [M+H] + m/z 221.1, found: 222.9.
Step 3: Synthesis of tert-butyl (S) -4- ( (1- (benzyloxy) -3-methyl-1-oxobutan-2-yl) (methyl) carbamoyl) -4-fluoropiperidine-1-carboxylate
To a solution of benzyl methyl-L-valinate HCl salt (362 mg, 1.41 mmol) in DMF (3 mL) was added 1- (tert-butoxycarbonyl) -4-fluoropiperidine-4-carboxylic acid (347 mg, 1.41 mmol) , HATU (770 mg, 2.03 mmol) and DIEA (404 mg, 4.00 mmol) at rt. The resulting mixture was stirred atrt overnight. After com-pletion, the reaction mixture was diluted with brine and extracted with EtOAc (2 x 10 mL) , the combined organic phase was washed with saturated NaCl (10 mL) , then dried over Na2SO4, following with concen-tration under reduced pressure to obtain crude product which was purified by silica gel column eluting with EtOAc/PE from 0%to 35%to afford tert-butyl (S) -4- ( (1- (benzyloxy) -3-methyl-1-oxobutan-2-yl) (methyl) carbamoyl) -4-fluoropiperidine-1-carboxylate (300 mg, 47 %) as a colorless oil.
LCMS (ESI) calcd. for C24H35FN2O5 [M+H] + m/z 450.3, found: 396.1.
Step 4: Synthesis of N- (1- (tert-butoxycarbonyl) -4-fluoropiperidine-4-carbonyl) -N-methyl-L-valine
To a solution of tert-butyl (S) -4- ( (1- (benzyloxy) -3-methyl-1-oxobutan-2-yl) (methyl) carbamoyl) -4-fluoropiperidine-1-carboxylate (300 mg, 0.667 mmol) in CH3OH (10 mL) was added Pd/C (60 mg, 10%) , the resulting mixture was stirred at rt for 3 hours under H2 atmosphere (1 atm) . The reaction was monitored by LCMS. After completion, the mixture was filtered and concentrated to give the N- (1- (tert-butoxycarbonyl) -4-fluoropiperidine-4-carbonyl) -N-methyl-L-valine (230 mg, 96%) as a color-less oil.
LCMS (ESI) calcd. for C17H29FN2O5 [M+H] + m/z 360.2, found: 361.6.
Step 5: Synthesis of tert-butyl 4- ( ( (2S) -1- ( ( (63S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (1, 3) -pyridazinacycloundeca-phane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) (methyl) carbamoyl) -4-fluoropiperidine-1-carboxylate
To a stirred solution of (63S, 4S, Z) -4-amino-11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (1, 3) -pyridazinacycloun decaphane-5, 7-dione (45 mg, 0.06 mmol) and N- (1- (tert-butoxycarbonyl) -4-fluoropiperidine-4-carbonyl) -N-methyl-L-valine (18 mg, 0.072 mmol) and 1-methyl-1H-imidazole (9 mg, 0.12 mmol) in ACN (2 mL) was added TCFH (21 mg, 0.078 mmol) at rt, the resulting mixture was stirred at rt for 1 h. The reaction mixture was concentrated to give the crude product. The crude product was purified by pre-HPLC (ACN/H2O (0.5 %NH4HCO3) from 20%to 95%in 30 min) to give tert-butyl 4- ( ( (2S) -1- ( ( (63S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (1, 3) -pyridazinacycloundeca-phane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) (methyl) carbamoyl) -4-fluoropiperidine-1-carboxylate (20 mg, 30%) as a white solid.
LCMS (ESI) calcd. for C58H78FN9O9S [M+H] + m/z 1095.6, found: 1097.1.
Step 6: Synthesis of N- ( (2S) -1- ( ( (63S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10 , 10-dimethyl-5, 7-dioxo-61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (1, 3) -pyridazinacycloundecaphane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) -4-fluoro-N-methylpiperidine-4-carboxamide TFA salt
To a stirred solution of tert-butyl 4- ( ( (2S) -1- ( ( (63S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (1, 3) -pyridazinacycloundeca-phane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) (methyl) carbamoyl) -4-fluoropiperidine-1-carboxylate (20 mg, 0.018 mmol) in dichloromethane (2 mL) was added TFA (1 mL) dropwisely at rt, the resulting mixture was stirred at rt for 1 h. The reaction mixture was concentrated and lyophilized to give N- ( (2S) -1- ( ( (63S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (1, 3) -pyridazinacycloundeca-phane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) -4-fluoro-N-methylpiperidine-4-carboxamide (21 mg, 100%) of its TFA salt as a yellow solid.
LCMS (ESI) calcd. for C53H70FN9O7S [M+H] + m/z 995.5, found: 997.1.
Step 7: Synthesis of 1-acryloyl-N- ( (2S) -1- ( ( (63S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyri din-3-yl) -10, 10-dimethyl-5, 7-dioxo-61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -thiazola-1 (5, 3) -indola-6(1, 3) -pyridazinacycloundecaphane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) -4-fluoro-N-methylpiperidine-4 -carboxamide
To a stirred solution of N- ( (2S) -1- ( ( (63S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (1, 3) -pyridazinacycloundeca-phane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) -4-fluoro-N-methylpiperidine-4-carboxamide TFA salt (21 mg, 0.019 mmol) in DCM (2 mL) was added DIEA (10 mg, 0.076 mmol) and acryloyl chloride (4 mg, 0.038 mmol) at rt, the resulting mixture was stirred at rt overnight. The reaction mixture was concentrated to give the crude product. The crude product was purified by pre-HPLC (ACN/H2O (0.5 %NH4HCO3) from 20%to 95%in 30 min) to give 1-acryloyl-N- ( (2S) -1- ( ( (63S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyrid-in-3-yl) -10, 10-dimethyl-5, 7-dioxo-61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (1, 3) -pyridazinacycloundecaphane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) -4-fluoro-N-methylpiperidine-4-carboxamide (6 mg, 30%) as a white solid.
LCMS (ESI) calcd. for C56H72FN9O8S [M+H] + m/z 1049.5, found: 1051.1.
1H NMR (400 MHz, DMSO-d6) δ 8.79 (d, J = 1.5 Hz, 1H) , 8.48 -8.42 (m, 2H) , 7.85 -7.78 (m, 2H) , 7.74 (d, J = 7.7 Hz, 1H) , 7.56 (d, J = 8.6 Hz, 1H) , 6.89-6.80 (m, 1H) , 6.15-6.10 (m, 1H) , 5.69 (d, J = 11.2 Hz, 1H) , 5.46-5.41 (m, 1H) , 5.09 -5.02 (m, 1H) , 4.70 (d, J = 11.5 Hz, 1H) , 4.38 -4.15 (m, 6H) , 4.12-3.94 (m, 3H) , 3.60-3.54 (m, 8H) , 3.24 (s, 3H) , 3.07 (d, J = 5.3 Hz, 4H) , 2.99 -2.92 (m, 2H) , 2.89-2.86 (m, 1H) , 2.79-2.70 (m, 1H) , 2.42-2.33 (m, 3H) , 2.23-1.92 (m, 6H) , 1.82-1.73 (m, 2H) , 1.56-1.43 (m, 2H) , 1.33 (d, J =6.0 Hz, 3H) , 1.01-1.98 (m, 1H) , 0.94 -0.81 (m, 9H) , 0.76 (d, J = 6.4 Hz, 3H) , 0.31 (s, 3H) .
Example 40
(2S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (22S, 64S, 4S) -12- (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11- (2, 2, 2-trifluoroethyl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -3-methylbutanamide
Substituting 4- (prop-2-yn-1-yl) morpholine with 4- (prop-2-yn-1-yl) thiomorpholine 1, 1-dioxide in the Step 1 of Example 37, the title compound was prepared by the same procedures as described for Example 37.
Example 41
(2S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (22S, 64S, 4S) -12- (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -3-methylbutanamide
Substituting 4- (prop-2-yn-1-yl) morpholine with 4- (prop-2-yn-1-yl) thiomorpholine 1, 1-dioxide in the Step 1 of Example 38, the title compound was prepared by the same procedures as described for Example 38.
Example 42
(2S) -2-cyclopentyl-2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (22S, 64S, 4S) -12- (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyeth yl) pyridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) acetamide
Substituting (S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -3-meth ylbutanoic acid with (S) -2-cyclopentyl-2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) acetic acid in the Step 3 of Example 41, the title compound was prepared by the same procedures as described for Example 41.
Example 43
(2S) -2-cyclopentyl-2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (22S, 64S, 4S) -12- (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyeth yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11- (2, 2, 2-trifluoroethyl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) acetamide
Substituting bromoethane with 2-bromo-1, 1, 1-trifluoroethane in the Step of Example 42, the title compound was prepared by the same procedures as described for Example 42.
Example 44
(2S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N - ( (22S, 64S, 4S) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- (4- (methylsulfonyl) piperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -3-methylbutanamide
Substituting 4- (prop-2-yn-1-yl) morpholine with 1- (methylsulfonyl) -4- (prop-2-yn-1-yl) piperazine in the Step 1 of Example 37, the title compound was prepared by the same procedures as described for Ex-ample 37.
Example 45
(2S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (22S, 64S, 4S) -12- (2- ( (S) -1-methoxyethyl) -5- (3- (4- (methylsulfonyl) piperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11- (2, 2, 2-trifluoroethyl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -3-methylbutanamide
Substituting bromoethane with 2-bromo-1, 1, 1-trifluoroethane in the Step of Example 44, the title compound was prepared by the same procedures as described for Example 44.
Example 46
(2S) -2-cyclopentyl-2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (22S, 64S, 4S) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- (4- (methylsulfonyl) piperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) acetamide
Substituting (S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -3-meth ylbutanoic acid with (S) -2-cyclopentyl-2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) acetic acid in the Step 3 of Example 44, the title compound was prepared by the same procedures as described for Example 44.
Example 47
(2S) -2-cyclopentyl-2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (22S, 64S, 4S) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- (4- (methylsulfonyl) piperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) acetamide
Substituting bromoethane with 2-bromo-1, 1, 1-trifluoroethane in the Step of Example 46, the title compound was prepared by the same procedures as described for Example 46.
Example 48
(2S) -4- (3- (5- ( (22S, 64S, 4S) -4- ( (S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspi-ro [4.4] nonan-2-yl) -3-methylbutanamido) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) -N, N-dimethylmorpholine-2-carboxamide
Substituting 4- (prop-2-yn-1-yl) morpholine with (S) -N, N-dimethyl-4- (prop-2-yn-1-yl) morpholine-2-carboxamide in the Step 1 of Example 37, the title compound was prepared by the same procedures as described for Example 37.
Example 49
(2S) -2-cyclopentyl-2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (22S, 64S, 4S) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- (4- (methylsulfonyl) piperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) acetamide
Substituting bromoethane with 2-bromo-1, 1, 1-trifluoroethane in the Step of Example 48, the title compound was prepared by the same procedures as described for Example 48.
Example 50
(2S) -4- (3- (5- ( (22S, 64S, 4S) -4- ( (S) -2-cyclopentyl-2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbon-yl) -2, 7-diazaspiro [4.4] nonan-2-yl) acetamido) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza -2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) -N, N-dimethylmorpholine-2-carboxamide
Substituting (S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -3-meth ylbutanoic acid with (S) -2-cyclopentyl-2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) acetic acid in the Step 3 of Example 48, the title compound was prepared by the same procedures as described for Example 48.
Example 51
(2S) -4- (3- (5- ( (22S, 64S, 4S) -4- ( (S) -2-cyclopentyl-2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbon-yl) -2, 7-diazaspiro [4.4] nonan-2-yl) acetamido) -10, 10-dimethyl-5, 7-dioxo-11- (2, 2, 2-trifluoroethyl) -11H-8 -oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) -N, N-dimethylmorpholine-2-carboxamide
Substituting bromoethane with 2-bromo-1, 1, 1-trifluoroethane in the Step of Example 50, the title compound was prepared by the same procedures as described for Example 50.
Example 52
1-acryloyl-N- ( (2S) -1- ( ( (63S, 4S, Z) -12- (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyeth-yl) pyridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (1, 3) -pyridazinacycloundecaphane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) -4-fluoro-N-methylpiperidine-4-carboxamide
Substituting 4- (prop-2-yn-1-yl) morpholine with 4- (prop-2-yn-1-yl) thiomorpholine 1, 1-dioxide in the Step of Example 39, the title compound was prepared by the same procedures as described for Example 39.
Example 53
1-acryloyl-N- ( (2S) -1- ( ( (64S, 4S, Z) -12- (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyeth-yl) pyridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola -6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) -4-fluoro-N-methylpiperidine-4-carboxamide
Substituting hexahydropyridazine-3-carboxylic acid with 2, 3-diazabicyclo [3.1.1] heptane-4-carboxylic acid in the Step of Example 52, the title compound was prepared by the same procedures as described for Example 52.
Example 54
1- (4- (dimethylamino) -4-methylpent-2-ynoyl) -N- ( (2S) -1- ( ( (63S, 4S, Z) -12- (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (1, 3) -pyridazinacycloundecaphane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) -4-fluoro-N-methylpiperidine-4-carboxamide
Substituting 4- (prop-2-yn-1-yl) morpholine with 4- (prop-2-yn-1-yl) thiomorpholine 1, 1-dioxide in the Step and substituting acrylic acid with 4- (dimethylamino) -4-methylpent-2-ynoic acid in the Step 7 of Ex-ample 39, the title compound was prepared by the same procedures as described for Example 39.
Example 55
1- (4- (dimethylamino) -4-methylpent-2-ynoyl) -N- ( (2S) -1- ( ( (64S, 4S, Z) -12- (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) -4-fluoro-N-methylpiperidine-4-carboxamide
Substituting 4- (prop-2-yn-1-yl) morpholine with 4- (prop-2-yn-1-yl) thiomorpholine 1, 1-dioxide in the Step 7 of Example 53, the title compound was prepared by the same procedures as described for Example 53.
Example 56
1- (4- (dimethylamino) -4-methylpent-2-ynoyl) -N- ( (2S) -1- ( ( (22S, 63S, 4S) -12- (5- (3- (1, 1-dioxidothiomorpholi-no) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (1, 3) -pyridazinacycloundecaphane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) -4-fluoro-N-methylpiperidine-4-carboxamide
Substituting tert-butyl ( (63S, 4S, Z) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (1, 3) -pyridazinacycloundecaphane-4-yl) carbamate with tert-butyl ( (22S, 63S, 4S) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (1, 3) -pyridazinacycloundecaphane-4-yl) c arbamate in the Step 1 of Example 54, the title compound was prepared by the same procedures as de-scribed for Example 54.
Example 57
1- (4- (dimethylamino) -4-methylpent-2-ynoyl) -N- ( (2S) -1- ( ( (64S, 4S, Z) -12- (5- (3- (1, 1-dioxidothiomorpholino) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) -4-fluoro-N-methylpiperidine-4-carboxamide
Substituting tert-butyl ( (64S, 4S, Z) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa -62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate with tert-butyl ( (22S, 64S, 4S) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carba mate in the Step 1 of Example 55, the title compound was prepared by the same procedures as described for Example 55.
Example 58
1- (4- (dimethylamino) -4-methylpent-2-ynoyl) -N- ( (2S) -1- ( ( (63S, 4S, Z) -11-ethyl-12- (5-ethynyl-2- ( (S) -1-methoxyeth-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -thiazola-1 (5, 3) -indo-la-6 (1, 3) -pyridazinacycloundecaphane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) -4-fluoro-N-methylpiperidine-4-carboxamide
Substituting tert-butyl ( (64S, 4S, Z) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate with tert-butyl ( (63S, 4S, Z) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (1, 3) -pyridazinacycloundecaphane-4-yl) carbamate in the Step 1 of Example 25, the title compound was prepared by the same procedures as described for Example 25.
Example 59
1- (4- (dimethylamino) -4-methylpent-2-ynoyl) -N- ( (2S) -1- ( ( (22S, 63S, 4S) -11-ethyl-12- (5-ethynyl-2- ( (S) -1-methoxyeth-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (1, 3) -pyridazinacycloundecaphane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) -4-fluoro-N-methylpiperidine-4-carboxamide
Substituting tert-butyl ( (64S, 4S, Z) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa -62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate with tert-butyl ( (22S, 63S, 4S) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (1, 3) -pyridazinacycloundecaphane-4-yl) carbamate in the Step 1 of Example 25, the title compound was prepared by the same procedures as de-scribed for Example 25.
Example 60
1-acryloyl-N- ( (2S) -1- ( ( (63S, 4S, Z) -11-ethyl-12- (5-ethynyl-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dime-thyl-5, 7-dioxo-61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (1, 3) -pyridazinacy cloundecaphane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) -4-fluoro-N-methylpiperidine-4-carboxamide
Substituting tert-butyl ( (64S, 4S, Z) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa -62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate with tert-butyl ( (63S, 4S, Z) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (1, 3) -pyridazinacycloundecaphane-4-yl) carbamate in the Step 1 and substituting acrylic acid with 4- (dimethylamino) -4-methylpent-2-ynoic acid in the Step 7 of Example 25, the title compound was prepared by the same procedures as described for Example 25.
Example 61
1-acryloyl-N- ( (2S) -1- ( ( (63S, 4S, Z) -11-ethyl-12- (5-ethynyl-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-di
me-thyl-5, 7-dioxo-61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (1, 3) -pyridazinacy cloundecaphane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) -4-fluoro-N-methylpiperidine-4-carboxamide
Substituting tert-butyl ( (63S, 4S, Z) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (1, 3) -pyridazinacycloundecaphane-4-yl) carbamate with tert-butyl ( (64S, 4S, Z) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa -62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate in the Step 1 of Example 60, the title compound was prepared by the same procedures as described for Example 60.
Example 62
1-acryloyl-N- ( (2S) -1- ( ( (63S, 4S) -11-ethyl-12- (5-ethynyl-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dime thyl-5, 7-dioxo-61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (1, 3) -pyridazinacycloundeca-phane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) -4-fluoro-N-methylpiperidine-4-carboxamide
Substituting tert-butyl ( (63S, 4S, Z) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (1, 3) -pyridazinacycloundecaphane-4-yl) carbamate with tert-butyl ( (22S, 63S, 4S) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (1, 3) -pyridazinacycloundecaphane-4-yl) c arbamate in the Step 1 of Example 60, the title compound was prepared by the same procedures as de-scribed for Example 60.
Example 63
1-acryloyl-N- ( (2S) -1- ( ( (64S, 4S) -11-ethyl-12- (5-ethynyl-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dime thyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) -4-fluoro-N-methylpiperidine-4-carboxamide
Substituting tert-butyl ( (63S, 4S, Z) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (1, 3) -pyridazinacycloundecaphane-4-yl) carbamate with tert-butyl ( (22S, 64S, 4S) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carba mate in the Step 1 of Example 60, the title compound was prepared by the same procedures as described for Example 60.
Example 64
(1r, 2R, 3S) -N- ( (64S, 4S, Z) -12- (5- (3- (1, 1-dioxidothiomorpholino) -4-hydroxybut-1-yn-1-yl) -2- ( (S) -1-methoxyeth-yl) pyridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6(2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
Substituting 4- (prop-2-yn-1-yl) morpholine with 4- (1-hydroxybut-3-yn-2-yl) thiomorpholine 1, 1-dioxide in the Step 1 of Example 16, the title compound was prepared by the same procedures as de-scribed for Example 16.
Example 65
(1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- (4- (oxetan-3-yl) piperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
Step 1: Synthesis of (S) -1- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) -4- (oxetan-3-yl) piperazine
To a stirred solution of (S) -3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl 4-methylbenzenesulfonate (300 mg, 0.71 mmol) in DCM (10 mL) was added 1- (oxetan-3-yl) piperazine (121 mg, 0.85 mmol) and DIEA (227 mg, 1.77 mmol) at r.t., the resulting mixture was stirred at r.t. for 8 h. The reaction was monitored by LCMS. After completion, the reaction mixture was diluted with DCM/MeOH (30 mL, V: V = 10: 1) , washed with water (10 mL x 3) and saturated NaCl (10 x 2 mL) , then dried over Na2SO4, following with concentration under reduced pressure to obtain crude product which was purified by silica gel column (eluting with MeOH/DCM from 0%to 7%) to give (S) -1- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) -4- (oxetan-3-yl) piperazine (227 mg, 81.6 %) as a yellow oil.
LCMS (ESI) calcd. for C18H24BrN3O2 [M+H] + m/z 394.1, found: 394.5.
Step 2: Synthesis of tert-butyl ( (64S, 4S, Z) -12- (2- ( (S) -1-methoxyethyl) -5- (3- (4- (oxetan-3-yl) piperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacy cloundecaphane-4-yl) carbamate
To a stirred solution of tert-butyl ( (64S, 4S, Z) -10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (200 mg, 0.28 mmol) , (S) -1- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) -4- (oxetan-3-yl) piperazine (111 mg, 0.28 mmol) and K2CO3 (117 mg, 0.85 mmol) in 1, 4-dioxane/H2O (7 mL, V: V= 6: 1 ) was added Pd (dppf) Cl2 (20 mg, 0.03 mmol) at r.t.. The reaction mixture was stirred at 85℃ under N2 for 8 h. After completion under reduced pressure to obtain crude product which was purified by rsilica gel column (elut-ing with MeOH/DCM from 0%to 5%) to give tert-butyl ( (64S, 4S, Z) -12- (2- ( (S) -1-methoxyethyl) -5- (3- (4- (oxetan-3-yl) piperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (240 mg, 95.1 %) as a brown solid.
LCMS (ESI) calcd. for C48H60N8O7S [M+H] + m/z 893.4, found: 893.9.
Step 3: Synthesis of tert-butyl ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- (4- (oxetan-3-yl) piperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] h eptanacycloundecaphane-4-yl) carbamate
To a stirred solution of tert-butyl ( (64S, 4S, Z) -12- (2- ( (S) -1-methoxyethyl) -5- (3- (4- (oxetan-3-yl) piperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (220 mg, 0.25 mmol) in DMF (5 mL) was added Cs2CO3 (241 mg, 0.74 mmol) and EtI (115 mg, 0.74 mmol) at 0℃., the resulting mixture was stirred at r.t. for 1 h. The reaction was monitored by LCMS. After completion, the reaction mixture was diluted with DCM/MeOH (11 mL, V:V = 10: 1) , washed with water (10 mL x 3) and saturated NaCl (10 x 2 mL) , then dried over Na2SO4, fol-lowing with concentration under reduced pressure to obtain crude product which was purified by rsilica gel column (eluting with MeOH/DCM from 0%to 5%) to give tert-butyl ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- (4- (oxetan-3-yl) piperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (60 mg, 26.4 %) as a white solid.
LCMS (ESI) calcd. for C50H64N8O7S [M+H] + m/z 921.5, found: 922.0.
Step 4: Synthesis of (64S, 4S, Z) -4-amino-11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- (4- (oxetan-3-yl) piperazin-1-yl) prop-1-yn-1-yl) pyri-din-3-yl) -10, 10-dimethyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione
To a stirred solution of tert-butyl ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- (4- (oxetan-3-yl) piperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (60 mg, 0.065 mmol) in dichloromethane (5 mL) was added TFA (1 mL) slowly at r.t., the resulting mixture was stirred at rt for 2 h. The reaction mixture was concen-trated to give the crude product (64S, 4S, Z) -4-amino-11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (pyrimidin-5-ylethynyl) pyridin-3-yl) -10, 10-dimethyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione (60 mg, crude ) as a light yellow solid.
LCMS (ESI) calcd. for C50H64N8O7S [M+H] + m/z 821.5, found: 822.0.
Step 5: Synthesis of (1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- (4- (oxetan-3-yl) piperazin-1-yl) prop-1-yn -1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
To a stirred solution of (64S, 4S, Z) -4-amino-11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (pyrimidin-5-ylethynyl) pyridin-3-yl) -10, 10-dimethyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione (60 mg, 0.065 mmol) and (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxylic acid (10 mg, 0.085 mmol) and NMI (21 mg, 0.260 mmol) in ACN/DMF (3.5 mL, V: V= 6: 1) was added TCFH (27 mg, 0.098 mmol) at rt, the resulting mixture was stirred at rt for 1 h. The reaction mixture was concentrated to give the crude product. The crude product was purified by HPLC (ACN/H2O (0.5 %NH4HCO3) from 40%to 95%in 30 min) to give (1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3- (4- (oxetan-3-yl) piperazin-1-yl) prop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide (19 mg, 31.9 %) as a white solid.
LCMS (ESI) calcd. for C51H64N8O6S [M+H] + m/z 917.5, found: 918.0.
1H NMR (400 MHz, DMSO-d6) δ 8.80 (d, J = 1.9 Hz, 1H) , 8.41 (d, J = 10.7 Hz, 2H) , 7.87 (d, J = 1.9 Hz, 1H) , 7.82 (s, 1H) , 7.75 (d, J = 8.5 Hz, 1H) , 7.58 (d, J = 8.7 Hz, 1H) , 5.93 (d, J = 11.2 Hz, 1H) , 5.38 (s, 1H) , 4.66 (d, J = 11.2 Hz, 1H) , 4.50 (dd, J = 13.0, 6.4 Hz, 3H) , 4.40 (t, J = 6.0 Hz, 2H) , 4.35 -4.24 (m, 2H) , 4.07 (dd, J = 14.7, 7.4 Hz, 1H) , 3.61 -3.51 (m, 4H) , 3.39 (dd, J = 12.6, 6.1 Hz, 1H) , 3.23 (s, 3H) , 3.14 (dd, J = 14.7, 7.2 Hz, 1H) , 2.94 (d, J = 14.3 Hz, 1H) , 2.67 -2.55 (m, 4H) , 2.42 (d, J = 14.9 Hz, 2H) , 2.36 -2.12 (m, 6H) , 1.57 (t, J = 9.1 Hz, 1H) , 1.36 (d, J = 6.0 Hz, 3H) , 1.24 (dt, J = 11.3, 5.8 Hz, 2H) , 1.20 -1.14 (m, 2H) , 1.13 -1.04 (m, 5H) , 0.92 -0.82 (m, 5H) , 0.33 (s, 3H) .
Example 66
(1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( (1'-methyl- [1, 4'-bipiperidin] -4-yl) ethynyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
Substituting 4- (prop-2-yn-1-yl) morpholine with 4-ethynyl-1'-methyl-1, 4'-bipiperidine in the Step 1 of Example 16, the title compound was prepared by the same procedures as described for Example 16.
Example 67
(1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( (1- (tetrahydro-2H-pyran-4-yl) azetidi n-3-yl) ethynyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -in dola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
Substituting 4- (prop-2-yn-1-yl) morpholine with 3-ethynyl-1- (tetrahydro-2H-pyran-4-yl) azetidine in the Step 1 of Example 16, the title compound was prepared by the same procedures as described for Ex-ample 16.
Example 68
(1r, 2R, 3S) -N- ( (64S, 4S, Z) -12- (5- (3- (4-cyclopropylpiperazin-1-yl) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
Step 1: Synthesis of (S) -1- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) -4-cyclopropylpiperazine
To a solution of 1-cyclopropylpiperazine (126 mg, 1.00 mmol) in DCM (5 mL) were added DIEA (129 g, 1.00 mmol) and a solution of (S) -3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl 4-methylbenzenesulfonate (212 mg, 0.500 mmol) in DCM (5 mL) at rt, the resulting mixture was stirred at rt for 2 h. After completion, the mixture was concentrated under reduced pressureto give a reside which was purified by silica gel column chromatography (eluting with EtOAc/PE from 0%to 40%) to obtain (S) -1- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) -4-cyclopropylpiperazine (180 mg, yield: 95%) as a yellow oil.
LCMS (ESI) calcd. for C18H24BrN3O [M+H] + m/z 377.1, found 378.5.
Step 2: Synthesis of tert-butyl ( (64S, 4S, Z) -12- (5- (3- (4-cyclopropylpiperazin-1-yl) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 1 0-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
A mixture of (S) -1- (3- (5-bromo-6- (1-methoxyethyl) pyridin-3-yl) prop-2-yn-1-yl) -4-cyclopropylpiperazine (100 mg, 0.26 mmol) , tert-butyl ( (64S, 4S, Z) -10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (200 mg, 0.286 mmol) , K2CO3 (106 mg, 0.78 mmol) and Pd (dppf) Cl2 (20 mg, 0.026 mmol) in dioxane (4 mL) and H2O (1 mL) was stirred at 85℃ under N2 for 8 h. After completion, the reaction mixture was concentrated to give the crude product which was purified by silica gel column (eluting with MeOH/DCM from 0%to 15%) to give tert-butyl ( (64S, 4S, Z) -12- (5- (3- (4-cyclopropylpiperazin-1-yl) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanac ycloundecaphane-4-yl) carbamate (120 mg, 53 %) as a yellow solid.
LCMS (ESI) calcd. for C48H60N8O6S [M+H] + m/z 876.4, found: 877.9.
Step 3: Synthesis of tert-butyl ( (64S, 4S, Z) -12- (5- (3- (4-cyclopropylpiperazin-1-yl) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
To a stirred solution of tert-butyl ( (64S, 4S, Z) -12- (5- (3- (4-cyclopropylpiperazin-1-yl) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanac ycloundecaphane-4-yl) carbamate (120 mg, 0.138 mmol) in DMF (3 mL) were added Cs2CO3 (90 mg, 0.276 mmol) and Iodoethane (42 mg, 0.276 mmol) at 0℃, the resulting mixture was stirred at rt overnight. After completion, the reaction mixture was diluted with EtOAc (20 mL) , washed with water (10 mL x 2) and saturated NaCl (10 mL) , then dried over Na2SO4, following with concentration under reduced pressure to obtain crude product which was purified by silica gel column chromatography (eluting with EtOAc/PE from 0%to 100%) to give tert-butyl ( (64S, 4S, Z) -12- (5- (3- (4-cyclopropylpiperazin-1-yl) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (50 mg, 40%) as a yellow solid.
LCMS (ESI) calcd. for C50H64N8O6S [M+H] + m/z 904.5, found: 906.0.
Step 4: Synthesis of (64S, 4S, Z) -4-amino-12- (5- (3- (4-cyclopropylpiperazin-1-yl) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3 -yl) -11-ethyl-10, 10-dimethyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] hept anacycloundecaphane-5, 7-dione
To a stirred solution of tert-butyl ( (64S, 4S, Z) -12- (5- (3- (4-cyclopropylpiperazin-1-yl) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (60 mg, 0.066 mmol) in dichloromethane (2 mL) was added TFA (1 mL) dropwisely at rt, the resulting mixture was stirred at rt for 1 h. The reaction mixture was concen-trated. The residue was dissolved in EtOAc (15 mL) . The mixture was washed with sat. NaHCO3 aquous (10 mL) and brine (10 mL) , then dried over Na2SO4, following with concentration under reduced pressure to obtain (64S, 4S, Z) -4-amino-12- (5- (3- (4-cyclopropylpiperazin-1-yl) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -11-ethyl-10, 10-dimethyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione (50 mg, yield: 94%) as a yellow solid.
LCMS (ESI) calcd. for C45H56N8O4S [M+H] + m/z 804.4, found: 805.9.
Step 5: Synthesis of (1r, 2R, 3S) -N- ( (64S, 4S, Z) -12- (5- (3- (4-cyclopropylpiperazin-1-yl) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
To a stirred solution of ( (64S, 4S, Z) -4-amino-12- (5- (3- (4-cyclopropylpiperazin-1-yl) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin -3-yl) -11-ethyl-10, 10-dimethyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione (50 mg, 0.062 mmol) and (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxylic acid (8 mg, 0.074 mmol) and 1-methyl-1H-imidazole (15 mg, 0.093 mmol) in ACN (2 mL) was added TCFH (26 mg, 0.093 mmol) at rt, the resulting mixture was stirred at rt for 1 h. The reaction mixture was concentrated to give the crude product. The crude prod-uct was purified by pre-HPLC (ACN/H2O (0.5 %NH4HCO3) from 20%to 95%in 30 min) to give (1r, 2R, 3S) -N- ( (64S, 4S, Z) -12- (5- (3- (4-cyclopropylpiperazin-1-yl) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) py ridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide as atropisomer 1 (16 mg, 29%) as a white solid and (1r, 2R, 3S) -N- ( (64S, 4S, Z) -12- (5- (3- (4-cyclopropylpiperazin-1-yl) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4 ) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide as atropisomer 2 (i.e., compound of Example 68) (10 mg, 18%) as a white solid.
LCMS (ESI) calcd. for C51H64N8O5S [M+H] + m/z 900.5, found: 901.9.
atropisomer 1: 1H NMR (400 MHz, DMSO-d6) δ 8.78 (d, J = 1.8 Hz, 1H) , 8.41-8.39 (m, 2H) , 7.97 (d, J = 1.9 Hz, 1H) , 7.80 (s, 1H) , 7.72 (d, J = 8.6 Hz, 1H) , 7.52 (d, J = 8.7 Hz, 1H) , 5.87 (d, J = 11.1 Hz, 1H) , 5.29 (t, J = 7.7 Hz, 1H) , 4.71 (d, J = 10.9 Hz, 1H) , 4.47 (dd, J = 9.9, 4.8 Hz, 1H) , 3.95 -3.87 (m, 2H) , 3.84 -3.77 (m, 1H) , 3.59 -3.47 (m, 4H) , 3.23 (d, J = 14.8 Hz, 1H) , 3.14 -2.99 (m, 5H) , 2.64-2.63 (m, 1H) , 2.53 (s, 8H) , 2.33-2.31 (m, 2H) , 2.13-2.11 (m, 1H) , 1.62-1.57 (m, 2H) , 1.25 -1.03 (m, 15H) , 0.89 (s, 3H) , 0.45 (s, 3H) , 0.39-0.35 (m, 2H) , 0.27 -0.21 (m, 2H) .
atropisomer 2 (i.e., compound of Example 68) : 1H NMR (400 MHz, DMSO-d6) δ 8.77 (d, J = 2.1 Hz, 1H) , 8.40-8.37 (m, 2H) , 7.83 (d, J = 1.9 Hz, 1H) , 7.80 (s, 1H) , 7.73 (d, J = 8.7 Hz, 1H) , 7.56 (d, J = 8.7 Hz, 1H) , 5.91 (d, J = 11.2 Hz, 1H) , 5.36 (t, J = 8.4 Hz, 1H) , 4.64 (d, J = 10.9 Hz, 1H) , 4.46 (dd, J = 8.8, 3.6 Hz, 1H) , 4.32 -4.22 (m, 2H) , 4.07-4.01 (m, 1H) , 3.59 -3.47 (m, 4H) , 3.21 (s, 3H) , 3.11 (dd, J = 14.7, 7.2 Hz, 2H) , 2.91 (d, J = 14.4 Hz, 2H) , 2.65-2.54 (m, 4H) , 2.45-2.38 (m, 6H) , 2.31-2.27 (m, 1H) , 2.16 -2.09 (m, 1H) , 1.61-1.52 (m, 2H) , 1.33 (d, J = 6.1 Hz, 3H) , 1.25 -0.98 (m, 10H) , 0.87-0.86 (m, 6H) , 0.37-0.24 (m, 7H) .
Example 69
(1r, 2R, 3S) -N- ( (64S, 4S, Z) -12- (5- (3- (1, 4-oxazepan-4-yl) prop-1-yn-1-yl) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
Substituting 4- (prop-2-yn-1-yl) morpholine with 4- (prop-2-yn-1-yl) -1, 4-oxazepane in the Step 1 of Example 16, the title compound was prepared by the same procedures as described for Example 16.
Example 70
(1r, 2R, 3S) -N- ( (22S, 64S, 4S) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
Substituting tert-butyl ( (64S, 4S, Z) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa -62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate with tert-butyl ( (22S, 64S, 4S) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carba mate in the Step 1 of Example 16, the title compound was prepared by the same procedures as described for Example 16.
Example 71
(1r, 2R, 3S) -N- ( (22R, 64S, 4S) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
Substituting tert-butyl ( (64S, 4S, Z) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa -62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate with tert-butyl ( (22R, 64S, 4S) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carba mate in the Step 1 of Example 16, the title compound was prepared by the same procedures as described for Example 16.
Example 72
(1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinobut-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
Substituting 4- (prop-2-yn-1-yl) morpholine with 4- (but-3-yn-2-yl) morpholine in the Step 1 of Exam-ple 16, the title compound was prepared by the same procedures as described for Example 16.
Example 73
(1S, 2S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( (R) -3- (4-methylpiperazin-1-yl) but-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2-methylcyclopropane-1-carboxamide
Substituting 4- (prop-2-yn-1-yl) morpholine with (R) -1- (but-3-yn-2-yl) -4-methylpiperazine in the Step 1 and substituting (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxylic acid with (1S, 2S) -2-methylcyclopropane-1-carboxylic acid in the Step 4 of Example 7, the title compound was pre-pared by the same procedures as described for Example 7.
Example 74
(1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( (R) -3- (4-methylpiperazin-1-yl) but-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
Substituting 4- (prop-2-yn-1-yl) morpholine with (R) -1- (but-3-yn-2-yl) -4-methylpiperazine in the Step 1 of Example 7, the title compound was prepared by the same procedures as described for Example 7.
Example 75
(1S, 2S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( (S) -3- (4-methylpiperazin-1-yl) but-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2-methylcyclopropane-1-carboxamide
Substituting (R) -1- (but-3-yn-2-yl) -4-methylpiperazine with (S) -1- (but-3-yn-2-yl) -4-methylpiperazine in the Step 1 of Example 73, the title compound was prepared by the same procedures as described for Example 73.
Example 76
(1S, 2S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( (S) -3- (4-methylpiperazin-1-yl) but-1-yn-1 -yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2-methylcyclopropane-1-carboxamide
Substituting (R) -1- (but-3-yn-2-yl) -4-methylpiperazine with (S) -1- (but-3-yn-2-yl) -4-methylpiperazine in the Step 1 of Example 74, the title compound was prepared by the same procedures as described for Example 74.
Example 77
(1S, 2S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( (S) -3- (4-methylpiperazin-1-yl) but-1-yn-1 -yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2-methylcyclopropane-1-carboxamide
Substituting (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxylic acid with (1S, 2S) -2-methylcyclopropane-1-carboxylic acid in the Step 19 of Example 78, the title compound was prepared by the same procedures as described for Example 78.
Example 78
(1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( (R) -3-morpholinobut-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
Step 1: Synthesis of (S) -but-3-yn-2-yl methanesulfonate
To a stirred solution of (S) -but-3-yn-2-ol 3 (300 mg, 4.28 mmol) in DCM (5mL) and DIEA (1.38g, 10.70 mmol) was added MsCl (736 mg, 6.42 mmol) , the resulting mixture was stirred at rt for 5 h. The re-action mixture was concentrated under reduced pressure to give the crude product which was purified by silica gel column (eluting with EtOAc/PE from 0%to 10%) to give (S) -but-3-yn-2-yl methanesulfonate (470 mg, 74.2%) as a brown oil.
Step 2: Synthesis of (R) -4- (but-3-yn-2-yl) morpholine
To a stirred solution of (S) -but-3-yn-2-yl methanesulfonate (230 mg, 1.55 mmol) in THF (4 mL) was addedmorpholine (677 mg, 7.77 mmol) dropwisely at rt, the resulting mixture was stirred at 75℃ for 5 h. After completion, the reaction solution was concentrated under reduced pressure to give the residual, the residual was purified by silica gel column (eluting with EtOAc/PE from 0%to 80%) to afford (R) -4- (but-3-yn-2-yl) morpholine (69 mg, 32.0 %) as a yellow oil.
LCMS (ESI) calcd. for C8H13NO [M+H] + m/z 140.1, found: 140.3.
Step 3: Synthesis of (S) -2- (1-methoxyethyl) -3- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyridine
To a stirred solution of (S) -3-bromo-2- (1-methoxyethyl) pyridine (80.1 g, 370.83 mmol) and B2Pin2 (113.1 g, 445.00 mmol) in 1, 4-dioxane (1000 mL) were added Pd (dppf) Cl2 (23.1 g, 31.52 mmol ) and KOAc (109.2 g, 1.11 mol) . The reaction was stirred at 100℃ for 18 h under N2 atmosphere. The reaction was monitored by LCMS. After completion, the reaction solution was concentrated under reduced pressure to give the residual which was purified by neutral Al2O3 column (eluting with EtOAc/PE from 0%to 100%) to afford (S) -2- (1-methoxyethyl) -3- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyridine (130.8 g) as a brown oil.
LCMS (ESI) calcd. for C14H22BNO3 [M+H] + m/z 264.2, found: 264.5.
Step 4: Synthesis of (S) -3- (5-bromo-2- (2- (1-methoxyethyl) pyridin-3-yl) -1H-indol-3-yl) -2, 2-dimethylpropyl acetate
To a stirred solution of 3- (5-bromo-2-iodo-1H-indol-3-yl) -2, 2-dimethylpropyl acetate (12.2 g, 27.17 mmol) , (S) -2- (1-methoxyethyl) -3- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyridine (11.4 g, 43.47 mmol) and K2CO3 (11.3 g, 81.51 mmol) in 1, 4-dioxane (180 mL) and H2O (45 mL) was added Pd (dppf) Cl2 (2.0 g, 2.72 mmol) at rt. The reaction mixture was stirred at 90℃ under N2 for 18 h. After completion, the reaction solution was concentrated under reduced pressure to remove 1, 4-dioxane to give the residual which was purified by silica gel column (eluting with EtOAc/PE from 0%to 80%) to afford (S) -3- (5-bromo-2- (2- (1-methoxyethyl) pyridin-3-yl) -1H-indol-3-yl) -2, 2-dimethylpropyl acetate (10.5 g, 84.1 %) as a yellow solid.
LCMS (ESI) calcd. for C23H27BrN2O3 [M+H] + m/z 459.1, found: 459.5.
Step 5: Synthesis of (S) -3- (5-bromo-1-ethyl-2- (2- (1-methoxyethyl) pyridin-3-yl) -1H-indol-3-yl) -2, 2-dimethylpropyl acetate
To a stirred solution of (S) -3- (5-bromo-2- (2- (1-methoxyethyl) pyridin-3-yl) -1H-indol-3-yl) -2, 2-dimethylpropyl acetate (12.4 g, 27.10 mmol) in DMF (120 mL) were added Cs2CO3 (17.6 g, 54.19 mmol) and Iodoethane (6.3 g, 40.64 mmol) at 0℃, the resulting mixture was stirred at rt for 18 h. The reaction was monitored by LCMS. After completion, the reaction mixture was diluted with EtOAc (200 mL) , washed with water (1000 mL x 2) and saturated NaCl (1000 mL) , then dried over Na2SO4, following with concentration under reduced pressure to obtain crude product which was purified by silica gel column (eluting with EtOAc/PE from 0%to 60%) to afford (S) -3- (5-bromo-1-ethyl-2- (2- (1-methoxyethyl) pyridin-3-yl) -1H-indol-3-yl) -2, 2-dimethylpropyl acetate (10.4 g, 79.0 %) as a brown solid.
LCMS (ESI) calcd. for C25H31BrN2O3 [M+H] + m/z 487.2, found: 487.6.
Step 6: Synthesis of (S) -3- (5-bromo-1-ethyl-2- (2- (1-methoxyethyl) pyridin-3-yl) -1H-indol-3-yl) -2, 2-dimethylpropan-1-ol
To a solution of (S) -3- (5-bromo-1-ethyl-2- (2- (1-methoxyethyl) pyridin-3-yl) -1H-indol-3-yl) -2, 2-dimethylpropyl acetate (10.4 g, 21.42 mmol) in THF (100 mL) , MeOH (50 mL) and H2O (50 mL) was added LiOH (2.7 g, 64.26 mmol) at 0-10℃, the resulting mixture was stirred at 0-10℃ for 20 h. The reaction was monitored by LCMS. After completion, the reaction mixture was concentrated to give the residue. The residue was di-luted with H2O and acidified to pH = 4-5 with 2 N HCl. The resulting mixture was extracted with EtOAc (100 mL x 2) , the combined organic phase was washed with saturated NaCl (100 mL) , then dried over Na2SO4 and concentrated to give the crude product which was purified by silica gel column (eluting with EtOAc/PE from 0%to 80%) to afford (S) -3- (5-bromo-1-ethyl-2- (2- (1-methoxyethyl) pyridin-3-yl) -1H-indol-3-yl) -2, 2-dimethylpropan-1-ol (5.0 g, 52.0 %) as a yellow solid.
LCMS (ESI) calcd. for C23H29BrN2O2 [M+H] + m/z 445.2, found: 445.6.
Step 7: Synthesis of (S) -3- (5-bromo-1-ethyl-2- (2- (1-methoxyethyl) pyridin-3-yl) -1H-indol-3-yl) -2, 2-dimethylpropyl acetate
To a stirred solution of (S) -3- (5-bromo-1-ethyl-2- (2- (1-methoxyethyl) pyridin-3-yl) -1H-indol-3-yl) -2, 2-dimethylpropan-1-ol (5.0 g, 11.15 mmol) in DCM (50 mL) were added DIEA (2.9 g, 22.30 mmol) , DMAP (136 mg, 1.11 mmol) and Ac2O (1.20 g, 11.71 mmol) at rt , t he resulting mixture was stirred at rt for 18 h. The reaction was moni-tored by LCMS. After completion, the reaction mixture was concetrated to give the crude product which was purified by silica gel column (eluting with EA/PE from 0%to 50%) to give (S) -3- (5-bromo-1-ethyl-2- (2- (1-methoxyethyl) pyridin-3-yl) -1H-indol-3-yl) -2, 2-dimethylpropyl acetate (5.1 g, 94.3 %) as a pale yellow solid.
LCMS (ESI) calcd. for C25H31BrN2O3 [M+H] + m/z 487.2, found: 487.6.
Step 8: Synthesis of (S) - (5- (3- (3-acetoxy-2, 2-dimethylpropyl) -5-bromo-1-ethyl-1H-indol-2-yl) -6- (1-methoxyethyl) pyridin-3-yl) boronic acid
To a stirred solution of (S) -3- (5-bromo-1-ethyl-2- (2- (1-methoxyethyl) pyridin-3-yl) -1H-indol-3-yl) -2, 2-dimethylpropyl acetate (5.1 g, 10.51 mmol) , B2Pin2 (4.0 g, 15.77 mmol) in THF (100 mL) was added [Ir (COD) Cl] 2 (353 mg, 0.53 mmol) and dtbpy (423 mg, 1.58 mmol) at rt , the resulting mixture was stirred at 85℃ under N2 for 24 h. The re-action was monitored by LCMS. After completion, the reaction mixture was concentrated to give the crude product which was used to next step without purification.
Step 9: Synthesis of (S) -3- (5-bromo-1-ethyl-2- (5-hydroxy-2- (1-methoxyethyl) pyridin-3-yl) -1H-indol-3-yl) -2, 2-dimethylpropyl acetate
To a stirred solution of crude (S) - (5- (3- (3-acetoxy-2, 2-dimethylpropyl) -5-bromo-1-ethyl-1H-indol-2-yl) -6- (1-methoxyethyl) pyridin-3-yl )boronic acid (4.5 g, 8.40 mmol) in THF (300 mL) was added H2O2 (4.8 g, 41.98 mmol) at r.t. The reaction mixture was stirred at r.t. for 2 h. After completion, reaction mixture was diluted with EtOAc (60 mL) , washed with water (50 mL x 2) and saturated NaCl (50 mL) , then dried over Na2SO4, following with con-centration under reduced pressure to obtain the crude (S) -3- (5-bromo-1-ethyl-2- (5-hydroxy-2- (1-methoxyethyl) pyridin-3-yl) -1H-indol-3-yl) -2, 2-dimethylpropyl acetate (3.4 g, 80.9 %) as a yellow solid.
LCMS (ESI) calcd. for C25H31BrN2O4 [M+H] + m/z 503.2, found: 503.7.
Step 10: Synthesis of (S) -3- (2- (5- (benzyloxy) -2- (1-methoxyethyl) pyridin-3-yl) -5-bromo-1-ethyl-1H-indol-3-yl) -2, 2-dimethylpropy l acetate
To a stirred solution of crude (S) -3- (5-bromo-1-ethyl-2- (5-hydroxy-2- (1-methoxyethyl) pyridin-3-yl) -1H-indol-3-yl) -2, 2-dimethylpropyl acetate (3.4 g, 6.79 mmol) in CH3CN (68 mL) were added K2CO3 (2.8 g, 20.38 mmol) and BnBr (1.7 g, 10.19 mmol) , the resulting mixture was stirred at 85℃ for 16 h. The reaction mixture was concentrated to give the crude product which was purified by silica gel column (eluting with EA/PE from 0%to 30%) to give (S) -3- (2- (5- (benzyloxy) -2- (1-methoxyethyl) pyridin-3-yl) -5-bromo-1-ethyl-1H-indol-3-yl) -2, 2-dimethylpropyl acetate (3.2 g, 79.4 %) as a yellow foam.
LCMS (ESI) calcd. for C32H37BrN2O4 [M+H] + m/z 593.2, found: 593.6.
Step 11: Synthesis of (S) -3- (2- (5- (benzyloxy) -2- (1-methoxyethyl) pyridin-3-yl) -1-ethyl-5- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -1H-indol-3-yl) -2, 2-dimethylpropyl acetate
To a stirred solution of (S) -3- (2- (5- (benzyloxy) -2- (1-methoxyethyl) pyridin-3-yl) -5-bromo-1-ethyl-1H-indol-3-yl) -2, 2-dimethylpropyl acetate (3.2 g, 5.39 mmol) and B2Pin2 (2.1 g, 8.08 mmol) in 1, 4-dioxane (45 mL) were added Pd (dppf) Cl2 (395 mg, 0.54 mmol ) and KOAc (1.6 g, 16.17 mol ) . The reaction mixture was stirred at 90℃ for 5 h under N2 atmosphere. The reaction was monitored by LCMS. After completion, the reaction solu-tion was concentrated under reduced pressure to give the residual which was purified by silica gel col-umn (eluting with EtOAc/PE from 0%to 30%) to afford (S) -3- (2- (5- (benzyloxy) -2- (1-methoxyethyl) pyridin-3-yl) -1-ethyl-5- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborola n-2-yl) -1H-indol-3-yl) -2, 2-dimethylpropyl acetate (3.7 g) as a yellow oil.
LCMS (ESI) calcd. for C38H49BrN2O6 [M+H] + m/z 641.4, found: 641.9.
Step 12: Synthesis of methyl (S) -2- ( (S) -3- (4- (3- (3-acetoxy-2, 2-dimethylpropyl) -2- (5- (benzyloxy) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -1-ethyl-1H-indol-5-yl) thiazol-2-yl) -2- ( (tert-butoxycarbonyl) amino) propanoyl) -2, 3-diazabicyclo [3.1.1] heptane-4-carboxylate
To a stirred solution of methyl (S) -2- ( (S) -3- (4-bromothiazol-2-yl) -2- ( (tert-butoxycarbonyl) amino) propanoyl) -2, 3-diazabicyclo [3.1.1] hepta ne-4-carboxylate (11.9 g, 24.31 mmol) , (S) -3- (2- (5- (benzyloxy) -2- (1-methoxyethyl) pyridin-3-yl) -1-ethyl-5- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborola n-2-yl) -1H-indol-3-yl) -2, 2-dimethylpropyl acetat (15.6 g, 24.31 mmol) and K2CO3 (10.1 g, 73.19 mmol) in toluene/1, 4-dioxane/H2O (350 mL, V: V: V = 3: 1: 1 ) was added Pd (dppf) Cl2 (1.8 g, 2.46 mmol) at rt. The reaction mixture was stirred at 90℃ under N2 for 22 h. After completion, the reaction solution was concen-trated under reduced pressure to remove 1, 4-dioxane and toluene, the residual was diluted with H2O and extracted with EtOAc (100 mL x 2) , the combined organic phase was washed with saturated NaCl (200 mL) , then dried over Na2SO4, following with concentration under reduced pressure to obtain crude product which was purified by silica gel column (eluting with EtOAc/PE from 0%to 80%) to afford methyl (S) -2- ( (S) -3- (4- (3- (3-acetoxy-2, 2-dimethylpropyl) -2- (5- (benzyloxy) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -1 -ethyl-1H-indol-5-yl) thiazol-2-yl) -2- ( (tert-butoxycarbonyl) amino) propanoyl) -2, 3-diazabicyclo [3.1.1] heptane-4-carboxylate (17.2 g, 76.8 %) as a brown foam.
LCMS (ESI) calcd. for C50H62N6O9S [M+H] + m/z 923.4, found: 924.7.
Step 13: Synthesis of (S) -2- ( (S) -3- (4- (2- (5- (benzyloxy) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -1-ethyl-3- (3-hydroxy-2, 2-dimethylpropyl) -1H-indol-5-yl) thiazol-2-yl) -2- ( (tert-butoxycarbonyl) amino) propanoyl) -2, 3-diazabicyclo [3.1.1] heptane-4-carboxylic acid
To a solution of methyl (S) -2- ( (S) -3- (4- (3- (3-acetoxy-2, 2-dimethylpropyl) -2- (5- (benzyloxy) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -1 -ethyl-1H-indol-5-yl) thiazol-2-yl) -2- ( (tert-butoxycarbonyl) amino) propanoyl) -2, 3-diazabicyclo [3.1.1] heptane-4-carboxylate (17.4 g, 18.66 mmol) in THF (170 mL) , MeOH (45 mL) and H2O (85 mL) was added LiOH (3.9 g, 93.28 mmol) at 0-10℃, the resulting mixture was stirred at rt for 17 h. The reaction was monitored by LCMS. After completion, the reaction mixture was concentrated to give the residue. The residue was diluted with H2O and acidified to pH = 4-5 with 2 N HCl. The resulting mixture was extracted with EtOAc (200 mL x 2) , the combined organic phase was washed with saturated NaCl (200 mL) , then dried over Na2SO4 and concentrated to give crude (S) -2- ( (S) -3- (4- (2- (5- (benzyloxy) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -1-ethyl-3- (3-hydroxy-2, 2-dimethyl pro-pyl) -1H-indol-5-yl) thiazol-2-yl) -2- ( (tert-butoxycarbonyl) amino) propanoyl) -2, 3-diazabicyclo [3.1.1] heptane -4-carboxylic acid (17.4 g) as a brown foam.
LCMS (ESI) calcd. for C47H58N6O8S [M+H] + m/z 867.4, found: 868.6.
Step 14: Synthesis of tert-butyl ( (64S, 4S, Z) -12- (5- (benzyloxy) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carba mate
To a solution of crude (S) -2- ( (S) -3- (4- (2- (5- (benzyloxy) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -1-ethyl-3- (3-hydroxy-2, 2-dimethyl pro-pyl) -1H-indol-5-yl) thiazol-2-yl) -2- ( (tert-butoxycarbonyl) amino) propanoyl) -2, 3-diazabicyclo [3.1.1] heptane -4-carboxylic acid (13.0 g, 15.01 mmol) , HOBt (10.1 g, 75.06 mmol) and DIEA (64.0 g, 495.38 mmol) in DCM (1300 mL) under ice-water bath was added EDCI (80.6 g, 420.32 mmol) in batches. The reaction was gradually improved temperature to room temperature and stirred for 22 h, the reaction was monitored by LCMS. After completion, the reaction was washed with H2O (700 mL x 3) and NH4Cl (aq) (700 mL x 3) , then dried over Na2SO4, following with concentration under reduced pressure to obtain crude product which was purified by silica gel column (eluting with EtOAc/PE from 0%to 65%) to afford tert-butyl ( (64S, 4S, Z) -12- (5- (benzyloxy) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H -8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carba mate (6.4 g, 50.3 %) as a yellow foam.
LCMS (ESI) calcd. for C47H56N5O6S [M+H] + m/z 849.4, found: 850.2.
Step 15: Synthesis of tert-butyl ( (64S, 4S, Z) -11-ethyl-12- (5-hydroxy-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
To a stirred solution of tert-butyl ( (64S, 4S, Z) -12- (5- (benzyloxy) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H -8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carba mate (6.2 g, 7.28 mmol) in IPA (180 mL) was added Pd (OH) 2/C (10%, 1.2 g) at rt , the resulting mixture was stirred at 85℃ for 60 h. The reaction was monitored by LCMS. After completion, the reaction mixture was filtered and the filtrate was concentrated to give crude tert-butyl ( (64S, 4S, Z) -11-ethyl-12- (5-hydroxy-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (6.6 g) as a white solid.
LCMS (ESI) calcd. for C40H50IN6O7S [M+H] + m/z 759.4, found: 760.2.
Step 16: Synthesis of 5- ( (64S, 4S, Z) -4- ( (tert-butoxycarbonyl) amino) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4 , 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl trifluoromethanesulfonate
To a solution of crude tert-butyl ( (64S, 4S, Z) -11-ethyl-12- (5-hydroxy-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbama te (6.60 g, 8.71 mmol) in DCM (130 mL) were added DIEA (2.2 g, 17.41 mmol) and PhN (Tf) 2 (3.1 g, 8.71 mmol) at 0-10℃. The resulting mixture was stirred at rt for 2 h. The reaction was monitored by LCMS. After completion, the reaction mixture was concentrated under reduced pressure to obtain crude product which was purified by silica gel column eluting with (EtOAc/PE from 0%to 60%) to afford 5- ( (64S, 4S, Z) -4- ( (tert-butoxycarbonyl) amino) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl trifluoromethanesulfonate (4.8 g, 61.9 %) as a white foam.
LCMS (ESI) calcd. for C41H49F3N6O9S2 [M+H] + m/z 891.3, found: 892.2.
Step 17: Synthesis of tert-butyl ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( (R) -3-morpholinobut-1-yn-1-yl) pyridin-3-yl) -10, 10-dime thyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate
A mixture of 5- ( (64S, 4S, Z) -4- ( (tert-butoxycarbonyl) amino) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl trifluoromethanesulfonate (256 mg, 0.29 mmol) , (R) -4- (but-3-yn-2-yl) morpholine (60 mg, 0.43 mmol) , CuI (9 mg, 0.043 mmol) , TEA (87 mg, 0.86 mmol) and Pd (PPh3) 2Cl2 (50 mg, 0.043 mmol) in DMF (4 mL) was stirred at 100℃ for 1 h under N2 atmosphere, the reaction was monitored by LCMS. After completion, the mixture was filtered and the filtrate was concentrated, the residue was purified by silica gel column chromatography (eluting with MeOH/DCM from 0%to 10%) to obtain tert-butyl ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( (R) -3-morpholinobut-1-yn-1-yl) pyridin-3-yl) -10, 10-dime-thyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (120 mg, 47.1%) as a brown foam.
LCMS (ESI) calcd. for C48H61N7O7S [M+H] + m/z 880.4, found: 881.4.
Step 18: Synthesis of (64S, 4S, Z) -4-amino-11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( (R) -3-morpholinobut-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundeca phane-5, 7-dione
To a stirred solution of tert-butyl ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( (R) -3-morpholinobut-1-yn-1-yl) pyridin-3-yl) -10, 10-dime-thyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (120 mg, 0.14 mmol) in dichloromethane (2 mL) was added TFA (0.6 mL) drop-wisely at rt, the resulting mixture was stirred at rt for 2 h. The reaction mixture was concentrated to give crude
(64S, 4S, Z) -4-amino-11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( (R) -3-morpholinobut-1-yn-1-yl) pyridin-3-yl) -1 0, 10-dimethyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione which was used to next step without purification.
LCMS (ESI) calcd. for C43H53N7O5S [M+H] + m/z 780.4, found: 781.4.
Step 19: Synthesis of (1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( (R) -3-morpholinobut-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] hept anacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
To a stirred solution of crude (64S, 4S, Z) -4-amino-11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( (R) -3-morpholinobut-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-5, 7-dione (106 mg, 0.14 mmol) and (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxylic acid (16 mg, 0.14 mmol) and NMI (112 mg, 1.37 mmol) in ACN (1.5 mL) and DCM (1.5 mL) was added TCFH (58 mg, 0.20 mmol) at rt, the resulting mixture was stirred at rt for 1 h. The reaction mixture was concentrated to give the crude product. The crude product was purified by pre-HPLC (eluting with CH3CN/H2O (0.1%NH4HCO3) from 20%to 80%) to give (1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( (R) -3-morpholinobut-1-yn-1-yl) pyridin-3 -yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide (26.8 mg) as a white solid.
LCMS (ESI) calcd. for C49H61N7O6S [M+H] + m/z 876.4, found: 876.5.
1H NMR (400 MHz, DMSO-d6) δ 8.76 (s, 1H) , 8.40 (d, J = 6.5 Hz, 2H) , 7.89 (s, 1H) , 7.81 (s, 1H) , 7.74 (d, J = 8.5 Hz, 1H) , 7.57 (d, J = 8.7 Hz, 1H) , 5.92 (d, J = 11.1 Hz, 1H) , 5.35 (s, 1H) , 4.65 (d, J = 10.9 Hz, 1H) , 4.46 (d, J = 4.7 Hz, 1H) , 4.37 -4.20 (m, 2H) , 4.05 (dd, J = 14.7, 7.6 Hz, 1H) , 3.89 (s, 2H) , 3.64 -3.46 (m, 6H) , 3.24 (d, J = 16.8 Hz, 3H) , 3.12 (dd, J = 15.1, 7.1 Hz, 1H) , 2.92 (d, J = 13.9 Hz, 1H) , 2.61 (d, J = 5.7 Hz, 1H) , 2.45 -2.36 (m, 2H) , 2.30 (d, J = 5.5 Hz, 1H) , 2.13 (t, J = 9.7 Hz, 1H) , 1.56 (s, 1H) , 1.48 (s, 5H) , 1.34 (d, J = 5.9 Hz, 3H) , 1.22 (s, 2H) , 1.16 (s, 2H) , 1.06 (dd, J = 10.5, 5.8 Hz, 5H) , 0.85 (d, J = 8.2 Hz, 5H) , 0.30 (s, 3H) .
Example 79
(1S, 2S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( (S) -3-morpholinobut-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2-methylcyclopropane-1-carboxamide
Substituting (R) -4- (but-3-yn-2-yl) morpholine with (S) -4- (but-3-yn-2-yl) morpholine in the Step 17 and substituting (1r, 2R, 3S) -2, 3-dimethylcyclopropane-1-carboxylic acid with (1S, 2S) -2-methylcyclopropane-1-carboxylic acid in the Step 19 of Example 78, the title compound was prepared by the same procedures as described for Example 78.
Example 80
(1r, 2R, 3S) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( (S) -3-morpholinobut-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -2, 3-dimethylcyclopropane-1-carboxamide
Substituting (R) -4- (but-3-yn-2-yl) morpholine with (S) -4- (but-3-yn-2-yl) morpholine in the Step 17 of Example 78, the title compound was prepared by the same procedures as described for Example 78.
Example 81
(2S) -2-cyclopentyl-2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (22S, 63S, 4S) -12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11- (2, 2, 2-trifluoroethyl) -61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (1, 3) -pyridazinacycloundecaphane-4-yl) acetamide
Substituting tert-butyl ( (22S, 64S, 4S) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carba mate with tert-butyl ( (22S, 63S, 4S) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (1, 3) -pyridazinacycloundecaphane-4-yl) c arbamate in the Step 1 of Example 23, the title compound was prepared by the same procedures as de-scribed for Example 23.
Example 82
(2S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N - ( (22S, 63S, 4S) -12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11- (2, 2, 2-trifluoroethyl) -61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -morpholina-1 (5, 3) -in dola-6 (1, 3) -pyridazinacycloundecaphane-4-yl) -3-methylbutanamide
Substituting (S) -2-cyclopentyl-2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) acetic acid with (S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -3-meth ylbutanoic acid in the Step 4 of Example 81, the title compound was prepared by the same procedures as described for Example 81.
Example 83
(2S) -2-cyclopentyl-2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (22S, 63S, 4S) -12- (2- ( (S) -1-methoxyethyl) -5- ( (R) -3- (4-methylpiperazin-1-yl) but-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11- (2, 2, 2-trifluoroethyl) -61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (1, 3) -pyridazinacycloundecaphane-4-yl) acetamide
Substituting 4- (prop-2-yn-1-yl) morpholine with (R) -1- (but-3-yn-2-yl) -4-methylpiperazine in the Step 1 of Example 81, the title compound was prepared by the same procedures as described for Example 81.
Example 84
(2S) -2-cyclopentyl-2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (22S, 64S, 4S) -12- (2- ( (S) -1-methoxyethyl) -5- ( (R) -3- (4-methylpiperazin-1-yl) but-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11- (2, 2, 2-trifluoroethyl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) acetamide
Substituting tert-butyl ( (22S, 63S, 4S) -10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11- (2, 2, 2-trifluor oethyl) -61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (1, 3) -pyridazinacyclound ecaphane-4-yl) carbamate with tert-butyl ( (22S, 64S, 4S) -10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11- (2, 2, 2-trifluor oethyl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate in the Step 1 of Example 83, the title compound was prepared by the same proce-dures as described for Example 83.
Example 85
(2S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (22S, 63S, 4S) -12- (2- ( (S) -1-methoxyethyl) -5- ( (R) -3- (4-methylpiperazin-1-yl) but-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11- (2, 2, 2-trifluoroethyl) -61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -morp holina-1 (5, 3) -indola-6 (1, 3) -pyridazinacycloundecaphane-4-yl) -3-methylbutanamide
Substituting 4- (prop-2-yn-1-yl) morpholine with (R) -1- (but-3-yn-2-yl) -4-methylpiperazine in the Step 4 of Example 82, the title compound was prepared by the same procedures as described for Example 82.
Example 86
(2S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N - ( (22S, 64S, 4S) -12- (2- ( (S) -1-methoxyethyl) -5- ( (R) -3- (4-methylpiperazin-1-yl) but-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11- (2, 2, 2-trifluoroethyl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -in dola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -3-methylbutanamide
Substituting (S) -2-cyclopentyl-2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) acetic acid with (S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -3-meth ylbutanoic acid in the Step 4 of Example 84, the title compound was prepared by the same procedures as described for Example 84.
Example 87
(2S) -2- (7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (pyrimidin-5-ylethynyl) pyridin-3-yl) -10, 10-dimet hyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -3-methylbutanamide
Step 1: Synthesis of 5- ( (64S, 4S, Z) -4-amino-11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl trifluoro-methanesulfonate
To a stirred solution of 5- ( (64S, 4S, Z) -4- ( (tert-butoxycarbonyl) amino) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl trifluoromethanesulfonate (330 mg, 0.37 mmol) in dichloromethane (10 mL) was added TFA (3 mL) dropwisely at rt, the resulting mixture was stirred at rt for 1 h. The reaction mixture was washed with sat. NaHCO3 (10 mL) , brine (10 mL) , then dried over Na2SO4, following with concentration under reduced pressure to obtain crude product 5- ( (64S, 4S, Z) -4-amino-11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl trifluoro-methanesulfonate (293 mg) as a yellow solid.
LCMS (ESI) calcd. for C36H41F3N6O7S2 [M+H] + m/z 791.2, found: 792.5.
Step 2: Synthesis of tert-butyl 7- ( (2S) -1- ( ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( ( (trifluoromethyl) sulfonyl) oxy) pyridin-3-yl) -1 0, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) -2, 7-diazaspiro [4.4] nonane-2-carboxylate
To a solution of (2S) -2- (7- (tert-butoxycarbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -3-methylbutanoic ac-id (121 mg, 0.37 mmol) in DMF (6 mL) were added HATU (183 mg, 0.37 mmol) and DIEA (239 mg, 1.85 mmol) at 0-10℃. The resulting mixture was stirred at RT for 30 min. Then 5- ( (64S, 4S, Z) -4-amino-11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl trifluoro-methanesulfonate (293 mg, 0.37 mmol) was added into the reaction mixture, the resulting mixture was stirred at rt for 2 h. The reaction was monitored by LCMS. After completion, the reaction mixture was di-luted with brine and extracted with EtOAc (2 x 30 mL) , the combined organic phase was washed with sat-urated NaCl (50 mL) , then dried over Na2SO4, following with concentration under reduced pressure to ob-tain crude product which was purified by silica gel column eluting with EtOAc/PE from 0%to 100%to afford tert-butyl 7- ( (2S) -1- ( ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( ( (trifluoromethyl) sulfonyl) oxy) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptan acycloundecaphane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) -2, 7-diazaspiro [4.4] nonane-2-carboxylate (280 mg, 69 %) as a yellow solid.
LCMS (ESI) calcd. for C53H69F3N8O10S2 [M+H] + m/z 1099.5, found: 1101.0.
Step 3: Synthesis of 5- ( (64S, 4S, Z) -11-ethyl-10, 10-dimethyl-4- ( (2S) -3-methyl-2- (2, 7-diazaspiro [4.4] nonan-2-yl) butanamido) -5, 7 -dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl trifluoromethanesulfonate
To a stirred solution of tert-butyl 7- ( (2S) -1- ( ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( ( (trifluoromethyl) sulfonyl) oxy) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptan acycloundecaphane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) -2, 7-diazaspiro [4.4] nonane-2-carboxylate (280 mg, 0.25 mmol) in dichloromethane (2 mL) was added TFA (0.5 mL) dropwisely at rt, the resulting mix-ture was stirred at rt for 1 h. The reaction mixture was washed with sat. NaHCO3 (10 mL) , brine (10 mL) , then dried over Na2SO4, following with concentration under reduced pressure to obtain crude product 5- ( (64S, 4S, Z) -11-ethyl-10, 10-dimethyl-4- ( (2S) -3-methyl-2- (2, 7-diazaspiro [4.4] nonan-2-yl) butanamido) -5, 7 -dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl trifluoromethanesulfonate (255 mg) as a yellow solid.
LCMS (ESI) calcd. for C48H61F3N8O8S2 [M+H] + m/z 999.4, found: 1000.1.
Step 4: Synthesis of 5- ( (64S, 4S, Z) -4- ( (2S) -2- (7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -3-methylbutanamido) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl tri-fluoromethanesulfonate
To a solution of lithium (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carboxylate (75 mg, 0.51 mmol) in DMF (3 mL) were added HATU (116 mg, 0.31 mmol) and DIEA (329 mg, 2.5 mmol) at 0-10℃. The resulting mixture was stirred at RT for 30 min. Then 5- ( (64S, 4S, Z) -11-ethyl-10, 10-dimethyl-4- ( (2S) -3-methyl-2- (2, 7-diazaspiro [4.4] nonan-2-yl) butanamido) -5, 7 -dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane -12-yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl trifluoromethanesulfonate (255 mg, 0.25 mmol) was added into the reaction mixture, the resulting mixture was stirred at rt for 2 h. The reaction was monitored by LCMS. After completion, the reaction mixture was diluted with brine and extracted with EtOAc (2 x 30 mL) , the combined organic phase was washed with saturated NaCl (50 mL) , then dried over Na2SO4, following with concentration under reduced pressure to obtain crude product which was purified by silica gel column eluting with MeOH/DCM from 0%to 7%to afford 5- ( (64S, 4S, Z) -4- ( (2S) -2- (7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -3-methylbutanamido) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5 , 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl tri-fluoromethanesulfonate (160 mg, 56 %) as an orange oil.
LCMS (ESI) calcd. for C53H69F3N8O10S2 [M+H] + m/z 1122.5, found: 1123.2.
Step 5: Synthesis of (2S) -2- (7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (pyrimidin-5-ylethynyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-1 1H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -3 -methylbutanamide
To a stirred solution of 5- ( (64S, 4S, Z) -4- ( (2S) -2- (7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -3-methylbutanamido) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5 , 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl tri-fluoromethanesulfonate (80 mg, 0.071 mmol) and 5-ethynylpyrimidine (15 mg, 0.14 mmol) , CuI (2 mg, 0.011 mmol) , TEA (0.5 mL) and Pd (PPh3) 4 (12 mg, 0.011 mmol) in DMF (2 mL) was stirred at 90℃ for 1 h under N2 atmosphere, the reaction was monitored by LCMS. After completion, the mixture was filtered and the filtrate was concentrated, the residue was purified by pre-HPLC (eluting with CH3CN/H2O (0.1%NH4HCO3) from 20%to 70%) to give (2S) -2- (7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (pyrimidin-5-ylethynyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo -11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -3-methylbutanamide (28 mg, 36 %) as a white solid.
LCMS (ESI) calcd. for C60H73N11O6S [M+H] + m/z 1076.6, found: 1077.5.
1H NMR (400 MHz, DMSO-d6) δ 9.24 (s, 1H) , 9.08 (s, 1H) , 8.99 (s, 1H) , 8.43 (s, 1H) , 8.37 -8.14 (m, 2H) , 8.09 (s, 1H) , 7.88 (d, J = 9.7 Hz, 1H) , 7.76 (t, J = 12.4 Hz, 1H) , 7.64 -7.55 (m, 1H) , 6.01 -5.89 (m, 1H) , 5.34 (dd, J = 13.5, 8.1 Hz, 1H) , 4.76 (d, J = 11.3 Hz, 1H) , 4.50 (s, 1H) , 4.41-4.35 (m, 2H) , 4.04 (dt, J = 63.0, 19.2 Hz, 2H) , 3.76 -3.36 (m, 5H) , 3.29 (s, 4H) , 3.13 -2.55 (m, 8H) , 2.53 (s, 5H) , 2.46 -2.06 (m, 6H) , 2.04 -1.54 (m, 6H) , 1.49 -1.18 (m, 5H) , 1.07 -0.71 (m, 9H) , 0.62-0.55 (m, 2H) , 0.39 -0.21 (m, 4H) , 0.19-0.15 (m, 1H) .
Example 88
(2S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N - ( (22S, 64S, 4S) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (pyrimidin-5-ylethynyl) pyridin-3-yl) -10, 10-dimet hyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -3-methylbutanamide
Step 1: Synthesis of methyl (S) -3- ( (S) -4- (3- (3-acetoxy-2, 2-dimethylpropyl) -2- (5- (benzyloxy) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -1-ethyl-1H-indol-5-yl) morpholin-2-yl) -2- ( (tert-butoxycarbonyl) amino) propanoate
To a stirred solution of (S) -3- (2- (5- (benzyloxy) -2- (1-methoxyethyl) pyridin-3-yl) -5-bromo-1-ethyl-1H-indol-3-yl) -2, 2-dimethylpropyl acetate (2.6 g, 4.4 mmol) , methyl (S) -2- ( (tert-butoxycarbonyl) amino) -3- ( (S) -morpholin-2-yl) propanoate (1.3 g, 4.4 mmol) and Cs2CO3 (3.6 g, 11 mmol) in 1, 4-dioxane (50 mL) were added RuphosG3Pd (367 mg, 0.4 mmol) and Ruphos (410 mg, 0.9 mmol at rt. The reaction mixture was stirred at 100 ℃ under N2 for 18 h. After completion, the reaction solution was concentrated under reduced pressure to remove 1, 4-dioxane to give the residual which was purified by silica gel column (eluting with EtOAc/PE from 0%to 80%) to afford methyl (S) -3- ( (S) -4- (3- (3-acetoxy-2, 2-dimethylpropyl) -2- (5- (benzyloxy) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -1-et hyl-1H-indol-5-yl) morpholin-2-yl) -2- ( (tert-butoxycarbonyl) amino) propanoate (2 g, 57 %) as a yellow oil.
LCMS (ESI) calcd. for C45H60N4O9 [M+H] + m/z 801.4, found: 802.6.
Step 2: Synthesis of (S) -3- ( (S) -4- (2- (5- (benzyloxy) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -1-ethyl-3- (3-hydroxy-2, 2-dimethylpropy l) -1H-indol-5-yl) morpholin-2-yl) -2- ( (tert-butoxycarbonyl) amino) propanoic acid
To a solution of methyl (S) -3- ( (S) -4- (3- (3-acetoxy-2, 2-dimethylpropyl) -2- (5- (benzyloxy) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -1-et hyl-1H-indol-5-yl) morpholin-2-yl) -2- ( (tert-butoxycarbonyl) amino) propanoate (1.7 g, 2.1 mmol) in THF (18 mL) and H2O (6 mL) was added LiOHH2O (267 mg, 6.4 mmol) at 0-10℃ , the resulting mixture was stirred at RT for 16 h. The reaction was monitored by LCMS. After completion, the reaction mixture was acidified to pH = 3-4 with 2 N HCl. The resulting mixture was extracted with EtOAc (300 mL x 2) , the combined organic phase was washed with saturated NaCl (500 mL) , then dried over Na2SO4 and concen-trated to give (S) -3- ( (S) -4- (2- (5- (benzyloxy) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -1-ethyl-3- (3-hydroxy-2, 2-dimethylpropyl) -1H-indol-5-yl) morpholin-2-yl) -2- ( (tert-butoxycarbonyl) amino) propanoic acid (1.58 g, 100 %) as a light yellow solid.
LCMS (ESI) calcd. for C42H56N4O8 [M+H] + m/z 745.4, found: 746.3.
Step 3: Synthesis of methyl (S) -2- ( (S) -3- ( (S) -4- (2- (5- (benzyloxy) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -1-ethyl-3- (3-hydroxy-2, 2-dimethylpro-pyl) -1H-indol-5-yl) morpholin-2-yl) -2- ( (tert-butoxycarbonyl) amino) propanoyl) -2, 3-diazabicyclo [3.1.1] heptane-4-carboxylate
To a solution of (S) -3- ( (S) -4- (2- (5- (benzyloxy) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -1-ethyl-3- (3-hydroxy-2, 2-dimethylpropyl) -1H-indol-5-yl) morpholin-2-yl) -2- ( (tert-butoxycarbonyl) amino) propanoic acid (1.58 g, 2.1 mmol) in DMF (20 mL) were added HATU (1.05 g, 2.8 mmol) and DIEA (1.37 g, 10.6 mmol) at 0-10℃. The re-sulting mixture was stirred at 0-10℃ for 10 min. Then methyl (S) -2, 3-diazabicyclo [3.1.1] heptane-4-carboxylate hydrochloride (531 mg, 2.8 mmol) was added into the reaction mixture, the resulting mixture was stirred at rt for 2 h. The reaction was monitored by LCMS. Af-ter completion, the reaction mixture was diluted with brine and extracted with EtOAc (2 x 30 mL) , the combined organic phase was washed with saturated NaCl (100 mL) , then dried over Na2SO4, following with concentration under reduced pressure to obtain crude product which was purified by silica gel col-umn eluting with EtOAc/PE from 0%to 100%to afford methyl (S) -2- ( (S) -3- ( (S) -4- (2- (5- (benzyloxy) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -1-ethyl-3- (3-hydroxy-2, 2-dimet hylpro-pyl) -1H-indol-5-yl) morpholin-2-yl) -2- ( (tert-butoxycarbonyl) amino) propanoyl) -2, 3-diazabicyclo [3.1.1] hept ane-4-carboxylate (750 mg, 40 %) as a light yellow solid.
LCMS (ESI) calcd. for C49H66N6O9 [M+H] + m/z 883.5, found: 884.2.
Step 4: Synthesis of (S) -2- ( (S) -3- ( (S) -4- (2- (5- (benzyloxy) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -1-ethyl-3- (3-hydroxy-2, 2-dimethylpro-pyl) -1H-indol-5-yl) morpholin-2-yl) -2- ( (tert-butoxycarbonyl) amino) propanoyl) -2, 3-diazabicyclo [3.1.1] hept ane-4-carboxylic acid
To a solution of (S) -2- ( (S) -3- ( (S) -4- (2- (5- (benzyloxy) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -1-ethyl-3- (3-hydroxy-2, 2-dimet hylpro-pyl) -1H-indol-5-yl) morpholin-2-yl) -2- ( (tert-butoxycarbonyl) amino) propanoyl) -2, 3-diazabicyclo [3.1.1] hept ane-4-carboxylate (750 mg, 0.85 mmol) in THF (6 mL) and H2O (2 mL) was added LiOH·H2O (107 mg, 2.55 mmol) at 0-10℃ , the resulting mixture was stirred at RT for 16 h. The reaction was monitored by LCMS. After completion, the reaction mixture was acidified to pH = 3-4 with 2 N HCl. The resulting mix-ture was extracted with EtOAc (30 mL x 2) , the combined organic phase was washed with saturated NaCl (50 mL) , then dried over Na2SO4 and concentrated to give (S) -2- ( (S) -3- ( (S) -4- (2- (5- (benzyloxy) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -1-ethyl-3- (3-hydroxy-2, 2-dimet hylpro-pyl) -1H-indol-5-yl) morpholin-2-yl) -2- ( (tert-butoxycarbonyl) amino) propanoyl) -2, 3-diazabicyclo [3.1.1] hept ane-4-carboxylic acid (700 mg, 95 %) as a light yellow solid.
LCMS (ESI) calcd. for C48H64N6O9 [M+H] + m/z 869.5, found: 870.6.
Step 5: Synthesis of tert-butyl ( (22S, 64S, 4S) -12- (5- (benzyloxy) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H -8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) c arbamate
To a solution of TCFH (905 mg, 3.2 mmol) and NMI (528 mg, 6.4 mmol) in ACN (100 mL) was added a solution of (S) -2- ( (S) -3- ( (S) -4- (2- (5- (benzyloxy) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -1-ethyl-3- (3-hydroxy-2, 2-dimet hylpro-pyl) -1H-indol-5-yl) morpholin-2-yl) -2- ( (tert-butoxycarbonyl) amino) propanoyl) -2, 3-diazabicyclo [3.1.1] hept ane-4-carboxylic acid (700 mg, 0.8 mmol) in ACN (40 mL) at 0-10℃, the reaction was gradually im-proved temperature to room temperature and stirred for 2 h. The reaction was monitored by LCMS. After completion, the reaction was concentrated under reduced pressure to give crude product which was puri-fied by silica gel column (eluting with EtOAc/PE from 0%to 100%to afford tert-butyl ( (22S, 64S, 4S) -12- (5- (benzyloxy) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11 H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (310 mg, 45 %) as a white solid.
LCMS (ESI) calcd. for C48H62N6O8 [M+H] + m/z 851.5, found: 852.4.
Step 6: Synthesis of tert-butyl ( (22S, 64S, 4S) -11-ethyl-12- (5-hydroxy-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carba mate
To a stirred solution of tert-butyl ( (22S, 64S, 4S) -12- (5- (benzyloxy) -2- ( (S) -1-methoxyethyl) pyridin-3-yl) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11 H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate (310 mg, 0.36 mmol) in MeOH (10 mL) was added Pd/C (10%, 31 mg) at rt , the resulting mix-ture was stirred at RT for 12 h. The reaction was monitored by LCMS. After completion, the reaction mix-ture was filtered and the filtrate was concentrated to give crude tert-butyl ( (22S, 64S, 4S) -11-ethyl-12- (5-hydroxy-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8 -oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carb amate (270 mg) as a light yellow solid.
LCMS (ESI) calcd. for C41H56N6O8 [M+H] + m/z 761.4, found: 762.5.
Step 7: Synthesis of 5- ( (22S, 64S, 4S) -4- ( (tert-butoxycarbonyl) amino) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyeth yl) pyridin-3-yl trifluoromethanesulfonate
To a solution of crude tert-butyl ( (22S, 64S, 4S) -11-ethyl-12- (5-hydroxy-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8 -oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carb amate (270 mg, 0.35 mmol) in DCM (5 mL) were added DIEA (137 mg, 1.06 mmol) and PhN (Tf) 2 (152 mg, 0.43 mmol) at 0-10℃. The resulting mixture was stirred at rt for 2 h. The reaction was monitored by LCMS. After completion, the reaction mixture was concentrated under reduced pressure to obtain crude product which was purified by silica gel column eluting with (EtOAc/PE from 0%to 100%) to afford 5- ( (22S, 64S, 4S) -4- ( (tert-butoxycarbonyl) amino) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyet hyl) pyridin-3-yl trifluoromethanesulfonate (290 mg, 91 %) as a white foam.
LCMS (ESI) calcd. for C42H55F3N6O10S [M+H] + m/z 893.4, found: 894.3.
Step 8: Synthesis of 5- ( (22S, 64S, 4S) -4-amino-11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3 ) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl trifluo-romethanesulfonate
To a stirred solution of 5- ( (22S, 64S, 4S) -4- ( (tert-butoxycarbonyl) amino) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyet hyl) pyridin-3-yl trifluoromethanesulfonate (80 mg, 0.09 mmol) in dichloromethane (3 mL) was added TFA (1 mL) dropwise at rt, the resulting mixture was stirred at rt for 1 h. The reaction mixture was washed with sat. NaHCO3 (3 mL) , brine (3 mL) , then dried over Na2SO4, following with concentration under reduced pressure to obtain crude product 5- ( (22S, 64S, 4S) -4-amino-11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl tri-fluoromethanesulfonate (91 mg, crude) as a yellow solid.
LCMS (ESI) calcd. for C37H47F3N6O8S [M+H] + m/z 793.3, found: 794.3.
Step 9: Synthesis of tert-butyl (5S) -7- ( (2S) -1- ( ( (22S, 64S, 4S) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( ( (trifluoromethyl) sulfonyl) oxy) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) -2, 7-diazaspiro [4.4] nonane-2-carboxy late
To a solution of (S) -2- ( (S) -7- (tert-butoxycarbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -3-methylbutanoic acid (44 mg, 0.13 mmol) in DMF (2 mL) were added HATU (51 mg, 0.13 mmol) and DIEA (58 mg, 0.45 mmol) at 0-10 ℃. The resulting mixture was stirred at 0-10℃ for 10 min. Then 5- ( (22S, 64S, 4S) -4-amino-11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl tri-fluoromethanesulfonate (71 mg, 0.89 mmol) was added into the reaction mixture, the resulting mixture was stirred at rt for 2 h. The reaction was monitored by LCMS. After completion, the reaction mixture was di-luted with brine and extracted with EtOAc (2 x 30 mL) , the combined organic phase was washed with sat-urated NaCl (100 mL) , then dried over Na2SO4, following with concentration under reduced pressure to obtain crude product which was purified by silica gel column eluting with EtOAc/PE from 0%to 100%to afford tert-butyl (5S) -7- ( (2S) -1- ( ( (22S, 64S, 4S) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( ( (trifluoromethyl) sulfonyl) oxy) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) -2, 7-diazaspiro [4.4] nonane-2-carbox ylate (70 mg, 71 %) as a light yellow solid.
LCMS (ESI) calcd. for C54H75F3N8O11S [M+H] + m/z 1101.5, found: 1102.4.
Step 10: Synthesis of 5- ( (22S, 64S, 4S) -11-ethyl-10, 10-dimethyl-4- ( (S) -3-methyl-2- ( (S) -2, 7-diazaspiro [4.4] nonan-2-yl) butanamido) -5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl trifluoromethanesulfonate
To a stirred solution of tert-butyl (5S) -7- ( (2S) -1- ( ( (22S, 64S, 4S) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( ( (trifluoromethyl) sulfonyl) oxy) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3. 1.1] heptanacycloundecaphane-4-yl) amino) -3-methyl-1-oxobutan-2-yl) -2, 7-diazaspiro [4.4] nonane-2-carbox ylate (70 mg, 0.06 mmol) in dichloromethane (3 mL) was added TFA (1 mL) dropwise at rt, the resulting mixture was stirred at rt for 1 h. The reaction mixture was washed with sat. NaHCO3 (3 mL) , brine (3 mL) , then dried over Na2SO4, following with concentration under reduced pressure to obtain crude product 5- ( (22S, 64S, 4S) -11-ethyl-10, 10-dimethyl-4- ( (S) -3-methyl-2- ( (S) -2, 7-diazaspiro [4.4] nonan-2-yl) butanamido ) -5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl trifluoromethanesulfonate (63 mg, crude) as a yellow solid.
LCMS (ESI) calcd. for C49H67F3N8O9S [M+H] + m/z 1001.5, found: 1002.3.
Step 11: Synthesis of (2S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (22 S, 64S, 4S) -11-ethyl-12- (5-hydroxy-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -3-methylbutanamide trifluoromethanesulfonate
To a solution of lithium (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carboxylate (19 mg, 0.13 mmol) in DMF (2 mL) were added HATU (29 mg, 0.08 mmol) and DIEA (81 mg, 0.63 mmol) at 0-10℃. The re-sulting mixture was stirred at 0-10℃ for 10 min. Then 5- ( (22S, 64S, 4S) -11-ethyl-10, 10-dimethyl-4- ( (S) -3-methyl-2- ( (S) -2, 7-diazaspiro [4.4] nonan-2-yl) butanamido ) -5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl trifluoromethanesulfonate (63 mg, 0.06 mmol) was added into the reaction mixture, the resulting mixture was stirred at rt for 2 h. The reaction was monitored by LCMS. After completion, the reaction mixture was diluted with brine and extracted with EtOAc (2 x 30 mL) , the combined organic phase was washed with saturated NaCl (100 mL) , then dried over Na2SO4, fol-lowing with concentration under reduced pressure to obtain crude product which was purified by silica gel column eluting with EtOAc/PE from 0%to 100%to afford tert-butyl (2S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (2 2S, 64S, 4S) -11-ethyl-12- (5-hydroxy-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -3-met hylbutanamide trifluoromethanesulfonate (25 mg, 31 %) as a light yellow solid.
LCMS (ESI) calcd. for C56H76F3N9O10S [M+H] + m/z 1124.5, found: 1125.4.
Step 12: Synthesis of (2S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (22 S, 64S, 4S) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (pyrimidin-5-ylethynyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -3-methylbutanamide
A mixture of (2S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (2 2S, 64S, 4S) -11-ethyl-12- (5-hydroxy-2- ( (S) -1-methoxyethyl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -3-met hylbutanamide trifluoromethanesulfonate (25 mg, 0.022 mmol) , 5-ethynylpyrimidine (4.6 mg, 0.044 mmol) , CuI (0.6 mg, 0.003 mmol) and Pd (PPh3) 4 (3.9 mg, 0.003 mmol) in TEA/DMF (0.5 mL/2 mL) was stirred at 100℃ for 1 h under N2 atmosphere, the reaction was monitored by LCMS. After completion, the mixture was filtered and the filtrate was concentrated, the residue was purified by pre-HPLC (ACN/H2O (0.5 %NH4HCO3) from 20%to 95%in 30 min) to give (2S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (2 2S, 64S, 4S) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (pyrimidin-5-ylethynyl) pyridin-3-yl) -10, 10-dimethyl-5, 7 -dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -3-methylbutanamide (3 mg, 12.5%) as a white solid.
LCMS (ESI) calcd. for C61H79N11O7 [M+H] + m/z 1078.6, found: 1079.4.
1H NMR (500 MHz, CDCl3) δ 9.15 (t, J = 1.7 Hz, 1H) , 8.79 (d, J = 1.8 Hz, 2H) , 8.36 (d, J = 12.1 Hz, 1H) , 8.31 (d, J = 2.0 Hz, 1H) , 8.04 (d, J = 2.0 Hz, 1H) , 7.36 (d, J = 7.9 Hz, 1H) , 7.21 (d, J = 2.2 Hz, 1H) , 7.01 (dd, J = 7.9, 2.2 Hz, 1H) , 5.24 (q, J = 5.4 Hz, 1H) , 4.90 (d, J = 8.6 Hz, 1H) , 4.47 (q, J = 11.9 Hz, 1H) , 4.15 (q, J = 4.4 Hz, 2H) , 4.12 -4.06 (m, 1H) , 4.06 -4.03 (m, 2H) , 3.99 -3.89 (m, 2H) , 3.89 -3.82 (m, 2H) , 3.82 -3.78 (m, 1H) , 3.68 (d, J = 6.0 Hz, 1H) , 3.65 -3.55 (m, 5H) , 3.45 (dd, J = 7.4, 4.8 Hz, 1H) , 3.37 (d, J = 1.3 Hz, 2H) , 3.33 (s, 3H) , 3.09 (s, 2H) , 2.99 -2.92 (m, 1H) , 2.88 -2.79 (m, 3H) , 2.79 -2.66 (m, 2H) , 2.41 (t, J = 1.5 Hz, 3H) , 2.25 -1.91 (m, 9H) , 1.89 -1.81 (m, 3H) , 1.67 (d, J = 5.3 Hz, 3H) , 1.37 (t, J = 4.4 Hz, 3H) , 1.21 (dd, J = 7.9, 4.9 Hz, 2H) , 1.15 (s, 3H) , 1.10 (s, 3H) , 1.05 (dd, J = 7.6, 5.0 Hz, 2H) , 0.95 (d, J = 5.7 Hz, 3H) , 0.90 (d, J = 5.5 Hz, 3H) .
Example 89
(2S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (63S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dime-thyl-5, 7-dioxo-61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (1, 3) -pyridazinacy cloundecaphane-4-yl) -3-methylbutanamide
Substituting tert-butyl ( (64S, 4S, Z) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa -62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate with tert-butyl ( (63S, 4S, Z) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (1, 3) -pyridazinacycloundecaphane-4-yl) carbamate in the Step 1 of Example 90, the title compound was prepared by the same procedures as described for Example 90.
Example 90
(2S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N - ( (64S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dime-thyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -3-methylbutanamide
Step 1: Synthesis of (2S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (64 S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -3-methylbutanamide
To a stirred solution of 5- ( (64S, 4S, Z) -4- ( (2S) -2- (7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -3-methylbutanamido) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-12-yl) -6- ( (S) -1-methoxyethyl) pyridin-3-yl tri-fluoromethanesulfonate (80 mg, 0.071 mmol) and 4- (prop-2-yn-1-yl) morpholine (18 mg, 0.14 mmol) , CuI (2 mg, 0.011 mmol) , TEA (22 mg, 0.21 mmol) and Pd (PPh3) 4 (12 mg, 0.011 mmol) in DMF (2 mL) was stirred at 90℃ for 1 h under N2 atmosphere, the reaction was monitored by LCMS. After completion, the mixture was filtered and the filtrate was concentrated, the residue was purified by pre-HPLC (eluting with CH3CN/H2O (0.1%NH4HCO3) from 20%to 70%) to give (2S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (6 4S, 4S, Z) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl -5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -3-methylbutanamide (19.1mg) as a white solid.
LCMS (ESI) calcd. for C61H80N10O7S [M+H] + m/z 1097.6, found: 1098.4.
1H NMR (400 MHz, DMSO-d6) δ 8.79 (s, 1H) , 8.40 (s, 1H) , 8.35 -8.18 (m, 1H) , 7.86 (s, 1H) , 7.75 (d, J = 8.3 Hz, 1H) , 7.58 (d, J = 8.5 Hz, 1H) , 5.93 (s, 1H) , 5.32 (s, 1H) , 4.73 (d, J = 10.2 Hz, 1H) , 4.49 (s, 1H) , 4.37 -4.20 (m, 2H) , 4.07 (d, J = 7.3 Hz, 1H) , 3.53 (dd, J = 26.3, 13.2 Hz, 5H) , 3.25 (s, 3H) , 2.88 -2.69 (m, 3H) , 2.67 -2.50 (m, 4H) , 2.36 (d, J = 14.5 Hz, 2H) , 2.27 -2.09 (m, 3H) , 2.02 -1.76 (m, 3H) , 1.66 (dd, J =33.9, 16.1 Hz, 3H) , 1.34 (d, J = 5.8 Hz, 3H) , 1.21 (s, 1H) , 0.99 -0.79 (m, 7H) , 0.58 (s, 1H) , 0.31 (d, J = 17.6 Hz, 3H) , 0.14 (s, 1H) .
Example 91
(2S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (22S, 63S, 4S) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dime-thyl-5, 7-dioxo-61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (1, 3) -pyridazin acycloundecaphane-4-yl) -3-methylbutanamide
Substituting tert-butyl ( (64S, 4S, Z) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa -62, 63-diaza-2 (4, 2) -thiazola-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carbamate with tert-butyl ( (22S, 63S, 4S) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (1, 3) -pyridazinacycloundecaphane-4-yl) c arbamate in the Step 1 of Example 90, the title compound was prepared by the same procedures as de-scribed for Example 90.
Example 92
(2S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N - ( (22S, 63S, 4S) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( (R) -3- (4-methylpiperazin-1-yl) but-1-yn-1-yl) pyri din-3-yl) -10, 10-dimethyl-5, 7-dioxo-61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -morpholina-1 (5, 3) -in dola-6 (1, 3) -pyridazinacycloundecaphane-4-yl) -3-methylbutanamide
Substituting 4- (prop-2-yn-1-yl) morpholine with (R) -1- (but-3-yn-2-yl) -4-methylpiperazine in the Step 1 of Example 91, the title compound was prepared by the same procedures as described for Example 91.
Example 93
(2S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (22S, 64S, 4S) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( (R) -3- (4-methylpiperazin-1-yl) but-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) -3-methylbutanamide
Substituting tert-butyl ( (22S, 63S, 4S) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (1, 3) -pyridazinacycloundecaphane-4-yl) carbamate with tert-butyl ( (22S, 64S, 4S) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carba mate in the Step 1 of Example 92, the title compound was prepared by the same procedures as described for Example 92.
Example 94
(2S) -2-cyclopentyl-2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (22S, 64S, 4S) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( (R) -3- (4-methylpiperazin-1-yl) but-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) acetamide
Substituting (S) -2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -3-meth ylbutanoic acid with (S) -2-cyclopentyl-2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) acetic acid in the Step 1 of Example 93, the title compound was prepared by the same procedures as described for Example 93.
Example 95
(2S) -2-cyclopentyl-2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (22S, 64S, 4S) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) acetamide
Substituting (R) -1- (but-3-yn-2-yl) -4-methylpiperazine with 4- (prop-2-yn-1-yl) morpholine in the Step 1 of Example 94, the title compound was prepared by the same procedures as described for Example 94.
Example 96
(2S) -2-cyclopentyl-2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (22S, 63S, 4S) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- (3-morpholinoprop-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (1, 3) -pyridazinacycloundecaphane-4-yl) acetamide
Substituting tert-butyl ( (22S, 64S, 4S) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -11H-8-oxa-62, 63-diaza-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (2, 4) -bicyclo [3.1.1] heptanacycloundecaphane-4-yl) carba mate with tert-butyl ( (22S, 63S, 4S) -11-ethyl-10, 10-dimethyl-5, 7-dioxo-12- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (1, 3) -pyridazinacycloundecaphane-4-yl) carbamate in the Step 1 of Example 95, the title compound was prepared by the same procedures as de-scribed for Example 95.
Example 97
(2S) -2-cyclopentyl-2- ( (S) -7- ( (2R, 3R) -3-cyclopropyl-1-methylaziridine-2-carbonyl) -2, 7-diazaspiro [4.4] nonan-2-yl) -N- ( (22S, 63S, 4S) -11-ethyl-12- (2- ( (S) -1-methoxyethyl) -5- ( (R) -3- (4-methylpiperazin-1-yl) but-1-yn-1-yl) pyridin-3-yl) -10, 10-dimethyl-5, 7-dioxo-61, 62, 63, 64, 65, 66-hexahydro-11H-8-oxa-2 (4, 2) -morpholina-1 (5, 3) -indola-6 (1, 3) -pyridazinacycloundecaphane-4-yl) acetamide
Substituting 4- (prop-2-yn-1-yl) morpholine with (R) -1- (but-3-yn-2-yl) -4-methylpiperazine in the Step 1 of Example 96, the title compound was prepared by the same procedures as described for Example 96.
The following exemplary compounds was synthesized from different starting materials according to the method above (such as method for Example 1-30)
Table 4A. Exemplary Compounds Prepared by Methods of the Present Invention
Note: values may differ slightly from values found elsewhere in this application due to different measure-ments and rounding.
BIOLOGICAL ACTIVITY RESULTS
The following were used as reference compounds.
Ras: RAF1 HTRF Assay
This example is to verify the binding between RAS protein and its interacting protein (SOS1 or RAF1) . When RAS exists with SOS1 or RAF, the two interact to make the energy donor and receptor bound to it close enough, and then when the donor is excited by an external light source, energy resonance transfer occurs, thereby exciting the receptor and emitting a specific wavelength of emitted light, the FRET signal is detected.
Specific experimental methods: 20 nM purified RAS protein (e.g. KRAS-G12C, G12D, G12V, G12S, WT) was incubated with the drug to be screened (the drug was diluted in a 3-fold gradient to a final con-centration of 1000 nM) at room temperature for 1 h. Subsequently, 30 nM purified SOS1-Cat was added and incubated at room temperature for 0.5-1 h. After adding the detection antibody (PerkinElmer, Cisbio, HTRF antibody) , the value was measured according to the manufacturer's instructions.
The representative compound results
The reference compound (Ref. 1) and the representative Compound A (Example 16) significantly in-hibited the interaction of RAF1 with active RASMutant protein or RASWT protein, and the result is shown in Figure 1.
The inhibitory IC50 of the compounds of the examples of the present invention was shown in Table 5.
Table 5. RAS-GTP &Raf Disruption/HTRF Assay
*Key: A: 1 nM <IC50 ≤ 10 nM; B: 10 nM < IC50 ≤ 100 nM; C: 100 nM < IC50 ≤ 1000 nM; D: IC50 >1000 nM; E: not tested.
Cell viability assay
Selective screening using RAS-driven and RAS-non-driven cells: (1) RAS-driven cells, including cells with persistent RAS activation due to RAS mutations and upstream EGFR activation. Cells that are continuously activated due to RAS mutations, such as KRAS G12D (AsPC-1, HPAC, AGS, etc. ) mutant cells, KRAS G12C (NCI-H358, MIAPaCa-2, etc. ) KRAS G13D (LOVO, HCT15, etc. ) mutant cells, etc. Cells with continuous activation of RAS due to EGFR mutations, such as EGFREx19del (PC-9, etc. ) mu-tant cells, EGFRT790M/L858R (NCI-H1975, etc. ) mutant cells, etc.; (2) Non-RAS driven cells, including BRAFV600E (A375, SK, ) -MEL-3, etc. ) mutant cells, RASWT (BxPC-3, etc. ) cells, etc.
Specific experimental methods: The above cells were seeded in a 96-well plate at a density of 1000-10000 per well. Drugs were diluted in DMSO to make a 10 mM stock solution. Subsequently, the drug was diluted in a 3-fold gradient to a final concentration of 1000 nM. The cells were treated with the drug for 5 days. Cell viability was then detected by the MTS method.
The representative compound results
The reference compound (Ref. 1) and the representative Compound A (Example 16) significantly in-hibited the cell viability of KRAS mutant cells: MIA PACA-2 (KRAS G12C) , NCI-H358 (KRAS G12C) , NCI-H2122 (KRAS G12C) HPAF II (KRAS G12D) , AGS (KRAS G12D) , ASPC-1 (KRAS G12D) , HPAC (KRAS G12D) , LS174T (KRAS G12D) , SW480 (KRAS G12V) , SW620 (KRAS G12V) , CAL-62 (KRAS G12R) , LOVO (KRAS G13D) Calu6 (KRAS Q61K) and HCC827 (EGFR) . The result is shown in Figure 2.
The inhibitory IC50 of the compounds of the examples of the present invention was shown in Table 6.
Table 6. Activity of Compounds of present invention in KRAS Cell viability assay
*Key: AA: IC50 ≤ 1 nM; A: 1 nM <IC50 ≤ 10 nM; B: 10 nM < IC50 ≤ 100 nM; C: 100 nM < IC50 ≤1000 nM; D: IC50 > 1000 nM; E: not tested.
Anti-proliferation IC50 Values of each cell line
Methods: The purpose of this cellular assay was to determine the effects of test compounds on the proliferation of human cancer cell lines NCI-H358 cells, AGS cells, HPAC cells, AsPC-1 cells, SW620 cells, and A375 cells over a 5-day treatment using Cell Counting Kit-8 (CCK8) . Cells were seeded at 2000 cells/well (NCI-H358) , 1000 cells/well (AGS) , 1500 cells/well (HAPC) , 3000 cells/well (AsPC-1) , 2000 cells/well (SW620) , 2000 cells/well (A375) in 96-well assay plates and incubated overnight. On the day of the assay, diluted compounds were then added in a final concentration of 0.5%DMSO. After 5 days incu-bation, a tenth of the volume of cell counting kit 8 (Dnjindo-CK04) was added into each well. Read the signal (OD450) using Biotek synergy H1 plate reader after incubation. IC50 was determined by Graphpad 8.0.
Table 6A. Anti-proliferation IC50 Values of reference compounds and Optimized compounds in each cell line
Results: Results from in vitro cell proliferation assays showed that Example 16, Example 17 and Example 18 exhibited a more potent inhibitory effect on cell growth compared to Ref. 1 and Ref. 2. Besides, the activity of Example 16 was more potent than Ref. 3; the activity of Example 17 was more potent than Ref. 4
In vivo efficacy and PD studies
After multiple batches of cell screening and activity testing, Compound A and B (Example 16 and Example 17) were selected for in vitro efficacy experiments with reference compounds Ref. 1 and Ref. 2 in this disclosure, and the differences and advantages of the disclosed compounds and clinical compounds were further compared.
Experimental Animal Models 1: Ref. 1 and Compound A in PDAC Tumor (HPACG12D)
Methods: Subcutaneous inoculation of 3 × 106 HPAC cells in 0.1 mL DPBS (Dulbecco‘s Phos-phate-Buffered Saline) was performed in the right cervical dorsal region of each Balb/c nude mice. Ex-perimental animals were marked with ear tag numbers during inoculation as the sole identification for subsequent experiments. The in vivo efficacy study commenced with group administration on day 20 post-cell inoculation, with 5 mice per group receiving continuous treatment for 21 days. Tumor volume and body weight were measured twice weekly. T/C %= T/C × 100; TGI (%) = [1- (Treatending day -Treat-grouping day) / (Vehicleending day -Vehiclegrouping day) ] × 100.
Results: On day 21 drug administration, tumor-bearing mice in the vehicle group exhibited a mean tumor volume of 723 mm3. Significant antitumor efficacy with statistical differences was observed in the following treatment groups: Ref. 1 5 mg/kg (tumor volume = 183 mm3, TGI = 89.32%, T/C = 25.37%, p <0.0001) ; Compound A 0.1 mg/kg (tumor volume = 167 mm3, TGI = 92.02%, T/C = 23.10%, p < 0.0001) , Compound A 0.5 mg/kg (tumor volume = 26 mm3, TGI = 115.38%, T/C = 3.53%, p < 0.0001) , Compound A 1 mg/kg (tumor volume = 20 mm3, TGI = 116.27%, T/C = 2.81%, p < 0.0001) . Notably, Compound A demonstrated a clear dose-dependent relationship in tumor growth inhibition. The result is shown in Figure 3.
Experimental Animal Models 2: Ref. 2 and Compound A in PDAC Tumor (PK59G12D)
Methods: Subcutaneous inoculation of 5 × 106 PK59 cells in 0.1 mL DPBS (Dulbecco's Phos-phate-Buffered Saline) was performed in the right cervical dorsal region of each Balb/c nude mice. Ex-perimental animals were marked with ear tag numbers during inoculation as the sole identification for subsequent experiments. The in vivo efficacy study commenced with group administration on day 17 post-cell inoculation, with 5 mice per group receiving continuous treatment for 21 days. Tumor volume and body weight were measured twice weekly. T/C %= T/C × 100; TGI (%) = [1- (Treatending day -Treat-grouping day) / (Vehicleending day -Vehiclegrouping day) ] × 100.
Results: On day 21 drug administration, tumor-bearing mice in the vehicle group exhibited a mean tumor volume of 915 mm3. Significant antitumor efficacy with statistical differences was observed in the following treatment groups: Ref. 2 0.1 mg/kg (tumor volume = 224 mm3, TGI = 86.68%, T/C = 24.46%, p < 0.0001) ; Compound A 0.02 mg/kg (tumor volume = 303 mm3, TGI = 76.82%, T/C = 33.05%, p <0.0001) , Compound A 0.05 mg/kg (tumor volume = 110 mm3, TGI = 100.92%, T/C = 12.05%, p < 0.0001) , Compound A 0.1 mg/kg (tumor volume = 16 mm3, TGI = 112.79%, T/C = 1.70%, p < 0.0001) , Compound A 0.3 mg/kg (tumor volume = 7 mm3, TGI = 113.82%, T/C = 0.81%, p < 0.0001) . Notably, Compound A demonstrated a clear dose-dependent relationship in tumor growth inhibition. The result is shown in Figure 4.
Experimental Animal Models 3: Ref. 1, Ref. 2, Compound A and Compound B in PDAC Tumor (PK59G12D)
Methods: Subcutaneous inoculation of 5 × 106 HPAC cells in 0.1 mL DPBS (Dulbecco's Phos-phate-Buffered Saline) mixed with 50%Matrigel was performed in the right cervical dorsal region of each Balb/c nude mice. Experimental animals were marked with ear tag numbers during inoculation as the sole identification for subsequent experiments. The in vivo efficacy study commenced with group administra-tion on day 17 post-cell inoculation, with 5 mice per group receiving continuous treatment for 23 days. Tumor volume and body weight were measured twice weekly. T/C %= T/C × 100; TGI (%) =[1- (Treatending day -Treatgrouping day) / (Vehicleending day -Vehiclegrouping day) ] × 100.
Results: On day 23 drug administration, tumor-bearing mice in the vehicle group exhibited a mean tumor volume of 1678 mm3. Significant antitumor efficacy with was observed across treatment groups ex-cept for Ref. 2 0.1 mg/kg (tumor volume = 1495 mm3, TGI = 11.93%) : Ref. 1 5 mg/kg (tumor volume = 674 mm3, TGI = 65.12%) ; Ref. 1 25 mg/kg (tumor volume = 50 mm3, TGI = 105.49%) ; , Ref. 2 0.3 mg/kg (tumor volume = 527 mm3, TGI = 74.51%) , Compound A 0.1 mg/kg (tumor volume = 392 mm3, TGI = 83.41%) , Compound A 0.3 mg/kg (tumor volume = 70 mm3, TGI = 104.13%) , Compound B 0.1 mg/kg (tumor vol-ume = 374 mm3, TGI = 84.48%) , Compound B 0.3 mg/kg (tumor volume = 82 mm3, TGI = 103.45%) . The result is shown in Figure 5.
Experimental Animal Models 4: Ref. 1, Ref. 3, Ref. 4, Compound A and Compound B in NSCLC Tumor (LU99G12C)
Methods: Subcutaneous inoculation of 5 × 106 LU99 cells in 0.1 mL DPBS (Dulbecco's Phos-phate-Buffered Saline) mixed with 50%Matrigel was performed in the right cervical dorsal region of each Balb/c nude mice. Experimental animals were marked with ear tag numbers during inoculation as the sole identification for subsequent experiments. The in vivo efficacy study commenced with group administra-tion on day 8 post-cell inoculation, with 6 mice per group receiving continuous treatment for 14 days. Tu-mor volume and body weight were measured twice weekly. T/C %= T/C × 100; TGI (%) = [1- (Treatending day -Treatgrouping day) / (Vehicleending day -Vehiclegrouping day) ] × 100.
Results: On day 14 drug administration, tumor-bearing mice in the vehicle group exhibited a mean tumor volume of 1069 mm3. Antitumor efficacy of Compound A (TGI=32.11%, Compound A 0.01 mg/kg; TGI=109.42%, Compound A 0.05 mg/kg) was more potent than that of Ref. 3 (TGI=13.37%, Ref. 3 0.01 mg/kg; TGI=87.45%, Ref. 3 0.05 mg/kg) ; Antitumor efficacy of Compound B (TGI=57.43%, Compound B 0.05 mg/kg) was more potent than that of Ref. 4 (TGI=33.62%, Ref. 4 0.05 mg/kg) . The result is shown in Figure 6.
Brain metastasis intracranial model
The purpose of this assay is to determine whether brain exposure of the compounds of current inven-tion was sufficient to mediate regression of tumors in experimental Brain Metastases (BM) models.
Methods: Female BALB/c nude mice were anesthetized with xylazine hydrochloride (8 mg/kg) and Zoletil 50 (35 mg/kg) , receiving pre-and post-operative analgesia through butorphanol tartrate (1 mg/kg) . Following head fixation in a stereotaxic apparatus, surgical sites were disinfected with 70%ethanol and draped sterilely. A 1 cm sagittal incision exposed the parietal and occipital bones, which were cleaned with 3%hydrogen peroxide. A burr hole was drilled 2 mm right of the bregma and 0.5 mm posterior to the cor-onal suture using a dental drill. The cell suspension containing 3×10 NCI-H1373-luc cells in 3 μL DPBS with 20%Matrigel Matrix was carefully loaded into a Hamilton syringe, with external surfaces wiped with ethanol to prevent ectopic tumor formation. The needle was vertically inserted to 3 mm depth for con-trolled infusion over 1 minute (0.05 μL/sec) , followed by 1-minute dwell time before slow withdrawal. The injection site was sealed with cyanoacrylate glue, and the scalp was repositioned and sutured. Postopera-tive analgesia with meloxicam was administered for three days, with ear tags applied for identification. The efficacy study commenced on day 4 post-implantation, utilizing five mice per group for 21-day treatment.
Results: On day 21 post-grouping, tumor-bearing mice in the vehicle control group demonstrated a mean tumor bioluminescence intensity of 5.12×10 photons/sec. Significant antitumor efficacy with sta-tistical significance (p < 0.0001) was observed across treatment groups: Ref. 1 25 mg/kg (2.45×10 pho-tons/sec, TGI=113.40%, T/C=4.79%) , Compound A 0.1 mg/kg (5.68×10 photons/sec, TGI=105.86%, T/C=11.09%) , Compound A 0.5 mg/kg (8.86×10 photons/sec, TGI=117.06%, T/C=1.73%) , and Com-pound A 1 mg/kg (1.05×10 photons/sec, TGI=116.63%, T/C=2.05%) . The result is shown in Figure 7 and Figure 8.
Metabolic Stability In Vitro
The purpose of this experiment was to investigate the metabolic stability of the test compounds in liv-er microsomes and plasma of human, mouse, rat, canine and cynomolgus monkey.
Liver microsomal stability assay
The original stock solution of the test compound and the positive compound (prepared with 1 mg/mL DMSO) was diluted with DMSO to prepare a 0.25 mM working solution. General formula: mg/mL =(1000/M) mmol/L, M is the relative molecular mass of the compound. The metabolic stability of com-pounds in liver microsomes was evaluated by incubation them with liver microsomes of different species in vitro. After the chemical was incubated with liver microsomes for different times (0 min, 5 min, 15 min, 30 min, 45 min, 60 min) , the reaction was terminated by precooled acetonitrile (containing internal stand-ard) . After sample processed (protein precipitation method) , it was loaded into LC-MS for quantitative analysis of the compounds.
The elimination rate constant and clearance rate of the test compound in liver microsomes were cal-culated using the following formula:
a) Residual Rate (%) = peak area ratio of compound to internal standard at any time point/peak area ratio of compound to internal standard at 0 min*100%;
b) T1/2 = 0.693/ke, where ke is the elimination rate constant and T1/2 is the half-life;
c) Liver Microsomal Intrinsic Clearance (CLint, in vitro) = 0.693/T1/2/microsomal protein concentration (mg/mL) ;
The representative compound results
The results of liver microsome stability (Table 7) showed that the intrinsic clearance rate of most of the compounds of the present invention in different species (human, mouse, rat, dog, cynomolgus monkey) was generally lower than that of the reference compound (Ref. 1) , indicating that it had better stability in the liver microsomes of the above species.
Table 7. Liver Microsome Stability Data for Selected Compounds
*/: not tested
Plasma stability assay
The compounds were incubated with the plasma of different species (human, CD-1 mouse, SD rat, beagle dog and cynomolgus monkey) to evaluate the metabolic stability of the compounds in plasma. After the compound was incubated with plasma for different time (0 min, 3 min, 5 min, 15 min, 30 min, 60 min, 120 min) , the reaction was terminated by precooling acetonitrile (including internal standard) . The termi-nated reaction solution was purified by protein precipitation method, and the compounds were quantita-tively analyzed by LC-MS.
The results of plasma stability (Table 8) showed that the metabolism of Ref. 1 in rat plasma was rela-tively unstable (T1/2=20.66 min) , while the compound of the present invention was T1/2>120 min in rat plasma, and at the same time, the T1/2 of the compounds of the present invention in the plasma of different species (human, CD-1 mouse, SD rat, beagle dog and cynomolgus monkey) were greater than 120 min.
Notably, the T1/2 of Example 16 and Example 17 in human, mouse, rat, monkey and dog plasma were >120 min, which indicates that Example 16 and Example 17 are metabolically stable in the plasma of different species. Example 16 exhibits superior stability compared to Ref. 3 in liver microsomal metabolic systems of all species (human, CD-1 mouse, SD rat, beagle dog and cynomolgus monkey) . Example 16 exhibits superior stability compared to Ref. 2 in liver microsomal metabolic systems of human, CD-1 mouse, and cynomolgus monkey (Table 8) . Example 17 exhibits superior stability compared to Ref. 4 in liver microsomal metabolic systems of SD rats (Table 8)
In summary, it showed that the compounds of the present invention had good plasma stability of mul-tiple genera, and at the same time, compared with the reference compound (Ref. 1) , it had more excellent plasma stability of rats.
Table 8. Plasma Stability Data for Selected Compounds
*/: not tested
Pharmacokinetic study
Oral bioavailability (F) of Ref. 1, Ref. 2, Ref. 3, Ref. 4, Example 5, Example 16 and Example 17 was compared in ICR mice.
The above compounds were administered via caudal vein at a dose of 2 mg/kg, and orally at a dose of 10 mg/kg. Whole blood was collected from cheek veins at 5 min, 15 min, 30 min, 1 h, 2 h, 4 h, 8 h, 24 h, and 48 h after administration. And plasma samples were also prepared for the pharmacokinetics study of Ref. 1, Example 5, Example 16.
The established and verified LC-MS method was used to quantitatively analyze the above compounds in plasma or blood. The mobile phase system of H2O (B) and acetonitrile (D) was utilized. The compounds were eluted by HPLC gradient with a rate of 0.8 mL·min-1. And the detection of the ions was carried out in the negative ESI mode and Selected Ion Record (SIR) mode. MS conditions are as follows: Capillary volt-age, 1.5 kV; Cone voltage, 50 V; Desolvation temperature, 400℃; Desolvation Gas, 800 L/Hr. Pharmaco-kinetic parameters were calculated by WinNonlin.
The representative compound results
The plasma pharmacokinetic parameters of Ref. 1, Example 5 and Example 16 after single-dose ad-ministration in mice were shown in Table 9. After a single oral dose, T1/2, Cmax, AUC (0-t) , AUC (0-∞) and bio-availability (F) of Example 5 were 10 times, 3 times, 11 times, 10 times and 1 times of that of Ref. 1, re-spectively; T1/2, Cmax, AUC (0-t) , AUC (0-∞) and bioavailability (F) of Example 16 were 9 times, 4 times, 24 times, 23 times and 1 times of that of Ref. 1, respectively.
Compared with Ref. 1, Example 5 and Example 16 exhibited better pharmacokinetics in mice.
Table 9. Plasma PK Parameters of Compounds in ICR Mice
The boold pharmacokinetic parameters of Ref. 1, Ref. 2, Ref. 3, Ref. 4 and Example 17 after single-dose administration in mice were shown in Table 9A.
After a single vein dose, Cmax, AUC (0-∞) of Example 16 were 2.3 times, 1.2 times of those of Ref. 2, respectively. And after a single oral dose, Cmax, AUC (0-t) , AUC (0-∞) and F of Example 16 were 1.7 times, 1.9 times, 1.8 times and 1.9 times of those of Ref. 2, respectively. And after a single vein dose, T1/2 and Vd of Example 16 were 1.9 times and 2.5 times of those of Ref. 3, after a single oral dose, T1/2 were 1.9 times of that of Ref. 3. The above data indicate that Example 16 (Plasma PK, Example 16 is evenly distributed in whole blood and plasma) has a higher peak concentration, higher exposure, higher bioavailability than Ref. 2 and a longer elimination half-life than Ref. 3 after a single dose administration in mice.
After a single vein dose, T1/2, Cmax, AUC (0-t) , AUC (0-∞) of Example 17 were 1.8 times, 1.5 times, 2.2 times and 2.3 times of those of Ref. 4; And after a single oral dose, T1/2, Cmax, AUC (0-t) , AUC (0-∞) and F of Example 17 were 2.1 times, 1.1 times, 2.7 times, 2.8 times and 1.2 times of those of Ref. 4. The above data indicate that Example 17 has a higher peak concentration, higher exposure, higher bioavailability and a longer elimination half-life than Ref. 4.
Table 9A. The Blood Pharmacokinetic Parameters of Compounds after Single-dose Administration in Mice.
All literatures mentioned in the present application are incorporated herein by reference, as though each one is individually incorporated by reference. Additionally, it should be understood that after reading the above teachings, those skilled in the art can make various changes and modifications to the present in-vention.These equivalents also fall within the scope defined by the appended claims.
Claims (21)
- A compound, or a stereoisomer thereof, a pharmaceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt thereof, wherein the compound is a compound of formula (I) ;
whereinn is 0, 1, 2, or 3;A is optionally substituted 5 to 6-membered heterocycloalkylene, optionally substituted 5 to 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene;B is selected from optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 3 to 6-membered heterocycloalkenylene, optionally substituted 4 to 11-membered bicyclic cycloalkylene, or optionally substituted 4 to 11-membered bicyclic heterocycloalkylene;G is optionally substituted C1-6 alkylene;R8 and R9 are each independently selected from hydrogen, deuterium, halogen, hydroxy, cyano, op-tionally substituted C1-3 alkyl, or R8 and R9 combine with the atoms to which they are attached to form an optionally substituted C3-6 cycloalkyl or a carbonyl;R10 and R11 are each independently selected from hydrogen, deuterium, halogen, hydroxy, cyano, op-tionally substituted C1-3 alkyl, or R10 and R11 combine with the atoms to which they are attached to form an optionally substituted C3-6 cycloalkyl or a carbonyl;R12 and R13 are each independently selected from hydrogen, deuterium, halogen, hydroxy, optionally substituted C1-3 alkyl, -O (C1-6 alkyl) , -S (C1-6 alkyl) or -N (C1-6 alkyl) (C1-6 alkyl) ; furthermore, the C1-6 alkyl may be further optionally substituted;RA is optionally substituted C3-6 cycloalkyl, optionally substituted C3-6 heterocycloalkyl (or 4 to 7-membered heterocycloalkyl) , optionally substituted 5 to 8-membered aryl, optionally substituted 5 to 8-membered heteroaryl, optionally substituted 8 to 10-membered fused bicyclic aryl or optionally substi-tuted 8 to 10-membered fused bicyclic heteroaryl;RB is selected from hydrogen, C1-6 alkyl, C1-6 alkoxyl, C1-6 haloalkyl, C1-6 haloalkoxyl, optionally sub-stituted C3-6 cycloalkyl, or optionally substituted C3-6 heterocycloalkyl (or 4 to 7-membered heterocycloal-kyl) ; or provided that when n exceeds 1, any two of RB combine with the atoms to which they are attached to form a 3 to 6-membered ring, wherein the ring can be optionally substituted with halogen, hydroxy, and C1-3 alkyl;X1 is N or C;X2 is N or -CRa-;X3 is N or -CRb-;X4 is N or -CRc-;X5 is N or C;X6 is S, O, N, or -CH-;Ra, Rb, and Rc are each independently hydrogen, halogen, cyano, C1-3 alkyl, C1-3 alkoxyl, C1-3 haloal-kyl, C1-3 haloalkoxyl, C3-6 cycloalkyl, or C3-6 heterocycloalkyl (or 4 to 7-membered heterocycloalkyl) ;R3 is absent, or selected from hydrogen, C1-6 alkyl, or C1-6 haloalkyl, provided that when X6 is O or S, R3 is absent;E is a bond, or selected from N, or -CRd-, wherein Rd is selected from hydrogen, halogen, hydroxy, cyano, carboxyl, optionally substituted C1-6 alkyl, optionally substituted C1-6 alkoxyl, C1-6 aminoalkyl, C1-6 hydroxyalkyl, or -NReRf; wherein Re and Rf are independently selected from hydrogen, or optionally sub-stituted C1-6 alkyl;R4 is hydrogen, optionally substituted C1-6 alkyl, optionally substituted C1-6 heteroalkyl, optionally substituted C3-10 cycloalkyl, optionally substituted C3-10 cycloalkenyl, optionally substituted C3-10 heterocy-cloalkyl (or 4 to 12-membered heterocycloalkyl) , optionally substituted 6 to 10-membered aryl, optionally substituted 5 to 10-membered heteroaryl, orR5 is hydrogen, C1-6 alkyl or C3-6 cycloalkyl, furthermore, the C1-6 alkyl or C3-6 cycloalkyl may be fur-ther optionally substituted;or R4 and R5 combine with the atoms to which they are attached to form a ring, wherein the ring is se-lected from optionally substituted 3 to 10-membered cycloalkyl, optionally substituted 3 to 10-membered heterocycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 4 to 11-membered bicyclic cycloalkyl, optionally substituted 4 to 11-membered bicyclic heterocycloalkyl;whereinL is absent, or selected from -CH2-, -C (O) -, -CHRg-or -C (Rg) 2-, wherein Rg is optionally substituted C1-6 alkyl;R6 is hydrogen, or optionally substituted C1-6 alkyl;R7 is optionally substituted C1-6 alkyl, optionally substituted C1-6 heteroalkyl, optionally substituted C3-10 cycloalkyl, optionally substituted C3-10 heterocycloalkyl (or 3 to 10-membered heterocycloalkyl) , op-tionally substituted 6 to 10-membered aryl, optionally substituted 5 to 10-membered heteroaryl;or L, R6 and R7 combine with the atoms to which they are attached to form a ring, wherein the ring is selected from optionally substituted 3 to 10-membered cycloalkyl, optionally substituted 3 to 10-membered heterocycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 4 to 11-membered bicyclic cycloalkyl, optionally substituted 4 to 11-membered bicyclic heterocycloalkyl. - The compound according to claim 1, or a stereoisomer thereof, a pharmaceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically ac-ceptable salt thereof, wherein the compound of formula (I) is a compound of formula (I-1)
wherein X1, X2, X3, X4, X5, X6, A, B, E, G, RA, RB, R3, R4, R5 and n are as defined in claim 1. - The compound according to claim 1, or a stereoisomer thereof, a pharmaceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically ac-ceptable salt thereof, whereinB is optionally substitutedandRA iswhereinR1 is selected from the group consisting of hydrogen, C1-6 alkyl, C1-6 alkoxyl, C1-6 haloalkyl, C1-6 haloalkoxyl, C1-6 hydroxyalkyl, C1-6 aminoalkyl, optionally substituted C3-6 cycloalkyl, optionally substi-tuted C3-6 heterocycloalkyl, optionally substituted 5 to 8-membered aryl, optionally substituted 5 to 8-membered heteroaryl, optionally substituted 8 to 10-membered fused bicyclic aryl, optionally substituted 8 to 10-membered fused bicyclic heteroaryl, optionally substituted -C1-2 alkylene-C3-6 cycloalkyl, optionally substituted -C1-2 alkylene-C3-6 heterocycloalkyl, optionally substituted -C1-2 alkylene-5 to 8-membered aryl, optionally substituted -C1-2 alkylene-5 to 8-membered heteroaryl, optionally substituted -C1-2 alkylene-8 to 10-membered fused bicyclic aryl, and optionally substituted -C1-2 alkylene-8 to 10-membered fused bicy-clic heteroaryl; furthermore, the C1-6 alkyl, C1-6 alkoxyl, C1-6 haloalkyl, C1-6 haloalkoxyl, C1-6 hydroxyalkyl may be further optionally substituted;in R1, when the C1-6 alkyl, C1-6 alkoxyl, C1-6 haloalkyl, C1-6 haloalkoxyl, or C1-6 hydroxyalkyl is further substituted, the substituent is selected from the group consisting of C1-6 alkyl, C1-6 alkoxyl, C1-6 haloalkyl, C1-6 haloalkoxyl, C1-6 hydroxyalkyl, C1-6 aminoalkyl, optionally substituted C3-6 cycloalkyl, optionally sub-stituted C3-6 heterocycloalkyl, optionally substituted 5 to 8-membered aryl, optionally substituted 5 to 8-membered heteroaryl, optionally substituted 8 to 10-membered fused bicyclic aryl, optionally substituted 8 to 10-membered fused bicyclic heteroaryl; preferably, the substituent is selected from the group consist-ing of optionally substituted C3-6 cycloalkyl, optionally substituted C3-6 heterocycloalkyl, optionally substi-tuted 5 to 8-membered aryl, optionally substituted 5 to 8-membered heteroaryl, optionally substituted 8 to 10-membered fused bicyclic aryl, optionally substituted 8 to 10-membered fused bicyclic heteroaryl; more preferably, the substituent is optionally substituted C3-6 heterocycloalkyl;Y is -CH-or N;R2 is optionally substituted C1-6 alkoxy, or optionally substituted C1-6 alkyl; preferably, R2 is optionally substituted C1-6 alkyl; andRM and RN are each independently selected from hydrogen, halogen, hydroxy, cyano, carboxyl, C1-6 alkyl, C1-6 alkoxyl, C1-6 haloalkyl, C1-6 haloalkoxyl, C1-6 hydroxyalkyl, C1-6 aminoalkyl, optionally substi-tuted C3-6 cycloalkyl, optionally substituted C3-6 heterocycloalkyl; furthermore, the C1-6 alkyl, C1-6 alkoxyl, C1-6 haloalkyl, C1-6 haloalkoxyl, C1-6 hydroxyalkyl may be further optionally substituted. In some embodi-ments, RM and RN are each independently selected from hydrogen, and C1-6 alkyl.
- The compound according to claim 3, or a stereoisomer thereof, a pharmaceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically ac-ceptable salt thereof, whereinR1 is optionally substituted C1-6 alkyl;and, in R1, the C1-6 alkyl is further substituted, and the substituent is selected from optionally substi-tuted C3-6 cycloalkyl, optionally substituted C3-6 heterocycloalkyl, optionally substituted 5 to 8-membered aryl, optionally substituted 5 to 8-membered heteroaryl, optionally substituted 8 to 10-membered fused bicyclic aryl, optionally substituted 8 to 10-membered fused bicyclic heteroaryl.
- The compound according to claim 3, or a stereoisomer thereof, a pharmaceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically ac-ceptable salt thereof, whereinR1 is optionally substituted C1-6 alkyl;in R1, the C1-6 alkyl is further substituted, and the substituent is optionally substituted C3-6 heterocy-cloalkyl.
- The compound of claim 3, or a stereoisomer thereof, a pharmaceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt thereof, wherein R1 is C1 alkyl; and the C1 alkyl is further substituted with optionally substituted C3-6 heter-ocycloalkyl, wherein the heterocycloalkyl is saturated and the heterocycloalkyl is attached to the rest of the molecule via the N atom on the ring.
- The compound of claim 3, or a stereoisomer thereof, a pharmaceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt thereof, wherein R1 is C1 alkyl; and the C1 alkyl is further substituted with optionally substituted C3-6 heter-ocycloalkyl, wherein the heterocycloalkyl is saturated and the heterocycloalkyl is attached to the rest of the molecule via the N atom on the ring. and the C3-6 heterocycloalkyl is 5 to 7 menbered heterocycloalkyl and has l to 2 heteroatoms.
- The compound according to claim 3, or a stereoisomer thereof, a pharmaceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically ac-ceptable salt thereof, whereinR1 is selected from the group consisting of
- The compound according to claim 3, or a stereoisomer thereof, a pharmaceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically ac-ceptable salt thereof, wherein R1 is C1 alkyl substituted by C6 heterocycloalkyl, optionally wherein the C6 heterocycloalkyl is piperazine or morpholine.
- The compound of claim 1, or a stereoisomer thereof, a pharmaceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically acceptable salt thereof, wherein R4 and R5 combine with the atoms to which they are attached to form a ring, wherein the ring is optionally substituted 3 to 10-membered cycloalkyl; preferably, is optionally substituted 3 to 6-membered cycloalkyl; more preferably, optionally substituted cyclopropyl.
- The compound according to claim 1, or a stereoisomer thereof, a pharmaceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically ac-ceptable salt thereof, whereinis selected from the group consisting of:is
- The compound according to claim 1, or a stereoisomer thereof, a pharmaceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically ac-ceptable salt thereof, whereinA is optionally substituted 5 to 6-membered heteroarylene, the heteroatom is independently selected from one or two of N, O and S, and the number of the heteroatom is independently 1 to 3preferably, A is optionally substituted 5-membered heteroarylene, the heteroatom is independently se-lected from N, O and S, and the number of the heteroatom is independently 1 to 2; preferably, one of the heteroatom is N, and the other, if present, is O or S;more preferably, A is selected from the group consisting of
- The compound according to claim 1, or a stereoisomer thereof, a pharmaceutically acceptable salt thereof, a metabolite thereof, a prodrug thereof, a solvate thereof or a solvate of the pharmaceutically ac-ceptable salt thereof, whereinX1 is C, and/or X2 is -CRa-, and/or X3 is -CRb, and/or X4 is -CRc-, and/or X5 is -C-.
- The compound according to claim 1, or the stereoisomer thereof, the pharmaceutically acceptable salt thereof, the metabolite thereof, the prodrug thereof, the solvate thereof or the solvate of the pharma-ceutically acceptable salt thereof, wherein, the compound is selected from any one of the compounds in Table 1.
- The compound according to claim 14, or the stereoisomer thereof, the pharmaceutically acceptable salt thereof, the metabolite thereof, the prodrug thereof, the solvate thereof or the solvate of the pharma-ceutically acceptable salt thereof, wherein, the compound is selected from compounds of Examples 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 27, 28, 29, 30, 31, 32, 33, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 84, 86, 87, 88, and 90 in Table 1.
- A pharmaceutical composition comprising the compound according to any one of claims 1 to 15, or the stereoisomer thereof, the pharmaceutically acceptable salt thereof, the metabolite thereof, the pro-drug thereof, the solvate thereof or the solvate of the pharmaceutically acceptable salt thereof, and a phar-maceutically acceptable carrier.
- A use of the compound according to any one of claims 1 to 15, or the stereoisomer thereof, the pharmaceutically acceptable salt thereof, the metabolite thereof, the prodrug thereof, the solvate thereof or the solvate of the pharmaceutically acceptable salt thereof, or the pharmaceutical composition according to claim 16 in the preparation of a medicament used for the treatment of cancer.
- The use according to claim 17, wherein the cancer is selected from the group consisting of small cell lung cancer (SCLC) , colorectal cancer (CRC) , pancreatic cancer, and cholangiocarcinoma, pancreatic cancer, appendiceal cancer, small bowel cancer, colorectal cancer, ampullary cancer, non-small cell lung cancer, cervical cancer, lung cancer, endometrial cancer, acute myeloid leukemia, gastrointestinal neuro-endocrine tumor, uterine endometrioid carcinoma, oesophagogastric cancer, bladder cancer, ovarian cancer, melanoma, multiple myeloma, thyroid gland adenocarcinoma, a myelodysplastic syndrome, and squamous cell lung carcinoma, or combinations thereof.
- The use according to claim 17, wherein the cancer is selected from the group consisting of non-small cell lung cancer, small cell lung cancer, colorectal cancer, pancreatic cancer, and cholangiocar-cinoma.
- The compound according to any one of claims 1 to 15, or the stereoisomer thereof, the pharma-ceutically acceptable salt thereof, the metabolite thereof, the prodrug thereof, the solvate thereof or the solvate of the pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising the same for use in treating cancer.
- A method for treating cancer in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of the compound according to any one of claims 1 to 15, or the stereoisomer thereof, the pharmaceutically acceptable salt thereof, the metabolite thereof, the prodrug thereof, the solvate thereof or the solvate of the pharmaceutically acceptable salt thereof, or the pharma-ceutical composition comprising the same.
Applications Claiming Priority (10)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| CN2024086658 | 2024-04-08 | ||
| CNPCT/CN2024/086658 | 2024-04-08 | ||
| CN2024114877 | 2024-08-27 | ||
| CNPCT/CN2024/114877 | 2024-08-27 | ||
| CNPCT/CN2024/133341 | 2024-11-20 | ||
| CN2024133341 | 2024-11-20 | ||
| CN2024137535 | 2024-12-06 | ||
| CNPCT/CN2024/137535 | 2024-12-06 | ||
| CN2024141123 | 2024-12-20 | ||
| CNPCT/CN2024/141123 | 2024-12-20 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2025214350A1 true WO2025214350A1 (en) | 2025-10-16 |
Family
ID=97349403
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/CN2025/087799 Pending WO2025214350A1 (en) | 2024-04-08 | 2025-04-08 | Macrocyclic compounds as ras inhibitors for the treatment of cancer |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2025214350A1 (en) |
Citations (7)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN113498342A (en) * | 2018-12-21 | 2021-10-12 | 锐新医药公司 | Compounds involved in synergistic binding and uses thereof |
| CN114867735A (en) * | 2019-11-04 | 2022-08-05 | 锐新医药公司 | RAS inhibitors |
| CN116457358A (en) * | 2020-09-15 | 2023-07-18 | 锐新医药公司 | Indole derivatives as RAS inhibitors for the treatment of cancer |
| WO2024060966A1 (en) * | 2022-09-19 | 2024-03-28 | 杭州阿诺生物医药科技有限公司 | Pan-kras inhibitor compound |
| WO2024153208A9 (en) * | 2023-01-19 | 2024-08-22 | 劲方医药科技(上海)有限公司 | Macrocyclic compounds, preparation method therefor, and use thereof |
| WO2025045233A1 (en) * | 2023-09-01 | 2025-03-06 | 劲方医药科技(上海)股份有限公司 | Macrocyclic compounds, preparation method therefor, and use thereof |
| WO2025051241A1 (en) * | 2023-09-06 | 2025-03-13 | 劲方医药科技(上海)股份有限公司 | Macrocyclic compound and antibody-drug conjugate thereof |
-
2025
- 2025-04-08 WO PCT/CN2025/087799 patent/WO2025214350A1/en active Pending
Patent Citations (7)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN113498342A (en) * | 2018-12-21 | 2021-10-12 | 锐新医药公司 | Compounds involved in synergistic binding and uses thereof |
| CN114867735A (en) * | 2019-11-04 | 2022-08-05 | 锐新医药公司 | RAS inhibitors |
| CN116457358A (en) * | 2020-09-15 | 2023-07-18 | 锐新医药公司 | Indole derivatives as RAS inhibitors for the treatment of cancer |
| WO2024060966A1 (en) * | 2022-09-19 | 2024-03-28 | 杭州阿诺生物医药科技有限公司 | Pan-kras inhibitor compound |
| WO2024153208A9 (en) * | 2023-01-19 | 2024-08-22 | 劲方医药科技(上海)有限公司 | Macrocyclic compounds, preparation method therefor, and use thereof |
| WO2025045233A1 (en) * | 2023-09-01 | 2025-03-06 | 劲方医药科技(上海)股份有限公司 | Macrocyclic compounds, preparation method therefor, and use thereof |
| WO2025051241A1 (en) * | 2023-09-06 | 2025-03-13 | 劲方医药科技(上海)股份有限公司 | Macrocyclic compound and antibody-drug conjugate thereof |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| EP4144732A1 (en) | Benzothiazolyl biaryl compound, and preparation method and use | |
| EP4365178A1 (en) | Nitrogen-containing heterocyclic compound, and preparation method therefor, intermediate thereof, and application thereof | |
| CN114656482B (en) | Macrocyclic heterocyclic compounds as EGFR inhibitors and application thereof | |
| EP4056563A1 (en) | Compound as shp2 inhibitor and use thereof | |
| ES2928169T3 (en) | 4-(3H-indol-5-yl)-N-(pyridin-2-yl)pyrimidin-2-amine derivatives as protein kinase inhibitors, and method of preparation and medical use thereof | |
| US12168058B2 (en) | Glutarimide-containing KRAS-mutant degrader compounds and uses thereof | |
| TWI751163B (en) | Fgfr4 inhibitor, its preparation method and use thereof | |
| WO2021046407A1 (en) | Rip1 inhibitory compounds and methods for making and using the same | |
| WO2024002373A1 (en) | Substituted pyrimidine-fused ring inhibitor, method for preparing same, and use thereof | |
| CN117917416A (en) | KRAS G12D degradation agent and preparation method and application thereof | |
| WO2024118966A1 (en) | Glutarimide-containing pan-kras-mutant degrader compounds and uses thereof | |
| WO2021180235A1 (en) | Inhibitor of enhancer of zeste homologue 2, and use thereof | |
| CN116143805A (en) | A class of nitrogen-containing heterocyclic biaryl compounds, preparation method and use | |
| WO2024235286A1 (en) | Tricyclic compounds and uses thereof | |
| JP2021511353A (en) | Substituted pyrazolo [1,5-a] pyrimidine macrocycle | |
| CA3150284A1 (en) | Aza-quinoline compounds and uses thereof | |
| CN116157401A (en) | Heterocyclic derivatives and their preparation and use | |
| WO2023143147A1 (en) | Pyridazopyridone compounds, pharmaceutical composition thereof and use thereof | |
| WO2018097162A1 (en) | Sulfonamide derivative having coumarin skeleton | |
| BR112020004563A2 (en) | heteroaromatic compounds substituted by cycloolefin and their use | |
| WO2025214350A1 (en) | Macrocyclic compounds as ras inhibitors for the treatment of cancer | |
| CN115433207A (en) | Macrocyclic heterocyclic compound as EGFR inhibitor and application thereof | |
| AU2022217319A9 (en) | Tricyclic compounds and uses thereof | |
| EP4308569A1 (en) | Selective modulators of ataxia telangiectasia mutated (atm) kinase and uses thereof | |
| TW202216728A (en) | Thienopyridine compound, comprising pharmacuetical composition and application thereof |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 25785876 Country of ref document: EP Kind code of ref document: A1 |