LU505465B1 - A pan-KRAS inhibitor compound - Google Patents

A pan-KRAS inhibitor compound Download PDF

Info

Publication number
LU505465B1
LU505465B1 LU505465A LU505465A LU505465B1 LU 505465 B1 LU505465 B1 LU 505465B1 LU 505465 A LU505465 A LU 505465A LU 505465 A LU505465 A LU 505465A LU 505465 B1 LU505465 B1 LU 505465B1
Authority
LU
Luxembourg
Prior art keywords
compound
int
mmol
alkylene
reaction
Prior art date
Application number
LU505465A
Other languages
German (de)
Inventor
Feifan Li
Canfeng Liu
Wanli Cheng
Meng Lv
Chaofan Jin
Shuaishuai Liu
Kaixuan Chen
Yufeng Chen
Peng Wu
Nanhai He
Han Yang
Zhao Sun
Original Assignee
Adlai Nortye Biopharma Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Adlai Nortye Biopharma Co Ltd filed Critical Adlai Nortye Biopharma Co Ltd
Application granted granted Critical
Publication of LU505465B1 publication Critical patent/LU505465B1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D513/00Heterocyclic 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
    • C07D513/22Heterocyclic 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic 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

Landscapes

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

Abstract

The present invention relates to a pan-KRAS inhibitor compound represented by formula (I) or formula (II) and a pharmaceutical composition containing the compound, and the use of compound of formula (I) or formula (II) for preventing and/or treating cancer, tumor, inflammatory disease, autoimmune disease or immune- mediated disease.

Description

BL-5786
A pan-KRAS inhibitor compound LU505465
Technical field
The present invention relates to a compound, in particular to a highly active pan-
KRAS inhibitor and its use.
Background technique
RAS is one of the most frequently mutated genes in human tumors, and its mutation occurs in about 30% of tumor patients, among which KRAS accounts for about 85% of RAS mutations, KRAS mutations exist in 38% of pancreatic cancer, 50% of colorectal adenocarcinoma, and 32% of lung adenocarcinoma. The development of targeted KRAS inhibilors has great clinical significance and value.
KRAS is a membrane-bound protein with GTPase activity, which cycles between a GDF-bound inactive conformation and a GTP-bound active conformation through nucleotide exchange, performing the function of a "molecular switch”, KRAX in the
GTP-bound state can activate multiple downstream signaling pathways including
RAF-MEK-ERK and PISK-AKT, and regulate life processes such as cell growth, proliferation, differentiation and apoptosis,
BRAS mutations (such as G12C, GI2D, GI2V, GI13D, etc.) will affect the hydrolysis of GTP mediated by GTPase activating proteins (GAPS), increase KRAS in the GTP-bound activated state, and over-activate downstream signaling pathways, eventually lead to the occurrence and development of tumors, However, since the
KRAS protein lacks a corresponding hydrophobic pocket suitable for drug binding, and tis affinity with GTP and GDP is at the picomolar level (~20pk), it is very difficult to develop inhibitors that competitively bind to KRAS. KRAS has been considered an undruggable target for the past decades.
In May 2021, AMOS 10 was approved by the FDA for the treatment of locally advanced or metastatic non-small cell lung cancer carrying the KRASS*PS mutation, breaking the history of KRAS being "undmiggable”. However, the GI2C mutation only accounts for a small portion of KRAS mutations. For mutations at other sites of
KRAS, there is still a lack of satisfactory and effective inhibitor compounds, and a large number of clinical needs have not been met. Therefore, the development of effective pan- KRAS inhibitor compounds are a need in the art.
Contents of the invention
The invention provides a pan-KRAS inhibitor. Such structures are different from 1
BL-5786 existing KRASY!2C inhibitors that act through covalent binding, but instead mediate the LUS05465 formation of a ternary complex between ubiquitous intracellular chaperones (such as
Cyclophilin A) and KRAS proteins. The formation of the ternary complex can block the combination of KRAS and its downstream effector molecules (such as RAF) through steric blockage, inhibit the activation of MAPK and PI3K-AKT signaling pathways, thereby inhibiting the occurrence and development of tumors, and play a role in the treatment of tumors and other diseases.
In one aspect, the present invention provides a compound with a structure of formula (I) or formula (II) or a pharmaceutically acceptable salt, isotopic derivative, or stereoisomer thereof: ee O O
R
O vx 4),
R
R, N= N 3
N UN
Re / \ J
O — N \ ! 8=N ReR
Rs R. 5 (D or os N° °, 0 (R) AS
R
R, N= s 3
N
Re / \ 7
O — N
Rga % — ' !
Res SN Re’ R;
WER,
Red (I) wherein:
Ri represents C1-C6 alkyl, -(Ci-Cs alkylene)-(Cs-Cs cycloalkyl), -(C1-C6 alkylene)-(4-8 membered heterocycloalkyl), -(C1-C6 alkylene)-ORa, -(C1-C6 alkylene)-
SRa or -(C1-C6 alkylene)-NRaRa’;
R represents halogen, cyano, Ci-Cs alkyl, -(Co-C¢ alkylene)-(Cs-Cs cycloalkyl), or -(Co-Cs alkylene)-(4-8 membered heterocycloalkyl), which may optionally be substituted by 0, 1 or 2 of the following substituents: -ORa, -SRa, or -
NRaRa’; 2
BL-5786
Rs represents hydrogen, -O(Co-Cs alkylene)Ra, -S(Co-Cs alkylene)Ra, -N(Co- LU505465
Cs alkylene)Ra(Co-Cs alkylene)Ra , which may optionally be substituted by 0, 1 or 2 of the following substituents: -ORa, -SRa, or NRaRa’;
Cy1 represents C3-C12 cycloalkyl or 4-12 membered heterocycloalkyl;
Rı each independently represents hydrogen, halogen, oxo, Ci-Cs alkyl, -(Co-
Cs alkylene)(Cs-Cs) cycloalkyl, -(Co-Cs alkylene)(4-8 membered) heterocycloalkyl, - (Co-Cs alkylene)ORa, -(Co-Cs alkylene)SRa, -(Co-Cs alkylene)NRaRa’, -(Co-Cs alkylene)CORa, -(Co-Cs alkylene)COORa, -(Co-Cs alkylene)CONRaRa’, -(Co-Cs alkylene) NRaCORa’, -(Co-Cs alkylene) OCONRaRa’, -(Co-Cs alkylene)
NRaCONRaRa’, -(Co-Cs alkylene)SORa, -(Co-Cs alkylene)S(O)»,Ra, -(Co-Cs alkylene)NRaS(O)»Ra’, -(Co-Cs alkylene)CN, -(Co-Cs alkylene)(C6-C10 aryl) or -(Co-
Cs alkylene)(5-12 membered heteroaryl); wherein, R4 on the two C atoms of Cy1 together with the C atoms connected thereto and the atoms between the two C atoms can form a 3-8 membered ring, the 3-8 membered ring optionally may contain 0, 1, 2 or 3 heteroatoms selected from N, O or S; or two R4 on the same C atom of Cy; together with the C atom connected thereto can form a 3-8 membered ring, the 3-8 membered ring optionally may contain 0, 1, 2 or 3 heteroatoms selected from N, O or S;
Rs, Re’ each independently represents hydrogen, halogen, Ci-Cs alkyl, C3-Cs cycloalkyl or -(Co-Cs alkylene)CN;
Rs, Rs’ each independently represents Ci-Cs alkyl, -(Co-Cs alkylene)(Cs-Cs cycloalkyl), -(Co-C¢ alkylene)(4-12 membered heterocycloalkyl), -(Ci-Cs alkylene)ORa, -(Ci-C¢ alkylene)SRa, -(Ci-Cs alkylene)NRaRa’, -(C1-C6 alkylene)CN, -(Ci-Cs alkylene)C(O)Ra, -(Co-Cs alkylene)C(O)ORa, -(Co-Cs alkylene)C(O)NRaRa’, -(Co-Cs alkylene)OC(O)NRaRa’, -(Co-Cs alkylene)NRaC(O)Ra’, -(Co-Cs alkylene)S(O)Ra, -(Co-Cs alkylene)S(O),Ra, which may optionally be substituted by 0, 1 or 2 substitutents selected from the following: - (C1-Cs alkylene)ORa, -(C1-C6 alkylene)SRa, -(Ci-Cs alkylene)NRaRa’;
W represents NR; or CR7R7’, wherein,
R7, R7’ each independently is selected from: hydrogen, Ci-Cs alkyl, -(Co-C6 alkylene)(Cs-Cs cycloalkyl), -(Co-Cs alkylene)(4-12 membered heterocycloalkyl), -(Co-
Cs alkylene)CN, -(Co-Cs alkylene)ORa, -(Co-Cs alkylene)SRa, -(Co-Cs alkylene)NRaRa’, -(Co-Cs alkylene)C(O)Ra, -(Co-Cs alkylene)C(O)ORa, -(Co-Cs alkylene)C(O)NRaRa’, -(Co-Cs alkylene)OC(O)NRaRa’, -(Co-Cs 3
BL-5786 alkylene)NRaC(O)Ra’, -(Co-Cs alkylene)S(O)Ra, -(Co-Cs alkylene)S(O):Ra, or R7, | LU505465
R7’ can form a 3-8 membered ring with the C atom connected thereto, and the ring may optionally contain 0, 1, 2 or 3 heteroatoms selected from N, O or S;
Rsa, Rgb, Rec, Red each independently is selected from: hydrogen, halogen, oxo,
Ci1-Cs alkyl, -(Co-Cs alkylene)(Cs-Cs cycloalkyl), -(Co-Cs alkylene)(4-8 membered heterocycloalkyl), -(Co-Cs alkylene)CN, -(Co-Cs alkylene)ORa, -(Co-Cs alkylene)SRa, -(Co-Cs alkylene) NRaRa’, -(Co-Cs alkylene)C(O)ORa, -(Co-Cs alkylene)C(O)NRaRa’, -(Co-Cs alkylene)OC(O)NRaRa’, -(Co-Cs alkylene)NRaC(O)Ra’, -(Co-Cs alkylene)S(O)Ra, -(Co-Cs alkylene)S(O)»Ra; or Rsa and Rsb or Rec and Rgd connected to the same C atom can form a 4-8 membered ring with the C atom connected thereto, and the ring may optionally contain 0, 1, 2 or 3 heteroatoms selected from N, O or S; or two of Rga, Rsb, Rgc, Red that are not connected to the same C atom can form a 4-8 membered ring with the C atoms connected thereto and the atoms between the two C atoms, and the ring may optionally contain 0, 1, 2 or 3 heteroatoms selected from N, O or S; or one of Rga, Rsb, Rec, Red forms a 4-8 membered ring with R7 or R7’ on adjacent or non-adjacent W, and the ring may optionally contain 0, 1, 2 or 3 heteroatoms selected from N, O or S, and when one of
Rsa, Rgb, Rec, Red forms a 4-8 membered ring with R7 or R7’ on the adjacent W, the ring may optionally contain 0, 1, 2 or 3 unsaturated bond; wherein, p represents 0, 1, 2, 3 or 4; m, n each independently represents 1, 2 or 3;
Ra, Ra’ each independently represents hydrogen, Ci-Cs alkyl, C3;-Cs cycloalkyl, 4-8 membered heterocycloalkyl; wherein, when Ra and Ra’ are connected to the same
N atom, the Ra and Ra’ and the commonly connected N atom can form a 4-8 membered ring, the 4-8 membered ring optionally may contain 0, 1, 2 or 3 heteroatoms selected from N, O or S; the alkyl, cycloalkyl, heterocycloalkyl, alkylene each independently can be substituted by 0, 1, 2, 3, 4, 5 or 6 halogen atoms.
In some embodiments, in the compound with the structure of formula (I) or formula (IT) or a pharmaceutically acceptable salt, isotopic derivative, stereoisomer thereof, Rı represents Ci-Cs alkyl, -(C1-Cs alkylene)-(C3-Cg cycloalkyl) or -(C1-C6 alkylene)-(4-8 membered heterocycloalkyl); preferably, Ri represents C1-C6 alkyl, 4
BL-5786 more preferably, R1 represents C1-Cs alkyl. LUS05465
In some embodiments, in the compound with the structure of formula (I) or formula (II) or a pharmaceutically acceptable salt, isotopic derivative, stereoisomer thereof, Ra represents Ci-Cs alkyl, which may optionally be substituted by 0, 1 or 2-
O—Ra
ORa substituents; preferably, Ro represents a ; further preferably, Ra represents —~ ve ; more preferably, Ra represents ; wherein, * represents Ra is connected to the part of formula (I) where connecting to the site .
In some embodiments, in the compound with the structure of formula (I) or formula (II) or a pharmaceutically acceptable salt, isotopic derivative, stereoisomer thereof, R3 represents hydrogen or -O(C1-Cs) alkyl, -O(Co-Cs alkylene) (C3-C3)H alkyl, -O(Co-C6 alkylene) (4-8 membered) heterocycloalkyl, which may optionally be substituted by 0 or 1 substituent selected from the following: ORa, -SRa, or NRaRa’.
In some embodiments, in the compound with the structure of formula (I) or formula (IT) or a pharmaceutically acceptable salt, isotopic derivative, stereoisomer thereof, Cy1 represents C;-Cg cycloalkyl or 4-8 membered heterocycloalkyl.
In some embodiments, in the compound with the structure of formula (I) or formula (IT) or a pharmaceutically acceptable salt, isotopic derivative, stereoisomer thereof, R4 each independently represents hydrogen, halogen, Ci-Cs alkyl, -(Co-C6 alkylene)ORa, -(Co-Cs alkylene)SRa, -(Co-Cs¢ alkylene)NRaRa’, -(Co-Cs alkylene)CONRaRa’, -(Co-Cs alkylene) NRaCORa’, -(Co-Cs alkylene) OCONRaRa’, - (Co-Cs alkylene)CN, -(Co-Cs alkylene)(5-12 membered heteroaryl), or R4 on the two C atoms of Cy: together with the C atoms connected thereto and the atoms between the two C atoms can form a 3-8 membered ring, and the 3-8 membered ring optionally may contain 0, 1, 2 or 3 heteroatoms selected from N, O or S; or two R4 on the same C atom of Cy: together with the C atom connected thereto can form a 3-8 membered ring, the 3-8 membered ring optionally may contain 0, 1, 2 or 3 heteroatoms selected from N, O or S. 5
BL-5786
LU505465
In some preferred embodiments, in the compound with the structure of formula (I) or formula (II) or a pharmaceutically acceptable salt, isotopic derivative, stereoisomer thereof, R4 each independently represents hydrogen, halogen, Ci-Cs alkyl, -(Co-C6 alkylene)CONRaRa’, -(Co-Cs alkylene)(5-12 membered heteroaryl), or R4 on the two
C atoms of Cy1 together with the C atoms connected thereto and the atoms between the two C atoms can form a 3-8 membered ring, the 3-8 membered ring optionally may contain 0, 1, 2 or 3 heteroatoms selected from N, O or S; or two R4 on the same C atom of Cy: together with the C atom connected thereto can form a 3-8 membered ring, the 3-8 membered ring optionally may contain 0, 1, 2 or 3 heteroatoms selected from N, O or S.
In some embodiments, in the compound with the structure of formula (I) or formula (II) or a pharmaceutically acceptable salt, isotopic derivative, stereoisomer thereof, Rs, Rs’ each independently represents hydrogen or Ci-C¢ alkyl; more preferably, Re, Re’ each independently represents hydrogen or methyl.
In some embodiments, in the compound with the structure of formula (I) or formula (II) or a pharmaceutically acceptable salt, isotopic derivative, stereoisomer thereof, Rs, Rs’ each independently represents C1-C6 alkyl, -(Co-Cs alkylene)(Cs-Cs cycloalkyl), -(Co-C¢ alkylene)(4-12 membered heterocycloalkyl), -(Ci-Cs alkylene)NRaRa’ substituted by 0 or 1 substituent selected from -(C1-C6 alkylene)ORa, -(C1-Cs alkylene)SRa.
Further, in some preferred embodiments, in the compound with the structure of formula (I) or formula (II) or a pharmaceutically acceptable salt, isotopic derivative, stereoisomer thereof, at least one of Rs, Rs’ contains at least one heteroatoms selected from N, O or S, preferably N atom, more preferably secondary amine or tertiary amine.
In some embodiments, in the compound with the structure of formula (I) or formula (II) or a pharmaceutically acceptable salt, isotopic derivative, stereoisomer thereof, R7, R7’ each independently represents hydrogen, Ci-Cs alkyl, -(Co-Cs alkylene)(Cs-Csg cycloalkyl), -(Co-Cs alkylene )(4-12 membered heterocycloalkyl), -(Co-
Cs alkylene)CN, -(Co-Cs alkylene)ORa, -(Co-Cs alkylene)SRa, -(Co-Cs 6
BL-5786 alkylene)NRaRa’, -(Co-Cs alkylene)C(O)NRaRa’, -(Co-Cs alkylene)OC(O)NRaRa’, - | LUS05465 (Co-Cs alkylene)NRaC(O)Ra’, -(Co-Cs alkylene)S(O):Ra, or R7, R7’ can form a 3-8 membered ring with the C atom connected thereto, and the ring may optionally contain 0, 1, 2 or 3 heteroatoms selected from N, O, S.
In some embodiments, in the compound with the structure of formula (I) or formula (II) or a pharmaceutically acceptable salt, isotopic derivative, stereoisomer thereof, Rsa, Rsb, Rec, Red each independently is selected from: hydrogen, halogen, C1-
Cs alkyl, -(Co-Cs alkylene)(Cs-Cs cycloalkyl), -(Co-Cs alkylene)(3-8 membered heterocycloalkyl), -(Co-Cs alkylene)CN, -(Co-Cs alkylene)ORa, -(Co-Cs alkylene)SRa, -(Co-Cs alkylene)NRaRa’, -(Co-Cs alkylene)C(O)NRaRa’; or Rsa and Rgb or Rec and
Rsd connected to the same C atom can form a 4-8 membered ring with the C atom connected thereto, and the ring may optionally contain 0, 1, 2 or 3 heteroatoms selected from N, O or S; or two of Rga, Rgb, Rsc, Red that are not connected to the same C atom can form a 4-8 membered ring with the C atoms connected thereto and the atoms between the two C atoms, and the ring may optionally contain 0, 1, 2 or 3 heteroatoms selected from N, O or S; or one of Rga, Rsb, Rec, Red forms a 4-8 membered ring with
R7 or R7° on adjacent or non-adjacent W, and the ring may optionally contain 0, 1, 2 or 3 heteroatoms selected from N, O or S, and when one of Rga, Rgb, Rec, Rsd forms a 4- 8 membered ring with R7 or R7° on the adjacent W, the ring may optionally contain 0, 1, 2 or 3 unsaturated bond, preferably, the ring is an aromatic heterocycle.
In some embodiments, in the compound with the structure of formula (I) or formula (II) or a pharmaceutically acceptable salt, isotopic derivative, stereoisomer thereof, when W is not NR7, R7, R7’, Rga, Rgb, Rec, Rsdat least one of or the ring formed between them includes at least one heteroatoms selected from N, O or S, preferably N atom, more preferably secondary amine or tertiary amine.
In some embodiments, in the compound with the structure of formula (I) or formula (II) or a pharmaceutically acceptable salt, isotopic derivative, stereoisomer thereof, p is preferably 0, 1 or 2, or m, n each independently is preferably 1 or 2.
In some embodiments, in the compound with the structure of formula (I) or 7
BL-5786 formula (IT) or a pharmaceutically acceptable salt, isotopic derivative, stereoisomer LUS05465 thereof, the structure Cy1-(R4)p in formula (I) is selected from the following:
F : 3 . 5e.
GONE RE el Sch SD
HA à Eee
UN (6 LL 0 AN © /
Safe) () (9) BR) 49) (P) «IN MM A0 MN wp A oO R) RR N BSD FZ av
N 3 X À N J A Ba Oh! AE ®
N
7 y 5 N ; “ee fo “NA AO “RG oH v 72) oH “Reo” / | So / —OH \ A HO, oO Le Be
Oo OT PR „N (R) 3 QG *, N RR (S) = >
A Se al La" a NP _ö >
O No P 0 0 =, %,,
NZ . .
N o © o7>N 07>N
DU wherein, * represents the site where -Cy1-(R4)p is connected to the part of formula (I) where connecting to -Cy1-(R4)p.
In some embodiments, in the above-mentioned compounds having the structure of formula (I) or the pharmaceutically acceptable salts, isotope derivatives, and
Q, _ /
Re tor stereoisomers thereof, the structure = Rs in formula (I) is selected from the following: 8
BL-5786
LU505465 ? # Q 7 ? / GO / Q + 0 ; —S=N —SEN —SSN ——S=N S=N
Il 7-S=N x x x x \ ( _N ) _N “SE N° ] J 5
O So HO
O O
O O Il > bh AE
Sy” ge A 5
CV N
O N
N od)
Q, _ /
Re tor wherein, * represents the site where ° Rs’ is connected to the part of formula
Q, _ /
Re tor (I) where connecting to © Rs .
In some embodiments, in the above-mentioned compounds having the structure of formula (II) or the pharmaceutically acceptable salts, isotope derivatives, and 0
Rga. m¥a_ +
Rep ca
W Roc stereoisomers thereof, the structure Rad in formula (ID) is selected from the following: 9
BL-5786
A Co N 0 0 , LU505465 (3° #4 S <> (OY I / of 8 C J —/ 6) 2 # À nN A N} 2 x a a =N =| =| =N7 = = or AT NT gv 4 y 7 HO F 7 C 0 Ho oO
Q > pré Ly 0 oO =" = i 7 ~~ KH " oN 5 FH"
X 57 À © y a oO , ?_ #ÿ oO
N HN y
LOU IS TES
© x ? # Q x NC o ., il 2. 7”
O N
>, A Gr ° Oh N >07 9 \
Q x Q x O 2 7 Q » 2 # o s= s= son =N = =N Q x / 4 / / NS ~~ © CN oO s \ \ \
O , o , 0 o ,
ON a $=N’ Lr F Lv y Oo Sen / A QO 47 X < 3 0 HN, HN, HN >N A oO 0 Q A oO Sa
SNS San“ Sn A ( oJ N 2 wo / TL # os
HN HN HN HN SN O NT
\ N N | \ 0
REN
WR wherein, * represents the site where Red is connected to the part of 0
REN
WR formula (II) where connecting to Red .
BL-5786
LU505465
In some embodiments, in the compound with the structure of formula (I) or formula (IT) or a pharmaceutically acceptable salt, isotopic derivative, stereoisomer thereof, the compound of formula (I) has the structure of formula (III), and the compound of formula (IT) has the structure of formula (IV):
O ©) O
To (R) R are
N= Rs
R, Ss
N NS
Re / \ 7
O — N
WN, ,
R AN Rg' R4 5 Rs formula (II),
SNS à
O (R) R re”
R, N=\ Rs
N S
Re / \ 7
O — N
R W ,
Rep SEN Re’ R4
WE Res
Red formula (IV)
In another aspect, the invention provides compounds having the following structures: oy Gh O6 ex 96 ex 99 7 Go) o Ha Jus o Huw ds o Hw As © Da do 7 ad <a 7 ie ds 7 NUE 79 ASE
AS FF = AA 7 NS 7 CN >»
CF J Ci J a J No —
S Qu q / ee 09 ex Oo , 5 "A J À ie Joa d _ N vv d N VE
Wo — oS
X G
© © 11
6 © X SS LU505465
Oy NSP à oy NMC 0 oy NO 0 oy NN à F / o "ok he / o Mend ® / °"® hee / o "ond od (5) (5) (5) Ss) “ae nA N Xe nÀ N ® Sw =H Xe nd 7 5 s 5
N ~ N ne N = N = rN Nf yo A oN 9 5 9 > 9 > 9 5 8 TE TE (8
N_/ N_/ N_/ N_/ / / / 7
Og GN à N Og LP à N Oy N20 à ° Oy NES © °
A He , 7 1 Hol Lol o He A à Hel A / ee / nm J NN ON o N Ji
Om NA, A Oo N H © N= © NA,
N = N x
LA > NA > AA NN 8 N 2 N 2 N 2 N (SN — an — 3 — an — nN Nn N_/ N / / 7 7 le IN Oy NT 0. SSP à Oy N° 99 , Rell Ag, , EC) ny ® , o Mel of , 0 ol Amo
N
0, © N= H oO 0, © N= H [D a N= F 9, ol N= A a, s 5 N 5 N 5
NA 7 > NS 7 PN 4 NS 2 N 2 N A N a À iN J $=N = =
CO (7 (J (7 / / / / 0.21 0 ole 0 oo 0 oon o
SON 5 5 SON 5 1 He 1 _N TON 5 5 ZN / © Oy Rom / © ONE on / © N La / © Ya 0 H © d BH Ve 9 H © o BH V®
Q N= 9 N= © N= 9, N= s s 5 5
N = N = N = N = yo Ny Ny rN Ny 9 5 9 5 9 5 9 +
EN 8 (SH eu nN nN N) N 4 4 7 7
Ou» ex 0 Os @ 0 Og ©) 0 Os X oO
J H @ 1 J H © 1 À H 8 3 © - J H © î of N NS of N NS of N Go J N 00 9 NH LUN a, N= L_o 3 NA, 3 N= N 0 s s s
N = N = N = N x
NA NA OS NA
2 5 2 5 2 A 2 7
TE TE (SN 4 nN nN N) N 7 7 ~ 4
X / lo X 6 © Ss) S ©
Oy in 99 9 os An 0 Ope in °0 0 y" on y
J 0 6 vie , 0 © Hod J 0 6 ve , 0 6 vey 3 N= Og N= à © N= X N Og N= ©
N = N = N p= N 5
SL ÿ Sd ON ÿ NS 9 + 9 5 9 + 9 + au $=N (ss $=N (J (J (7 (J / 7 7 7
On oa 0 Os oa 0 Os GA Oo N 0. ea 0
SON 9 SON 9 SON 9 Sr 5 / °° 1e / oo a / AN 0" SE, / N
H 9 H
Ca N= Og n=l 3) N= (Le Ce NE 0 5 5 5
N x = N =
Mg Mg MN A > a N a N 9 N 0 N (SN — (SH At Te — a J
NS No Nn NS 7 x s 4 ole) 0 61 Os ex So Os ex
FN N00 J N °0 / J Ho In À N 99 0 / o "eu J / 0 ol JL o N / 0 ©) 9 NNT 4 NONE © N H = N © H le © N= N = o N=
N n=l en A = NA 5 = = ©
NV J = 0 7 0 \_… ° J a NON NU a
CH 3 3 Sul
N N 7 N 0 © N | 6) 0 6)
SON 0 Os 61 Oe a @ No y J N { / 3 not °° 0 TON V0 0 , J N î So o © wl oO of 0 © AA of 0 © Ae CM o © A © H N N © H ° (VA > À VS ARN; 2S ° / = or — 0 (_ 0 (_ — i N q N 2 N I N
I (TSN S=N NU — “m —
A A NV N en oe o A SANS —=N o Jad o oO ex o oH Ty ? o Hy ST
N 9? —oh 3 N Oo Hz / o Hg A EAN J Wo
I) (8) Ay / oO ml A 3 9 À 15) 5 N H / N WB) 9 NN fs N= N Us 9 = (8) N=! N s # À > & N MN = zZ
SS N 8 Z 0 HN
Mg { q LN sn J / 9 ZN CN cn 4 AT 3 [EN — Wo 7 7 7 ‘
N old, a... ou fo) TN 9 o Hy L ou lo) ° 3 N 29 N / o Fe I 2e J A N © 3 N 1 / o SR Se À 9 N NTA ml s NOR ®) / (8) oO N = ! 9 N wb is) n= H 8 nN hy Ss ® A 7 =” n°9 N= AEN MN 4 8 CN 9 AN > 0 = S=N 9° (+ > 2 = N (TSN N 3 —
I N (8 J J N (CSN _J 7 N
N A
NS 7 7 ou lo
Ol Ng SON o . 7 o 0, À. >
Ou No 3 N 9 y o Bel À # 9 N
SON 9 / oO ol, À 8, 9 Se NON ©) NAT o Be o SO NON o SH N / 0, NT 8 fs) N= Ÿ aS 9 — N 8) is) N= iS N 8 4 7 s) N= æ MN J LS NA 28 = \
N s 9 = S=N
AK 4 Bas CN sa‘ CAT 3 4 (sy J N
N
N) 7
A slo
Os" ei 0 TON 9 0 6) Ton 9 o Fe oO ©) 0 Ÿ y" 90 / 9 "ol Je od = ©
TN 9 / 0 of Ne q N © © N= TT Ym / 0 a de 9 N © ) N= ® N 5 a N © ) N= ® N 5 oN 6 N= ® N 5 0 NX 0 N
N ¢ SL + a => 0 Wr i. N (3 N + 0 = $= ( N)
N ISN J py
SN J N_/ N Ho 7 a
CN
6) 67
Gù old a ne 0, S) 0 () .
Og ©) 0 DI (AVC / o Hg de o = 2 ©
SN 9 o Aal 1 9 N © A
H | / ol Je N © N= ® / 0 oly Je 0 N © 6) N= © N Ss 9 N ©) © N: H ® N 5 a, > & N= ® N: a (NV / = 0 \
A A SS GNS > MS a op o \_, 2 = TSN n_ N S=N 4 N
NU SS N © nN N Ho o ~~ ou lo) 0 Go) TNA ou lo) oO SN 90 d Fey
Os ON TON a o Hl J / NUL
SNL 0 o Hel i / N 9 N ©
N | / NT 9 N © 6 N= ® / O À de Q N © ©) N= ® N 5 q N © &, N= æ N 5 NA = 6 N= ® N LS a, = 0 #44
N Ss / 7 0 4 a, > — Un N S=N J a 3 = Sal 1 2 N N y N
LAN A > - N A &
N 6) o 61 a oo TON 0 6) oo bdo, JN y "LS,
Or No H | / © SL Ao 9, N (5) 3% ° y 0 ol a 0 N © & TE Li / o Hyg Le 0 N (5) 5 + H ® a H 6 ©) N N Ng) ° N 5 ©) N= N LS (NX 9 & N= ®) N 4 7 Ss WON 9 N
A = 8 0 \— =I —s=N J
Ÿ 2 N —s=N" _ 7 9 N= TSN J —s=N J N ON
N A 7 DD
N J
C3 “e HO o°
Ga 0 wo 5 0 © 0 62 o Fe J, 0 a rip, ; ce
QC N 9 ol N 9
TF9 1° "A Le of yh 9 Ne ®
Ô 5) a IN H (5) 5 5) 174 A 5 wa YA 15 A fs 5 CL N (3) N= (RY N 5 x N
N x IH = oO (4 (i — ¢ Wot 9 == a N 2 N Sa’) ~ 2 ~Sa _7 | ,
Q A x
N
\ oC Aad © So SWS
OO Oe N 0 o Hl J / & MS
SA 0 > N 9 of INH q N 7 o Hl / 0 A due H 19 s) N= (R)
X 6) ee o N a 5 N (9) N= (i) N os ) wed A a WY p= 5 ’ J 5 0 $_/
N N 0 N € XS 0 Ÿ N As > 8 N _ \ 7 N VON os ca CE J N HN 7
Nn / 5 N
N AS N \
UT ve og le) oO ° 9 6 o Hl J, 6) I a on SA 09 J wor
Ogu (SI H 5)
Io , DO J © SP 6) n= ff / 0 Ju, o N © N 5) j { 5 of N (5) (s), Nes (21 (9 Na (21 — ° J 1 Na ® a 5 a, N N a ¢ NSG > 0 = 0 N ©
I N Az N. 2 N (SN J a N _/ 0 (Sn J N + N L \
N C o. 0 ty Fu N
N \ \ oo) 0 ©; SQN 0 o. ©) SN 0 3 N a 0, fo) so SON J > 8 0, ; I He Ÿ J à Hog 5e J ©) yz / 0 of de o NE HM © ©) N= ® a N © H ©) 6) N= ® N 5 bo n= f A nŸ 0S A J SS o < J >
N 5 = —_ au; = ° © 7 8 N Ca À = N SN y CN — (3 — (© N N
N ~
N / L 9 y = 50 TN
N
\ Q
O00 ® No 9 lo SON Pa / o Hel m 0, > SN 0 / o Hel dg 9 N ©
FN / 9 Pel Je a N © © N= œ / à "au À 9 N © ©) N= ® N > 9 NL A N 5 CN
N © N ©) N= ®) À 7 = a _ 19 N= ® AR 5 o N In 5 oN gp = 9 N eu J ® $ N Ru N / eu J y De /
A â
CN 4 0. ©) Os ol Pa / d "0 1,
Oy NO Os ON / à Hel 9 N ©
J N 9 I N 0g d TR © © n=l HH AS o 0 © A < J 0 ol, lo 19 nd RH 8 AS J = 1.9 N4 ® Pa n=l fH VF (0 s GEN aw => AA} 5 2 N eo al WS Sl 5 = 5=N J 7 oO as CN CF3 / oO 0 © Os An 8
Ox ® IN _g / Q & 0)
Os en a J N © X 9 N ©
Q 61 7 N 9 J s) N= ®
Ser N °° J Q © eo 04 S a ze N= } 5
J o ol de Pc n=l A As MN J 5 9 N © = © nA A CC AA 5 0 Sr Os
MA sv 2 N VIN © 9 = S=N J F 2 N -
S=N y (J HN N
HN
HN 0
Ol No
Q. 6) TON 9 061 an Le
Son / DE o N°05
GC y o Hel Le 8 N @ 19 p= = og. l® qo NSE N= N
NN, ©) =H a, N 8 a o Hel 1 N N | CN 2 N
J ol Ao Z aay 5 and ads =) nt 1 ® QO S=N J SA
AS ADs oH ( A = /
S=N J HN vo > Sy No qd N 7 / 5 Hel Je J 0 oe
He 5 = ® > N 09 > NN Og nN ha A A 5
J o > wl © o o Hg ws © a, = 0 (_ 5) 0 Ne an N ) NH 1 ) ©) n= N p doy MN EN
A J a OP A J SS oy iv ON 7 20 0 NT
S=N _ S=N — N ve HN !
HN-
BL-5786 6 6 6) oY LU505465
Ve Ve he a. / o Hg Ne a 0 Hg Ne J 9 To Mo X 0 To de > N: H Ca ) = H CE 9 N= ob a “ N= oF ©
RE SEE ERE Sd Rw 0 6) 0 © 0 fo oy ad 09 NZ
SANS SN à Ÿ y" 99 / 0 "Ay 0
J 0 Mg ne J 0 Hg em J © 9 A 7 © N= H ea
HEE EN LES dé CH AO à de 9, 61 ole) ole 0
TDR À LCR 3 NLS o 0 © A à J 0 & y" + x ©) yt 7 oe oN oe oN Sh oN
In yet another aspect, the present invention also provides a pharmaceutical composition, which includes at least one of the aforementioned compounds or pharmaceutically acceptable salts, isotope derivatives, stereoisomers thereof.
In yet another aspect, the invention also provides use of the aforementioned compounds or the pharmaceutically acceptable salts, isotope derivatives, stereoisomers or pharmaceutical compositions thereof in the preparation of medicine for preventing and/or treating cancer, tumor, inflammatory disease, autoimmune disease or immune mediated disease.
In yet another aspect, the invention also provides a method for preventing and/or treating cancer, tumors, inflammatory diseases, autoimmune diseases, or immune- mediated diseases, comprising administering to a patient in need a therapeutically effective amount of the aforementioned compounds or pharmaceutically acceptable salts, isotope derivatives, stereoisomers thereof and/or pharmaceutical composition.
It should be noted that, in this article, when referring to the "compound" of formula (D, formula (II), formula (III) and formula (IV), it generally also covers its stereoisomers, diastereomers,enantiomers, racemic mixtures and isotopic derivatives.
It is well known to those skilled in the art that a compound's salt, solvate, and hydrate are alternative forms of the compound, and they can all be converted into the compound under certain conditions. When referring to the compounds of formula (I) , formula (II), formula (III) and formula (IV), generally, their pharmaceutically acceptable salts are also included, and furthermore, their solvates and hydrates are also included. 15
BL-5786
Similarly, when referring to a compound herein, its prodrugs, metabolites and LUS05465 nitroxides are also generally included.
The pharmaceutically acceptable salts of the present invention may be formed using, for example, the following inorganic or organic acids. "Pharmaceutically acceptable salt" means a salt which, within the scope of reasonable medical judgment, is suitable for use in contact with tissues of humans and lower animals, without undue toxicity, irritation, allergic reaction, etc., can be called a reasonable benefit / risk ratio.
The salts can be prepared in situ during the final isolation and purification of the compounds of the invention, or alone by reacting the free base or acid with a suitable reagent, as outlined below. For example, a free base function can be reacted with a suitable acid. Examples of pharmaceutically acceptable inorganic acid addition salts are amino acids with inorganic acids (e.g., hydrochloric, hydrobromic, phosphoric, sulfuric, and perchloric) or organic acids (e.g., acetic, oxalic, maleic, tartaric, lemon acid, succinic acid or malonic acid), or by using other methods known in the art such as ion exchange. Other pharmaceutically acceptable salts include adipate, sodium alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, lauryl sulfate, ethylate, formate, fumarate, glucoheptonate, glycerin phosphate, gluconate, hernisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectate, persulfate, 3-phenylpropionate, phosphate, bitter salt, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluene sulfonate, undecanoate, valerate, etc. Representative alkali or alkaline earth metal salts include those of sodium, lithium, potassium, calcium, magnesium, and the like. Other pharmaceutically acceptable salts include, where appropriate, nontoxic ammonium salts, quaternary ammonium salts, and amine cations formed with counterions, for example, halides, hydroxides, carboxylates, sulfates, phosphates, nitrates, lower alkylsulfonates and arylsulfonates.
The pharmaceutically acceptable salts of the present invention can be prepared by conventional methods, for example, by dissolving the compound of the present invention in a water-miscible organic solvent (such as acetone, methanol, ethanol and acetonitrile), adding an excess of organic acid or inorganic acid aqueous solution, so 16
BL-5786 that the salt is precipitated from the resulting mixture, the solvent and remaining free LUS05465 acid are removed therefrom, and the precipitated salt is isolated.
The precursors or metabolites described in the present invention may be precursors or metabolites known in the art, as long as the precursors or metabolites are transformed into compounds through in vivo metabolism. For example, "prodrugs" refer to those prodrugs of the compounds of the present invention which, within the scope of sound medical judgment, are suitable for use in contact with human and lower animal tissues without undue toxicity, irritation, allergic response, etc., qualified as having a reasonable benefit/risk ratio and valid for its intended use. The term "prodrug" refers to a compound that is rapidly transformed in vivo to yield the parent compound of the above formula, for example by in vivo metabolism, or N-demethylation of a compound of the invention. "Solvate" as used herein means a physical association of a compound of the present invention with one or more solvent molecules, whether organic or inorganic. This physical association includes hydrogen bonding. In some cases, for example, when one or more solvent molecules are incorporated into the crystal lattice of a crystalline solid, solvates will be able to be isolated. Solvent molecules in solvates may exist in regular and/or disordered arrangements. Solvates may contain stoichiometric or non- stoichiometric amounts of solvent molecules. "Solvate" encompasses both solution- phase and isolatable solvates. Exemplary solvates include, but are not limited to, hydrates, ethanolates, methanolates, and isopropanolates. Solvation methods are well known in the art.
The "stereoisomerism" described in the present invention is divided into conformational isomerism and configurational isomerism, and configurational isomerism can also be divided into cis-trans isomerism and optical isomerism (that is, optical isomerism). Due to the rotation or twisting of carbon and carbon single bonds in organic molecules of a certain configuration, a stereoisomerism phenomenon in which each atom or atomic group of the molecule has a different arrangement in space, the common structures are alkanes and cycloalkanes. Such as the chair conformation and boat conformation that appear in the structure of cyclohexane. "Stereoisomer" means when a compound of the present invention contains one or more asymmetric centers and is thus available as racemates and racemic mixtures, single enantiomers, diastereomeric mixtures and single diastereomers. The compound of the present 17
BL-5786 invention has an asymmetric center, and each asymmetric center can produce two LUS05465 optical isomers, and the scope of the present invention includes all possible optical isomers and diastereoisomer mixtures and pure or partially pure compounds . The compounds described herein may exist in tautomeric forms having different points of attachment of hydrogens by displacement of one or more double bonds. For example, a ketone and its enol form are keto-enol tautomers. Each tautomer and mixtures thereof are included in the compounds of the present invention. All enantiomers, diastereoisomers, racemates, mesoforms, cis-trans isomers, tautomers, geometric isomers, epimers of compounds of formula (I) to formula (IV) and mixtures thereof are included in the scope of the present invention.
An "isotopic derivative" of the present invention refers to a molecule in which a compound is isotopically labeled. Isotopes commonly used for isotopic labeling are: hydrogen isotopes, 2H and *H; carbon isotopes: ''C, 1*C and '*C; chlorine isotopes: CI and ?’Cl; fluorine isotopes: !$F; iodine isotopes: !°I and !?°T; nitrogen isotopes: '’N and °N ; Oxygen isotopes: !°O, 70 and !®O and sulfur isotope ‘PS. These isotope-labeled compounds can be used to study the distribution of pharmaceutical molecules in tissues.
Especially deuterium *H and carbon ‘°C are more widely used due to their easy labeling and convenient detection. Substitution of certain heavy isotopes, such as deuterium (CH), can enhance metabolic stability, prolong half-life and thus achieve the purpose of reducing doses and provide therapeutic advantages. Isotopically labeled compounds are generally synthesized starting from labeled starting materials and carried out in the same way as non-isotopically labeled compounds using known synthetic techniques.
The present invention also provides the use of the compound of the present invention in the preparation of medicaments for preventing and/or treating cancer, tumor, inflammatory disease, autoimmune disease or immune-mediated disease.
In addition, the present invention provides a pharmaceutical composition for preventing and/or treating cancer, tumor, inflammatory disease, autoimmune disease, neurodegenerative disease, attention-related disease or immune-mediated disease, which comprises the present invention compounds as active ingredients. The pharmaceutical composition may optionally comprise a pharmaceutically acceptable carrier.
In addition, the present invention provides a method of preventing and/or treating 18
BL-5786 cancer, tumor, inflammatory disease, autoimmune disease, neurodegenerative disease, LU505465 attention-related disease or immune-mediated disease, which comprises administering the compound of the invention to the mammals in need thereof.
Representative examples of inflammatory, autoimmune, and immune-mediated diseases may include, but are not limited to, arthritis, rheumatoid arthritis, spondyloarthritis, gouty arthritis, osteoarthritis, juvenile arthritis , Other Arthritis
Conditions, Lupus, Systemic Lupus Erythematosus (SLE), Skin Related Disorders,
Psoriasis, Eczema, Dermatitis, Atopic Dermatitis, Pain, Pulmonary Disease, Lung
Inflammation, Adult Respiratory Distress Syndrome (ARDS) , pulmonary sarcoidosis, chronic pulmonary inflammatory disease, chronic obstructive pulmonary disease (COPD), cardiovascular disease, atherosclerosis, myocardial infarction, congestive heart failure, myocardial ischemia-reperfusion injury, inflammatory bowel disease,
Crohn's disease, ulcerative colitis, irritable bowel syndrome, asthma, Sjogren's syndrome, autoimmune thyroid disease, urticaria (rubella), multiple sclerosis, scleroderma, organ transplant rejection, xenograft, idiopathic thrombocytopenic purpura (ITP), Parkinson's disease, Alzheimer's disease, diabetes-related diseases, inflammation, pelvic inflammatory disease, allergic rhinitis, allergic bronchitis, allergic sinusitis, leukemia, lymphoma, B Cell Lymphoma, T Cell Lymphoma, Myeloma, Acute
Lymphocytic Leukemia (ALL), Chronic Lymphocytic Leukemia (CLL), Acute
Myeloid Leukemia (AML), Chronic Myelogenous Leukemia (CML), Hairy Cell
Leukemia, He Jie King's disease, non-Hodgkin's lymphoma, multiple myeloma, myelodysplastic syndrome (MDS), myeloproliferative neoplasm (MPN), diffuse large
B-cell lymphoma, and follicular lymphoma.
Representative examples of cancer or tumor may include, but are not limited to, skin cancer, bladder cancer, ovarian cancer, breast cancer, stomach cancer, pancreatic cancer, prostate cancer, colon cancer, lung cancer, bone cancer, brain cancer, neuroblastoma, rectal cancer, colon cancer, familial adenomatous polyposis carcinoma, hereditary nonpolyposis colorectal cancer, esophagus cancer, lip cancer, larynx cancer, hypopharyngeal cancer, tongue cancer, salivary gland cancer, stomach cancer, adenocarcinoma, medullary thyroid cancer, Papillary thyroid cancer, renal cancer, renal parenchymal cancer, ovarian cancer, cervical cancer, uterine body cancer, endometrial cancer, choriocarcinoma, pancreatic cancer, prostate cancer, testicular cancer, urinary cancer, melanoma, brain tumors such as Glioblastoma, astrocytoma, meningioma, 19
BL-5786 medulloblastoma and peripheral neuroectodermal tumor, Hodgkin lymphoma, non- LUS05465
Hodgkin lymphoma, Burkitt lymphoma, acute lymphoblastic leukemia ( ALL), chronic lymphocytic leukemia (CLL), acute myeloid leukemia (AML), chronic myelogenous leukemia (CML), adult T-cell leukemia lymphoma, diffuse large B-cell lymphoma (DLBCL), hepatocellular carcinoma, gallbladder Carcinoma, bronchial carcinoma, small cell lung cancer, non-small cell lung cancer, multiple myeloma, basal cell tumor, teratoma, retinoblastoma, choroidal melanoma, seminoma, rhabdomyosarcoma, craniopharyngioma, Osteosarcoma, chondrosarcoma, sarcoma, liposarcoma, fibrosarcoma, Ewing sarcoma, or plasmacytoma.
When the compound of the present invention or a pharmaceutically acceptable salt thereof is administered in combination with another anticancer agent or immune checkpoint inhibitor for the treatment of cancer or tumors, the compound of the present invention or a pharmaceutically acceptable salt thereof can provide enhanced anticancer effects .
Representative examples of anticancer agents useful in the treatment of cancer or tumors may include, but are not limited to, cell signal transduction inhibitor, chlorambucil, melphalan, cyclophosphamide, ifosfamide, busulfan, carmustine, lomustine, streptozotocin, cisplatin, carboplatin, oxaliplatin, dacarbazine, temozolomide, procarbazine, methotrexate, fluorouracil, cytarabine, gemcitabine,
Mercaptopurine, fludarabine, vinblastine, vincristine, vinorelbine, paclitaxel, docetaxel, topotecan, irinotecan, etoposide, trabectedin, dactinomycin, doxorubicin , epirubicin, daunorubicin, mitoxantrone, bleomycin, mitomycin C, ixabepilone, tamoxifen, flutamide, gonadorelin analogs, methadone progesterone, prednisone, dexamethasone, methylprednisolone, thalidomide, interferon alpha, leucovorin, sirolimus, sirolimus ester, everolimus, afatinib, alisertib, amuvatinib, apatinib, axitinib, bortezomib, bosutinib, britinib, cabozantinib, cediranib, crenolanib, kezhuotinib, dabrafenib, dabrafenib, Cotinib, danucitinib, dasatinib, dovitinib, erlotinib, foretinib, ganetespib, gefitinib, ibrutinib, icotinib, imatinib, iniparib, la Patinib, lenvatinib, linifanib, linsitinib, masitinib, momelotinib, motisanib, neratinib, nilotinib, niraparib, oprozomib, olaparib, pazopanib, pictilisib, ponatinib, quizartinib, regorafenib, rigosertib, rucaparib, ruxolitinib, saracatinib, saridegib, sorafenib, sunitinib, tiratinib, tivantinib, tivozanib, tofacitinib, trametinib, vandetanib, veliparib, vemurafenib, vimodegib, volasertib, alemtuzumab, bevacizumab, berentuzumab vedotin, 20
BL-5786 catumaxumab Antibodies, cetuximab, denosumab, gemtuzumab, ipilimumab, LU505465 nimotuzumab, ofatumumab, panitumumab, rituximab, tositumomab, trastuzumab,
PI3K inhibitor, CSF1R inhibitor, A2A and/or A2B receptor antagonist, IDO inhibitor, anti-PD-1 antibody, anti-PD-L1 antibody, LAG3 antibody, TIM-3 antibody and an anti-
CTLA-4 antibody, or any combination thereof.
When administered in combination with other therapeutic agents for the treatment of inflammatory disease, autoimmune disease and immune-mediated disease, the compounds of the present invention, or pharmaceutically acceptable salts thereof provide enhanced therapeutic effect.
Representative examples of therapeutic agents useful in the treatment of inflammatory, autoimmune, and immune-mediated diseases can include, but are not limited to, steroidal agents (eg, prednisone, prednisone, prednisone, methylphenidate, cortisone, hydroxycortisone, betamethasone, dexamethasone, etc.), methotrexate, leflunomide, anti-TNFa agents (eg, etanercept, infliximab, adalib monoclonal antibody, etc.), calcineurin inhibitors (e.g., tacrolimus, pimecrolimus, etc.), and antihistamines (e.g., diphenhydramine, hydroxyzine, loratadine, ebastine, ketotifen, cetirizine, levocetirizine, fexofenadine, etc.), and at least one or more therapeutic agents selected from them can be included in the pharmaceutical composition of the present invention.
In addition, the present invention also provides a method for preventing and/or treating tumors, cancers, viral infections, organ transplant rejection, neurodegenerative diseases, attention-related diseases or autoimmune diseases, which comprises administering the compound of the invention or the pharmaceutical composition of the invention to a mammal in need thereof.
The pharmaceutical composition of the present invention can be formulated into dosage forms for oral administration or parenteral administration (including intramuscular, intravenous and subcutaneous routes, intratumoral injection) according to any of the conventional methods, such as tablets, granules, powders , capsules, syrups, emulsions, microemulsions, solutions or suspensions.
Pharmaceutical compositions of the present invention for oral administration can be prepared by mixing the active ingredient with carriers such as cellulose, calcium silicate, corn starch, lactose, sucrose, dextrose, calcium phosphate, stearic acid, stearic 21
BL-5786 acid, magnesium stearate, calcium stearate, gelatin, talc, surfactant, suspending agent, LU505465 emulsifier and diluent.
Examples of carriers employed in the pharmaceutical composition for injection administration of the present invention can be water, saline solution, glucose solution, glucose-like solution (glucose-like solution), alcohol, glycol, ether (for example, polyethylene glycol 400 ), oils, fatty acids, fatty acid esters, glycerides, surfactants, suspending agents and emulsifiers.
Other features of the invention will become apparent in the course of the description of exemplary embodiments of the invention which are given to illustrate the invention and are not intended to be limiting thereof. In the following examples the methods disclosed in the invention were used for preparation, separation and characterization.
The compounds of the present invention can be prepared in a variety of methods known to those skilled in the art of organic synthesis, the methods described below as well as synthetic methods known in the art of synthetic organic chemistry or by variations thereof known to those skilled in the art may be used for the synthesis of the compounds of the present invention. Preferred methods include, but are not limited to, those described below. Reactions are performed in solvents or solvent mixtures appropriate to the kit materials used and to the transformations effected. Those skilled in the art of organic synthesis will appreciate that the functionality present on the molecule is consistent with the proposed transitions. This sometimes requires judgment to alter the order of synthetic steps or starting materials to obtain the desired compound of the invention.
Embodiments
Terms
Unless otherwise stated, the terms used in the present application, including the specification and claims, are defined as follows. If not stated otherwise, conventional methods of mass spectrometry, NMR, HPLC, protein chemistry, biochemistry, recombinant DNA techniques and pharmacology are used. In this application, the use of "or" or "and" means "and/or" if not stated otherwise.
In the specification and claims, a given chemical formula or name shall encompass all stereoisomers and optical isomers thereof and racemates in which such isomers exist. 22
BL-5786
Unless otherwise indicated, all chiral (enantiomers and diastereoisomers) and racemic LUS05465 forms are within the scope of the invention. Various geometric isomers of C=C double bonds, C=N double bonds, ring systems, etc. may also exist in the compounds, and all such stable isomers are encompassed by the present invention. The present invention describes cis- and trans- (or E- and Z-) geometric isomers of the compounds of the invention and which may be isolated as a mixture of isomers or as separated isomeric forms. The compounds of the invention may be isolated in optically active or racemic forms. All methods used to prepare the compounds of the invention and intermediates prepared therein are considered part of the invention. When preparing enantiomeric or diastereomeric products, they may be separated by customary methods, for example by chromatography or fractional crystallization. Depending on the process conditions, the end products of the invention are obtained in free (neutral) or salt form. The free forms and salts of these end products are within the scope of the present invention. A compound can be converted from one form to another, if desired. A free base or acid can be converted into a salt; a salt can be converted into the free compound or another salt; a mixture of isomeric compounds of the invention can be separated into the individual isomers. The compounds of the invention, their free forms and salts, may exist in various tautomeric forms in which the hydrogen atoms are transposed to other parts of the molecule and thus the chemical bonds between the atoms of the molecule are rearranged. It is to be understood that all tautomeric forms which may exist are included within the present invention.
In the present invention, when the listed connecting groups do not specify the connecting direction, the connecting direction is arbitrary, for example when the L in
OL) is -C(O)NH-, -C(O)NH- can both connect phenyl and cyclohexyl in the
OX reading order from left to right to form NC). and connect phenyl and an cyclohexyl in the opposite reading order from left to right to form 70 The combination of the linking group and the connected group is only allowed if it results in a stable compound. In some preferred embodiments of the present invention, the reading order is from left to right.
Unless otherwise defined, the definitions of the substituents in the present invention are independent and not interrelated, for example (listed but not exhaustive), 23
BL-5786 in one aspect, for the substituents Ra (or Ra”) in different definitions of the substituents, LUS05465 the definitions are independent of each other. Specifically, when one definition is selected for R* (or R¥) in one substituent, it does not mean that R* (or R*) has the same definition in other substituents. More specifically, for example (but not exhaustively) for NR°R*, when R? (or R?) is defined from hydrogen, it does not mean that in -C(O)-
NR*R?, R* (or R¥) must be hydrogen. In another aspect, when there is more than one
R? (or R?) in a certain substituent, these R* (or R*) are also independent. For example, in the substituent -(CR*R*)m-O-(CR*R*)n-, when m+n is greater than or equal to 2, the m+n pieces of R* (or R*) are independent, and they can be have the same or different meanings.
Unless otherwise defined, when a substituent is noted as "optionally substituted", the substituent is selected from, for example, the following substituents, such as alkyl, cycloalkyl, aryl, heterocyclyl, halogen, hydroxy, alkoxy, oxo, alkanoyl, aryloxy, alkanoyloxy, amino, alkylamino, arylamino, arylalkylamino, disubstituted amino (wherein 2 amino substituents are selected from alkyl radical, aryl or arylalkyl), alkanoylamino, aroylamino, aralkanoylamino, substituted alkanoylamino, substituted arylamino, substituted aralkanoylamino, thio, alkylthio, arylthio, arylalkylthio, arylthiocarbonyl, arylalkylthiocarbonyl, alkylsulfonyl, arylsulfonyl, arylalkylsulfonyl, aminosulfonyl such as -SO2NH2, substituted sulfonylamino, nitro, cyano, carboxyl, carbamoyl such as -CONH2, substituted carbamoyl such as -CONHalkyl, -CONHaryl, -CONHarylalkyl or two optional In the case of substituents from alkyl, aryl or arylalkyl, alkoxycarbonyl, aryl, substituted aryl, guanidino, heterocyclic, e.g. indolyl, imidazolyl, furyl, thienyl , Thiazolyl, pyrrolidinyl, pyridyl, pyrimidinyl, pyrrolidinyl, piperidinyl, morpholinyl, piperazinyl, homopiperazinyl, etc. and substituted heterocyclyl.
The term "alkyl" as used herein is intended to include branched and straight chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms.
For example, "C1-C6 alkyl" means an alkyl group having 1 to 6 carbon atoms. Examples of alkyl groups include, but are not limited to, methyl (Me), ethyl (Et), propyl (such as n-propyl and isopropyl), butyl (such as n-butyl, isobutyl, tert-butyl), and pentyl (e.g. n- pentyl, isopentyl, neopentyl). Alkyl may be unsubstituted or substituted, and when substituted, it may be substituted at any available point of attachment, the substituents being preferably selected from one or more of deuterium, halogen, hydroxyl, amino, cyano, alkyl, alkoxy, haloalkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl. In this context, the alkyl group is preferably an alkyl group having 1 to 6, more preferably 1 to 24
BL-5786 4 carbon atoms. LUS05465
The term "alkylene" as used herein is intended to include branched, straight-chain, saturated aliphatic hydrocarbon radicals having the specified number of carbon atoms, with or without cyclic alkyl groups, either from the same carbon atom or two different carbon atoms from the parent alkyl group. A residue derived from removing two hydrogen atoms from a carbon atom. For example, "Co-Cs alkylene" means an alkylene group having 0 (i.e, bonds), 1, 2, 3, 4, 5, or 6 carbon atoms. Examples of alkylene include, but are not limited to, methylene (-CH»-), ethylene (-CH:CHz-), propylene (e.g.,-(CH»)s-, -(CHCH3)CH;-, -(CHCH.CH)-), butylene (eg, -(CHa)4-, -
CH:CH(CH:CH;)-, -CH,(CHCH:CH)- etc), pentylene (eg, -(CH»)s-, -
CH,CH(CH(CH3),)-, -CH,(CHCH:CH)CHz- etc), hexylene (eg, -(CH»)s-, -
CH,CH,CH(CH(CH:)»)-, -CH,(CHCH(CH;)CH)CHz-etc. In this disclosure, the alkylene group is preferably an alkylene group having 0-6, 0-4, 0-3, 1-6, 1-4, or 1-3 carbon atoms. In this disclosure, alkylene is preferably an alkylene group which does not contain a cyclic alkyl group.
The term "cycloalkyl" refers to a monocyclic, polycyclic or branched cyclic alkyl group. For example, Cz-C12 cyclic alkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and norbornyl. Branched cycloalkyl groups such as 1-methylcyclopropyl and 2-methylcyclopropyl are included in the definition of "cycloalkyl". Polycyclic, such as bicyclic and tricyclic cyclic alkyl groups include bridged, spiro or fused cyclic cycloalkyl groups. Cycloalkyl may be unsubstituted or substituted, and when substituted, it may be substituted at any available point of attachment, the substituents being preferably selected from one or more of halogen, hydroxyl, amino, cyano, oxo, alkyl, alkyl, oxy, haloalkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl. In the invention, the cycloalkyl group is preferably a C3-C12 cycloalkyl group, and more preferably a Cs-Cs cycloalkyl group.
Similarly, the term "heterocycloalkyl" refers to a one-membered cyclic structure in which at least one carbon atom of the cycloalkyl ring is replaced by a heteroatom selected from the group consisting of N, O, S and P. The N atoms may be optionally quaternized, and the N and S atoms may be optionally oxidized (i.e., NO, SO, and SO»).
It includes monocyclic heterocyclic, bicyclic heterocyclic and tricyclic heterocyclic systems, wherein the bicyclic heterocyclic and tricyclic heterocyclic systems include spirocyclic heterocyclic, paracyclic heterocyclic and bridged heterocyclic.
Heterocycloalkyl may be unsubstituted or substituted, and when substituted, it may be 25
BL-5786 substituted at any available point of attachment, the substituents being preferably LUS05465 selected from one or more of halogen, hydroxyl, amino, cyano, oxo, alkyl, alkoxy, haloalkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl. In the present invention, the heterocycloalkyl group is preferably a 4-12-membered heterocycloalkyl group, and more preferably a 4-8-membered heterocycloalkyl group.
In the present invention, the term “cyclic ring” refers to a polycyclic group formed by two or more cyclic structures sharing two adjacent atoms with each other.
In the present invention, the term “bridged ring” refers to a polycyclic group in which two rings in the system share two or more ring atoms.
In the present invention, the term “spiro ring” refers to a polycyclic group in which one carbon atom (called a spiro atom) is shared between single rings.
The term "alkenyl" denotes a straight or branched chain hydrocarbon group containing one or more double bonds and generally having a length of 2 to 20 carbon atoms. For example, "C,-Cs alkenyl" contains two to six carbon atoms. Alkenyl groups include, but are not limited to, e.g. vinyl, propenyl, butenyl, 1-methyl-2-buten-1-yl, and the like. Herein, alkenyl is preferably C,-Cs alkenyl.
The term "cycloalkenyl" refers to a monocyclic or bicyclic cyclic alkenyl group.
Monocyclic cyclic alkenyl refers to C3-Cg cyclic alkenyl, including but not limited to cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl and norbornenyl. Branched cycloalkenyl groups such as 1-methylcyclopropenyl and 2-methylcyclopropenyl are included in the definition of "cycloalkenyl". Bicyclic cyclic alkenyl groups include bridged, spiro or condensed ring cyclic alkenyl groups.
The term "alkynyl" denotes a straight or branched chain hydrocarbon group containing one or more triple bonds and generally having a length of 2 to 20 carbon atoms. For example, "C,-Cs alkynyl" contains two to six carbon atoms. Representative alkynyl groups include, but are not limited to, for example, ethynyl, 1-propynyl, 1- butynyl, and the like. Herein, the alkynyl group is preferably a C,-Cs alkynyl group.
The term "alkoxy" or "alkyloxy" refers to -O-alkyl. " C1-C6 alkoxy" (or alkyloxy) is intended to include C1, Cs, C3, Ca, Cs, Cs alkoxy. Examples of alkoxy include, but are not limited to, methoxy, ethoxy, propoxy (eg, n-propoxy and isopropoxy), and tert- butoxy. Herein, the alkoxy group is preferably an alkoxy group having 1 to 6, more preferably 1 to 4 carbon atoms. Similarly, "alkylthio" or "thioalkoxy" means a sulfur- bridged alkyl group as defined above having the specified number of carbon atoms; for example, methyl-S- and ethyl-S-. Alkoxy may be unsubstituted or substituted, and when 26
BL-5786 substituted, it may be substituted at any available point of attachment, the substituents LUS05465 being preferably selected from one or more of deuterium, halogen, hydroxyl, amino, cyano, alkyl, alkoxy, haloalkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl.
The term “carbonyl” refers to an organic functional group (C=0) consisting of two atoms, carbon and oxygen, linked by a double bond.
The term "aryl", alone or as part of a larger moiety such as "aralkyl", "arylalkoxy" or "aryloxyalkyl", refers to a single cyclic, bicyclic or tricyclic ring system having a total of 5 to 12 ring members, wherein at least one ring in the system is aromatic and wherein each ring in the system contains 3 to 7 ring members. In certain embodiments of the present invention, "aryl" refers to an aromatic ring system including, but not limited to, phenyl, biphenyl, indanyl, 1-naphthyl, 2-naphthyl, and tetralin base. The term "aralkyl" or "arylalkyl" refers to an alkyl residue attached to an aryl ring, non- limiting examples of which include benzyl, phenethyl, and the like. The fused aryl group can be attached to another group at a suitable position on the cycloalkyl ring or aromatic ring. The dashed lines drawn from the ring system indicate that bonds can be attached to any suitable ring atom. Aryl groups may be unsubstituted or substituted, and when substituted, they may be substituted at any available point of attachment, the substituents being preferably selected from one or more of deuterium, halogen, hydroxyl, amino, cyano, alkyl, alkoxy, haloalkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl.
The term "heteroaryl" means a stable 5-membered, 6-membered, or 7-membered aromatic monocyclic ring or aromatic bicyclic ring or a 7-membered, 8-membered, 9- membered, 10-membered, 11-membered, 12-membered aromatic polycyclic heterocycle, which is fully unsaturated or partially unsaturated, and contains carbon atomsand 1, 2, 3 or 4 heteroatoms independently selected from N, O and S; and includes cycloalkane or heterocycloalkane with a structure in which aromatic rings such as benzene rings or heteroaromatic rings such as pyridine are condensed. The position of the structure as a substituent may be located on a cycloalkane, a heterocycloalkane, an aromatic ring or a heteroaryl ring. Nitrogen and sulfur heteroatoms can be optionally oxidized. The nitrogen atom is substituted or unsubstituted (i.e. N or NR, where R is H or another substituent if defined). A heterocycle can be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure. The heterocyclyl groups described herein may be substituted on carbon or nitrogen atoms if the resulting compound is stable. The nitrogen in the heterocycle can optionally be quaternized. 27
BL-5786
Preferably, when the total number of S and O atoms in the heterocycle exceeds 1, then LUS05465 these heteroatoms are not adjacent to each other. Preferably, the total number of S and
O atoms in the heterocycle is not greater than one. Heteroaryl groups may be unsubstituted or substituted, and when substituted, they may be substituted at any available point of attachment, the substituents being preferably selected from one or more of halogen, hydroxyl, amino, cyano, alkyl, alkoxy, haloalkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl. When the term "heterocycle" is used, it is intended to include heteroaryl. Examples of heteroaryl groups include, but are not limited to, acridinyl, azetidinyl, aziocinyl, benzimidazolyl, benzofuryl, benzothiofuranyl, benzothienyl, benzooxa azolyl, benzoxazolyl, benzothiazolyl, benzotriazolyl, benzotetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolyl, carbazolyl, 4aH-carbazolyl, carbolinyl, chromanyl, chromenyl, cinnolinyl, decahydroquinolyl, 2H,6H-1,5,2-dithiazinyl, dihydrofuro[2, 3-b] tetrahydrofuryl, furyl, furanyl, imidazolidinyl, imidazolinyl, imidazolyl, 1H-indazolyl, imidazopyridyl, indolenyl, indolinyl, Indolazinyl, indolyl, 3H-indolyl, isatinoyl, isobenzofuryl, isochromanyl, isoindazolyl, isoindolinyl, isoindolyl, isoindolyl quinolinyl, isothiazolyl, isothiazolopyridyl, isoxazolyl, isoxazolopyridyl, methylenedioxyphenyl, morpholinyl, diazanaphthyl, octahydroisoquinoline oxadiazolyl, 1,2,3-oxadiazolyl, 1,2,4- oxadiazolyl, 1,2,5-oxadiazolyl, 1,3,4-oxadiazolyl , oxazolidinyl, oxazolyl, oxazolopyridyl, oxazolidinyl, diazaphenyl, oxindolyl, pyrimidinyl, phenanthridinyl, phenanthrolinyl, phenazinyl , phenothiazinyl, phenoxathiyl, phenoxazinyl, phthalazinyl, piperazinyl, piperidinyl, piperidinyl, 4-piperidinyl, piperonyl, pteridinyl, purinyl, pyranyl, pyrazinyl, pyrazolidinyl, pyrazolinyl, pyrazolopyridyl, pyrazolyl, pyridazinyl, pyridoxazolyl, pyridimidazolyl, pyridothiazolyl, pyridyl , pyrimidinyl, pyrrolidinyl, pyrrolinyl, 2-pyrrolidonyl, 2H-pyrrolyl, pyrrolyl, quinazolinyl, quinolinyl, 4H-quinazinyl, quinoxalinyl, quinuclidinyl, tetrazolyl, tetrahydrofuryl, tetrahydroisoquinolyl, tetrahydroquinolyl, 6H-1,2,5-thiadiazinyl, 1,2,3-thiadiazolyl, 1,2,4-thiadiazolyl, 1,2,5-thiadiazolyl, 1,3,4-thiadiazolyl, thianthryl, thiazolyl, thienyl, thiazolopyridyl, thienothiazolyl, thienooxa Azolyl, thienoimidazolyl, thienyl, triazinyl, 1,2,3-triazolyl, 1,2,4-triazolyl, 1,2,5-triazolyl, 1,3,4-triazolyl and xanthenyl, quinolinyl, isoquinolyl, phthalazinyl, quinazolinyl, indolyl, isoindolyl, indolinyl, 1H-indazolyl, benzo Imidazolyl, 1,2,3,4-tetrahydroquinolinyl, 1,2,3,4-tetrahydroisoquinolinyl, 5,6,7,8-tetrahydro-quinolinyl, 2,3 -Dihydro-benzofuryl, chromanyl, 1,2,3,4-tetrahydro- quinoxalinyl and 1,2,3,4-tetrahydro-quinazolinyl. The term "heteroaryl" may also 28
BL-5786 include biaryl structures formed by the above-defined "aryl" and a monocyclic LUS05465 "heteroaryl", such as but not limited to "-phenyl bipyridyl-", "- "phenyl bipyrimidyl", "-pyridyl biphenyl", "-pyridyl bipyrimidyl-", "-pyrimidyl biphenyl-"; wherein the present invention also includes condensed ring and spiro compound containing, for example, the above-mentioned heterocycles.
The term "substituted" as used herein means that at least one hydrogen atom is replaced by a non-hydrogen group, provided that normal valences are maintained and that the substitution results in a stable compound. A cyclic double bond, as used herein, is a double bond formed between two adjacent ring atoms (e.g., C=C, C=N or N=N).
In this disclosure, one or more halogens may be each independently selected from fluorine, chlorine, bromine, and iodine. "Halo" or "halogen" includes fluoro, chloro, bromo and iodo. "Haloalkyl"/"haloalkylene" is intended to include branched and straight chain saturated alkyl/alkylene groups having the indicated number of carbon atoms substituted with one or more halogens. Examples of haloalkyl include, but are not limited to, fluoromethyl, difluoromethyl, trifluoromethyl, trichloromethyl, pentafluoroethyl, pentachloroethyl, 2,2 2-trifluoroethyl, heptafluoro propyl and heptachloropropyl.
Examples of haloalkyl also include "fluoroalkyl" intended to include branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms and substituted with 1 or more fluorine atoms. "Halocycloalkyl"/"haloheterocycloalkyl" is intended to include cycloalkyl/heterocycloalkyl having the indicated number of carbon atoms substituted with one or more halogens. In the present invention, the halogen atom is preferably fluorine or chlorine, more preferably fluorine. "Haloalkoxy" or "haloalkyloxy" means a haloalkyl group as defined above having the indicated number of carbon atoms attached through an oxygen bridge. For example, "haloC1-C6 alkoxy" is intended to include Ci, Cs, Cs, Ca, Cs, Cs haloalkoxy. Examples of haloalkoxy include, but are not limited to, trifluoromethoxy, 2,2, 2-trifluoroethoxy, and pentafluoroethoxy. Similarly, "haloalkylthio" or "thiohaloalkoxy" denotes a haloalkyl group as defined above having the indicated number of carbon atoms attached through a sulfur bridge; for example trifluoromethyl-S- and pentafluoroethyl -S-.
In the present disclosure, the expression Cx1-Cx is used when referring to some substituent groups, which means that the number of carbon atoms in the substituent groups may be x1 to x2. For example, Co-Cs means that the group contains 0, 1, 2, 3, 4, 29
BL-5786 5, 6, 7 or 8 carbon atoms, C;-Cg means that the group contains 1, 2, 3,4, 5,6, 7or 8 LU505465 carbon atoms, C,-Cg means that the group contains 2, 3, 4, 5, 6, 7 or 8 carbon atoms,
C3-Cg means that the group contains 3, 4, 5 , 6, 7 or 8 carbon atoms, C4-Cg means that the group contains 4, 5, 6, 7 or 8 carbon atoms, Co-Cs means that the group contains 0, 1,2,3,4, 5 or 6 carbon atoms, C1-C¢ means that the group contains 1, 2, 3, 4, Sor 6 carbon atoms, C2-Cs means that the group contains 2, 3, 4, 5 or 6 carbon atoms, C3-C6 means that the group contains 3, 4, 5 or 6 carbon atoms.
In this disclosure, the expression "x1-x2 membered ring" is used when referring to cyclic groups such as aryl, heteroaryl, cycloalkyl, and heterocycloalkyl, which means that the number of the ring atoms of the group can be x1 to x2. For example, the 3-12 membered cyclic group may be a 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12 membered ring, and the number of ring atoms may be 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12; 3-6-membered ring means that the cyclic group can be 3, 4, 5 or 6-membered ring, and the number of ring atoms can be 3, 4, 5 or 6 ; 3-8 membered ring means that the cyclic group can be 3, 4, 5, 6, 7 or 8-membered ring, and the number of ring atoms can be 3, 4, 5, 6, 7 or 8; 3-9 membered ring means that the cyclic group can be a 3, 4, 5, 6, 7, 8 or 9-membered ring, and the number of ring atoms can be 3, 4, 5, 6, 7, 8 or 9; 4-7 membered ring means that the cyclic group can be a 4, 5, 6 or 7-membered ring, and the number of ring atoms can be 4, 5, 6 or 7; 5-8-membered ring means that the cyclic group can be 5, 6, 7 or 8- membered ring, the number of ring atoms can be 5, 6, 7 or 8; 5-12 membered ring means that the ring group can be 5, 6, 7, 8, 9, 10, 11 or 12-membered ring, the number of ring atoms can be 5, 6, 7, 8, 9, 10, 11 or 12; 6-12 membered ring means that the ring group can be 6, 7, 8, 9, 10, 11- or 12-membered rings, the number of ring atoms may be 6, 7, 8, 9, 10, 11 or 12. The ring atoms may be carbon atoms or heteroatoms, for example selected from N, O and S. When the ring is heterocyclic, the heterocyclic ring may contain 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more ring heteroatoms, for example selected from
N, O and S of heteroatoms.
Where nitrogen atoms (e.g. amines) are present on compounds of the invention, these nitrogen atoms can be converted to N-oxides by treatment with oxidizing agents (e.g. mCPBA and/or hydrogen peroxide) to obtain other compounds of the invention .
Accordingly, both shown and claimed nitrogen atoms are considered to cover both the shown nitrogen and its N-oxides to obtain the derivatives of the present invention.
When any variable occurs more than one time in any composition or formula of a compound, its definition on each occurrence 1s independent of its definition at every 30
BL-5786 other occurrence. Thus, for example, if a group is shown to be substituted with 0-3 R, LUS05465 then said group may be optionally substituted with up to three R groups, and R at each occurrence is independently selected from the definition of R. Also, combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
The term "patient" as used herein refers to an organism being treated by the methods of the present invention. Such organisms preferably include, but are not limited to, mammals (e.g., murine, ape, monkey, equine, bovine, porcine, canine, feline, etc.) and most preferably refer to humans.
The term "effective amount" as used herein means an amount of a drug or agent (i.e., a compound of the invention) that will elicit a biological or medical response in a tissue, system, animal or human being sought, e.g, by a researcher or clinician.
Furthermore, the term "therapeutically effective amount" means an amount which results in improved treatment, cure, prevention or alleviation of a disease, disorder or side effect, or a reduction in the rate of disease or disorder progression. An effective amount may be given in one or more administrations, applications or doses and is not intended to be limited by a particular formulation or route of administration. The term also includes within its scope amounts effective to enhance normal physiological function.
As used herein, the term "treating" includes any effect that results in amelioration of a condition, disease, disorder, etc., such as alleviation, reduction, regulation, amelioration or elimination, or amelioration of the symptoms thereof.
The term "pharmaceutically acceptable" is used herein to refer to those compounds, substances, compositions and/or dosage forms: within the scope of sound medical judgment, they are suitable for use in contact with human and animal tissues without excessive toxicity, irritation, allergic reactions, and/or other problems or complications, commensurate with a reasonable benefit/risk ratio.
The phrase "pharmaceutically acceptable carrier" or “pharmaceutical carrier” as used herein means a pharmaceutical substance, composition or vehicle, such as a liquid or solid filler, diluent, excipient, manufacturing aid (e.g. lubricant, talc, magnesium stearate, calcium stearate, or zinc stearate, or stearic acid) or solvent-encapsulated substances involved in the carrying or transport of a subject compound from one organ or body part to another. Each carrier must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient. 31
BL-5786
The term "pharmaceutical composition" means a composition comprising a LUS05465 compound of the present invention together with at least one other pharmaceutically acceptable carrier. "Pharmaceutically acceptable carrier" means a medium generally accepted in the art for delivering a biologically active agent to an animal, particularly a mammal, including (ie.) an adjuvant, excipient or vehicle, such as a diluent , preservatives, fillers, flow regulators, disintegrants, wetting agents, emulsifiers, suspending agents, sweeteners, flavoring agents, fragrances, antibacterial agents, antifungal agents, lubricants and dispersants, which depends on the mode of administration and the nature of the dosage form.
Certain pharmaceutical and medical terms
The term "acceptable", as used herein, means that a formulation ingredient or active ingredient does not have an undue adverse effect on health for the general purpose of treatment.
The term "cancer", as used herein, refers to an abnormal growth of cells that cannot be controlled and, under certain conditions, is capable of metastasizing (spreading).
Cancers of this type include, but are not limited to, solid tumors (e.g., bladder, bowel, brain, chest, uterus, heart, kidney, lung, lymphoid tissue (lymphoma), ovary, pancreas or other endocrine organs (e.g., thyroid), prostate , skin (melanoma), or blood cancer (such as non-leukemic leukemia).
The term "administration in combination" or similar terms, as used herein, refers to the administration of several selected therapeutic agents to a patient, in the same or different modes of administration at the same or different times.
The term "enhancing" or "capable of enhancing", as used herein, means that a desired result is capable of being increased or prolonged, either in potency or duration.
Thus, in relation to enhancing the therapeutic effect of a drug, the term "capable of potentiating” refers to the ability of the drug to increase or prolong its potency or duration in the system. As used herein, "potency value" refers to the ability to maximize the enhancement of another therapeutic drug in an ideal system.
The term "immune disease" refers to a disease or condition of an adverse or deleterious reaction to an endogenous or exogenous antigen. The result is usually dysfunction of the cells, or destruction thereof and dysfunction, or destruction of organs or tissues that may produce immune symptoms.
The terms "kit" and "product packaging" are synonymous.
The term "subject" or "patient" includes mammals and non-mammals. Mammals 32
BL-5786 include, but are not limited to, mammals: humans, non-human primates such as LU505465 orangutans, apes, and monkeys; agricultural animals such as cattle, horses, goats, sheep, and pigs, domestic animals such as rabbits and dogs; experimental animals include rodents, such as rats, mice and guinea pigs. Non-mammalian animals include, but are not limited to, birds, fish, and the like. In a preferred aspect, the selected mammal is a human.
The term "treatment", "course of treatment" or "therapy" as used herein includes alleviating, suppressing or ameliorating the symptoms or conditions of a disease; inhibiting the development of complications; ameliorating or preventing the underlying metabolic syndrome; inhibiting the development of diseases or symptoms, such as controlling the development of a disease or condition; alleviating a disease or a symptom; causing a disease or a symptom to regress; alleviating a complication caused by a disease or a symptom, or preventing and/or treating a sign caused by a disease or a symptom.
As used herein, a certain compound or pharmaceutical composition, after administration, can improve a certain disease, symptom or condition, especially improve its severity, delay the onset, slow down the progression of the disease, or reduce the duration of the disease. Circumstances that may be attributable to or related to the administration, whether fixed or episodic, continuous or intermittent.
Suitable routes of administration include, but are not limited to, oral, intravenous, rectal, aerosol, parenteral, ocular, pulmonary, transdermal, vaginal, ear canal , nasal administration and topical administration. In addition, by way of example only, parenteral administration includes intramuscular injection, subcutaneous injection, intravenous injection, intramedullary injection, intraventricular injection, intraperitoneal injection, intralymphatic injection, and intranasal injection.
In one aspect, the compounds described herein are administered locally rather than systemically. In certain embodiments, the depot formulation is administered by implantation (e.g., subcutaneously or intramuscularly) or by intramuscular injection.
Furthermore, in another specific embodiment, the drug is administered via a targeted drug delivery system. For example, liposomes coated with organ-specific antibodies. In such embodiments, the liposomes are selectively directed to and taken up by specific organs.
Pharmaceutical Composition and Dosage
The present invention also provides pharmaceutical compositions comprising a 33
BL-5786 therapeutically effective amount of one or more compounds of the present invention LUS05465 formulated together with one or more pharmaceutically acceptable carriers (additives) and/or diluents, and optionally one or more of the other therapeutic agents described above. The compounds of the present invention may be administered for any of the above uses by any suitable means, for example orally, such as tablets, pills, powders, granules, elixirs, tinctures, suspensions (including nanosuspensions, microsuspensions, spray-dried dispersions), syrups and emulsions; sublingually; buccally; parenterally, such as by subcutaneous, intravenous, intramuscular or intrasternal injection or infusion techniques (e.g., as sterile injectable aqueous or nonaqueous solutions or suspensions liquid form); nasally, including to the nasal membranes, such as by inhalation spray; topically, such as in the form of a cream or ointment; or rectally, such as in the form of a suppository; or by intratumoral injection. They can be administered alone, but generally will be administered using a pharmaceutical carrier selected on the basis of the chosen route of administration and standard pharmaceutical practice.
Pharmaceutical carriers include aqueous and non-aqueous liquid media and various solid and semisolid pharmaceutical carriers. Such carriers may include many different ingredients and additives besides the active agent, which are included in the formulation for various reasons known to those skilled in the art, such as stabilizers, binders, and the like. A description of suitable pharmaceutical carriers and the factors involved in the selection of the carrier can be found in several readily available sources, such as Allen L.V.Jr. et al. Remington: The Science and Practice of PharmaCy1l (2
Volumes), 22nd Edition (2012), Pharmaceutical Press.
Dosage regimens for the compounds of the present invention will of course vary depending on known factors such as the pharmacodynamic properties of the particular agent and its mode and route of administration; the species, age, sex, health, medical condition and weight of the recipient ; nature and extent of symptoms; type of concomitant therapy; frequency of therapy; route of administration, patient's renal and hepatic function, and desired effects. As a general guide, the daily oral dosage of each active ingredient should be from about 0.001 mg/day to about 10-5000 mg/day, preferably from about 0.01 mg/day to about 1000 mg/day, and most preferably From about 0.1 mg/day to about 250 mg/day. The most preferred dose intravenously will be about 0.01 mg/kg/minute to about 10 mg/kg/minute during a constant rate infusion. The compounds of the present invention may be administered in a single daily dose, or the total daily dose may be administered in divided doses of two, three or four times daily. 34
BL-5786
The compounds are usually formulated with a suitable pharmaceutical diluent, LUS05465 excipient or carrier (herein collectively referred to as drug carriers) in the form of mixtures for administration.
Dosage forms (pharmaceutical compositions) suitable for administration may contain from about 1 milligram to about 2000 milligrams of active ingredient per dosage unit. In these pharmaceutical compositions, the active ingredient will generally be present in an amount of about 0.1-95% by weight, based on the total weight of the composition.
Included within the scope of the present invention are pharmaceutical compositions comprising (alone or in combination with a pharmaceutical carrier) a therapeutically effective amount of at least one compound of the present invention as an active ingredient. Optionally, compounds of the invention may be used alone, in combination with other compounds of the invention, or in combination with one or more other therapeutic agents (e.g., anticancer agents or other pharmaceutically active substances).
Irrespective of the chosen route of administration, the compounds of the invention (which may be used in suitably hydrated form) and/or the pharmaceutical compositions of the invention are formulated into pharmaceutical dosage forms by conventional methods known to those skilled in the art.
The actual dosage levels of the active ingredients in the pharmaceutical compositions of the present invention can be varied to obtain an amount of the active ingredient that is effective in achieving the desired therapeutic response, composition, and mode of administration for a particular patient without being toxic to the patient.
The selected dosage level will depend on a variety of factors, including the activity of the particular compound of the invention employed, or its ester, salt or amide; the route of administration; the time of administration; the rate of excretion of the particular compound employed; and the rate and extent of absorption; duration of treatment; other drugs, compounds and/or substances used in combination with the particular compound used; factors well known in the medical art such as age, sex, weight, condition, general health and prior medical history of the patient being treated.
A physician or veterinarian having ordinary skill in the art can determine and prescribe the effective amount of the pharmaceutical composition required. In general, a suitable daily dose of a compound of the invention will be that amount of the compound at the lowest dose effective to produce a therapeutic effect. Such effective
BL-5786 dosage will generally depend on the factors mentioned above. Typically, oral, LUS05465 intravenous, intracerebroventricular and subcutaneous doses of the compounds of this invention to patients range from about 0.01 to about 50 mg/kg body weight/day. If desired, an effective daily dose of the active compound may be administered in two, three, four, five, six or more sub-doses at appropriate intervals throughout the day, optionally in unit dosage form. In certain aspects of the invention, the administration is once daily.
Although the compounds of the present invention may be administered alone, it is preferred to administer the compounds in the form of pharmaceutical formulations (compositions).
The above-mentioned features mentioned in the present invention, or the features mentioned in the embodiments can be combined arbitrarily. All the features disclosed in the specification of this case can be used in combination with any combination, and each feature disclosed in the specification can be replaced by any alternative feature that can provide the same, equivalent or similar purpose. Therefore, unless otherwise stated, the disclosed features are only general examples of equivalent or similar features.
Below in conjunction with specific embodiment, further illustrate the present invention. It should be understood that these examples are only used to illustrate the present invention and are not intended to limit the scope of the present invention. For the experimental methods without specific conditions indicated in the following examples, usually follow the conventional conditions or the conditions suggested by the manufacturer. All percentages, ratios, ratios, or parts are by weight unless otherwise indicated.
The unit of weight volume percentage in the present invention is well known to those skilled in the art, for example, it refers to the weight (g) of solute in 100 ml of solution. Unless otherwise defined, all professional and scientific terms used herein have the same meanings as are familiar to those skilled in the art. In addition, any methods and materials similar or equivalent to those described can be applied to the method of the present invention. The preferred implementation methods and materials described herein are for demonstration purposes only.
Example
General process 36
BL-5786
When the preparation route is not included, the raw materials and reagents used in LUS05465 the present invention are known products, which can be synthesized according to methods known in the art, or can be obtained by purchasing commercially available products. All commercially available reagents were used without further purification.
Room temperature means 20-30°C.
There is no special description in the reaction examples, and the reactions are all carried out under a nitrogen atmosphere. The nitrogen atmosphere refers to a nitrogen balloon of about 1 L connected to the reaction flask.
The hydrogenation reaction is usually vacuumized and filled with hydrogen, and the operation is repeated 3 times. The hydrogen atmosphere means that the reaction bottle is connected with a hydrogen balloon of about 1L.
Microwave reactions use the Biotage® Initiator+ Microwave Reactor.
The structures of the compounds of the present invention were determined by nuclear magnetic resonance (NMR) and mass spectroscopy (MS). NMR shifts (6) are given in units of 10° (ppm). The determination of NMR is to use (Bruker Ascend TM 500 type) nuclear magnetic analyzer, the measurement solvent is deuterated dimethyl sulfoxide (DMSO-d6), deuterated chloroform (CDCI3), deuterated methanol (CD3OD), and the internal standard is tetramethylsilane (TMS). The following abbreviations are used for the multiplicity of NMR signals: s = singlet, brs = broad singlet, d = doublet, t = triplet, m = multiplet. Coupling constants are listed as J values, measured in Hz.
As to reverse-phase preparative chromatography, a Thermo (UltiMate 3000) reverse-phase preparative chromatograph was used. The flash column chromatography uses Agela (FS-9200T) automatic column passing machine, and the silica gel prepacked column uses Santai SEPAFLASH® prepacked column. Yantai Huanghai HSGF254 or
Qingdao GF254 silica gel plates are used for thin-layer chromatography silica gel plates, and the specifications used for thin-layer chromatography separation and purification products are 0.4mm to 0.5mm.
LC-MS analysis method is as follows: 1) Mass spectrometry method: Thermo Fisher MSQ PLUS mass spectrometer, ESI source, positive ion mode. Ion source parameter settings: drying gas temperature is 350 °C; drying gas flow rate is 10 L/min; MS Range: 120-1000. 2) Liquid phase conditions: Chromatographic column: Waters XBridge (3.5um, 5S0mmx4.6mm); mobile phase A is an aqueous solution containing 0.1% ammonium 37
BL-5786 bicarbonate, mobile phase B is an acetonitrile solution, and perform linear gradient LUS05465 elution in the following table 1; Flow rate: 2 mL/min; column temperature: 30°C; UV detection wavelength: 214nm, 254nm, 280nm; injection volume 2uL.
Table 1. Gradient elution conditions
Tim mobile mobile min phase A/% phase B/% 0 9% 5 1.4 5 95 2.8 5 95 2.820 95 5 3 95 5
HPLC analysis method is as follows:
Chromatographic column: Waters XBridge phenyl (3.5um, 150mmx4.6mm); mobile phase A is aqueous solution containing 0.1% ammonium bicarbonate, mobile phase B is acetonitrile solution, and linear gradient elution is carried out in the following table 2; flow rate: 1 mL/ min; column temperature: 30°C; UV detection wavelength: 214nm, 254nm, 280nm; injection volume 2uL.
Table 2. Gradient elution conditions mobile mobile
Time/min phase A/% phase B/% 0 95 5 5 95 5 95 20.2 95 5 24 95 5 15 The synthetic method of some intermediates in the invention is as follows:
Intermediate 1 38
BL-5786 / LU505465
AcO © c (S) ©
N={ HN—Boc
O 7”
B 7
O N
H
Intermediate 1 was prepared by the following steps: o TBDPSCI © 9 9 imidazol x NaOH TBDPS, SOCI TBDPS,
Ho > 0” ST 16078 oo MeOHMLO 0 on DOM. Ho
INT-1a INT-1b INT-1c INT-1d
TBDPS JBDPS
"CD Ö O, HO,
N Acetic anhydride
INT-te > oO 1) LiBH,, THF Br TBAF Br TEA,DMAP
AIEbCI N 2) diludine, TSOH,DCM = THF = DCM
DCM N 3)LIOH, MeOH H,0 N N
INT-1f INT-19 INT-1h o INTAK 97 ©
S— X Ne S) » d a © IS q oO
Pd(dppf)Cla, ACOK © Boc HN-Boc dioxane o-B Pd(dppf)Cl2, K3PO4 N=
Br A Ÿ dioxane/H,0 J >
N N N
INT-i INT-1j INT-1I
Q Q
A d A d
Q LEE Pinacolborane Q LEE
NIS S-Phos, Pd,dbas, ACOK
TOME N= HN-Boc TT n= HN-Boc = o SS — x ad
N O N
H H
INT-1m INT-1
Step 1: 2.2-dimethyl-3-hydroxypropionate methyl ester INT-1a (100 g, 756.7 mmol) was dissolved in 1L N,N-dimethylformamide, imidazole (128.8 g, 1.89 mol) was added, stirred to dissolve, and tert-butyldiphenylchlorosilane (228.8 g, 832.3 mmol) was added dropwise at room temperature. After the addition was complete, stirring was continued for 4 hours. After the reaction was complete, the reaction solution was poured into 3L of ice water, the suspension was extracted with ethyl acetate (1L*2), the organic phase was washed with water three times, and then concentrated under reduced pressure to obtain INT-1b as a colorless oil. It was directly used in the next 39
BL-5786 step without purification. ESI-MS (m/z): 371.2 [M+H]". LUS05465
Step 2: Add the raffinate INT-1b obtained in the previous step into 2L of methanol, add 360 g of 33% aqueous sodium hydroxide solution, and stir at room temperature for 17 hours. After the reaction was complete, add 1L of water, remove methanol under reduced pressure, extract the residue with petroleum ether (1L*5), adjust the pH value of the aqueous phase to 4~5 with hydrochloric acid after extraction, continue to stir for 30 minutes, filter with suction, and dry to obtain white solid INT-1¢ (269 g, yield 90%).
ESI-MS (m/z): 357.8 [M+H]".
Step 3: INT-1c (130 g, 364.6 mmol) was dissolved in 500 mL of dichloromethane, thionyl chloride (130.1 g, 1.09 mol, 79.4 mL) was added at room temperature, stirred at 60 °C for 3 hours. After the reaction was complete, dichloromethane and remaining thionyl chloride were removed under reduced pressure to obtain INT-1d as a light yellow oil, without purification, 200 mL of dichloromethane was added for use.
Step 4: INT-1e (64.8 g, 331 mmol) was dissolved in 400 mL of dichloromethane, 198 mL of diethylaluminum chloride solution (2M in hexanes) was added dropwise at 0 °C. During the dropping process, the temperature was controlled to be no more than 5°C. Stir the solution for 30 minutes after the dropwise addition, and add the obtained
INT-1d dichloromethane solution dropwise into the reaction flask. During the dropping process the temperature was controlled to be no more than 10 °C. After the dropping was complete, continue the stirring for 2 hours. After the reaction was complete, the reaction solution was poured into 1L of ice water, stirred for 30 minutes, concentrated under reduced pressure to remove dichloromethane. The residue was extracted with ethyl acetate (1L*2), washed with water, and the organic phase was spin evaporated to obtain a brown oil. The oil was added to 2L of petroleum ether/ethyl acetate=10/1 mixed solution, the solid was precipitated by stirring, and filtered by suction to obtain INT-1f as a yellow solid (139 g, yield 78%). ESI-MS (m/z): 534.8 [M+H]".
Step 5: INT-1f (100 g, 187.1 mmol) was dissolved in 500 mL tetrahydrofuran, lithium borohydride (12.2 g, 561.2 mmol) was added, and stirred overnight at 60 °C.
After the raw materials disappeared, the reaction solution was added to 200 mL of ice water to quench, extracted with ethyl acetate (500 mL*3), and the organic phase was
BL-5786 washed with water, dried, concentrated under reduced pressure, and the residue was LU505465 dissolved in 500 mL of dichloromethane, then 2,6-dimethyl- Diethyl 1,4-dihydro-3,5- pyridinedicarboxylate (28.4 g, 112.2 mmol) and p-toluenesulfonic acid (21.4 g, 112.2 mmol) were added thereto, and the reaction solution was stirred at room temperature for 3 hours. After the reaction was complete, the reaction solution was concentrated under reduced pressure to remove methylene chloride, and the residue was dissolved in 500 mL of methanol, 14% lithium hydroxide aqueous solution (100 mL) prepared in advance was added, and the solution was stirred at room temperature for 3 hours, and filtered with suction to obtain a yellow solid INT-1g (84 g, collected rate of 86.3%).
ESI-MS (m/z): 520.2 [M+H]".
Step 6: INT-1g (50 g, 96 mmol) was dissolved in 250 mL tetrahydrofuran, tetrabutylammonium fluoride (1M inTHF, 197mL) was added, stirred overnight at 60°C. After the reaction was complete, the reaction solution was added to 300 mL of water, extracted with ethyl acetate (200 mL*3), washed with water, and concentrated under reduced pressure to obtain a brown oil. Dissolve the obtained residue in 40 mL of methanol, add 20 mL of water, wash the mixed solution with petroleum ether (40 mL*5), concentrate under reduced pressure to remove methanol, and extract the residue with ethyl acetate (50 mL*2). The organic phase was washed with water and dried to obtain INT-1h (25 g, yield 90.4%) as a pale yellow oil. ESI-MS (m/z): 282.8 [M+H]".
Step 7: Dissolve compound INT-1h (22 g 77 mmol) in 100 mL of dichloromethane. Add 4-dimethylaminopyridine (467 mg, 3.82 mmol), triethylamine (23.2 g, 230 mmol), and add acetic anhydride (7.9 g, 77 mmol) dropwise at 0 °C. After the dropwise addition, the temperature was raised naturally, and the solution was stirred overnight. After the reaction was complete, the reaction solution was washed with water, dried, and concentrated to obtain a brown oil, which was purified by silica gel column chromatography (petroleum ether/ethyl acetate=4/1) to obtain a light yellow oil
INT-1i (22.5 g, Yield 90.7%). ESI-MS (m/z): 324.2 [M+H]".
Step 8: Compound INT-1i (40 g, 123.4 mmol) was dissolved in dioxane (400 mL), potassium acetate (30.3 g, 308.4 mmol), [1,1'-bis(diphenyl
Phosphine)ferrocene]palladium dichloride (10 g, 12.3 mmol), pinacol diborate (78.3 g, 308.4 mmol) were added to react at 90 °C for 3 hours under nitrogen protection, LCMS 41
BL-5786 monitored the complete reaction of the raw materials, and the reaction solution was LUS05465 directly concentrated under reduced pressure. The residue was dissolved in ethyl acetate (300 mL), washed with water and brine, and the organic phase was purified by silica gel column chromatography to obtain white solid compound INT-1j (35 g, yield 76.4%). ESI-MS (m/z): 372.5 [M+H]".
Step 9: Dissolve compound INT-1j (35 g, 94.3 mmol) and compound INT-1k (37.9 g, 103.7 mmol) in dioxane (300 mL) and water (30 mL), add potassium phosphate ( 50 g, 235.7 mmol) and [1,l'-bis(diphenylphosphino)ferrocene]palladium dichloride (6.89 10g, 9.43 mmol) to react overnight at 90 °C under nitrogen protection. LCMS monitored the complete reaction of the raw material, the reaction solution was directly concentrated under reduced pressure, the residue was dissolved in ethyl acetate (300 mL), washed with water and brine, and the organic phase was purified by silica gel column chromatography to obtain yellow oily compound INT-11 (28 g, yield 56.08%).
ESI-MS (m/z): 530.7 [M+H]".
Step 10: Dissolve compound INT-11 (28 g 529 mmol) in N,N- dimethylformamide (280 mL), add N-iodosuccinimide (11.9 g, 52.9 mmol) to react at 50°C for 2 hours. LCMS monitored the complete reaction of the raw materials, and the reaction solution was poured into water (800 mL), extracted with ethyl acetate (200 mL*2). The organic phase was washed with saturated brine, dried, filtered, and purified through a silica gel column chromatography to give a yellow solid compound INT-1m (22 g, yield 63.5%). ESI-MS (m/z): 656.6 [M+H]*.
Step 11: Compound INT-1m (5 g, 7.63 mmol), 2-dicyclohexylphosphine-2',6"- dimethoxy-biphenyl (939.4 mg, 2.29 mmol), tris(dimethoxy benzylacetone) dipalladium (838.1 mg, 0.915 mmol), potassium acetate (2.6 g, 26.7 mmol) were dissolved in toluene (100 mL), and pinacol borane (4.9 g, 38.1 mmol) was added under nitrogen protection, after the addition was complete, the reaction was carried out at °C for 5 hours under the protection of nitrogen. LCMS monitored the complete reaction of the raw materials. The reaction solution was filtered and purified by silica gel column chromatography to obtain a yellow oily compound INT-1 (4.5 g, yield 90%). ESI-MS (m/z): 656.5 [M+H]". 42
BL-5786
Intermediate 2 LUS05465
O
“ods )
HN—N 6 s/ ©
N= HN—Boc
O ~°
BB
O N
H
Intermediate 2 was prepared by the following steps: 0 INT-2b Ale a 5 O 8 © © Sor) HO 6 un o
VAS LIOH'H,0 n={ HN-Boc CO, HNONATFA A LIOH'H,0
N= > oc THF/H2O Ss TCFH, NMI N= > HN-Boc THF/H20 = — ACN } =
N
INT-1m INT-2a INT-2c
HO HO HN-N Oo / HNN O Pinacolborane / HNON oO
A orale o A S-Phos, Pd2dba3, AcOK o A n= HN-Boc Tou 0 N={ HN-Boc THF n= HN-Boc
SS SS SS
N H SH
INT-2d INT-2e INT-2
Step 1: Compound INT-1m (12 g, 18.3 mmol) was dissolved in tetrahydrofuran (120 mL) and water (20 mL), lithium hydroxide monohydrate (3.84 g, 91.5 mmol) was added to react overnight at room temperature, LCMS monitored the complete reaction of raw materials, the reaction solution was directly concentrated under reduced pressure, the residue was dissolved in water (100 mL), the pH was adjusted to 4~5 with 4M hydrochloric acid, the solution was extracted with dichloromethane (100 mL*3), the organic phase was washed with water, and washed with salt water, dried over anhydrous sodium sulfate, filtered, and concentrated to obtain white solid compound
INT-2a (10.6 g, yield 96.6%). ESI-MS (m/z): 600.7 [M+H]".
Step 2: Compound INT-2a (9.5 g, 15.9 mmol) and compound INT-2b (11.7 g, 31.7 mmol) were dissolved in acetonitrile (190 mL), NNNN '-
Tetramethylchloroformamidine hexafluorophosphate (6.67 g, 23.8 mmol) and 1- methylimidazole (6.51 g, 79.2 mmol) were added to react at 0 °C for 1 hour, and the 43
BL-5786 reaction of the raw materials was monitored by LCMS to be complete. The reaction LUS05465 solution was poured into water (200 mL), extracted with dichloromethane (100 mL*3), the organic phase was washed with water, and purified by silica gel column chromatography to obtain compound INT-2¢ (9.6 g, yield 83.5%) as a yellow solid.
ESI-MS (m/z): 726.3 [M+H]".
Step 3: Dissolve compound INT-2¢ (9.6 g, 13.2 mmol) in tetrahydrofuran (100 mL) and water (10 mL), add lithium hydroxide monohydrate (1.39 g, 33.1 mmol), and react at room temperature for 4 hours. LCMS monitored the complete reaction of the raw materials. The reaction solution was directly concentrated under reduced pressure, the residue was dissolved in water (100 mL), the pH was adjusted to 4~5 with 4M hydrochloric acid, and a white solid precipitated. After being filtered, washed with water, and dried a white solid compound INT- 2d (8.3 g, 88.2% yield) was obtained.
ESI-MS (m/z): 712.6 [M+H]".
Step 4: Dissolve compound INT-2d (3.5 g, 4.9 mmol), 1-hydroxybenzotriazole (1.99 g, 14.8 mmol), 4-dimethylaminopyridine (1.8 g, 14. mmol) in dichloromethane (170 mL), N,N-diisopropylethylamine (6 mL, 34.4 mmol) was added at 0°C, then 1-(3- dimethylaminopropyl)-3-ethylcarbodieneamine hydrochloride (4.71 g, 24.6 mmol) was added to react overnight at room temperature. LCMS monitored the complete reaction of the raw materials. The reaction solution was washed with saturated ammonium chloride aqueous solution, dried over sodium sulfate, and purified by silica gel column chromatography to obtain a yellow solid compound INT-2e ( 2 g, yield 58.6%). ESI-
MS (m/z): 694.6 [M+H]".
Step 5: Compound INT-2e (500 mg, 0.721 mmol), 2-dicyclohexylphosphine- 2',6"-dimethyl-biphenyl (88.8 mg, 0.216 mmol), tris(dibenzylidene acetone) dipalladium (79 mg, 0.086 mmol), potassium acetate (247 mg, 2.52 mmol) were dissolved in tetrahydrofuran (20 mL), and pinacolborane (461 mg, 3.6 mmol) was added under nitrogen protection. After the addition was complete, the reaction solution was allowed to react at 50 °C for 3 hours under the protection of nitrogen.
LCMS monitored the complete reaction of the raw materials. The reaction solution was filtered and purified by silica gel column chromatography to obtain a yellow solid compound INT-2 (400 mg, yield 80%). ESI-MS (m/z): 694.6 [M+H]". 44
BL-5786
LU505465
Intermediate 3
O
11: (S) >
HN—N © (S) 9
S “Cs xe zz
BH
OÖ N
H
Intermediate 3 was prepared by the following steps:
INT-3b
HN-N HN-N Ho HN-N
O Oo NS O © LE TFA o as > © LÀ, D
N= TNTP0 pou N= Ne TOFH,NMI N= i 7 = 4 = ACN 4 >
INT-2e INT-3a INT-3c
HN=N
Pinacolborane A
S-Phos, Pd2dba3, AcOK © 0
EA NX HNL
X SS
5 Z
ON
H
INT3
Step 1: Dissolve compound INT-2e (1.7 g, 2.45 mmol) in dichloromethane (20 mL), add trifluoroacetic acid (5 mL), and react at room temperature for 2 hours. LCMS monitored that the reaction of the raw materials was complete, and the reaction solution was directly concentrated under reduced pressure, the residue was dissolved in DCM (50 mL), washed twice with saturated NaHCO3 aqueous solution, the organic phase was washed with water, dried over sodium sulfate, filtered, and concentrated to obtain compound INT-3a (1.3 g, yield 89.4%) as a yellow solid. ESI-MS (m/z): 594.7 [M+H]".
Step 2: Compound INT-3a (1.3 g, 2.19 mmol) and compound INT-3b (0.24 g, 2.41 mmol) were dissolved in acetonitrile (30 mL), NN,N'N "- tetramethylchloroformamidine hexafluorophosphate (921.9 mg, 3.29 mmol), and 1-
BL-5786 methylimidazole (414 mg, 5.04 mmol) were added to react at 0 °C for 1 hour, LCMS LUS05465 monitored the complete reaction of the raw materials, and the reaction solution was poured into water (50 mL), extracted with dichloromethane (50 mL*3). Wash the organic phase with water, mix the sample and pass through the column for purification to obtain white solid compound INT-3¢ (1.3 g, yield 87.9%). ESI-MS (m/z): 675.7 [M+H].
Step 1: Compound INT-3¢ (1.1 g, 1.63 mmol), 2-dicyclohexylphosphine-2',6'- dimethyl-biphenyl (200.5 mg, 0.188 mmol), tris(dibenzylidene acetone) dipalladium (179 mg, 0.195 mmol), potassium acetate (559 mg, 5.7 mmol) were dissolved in toluene (30 mL), and pinacol borane (1.04 g, 8.14 mmol) was added under nitrogen protection. The solution was allowed to react at 50 °C for 3 hours under nitrogen protection after the addition was complete. LCMS monitored the complete reaction of the raw materials, the reaction solution was filtered, and purified by silica gel column chromatography to obtain a yellow solid compound INT-3 (990 mg, yield 90%). ESI-
MS (m/z): 676.9 [M+H]".
Intermediate 4
O nes)
HN—N
S (S) 9 ° 5S es
B 7 (R)
O N
H
Intermediate 4 was prepared by the following steps:
HN-N “ HN-N HN=N as J LE i AL
Oo oO oO Pinacolborane O Oo
N= Nz opm, mi NA, HN, S-Phos, Pdadbas, NA HN, 2 =~ ACN 7 =~ È AcOK, THF ny } x Va
N N od N
INT-3a INT-4b INT4
Step 1: Compound INT-3a (2.2 g, 3.71 mmol) and compound INT-4a (0.47 g, 4.08 mmol) was dissolvd in dichloromethane (50 mL), N,N,N' at 0 °C , N'- 46
BL-5786 tetramethylchloroformamidine hexafluorophosphate (1.56 g, 5.56 mmol), and 1- LUS05465 methylimidazole (0.70 g, 8.53 mmol) were added to react at 0 °C for 1 hour. LCMS monitored the complete reaction of the raw materials, and the reaction solution was poured into water (50 mL), extracted with dichloromethane (50 mL*3), the organic phase was washed with water, and the sample was mixed and purified by column to obtain white solid compound INT-4b (2.3 g, yield 90.0%). ESI-MS (m/z): 690.2 [M+H].
Step 2: Compound INT-4b (2.1 g, 3.05 mmol), 2-dicyclohexylphosphine-2',6'- dimethyl-biphenyl (375.0 mg, 0.91 mmol), tris(dibenzylidene acetone) dipalladium (334.6 mg, 0.365 mmol), potassium acetate (1.05 g, 10.7 mmol) were dissolved in toluene (30 mL), and pinacolborane (1.95 g, 15.2 mmol) was added under nitrogen protection. After the addition was complete, the reaction system was allowed react at 50 °C for 3 hours under the protection of nitrogen. LCMS monitored the complete reaction of the raw materials. The reaction solution was filtered and purified by silica gel column chromatography to obtain a yellow solid compound INT-4 (1.8 g, yield 85.7%). ESI-MS (m/z): 690.3 [M+H]".
Intermediate 5 —O
N
N, (SP x
Cbz
Intermediate 5 was prepared by the following steps: >
Nx (8) ©
HN. © | Br
S s ; 27 A INT-5d NT
Cbz-CI, DIPEA ( ) Ammonium carbamate Ss I Br lL ® THF N (Diacetoxyiodo)benzene, C J Cul, K3PO4,
H te MeOH, RT, 16h N N, N-Dimethylethylenediamine, Nsg20 ‘ Cbz DMF,80 °C C J
INT-5a INT-5b INT-5¢ N
Cbz
INT-5
Step 1: Compound INT-Sa (2 g, 19.42 mmol) and benzyl chloroformate (3.3 g, 47
BL-5786 19.42 mmol) were dissolved in tetrahydrofuran (20 mL), at room temperature N,N- LUS05465 diisopropylethylamine (5.0 g, 38.76 mmol) was added, and the reaction was allowed to react for 16 hours at room temperature. LCMS monitored the complete reaction of raw materials. The reaction solution was diluted with ethyl acetate (60 mL), washed three times with saturated aqueous ammonium chloride solution, and the organic phase was dried with anhydrous sodium sulfate and spin-dried to obtain a yellow oily compound
INT-5b (3.9 g, yield 84.7%). ESI-MS (m/z): 238.2 [M+H]".
Step 2: Compound INT- Sb (3.9 g, 16.45 mmol) was dissolved in methanol (30 mL), ammonium carbamate (1.9 g, 24.35 mmol) and iodobenzene acetate (10.6 g, 32.91 mmol) were added, and the reaction solution was stirred at room temperature for 16 h.
After the reaction was complete, the reaction solution was concentrated. The residue was purified by silica gel column chromatography (dichloromethane/methanol=30/1) to obtain compound INT-5¢ (3.5 g, yield 79.5%). ESI-MS (m/z): 269.3 [M+H]".
Step 3: Compound INT- 5e (600 mg, 2.24 mmol) and compound INT- 5d (761 mg, 2.24 mmol) were dissolved in N,N-dimethylformamide (10 mL), cuprous iodide (85 mg, 0.45 mmol), N,N-dimethylethylenediamine (40 mg, 0.45 mmol) and potassium phosphate (1.42 g, 6.72 mmol) were added. The reaction system was replaced with nitrogen and heated to 80 °C and stirred for 16 hours. After the reaction was complete, the reaction system was added with water (80 mL), extracted with ethyl acetate (80 mL*2), the organic phases were combined and washed with saturated brine, dried over anhydrous sodium sulfate, filtered and the filtrate was concentrated. the residue was purified by silica gel column chromatography (dichloromethane/methanol=20/1) to obtain compound INT-S (615 mg, yield 57.1%). ESI-MS (m/z): 482.6 [M+H]".
Intermediate 6
J
(S)
No
AN
48
BL-5786
LU505465
Intermediate 6 was prepared by the following steps: / 0
J (©) ©) Cul, KPO, A Br
N \ + HN, zo N,N-dimethylethylenediamine — ya À DMF, 85°C,16 h No
AS
INT-5d INT-6a \ INT
Step 1: Compound INT- 5d (500 mg, 1.46 mmol) and compound INT- 6a (150 mg, 1.61 mmol) was dissolved in N,N-dimethylformamide (5 mL), cuprous iodide (56 mg, 0.29 mmol), N,N-dimethylethylenediamine (26 mg, 0.29 mmol) and potassium phosphate (931 mg, 4.4 mmol) were added. The reaction system was replaced with nitrogen and heated to 85 °C and stirred for 16 hours. After the reaction was complete, water (50 mL) was added, and the reaction solution was extracted with ethyl acetate (50 mL*2), the organic phases were combined and washed with saturated brine, dried over anhydrous sodium sulfate, filtered and the filtrate was concentrated. the residue was purified by silica gel column chromatography (dichloromethane/methanol=20/1) to obtain compound INT-6 (360 mg, yield 80.2%). ESI-MS (m/z): 307.4 [M+H]".
Intermediate 7
J
(S)
N
/ \ Br © _ ar ( 6)
Intermediate 7 was prepared by the following steps: 49
BL-5786 ~ + LU505465
NS pr
HN» s Ou LL INT-5d - \ > , DCE, RT, 4h (J MeOH, RT, 16h CO N, N-Dimelhyieihyienediamine,
INT-7b INT-7c (J
INT-7
Step 1: Compound INT- 7a (500 mg, 5.67 mmol) and morpholine (989 mg, 11.35 mmol) were dissolved in 1,2-dichloroethane (5 mL), acetic acid (34 mg, 0.57 mmol) was added, and the reaction solution was stirred at room temperature for 1 h. Then triacetyl sodium oxyborohydride (2.4 g, 11.35 mmol) was added, and the stirring continued for 3 h. After the reaction was complete, the reaction solution was added with saturated sodium bicarbonate (30 mL), extracted with ethyl acetate (30 mL*2), and the the organic phases were combined and washed with saturated brine, dried over anhydrous sodium sulfate, filtered and the filtrate was concentrated. The residue was purified by silica gel column chromatography (dichloromethane/methanol=40/1) to obtain compound INT-7b (650 mg, yield 71.9%). ESI-MS (m/z): 159.9 [M+H]".
Step 2: Compound INT- 7b (600 mg, 3.77 mmol) was dissolved in methanol (10 mL), ammonium carbamate (441 mg, 5.65 mmol) and iodobenzene acetate (2.43 g, 7.54 mmol) were added, and the reaction solution was stirred at room temperature for 16 h.
After the reaction was complete, the reaction solution was concentrated. the residue was purified by silica gel column chromatography (dichloromethane/methanol=20/1) to obtain compound INT-7¢ (670 mg, yield 93.5%). ESI-MS (m/z): 191.3 [M+H]*. 'H
NMR (500 MHz, DMSO-d6) & 4.65 (s, 1H), 4.05 — 3.94 (m, 2H), 3.91 — 3.82 (m, 2H), 3.60 —3.55 (m, 4H), 3.14 — 3.06 (m, 1H), 2.35 — 2.29 (m, 4H).
Step 3: Compound INT- 7¢ (55.6 mg, 0.29 mmol) and compound INT- 5d (100 mg, 0.29 mmol) were dissolved in N,N-dimethylformamide (5 mL), cuprous iodide (11 mg, 0.058 mmol), N,N-dimethylethylenediamine (5 mg, 0.058 mmol) and potassium phosphate (186 mg, 0.87 mmol) were added. The reaction system was replaced with
BL-5786
LU505465 nitrogen and heated to 80 °C and stirred for 16 hours. After the reaction was complete, water (50 mL) was added, and the reaction solution was extracted with ethyl acetate (50 mL*2), the organic phases were combined and washed with saturated brine, dried over anhydrous sodium sulfate, filtered and the filtrate was concentrated. the residue was purified by silica gel column chromatography (dichloromethane/methanol=20/1) to obtain compound INT-7 (65 mg, yield 55%). ESI-MS (m/z): 404.6 [M+H]".
Intermediate 8 —0
N
N,
IS) 1
Bod
Intermediate 8 was prepared by the following steps: >
Na À ES s s HN, ‚9 | INT5d NSE (Boc),0, DIPEA C J Ammonium carbamate Ss | Br | > ® DCM N (Diacetoxyiodo)benzene, C J Cul, K3PO4,
H 5 MeOH, RT, 16h N N, N-Dimethylethylenediamine, Nsg20 0e Boc DMF,80 °C C J
INT-5a INT-8b INT-8¢ N
Boc
INT-8
Use di-tert-butyl dicarbonate to replace benzyl chloroformate in Step 1 of the synthesis step of Intermediate INT-5, and use similar methods and reaction steps to obtain compound INT-8. ESI-MS (m/z): 448.6 [M+H]".
Intermediate 9 / o (S)
N
/ N Br
LI cbz N
Intermediate 9 was prepared by the following steps: 51
BL-5786 - | LU505465 0 u, 0
NL © ©
O, NH joa ot
Ss S Ss A INT-5d
Cbz-Cl, DIPEA Ammonium carbamate | Br A x THF x (Diacetoxyiodo)benzene, x Cul, KsPO4, NO
N N MeOH, RT, 16h N N, N-Dimethylethylenediamine, SC
H Cbz Cbz DMF,80 °C
INT-92 INT-9b INT-9¢
N
Cbz
INT-9
Use INT-9a to replace INT-Sa in Intermediate INT-5, and use similar methods and reaction steps to obtain compound INT-9. ESI-MS (m/z): 494.4 [M+H]".
Intermediate 10 /
Og) 7 er 9 —
CC
6)
Intermediate 10 was prepared by the following steps: ~o >
Oo NH N. (9 vo) Br ° ARN S = | INT-5d N 2 ® \ / Ammonium carbamate OO I Br \
AcOH, NaBH(OAc)s, Diacetoxyiodo)b ,Ç oN oman ™ oh RT an A Psd Xartphos, 5
INT-10a Oo (J ©
INT-10b INT-10c N
J
INT-10
Step 1: Compound INT-10a (1.3 g, 11.19 mmol) and morpholine (974.8 mg, 11.19 mmol) was dissolved in dichloromethane (10 mL), acetic acid (67 mg, 1.12 mmol) was added, and the reaction solution was stirred at room temperature for 1 h. Then triacetyl sodium oxyborohydride (4.74 g, 22.38 mmol) was added, and the stirring continued for 3 h. After the reaction was complete, the reaction system was added with saturated sodium bicarbonate (30 mL), extracted with ethyl acetate (50 mL*2), and the organic phases were combined and washed with saturated brine, dried over anhydrous sodium sulfate, filtered and the filtrate was concentrated. The residue was purified by silica gel column chromatography (dichloromethane/methanol= 40/1) to obtain compound INT- 52
BL-5786 10b (1.8 g, yield 85.9%). ESI-MS (m/z): 188.6 [M+H]". 17005685
Step 2: Compound INT-10b (1.8 g, 9.61 mmol) was dissolved in methanol (15 mL), ammonium carbamate (1.88 g, 24.03 mmol) and iodobenzene acetate (7.74 g, 24.03 mmol) were added, and the reaction solution was stirred at room temperature for 16 h. After the reaction was complete, the reaction solution was concentrated. the residue was purified by silica gel column chromatography (dichloromethane/methanol=20/1) to obtain compound INT-10c (1.5 g, yield 71.5%).
ESI-MS (m/z): 219.4 [M+H]".
Step 3: Compound INT-10¢ (127.7 mg, 0.58 mmol) and compound INT-Sd (200 mg, 058 mmol) were dissolved in 1, 4-dioxane (5 mL), tris(dibenzylideneacetone)dipalladium (53.6 mg, 0.058 mmol), xantphos (33.8 mg, 0.058 mmol) and cesium carbonate (381.1 mg, 1.17 mmol) were added. The reaction system was replaced with nitrogen and heated to 80 oC and stirred for 16 hours. After the reaction was complete, water (50 mL) was added, and the reaction solution was extracted with ethyl acetate (50 mL*2), the organic phases were combined and washed with saturated brine, dried over anhydrous sodium sulfate, filtered and the filtrate was concentrated. the residue was purified by silica gel column chromatography (dichloromethane/methanol=20/1) to obtain compound INT-10 (180 mg, yield 71.2%).
ESI-MS (m/z): 432.6 [M+H]+.
Intermediate 11
Jd (S) a 0 =
S=N
O pe
INT-11
Intermediate 11 was prepared by the following steps: 53
BL-5786 \ LU505465 > O s NH NS EB ° /\ os oe | No > NP = Ammonium carbamate I Z er INT-5d 4
AcOH, NaBH(OAC)3, (Diacetoxyiodo)benzene, Pdz(dba)s, Xantphos oN s DCM, RT © MeOH, RT, 16h a C82COs, dioxane Ss
INT-11a © , y
INT-11b - N
INT-11c IS
INT-11
Use INT-11a to replace INT-10a in Intermediate INT-10, and use similar methods and reaction steps to obtain compound INT-11. ESI-MS (m/z): 446.4 [M+H]".
The synthetic method of embodiment compound among the present invention is as follows:
Example 1 (1S,2S)-N-((63R,4S,Z))-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-((4-methyl-1- oxido-116-thiomorpholin-1-ylidene)amino)pyridin-3-yl)-10,10-dimethyl-5,7- dioxo-61,62,63,64,65,66-hexahydro-11 H-8-oxa-2(4,2)-thiazola-1(5,3)-indola- 6(1,3)-pyridazinacycloundecaphane-4-yl)-2-methylcyclopropane-1-carboxamide
O
N (9 )
HN—N © (S) >. 0 NA HN
S Xe
N > rN — N
N° — (CP 7 /
Example 1 was prepared by the following steps: 54
BL-5786 ~0 LU505465 a A 7 X
So Br Q
N Mods
Q fo) 820 / HN—N 7 HN=N {5 © ©) On
A, 9 pod INT-8 7 N= NE I n= HN ©. PdEppACly KPO4 NA À 7 = Cs2CO3, o aS p= dioxane, H,O, 70 °C _ N DMF, rt x BL N H dN S=0
H
INT-3 €) 1a
Bod
Q, if ) Da >
HN—N HNN o e/ 79 5 © e/ 70 5 Loto = N HN = © N= AN NaBH(OAc) ©) = (5) S) “Ss a C)3, a, SS = = mes
No NN — 5=0 8-0 ¢ 2 1b ©) 1c
Bod
Df > 8 >
HN—N HNN
O Da 5 0 ©) 90 —Q HN—4 + = A HN (s) N= > “9 ©) = > “Sg,
Ad > >= CC / >= — — N
N _ ND — ‘S=0 8-0 ¢ 2 1 ( DD) 1 / /
Step 1: Intermediate INT- 3 (60 mg, 13.4 umol) and Intermediate INT- 8 (99 mg, 14.7 umol) were dissolved in a mixed solvent of 1,4-dioxane (4 mL) and water (0.4 mL), [1,1’-bis(diphenylphosphino)ferrocene]palladium dichloride (19 mg, 0.26 mmol) and potassium phosphate (57 mg, 0.54 mmol) were added. The reaction system was replaced with nitrogen and heated to 70 °C to stir for 12 h. LCMS monitored that the reaction was complete. The solvent was evaporated under reduced pressure, diluted with ethyl acetate, and the mixture was filtered through diatomaceous earth, and the filtrate was concentrated and purified by preparative thin-layer chromatography (dichloromethane/methanol = 20/1) to obtain to obtain compound la (88 mg, yield 71%). ESI-MS (m/z): 917.5 [M+H]".
BL-5786
Step 2: Compound 1a (88 mg, 96 umol) was dissolved in DMF (3 mL), thereto HUS05465 was added with cesium carbonate (94 mg, 288 umol), and then ethyl iodide (75 mg, 0.48 mmol) was added dropwise to the reaction solution. The reaction solution was at room temperature stirred for 16 hours. LCMS detected that the reaction was complete.
The reaction solution was added with saturated brine, extracted with ethyl acetate, the organic phases were combined and washed with saturated brine, dried over anhydrous sodium sulfate, and the organic phase was concentrated to obtain a crude product 1b.
ESI-MS (m/z): 945.5 [M+H]".
Step 3: Trifluoroacetic acid (1 mL) was added dropwise to the crude product 1b in dichloromethane (3 mL). The reaction solution was stirred at room temperature for 30 minutes. LCMS monitored that the reaction was complete. The reaction solution was concentrated by distillation under reduced pressure, saturated aqueous sodium carbonate solution was added thereto, and extracted with ethyl acetate, the organic phases were combined and washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to obtain a crude product 1¢. ESI-MS (m/z): 845.5 [M +H]".
Step 4: Aqueous solution of formaldehyde (0.05 mL, 37%) was added dropwise to compound 1c (67 mg, 79 umol) in 1,2-dichloroethane (2 mL). The reaction solution was stirred at room temperature for 20 minutes, and then thereto was added triacetyl sodium oxyborohydride (101 mg, 476 umol). The reaction solution was continued to stir at room temperature for 30 minutes. LCMS monitored that the reaction was complete. Saturated ammonium chloride aqueous solution was added to quench the reaction, the reaction solution was extracted with ethyl acetate, the organic phases were combined and washed with saturated brine, dried over anhydrous sodium sulfate, concentrated and purified by preparative thin layer chromatography (dichloromethane/methanol = 20/1) to obtain a white solid compound 1 (5 mg, yield 7%) and epimer 1° (9 mg, yield 13%). The absolute configurations drawn by the two compounds are based on empirical assumptions, compound 1 is an isomer with relatively long retention time in LC-MS and HPLC, and 1° is an isomer with relatively short retention time in LC-MS and HPLC. 56
BL-5786
Compound 1: LUS05465
ESI-MS (m/z): 859.6 [M+H]'; LC-MS retention time RT=1.60 min. HPLC retention time RT= 10.04 min. '"H NMR (500 MHz, DMSO-d6) 6 8.49-8.38 (m, 2H), 8.31 (d, J = 2.6 Hz, 1H), 7.72 (s, 1H), 7.69-7.63 (m, 1H), 7.49 (d, J= 8.7 Hz, 1H), 7.17 (d, /= 2.6 Hz, 1H), 5.50 (t, J=9.1 Hz, 1H), 5.03-4.98 (m, 1H), 4.28-4.06 (m, SH), 3.55-3.47 (m, 3H), 3.16 (s, 3H), 3.11-3.05 (m, 1H), 2.93-2.63 (m, 7H), 2.37-2.32 (m, 1H), 2.23 (s, 3H), 2.01 (d, J = 9.6 Hz, 1H), 1.76-1.64 (m, 2H), 1.49-1.40 (m, 2H), 1.26 (d, J = 6.0 Hz, 3H), 1.03- 0.95 (m, 4H), 0.86-0.77 (m, 7H), 0.51-0.44 (m, 1H), 0.28 (s, 3H).
Compound 1’:
ESI-MS (m/z): 859.5 [M+H]". LC-MS retention time RT=1.59 min. HPLC retention time RT= 10.03 min. '"H NMR (500 MHz, DMSO-ds) 6 8.56-8.49 (m, 2H), 8.38 (d, J = 2.5 Hz, 1H), 7.80 (s, 1H), 7.72 (dd, J = 8.7, 1.7 Hz, 1H), 7.51 (d, J = 8.7 Hz, 1H), 7.38 (d, J = 2.6
Hz, 1H), 5.54 (t, J = 9.2 Hz, 1H), 5.08-5.02 (m, 1H), 4.22 (t, J = 12.2 Hz, 2H), 3.93- 3.88 (m, 1H), 3.66 (d, J= 10.9 Hz, 1H), 3.54 (d, J= 10.9 Hz, 1H), 3.38 (d, J = 4.8 Hz, 1H), 3.18-3.11 (m, 1H), 3.08 (s, 3H), 3.02 (d, J= 14.4 Hz, 1H), 2.93-2.85 (m, 2H), 2.84- 2.71 (m, 3H), 2.45-2.40 (m, 1H), 2.30 (s, 3H), 2.15-2.08 (m, 1H), 1.84-1.77 (m, 2H), 1.55-1.48 (m, 2H), 1.21 (d, J = 6.2 Hz, 3H), 1.14-1.05 (m, 6H), 0.94 (s, 3H), 0.89-0.83 (m, 1H), 0.59-0.53 (m, 1H), 0.51 (s, 3H).
Example 2 (1S,25)-N-((63S,4S,Z))-12-(5-((dimethyl(oxo0)-16-sulfaneylidene)amino)-2-((S)- 1-methoxyethyl)pyridin-3-yl)-11-ethyl-10,10-dimethyl-5,7-dioxo- 61,62,63,64,65,66-hexahydro-11 H-8-oxa-2(4,2)-thiazola-1(5,3)-indola-6(1,3)- pyridazinacycloundecaphane-4-yl)-2-methylcyclopropane-1-carboxamide 57
BL-5786
LU505465
O ©) O
NON 0 / o "© His © va
N= (S) S
N >
AN
— N
No —
Ss”
AN
Example 2 was prepared by the following steps: o” ©) | o ip oO
Oy ON Oo N Apr S N © o "wm —s=0 / © NHBoc
NHBoc | INT 3 N
N= -_— (8) N= ——
S Pd(dppf)Clz, K3PO4, N aS Cs,CO;3, ny J = dioxane, H,0,70°C, 16h U N_¢/ DMF, rt
B — N oN M, *
INT-2 AS 2a 0
À ox lo) Oo a © He “Ss 1 Hos 2 Hos >= d NHBoc TFA d NH, INT-3b © N= DCM, rt ©) N= COMU, DIEA,
RO RON our pi =)
N N,
BP AP
\ 2b \ 2c we 0 9 ou °
O S bi, (9) O (S) L.(S)
Rv J A ve ©) N= + ©) N= >> "Lv NA =
Da 34
N, N,
AP 5° \ 2 \ 2'
Step 1: Compound INT-2 (108.4 mg, 0.16 mmol) was dissolved in a mixed solution of 1,4-dioxane (5 mL) and water (0.5 mL), INT-6 (40 mg, 0.13 mmol), [1,1”- bis(diphenylphosphine)ferrocene]palladium dichloride (9.5 mg, 0.013 mmol) and potassium phosphate (82.9 mg, 0.39 mmol) were successively added. The reaction 58
BL-5786 mixture was stirred at 70 °C for 16 hours under nitrogen protection. After the reaction LUS05465 was complete, the reaction system was added with water (20 mL), extracted with ethyl acetate (30 mL*2), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated, the residue was purified by preparative thin layer chromatography (dichloromethane/methanol=20:1) to obtain
Ra yellow solid compound 2a (55 mg, yield 53.2%). ESI-MS (m/z): 794.5 [M+H]".
Step 2: Compound 2a (55 mg, 0.069 mmol) was dissolved in N,N- dimethylformamide (2 mL), thereto were added cesium carbonate (45.1 mg, 0.138 mmol) and ethyl iodide (16.2 mg, 0.104 mmol). the reaction mixture was stirred at room temperature for 16 hours. After the reaction was complete, the reaction system was added with water (30 mL), extracted with ethyl acetate (30 mL*2), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated, the residue was purified by preparative thin layer chromatography (dichloromethane/methanol=30:1) to obtain a light yellow solid compound 2b (45 mg, yield 79.0%). ESI-MS (m/z): 822.4 [M+H]".
Step 3: Compound 2b (45 mg, 0.055 mmol) was dissolved in dichloromethane (2 mL), thereto were added trifluoroacetic acid (0.5 mL). the reaction mixture was stirred at room temperature for 2 h. After the reaction was complete, under ice bath the reaction system was added with saturated sodium bicarbonate solution (30 mL) , extracted with dichloromethane (30 mL*2), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to obtain a light yellow solid compound 2c (40 mg, yield 100%). ESI-MS (m/z): 722.4 [M+H]".
Step 4: Compound 2¢ (40 mg, 0.055 mmol) was dissolved in N,N- dimethylformamide (2 mL), thereto were added (1S,2S)-2- methylcyclopropanecarboxylic acidINT-3b (111 mg 0.11 mmol), N,N- diisopropylethylamine (21.5 mg, 0.166 mmol) and (2-ethyl 2-oximino-cyanoacetate)-
N,N-dimethyl-morpholinourea hexafluoro phosphate (47.5 mg, 0.11 mmol). the reaction mixture was stirred at ice bath for 1 h. After the reaction was complete, the reaction system was added with water (20 mL), extracted with ethyl acetate (30 mL*2), the organic phases were combined, washed with saturated brine, dried over anhydrous 59
BL-5786 sodium sulfate, filtered and concentrated, the residue was purified by preparative liquid LUS05465 chromatography to obtaina white solid compound 2 (5 mg, yield 11.2%) and epimer 2’ (8 mg, yield 18.0%). The absolute configurations drawn for the two compounds are based on empirical assumptions. Compound 2 is an isomer with relatively long retention time in LC-MS and HPLC, and 2’ is an isomer with relatively short retention time in LC-MS and HPLC.
Compound 2:
ESI-MS (m/z): 804.5 [M+H]'; LC-MS retention time RT=1.56 min. HPLC retention time RT= 11.12 min. 'H NMR (500 MHz, DMSO-d6) § 8.53 — 8.50 (m, 1H), 8.49-8.47 (m, 1H), 8.33 (d, J=2.5 Hz, 1H), 7.80 (s, 1H), 7.75-7.71 (m, 1H), 7.59-7.54 (m, 1H), 7.20 (d, J=2.5
Hz, 1H), 5.60-5.53 (m, 1H), 5.11-5.05 (m, 1H), 4.33-4.17 (m, 5H), 3.61-3.54 (m, 2H), 3.32-3.31 (m, 3H), 3.29 (s, 3H), 3.22 (s, 3H), 3.17-3.12 (m, 1H), 2.98-2.92 (m, 1H), 2.79-2.73 (m, 1H), 2.44-2.40 (m, 1H), 2.10-2.05 (m, 1H), 1.82-1.75 (m, 2H), 1.53-1.48 (m, 2H), 1.33 (d, J= 6.0 Hz, 3H), 1.24-1.19 (m, 1H), 1.14-1.06 (m, 5H), 0.94-0.85 (m, 6H), 0.57-0.52 (m, 1H), 0.39-0.33 (m, 3H).
Compound 2’:
ESI-MS (m/z): 804.5 [M+H]'; LC-MS retention time RT=1.56 min. HPLC retention time RT= 11.09 min. 'H NMR (500 MHz, DMSO-d6) & 8.55-8.51 (m, 2H), 8.34 (d, J= 2.5 Hz, 1H), 7.80 (s, 1H), 7.74-7.70 (m, 1H), 7.53-7.49 (m, 1H), 7.34 (d, J= 2.5 Hz, 1H), 5.54 (t, J = 9.0 Hz, 1H), 5.05 (d, J= 12.0 Hz, 1H), 4.29-4.18 (m, 2H), 3.99-3.91 (m, 1H), 3.88-3.80 (m, 2H), 3.70-3.64 (m, 1H), 3.59-3.51 (m, 1H), 3.31 (s, 3H), 3.28 (s, 3H), 3.18-3.12 (m, 1H), 3.07 (s, 3H), 3.04-3.00 (m, 1H), 2.80-2.74 (m, 1H), 2.43 (d, J= 14.0 Hz, 1H), 2.16-2.11 (m, 1H), 1.85-1.78 (m, 2H), 1.55-1.47 (m, 2H), 1.25-1.17 (m, 4H), 1.13-1.05 (m, 7H), 0.94 (s, 3H), 0.90-0.85 (m, 1H), 0.60-0.48 (m, 4H).
Example 3 (18,2S)-N-((63S,48,Z)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-((6-methyl-2- oxido-216-thia-6-azaspiro[3.3]heptan-2-ylidene)amino)pyridin-3-yl)-10,10- 60
BL-5786
LU505465 dimethyl-5,7-dioxo-61,62,63,64,65,66-hexahydro-11 H-8-oxa-2(4,2)-thiazola- 1(5,3)-indola-6(1,3)-pyridazinacycloundecaphane-4-yl)-2-methylcyclopropane-1- 6) os N26 o He Je
J Ne
N= (S) S
N NS
NI
— N
N X
N carboxamide /
Example 3 was prepared by the following steps: 61
BL-5786
J LU505465 (s)
PN er
N
57° ©) o x Ou" & „NO ya ) NT INT-9 J He
HN-N ! / NHBoc
ESP Pd(dpphClz, KsPOs, N SS CszCOs,
NF > “(js dioxane, H2O,70°C,16h Ÿ X ZZ DMF, rt 0 = > =/ N x BH No " d N >
INT- 3 x 3a
N cb?
GA °9 on oh °9
O (s) (9) O (s) (SD d va { Ne (s) N= s TFA (s) N= > HCHO
N = DCM, N = NaBH(OAc) , NZZ tt #4 NN res + +
N. N, 57° $20 3b 5 3c
Cbz H
GA ° GA ° oO (S) (5) O (S) (5)
J Ne J aval © NA, © N=
N = N =
UN ZZ + NL >; = J
N N
870 870
N N
/ /
Use INT- 9 to replace INT-8 in the systhesis step of compound 1, and use similar methods and reaction steps to obtain compound 3. Compound 3 and its epimer 3’ cannot 5 be separated and purified by reverse preparative liquid chromatography and normal phase column. Both epimers have the same retention time in LC-MS and HPLC.
Compound 3+3°:
ESI-MS (m/z): 871.5 [M+H]'; LC-MS retention time RT=1.58 min. HPLC retention time RT=11.23 min. 62
BL-5786
Example 4 LU505465 (1S,2S)-N-((63S,4S,Z)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-((4-(2- methoxyethyl)-1-oxido-116-thiomorpholin-1-ylidene)amino)pyridin-3-yl)-10,10- dimethyl-5,7-dioxo-61,62,63,64,65,66-hexahydro-11 H-8-oxa-2(4,2)-thiazola- 1(5,3)-indola-6(1,3)-pyridazinacycloundecaphane-4-yl)-2-methylcyclopropane-1-
Oye VNC 6 o He Js
Jd vA (S) N=
S
N = rN — N
N
(SS \ ~~ = carboxamide —O
Example 4 was prepared by the following steps: /
N
/ \ Br oO N a N O meds) > \3=0 © Hs)
HN=N C J / NHBoc oO (y= N=" INTs xe N=
HN-Boe —— N 7 ah
NS Pd(dppf)Clz, K,PO4 rN Cs,CO0;, o À = dioxane, H;0, 70 °C, 16h — N DMF, rt
Ss N No
H ©
INT-2 N 4a
Cbz el Oo ol} Oo pl TH 9 0 (s) 0 (s) 7
J NHBoc J NH, Ho-4 5) - N= TFA - NS > "€ | > DCM, rt A COMU, DIEA, DMF =/ N — N cod 4b cot 4c 63
BL-5786
LU505465
O (s) hs) O (s) bis)
Sey "A Vo Sey = N Vo
Ss Pd(OH)2, Ha Ss AS
N = —_— N \ Yi = _— > a Ÿ THF { N CszCOs, KI,
No J No J DMF, 60°C, 16h 5? 5% @ 4d @ 4e
Cbz H on Loh 9 0 oe 99 o Hel Jus o Me Jus de A H Vo de N= H Vo
N > S N x S
ON ZZ + AN —/ N pe N
LOR LOR
BS N°
Ww, 7 § 4 § 4 oO oO \ \
Step 1: Compound INT-2 (155.9 mg, 0.225 mmol) was dissolved in a mixed solution of 1,4-dioxane (5 mL) and water (0.5 mL), INT-S (108.2 mg, 0.225 mmol), [1,1’-bis(diphenylphosphine)ferrocene]palladium dichloride (16.8 mg, 0.023 mmol) and potassium phosphate (95.4 mg, 0.45 mmol) were successively added. The reaction mixture was stirred at 70 °C for 16 hours under nitrogen protection. After the reaction was complete, the reaction system was added with water (20 mL), extracted with ethyl acetate (30 mL*2), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated, the residue was purified by preparative thin layer chromatography (dichloromethane/methanol=20:1) to obtain a light yellow solid compound 4a (130.5 mg, yield 60.0%). ESI-MS (m/z): 969.5 [M+H].
Step 2: Compound 4a (130.5 mg, 0.135 mmol) was dissolved in N,N- dimethylformamide (2 mL), thereto were added cesium carbonate (88 mg, 0.27 mmol) and ethyl iodide (42.1 mg, 0.27 mmol). The reaction mixture was stirred at room temperature for 16 hours. After the reaction was complete, the reaction system was added with water (30 mL), extracted with ethyl acetate (30 mL*2), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, 64
BL-5786 filtered and concentrated, the residue was purified by preparative thin layer LUS05465 chromatography (dichloromethane/methanol=30:1) to obtain a light yellow solid compound 4b (116.6 mg, yield 86.8%). ESI-MS (m/z): 997.5 [M+H]".
Step 3: Compound 4b (116.6 mg, 0.117 mmol) was dissolved in dichloromethane (2 mL), thereto was added trifluoroacetic acid (0.5 mL). the reaction mixture was stirred at room temperature for 2 h. After the reaction was complete, under ice bath the reaction system was added with saturated sodium bicarbonate solution (30 mL) , extracted with dichloromethane (30 mL*2), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to obtain a light yellow solid compound 4c (105.5 mg, yield 100%). ESI-MS (m/z): 897.5 [M+H]".
Step 4: Compound 4c (1055 mg, 0.117 mmol) was dissolved in N,N- dimethylformamide (3 mL), thereto were added (1S,2S)-2- methylcyclopropanecarboxylic acidINT-3b (176 mg, 0.176 mmol), N,N- diisopropylethylamine (30.2 mg, 0.234 mmol) and (2-ethyl 2-oximino-cyanoacetate)-
N,N-dimethyl-morpholinourea hexafluoro phosphate (75.4 mg, 0.176 mmol). the reaction mixture was stirred at ice bath for 1 h. After the reaction was complete, the reaction system was added with water (20 mL), extracted with ethyl acetate (30 mL*2), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated, the residue was purified by preparative thin layer chromatography to obtain a light yellow solid compound 4d (100 mg, yield 85%).
ESI-MS (m/z): 979.6[M+H]".
Step 5: Compound 4d (100 mg, 0.10 mmol) was dissolved in tetrahydrofuran (6 mL), thereto were added Pd(OH),/C (15 mg). The reaction solution was stirred at room temperature under hydrogen for 16 hours. After the reaction was complete, the reaction solution was filtered, and the filtrate was spin-driedto obtain a yellow oily compound 4e (80 mg, yield 93%). ESI-MS (m/z): 845.3 [M+H].
Step 6: Compound 4e (30 mg, 0.04 mmol) was dissolved in DMF (2 mL), thereto were added 1-iodo-2-methoxyethane (13 mg, 0.07 mmol), cesium carbonate (23 mg, 0.07 mmol) and potassium iodide (1.4 mg, 0.008 mmol). At 60 degree the reaction solution was stirred for 16 hours. After the reaction was complete, the reaction solution 65
BL-5786 was filtered and concentrated, the residue was purified by preparative liquid LUS05465 chromatography to obtaina white solid compound 4 (6 mg, yield 18.7%) and epimer 4° (9 mg, yield 28.1%). The absolute configurations drawn for the two compounds are based on empirical assumptions. Compound 4 is an isomer with relatively long retention time in LC-MS and HPLC, and 4’ is an isomer with relatively short retention time in LC-MS and HPLC.
Compound 4:
ESI-MS (m/z) : 903.5 [M+H]"; LC-MS retention time RT=1.63 min. HPLC retention time RT= 11.69 min. '"H NMR (500 MHz, DMSO-ds) 6 8.54-8.47 (m, 2H), 8.37 (d, J = 2.6 Hz, 1H), 7.79 (s, 1H), 7.73 (dd, J = 8.7, 1.6 Hz, 1H), 7.56 (d, J = 8.7 Hz, 1H), 7.24 (d, J = 2.6
Hz, 1H), 5.56 (t, J= 9.1 Hz, 1H), 5.10-5.05 (m, 1H), 4.35-4.12 (m, 6H), 3.27 (s, 3H), 3.22 (s, 3H), 3.18-3.12 (m, 2H), 3.06-2.92 (m, 6H), 2.82-2.74 (m, 2H),, 2.69 (t, J= 5.5
Hz, 2H), 2.66-2.62 (m, 1H), 2.44-2.35 (m, 2H), 2.11-2.04 (m, 1H), 1.82-1.74 (m, 2H), 1.55-1.46 (m, 2H), 1.33 (d, J = 6.0 Hz, 3H), 1.31-1.25 (m, 2H), 1.24 (s, 3H), 1.06 (s, 3H), 0.91-0.84 (m, 6H), 0.57-0.53 (m, 1H), 0.35 (s, 3H).
Compound 4’:
ESI-MS (m/z) : 903.6 [M+H]". LC-MS retention time RT=1.62 min. HPLC retention time RT= 11.67 min. "H NMR (500 MHz, DMSO-de) 5 8.57-8.49 (m, 2H), 8.38 (d, J = 2.6 Hz, 1H), 7.79 (s, 1H), 7.72 (dd, J = 8.7, 1.7 Hz, 1H), 7.51 (d, J = 8.7 Hz, 1H), 7.37 (d, J=2.6
Hz, 1H), 5.54 (t, J= 9.2 Hz, 1H), 5.07-5.01 (m, 1H), 4.27-4.18 (m, 2H), 3.97-3.79 (m,
SH), 3.23 (s, 3H), 3.07 (s, 3H), 3.05-2.92 (m, SH), 2.81-2.73 (m, 2H), 2.69 (t, J= 5.5
Hz, 3H), 2.42 (d, J= 14.3 Hz, 1H), 2.16-2.09 (m, 1H), 1.83-1.77 (m, 2H), 1.57-1.48 (m, 2H), 1.24 (s, 2H), 1.21 (d, J= 6.2 Hz, 3H), 1.11 (t, J=7.1 Hz, 3H), 1.07 (s, 3H), 0.94 (s, 3H), 0.90-0.85 (m, 2H), 0.56 (d, J= 6.6 Hz, 1H), 0.51 (s, 3H).
Example 5 (1S,2S)-N-((63S,4S,Z)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-((3-morpholino- 1-oxido-116-thietan-1-ylidene)amino)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo- 61,62,63,64,65,66-hexahydro-11 H-8-oxa-2(4,2)-thiazola-1(5,3)-indola-6(1,3)- pyridazinacycloundecaphane-4-yl)-2-methylcyclopropane-1-carboxamide 66
BL-5786 ©) LU505465
Os N__O rs / O (S) A o H Vo (S) N=
S
N - y NO
O _ N
J
CN
6)
Example 5 was prepared by the following steps: /
Os)
N ©) ° 220 Oe ) NO 1 {(S) > Wo / oO H ©) NHBoc
HN-N N du dS ©) ° C J) INT-7 N Ss A nl HN-50e ——: N\ / =>
Ss Pd(dppf)Cl2, K3PO4, — N Cs,CO3, % J dioxane, H,0, 70 °C, 16h No 7 DMF, rt
Jd N Ss
H 9
INT-2 C 5a
J a 0 o © 0 0 pi Hs J Hs Ho 5 / NHBoc / NH, pa
Os) N= As N=
TFA INT-3b
N > — N x” ——— rN DCM, rt , NL COMU, DIEA, DMF = N = N
N N,
S $0 0) 5b C5 5c oO o oh 0 4 ooh O6 o "Tel he o He Jo < NE d NS (S) N== (S) N="
Ss Ss
AA = + NN =
N Cs N
N, N, © ) Sh )
N 5 N 5' ©) ©) 67
BL-5786
LU505465
Use INT-7 to replace INT-6 in the synthesis step of compound 2, and use similar methods and reaction steps to obtain compound 5 and epimer 5’. The absolute configurations drawn for the two compounds are based on empirical assumptions.
Compound 5 is an isomer with relatively long retention time in LC-MS and HPLC, and 5’ is an isomer with relatively short retention time in LC-MS and HPLC.
Compound 5:
ESI-MS (m/z): 901.8 [M+H]"; LC-MS retention time RT=1.62 min. HPLC retention time RT= 11.78 min. !H NMR (500 MHz, DMSO-d6) 6 8.52-8.47 (m, 2H), 8.35-8.30 (m, 1H), 7.81- 7.79 (m, 1H), 7.75-7.71 (m, 1H), 7.58-7.54 (m, 1H), 7.19-7.15 (m, 1H), 5.57 (t, J=9.5
Hz, 1H), 5.10-5.05 (m, 1H), 4.46-4.40 (m, 2H), 4.33-4.15 (m, 7H), 3.61-3.54 (m, 6H), 3.31-3.27 (m, 1H), 3.23 (s, 3H), 3.18-3.11 (m, 1H), 2.96-2.91 (m, 1H), 2.79-2.72 (m, 1H), 2.42-2.35 (m, 5H), 2.10-2.05 (m, 1H), 1.83-1.74 (m, 2H), 1.53-1.47 (m, 2H), 1.33 (d,J=6.0 Hz, 3H), 1.25-1.20 (m, 1H), 1.08-1.04 (m, 4H), 0.95-0.85 (m, 7H), 0.56-0.52 (m, 1H), 0.36 (s, 3H).
Compound 5°:
ESI-MS (m/z): 901.9 [M+H]". LC-MS retention time RT=1.62 min. HPLC retention time RT= 11.76 min. !H NMR (500 MHz, DMSO-d6) § 8.55-8.50 (m, 2H), 8.36-8.31 (m, 1H), 7.80 (s, 1H), 7.74-7.70 (m, 1H), 7.54-7.48 (m, 1H), 7.36-7.28 (m, 1H), 5.55 (t, J= 9.5 Hz, 1H), 5.07-5.02 (m, 1H), 4.47-4.32 (m, 3H), 4.29-4.20 (m, 3H), 3.99-3.92 (m, 1H), 3.88-3.79 (m, 2H), 3.70-3.65 (m, 1H), 3.61-3.48 (m, 6H), 3.19-3.11 (m, 1H), 3.09-3.06 (m, 3H), 3.05-3.00 (m, 1H), 2.80-2.73 (m, 1H), 2.43-2.35 (m, SH), 2.15-2.10 (m, 1H), 1.83-1.77 (m, 2H), 1.56-1.48 (m, 2H), 1.24-1.20 (m, 3H), 1.13-1.05 (m, 7H), 0.97-0.85 (m, SH), 0.59-0.50 (m, 4H).
Example 6 (1S,25)-N-((63S,4S,Z))-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-((4-morpholino- 1-oxidotetrahydro-2H-116-thiopyran-1-ylidene)amino)pyridin-3-yl)-10,10- dimethyl-5,7-dioxo-61,62,63,64,65,66-hexahydro-11 H-8-oxa-2(4,2)-thiazola- 1(5,3)-indola-6(1,3)-pyridazinacycloundecaphane-4-yl)-2-methylcyclopropane-1- 68
BL-5786 ©) LU505465
On" NO
SP ON 0 o Mel es / N™
Q H =) (S) N=
S
N x rN
Qo = N a © carboxamide 6
Example 6 was prepared by the following steps:
TO
(S)
N és
So Br
Noo = QO, an © DE > A } ; HN=N °
HN-N C SY To o Ie o_/ INT-10 78 LES AN oO = TA (s) 3 Sg) ZI
N= HN Ss) Pa(dppfCla, K3PO4, N \ / > > CsCO3, ~° | dioxane, HO, 70 °C _ DMF, rt a6. J > joxane, Ha Ç N dN S=0
INT-3 ¢ 6a " o/
O, O,
Da ) NE )
HN=N HN=N 0 ©) 90 0 ©) 90 78 HN 79 HN © N= “9 © N= “9
SN + xx
PN >= PN >=
Dai (54
NN 0 NN 0 © ; ("
Use INT- 10 to replace INT-8 in the synthesis step of compound 1, and use similar methods and reaction steps to obtain compound 6. Compound 6 and its epimer 6° cannot be separated and purified by reverse preparative liquid chromatography and normal phase column. Both epimers have the same retention time in LC-MS and HPLC.
Compound 6+6’: 69
BL-5786
ESI-MS (m/z): 929.8 [M+H]*; LC-MS retention time RT=1.58 min. HPLC LU505465 retention time RT= 11.28 min.
Example 7 (1S,2S)-N-((63S,4$,Z)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-((4- (morpholinomethyl)-1-oxidotetrahydro-2H-116-thiopyran-1- ylidene)amino)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-61,62,63,64,65,66- hexahydro-11 H-8-oxa-2(4,2)-thiazola-1(5,3)-indola-6(1,3)- pyridazinacycloundecaphane-4-yl)-2-methylcyclopropane-1-carboxamide 6)
Oye NO à o Hel Le
J À Vo (S) N=
S
N Is
NL
0 = N
S=N —
I pa
Example 7 was prepared by the following steps: /
Qs as
Noo s= ° le oO
JR / / 4 hs
HN—N N d oc
O (S) =
N=X HN-Boc ————————— NS ——
S Pd(dppfCla, K3PO4, _ Cs2COs, 0, > dioxane, H,0, 70 °C, 16h N DMF, rt x BL No " dN 5?
H
INT-2 y 7a
N
U
70
BL-5786
LU505465
Oo en O O. le Oo 0
J N (S) à N (S) Ho—4 oO Oo Ss) / NHBoc / NH; Do.
Ae N= A N=
Ss TFA Ss INT-3b
PN DCM, rt NN COMU, DIEA, DMF — N — N
Noo Ç Noo Ç
N N
O3 O3 oe 9 a 99 o MeN Jus o "Mel Je de N 7 NTS des N 176
N SS N ~°
NL + NI
D ei
N
No \ $=0 N ; 7 ( 7
CN a
Lo Lo
Use INT- 11 to replace INT-6 in the synthesis step of Example 2, and use similar methods and reaction steps to obtain compound 7 and epimer 7’. The absolute configurations drawn for the two compounds are based on empirical assumptions.
Compound 7 is an isomer with relatively long retention time in LC-MS and HPLC, and 7’ is an isomer with relatively short retention time in LC-MS and HPLC.
Compound 7:
ESI-MS (m/z): 943.8 [M+H]'; LC-MS retention time RT=1.66 min. HPLC retention time RT= 11.92 min. 'H NMR (500 MHz, DMSO-d6) 8.55-8.44 (m, 2H), 8.39-8.33 (m, 1H), 7.81- 7.76 (m, 1H), 7.75-7.69 (m, 1H), 7.60-7.49 (m, 1H), 7.25-7.19 (m, 1H), 5.60-5.51 (m, 1H), 5.11-5.01 (m, 1H), 4.34-4.12 (m, 4H), 3.99-3.79 (m, 1H), 3.60-3.51 (m, 6H), 3.49- 3.42 (m, 2H), 3.26-3.19 (m, 3H), 3.18-3.10 (m, 1H), 3.10-3.05 (m, 1H), 3.00-2.89 (m, 1H), 2.82-2.70 (m, 1H), 2.43-2.26 (m, 6H), 2.21-2.16 (m, 2H), 2.14-1.98 (m, 4H), 1.97- 1.89 (m, 1H), 1.83-1.71 (m, 2H), 1.63-1.47 (m, 4H), 1.36-1.30 (m, 2H), 1.25-1.19 (m, 1H), 1.14-1.08 (m, 1H), 1.08-1.03 (m, 4H), 0.97-0.93 (m, 1H), 0.91-0.84 (m, SH), 0.58- 0.47 (m, 2H), 0.41-0.29 (m, 2H).
Compound 7’: 71
BL-5786
ESI-MS (m/z): 943.9 [M+H]". LC-MS retention time RT=1.66 min. HPLC LU505465 retention time RT= 11.84 min. "H NMR (500 MHz, DMSO-d6) 6 8.57-8.47 (m, 2H), 8.39-8.33 (m, 1H), 7.80 (s, 1H), 7.72 (dd, J = 8.7, 1.7 Hz, 1H), 7.54-7.46 (m, 1H), 7.39-7.32 (m, 1H), 5.61-5.48 (m, 1H), 5.10-4.99 (m, 1H), 4.30-4.15 (m, 2H), 3.99-3.89 (m, 1H), 3.89-3.76 (m, 2H), 3.71-3.63 (m, 1H), 3.60-3.52 (m, SH), 3.49-3.41 (m, 2H), 3.18-3.12 (m, 1H), 3.09-3.05 (m, 3H), 3.04-2.99 (m, 1H), 2.81-2.72 (m, 1H), 2.48-2.39 (m, 2H), 2.36-2.28 (m, 4H), 2.20-2.17 (m, 1H), 2.16-2.01 (m, 4H), 1.96-1.89 (m, 1H), 1.84-1.75 (m, 2H), 1.62-1.48 (m, 4H), 1.24-1.19 (m, 3H), 1.12-1.04 (m, 7H), 0.94 (s, 3H), 0.90-0.86 (m, 1H), 0.58- 0.54 (m, 1H), 0.54-0.48 (m, 3H).
Example 8 (1S,25)-N-((63S,4S,Z))-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-((4-(oxetan-3- yl)-1-oxido-116-thiomorpholin-1-ylidene)amino)pyridin-3-yl)-10,10-dimethyl-5,7- dioxo-61,62,63,64,65,66-hexahydro-11 H-8-oxa-2(4,2)-thiazola-1(5,3)-indola- 6(1,3)-pyridazinacycloundecaphane-4-yl)-2-methylcyclopropane-1-carboxamide
O SL hs d va (S) N= . o = N
D —
N uf
Example 8 was prepared by the following steps: 72
BL-5786
LU505465
N gg: de N 7 va if 5°
RO” ps
N, 0 — ° +
A
N.
S=0 ® ;
Step 1: Compound 4e (30 mg, 0.04 mmol) was dissolved in DCM (5 mL), thereto were added 3-oxetanone (13 mg, 0.18 mmol), triacetyl sodium oxyborohydride (23 mg, 5 0.11 mmol) and glacial acetic acid (2 mg, 0.04 mmol). At room temperature the reaction solution was stirred for 16 hours. After the reaction was complete, the reaction solution was added with DCM (15 mL), and the organic phase was washed twice with water and concentrated. The residue was purified by preparative liquid chromatography to obtain a white solid compound 8 (4 mg, yield 12.9%) and epimer 8° (6 mg, yield 18.7%). The absolute configurations drawn for the two compounds are based on empirical assumptions. Compound 8 is an isomer with relatively long retention time in LC-MS and HPLC, and 8’ is an isomer with relatively short retention time in LC-MS and
HPLC.
Compound 8:
ESI-MS (m/z): 901.9 [M+H]'; LC-MS retention time RT=1.58 min. HPLC retention time RT=11.19 min. 'H NMR (500 MHz, DMSO-d6) & 8.51 (d, J = 9.0 Hz, 1H), 8.48 (d, J = 1.6 Hz, 1H), 8.38 (d, J = 2.6 Hz, 1H), 7.79 (s, 1H), 7.73 (dd, J = 8.7, 1.7 Hz, 1H), 7.56 (d, J = 8.6 Hz, 1H), 7.25 (d, J = 2.6 Hz, 1H), 5.57 (t, J= 9.2 Hz, 1H), 5.11-5.05 (m, 1H), 4.54 (t,J=6.6 Hz, 2H), 4.42 (t,J = 6.1 Hz, 2H), 4.36-4.11 (m, 5H), 3.76-3.68 (m, 1H), 3.61- 73
BL-5786 3.54 (m, 2H), 3.49-3.38 (m, 4H), 3.22 (s, 3H), 3.18-3.11 (m, 1H), 2.97-2.93 (m, 1H), | L|U505465 2.87-2.73 (m, 5H), 2.44-2.35 (m, 1H), 2.10-2.05 (m, 1H), 1.84-1.72 (m, 2H), 1.57-1.43 (m, 2H), 1.33 (d, J = 6.1 Hz, 3H), 1.09-1.01 (m, 4H), 0.92-0.83 (m, 6H), 0.57-0.52 (m, 1H), 0.35 (s, 3H).
Compound 8’:
ESI-MS (m/z): 901.9 [M+H]'; LC-MS retention time RT=1.58 min. HPLC retention time RT= 11.19 min. 'H NMR (500 MHz, DMSO-d6) 5 8.56-8.50 (m, 2H), 8.38 (d, J = 2.6 Hz, 1H), 7.80 (s, 1H), 7.72 (dd, J = 8.6, 1.7 Hz, 1H), 7.51 (d, J = 8.6 Hz, 1H), 7.38 (d, J= 2.7
Hz 1H), 5.54 (t, J= 9.2 Hz, 1H), 5.08-5.04 (m, 1H), 4.54 (t, J = 6.6 Hz, 2H), 4.42 (t, J = 6.2 Hz, 2H), 4.28-4.17 (m, 2H), 3.98-3.78 (m, 3H), 3.75-3.63 (m, 2H), 3.58-3.52 (m, 1H), 3.45-3.38 (m, 4H), 3.19-3.11 (m, 1H), 3.08 (s, 3H), 3.05-3.00 (m, 1H), 2.87-2.69 (m, SH), 2.45-2.40 (m, 1H), 2.15-2.10 (m, 1H), 1.86-1.76 (m, 2H), 1.59-1.45 (m, 2H), 1.21 (d, J = 6.3 Hz, 3H), 1.13-1.04 (m, 6H), 0.94 (s, 3H), 0.90-0.84 (m, 1H), 0.58-0.53 (m, 1H), 0.51 (s, 3H).
BIOLOGICAL SCREENING AND RESULTS OF RAS INHIBITORS
Test Example 1: In vitro cell proliferation inhibition test
Due to the diversity of RAS mutations and in order to evaluate the activity of compounds in different RAS mutant cell lines, we selected KRASYT KRASS!?C,
KRASS!P, KRASS!?Y and BRAF mutant cell lines (see the table below) for in vitro activity evaluation and screening of compounds. 74
BL-5786
Experimental protocol: CellTiter-Glo® Cell Luminescent Viability Assay (Promega)
According to the doubling time of different cell lines, different numbers of cells (1000-5000 cells/well) were inoculated into 96-well plates containing 180 ul of the corresponding medium, and cultured overnight in a 37°C cell incubator containing 5%
COs. On the second day, the compound to be tested was pre-diluted 3-fold with the medium, the highest concentration was 100 uM, and there were 10 concentration gradients in total; then 20 pl of the medium containing different concentrations of the compound was added to the cells in the 96-well plate to ensure the final concentration ofthe compound to be up to 10 uM, with 10 concentration gradients of 3-fold dilutions. After co-incubating cells and compounds for 72 hours, the 96-well plate was taken out of the incubator, placed at room temperature for 30 minutes, and then 25 ul
CellTiter-Glo® Reagent was added to each well to mix well, and incubated at room temperature for 10 minutes, and then 100 ul of the sample was transferred to a white 96-well plate (OptiPlateTM-96, PerkinElmer), the fluorescent signal value was read using a multi-functional microplate reader (SpectraMax® 13x, Molecular devices).
Subsequently, the signal value was standardized, and the four-parameter fitting regression equation was used for curve fitting to calculate the half maximal inhibitory concentration (IC50) of the compound on the cell line.
Table 1: Antiproliferative activity of compounds of the present invention on
KRAS cell mutants
NCI- NCI- | NCL
PaCa- | LS513 | AsPC-1 | HCC1588 | SW480 HT-29
H358 H727 | H520 2 75
BL-5786
LU505465
La [ow [wo [we oe pe | ew [oe [ow
Lew [ew [wo [ww
To rw [a [ow [wo [wr Jem]
Lo [ow Jon ow [om ww [wr * NT MEANS NOT TESTED 76

Claims (6)

Claims
1. A compound having the structure: 9 Og y" oO î ou nt °9 Og nt oO 9 7 HN-N ° OÖ mi) oO IS) (5) oO © iS) o 3 d û û o de N vo d N Vo d N WA —o # Cl Ne 9 Ne 9 N= NA HN 5) Ss Ss Ss ia N = N: xy, N- te, N: >, 4 N¢ rN rN CN 9 N o N 9 N N C4 À Ce — sen À MN — M7 (J : $ — N © 3 / Ca GA or y" o 3 OS yt °9 0 ns © & Hig d NT 4 N = 5, Ne N > N x rN IN N N No — N, + N © (> © ° Oy © °0 oy © 99 Os ©) 99 Os © 99 Fr H H H H 0 ol, Me 0 ol, Ao 0 ol Mo 0 ol de (3 N= ® (9 N= ® © N= (9 N= =n 5 > 5 = 5 = 5 NS NA NA A 9 > 9 > 9 > 9 > $=N $=N §=N $=N / / / / oy we Pa 7 Sy i Pa ÿ Os © © 9 J N ©) i 1 / © © Ao / © © Heb / © 9 Ney J © NT Ji Om nA Og nÀ Se n=l " © N= N > N > N = N = Ny A rN rN rN A 2 N 2 N 2 A 2 > S=N — S=N — an TN © (J (J A 7 A 7 Os el 0 Oy © Oo Oy © 99 Oy ©) 0 UN 8 > : H F H ; J Te Anim / °o "a vus / © Oy Ah CF / © 9 ve Sa N H a) Sw N= F Xe N= H a NE Ss SS = 7 NA 7 > PA NA 7 NS 7 0 N 2 N 2 7 3 _ 0 _ 4 + S=N — S=N — 3 N a N © ow M7 he / © © ; ©) Oy “y” °4 Oy N° °4 Op a) °5 N Oy y" °8 N 0 & . Ô & 3 U 0 (3) 3 i Ô ©) l. SF J NR on d N Rio d VE J Ne Q N= 9 N= © N= 9, N= am 5 = 5 = = 5 NA Mg NA A 7 0 N oO N 0 5 0 + §=N — NJ $=N $=N 4 4 7 7 Os © 99 Os ©) 99 Os © 90 Oy GA 99 ; à He Ads , A) Ads ; He nt © or , à Hg A 0 © 0 H 0 H 0 H © 0 H Ss) N= LUNG S) N= L_o 2 N= © NA æx-0 5 Ss = x NA NA A NA 2 5 2 5 2 A 2 7 §=N §=N A $=N (J (J (7 (J 7 7 ~ 4 Oy GA °0 of Oy GA 99 op lad 90 Oy GA 99 / / © "a / o "9 Ad / © es / © AE Og N= A ea n=l " 8 0 Og N= A wo N “a n=l " © = 5 = 5 NE Ng > NE 7 a > 9 + 9 5 9 + 9 + $=N $=N $=N $=N ea OS Os GA 99 N SON 0 ’ N ’ 4 F oO (5), CS TN i 0 A / N 0 ; N S) / « J He / oO ro o _ N @ | _ / N 9 Ne = i - Ny 3 N= = Wel ; , 0 N > © 4 My } 0 zz ) î J (TE J 3 { J §=N 3 ( ho ‘J  CO Ç N 7 61 0 Oy ba 00 > on 0, à OO No JW, N ; i L ’ 1 4 H 1 J N 0 H I iN / 0 Foy À ) ©) N H ©) N= 0 6 H A 0 NT ONS) _ 2 Ç a ! N > ESS 9 N Ne ©) N= py) N N ; " 4 ©) N= AON I = 9 4 12 MN À SS o \+ in _J A ‘ = 7 i 5h N (8 y  ) Ce 13 nN / © 5 GA 99 0 ON N / y sy © 5 J N ‘a / 0 © A OO N TON 0 J ol 100 ‘ ; . ° N , ; TON © o Hel 1 9 N 0 2 N H | / 6 N N i ‘ / Oo ©) Ao S o N 9 _ 8 vi) - © N H ©, N= LUN N oy = Na / > 4 ° u N 5 rN = 0 LT Ng = o \— N 4 5 3 he hr 3 3 qd ‘ nN 7 Gp © GA oy lad 99 7 1 Ny 1 —=N Og ® NO ° > NH 99 oH o Hy de As, / NON 5° u i 1 / N° N 9 À Hog À _7 oO so, À ÿ 0 N x E N o NN J Ay © . / J H H N N 7) - & N= 1s) N= 7) - LS NY 8 9 SI 2 (3 S=N AG 9 AN CN A BD o N (és 3 s=N J 3 ) CO A N 7 © u ze X / fa o. fo) SN N 0 6 X / o Fe I B05 g _o a Z SCI / ° Mg "M 9 ROM (8) N= ® ’ N 3. {OR bo ’ en ÿ 9 N @ N Ss / N N #87 H 5 ) : J - o H ) Ne me = { Ç es N 5 / / 9 =
N . MY = ; ; N SS / 0 (4, me oN 9 = N (Sen J 3 I N 1a ot 7 ; It A ; N EL fo) oy où ° > NN °% N j
0. Ss PONT o © , à he 6 ° Go EN / o 5. ol, À _7 o N SU NS / o Huw À @ 9 Ps NS 3 © o Hl 0 9 ~~ NTN 5 = ne x = / oN, hm oN wi) 9 SN © © Ne +, x = In 1s), N= RR} N { 5 ° } 9 ( N oS ° J 0 0 ~ = rN 0 = A A 2 = N ay _J G ; CN 3 ; N NS A on lol, 3 © SWS , à Folie Ogio SNS 0 / 0 [CI 9 N © T N" 9 / à Fo Je 0 N 4 8 _ A ’ PN 5 N 5 ©) N= N lB / © y ks 9 N © © N= 6 "4 = { iL © A > 0 4 sn N N = MN J => 0 = WW A Q = U N es ( = or (3 - s=N J 3 - \ N <7 0 Co 6) N
0. (5) ST 20 : oo J N °3 / 0 Hel Je OO BAZ / 0 ol Je ° N 4 ar ) ; N Ho Ye À N 9 / 0 olf, Mg N “ 2 _ 0 al de oO N © 6) Ne 6 N = / N ji q N © ©) N= ™ = = J ©) N= ® N = | EU N 5 Ng 2 =r ND (NA 0 (_ 7. = g = U N — ( $ N s=N J [ (JT - § ; & 7 eo wo 44 ) 2 N 3 4 0 Hg de Os No TON a y o "ol Je ° , / SN 0 / 0 [a 5 N Z 3 \ A , © Pe 5 N 5 ©) N= ® N oS q N © 8 N= ® N = wT © N= ® N 5 NE 2 (9 N \ SS 4 J 9 { q = a < / = Ios 3 I J! oe , jaan N x " 5)
LU505465A 2022-11-16 2023-11-09 A pan-KRAS inhibitor compound LU505465B1 (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202211463547 2022-11-16

Publications (1)

Publication Number Publication Date
LU505465B1 true LU505465B1 (en) 2024-05-14

Family

ID=89791270

Family Applications (1)

Application Number Title Priority Date Filing Date
LU505465A LU505465B1 (en) 2022-11-16 2023-11-09 A pan-KRAS inhibitor compound

Country Status (2)

Country Link
CN (1) CN117534685A (en)
LU (1) LU505465B1 (en)

Also Published As

Publication number Publication date
CN117534685A (en) 2024-02-09

Similar Documents

Publication Publication Date Title
TWI723511B (en) Highly active sting protein agonist compound
WO2022089454A1 (en) High-activity wnt pathway inhibitor compound
CN111989313A (en) A2A and/or A2B receptor antagonists
WO2020038387A1 (en) High activity sting protein agonist
US20230399327A1 (en) High activity hpk1 kinase inhibitor
EP4083032A1 (en) Pd-l1 antagonist compound
JP7281834B2 (en) PD-L1 antagonist compounds
LU505465B1 (en) A pan-KRAS inhibitor compound
LU505620B1 (en) A pan-KRAS inhibitor compound
LU505464B1 (en) A pan-KRAS inhibitor compound
LU505117B1 (en) A pan-KRAS inhibitor compound
CN111440148B (en) Preparation method of adenosine receptor antagonist
US20220267350A1 (en) Adenosine receptor antagonist
WO2024022365A1 (en) Wnt pathway inhibitor compound
WO2024104364A1 (en) Pan-kras inhibitor compound
TWI810547B (en) Pd-l1 antagonist compound
WO2024060966A1 (en) Pan-kras inhibitor compound
WO2022199561A1 (en) Hpk1 kinase inhibitor compound
WO2022214008A1 (en) Highly active hpk1 kinase inhibitor
WO2023005894A1 (en) Inhibitor compound for wnt pathway
TW202333697A (en) Wnt pathway inhibitor compound