LU505117B1 - A pan-KRAS inhibitor compound - Google Patents

A pan-KRAS inhibitor compound Download PDF

Info

Publication number
LU505117B1
LU505117B1 LU505117A LU505117A LU505117B1 LU 505117 B1 LU505117 B1 LU 505117B1 LU 505117 A LU505117 A LU 505117A LU 505117 A LU505117 A LU 505117A LU 505117 B1 LU505117 B1 LU 505117B1
Authority
LU
Luxembourg
Prior art keywords
int
mmol
compound
reaction
esi
Prior art date
Application number
LU505117A
Other languages
French (fr)
Inventor
Yufeng Chen
Feifan Li
Meng Lv
Wanli Cheng
Kaixuan Chen
Canfeng Liu
Han Yang
Shuaishuai Liu
Nanhai He
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
Priority to LU505117A priority Critical patent/LU505117B1/en
Application granted granted Critical
Publication of LU505117B1 publication Critical patent/LU505117B1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D515/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen, oxygen, and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D515/22Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen, oxygen, 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
    • 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)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

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

Description

BL-5758
A pan-KRAS inhibitor compound LUS05117
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 Us mutation occurs in about 30% of tumor patients, among which KRAS accounts for about 85% of RAS mutations, KRAS mutations exist in 88% of pancreatic cancer, 50% of colorectal adenocarcinoma, and 32% of lung adenocarcinoma. The development of targeted KRAS inlubitors has great clinical significance and value.
KRAS is a membrane-bound protein with GTPase activity, which cycles between a GDP-bound inactive conformation and a GTP-bound active conformation through nucleotide exchange, performing the function of a "molecular switch”, KRAS in the
GTP-bound state can activate multiple downstream signaling pathways including
RAF-MEK-ERK and PI3K-AKT, and regulate life processes such as cell growth, proliferation, differentiation and apoptosis.
ERAS mutations {such as GI2C, GI2D, GI2V, GI3D, etc.) will affect the hydrolysis of GTP mediated by GTPase activating proteins {3APs), 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 (-Z0pM), 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, AMG510 was approved by the FDA for the treatment of locally advanced or metastatic non-small cell lung cancer carrying the KRAS*PS mutation, breaking the history of KRAS being "undruegable”. However, the G12C mutation oniy accounts for a small portion of KRAS mutations. For mutations at other sites of
BRAK, 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. 1
BL-5758
Contents of the invention LUSOS117
The invention provides a pan-KRAS inhibitor. Such structures are different from existing KRASS!? inhibitors that act through covalent binding, but instead mediate the 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 having a structure of formula (I) or a pharmaceutically acceptable salt, isotopic derivative, or stereoisomer thereof’ y X Ay
N p
N NY
Re— NH a
J Re’ Ry
Ra N
R7 R7' © wherein:
Rı represents Ci-Cs alkyl, -(C1-C6 alkylene)-(Cs-Cs cycloalkyl) or -(C1-C6 alkylene)-(3-8 membered heterocycloalkyl);
Rz represents halogen, cyano, Ci-Cs alkyl, -(Co-Cs alkylene)-(Cs-Cs cycloalkyl), or -(Co-Cs alkylene)-(3-8 membered heterocycloalkyl), which may optionally be substituted by 0, 1 or 2 of the following substituents: -ORa, -SRa or -
NRaRa';
Rs represents hydrogen, -O (Co-Cs alkylene) Ra, -S (Co-Cs alkylene) Ra, -N (Co-Cs alkylene) Ra (Co-Cs alkylene) Ra’, -O (Cz-C6 alkylene) Ry, -S (C2-Cs alkylene)
Ri, -N (Cz-C6 alkylene) Ru (C2-Cs alkylene) Rr, wherein, Rr, Ru each independently 2
BL-5758 represents -ORa, -SRa, or NRaRa', LU505117
Cy1 represents C3-C12 cycloalkyl or 3-12 membered heterocycloalkyl;
R4 represents hydrogen, halogen, oxo, C1-Cs alkyl, -(Co-Cs alkylene) (C3-C6) cycloalkyl, -(Co-Cs alkylene) (3-8 membered) heterocycloalkyl, -(Co-Cs alkylene) ORa, -(Co-Cs alkylene) SRa, -(Co-Cs alkylene) NRaRa', -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-C6 alkylene) (Cs-Cio 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, and the 3-8 membered ring optionally can contain 0, 1, 2 or 3 members selected from A heteroatom of N, O or S; or two R4 on the same C atom of Cy1 together with the C atom connected thereto can form a 3-8 membered ring, and the 3-8 membered ring can optionally contain 0, 1, 2 or 3 heteroatoms selected from N, O or S.
Rs represents-Cy2-(Rs)q Or-NRoRs', wherein,
Cys represents C3-C12 cycloalkyl, 3-12 membered heterocycloalkyl, Cs-Cio aryl or 5-12 membered heteroaryl;
Rs represents hydrogen, halogen, oxo, C1-C6 alkyl, -(Co-Cs alkylene) ORa, -(Co-
Cs alkylene) SRa, -(Co-Cs alkylene) NRaRa', or Rs 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 can contain O, 1, 2 or 3 heteroatoms selected from N, O or S; or two Rs on the same C atom of Cy; together with the C atom connected thereto 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 at least one atom on the Cy» ring is substituted by S(FO)FNRa) or S( =0);;
Ro and Ro’ each independently represent C1-C6 alkyl, C3-Cs cycloalkyl, 3-8 membered heterocycloalkyl, Cs-Cio aryl or 5-12 membered heteroaryl which may be optionally substituted by q RS;
Rs and Re each independently represent hydrogen, halogen, C1-C6 alkyl, C3-Cs cycloalkyl or -(Co-Cs alkylene)CN
R7 and R7 each independently represent hydrogen, halogen, C1-C6 alkyl, C3-Cs cycloalkyl, 3-8 membered heterocycloalkyl; or R7 and R7 form a 3-8 membered ring, which may optionally contain 0, 1, 2 or 3 heteroatoms selected from N, O, S. 3
BL-5758 wherein, p and q independently represent 0, 1, 2, 3 or 4; LUSOS117 m represents 0, 1, 2 or 3;
Ra and Ra' independently represent hydrogen, C1-Cs alkyl, and Cs-Cs cycloalkyl; wherein, if Ra and Ra' are connected to the same N atom, the Ra and Ra' can be connected to the common N atom to form a 4-8 membered ring, which optionally may contain 0, 1, 2 or 3 heteroatoms selected from N, O or S. each of the alkyl, cycloalkyl, heterocycloalkyl and alkylene can be independently substituted by 0, 1, 2, 3, 4, 5 or 6 halogen atoms.
Furthermore, the present invention provides a compound having the structure of formula (I) as described above or a pharmaceutically acceptable salt, isotopic derivative, or stereoisomer thereof, wherein R; represents a C1-Cs alkyl, preferably a C1 -Cz3 alkyl.
Furthermore, the present invention provides a compound having the structure of formula (I) as described above or a pharmaceutically acceptable salt, isotopic derivative, or stereoisomer thereof, wherein Ra represents C1-C6 alkyl, which can optionally be substituted with 0, 1 or 2 -ORa substituents. More preferably, Ra represents, preferably, wherein, * represents the site where Ra is linked to the linking site in formula (I).
Further, the present invention provides a compound having the structure of formula (I) as described above or a pharmaceutically acceptable salt, isotopic derivative, or stereoisomer thereof, wherein Rs represents -O(C1-C6) alkyl, -O(Co-Cs alkylene) (C3-C8) cycloalkyl, -O(Co-Cs alkylene) (3-8 member) heterocycloalkyl, -O(Cz-C6) RL or hydrogen
Further, the present invention provides a compound having the structure of formula (I) as described above or a pharmaceutically acceptable salt, isotopic derivative, or stereoisomer thereof, wherein Cy; represents a C3-Cg cycloalkyl or 3 -8 membered heterocycloalkyl.
Further, the present invention provides a compound having the structure of formula (I) as described above or a pharmaceutically acceptable salt, isotopic derivative, or stereoisomer thereof, wherein R4 represents hydrogen, halogen, Ci-Cs alkyl , -(Co-
Cs alkylene) CONRaRa', -(Co-Cç alkylene) NRaCORa', -(Co-Cs alkylene) OCONRaRa', 4
BL-5758 -(Co-Cs alkylene) CN, -(Co-Cs alkylene) (5-12 membered heteroaryl), or R4 on the two LUS0S117
C atoms of Cy1 together with the C atom connected thereto and the atoms between the two C atoms can form a 3-8 membered ring, and the 3-8 membered ring optionally can 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 may form a 3-8 membered ring, and the 3-8 membered ring may optionally contain 0, 1, 2 or 3 heteroatoms selected from N, O or S.
Further, the present invention provides a compound having the structure of formula (I) as described above or a pharmaceutically acceptable salt, isotopic derivative, or stereoisomer thereof, wherein R4 represents hydrogen, halogen, Ci-Cs alkyl , -(Co-
Cs alkylene) CONRaRa', -(Co-Cs alkylene) (5-12 membered heteroaryl), or R4 on the two C atoms of Cy; together with the C atom connected thereto and the atoms between the two C atoms can form a 3-8 membered ring, and the 3-8 membered ring optionally can contain 0, 1, 2 or 3 heteroatoms selected from N, O or S; or two R4 on the same C atom of Cy1 together with the C atom connected thereto can form a 3-8 membered ring, and the 3-8 membered ring can optionally contain 0, 1, 2 or 3 heteroatoms selected from N, O or S.
Further, the present invention provides a compound having the structure of formula (I) as described above or a pharmaceutically acceptable salt, isotopic derivative, or stereoisomer thereof, wherein Cy, represents a 3-8 membered heterocycloalkyl or 5- 12 membered heteroaryl.
Further, the present invention provides a compound having the structure of formula (I) as described above or a pharmaceutically acceptable salt, isotopic derivative, or stereoisomer thereof, wherein Rs represents hydrogen, halogen, C1-Cs alkyl, -(Co-Cs alkylene) ORa, -(Co-Cs alkylene) NRaRa'; or Rs on the two C atoms of Cy, together with the C atom connected thereto and the atoms between the two C atoms can form a 3-8 membered ring, and the 3-8 membered ring optionally can contain O, 1, 2 or 3 heteroatoms selected from N, O or S; or two RS on the same C atom of Cy2 together with the C atom connected thereto can form a 3-8 membered ring, and the 3-8 membered ring optionally can contain 0, 1, 2 or 3 heteroatoms selected from N, O or S;
BL-5758
Furthermore, the present invention provides a compound having the structure of LUSOS117 formula (I) as described above or a pharmaceutically acceptable salt, isotopic derivative, or stereoisomer thereof, wherein at least one of Ro and Ro represents Ci-Cs alkyl substituted by q RS.
Furthermore, the present invention provides a compound having the structure of formula (I) as described above or a pharmaceutically acceptable salt, isotopic derivative, or stereoisomer thereof, wherein Rs and Re: each independently represent hydrogen or
C1-Cs alkyl; more preferably, Rs and Re: each independently represent hydrogen or methyl.
Furthermore, the present invention provides a compound having the structure of formula (I) as described above or a pharmaceutically acceptable salt, isotopic derivative, or stereoisomer thereof, wherein R7 and R7 each independently represent hydrogen, C1-
Cs alkyl; or R7, R7 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. More preferably, R7 represents hydrogen.
Further, the present invention provides a compound having the structure of formula (I) as described above or a pharmaceutically acceptable salt, isotopic derivative, or stereoisomer thereof, wherein m, p, and q are each independently preferably 0 , 1 or 2.
Further, the present invention provides a compound having the structure of formula (I) as described above or a pharmaceutically acceptable salt, isotopic derivative, or stereoisomer thereof, wherein the structure of -Cyi-(R4)p is selected from the following: 6
BL-5758 on Fo ; : SE LU505117
BD ns uf . i i, oy ve 7 MOT EEE RE PS TS
SN A # N A * N A N A Ra © > > ©
LUN (0 10 ON ©)
N N (S) N
ARE BD we ©\_0 ONNL © © Du ®" ©
N
N N _ y AN ! #.(9 HAN 5.05) - “er #45) NT =,(S) A(S) 76 oH rm OH Vo © / x
HQ | ° o— ~—OH \ = A 0 +, * a > * (R) N
VO Ee À » 0 5 © 3
Pet oO 9 N _ 0 0 * N= , "a we ao N = | Er EY (S) "
Un Lo wherein, * represents the site where -Cy1-(R4)p is linked to the linking site in formula (I).
Further, the present invention provides a compound having the structure of formula (I) as described above or a pharmaceutically acceptable salt, isotopic derivative, or stereoisomer thereof, wherein the structure of -Cy2-(Rs)q in formula (I) is selected from the following: 7
BL-5758 . ; x LU505117 * x x ; # (R) & ) ct ) 2 (S) dde & oO Fg
N N ~ ~ / / \ \
SS ae Dr OD CO ©
N—= + OO N-= — + \ / \_/
N
A / > \ = oo a ha GE Ö ) X
Ca RN OQ) N—= a
ON ON 4 4 Cg (Ug ~~ =
Le CC a N Ss. N-=
J N-s GO SOON 6) \ N°
IN OR
_ JUN IN / N o _N-= 7 TU —* N—= — x — x O N—* — x
Re D
OH 0 HO Ho— OH / \ HO
HO,(9) HOw 0 _ /\ ee De De 3,00 HO- ro + HO N N A bs
Os \ Os¢/ Ws WS = Ç , Ç y NNN y
HN Nos oN °N N 4 N —_ EN
SS \ 2 | / / \
N-N NH, N ~ wherein, * represents the site where -Cy2-(Rs)q is linked to the linking site in formula (I).
Furthermore, the present invention provides a compound having the structure of formula (I) or a pharmaceutically acceptable salt, isotopic derivative, or stereoisomer thereof, wherein the compound of formula (I) is the compound of formula (II) structure: 8
BL-5758
LU505117
S)
Ou" .N__O a N O
V1
R H
R, N=\ F3
N SS
Re / \ Ÿ —_ N
VA Rg' R4
Rs
R7 R7'
Formula (IT)
In another aspect, the present invention also provides compounds having the following structure:
Os GX 9, Os (5 9, Os GA 90 Os, X 0 1 Fol de Ï Fon Ne 1 Tol Js F 1 N © 3» LF d, Ne N wa J, na N © d, Ne N a J, N N og s 5 s >
NS = NS > A > N= > — — J = —/ 7 A / —
ON ON qu, oN
Oxy (A 20 A Oy GA 20 N oa © oO 0 Oy © 9 oO oO
J, 0 Aer 4, 0 "hed 4, 9 Fe 4, 9 TA Er
AR RO SR SR
A A A N
— 7 — À I 7 ~~ Ÿ oN oN \ A
Oxy (A °° Oxy GA Og . Oy GA 99 Os Ga °4 > 5 : Ë © F 5 .
Ne 5 _ a _ =
AR SR SR RO
N N N N
A — J A dh / J / J / dh /
NS NS NS NS oy lad O9 Oy GA 99 Oy GA 99 Os ex 00
Fe, de Fe Le, o Tel Je 2N Hog MN d, ) "A GO om dr ) Then oH J, A RE J, ° hes
NA 7 N SS MN 4 = ) N ° i 7 AN 4 rN
A 5 — J +} — f ay Ÿ — 7 oN oN \ oN
On © 0 Os ex 0 Os GA o Os Ge 0
TW THR TRL TNL
/ oO (5), N ws / oO N N y Ô N ao / Ô N © a TU ALU RL da LS
S
NA | > Pg > Aa > NA =
À > a N < N y < N y < N y < N /
Ox Ge 0 J Os Ge 9, Oz GA 9, Oy Ge °, ,
Pa © i 3 Fel JL + Hol los o Tw N / © eno / NN / N / N eo
Go Ln fo LN fe LVR af AY 5 5 SS N =
NA | > AN a A — — 5 —
J
J / oN / of N / oN (
Nf 9
» | hoo 8 N_0 LN PSN 8, à 0. © o Oy N 9 3 H © 2
Os) N_o STUN 9 ° oN Aly / N 5
SN 9 o Muy / N 0 o Hu N / N a, N= L_o S NEY << H Og N= oS N >
S N= 8 N \ À ON
N 4 = NA N N
HN N N / J ÿ — Ÿ CN J ÿ IN q \/ oO N A X fo ° Ox TP 5 à o S NO 3 N 9 o 3) N / N >
SON 9 o Ly S / H o Og N= 6 Huy ons J N Os N= SS / ñ N= SS N a, A, ON N = LS Ng ay € > rN, N
A N J — J — Y — IN 7 d û SN Oo N \
À ff N \ oN \ \ X
ON Oho fo | ° oa o TN 9 Se tN o shy À,
Ss) N Our y 99 IN De / © N o° NO i TOR 9 N
SON 9 5 sh, A / N 0 OS N= o Ful Jets / NON OS N= © 8 o” / N O NE UN . SS NA À
Og N= N AS NA À N
N > 7%] N J
AN N J Ÿ
N A — I / CN — y N \ 7 Jd \
I 3
N ole) 0 = Oy NSP 0 —=N 0. © NO HO SON 9 ] J Hg py 0.9 No SON o = o Mel Je SN / NON
SONT 9 —OH o Hel J em / N © J H o Hel J / NONTTH d = 9 NA, / N m J H 2 s = d H 8 N= N 3 = NA Ms ¢
NA 7 = NE N À ¢ 4 N J — Ÿ — Ÿ ON ON ÿ I 0 ns
VAR dN \ <N \ ©} Os NO à 8 0 N
N Os X 04 | Oy y” 1 Se q H 0 A
OS N_o SON SN 0 NN 0 / ON ©
SON 9 0 ® 9 à / NON Og NS |® "0 Hod J 750 Og N= ® 5 / = 5 N
On we, N N 2 = NC 7 = a $
N NN N
NA 7 N + Vo
N J Ÿ I
A Ÿ I oN 7 an dp \ — dN
ON —
Q 0a THLE 0
Gi» ee à TLL To aN oy ° OÖ Ho A / ON A 0 NEO à Hel lg, / ON 0 Ce n=O oS / OLN © Og N= 9 © Sa NA À 0 H ® Le N = ÿ 3 NS) N = NI N / rN J
N N rN N J Y
N J Ÿ an
Ÿ I N
/ — I oN \ —~ J ON \ oN i \ ©) 5) oO ©. N o. Ga o SON 0
LS N LO SON 9 Hs) |, oO. N oO Os N 9 d Fol À, © / © N° te
TN 9 o "ol le / N a d H o Hoel Je / N Vo o H 5 N= / N Vo J H 5 N= NJ]
J H S N= = N 5) N= _ NA À LS oS N i N
N A N rN N J / —
N À Ÿ \ — Ÿ \ N°(@
Ÿ NS C
_ o
N o /
JO N_o Hol Le © ol lod ° 0 Oy N 1 / ° N a 0 NO Q SON o "ol Le oO
TN o Mul Le / N a 5 N= 6 / NE 6 N N SS
J H © Ou N= = Ss rN
N= SS N > N
SS N AN J
N rN 7
N Ÿ
Sd N J
N J Ÿ
Jo / 7 N
O1 PO N / & a
N \ 6)
OE N_O 0 lod oO 5 N
MS NO SOON ° sh Mo 0... © 96 Os N 9 7 Hol le / © N LE
SON 1 o Hol Le / NE 0 N= ° À de / N Vo o H = s / HN ® 0, H > N= Ny 0 H 3 N= Ss Ng
Ss) N= Ss N J 4
N AS NA) a N
LS ç N 4 J
N _Ù Ÿ — 7 = U ®
Ÿ ® © CS © - N
N— N \
N
©. fen o o © o oa ©}
N° o Se N° o Oy N NO Oy NN 0 y o He We / o He n°49 , o Hel Ja , o Ho ol 5 o H Vo 0 H Vo o N To d À Vo © N= 5) N=! 5 N= 2) N=
SS SS Ng 8
NA Ms NN AR — — A N 7 7 ÿ 7 — = (9 ®
HN. 7 NG N Un
Os GA 99 oy GA °e Os GA 99 Oy GA 90 o Hol Je , o Hal Jy o Hal lg ; o "el ly
J NE à NRE d N°6 d NE © NE > 3) N= > 3) N= s 3) N= > = = we <
NS 9 A Mg NA
N N N N
PIN Vo Jo Jo oN ° N CN on \@ J
Oy GA 96 DR 9 Oy GA 9 Os GA °,
H H H | Hol Jl ° À, is ° sn À. o ol J 0 © „(9 / N
SA s N <8 N Ss
NS 9 NA > A NA
N N N N
A / NT / /
Tw Og N N N
HN
OH ° \
Ps, GA 99 oy °0 Oss (GA 99 Os GA 99 hp Hol Je , o Hao wl © d Hol Je y o Te Ae dg H 7 Os N N Ve dg N H rE Os N N va
N= = = =
N = N => N SS N VS rN Nd rN a4
N N N N
# & Ÿ N # & Ÿ Q
VN y Le M
X Os GA 99 Os GA 20 Oy ex °°
Og GND à Hk Je JON à * A de
Th sl, { N Lo / © Tw TE / N RA / © N RA Ag N= 9 H © Os A A
Sg N N © N => < ls 3 () 5 FY / Oo Ng oO > A
A N N ï ÿ IN # « Ÿ k / a - — (ON ~ A
NN N ~ NH, 3 N I) 3 (5) oy (A 0 oO Oy © 9 oO Oy (A 9 oO
J N va o Mog Je o Hell le o Hel Je a N= J N D dd N Lo dd N Le
N SS 5) N= 5) N= 5) N=
NV N 5 N 5 N eS
N ON 7 NN NN ÿ — Bi Bi Bi / ÿ > À A œ v EN / Yer /
Q N Q N Q N a, \ \ \ oO
6 © . 6) N ole) ° LU505117 oy N20 à Oy NNO à 5 N“ a SON Î
N ©) 0 oN Ah o Meg 0 À de o IS / N (5 7 N / N
Ce A id dg N= N va A nA % “ge NA”
N = SS N = N = (NA NA 4 CN rN
N N N N
= J J — y — . A / 2
Jar) / ~ / dN ( od aN a} Da ww,
X HO HO—
Os GA 99 oy GA °e Oy GA 99 Os © 99
À o oly Js H H b sl À, © a oN, oO of À.
LAN ee à Head 4 0 hee 4 Ver (5) = = (5) N=" (5) N="
N X 5 N = N 5 N 5
NN 7 0 (44 NS
N
N N N
— # — A A / A / — dN 9 ay N Cw 2 VE ®) lo
OH wd OH —oH
Oy GA 99 Os GA ° 0 Oy GA 99 Our GA 99 o Hel Je d Fol Je o Hel le d FA de
Jd iv Jd Ne Jd NE d We © NA © N= © N > N= > >
AA NA > AA 7 > NS =
Bi > N
Ÿ FC
N ( 24 / Ow / dW / ’ od oN Ny © (8) . ° 1s) ° 1s) ° 1s) 4 rtf 4 rt 4 rt 4 WA 3 NA 3 NA 3 NA, 3 NA
NA 7 > NE 7 > "A > MN 7 > _— _— _— _—
HO.45) Ÿ HOW) Ÿ EF. 7 Fa® 7
DO Tn © +,
THs "Tes TCS TCS © (8), 8 à Hk U, y ° SNe) , D Ads qd, Ty 5 qd, ne N Oy wl #H vi de pl # 5
N LS % N 8 N 5 (NI NA À CN SN
N N N —
Va J / / oN N: N or lh So Oxy Go ° 0 ole © u o
Hl J N NN © SON © / o NN a ; o Hu ue (8) 6 He Jeg o Fe Le
Cy n=, N Ss H vo d N 5 d H wo
N 8 2 ha a NA 3 NA
N: =
C= PN a > Ne
A N N N
9 JO / > ÿ - Ou ÿ — Ÿ -
N N. — ©
BL-5758
GO LU505117 oko 0 oo o oko o / 2 He ea od wel," © 0 va © a wd YF =. =
NA € > PY $ > NS { = / > ÿ — Ÿ — ÿ — N
Ln L ON D oy ‘9 oy Po op lt) 99 oi lo 0 dr 9 "des d o Aer J o Hee J A de ai = a 7 I = NA >
N WV — . fl J . / À | J _ <q o i ol lo o oy ld °° ole) 0 61 0
TH OLA à Hel Je TNL TLS go wer dr UE J NRA < A
PN 7 xd > NS 7 NA au 9 — J 2 Sa
In yet another aspect, the present invention also provides a pharmaceutical composition, which includes any of the aforementioned compounds or pharmaceutically acceptable salts, isotopic derivatives, and stereoisomers thereof.
In one aspect, the present invention also provides use of the aforementioned compounds or pharmaceutically acceptable salts, isotopic derivatives, stereoisomers thereof and pharmaceutical compositions in the preparation of medicine for preventing and/or treating cancers, tumors, inflammatory diseases, autoimmune immune diseases or immune-mediated diseases.
It should be noted that, in this article, when referring to the "compound" of formula (D and formula (II), 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) and formula (II), generally, their pharmaceutically acceptable salts are also included, and furthermore, their solvates and hydrates are also included.
Similarly, when referring to a compound herein, its prodrugs, metabolites and 13
BL-5758 nitroxides are also generally included. LUS0S117
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 that the salt is precipitated from the resulting mixture, the solvent and remaining free 14
BL-5758 acid are removed therefrom, and the precipitated salt is isolated. LUS0S117
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
BL-5758 invention has an asymmetric center, and each asymmetric center can produce two LUSOS117 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 (IIT) 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 >I; nitrogen isotopes: ’N and
IN ; Oxygen isotopes: °0, 170 and '®O and sulfur isotope *°S. 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 (°H), 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 16
BL-5758 cancer, tumor, inflammatory disease, autoimmune disease, neurodegenerative disease, LU505117 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, 17
BL-5758 medulloblastoma and peripheral neuroectodermal tumor, Hodgkin lymphoma, non- LUSOS117
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, catumaxumab Antibodies, 18
BL-5758 cetuximab, denosumab, gemtuzumab, ipilimumab, nimotuzumab, ofatumumab, LU505117 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 (e.g., 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 acid, magnesium stearate, calcium stearate, gelatin, talc, surfactant, suspending agent, 19
BL-5758 emulsifier and diluent. LUS0S117
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.
Unless otherwise indicated, all chiral (enantiomers and diastereoisomers) and racemic
BL-5758 forms are within the scope of the invention. Various geometric isomers of C=C double LUSOS117 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.
Unless otherwise defined, the definitions of the substituents in the present invention are independent and not interrelated, for example (listed but not exhaustive), in one aspect, for the substituents Ra (or Ra") in different definitions of the substituents, 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, 21
BL-5758 cycloalkyl, aryl, heterocyclyl, halogen, hydroxy, alkoxy, oxo, alkanoyl, aryloxy, LUS0S117 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" or "alkylene" as used herein is intended to include both branched and straight chain saturated aliphatic hydrocarbon groups having the indicated 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, t-butyl), and pentyl (such as n-pentyl, isopentyl, neopentyl). Herein, the alkyl group is preferably an alkyl group having 1 to 6, more preferably 1 to 4 carbon atoms.
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 "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,-C¢ 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, Cz, C3, Ca, Cs, Ce alkoxy. Examples of alkoxy include, but are not limited to, methoxy, ethoxy, propoxy (e.g., 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 "thiothio" denotes an alkyl 22
BL-5758 group as defined above having the indicated number of carbon atoms attached through LUSOS117 a sulfur bridge; e.g. methyl-S- and ethyl-S-.
The term "carbonyl" refers to an organic functional group (C=O) 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 monocyclic, 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 invention, "aryl" refers to an aromatic ring system which includes, but is not limited to, phenyl, biphenyl, indanyl, 1-naphthyl, 2-naphthyl, and tetrahydronaphthyl. 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. A fused aryl group can be attached to another group at a suitable position on the cycloalkyl ring or aromatic ring.
Dashed lines drawn from ring systems indicate that bonds may be attached to any suitable ring atom.
The term "cycloalkyl" refers to monocyclic, bicyclic or branched cyclic alkyl groups. Monocyclic cycloalkyl refers to C3-C12 cycloalkyl, including but not limited to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and norbornyl. Branched cycloalkyl groups such as 1-methylcyclopropyl and 2-methylcyclopropyl are included within the definition of "cycloalkyl". Bicyclic cycloalkyl groups include bridged, spiro or fused cycloalkyl groups. Herein, the cycloalkyl group is preferably a C3-C12 cycloalkyl group, more preferably a C3-Cs cycloalkyl group.
Similarly, the term "heterocycloalkyl" refers to a 3-12 membered ring structure in which at least one carbon atom on a cycloalkyl group is replaced by a heteroatom selected from N, O, S and P, which may be a monocyclic , bicyclic or branched ring structures. Herein, the heterocycloalkyl group is preferably a 3-12 membered heterocycloalkyl group, more preferably a 3-8 membered heterocycloalkyl group.
The term "bridged cycloalkyl" as used herein refers to polycyclic compounds sharing two or more carbon atoms. “Bbridged cycloalkyl” can be divided into bicyclic bridged ring hydrocarbons and polycyclic bridged ring hydrocarbons. The former is composed of two alicyclic rings sharing more than two carbon atoms; the latter is a bridged ring hydrocarbon composed of more than three rings.
The term "spirocycloalkyl" as used herein refers to polycyclic hydrocarbons in which the monocyclic rings share one carbon atom (called the spiro atom). 23
BL-5758
The term "bridged heterocycle" as used herein refers to a polycyclic compound LUSOS117 sharing two or more carbon atoms, and the ring contains at least one heteroatom selected from O, N and S atoms. It can be divided into bicyclic bridged heterocycles and polycyclic bridged heterocycles.
The term "heterospirocyclyl" as used herein refers to a polycyclic hydrocarbon that shares one carbon atom (called a spiro atom) between monocyclic rings, and the ring contains at least one heteroatom selected from O, N and S atoms.
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).
The term "cycloalkenyl" refers to a monocyclic or bicyclic cyclic alkenyl group.
Monocyclic cycloalkenyl refers to C;-Cs 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 within the definition of "cycloalkenyl". Bicyclic cycloalkenyl groups include bridged, spiro, or fused cycloalkenyl groups. "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 C1, Ca, C3, Ca, Cs, Cs haloalkoxy. Examples of haloalkoxy include, but are not limited to, trifluoromethoxy, 2,2, 2-trifluoroethoxy, 24
BL-5758 and pentafluoroethoxy. Similarly, "haloalkylthio" or "thiohaloalkoxy" denotes a LUSOS117 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, 5, 6, 7 or 8 carbon atoms, C1-Cg means that the group contains 1, 2, 3, 4, 5, 6,7 or 8 carbon atoms, C,-Cg means that the group contains 2, 3, 4, 5, 6, 7 or 8 carbon atoms,
C3-Cs 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-C6 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.
In the present disclosure, the one or more halogens may each be independently
BL-5758 selected from fluorine, chlorine, bromine and iodine. LU505117
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 atoms and 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.
Preferably, when the total number of S and O atoms in the heterocycle exceeds 1, then 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. 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, 26
BL-5758 pyridoxazolyl, pyridimidazolyl, pyridothiazolyl, pyridyl , pyrimidinyl, pyrrolidinyl, ~~ LUVS05117 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 include biaryl structures formed by the above-defined "aryl" and a monocyclic "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.
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 other occurrence. Thus, for example, if a group is shown to be substituted with 0-3 R, 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 27
BL-5758 tissue, system, animal or human being sought, e.g, by a researcher or clinician. LUSOS117
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" 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.
The term "pharmaceutical composition" means a composition comprising a 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 28
BL-5758 active ingredient does not have an undue adverse effect on health for the general LUSOS117 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 include, but are not limited to, mammals: humans, non-human primates such as 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 29
BL-5758 symptom; causing a disease or a symptom to regress; alleviating a complication caused LUSOS117 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 therapeutically effective amount of one or more compounds of the present invention 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;
BL-5758 topically, such as in the form of a cream or ointment; or rectally, such as in the form of LUSOS117 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 PharmaCy1 (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.
The compounds are usually formulated with a suitable pharmaceutical diluent, 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 31
BL-5758 therapeutically effective amount of at least one compound of the present invention as LUSOS117 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 dosage will generally depend on the factors mentioned above. Typically, oral, 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). 32
BL-5758
Kit/Product Packaging LUS0S117
Kits/product packaging are also described herein for use in the treatment of the above indications. These kits may consist of transporters, packs, or container boxes, which may be divided into compartments to accommodate one or more types of containers, such as vials, test tubes, and the like, with each container containing a single component in the method described above. Suitable containers include bottles, vials, syringes, test tubes and the like. Containers are made of acceptable materials such as glass or plastic.
For example, a container may contain one or more compounds described herein, either as a pharmaceutical compound or in admixture with other ingredients described herein. The container can have a sterile outlet (e.g., the container can be an IV bag or bottle, the stopper of which can be pierced by a hypodermic needle). Such a kit may contain a compound, together with instructions for use, labels or instructions for the methods described herein.
À typical kit may include one or more containers, each containing one or more materials (such as reagents, concentrated stock solutions, and/or or equipment). These materials include, but are not limited to, buffers, diluents, filters, needles, syringes, transporters, bags, containers, bottles and/or test tubes, accompanied by a list of contents and/or instructions for use, and inner packaging also accompanied by instructions. Instructions for the entire set are to be included.
Labels can be displayed on or closely associated with the container. The appearance of the label on the container means that the label letters, numbers or other features are pasted, molded, or engraved on the container; the label can also appear in the container box or shipping box containing various containers, such as in the product insert. À label may be used to indicate a specific therapeutic use of the contents. The label may also bear instructions for use ofthe contents, such as described in the methods above.
All features described in this specification (including any stated claims, abstract and drawings), and/or all steps involved in any method or process, may exist in any combination, unless certain features or steps are mutually exclusive in the same combination.
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 33
BL-5758 each feature disclosed in the specification can be replaced by any alternative feature LUSOS117 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
When the preparation route is not included, the raw materials and reagents used in 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 34
BL-5758 nuclear magnetic resonance (NMR) and mass spectroscopy (MS). NMR shifts (6) are LUSOS117 given in units of 10% (ppm). The determination of NMR is to use (Bruker AscendTM 500 type) nuclear magnetic analyzer, the measurement solvent is deuterated dimethyl sulfoxide (DMSO-d6), deuterated chloroform (CDC13), 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 bicarbonate, mobile phase B is an acetonitrile solution, and perform linear gradient 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 pine Mobile Mobile phase A/% phase B/% 0 9 5 1.4 5 95 2.8 5 95 2.820 95 5 3 95 5
BL-5758
HPLC analysis method is as follows: LUS0S117
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 phase Mobile phase
Time/min
A/% B/% 0 95 5 5 95 5 95 20.2 95 5 24 95 5
The synthetic method of some intermediates in the invention is as follows:
Intermediate 1 /
AcO 2 c (S) ©
N= HN—Boc
O °°
BC
O N
H
Intermediate 1 was prepared by the following steps: 36
BL-5758 9 TBDPSCI 0 9 0 LU505117 imidazol . NaOH TBDPS,, SOC! TBDPS., 1007 MF == TERS > VeOHH0 0 on DOM. Xe
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
AIERCI N 2) diludine, TSOH,DCM = THF S DCM
DCM N 3)LIOH, MeOH, HO N N
INT-1f INT-1g INT-1h 0 INTAK 97 © bal o=( NS NS A Jd
B > 0 D ef a 0
Pd(dppf)Cly, AcOK © Boc P—Boc dioxane o-B Pd(dppf)Cl2, K,PO4 N=
Br A Ÿ dioxane/H20 J >
N N N
INT-1i INT-1j INT-11
Q Q
> d I d
Q LL Pinacolborane Q LE
NIS S-Phos, Pd,dba;, ACOK ovr N= HN-Boc — N= HN—Boc — I 7
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 step without purification. ESI-MS (m/z): 371.2 [M+H]".
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 37
BL-5758 30 minutes, filter with suction, and dry to obtain white solid INT-1¢ (269 g, yield 90%). LUSOS117
ESI-MS (m/z): 357.8 [M+H]".
Step 3: INT-1¢ (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 © 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 washed with water, dried, concentrated under reduced pressure, and the residue was 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 38
BL-5758 advance was added, and the solution was stirred at room temperature for 3 hours, and LUSOS117 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 (IM 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 monitored the complete reaction of the raw materials, and 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 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 39
BL-5758 g, 103.7 mmol) in dioxane (300 mL) and water (30 mL), add potassium phosphate ( 50 LUS0S117 g, 235.7 mmol) and [1,1'-bis(diphenylphosphino)ferrocene]palladium dichloride (6.89 g, 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-Im (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 50 °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]".
BL-5758
Intermediate 2 LUS0S117
O
Nols)
HN—N ° s/ ©
N= HN-Boc
O =
B—
O N
H
Intermediate 2 was prepared by the following steps: 0 / INT-2b Sud N > Fa 8 Pe Su @ HO J HIN o © HN-NHTFA (s) ;
LiOH*H,O N= HN—Boc \ LIOH'H20
N= NB To 5 TCFH, NMI RS THF/H20
J = +7 ACN } NP
N
INT-1m INT-2a INT-2c
HO HO HN-N HNN Oo Pinacolborane / HNON o
A © ohare o A S-Phos, Pd2dba3, AcOK o A
Jone FIN Boe DCM rh HN—Boc THF © fos HN-—Boc 0 0 x 8
N H ° À
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 41
BL-5758 reaction of the raw materials was monitored by LCMS to be complete. The reaction LUSOS117 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-2c (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]". 42
BL-5758
LU505117
Intermediate 3
O
“ods )
HN—N 6 (S) >
N= HN s “Le x To
BY
OÖ N
H
Intermediate 3 was prepared by the following steps:
INT-3b
HN-N HN-N Ho HN-N
V4 0 oO Ne o
Jd s. TFA Jd À > o X 9
N= HN—Boc DCM N= Nz TCFH,NMI N= WL 7 = 0 = ACN 4 pa
INT-2e INT-3a INT-3c
HN=N
Pinacolborane ©), O
S-Phos, Pd2dba3, AcOK © ea L
THF VA, Mg
À te S > 3 J 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 NaHCO; 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) NNNN "- tetramethylchloroformamidine hexafluorophosphate (921.9 mg, 3.29 mmol), and 1- 43
BL-5758 methylimidazole (414 mg, 5.04 mmol) were added to react at 0 °C for 1 hour, LCMS LUSOS117 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
“ods )
HN—N 6 (S) >
NX HN
° 8 Le
BL (R)
O N
H
Intermediate 4 was prepared by the following steps:
INT4a oO 0 oO oO / su or a 764 Pinacolborane Jd o A © Jo o A 6 SPhos, Pdzdba3, AcOK oO A ©
N=, Ni Ton N= WN THF N= WNL 4 = ACN dû = fes nu, } = Ja
H N oN
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'- 44
BL-5758 tetramethylchloroformamidine hexafluorophosphate (1.56 g, 5.56 mmol), and 1- LUS0S117 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 S or
NS
S
O N
7) ZZ Br
Intermediate 5 was prepared by the following steps:
INT-5b —0 oO re gE Fr
N - =
VS Br Cul, TEA, Pd(PPhy);Cl, OT, A — LA Br
THF, rt, 3h UN LA
INT-5a INT-5
Step 1: Dissolve (S)-3-bromo-5-iodo-2-(1-methoxyethyl)pyridine INT-Sa (2.0 g, 5.85 mmol) in tetrahydrofuran (20 mL), and add cuprous iodide (111.4 mg, 0.585 mmol), bistriphenylphosphinepalladium dichloride (4105 mg, 0.585 mmol), triethylamine (1.18 g, 11.7 mmol), and 4-propyne-1-morpholine INT -Sb (878.5 mg,
BL-5758 7.02 mmol). The reaction mixture was stirred at room temperature for 3 hours under LUSOS117 nitrogen protection. After the reaction was complete, the reaction solution was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (dichloromethane/ethyl acetate = 1/1) to obtain light yellow oily compound INT-5 (1.8 g, yield 90.7%). ESI-MS (m/z): 339.4 [M+H]".
Intermediate 6 or
N P ei FZ S Br
Intermediate 6 was prepared by the following steps:
INT-5a al
DMF, . Ce GANA N
INT-6a INT-6b INT-6
Step 1: (S)-2-methylmorpholine hydrochloride INT-6a (1.00 g, 9.27 mmol), 3- bromopropyne (1.04 g, 8.72 mmol), potassium carbonate (3.01 g, 21.80 mmol ) were dissolved in N,N-dimethylformamide (15 mL), and the reaction solution was stirred overnight at room temperature. After the reaction was complete, the reaction solution was extracted with ethyl acetate, and the organic phase was dried and concentrated to obtain a crude product. The crude product was purified by column chromatography (petroleum ether/ethyl acetate = 1/1) to obtain light yellow oily compound INT-6b (450 mg, yield 44.5%). ESI-MS (m/z): 140.3 [M+H]".
Step 2: INT-6b (268.6 mg, 1.93 mmol), INT-5a (600 mg, 1.75 mmol), cuprous iodide (334 mg, 0.18 mmol), triethylamine (355.1 mg, 3.51 mmol), bistriphenylpalladium dichloride (123.0 mg, 0.18 mmol) were dissolved in tetrahydrofuran (10 mL), and the reaction solution was stirred overnight at room temperature. After the reaction was complete, the reaction solution was filtered, and the filtrate was concentrated to obtain a crude product. The crude product was purified by 46
BL-5758 column chromatography (petroleum ether/ethyl acetate = 1/1) to obtain light yellow LUSOS117 oily compound INT-6 (360 mg, yield 58.10%). ESI-MS (m/z): 353.5 [M+H]".
Intermediate 7 or
NS
S
N
ZZ Br
ON
EU
Intermediate 7 was prepared by the following steps: ° INT-Sa
A0
XL p 400008 © La,
INT K2CO3 N Cul, TEA, Pd(PPhz)zCl> ZZ Np © MeCN, 90°C, 16h C J THE, rt, 3h (ON oO 0”
INT-7a INT-7b INT-7
Step 1: Compound INT-7a (1 g, 4.46 mmol) was dissolved in acetonitrile (10 mL), and morpholine (1.16 g, 13.3 mmol) and potassium carbonate (1.23 g, 8.92 mmol) were added in sequence. The reaction mixture was stirred at 90 °C for 16 hours under nitrogen protection. After the reaction was complete, the reaction solution was filtered with diatomaceous earth, the filtrate was washed twice with ammonium chloride aqueous solution and then concentrated to obtain a yellow oily compound INT-7b (450 mg, yield 72.6%). ESI-MS (m/z): 140.1 [M+H]".
Step 2: Dissolve (S)-3-bromo-5-iodo-2-(1-methoxyethyl)pyridine INT-5a (920 mg, 2.69 mmol) in tetrahydrofuran (20 mL), and add cuprous iodide (51 mg, 0.27 mmol), bistriphenylphosphine palladium dichloride (188 mg, 0.27 mmol), triethylamine (540 mg, 5.34 mmol) and INT-7b (450 mg, 3.24 mmol) . The reaction mixture was stirred at room temperature for 3 hours under nitrogen protection. After the reaction was complete, the reaction solution was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (dichloromethane/ethyl acetate = 1/1) to obtain light yellow oily compound INT-7 (850 mg, yield 89.2%). ESI-MS (m/z): 353.6 [M+H]". 47
BL-5758
LU505117
Intermediate 8
Jd (S)
N
: —N (S) 9 \ / ;
Intermediate 8 was prepared by the following steps: —O 1 b / N Br
O OH Dess-martin O = onMe O zZ — ( (R) periodinane C (R) O N» C (S) | INT-5a
N DCM, rt N K,CO3 N Cul, Pd(PPhs),Cl,, TEA
Boc Boc MeOH, rt Boc THF, rt
INT-8a INT-8b INT-8c —0 _o —Q
N N
\ N / N a. TFA / N—gr HCHO, NaBH(OAc)s _ Br
DCM, rt — DCE, rt 0 © INT-8d 05) INT-8e © INT-8
N Ca N
Boc
Step 1: Add Dess Martin reagent (5.08 g, 11.97 mmol) to a solution of INT-8a (2.0 g, 9.21 mmol) in dichloromethane (40 mL) at 0 °C. After the reaction solution was warmed to room temperature, stirring was continued for 5 hours. After the reaction was complete, saturated aqueous sodium bicarbonate and sodium thiosulfate were added to quench the reaction, the reaction solution was extracted with dichloromethane, the organic phases were combined and washed with saturated brine, dried over anhydrous sodium sulfate, and the organic phase was concentrated to obtain the crude product
INT-8b. 48
BL-5758
Step 2: The above crude product INT-8b and potassium carbonate (3.81 g, 27.60 12505117 mmol) were added to methanol (40 mL), and (1-diazo-2-oxopropyl) dimethyl phosphonate (3.53 g, 18.40 mmol) was added dropwise thereto at 0 °C. After the reaction solution was stirred at room temperature for 4 hours, the reaction was monitored by TLC to be complete. The reaction was quenched by adding water, the reaction solution was extracted with ethyl acetate, and the combined organic phases were washed with saturated brine, dried over anhydrous sodium sulfate, then the organic phase was concentrated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 6:1) to obtain colorless transparent solid INT-8¢ (1.20 g, two-step yield 62.0%). ESI-MS (m/z): 212.3 [M+H]".
Step 3: Under nitrogen atmosphere, add (S)-3-bromo-5-iodo-2-(1- methoxyethyl)pyridine INT-Sa (300 mg, 0.88 mmol) and cuprous iodide (17 mg, 0.088 mmol), bistriphenylphosphine palladium dichloride (62 mg, 0.088 mmol), triethylamine (178 mg, 1.75 mmol), compound INT-8¢ (222 mg, 1.05 mmol) and tetrahydrofuran ( 10 mL) successively into the reaction flask. After the reaction solution was stirred at room temperature for 4 hours, the reaction was monitored by LCMS to be complete. The reaction solution was concentrated under reduced pressure and purified by silica gel column chromatography (petroleum ether/ethyl acetate=3/1) to obtain a pale yellow oily liquid Int-8d. ESI-MS (m/z): 425.7 [M+H]".
Step 4: The above product Int-8d was dissolved in dichloromethane (6 mL), and trifluoroacetic acid (2 mL) was added dropwise thereto. After the reaction solution was stirred at room temperature for 30 minutes, the reaction was monitored by LCMS to be complete. The reaction solution was concentrated under reduced pressure to obtain compound Int-8e. ESI-MS (m/z): 325.5 [M+H]".
Step 5: Dissolve the above product Int-8e in 1,2-dichloroethane (10 mL), add formaldehyde aqueous solution (213 mg, 2.63 mmol, 37%) dropwise thereto, react for minutes, then add triacetyl sodium oxyborohydride (1.11 g, 5.26 mmol). The reaction solution was stirred at room temperature for 30 minutes, and LCMS monitored the completion of the reaction. The reaction was quenched by adding saturated aqueous 49
BL-5758 sodium bicarbonate solution, the reaction solution was extracted with dichloromethane LU505117 and methanol, the organic phases were combined and washed with saturated brine, dried over anhydrous sodium sulfate, then the organic phase was concentrated and purified by silica gel column chromatography (dichloromethane:methanol=10: 1) to obtain Int-8 (269 mg, 90.5% yield in three steps) as light yellow oily liquid. ESI-MS (m/z): 339.6 [M+H]".
Intermediate 9
Oo”
NS
4 )
Intermediate 9 was prepared by the following steps:
NS
DMF, rt THF
INT-9a INT-9b INT-9
Step 1: cis-2,6-dimethylmorpholine INT-9a (1.06 g, 9.20 mmol), 3-bromopropyne (1.09 g, 9.20 mmol), cesium carbonate (3.82 g, 27.6 mmol) were dissolved in N, N- dimethylformamide (10 mL), and the reaction solution was stirred at room temperature overnight. After the reaction was complete, the reaction solution was extracted with ethyl acetate, and the organic phase was dried and concentrated to obtain a crude product. The crude product was purified by column chromatography (petroleum ether/ethyl acetate = 1/1) to obtain light yellow oily compound INT-9b (600 mg, yield 42.6%). ESI-MS (m/z): 154.2 [M+H]".
Step 2: INT-9b (295.7 mg, 1.93 mmol), INT-5a (600 mg, 1.75 mmol), cuprous iodide (334 mg, 0.18 mmol), triethylamine (355.1 mg, 3.51 mmol), bistriphenylpalladium dichloride (123.0 mg, 0.18 mmol) were dissolved in tetrahydrofuran (10 mL), and the reaction solution was stirred overnight at room temperature. After the reaction was complete, the reaction solution was filtered, and the filtrate was concentrated and spin-dried to obtain a crude product. The crude product
BL-5758 _ : LU505117 was purified by column chromatography (petroleum ether/ethyl acetate=1/1) to obtain light yellow oily compound INT-9 (600 mg, yield 93.1%). ESI-MS (m/z): 367.3 [M+H].
Intermediate 10
So
NS
ZZ > Br
ON en
Intermediate 10 was prepared by the following steps:
Oo oO “© oO a 2 a Lk ! "OMe LA or A MS oP Lira Ca
SON KoCO, PE Cul, TEA, Pd(PPha)Cla NAB
O MeOH, rt Oo THF | NZNO
INT-10a INT-10b INT-10c
NH NT
HCI-dioxane Le HCHO, NaBH3CN Le
DCM = x Br MeOH, rt S Br
N° S) Oo | NZ (S) Oo
INT-10d INT-10
Step 1: (R)-3-formylmorpholine-4-carboxylate tert-butyl ester INT-10a (4.06 g, 18.9 mmol) and (1-diazo-2-oxopropyl) dimethyl phosphonate (5.44 g, 28.3 mmol) were dissolved in methanol (30 mL), and potassium carbonate was added to the above reaction solution at room temperature. The reaction solution was stirred overnight at room temperature. After the reaction was complete, the reaction solution was extracted with ethyl acetate, and the organic phase was dried and concentrated to obtain a crude product. The crude product was purified by column chromatography (petroleum ether/ethyl acetate = 3/1) to obtain light yellow oily compound INT-10b (3.2 g, yield 80.3%). ESI-MS (m/z): 212.4 [M+H]".
Step 2: INT-10b (7104 mg, 3.36 mmol), INT-5a (1.00 g, 2.92 mmol), triethylamine (591.8 mg, 5.58 mmol), cuprous iodide (55.7 mg, 0.29 mmol), 51
BL-5758 bistriphenylpalladium dichloride (205.3 mg, 0.29 mmol) were dissolved in LUS0S117 tetrahydrofuran (10 mL), and the reaction solution was stirred overnight at room temperature. After the reaction was complete, the reaction solution was filtered, and the filtrate was concentrated to obtain a crude product. The crude product was purified by column chromatography (dichloromethane/methanol = 10/1) to obtain light yellow solid compound INT-10c (1.05 g, yield 84.4%). ESI-MS (m/z): 425.5 [M+H]".
Step 3: Dissolve INT-10c (1.20 g, 2.82 mmol) in dichloromethane (15 mL), and add hydrochloric acid-dioxane (7.05 mL, 4M) to the above reaction solution. The reaction was stirred at room temperature for 4 hours. After the reaction was complete, a saturated aqueous sodium bicarbonate solution was added to the reaction solution.
The organic phases were combined, dried and concentrated to obtain light yellow solid compound INT-10d (917.5 mg, yield 100.00%). ESI-MS (m/z): 325.5 [M+H]".
Step 4: INT-10d (917.06 mg, 2.82 mmol), aqueous formaldehyde (288.2 mg, 8.46 mmol), and sodium cyanoborohydride (531.6 mg, 8.46 mmol) were dissolved in methanol (10 mL), and the reaction solution was stirred overnight at room temperature.
After the reaction was complete, a saturated aqueous sodium bicarbonate solution was added to the reaction solution, followed by extraction with dichloromethane. The combined organic phases were dried and concentrated to obtain a crude product, which was purified by column chromatography (dichloromethane/methanol = 10/1) to obtain light yellow oily compound INT-10 (650 mg, yield 68.0%). ESI-MS (m/z): 339.2 [M+H]".
Intermediate 11 —O (S)
A
©
N
Intermediate 11 was prepared by the following steps: 52
BL-5758 —0 LU505117 oo N
Il 7 \ Br
P.
Cl “OH periodinane. Cl So ac Gl INT-Sa
N DCM, rt N KaCO3 N Cul, Pd(PPha)2Cl2, TEA
Boc Boc MeOH, rt Boc THF, rt
INT-11a INT-11b INT-11¢ 79 —0 —Q
N
7 \ Br N \ Br a \ Br — TFA _ HCHO, NaBH(OAC) 3 —
Ÿ DCM, rt J DCE, rt J 0X os os
N (R) (R)
Cx INT-11d wi INT-11e CY INT-11
Boc \
Step 1: Add Dess Martin reagent (5.08 g, 11.97 mmol) to a solution of INT-11a (2.0 g, 9.21 mmol) in dichloromethane (40 mL) at 0 °C. After the reaction solution was warmed to room temperature, stirring was continued for 5 hours. After the reaction was complete, saturated aqueous sodium bicarbonate and sodium thiosulfate were added to quench the reaction. The reaction solution was extracted with dichloromethane, and the organic phases were combined and washed with saturated brine, dried over anhydrous sodium sulfate, and the organic phase was concentrated to obtain the crude product
INT-11b.
Step 2: The above crude product INT-11b and potassium carbonate (3.81 g, 27.60 mmol) were added to methanol (40 mL), and (1-diazo-2-oxo Dimethyl phosphonate (3.53 g, 18.40 mmol) was added thereto at 0 °C. After the reaction solution was stirred at room temperature for 16 hours, the reaction was monitored by TLC to be complete.
The reaction was quenched by adding water, the reaction solution was 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 and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 6/1) to obtain colorless transparent solid INT-11¢ (1.38 g, 70.8% yield in two steps). ESI-MS (m/z): 212.2 [M+H]". 53
BL-5758
Step 3: Under nitrogen atmosphere, add (S)-3-bromo-5-iodo-2-(1- LUSOS117 methoxyethyl)pyridine INT-Sa (500 mg, 1.46 mmol) and cuprous iodide (28 mg, 0.146 mmol), bistriphenylphosphine palladium dichloride (103 mg, 0.146 mmol), triethylamine (296 mg, 2.92 mmol), compound INT-11¢ (402 mg, 1.90 mmol) and tetrahydrofuran (10 mL) successively into the reaction flask. After the reaction solution was stirred at room temperature for 16 hours, the reaction was monitored by LCMS to be complete. The reaction solution was concentrated under reduced pressure and purified by silica gel column chromatography (petroleum ether/ethyl acetate=3/1) to obtain a pale yellow oily liquid Int-11d. ESI-MS (m/z): 425.6 [M+H]".
Step 4: The above product Int-11d was dissolved in dichloromethane (9 mL), and trifluoroacetic acid (3 mL) was added dropwise thereto. After the reaction solution was stirred at room temperature for 30 minutes, the reaction was monitored by LCMS to complete. The reaction solution was concentrated under reduced pressure to obtain compound Int-11e. ESI-MS (m/z): 325.3 [M+H]".
Step 5: Dissolve the above product Int-11e in 1,2-dichloroethane (10 mL), add formaldehyde aqueous solution (356 mg, 4.38 mmol, 37%) dropwise thereto, react for minutes, then add triacetyl sodium oxyborohydride (1.86 g, 8.76 mmol). The reaction solution was stirred at room temperature for 30 minutes, and LCMS monitored the completion of the reaction. The reaction was quenched by adding saturated aqueous sodium bicarbonate solution, the reaction solution was extracted with dichloromethane and methanol, the organic phases were combined and washed with saturated brine, dried over anhydrous sodium sulfate, and the organic phase was concentrated and purified by silica gel column chromatography (dichloromethane/methanol=10/ 1) to obtain Int-11 (481 mg, three-step yield 97.1%) as light yellow oily liquid. ESI-MS (m/z): 339.4 [M+H]".
Intermediate 12 >o
NS
S
A
Boe” 54
BL-5758
LU505117
Intermediate 12 was prepared by the following steps: —0 (S) —0
N (S)
Dai LÀ 7 | INT-5a _/ Br
Boo” Cul, Pd(PPh3),Cl, Ÿ
INT-12a TEA, THF, it
Bod INT-12
Compound INT-12 can be obtained by replacing INT-Sb in intermediate 5 with
INT-12a and using similar methods and reaction steps. ESI-MS (m/z): 395.5 [M+H]".
Intermediate 13
So
LE
Boc > Br pn?
Intermediate 13 was prepared by the following steps: 9 8
Ar Tome
Dess-marti
Boe. =, OH periodinane Boe, 30 No) © Boc, Zz = DCM, rt L] K,CO4, MeOH, rt Ye
INT-13a INT-13b INT-13c —0 —0 = PN 1 INT-5a _/ Br
Cul, Pd(PPha)2Cl2
TEA, THF, rt Boc 7
NP
INT-13
Compound INT-13 can be obtained by replacing INT-8a in intermediate 8 with
INT-13a and using similar methods and reaction steps. ESI-MS (m/z): 395.5 [M+H]".
Intermediate 14
BL-5758
So LU505117
NS
UN
B es, ® ZZ Br
Intermediate 14 was prepared by the following steps: 7 9%
Ar ome
Dess-marti
Boc, OH periodinane Boc, =0 NZ) © Boc, 7
DCM, tt K2COs, MeOH, rt
INT-14a INT-14b INT-14c —o
N —o a=" = A ’ INT-5a _/ Br
Cul, Pd(PPhs)2Cl2 J
TEA, THF, rt Boo “NA
INT-14
Compound INT-14 can be obtained by replacing INT-8a in intermediate 8 with
INT-14a and using similar methods and reaction steps. ESI-MS (m/z): 395.5 [M+H]".
Intermediate 15
O0 (S)
N \ —_— Br
Boc, 4
N (R)
Intermediate 15 was prepared by the following steps: 56
BL-5758
Lo LU505117
Q 0 (S) _ +O Np Ne 9 N — A
Boc, _\ N Boc Ÿ | INT- 5a Br
WW AN {09010
K,CO4 PdCl-(PPha)>, Cul, TEA Boc À
MeOH, rt THF, rt N (R) - INT- 15b
INT- 15a INT- 15
Step 1: Dissolve compound INT-15a (600 mg, 3 mmol) in methanol (5 mL), add potassium carbonate (1.25 g 9 mmol) and (1-diazo-2-oxopropyl) dimethyl phosphonate (1.16 g, 6 mmol) under room temperature. The reaction solution was stirred at room temperature for 12 h. TLC detected that the reaction was complete.
Saturated brine was added to the reaction system, and the reaction solution was extracted with dichloromethane, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to obtain the crude product of compound INT-15b (587 mg, yield 99%).
Step 2: Dissolve compound INT-15b (570 mg, 2.92 mmol) and compound INT-
Sa (1 g, 2.92 mmol) in tetrahydrofuran (8 mL), add bistriphenylphosphine palladium dichloride (204 mg, 0.29 mmol), cuprous iodide (56 mg, 0.29 mmol) and triethylamine (591 mg, 5.85 mmol). The reaction system was stirred at room temperature for 8 h after nitrogen replacement. LCMS detected that the reaction was complete. Saturated brine was added to the reaction system, and the reaction solution was extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated by filtration, and the residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=5/ 1) to obtain INT- (1.08 g, yield 90%) as a yellow oily liquid. ESI-MS (m/z): 409.6 [M+H]".
Intermediate 16
O0 (S)
N \ —_— Br
Boc, J 57
BL-5758
Intermediate 16 was prepared by the following steps: LUS0S117 —Q
Q 0 (S) —
Ar No” NX (s)
NON Br N
O Il — 4
Bog, (S) À N Boc, 8) 7 | INT- 5a VS Br
CO K,CO5 OÙ PdCI(PPha), Cul, TEA Bo A
MeOH, rt THF, rt N (SS
INT- 16a INT- 16b J INT- 16
The INT-15a in the intermediate INT-15 was replaced by INT-16a, and the compound INT-16 can be obtained by a similar method and reaction steps. ESI-MS (m/z): 409.5 [M+H]".
Intermediate 17 —O (S)
N
Boc”N
Intermediate 17 was prepared by the following steps:
Oo © 2
Il (S) —0o
Ar Ne NX (S) +O Br N 0 N — 7 \
WY af wre | Br
K,CO PdCIz(PPhs)z, Cul, TEA Ÿ soc) MeOH, rt soc ; THE À 0
INT- 17a INT- 17b soc) INT- 17
The INT-15a in the intermediate INT-15 was replaced by INT-17a, and the compound INT-17 can be obtained by a similar method and reaction steps. ESI-MS (m/z): 409.3 [M+H]".
Intermediate 18 58
BL-5758 or LU505117
N. (9)
SNS a” ‘
Intermediate 18 was prepared by the following steps: —0, © _
N 0 / pr NS — _ or
X_o_o | INT-6a Le 4 Aer
UV Cul, TEA, Pd(PPhs),Cl, Oi
INT-18a THF, 13h INT-18
The INT-5b in the intermediate INT-5 was replaced by INT-18a, and the compound INT-18 can be obtained by a similar method and reaction steps. ESI-MS (m/z): 354.3 [M+H]".
Intermediate 19 ex 9 6 He (s de N H Vo
S
NI S
— N
J —
MsO
Intermediate 19 was prepared by the following steps: 59
BL-5758 07 LU505117 0 € | ey 0 oO u. N o. 0. Æ Ng / O her
HN-N ©
Je D we | 2 N casouen
N= HN Pd(dppf)Cl2, KsPO4 rN DMF t ° ~° Ze dioxane/H2O, 70°C, 16h — N x =~ / oO N
INT-3 INT-19a eo °9 Q / O N Le Og NP à % N== H © / O He ve 44 = _Ts0H _ As A SC DIEA_ — N MeOH, rt } \ / > DCM
Jy — =,
Oo P A (5 INT-19b HO INT-19¢ ole Oo o Hel Je < Ne (S) N=
N >
UN ZZ
— N yj —
MsO
INT-19
Step 1: Dissolve compound INT-3 (300 mg, 0.44 mmol) in a mixed solution of 1,4-dioxane (5 mL) and water (1 mL), and add INT-18 (170 mg, 0.48 mmol), [1,1'- bis(diphenylphosphino)ferrocene]palladium dichloride (32 mg, 0.04 mmol) and potassium phosphate (188 mg, 0.88 mmol) successively. The reaction mixture was stirred at 70 °C for 16 hours under nitrogen protection. After the reaction was complete, the reaction solution was filtered with celite, and the concentrated residue was purified by silica gel column chromatography (dichloromethane/methanol = 20/1) to obtain light yellow oily compound INT-19a (260 mg, yield 71.2%) . ESI-MS (m/z): 823.1 [M+H]".
Step 2: Dissolve compound INT-19a (260 mg, 031 mmol) in N,N- dimethylformamide (4 mL), add cesium carbonate (205 mg, 0.63 mmol) and ethyl iodide ( 145 mg, 0.93 mmol). The reaction mixture was stirred at room temperature for 16 hours. After the reaction was complete, water (20 mL) was added to the reaction system, the reaction solution was extracted with ethyl acetate (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 INT-19b (110 60
BL-5758 mg, yield 41.0%). ESI-MS (m/z): 851.2 [M+H]". LUS05117
Step 3: Compound INT-19b (110 mg, 0.13 mmol) was dissolved in methanol (3 mL), and p-toluenesulfonic acid monohydrate (123 mg, 0.65 mmol) was added thereto.
The reaction mixture was stirred at room temperature for 3 hours. After the reaction was complete, water (30 mL) was added to the reaction system, the reaction solution was extracted with ethyl acetate (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 INT-19¢ (80 mg, yield 80.8%). ESI-MS (m/z): 767.5 [M+H]".
Step 4: Dissolve compound INT-19¢ (80 mg, 0.10 mmol) in dichloromethane (4 mL), add methanesulfonic anhydride (54 mg, 0.31 mmol) and diisopropylethylamine (68 mg, 0.53 mmol). The reaction mixture was stirred at room temperature for 2 hours.
After the reaction was complete, dichloromethane (30 mL) was added to the reaction system, the reaction solution was washed with water (15 mL*2) and saturated brine, the organic phase was dried over anhydrous sodium sulfate, filtered and concentrated to obtain light yellow solid compound INT-19 (70 mg, yield 79.5%). ESI-MS (m/z): 845.5 [M+H]".
Intermediate 20
Jd (S)
N
7 \ er
Wa
Intermediate 20 was prepared by the following steps: 61
BL-5758 0 LU505117 (S)
N or
I / À Br NS
INT-5a «
NS Y— ZZ Br
Cul, TEA, Pd(PPh3),Cl, aN
THF, rt, 3 h | _N
INT-20a
INT-20
The INT-5b in the intermediate INT-5 was replaced by INT-20a, and the compound INT-20 can be obtained by a similar method and reaction steps. ESI-MS (m/z): 317.3 [M+H]".
Intermediate 21 /
Os)
N
7 / \ N
Intermediate 21 was prepared by the following steps: —0
N ~~
Il an N 2
INT-5a e
S _ ZZ Ser
ZN Cul, TEA, Pd(PPhz)2Cl S
THF, rt, 3h »
INT-21a N
INT-21
The compound INT-21 can be obtained by using INT-21a to replace INT-5b in the intermediate INT-5, and using similar methods and reaction steps. ESI-MS (m/z): 317.3 [M+H]".
The synthetic method of embodiment compound among the present invention is as follows: 62
BL-5758
LU505117
Example 1 (18,2S)-N-((63S,4S,Z.)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(3- morpholinoprop-1-yn-1-yl )pyridin-3-yl)-10,10-dimethyl-S,7-dioxo- 61,62,63,64,65,66-hexahydro-11H-8-oxa-2(4,2)-thiazola-1 (5,3)-indola-6(1,3)- pyridazinacyclonecaphane-4-yl)-2-methylcyclopropane-1-carboxamide 6) os NN O o o Tel Js d NINE (S) N=
S
N UN
IN
— N ya
O N
\/
Example 1 is prepared by the following steps: e 0 0
Il "ar "a / (5) )~=NHBoc (5) =NHBoc
J N o orc ng OH Ag
AcO © oO $, INT4 — —
N fisc — 59 —OH , OS
Ts PA(dppf)Cl 2, K,CO; | THF/H20, rt
X Ÿ > doxaneH,0, 80°C NTN NTN oO N = =
INT-1 | 1a il 1b oO oO oO od poe ° fn oes o= = =
OH ng oHOH Ds ° Ns
Os ER Ne N EDCI, HOBt, | a oT INT2b d LioH 4 DMAP, DEA _ Og
EDCI, HOB, DIEA (9) | THF/H20, rt (8 | DCM, rt
DCM, rt N SAN NSN NS l H | > H =
I
63
BL-5758 0 0 LU505117
Ca (s)=NHBoc (on (s)}—=NH2 ©) 9 9 o WT-3b Oy N° °3 7 7 Le) 0 LS)
Cs,CO;, Etl | NF TEA | > vo du A VO
DMF, ıt 0 pm © T COMU,DIEA N =S
DMF rN
NS N N > N = N
J J / J
SN
Il Il UN 1 1f 1g + 1 N 1e a A A
NA > # 7
X) ,
Step 1: Dissolve compound INT-4 (600 mg, 1.77 mmol) in a mixed solution of 1,4-dioxane (10 mL) and water (1 mL), and successively add INT-1 (1.16 g, 1.77 mmol), [1,1'-bis(diphenylphosphino)ferrocene]palladium dichloride (129.3 mg, 0.177 mmol) and potassium carbonate (1.18 g, 4.42 mmol). The reaction mixture was stirred at 80 °C for 16 hours under the protection of nitrogen. After the reaction was complete, the reaction solution was filtered with celite and concentrated to obtain the black crude compound 1a (1.39 g, yield 100%), which was directly used in the next reaction without purification. ESI-MS (m/z): 788.1 [M+H]".
Step 2: the crude compound 1a (1.39 g, 1.77 mmol) was dissolved in a mixed solution of tetrahydrofuran (3 mL) and water (3 mL), and lithium hydroxide (211.2 mg, 8.82 mmol) was added thereto. The reaction mixture was stirred at room temperature for 3 hours. After the reaction was complete, adjust the pH of the reaction solution to 6 with 6N hydrochloric acid solution, the reaction solution was extracted with ethyl acetate (40 mL*2), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated by filtration. The residue was purified by silica gel column chromatography (dichloromethane/methanol = 30/1) to obtain compound 1b as a pale yellow solid (1.0 g, yield 77.3%). ESI-MS (m/z): 732.1 [M+H].
Step 3: Compound 1b (1.0 g, 1.37 mmol) was dissolved in dichloromethane (20 mL), and INT-2b (1.01 g, 2.73 mmol, TFA salt), 1-(3-dimethylamino propyl)-3- ethylcarbodiimide hydrochloride (523.9 mg, 2.73 mmol), 1-hydroxybenzotriazole 64
BL-5758 (369.2 mg, 2.73 mmol) and N,N diisopropylethylamine (882.9 mg , 6.83 mmol) were LUS0S117 added thereto. The reaction mixture was stirred at room temperature for 16 hours. After the reaction was complete, water (50 mL) was added to the reaction system, the reaction system was extracted with dichloromethane (50 mL*2), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated by filtration, and the residue was purified by silica gel column chromatography (dichloromethane/methanol = 30/1) to obtain compound 1c (700 mg, yield 59.7%) as a pale yellow solid. ESI-MS (m/z): 858.2 [M+H]".
Step 4: Compound 1c (700 mg, 0.815 mmol) was dissolved in a mixed solution of tetrahydrofuran (3 mL) and water (3 mL), and lithium hydroxide (195.4 mg, 8.16 mmol) was added thereto. The reaction mixture was stirred at room temperature for 4 hours.
After the reaction was complete, adjust the pH of the reaction solution to 6 with 6N hydrochloric acid solution. The reaction solution was extracted with ethyl acetate (40mL*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 1d (640 mg, yield 93.0%). ESI-MS (m/z): 844.2 [M+H]".
Step 5: Dissolve compound 1d (370 mg, 0.44 mmol) in dichloromethane (8 mL), add thereto 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (840.4 mg, 44 mmol), 1-hydroxybenzotriazole (2962 mg, 22 mmol), 4- dimethylaminopyridine (267.8 mg, 2.2 mmol) and N,N diisopropylethylamine (849.8 mg , 6.6 mmol). The reaction mixture was stirred at room temperature for 16 hours.
After the reaction was complete, water (30 mL) was added to the reaction system. The reaction solution was extracted with dichloromethane (30 mL*2), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated by filtration, and the residue was purified by silica gel column chromatography (dichloromethane/methanol = 30/1) to obtain compound 1e as a pale yellow solid (120 mg, yield 33.1%). ESI-MS (m/z): 826.2 [M+H]".
Step 6: Dissolve compound 1e (150 mg, 0.18 mmol) in N,N-dimethylformamide (3 mL), add cesium carbonate (118.3 mg, 0.36 mmol) and iodoethane (42.5 mg , 0.27 mmol). The reaction mixture was stirred at room temperature for 16 hours. After the reaction was complete, water (30 mL) was added to the reaction system, extracted with 65
BL-5758 ethyl acetate (30 mL*2), the organic phases were combined, washed with saturated LUSOS117 brine, dried over anhydrous sodium sulfate, filtered and concentrated to obtain a light yellow solid compound 1f (150 mg, yield 96.7%). ESI-MS (m/z): 854.2 [M+H]".
Step 7: Compound 1f (120 mg, 0.14 mmol) was dissolved in dichloromethane (2 mL), and trifluoroacetic acid (0.5 mL) was added thereto. The reaction mixture was stirred at room temperature for 2 hours. After the reaction was complete, add saturated sodium bicarbonate solution (30 mL) to the reaction system under ice bath. The reaction solution was extracted with dichloromethane (30 mL*2), the organic phases were combined, washed with saturated brine, and dried over anhydrous sodium sulfate, filtered and concentrated to obtain light yellow solid compound 1g (100 mg, yield 94.4%). ESI-MS (m/z): 754.2 [M+H]*.
Step 8: Dissolve compound 1g (70 mg, 0.093 mmol) in N,N-dimethylformamide (3 mL), add thereto (1S,2S)-2-methylcyclopropanecarboxylic acid INT- 3b (18.6 mg, 0.186 mmol), diisopropylethylamine (36.0 mg, 0.278 mmol) and (ethyl 2-oximino- cyanoacetate)-N,N-dimethyl-morpholinourea hexafluoro phosphate (79.5 mg, 0.186 mmol). The reaction mixture was stirred under ice bath for 1 hour. After the reaction was complete, water (20 mL) was added to the reaction system. The reaction solution was extracted with ethyl acetate (30 mL*2), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated by filtration, and the residue was purified by preparative liquid chromatography to obtain white solid compound 1 (15 mg, yield 19.3%) and epimer 1' (20 mg, yield 25.8%). The absolute configurations drawn by the two compounds are based on empirical assumptions. In the existing analytical methods, 1 is a compound with relatively low polarity and relatively long retention time in LC-MS and HPLC, and 1' is compound with relatively high polarity and relatively short retention time in LC-MS and HPLC.
Compound 1:
ESI-MS (m/z): 836.5 [M+H]'; LC-MS retention time RT=1.80 min. HPLC retention time RT=11.63 min. 'H NMR (500 MHz, DMSO-d6) 5 8.81 (d, J = 2.0 Hz, 1H), 8.54 — 8.48 (m, 2H), 7.86 (d, J = 2.0 Hz, 1H), 7.81 (s, 1H), 7.77 — 7.73 (m, 1H), 7.58 (m, 1H), 5.56 (t, J = 9.0 Hz, 1H), 5.10 — 5.04 (m, 1H), 4.36 — 4.15 (m, 4H), 4.14 — 4.01 (m, 1H), 3.61 (t, J = 5.0 Hz, 4H), 3.59 — 3.53 (m, 4H), 3.32 — 3.29 (m, 1H), 3.25 (s, 3H), 3.18 — 3.11 (m, 66
BL-5758
IH ), 3.00 — 2.94 (m, 1H), 2.79 — 2.73 (m, 1H), 2.56 — 2.52 (m, 4H), 2.40 — 2.35 (m, | LU505117 1H), 2.11 — 2.06 (m, 1H), 1.85 — 1.73 (m, 2H), 1.57 — 1.47 (m, 2H), 1.35 (d, J = 6.0 Hz, 3H), 1.11 — 1.03 (m, 4H), 0.91 (s, 3H), 0.90 — 0.85 (m, 4H ), 0.59 — 0.53 (m, 1H), 0.34 (s, 3H).
Compound 1":
ESI-MS (m/z): 836.5 [M+H]'; LC-MS retention time RT=1.77 min. HPLC retention time RT=11.27 min. 'H NMR (500 MHz, DMSO-d6) 5 8.83 (d, J = 2.0 Hz, 1H), 8.56 — 8.51 (m, 2H), 8.00 (d, J = 2.0 Hz, 1H), 7.81 (s, 1H), 7.77 — 7.71 (m, 1H), 7.57 — 7.52 (m, 1H), 5.55 (t,
J=9.0 Hz, 1H), 5.10 — 5.04 (m, 1H), 4.28 — 4.17 (m, 2H), 4.00 — 3.91 (m, 2H), 3.88 — 3.79 (m, 1H), 3.69 — 3.53 (m, 9H), 3.18 — 3.13 (m, 1H), 3.10 (s, 3H), 3.07 — 3.02 (m, 1H) , 2.81 — 2.74 (m, 1H), 2.57 — 2.52 (m, 4H), 2.35 — 2.29 (m, 1H), 2.16 — 2.07 (m, 1H), 1.84 — 1.76 (m, 2H), 1.56 — 1.48 ( m, 2H), 1.22 (d, J = 6.0 Hz, 3H), 1.11 (t, J = 7.0
Hz, 3H), 1.08 — 1.05 (m, 4H), 0.98 — 0.92 (m, 3H), 0.90 — 0.85 (m, 1H), 0.58 — 0.54 (m, 1H), 0.50 (s, 3H).
Example 2 (1r,2R,3S)-N-((638,4S,Z)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(3- morpholinoprop-1-yn-1 -yl)pyridin-3-yl)-10,10-dimethyl-58,7-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)-2,3-dimethylcyclopropane-1-carboxamide ey 06 o "Ok Mo
EE
(S) N= . (R) — N
J — dN \_/
Example 2 is prepared by the following steps: 67
BL-5758
LU505117 65 ° © / o "© du od NH (SEN © INT4a { 2 ç > Ds Ho" Ÿ — = VF d Nn 2 - = . /
Og | COMU, DIEA, DMF +
N N
NG / on lo h 9
Il / o "6 Fe
ON 19 Ors N= , H YF ot NA = —S N —
A
Q N
/
Step 1: Dissolve compound 1g (30 mg, 0.04 mmol) in N,N-dimethylformamide (3 mL), add (2R,3S)-2,3-dimethylcyclopropanecarboxylic acid 2a (9.1 mg, 0.08 mmol),
N,N diisopropylethylamine (15.4 mg, 0.12 mmol) and (ethyl 2-oximino-cyanoacetate)-
N,N-dimethyl-morpholineurea hexafluorophosphate (34.1 mg, 0.08 mmol). The reaction mixture was stirred under ice bath for 1 hour. After the reaction was complete, water (20 mL) was added to the reaction system. The reaction solution was extracted with ethyl acetate (30 mL*2), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated by filtration, and the residue was purified by preparative liquid chromatography to obtain white solid compound 2 (10 mg, yield 29.6%) and epimer 2' (15 mg, yield 44.4%). The absolute configurations drawn by the two compounds are based on empirical assumptions. In the existing analytical methods, 2 is a compound with relatively low polarity and relatively long retention time in LC-MS and HPLC, and 2' is compound with relatively high polarity and relatively short retention time in LC-MS and HPLC.
Compound 2:
ESI-MS (m/z): 850.6 [M+H]'; LC-MS retention time RT=1.86 min. HPLC retention time RT=12.10 min. 'H NMR (500 MHz, DMSO-de) & 8.81 (d, J = 2.0 Hz, 1H), 8.51 — 8.49 (m, 1H), 8.39 (d, J = 9.0 Hz, 1H), 7.86 (d, J = 2.0 Hz, 1H), 7.82 (s, 1H), 7.77 — 7.73 (m, 1H), 7.58 (d, J = 9.0 Hz, 1H), 5.56 (t, J = 9.0 Hz, 1H), 5.10 — 5.04 ( m, 1H), 4.36 — 4.04 (m, 68
BL-5758 5H), 3.63 — 3.59 (m, 4H), 3.59 — 3.55 (m, 4H), 3.31 — 3.29 (m, 1H), 3.25 (s, 3H), 3.18 1LU505117 — 3.12 (m, 1H), 2.99 — 2.92 (m, 1H), 2.79 — 2.72 (m, 1H), 2.56 — 2.52 (m, 4H), 2.42 — 2.36 (m, 1H), 2.11 — 2.05 (m, 1H ), 1.83 — 1.70 (m, 2H), 1.56 — 1.47 (m, 1H), 1.35 (d,
J = 6.0 Hz, 3H), 1.26 — 1.13 (m, 4H), 1.10 — 1.04 (m, 5H), 0.91 (s, 3H), 0.88 (t, J = 7.0
Hz, 3H), 0.34 (s, 3H).
Compound 2":
ESI-MS (m/z): 850.6 [M+H]'; LC-MS retention time RT=1.83 min. HPLC retention time RT=11.76 min. 'H NMR (500 MHz, DMSO-d) & 8.82 (d, J = 2.0 Hz, 1H), 8.54 — 8.51 (m, 1H), 8.41 (d, J = 9.0 Hz, 1H), 8.00 (d, J = 2.0 Hz, 1H), 7.82 (s, 1H), 7.76 — 7.71 (m, 1H), 7.54 (d, J= 9.0 Hz, 1H), 5.53 (t, J = 9.0 Hz, 1H), 5.07 — 5.02 (m, 1H), 4.26 — 4.18 (m, 2H), 4.00 — 3.91 (m, 2H), 3.87 — 3.79 (m, 1H), 3.69 — 3.65 (m, 1H), 3.61 (t, J = 5.0 Hz, 4H), 3.59 — 3.52 (m, 3H), 3.31 — 3.29 (m, 1H), 3.19 — 3.12 (m, 1H), 3.09 (s, 3H), 3.06 — 3.02 (m, 1H), 2.81 — 2.73 (m, 1H), 2.56 — 2.52 (m, 4H), 2.34 — 2.30 (m, 1H), 2.15 — 2.06 (m, 1H), 1.84 — 1.75 (m, 2H), 1.57 — 1.47 (m, 1H), 1.25 — 1.20 (m, 4H), 1.20 — 1.15 (m, 2H), 1.13 — 1.08 (m, 6H), 1.08 — 1.05 (m, 3H), 0.93 (s, 3H), 0.49 (s, 3H).
Example 3 (1S,2S)-N-((63S,4S,Z)-12-(2-((S)-1-methoxyethyl)-5-(3-morpholinoprop-1-yn- 1-yl)pyridin-3 -yl)-10,10-dimethyl-5,7-dioxo-11-(2,2,2-trifluoroethyl)- 61,62,63,64,65,66-hexahydro-11H-8-oxa-2( 4,2)-thiazola-1(5,3)-indola-6(1,3)- pyridazinacyclonecaphane-4-yl)-2-methylcyclopropane-1-carboxamide ey O5 oo d A 76 (S) N= . — N
VA Fc” dN \_/
Example 3 is prepared by the following steps: 69
BL-5758
Oo o 0 LU505117
N
Lu gr do o=x X o= a. a. pus — Cs2COs, == = ba LFLCR OT, & © TIA, ba
DMF, rt | DCM
N Ss N N > N N xX N 7 A re’ A re’
Il I I
N N N
IS 1e iS 3a IS 3b ey se, 0,6 se, o \NT-36 o Mao Je o Hel le / N° / N° vo Lg Qe "A VS A ve val
VL, N Ss + N Ss
COMU, DIEA NL NL
DMF — N — N
À 5
FiC FsC
IN fl IN + \/ 3 \_/ 3
Step 1: Compound 1g (30 mg, 0.036 mmol) was dissolved in N,N- dimethylformamide (2 mL), and cesium carbonate (23.7 mg, 0.072 mmol) and 2,2,2- trifluoroethyl trifluoromethanesulfonate (12.6 mg, 0.054 mmol) were added thereto.
The reaction mixture was stirred at room temperature for 16 hours. After the reaction was complete, water (30 mL) was added to the reaction system. The reaction solution was extracted with ethyl acetate (30 mL*2), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated by filtration, and the residue was purified by preparative thin-layer chromatography (DCM:MeOH=30/1) to obtain light yellow solid compound 3a (10 mg, yield 30.2%).
ESI-MS (m/z): 908.2 [M+H]".
Step 2: Compound 3a (10 mg, 0.011 mmol) was dissolved in dichloromethane (2 mL), and trifluoroacetic acid (0.5 mL) was added thereto. The reaction mixture was stirred at room temperature for 2 hours. After the reaction was complete, saturated sodium bicarbonate solution (30 mL) was added to the reaction system under ice bath.
The reaction system was extracted with dichloromethane (30 mL*2), and the organic phases were combined, washed with saturated brine, and dried over anhydrous sodium sulfate, filtered and concentrated to obtain light yellow solid compound 3b (7 mg, yield 70
BL-5758 78.7%). ESI-MS (m/z): 808.2 [M+H]". LUS05117
Step 3: Compound 3b (7 mg, 0.011 mmol) was dissolved in N,N- dimethylformamide (2 mL), and (1S,2S)-2-methylcyclopropanecarboxylic acid INT- 3b (2.2 mg, 0.022 mmol), N,N-diisopropylethylamine (4.2 mg, 0.033 mmol) and (ethyl 2-oximino-cyanoacetate)-N,N-dimethyl-morpholine urea hexafluorophosphate (9.4 mg, 0.022 mmol) were added thereto. The reaction mixture was stirred under ice bath for 1 hour. After the reaction was complete, water (20 mL) was added to the reaction system, the reaction system was extracted with ethyl acetate (30 mL*2), and the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated by filtration, and the residue was purified by preparative liquid chromatography to obtain white solid compound 3 (0.25 mg, yield 4.5%) and epimer 3' (0.25 mg, yield 4.5%). The absolute configurations drawn by the two compounds are based on empirical assumptions. In the existing analytical methods, 3 is a compound with relatively low polarity and relatively long retention time in LC-MS and HPLC, and 3' is compound with relatively high polarity and relatively short retention time in
LC-MS and HPLC.
Compound 3:
ESI-MS (m/z): 890.2 [M+H]'; LC-MS retention time RT=1.84 min. HPLC retention time RT=13.37 min. 'H NMR (500 MHz, DMSO-d6) 5 8.85 — 8.82 (m, 1H), 8.55 — 8.50 (m, 2H), 7.87 (s, 1H), 7.86 — 7.81 (m, 2H), 7.79 — 7.75 (m , 1H), 5.63 — 5.51 (m, 2H), 5.09 — 5.04 (m, 1H), 4.89 — 4.81 (m, 1H), 4.27 — 4.20 (m, 3H), 3.63 — 3.56 (m, 8H), 3.30 — 3.27 (m, 1H), 3.19 — 3.11 (m, 2H), 3.06 — 3.00 (m, 1H), 2.79 — 2.72 (m, 1H), 2.55 — 2.52 (m, 2H), 2.40 — 2.36 (m, 1H), 2.12 — 2.08 (m, 1H), 2.03 — 1.97 (m, 1H), 1.82 — 1.78 (m, 2H), 1.54 — 1.49 (m, 2H), 1.36 (d, J = 6.0 Hz, 3H), 1.27 — 1.19 (m, 2H), 1.10 — 1.05 (m, 4H), 0.94 (s, 3H), 0.89 — 0.84 (m, 2H), 0.58 — 0.53 (m, 1H), 0.29 (s, 3H) .
Compound 3":
ESI-MS (m/z): 890.2 [M+H]'; LC-MS retention time RT=1.80 min. HPLC retention time RT=12.95 min. 'H NMR (500 MHz, DMSO-de) 5 8.82 (d, J = 2.0 Hz, 1H), 8.55 — 8.51 (m, 2H), 7.95 (d, J = 2.0 Hz, 1H), 7.87 (s, 1H) , 7.84 — 7.80 (m, 1H), 7.71 — 7.67 (m, 1H), 5.54 71
BL-5758
LU505117 (t J= 9.0 Hz, 1H), 5.07 — 5.02 (m, 1H), 4.98 — 4.79 (m, 2H), 4.27 — 4.19 (m, 2H), 3.90 (q, J = 6.0 Hz, 1H), 3.68 — 3.65 (m, 1H), 3.64 — 3.59 (m, 4H), 3.58 (s, 2H), 3.53 — 3.47 (m , 1H), 3.19 — 3.12 (m, 1H), 3.09 (s, 3H), 3.04 (d, J = 14.5 Hz, 1H), 2.80 — 2.73 (m, 1H), 2.56 — 2.52 (m, 4H), 2.47 — 2.42 (m, 1H), 2.16 — 2.09 (m, 1H), 1.84 — 1.77 (m, 2H), 1.54 — 1.49 (m, 2H), 1.24 (d, J = 6.0 Hz, 3H), 1.09 — 1.06 (m, 4H), 0.93 (s, 3H), 0.90 — 0.83 (m, 2H), 0.58 — 0.54 (m, 1H), 0.46 (s, 3H).
Example 4 (1S,2S)-N-((63S,4S,Z)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(3-((S)-2- methylmorpholino) prop-1-yn-1-yl)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)- pyridazinacyclonecaphane-4-yl)-2-methylcyclopropane-1-carboxamide ©)
Oy NO 6 o Pel Mle d N 76 (S) N=
S
N S
NL
— N —
O N
\S) /
Example 4 is prepared by the following steps: 72
BL-5758
LU505117
O O
(ee Lu
QO, | > 0X
Dan NA pr Ns Ps oO © INTA | = | = 9 | 0, Cs2COs, Et 6
N= HN—-Boc ——" I LA ï ne es IN N DMF. rt NTN od : HO, H | H
N 70°C, 16h A A
H
INT2 Il Il
J À © J © ae N 76
GN Oo (S) N=!
INH (9 Y=NHBoc AA 7 3 Ps o INT-3b 4 5 = Ho” “Re IN 7 _TFA - 05 v7, 8 / 4
DCM ï COMU, DIEA $ + x N DMF \ A N O 6 O
Li
S 0 (9)
I de ° N N Vo
N yg Ss
Se NL : 4c — _
IN y a \o / N x
Step 1: Dissolve compound INT-2 (102.1 mg, 0.15 mmol) in a mixed solution of 1,4-dioxane (3 mL) and water (0.3 mL), and add successively INT-6 (40 mg, 0.11 mmol), [1,1'-bis(diphenylphosphino)ferrocene]palladium dichloride (8.3 mg, 0.011 mmol) and potassium phosphate (72.1 mg, 0.33 mmol). The reaction mixture was stirred at 70 °C for 16 hours under nitrogen protection. After the reaction was complete, water (20 mL) was added to the reaction system, the reaction system was extracted with ethyl acetate (30 mL*2), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated by filtration, and the residue was purified by preparative thin-layer chromatography (dichloromethane/methanol=20:1) to obtain light yellow solid compound 4a (60 mg, yield 63.1%). ESI-MS (m/z): 840.2 [M+H]".
Step 2: Compound 4a (60 mg, 0.071 mmol) was dissolved in N,N- 73
BL-5758 dimethylformamide (2 mL), and cesium carbonate (46.3 mg, 0.142 mmol) and LUS0S117 iodoethane (16.6 mg , 0.106 mmol) were added thereto. The reaction mixture was stirred at room temperature for 16 hours. After the reaction was complete, water (30 mL) was added to the reaction system, the reaction system was extracted with ethyl acetate (30 mL*2), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated by filtration, and the residue was purified by preparative thin-layer chromatography (dichloromethane/methanol=30/1) to obtain light yellow solid compound 4b (50 mg, yield 81.2%). ESI-MS (m/z): 868.6 [M+H]".
Step 3: Compound 4b (50 mg, 0.058 mmol) was dissolved in dichloromethane (2 mL), and trifluoroacetic acid (0.5 mL) was added thereto. The reaction mixture was stirred at room temperature for 2 hours. After the reaction was complete, saturated sodium bicarbonate solution (30 mL) was added to the reaction system under ice bath.
The reaction system was extracted with dichloromethane (30 mL*2), and the organic phases were combined, washed with saturated brine, and dried over anhydrous sodium sulfate, filtered and concentrated to obtain light yellow solid compound 4¢ (40 mg, yield 90.4%). ESI-MS (m/z): 768.4 [M+H]".
Step 4: Dissolve compound 4¢ (40 mg, 0.052 mmol) in N,N-dimethylformamide (2 mL), add thereto (1S,2S)-2-methylcyclopropanecarboxylic acid INT-3b (10.4 mg, 0.104 mmol), N,N-Diisopropylethylamine (20.2 mg, 0.156 mmol) and (ethyl 2- oximino-cyanoacetate)-N,N-dimethyl-morpholine urea hexafluorophosphate (55.8 mg, 0.104 mmol). The reaction mixture was stirred under ice bath for 1 hour. After the reaction was complete, water (20 mL) was added to the reaction system, the reaction system was extracted with ethyl acetate (30 mL*2), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated by filtration, and the residue was purified by preparative liquid chromatography to obtain white solid compound 4 (6 mg, yield 13.6%) and epimer 4' (9 mg, yield 20.3%). The absolute configurations drawn by the two compounds are based on empirical assumptions. In the existing analytical methods, 4 is a compound with relatively low polarity and relatively long retention time in LC-MS and HPLC, and 4' is compound with relatively high polarity and relatively short retention time in LC-MS and HPLC. 74
BL-5758
Compound 4: LUSOS117
ESI-MS (m/z): 850.4 [M+H]'; LC-MS retention time RT=1.88 min. HPLC retention time RT=13.62 min.
TH NMR (500 MHz, DMSO-ds) & 8.81 (d, J = 2.0 Hz, 1H), 8.53 — 8.48 (m, 2H), 7.86 (d, J = 2.0 Hz, 1H), 7.81 (s, 1H) , 7.77 — 7.73 (m, 1H), 7.58 (d, J = 8.5 Hz, 1H), 5.56 (t, J= 9.0 Hz, 1H), 5.10 — 5.06 (m, 1H), 4.36 — 4.13 (m, 4H ), 4.12 — 4.02 (m, 1H), 3.80 — 3.75 (m, 1H), 3.61 — 3.48 (m, 6H), 3.25 (s, 3H), 3.18 — 3.11 (m, 1H), 2.99 — 2.94 (m, 1H), 2.81 — 2.70 (m, 3H), 2.40 — 2.36 (m, 1H), 2.30 — 2.24 (m, 1H), 2.11 — 2.05 (m, 1H), 2.00 — 1.94 (m, 2H), 1.83 — 1.75 (m, 2H), 1.53 — 1.47 (m, 2H), 1.35 (d, J = 6.0
Hz, 3H), 1.08 — 1.04 (m, 7H), 0.92 — 0.85 (m, 8H), 0.57 — 0.53 (m, 1H), 0.34 (s, 3H).
Compound 4":
ESI-MS (m/z): 850.4 [M+H]'; LC-MS retention time RT=1.85 min. HPLC retention time RT=13.25 min.
TH NMR (500 MHz, DMSO-d) & 8.82 (d, J = 2.0 Hz, 1H), 8.56 — 8.49 (m, 2H), 7.99 (d, J = 2.0 Hz, 1H), 7.81 (s, 1H) , 7.76 — 7.72 (m, 1H), 7.54 (d, J = 8.5 Hz, 1H), 5.55 (t, J= 9.0 Hz, 1H), 5.07 — 5.02 (m, 1H), 4.28 — 4.17 (m, 2H ), 4.00 — 3.91 (m, 2H), 3.87 — 3.75 (m, 2H), 3.70 — 3.64 (m, 1H), 3.60 — 3.48 (m, 4H), 3.20 — 3.02 (m, SH), 2.81 — 2.68 (m, 3H), 2.35 — 2.24 (m, 2H), 2.15 — 2.09 (m, 1H), 2.03 — 1.93 (m, 2H), 1.84 — 1.75 (m, 2H), 1.56 — 1.47 (m, 2H) , 1.42 — 1.38 (m, 1H), 1.27 — 1.19 (m, 7H), 1.14 — 1.05 (m, 6H), 0.93 (s, 3H), 0.89 — 0.82 (m, 2H), 0.59 — 0.53 (m, 1H), 0.50 (s, 2H).
Example 5 (1S,2S)-N-((63S,4S,Z)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4- morpholinobut-1-yn-1-yl )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)- pyridazinacyclonecaphane-4-yl)-2-methylcyclopropane-1-carboxamide 75
BL-5758 ©) LU505117
Ou NO
Nz N 0 o Hel Je / N°" 6 H Vo (S) N=
S
N Vs rr NO] — À a 7
Example 5 is prepared by the following steps:
O
N
0 0
Se (s) =NHBoc “ei (s) F=NHBoc 0, N I S = = / NN f, Br NP NTS =O INT-7 9 LE ? 6 5) Cs2COs, Et | 5)
NA ee (dppACla, KaPOa, | DMF, rt > Ÿ = dioxane, HO, N = N N N
N 70°C, 16h FZ A
H
INT2 Il Il 5a 5b
O O
0 0 «= 99 / o "9 u oO ° de ve Vo
N
SH @)=NHBoc AN, 7
PS VAS o INT-3b 5 =e 7
FA. dg TNE N 5
DCM 7 COMU, DIEA {5 +
NTN DMF 0
H
TN X oO (s) SD
I de N 7 N Vo
N ) NN (J = N
O / A
C ’ 7
Step 1: Dissolve compound INT-2 (100 mg, 0.14 mmol) in a mixed solution of 76
BL-5758 1,4-dioxane (5 mL) and water (1 mL), and add INT-7 (51 mg, 0.14 mmol), [1,1"- LUS0S117 bis(diphenylphosphino)ferrocene]palladium dichloride (10 mg, 0.01 mmol) and potassium phosphate (60 mg, 0.28 mmol) successively. The reaction mixture was stirred at 70 °C for 16 hours under nitrogen protection. After the reaction was complete, the reaction solution was filtered with celite, and the concentrated residue was purified by silica gel column chromatography (dichloromethane/methanol = 20/1) to obtain light yellow oily compound Sa (65 mg, yield 53.7%). ESI-MS (m/z): 840.1 [M+H]";
Step 2: Compound Sa (60 mg, 0.07 mmol) was dissolved in N,N- dimethylformamide (2 mL), and cesium carbonate (46 mg, 0.14 mmol) and iodoethane (22 mg , 0.14 mmol) were added thereto. The reaction mixture was stirred at room temperature for 16 hours. After the reaction was complete, water (15 mL) was added to the reaction system, the reaction system was extracted with ethyl acetate (20 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 5b (25 mg, yield 40.3%). ESI-MS (m/z): 868.2 [M+H]";
Step 3: Compound 5b (25 mg, 0.03 mmol) was dissolved in dichloromethane (2 mL), and trifluoroacetic acid (0.5 mL) was added thereto. The reaction mixture was stirred at room temperature for 2 hours. After the reaction was complete, saturated sodium bicarbonate solution (20 mL) was added to the reaction system under ice bath.
The reaction system was extracted with dichloromethane (20 mL*2), and the organic phases were combined, washed with saturated brine, and dried over anhydrous sodium sulfate, filtered and concentrated to obtain light yellow solid compound Se (20 mg, yield 90.9%). ESI-MS (m/z): 768.2 [M+H]";
Step 4: Dissolve compound Se (20 mg, 0.03 mmol) in N,N-dimethylformamide (1 mL), add thereto (1S,2S)-2-methylcyclopropanecarboxylic acid INT-3b (5 mg, 0.05 mmol), N,N-diisopropylethylamine (10 mg, 0.07 mmol) and (ethyl 2-oximino- cyanoacetate)-N,N-dimethyl-morpholineurea hexafluorophosphate (11 mg, 0.03 mmol). The reaction mixture was stirred under ice bath for 1 hour. After the reaction was complete, water (20 mL) was added to the reaction system, the reaction system was extracted with ethyl acetate (20 mL*2), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated by filtration, and the 77
BL-5758 residue was purified by preparative liquid chromatography to obtain white solid LUSOS117 compound 5 (5 mg, yield 22.7%) and epimer 5' (7 mg, yield 31.8%). The absolute configurations drawn by the two compounds are based on empirical assumptions. In the existing analytical methods, 5 is a compound with relatively low polarity and relatively long retention time in LC-MS and HPLC, and 5' is compound with relatively high polarity and relatively short retention time in LC-MS and HPLC.
Compound 5:
ESI-MS (m/z): 850.6 [M+H]'; LC-MS retention time RT=1.89 min. HPLC retention time RT=13.37 min. 'H NMR (500 MHz, DMSO-de) & 8.75 (d, J = 2.1 Hz, 1H), 8.55 — 8.48 (m, 2H), 7.81 (s, 1H), 7.78 — 7.73 (m, 2H), 7.58 ( d, J = 8.7 Hz, 1H), 5.56 (t, J = 9.1 Hz, 1H), 5.10 — 5.06 (m, 1H), 4.37 — 4.04 (m, 5H), 3.60 — 3.54 (m, 6H), 3.25 (s, 3H), 3.18 — 3.11 (m, 1H), 2.98 — 2.94 (m, 1H), 2.80 — 2.72 (m, 1H), 2.69 — 2.57 (m, 4H), 2.44 (s, 4H), 2.39 — 2.35 (m, 1H), 2.11 — 2.07 (m, 1H), 1.83 — 1.76 (m, 2H), 1.53— 1.46 (m, 2H), 1.34 (d, J= 6.0 Hz, 3H), 1.07 (s , 3H), 0.91 (s, 3H), 0.87 (t, J = 7.0 Hz, 3H), 0.57 — 0.53 (m, 1H), 0.33 (s, 3H).
Compound 5":
ESI-MS (m/z): 850.6 [M+H]'; LC-MS retention time RT=1.83 min. HPLC retention time RT=13.01 min. 'H NMR (500 MHz, DMSO-de) & 8.76 (d, J = 2.1 Hz, 1H), 8.56 — 8.50 (m, 2H), 7.91 (d, J= 2.2 Hz, 1H), 7.81 (s, 1H), 7.74 (dd, J = 8.7, 1.7 Hz, 1H), 7.54 (d, J = 8.7
Hz, 1H), 5.54 (t, J = 9.2 Hz, 1H), 5.10 — 5.06 (m, 1H), 4.27 — 4.15 (m, 2H), 3.97 — 3.88 (m, 2H), 3.84 — 3.77 (m, 1H), 3.70 — 3.66 (m, 1H), 3.59 — 3.55 (m, 5H), 3.18 — 3.14 (m, 1H ), 3.08 (s, 3H), 3.07 — 3.02 (m, 1H), 2.80 — 2.73 (m, 1H), 2.68 — 2.57 (m, SH), 2.47 — 2.42 (m, 4H), 2.34 — 2.28 (m , 1H), 2.15 — 2.10 (m, 1H), 1.84 — 1.77 (m, 2H), 1.56 — 1.47 (m, 2H), 1.21 (d, J = 6.2 Hz, 3H), 1.10 (t, J = 7.2 Hz, 3H), 1.07 (s, 3H), 0.93 (s, 3H), 0.90 — 0.84 (m, 2H), 0.55 (d, J = 6.1 Hz, 1H), 0.49 (s, 3H).
Example 6 (1S,28)-N-((63S,4S,Z)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(((S)-4- methylmorpholin-2- yl)ethynyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo- 61,62,63,64,65,66-hexahydro-11H-8-oxa-2(4,2)- Thiazola-1(5,3)-indola-6(1,3)- 78
BL-5758 . . . LU505117 pyridazinacyclonecaphane-4-yl)-2-methylcyclopropane-1-carboxamide
O GA O
NON 0 o Hel Je / N°
O H (S) (S) N=
S
N J
NI] — À / SN —N (S) 0 \ /
Example 6 is prepared by the following steps: 5.0
We 0 (a (s) =NHBoc (ex (s) = NHBoc ° NS = =
J N f Br NTs NTs +0 INT-8 9 A ? & 5 Cs2COs, Etl & 5)
NEA HN Bee d(dppNCla, K:PO4 | DMF, rt
Oo, Ÿ = dioxane, H,0, N = N N S N °° N 70°C, 16h A A
H
INT-2 0% a 05 6b ne EN «= 99 / 0 H (S) A ° de LH ve
N
Ca (s) F=NHBoc N \ / 7 =, Pe © INT-3b 5 = Hue y
FA. bg TNE © 6
DCM 7 COMU, DIEA N +
NTN DMF tr N oO. 6) oO
TVS oO (S) 1.(S) dd. N 7 N VS ow N Ss
N 1 \ 7 6c —= N / À
SS
N
\
Step 1: Dissolve compound INT-2 (119.6 mg, 0.172 mmol) in a mixed solution 79
BL-5758 of 1,4-dioxane (3 mL) and water (0.3 mL), and add successively INT-8 (45 mg, 0.133 12505117 mmol), [1,1'-bis(diphenylphosphino)ferrocene]palladium dichloride (9.7 mg, 0.013 mmol) and potassium phosphate (84.5 mg, 0.399 mmol). The reaction mixture was stirred at 70 °C for 16 hours under nitrogen protection. After the reaction was complete, water (20 mL) was added to the reaction system, the reaction system was extracted with ethyl acetate (30 mL*2), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated by filtration, and the residue was purified by preparative thin-layer chromatography to obtain compound 6a (65 mg, yield 59.3%) as a pale yellow solid. ESI-MS (m/z): 826.2 [M+H]".
Step 2: Compound 6a (65 mg, 0.079 mmol) was dissolved in N,N- dimethylformamide (2 mL), and cesium carbonate (51.3 mg, 0.157 mmol) and iodoethane (18.4 mg, 0.118 mmol) were added thereto. The reaction mixture was stirred at room temperature for 16 hours. After the reaction was complete, water (30 mL) was added to the reaction system, the reaction system was extracted with ethyl acetate (30 mL*2), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated by filtration, and the residue was purified by preparative thin-layer chromatography (dichloromethane/methanol=30/1) to obtain light yellow solid compound 6b (50 mg, yield 74.4%). ESI-MS (m/z): 854.4 [M+H]".
Step 3: Compound 6b (50 mg, 0.059 mmol) was dissolved in dichloromethane (2 mL), and trifluoroacetic acid (0.5 mL) was added thereto. The reaction mixture was stirred at room temperature for 2 hours. After the reaction was complete, saturated sodium bicarbonate solution (30 mL) was added to the reaction system under ice bath.
The reaction system was extracted with dichloromethane (30 mL*2), and the organic phases were combined, washed with saturated brine, and dried over anhydrous sodium sulfate, filtered and concentrated to obtain light yellow solid compound 6¢ (40 mg, yield 90.6%). ESI-MS (m/z): 754.4 [M+H]". 80
BL-5758
Step 4: Dissolve compound 6¢ (40 mg, 0.053 mmol) in N,N-dimethylformamide LUSOS117 (2 mL), add thereto (1S,2S)-2-methylcyclopropanecarboxylic acid INT-3b (10.6 mg, 0.106 mmol), N,N-Diisopropylethylamine (20.6 mg, 0.159 mmol) and (ethyl 2- oximino-cyanoacetate)-N,N-dimethyl-morpholineurea hexafluorophosphate (45.4 mg, 0.106 mmol). The reaction mixture was stirred under ice bath for 1 hour. After the reaction was complete, water (20 mL) was added to the reaction system, the reaction system was extracted with ethyl acetate (30 mL*2), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated by filtration, and the residue was purified by preparative liquid chromatography to obtain white solid compound 6 (8 mg, yield 18.0%) and epimer 6' (10 mg, yield 22.5%). The absolute configurations drawn by the two compounds are based on empirical assumptions. In the existing analytical methods, 6 is a compound with relatively low polarity and relatively long retention time in LC-MS and HPLC, and 6' is compound with relatively high polarity and relatively short retention time in LC-MS and HPLC.
Compound 6:
ESI-MS (m/z): 836.4 [M+H]'; LC-MS retention time RT=1.80 min. HPLC retention time RT=13.34 min. 'H NMR (500 MHz, DMSO-d) & 8.81 (d, J = 2.0 Hz, 1H), 8.54 — 8.48 (m, 2H), 7.86 (d, J = 2.0 Hz, 1H), 7.81 (s, 1H) , 7.77 — 7.74 (m, 1H), 7.58 (d, J = 8.5 Hz, 1H), 5.56 (t, J= 9.0 Hz, 1H), 5.10 — 5.05 (m, 1H), 4.63 — 4.57 (m, 1H ), 4.37 — 4.16 (m, 4H), 4.11 — 4.04 (m, 1H), 3.90 — 3.83 (m, 1H), 3.61 — 3.54 (m, 3H), 3.25 (s, 3H), 3.18 — 3.11 (m , 1H), 3.00 — 2.93 (m, 1H), 2.80 — 2.72 (m, 2H), 2.48 — 2.44 (m, 1H), 2.40 — 2.36 (m, 1H), 2.34 — 2.28 (m, 1H), 2.22 — 2.17 (m, 4H), 2.11 — 2.04 (m, 1H), 1.83 — 1.75 (m, 2H), 1.53 — 1.47 (m, 2H), 1.35 (d, J = 6.0 Hz, 3H), 1.10 — 1.04 (m, 5H), 0.92 (s, 3H), 0.90 — 0.85 (m, 4H), 0.57 — 0.52 (m, 1H), 0.33 (s, 3H).
Compound 6":
ESI-MS (m/z): 836.4 [M+H]'; LC-MS retention time RT=1.76 min. HPLC retention time RT=12.94 min. 'H NMR (500 MHz, DMSO-d) & 8.82 (d, J = 2.0 Hz, 1H), 8.56 — 8.50 (m, 2H), 8.00 (d, J = 2.0 Hz, 1H), 7.81 (s, 1H) , 7.76 — 7.71 (m, 1H), 7.54 (d, J = 8.5 Hz, 1H), 5.54 (t, J= 9.0 Hz, 1H), 5.07 — 5.02 (m, 1H), 4.64 — 4.58 (m, 1H ), 4.28 — 4.18 (m, 2H), 3.98 — 3.91 (m, 2H), 3.88 — 3.79 (m, 2H), 3.71 — 3.64 (m, 1H), 3.62 — 3.50 (m, 2H), 81
BL-5758 3.31 — 3.27 (m, 1H), 3.18 — 3.12 (m, 1H), 3.09 (s, 3H), 3.07 — 3.02 (m, 1H), 2.80 —2.74 | LU505117 (m, 2H), 2.2.48 — 2.2.43 (m, 1H), 2.34 — 2.26 (m, 2H), 2.21 (s, 3H), 2.19 — 2.09 (m, 2H), 1.85 — 1.75 (m, 2H), 1.54 — 1.48 (m, 2H), 1.21 (d, J= 6.0 Hz, 3H), 1.13 — 1.06 (m, 7H), 0.93 (s, 3H), 0.90 — 0.86 (m, 1H), 0.58 — 0.54 (m, 1H), 0.49 (s, 3H).
Example 7 (18,2S)-N-((63S,48,Z)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(((R)-4- methylmorpholin-3- yl)ethynyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo- 61,62,63,64,65,66-hexahydro-11H-8-oxa-2(4,2)- Thiazola-1(5,3)-indola-6(1,3)- pyridazinacyclonecaphane-4-yl)-2-methylcyclopropane-1-carboxamide
O nes) )
HN—N 0 (S) © 9 n= HN (S) s “(Ss
N UN | >
NO] — N y —
J (R) N— /
Example 7 is prepared by the following steps: 82
BL-5758 o™ LU505117
SIN
O O ç ° res pres = ot 3» C > > >
HNN © Br NT NT
J ea ? INT-10
OS, Cs2COs, Etl OS)
N= FN Boc Pd(dppfCla, KsPO. | DMF, rt 0 SS ; ve NS N ' NT N x 5 J dioxane, H20, | 2 H | H
J WN 70°C, 12h CG
H
INT2 Il ll hg \ 7a A 7b ole) Oo / o Me ld © Oe wa vd
Co, (© )=NHBoc A si a Ä o INT-3b BD = Je A
TFA | Ho ve NS
A OS ———— 7
DCM i COMU, DIEA Cs +
SON DMF
\ A" 0 Go 0 ane
O ©), (5)
Le 7 N Vo
NG = S
Ko NA 7c — N \ 7 © 7
Step 1: Dissolve intermediate INT-2 (98 mg, 0.14 mmol) and intermediate INT- (40 mg, 0.12 mmol) in a mixed solvent of 1,4-dioxane (2 mL) and water (0.2 mL), and add [1,1'-bis(diphenylphosphino)ferrocene]palladium dichloride and potassium phosphate (75 mg, 0.35 mmol). The reaction system was replaced with nitrogen and then heated to 70 °C and stirred for 12 hours. After the reaction solution was cooled to room temperature, the reaction solution was filtered through celite, and the filtrate was concentrated. The residue was purified by preparative thin-layer chromatography (dichloromethane/methanol=30/1) to obtain compound 7a (92 mg, yield 94%). ESI-MS (m/z): 826.5 [M+H]".
Step 2: Compound 7a (92 mg, 0.11 mmol) was dissolved in N,N- 83
BL-5758 dimethylformamide (2 mL), cesium carbonate (73 mg, 0.22 mmol) was added, 12505117 subsequently ethyl iodide (35 mg , 0.22 mmol) was added dropwise to the reaction solution, and the reaction solution was stirred at 50 °C for 6 hours. After the reaction solution was cooled to room temperature, saturated brine (20 mL) was added to the reaction system. The reaction solution was extracted with ethyl acetate (30 mL*2), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to obtain the crude product of compound 7b (80 mg, yield 84%). ESI-MS (m/z): 854.5 [M+H]".
Step 3: Dissolve compound 7b (80 mg, 0.09 mmol) in dichloromethane (1 mL), cool the reaction solution to 0 °C, then add trifluoroacetic acid (53 mg, 0.47 mmol) dropwise to the reaction solution . The reaction continued to be stirred at 0 °C for 1 hour. The reaction solution was quenched with saturated aqueous sodium bicarbonate solution, extracted with dichloromethane, separated, the organic phase was dried over anhydrous sodium sulfate, filtered and concentrated to obtain the crude product of compound 7¢ (70 mg, yield 99%). ESI-MS (m/z): 754.4 [M+H]".
Step 4: Dissolve compound 7c (70 mg, 0.09 mmol) and intermediate INT- 3 (18 mg, 0.19 mmol) in N,N-dimethylformamide (3 mL), add N,N-diisopropylethylamine (36 mg, 028 mmol) and (2-oximino-ethyl cyanoacetate)-N,N-dimethyl- morpholinourea hexafluorophosphate (36 mg, 0.19 mmol). The reaction solution was stirred at 0 °C for 30 minutes. Saturated brine (20 mL) was added to the reaction system, the reaction system was extracted with ethyl acetate (30 mL*2), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated, and the residue was purified by preparative liquid phase chromatography to obtain white solid compound 7 (7 mg, yield 9%) and epimer 7' (10 mg, yield 12.9%). The absolute configurations drawn by the two compounds are based on empirical assumptions. In the existing analytical methods, 7 is a compound with relatively low polarity and relatively long retention time in LC-MS and HPLC, and 7' is compound with relatively high polarity and relatively short retention time in LC-MS and HPLC. 84
BL-5758
LU505117
Compound 7:
ESI-MS (m/z): 836.6 [M+H]". LC-MS retention time RT=2.09 min. HPLC retention time RT=13.10 min. "H NMR (500 MHz, DMSO) 8 8.81 (d, J = 2.1 Hz, 1H), 8.51 (dd, J = 9.3, 5.0 Hz, 2H), 7.85 (d, J = 1.9 Hz, 1H), 7.81 (s, 1H), 7.75 (dd, J = 8.6, 1.5 Hz, 1H), 7.58 (d, J = 8.7 Hz, 1H), 5.56 (t, J = 9.3 Hz, 1H), 5.09 — 5.05 (m, 1H) , 4.35 — 4.16 (m, 4H), 4.10 — 4.05 (m, 1H), 3.79 — 3.75 (m, 1H), 3.72 — 3.56 (m, SH), 3.51 (s, 1H), 3.31 (s, 2H) , 3.25 (s, 3H), 3.17 — 3.08 (m, 2H), 3.00 — 2.95 (m, 1H), 2.79 — 2.72 (m, 1H), 2.68 — 2.62 (m, 1H), 2.40 — 2.33 (m, 4H), 2.32 — 2.27 (m, 1H), 2.11 — 2.07 (m, 1H), 1.83 — 1.74 (m, 2H), 1.54 — 1.46 (m, 2H), 1.35 (d, J = 6.1 Hz, 3H), 1.23 (s, 1H), 1.06 (s, 3H), 0.92 (s, 3H), 0.87 (t, J = 7.0 Hz, 3H), 0.55 (dd, J = 7.7, 5.5 Hz, 1H), 0.34 (s, 3H).
Compound 7":
ESI-MS (m/z): 836.6 [M+H]". LC-MS retention time RT=2.06 min. HPLC retention time RT=12.76 min. "HNMR (500 MHz, DMSO) & 8.84 — 8.80 (m, 1H), 8.53 (d, J = 9.2 Hz, 2H), 7.99 (d,J= 1.5 Hz, 1H), 7.81 (s, 1H), 7.74 (dd, J = 8.7, 1.5 Hz, 1H), 7.54 (d, J = 8.7 Hz, 1H), 5.54 (t, J= 9.3 Hz, 1H), 5.07 — 5.02 (m, 1H), 4.25 — 4.20 (m, 1H), 3.98 — 3.92 (m, 1H), 3.86 — 3.82 (m, 1H), 3.78 — 3.72 (m, 1H), 3.70 — 3.65 (m, 3H), 3.63 — 3.59 (m, 1H), 3.57 — 3.49 (m, 2H), 3.31 (s, 2H), 3.18 — 3.12 (m, 1H), 3.09 (s, 3H), 3.07 — 3.02 (m, 1H), 2.79 — 5.75 (m, 1H), 2.68 — 2.61 (m, 1H), 2.33 (d, J = 4.1 Hz, 3H), 2.32 — 2.25 (m, 2H), 2.14 — 2.09 (m, 1H), 1.85 — 1.75 (m, 2H), 1.51 (dt, J= 8.0, 6.9 Hz, 2H), 1.21 (d, J = 6.3 Hz, 3H), 1.11 (t J= 7.1 Hz, 3H), 1.07 (s, 4H), 0.93 (s, 3H), 0.90 — 0.85 (m, 1H), 0.56 (dd, J = 7.7, 5.4 Hz, 1H), 0.50 (s, 3H).
Example 8 (1S,2S)-N-((63S,4S,Z)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(((R)-4- methylmorpholin-2- yl)ethynyl)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)- 85
BL-5758 . . . LU505117 pyridazinacyclonecaphane-4-yl)-2-methylcyclopropane-1-carboxamide
O
111-Ç(S) >
HN—N
O (S) 9 78 nA HN (S) 5 =(S)
N UN | San
NL
— À
O—\ ( A
N
\
Example 8 is prepared by the following steps: >N z 0 0
Il N N (a (s) =NHBoc (a (s) =NHBoc ° oO ON = o={
J N f Br NTS © Ns og Cs2COz, Etl OE
N= NB do Gla, KsPO4 Ï DMF, rt
Oo, Ÿ = dioxane, H20, N = N N S N
SN 70°C, 16h A D
H
INT-2 Il I o® oR 8b
O (S) (5) ° de A Vs
N s
Ge} (s) F=NHBoc A Z > =, A, o INT-3b BD
Nee Aw LA
HO Vo ot
TFA os © CP) 8
DCM 7 COMU, DIEA N +
NTN DMF
= ) O © 0
VS oO (S) (5)
I d, N Vo
RN 5
N PN
8c — N / — @
N
\ 86
BL-5758
Step 1: Dissolve intermediate INT-2 (98 mg, 0.14 mmol) and intermediate INT- 12505117 11 (40 mg, 0.12 mmol) in a mixed solvent of 1,4-dioxane (2 mL) and water (0.2 mL), and add [1,1'-bis(diphenylphosphino)ferrocene]palladium dichloride (9 mg, 0.01 mmol) and potassium phosphate (75 mg, 0.35 mmol). The reaction system was replaced with nitrogen and then heated to 70 °C and stirred for 12 hours. After the reaction solution was cooled to room temperature, the reaction solution was filtered through celite, and the filtrate was concentrated. The residue was purified by preparative thin-layer chromatography (dichloromethane/methanol=30/1) to obtain compound 8a (37 mg, yield 38%). ESI-MS (m/z): 826.5 [M+H]".
Step 2: Compound 8a (37 mg, 0.05 mmol) was dissolved in N,N- dimethylformamide (2 mL), cesium carbonate (29 mg, 0.09 mmol) was added, then ethyl iodide (14 mg, 0.09 mmol) was added dropwise to the reaction solution, and the reaction solution was stirred at 50 °C for 6 hours. After the reaction solution was cooled to room temperature, saturated brine (20 mL) was added to the reaction system, the reaction solution was extracted with ethyl acetate (30 mL*2), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated, to obtain the crude product of compound 8b (30 mg, yield 78%). ESI-
MS (m/z): 854.3 [M+H]".
Step 3: Dissolve compound 8b (30 mg, 0.04 mmol) in dichloromethane (1 mL), cool the reaction solution to 0 °C, then add trifluoroacetic acid (20 mg, 0.18 mmol) dropwise to the reaction solution . The reaction continued to be stirred at 0 °C for 1 hour. The reaction solution was quenched with saturated aqueous sodium bicarbonate, extracted with dichloromethane, separated, the organic phase was dried over anhydrous sodium sulfate, filtered and concentrated to obtain the crude product of compound 8c (26 mg, yield 99%). ESI-MS (m/z): 754.5 [M+H]".
Step 4: Dissolve compound 8c (26 mg, 0.04 mmol) and intermediate INT-3 (7 mg, 0.07 mmol) in N,N-dimethylformamide (2 mL), add N,N-di Isopropylethylamine (13 87
BL-5758 mg, 0.10 mmol) and (2-oximino-cyanoacetate ethyl)-N,N-dimethyl-morpholinourea LUS0S117 hexafluorophosphate (14 mg, 0.07 mmol), The reaction solution was stirred at 0°C for minutes. Saturated brine (20 mL) was added to the reaction system, the reaction system was extracted with ethyl acetate (30 mL*2), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated, and the residue was purified by preparative liquid phase chromatography to obtain white solid compound 8 (3 mg, yield 10%) and epimer 8' (5 mg, yield 17.8%).
The absolute configurations drawn by the two compounds are based on empirical assumptions. In the existing analytical methods, 8 is a compound with relatively low polarity and relatively long retention time in LC-MS and HPLC, and 8' is compound with relatively high polarity and relatively short retention time in LC-MS and HPLC.
Compound 8:
ESI-MS (m/z): 836.2 [M+H]". LC-MS retention time RT=1.81 min. HPLC retention time RT=13.43 min. 'H NMR (500 MHz, DMSO) 5 8.81 (d, J = 1.9 Hz, 1H), 8.55 — 8.50 (m, 2H), 7.85 (d, J= 1.9 Hz, 1H), 7.81 (s, 1H), 7.75 (d, J = 8.7 Hz, 1H), 7.58 (d, J = 8.6 Hz, 1H), 5.56 (t, J= 9.1 Hz, 1H), 5.09 — 5.05 (m, 1H), 4.60 (dd, J = 8.0, 2.5 Hz, 1H), 4.38 — 4.15 (m, 5H), 4.10 — 4.04 (m, 1H), 3.88 — 3.83 (m, 1H), 3.61 — 3.54 (m, 3H), 3.32 — 3.31 (m, 2H), 3.25 (s, 3H), 3.17 — 3.08 (m, 2H), 2.99 — 2.95 (m, 1H), 2.80 — 2.72 (m, 2H), 2.40 — 2.35 (m, 1H), 2.32 — 2.28 ( m, 1H), 2.20 (s, 3H), 2.11 — 2.05 (m, 1H), 1.81 — 1.76 (m, 2H), 1.53 — 1.46 (m, 2H), 1.35 (d, J = 6.0 Hz, 3H), 1.23 (s, 1H), 1.06 (s, 3H), 0.91 (s, 3H), 0.87 (t, J = 7.1 Hz, 3H), 0.55 (dd, J= 7.5, 5.4 Hz, 1H), 0.33 (s, 3H).
Compound 8":
ESI-MS (m/z): 836.2 [M+H]". LC-MS retention time RT=1.78 min. HPLC retention time RT=13.01 min. 'H NMR (500 MHz, DMSO) 8 8.82 (d, J = 2.1 Hz, 1H), 8.53 (d, J = 9.2 Hz, 2H), 8.00 (d, J= 2.1 Hz, 1H), 7.81 (s, 1H ), 7.74 (dd, J = 8.7, 1.5 Hz, 1H), 7.54 (d, J = 8.7
Hz, 1H), 5.54 (t, J = 9.4 Hz, 1H), 5.07 — 5.02 (m, 1H), 4.60 (dd, J = 8.1, 2.8 Hz, 1H), 4.25 — 4.20 (m, 2H), 4.00 — 3.90 (m, 2H), 3.88 — 3.79 (m, 2H), 3.68 — 3.62 (m, 1H), 88
BL-5758
LU505117 3.60 — 3.57 (m, 1H), 3.58 — 3.50 (m, 1H), 3.31 (d, J= 1.8 Hz, 2H), 3.18 — 3.14 (m, 1H), 3.09 (s, 3H), 3.08 — 3.02 (m , 1H), 2.78 — 2.73 (m, 2H), 2.50 — 3.45 (m, 1H), 2.32 — 2.25 (m, 2H), 2.20 (d, J= 8.0 Hz, 3H), 2.19 — 2.06 (m, 2H ), 1.83 — 1.77 (m, 2H), 1.56 — 1.47 (m, 2H), 1.21 (d, J = 6.3 Hz, 3H), 1.12 — 1.17 (m, 6H), 0.93 (s, 3H), 0.89 — 0.85 (m, 1H), 0.56 (dd, J = 7.6, 5.5 Hz, 1H), 0.49 (s, 3H).
Example 9 (1S,2S)-N-((63S,4$,Z)-12-(5-(3-((2S,6R)-2,6-dimethylmorpholino)prop-1-yn- 1-y1l)-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)- pyridazinacyclonecaphane-4-yl)-2-methylcyclopropane-1-carboxamide ©)
OS VN 6
Oo Jo Ms / N VW
J N H (S) (S) -
N ~"
NO] — N \ J 7 (S)
O (R) N
Example 9 is prepared by the following steps: 89
BL-5758
LU505117
A SN
© © ç be bg
Q, | 0
J ea 9 INT-9 _ OLE Cs2CO3, Etl OS,
N= Boe a dppClz, KaPOs, Ï DMF, rt 9 J = dioxane, H20, N N N S N
J N 70°C, 16h Z Z
H
INT-2 Il Il
S)
J À © Js
G © de = N ve en 5)}=NHBoc N ~° a I o INT-3b + 3 > yo Yon A _TFA & S) TNE Ar NN 9
DCM | COMU, DIEA I + > N DMF \ po" O 6) 0
J N © J © © N N ~S
Or N ¢ ¢ 9c NS N
ON T
D 9
Step 1: INT-3 (85.0 mg, 0.12 mmol), INT-9 (30 mg, 0.08 mmol), 1,1- bis(diphenylphosphine) dioxonium dichloride palladium (6.0 mg, 0.008 mmol), potassium phosphate (52.0 mg, 0.25 mmol) were dissolved in a mixed solution of 1,4- dioxane (5 mL) and water (1 mL), and the reaction solution was stirred overnight at 70 °C under a nitrogen atmosphere. After the reaction was complete, the reaction solution was filtered, and the filtrate was concentrated to obtain a crude product. The crude product was purified by column chromatography (dichloromethane/methanol = 10/1) to obtain brown oily compound 9a (69.76 mg, yield 100.00%). ESI-MS (m/z): 854.3 [M+H]";
Step 2: Dissolve compound 9a (60.00 mg, 0.07 mmol), cesium carbonate (45.78 90
BL-5758 mg, 0.14 mmol), iodoethane (21.91 mg, 0.14 mmol) in N,N-dimethylformamide (2 505717 mL), the reaction solution was stirred overnight at room temperature. After the reaction, the reaction solution was extracted with ethyl acetate. Then the organic phases were combined, dried and concentrated to obtain light yellow oily compound 9b (61.97 mg, yield 100.00%). ESI-MS (m/z): 882.5 [M+H]*;
Step 3: Compound 9b (60 mg, 0.07 mmol) was dissolved in dichloromethane (2 mL), trifluoroacetic acid (77.55 mg, 0.68 mmol) was added to the above reaction solution, and the reaction solution was stirred overnight at room temperature. After the reaction was complete, saturated aqueous sodium bicarbonate solution was added to the reaction solution. The neutralized reaction solution was extracted with dichloromethane, and the combined organic phases were dried and concentrated to obtain compound 9c (53.19 mg, yield 100.00%). ESI-MS (m/z): 782.5 [M+H]";
Step 4: Compound 9¢ (60 mg, 0.08 mmol), INT-3b (15.4 mg, 0.15 mmol), N- diisopropylethylamine (496 mg, 0.38 mmol) were dissolved in N,N- dimethylformamide (2 mL), and (2-oxime-ethyl cyanoacetate)-N,N-dimethyl- morpholinylurea hexafluorophosphate (65.7 mg, 0.15 mmol) was added to the reaction solution under 0°C. The reaction solution reacted at 0 °C for 2 hours. After the reaction was complete, the reaction solution was extracted with ethyl acetate, the combined organic phases were dried and concentrated to obtain the crude product, which was purified by preparative liquid chromatography to obtain compound 9 (4 mg, yield 6.0%) and epimer 9' (6 mg, yield 9.0%). The absolute configurations drawn by the two compounds are based on empirical assumptions. In the existing analytical methods, 9 is a compound with relatively low polarity and relatively long retention time in LC-MS and HPLC, and 9' is compound with relatively high polarity and relatively short retention time in LC-MS and HPLC.
Compound 9:
ESI-MS (m/z): 864.6 [M+H]"; LC-MS retention time RT = 1.95 min. HPLC retention time RT = 14.28 min. 91
BL-5758
LU505117 "H NMR (500 MHz, DMSO-ds) 8 8.81 (d, J = 2.0 Hz, 1H), 8.53-8.49 (m, 2H), 7.85 (d, J = 2.5 Hz, 1H), 7.81 (s, 1H) , 7.76-7.74 (m, 1H), 7.59-7.57 (m, 1H), 5.56 (t, J = 9.5 Hz, 1H), 5.10 — 5.05 (m, 1H), 4.40-4.15 (m, SH), 4.12 -4.05 (m, 1H), 3.62-3.55 (m, 7H), 3.25 (s, 3H), 3.17-3.12 (m, 1H), 2.98-2.95 (m, 1H), 2.78-2.75 (m, 3H) , 2.40 — 2.35 (m, 1H), 2.09-2.07 (m, 1H), 1.93 — 1.88 (m, 2H), 1.79-1.76 (m, 2H), 1.60-1.45 (m, 3H), 1.35 (d, J= 6.0 Hz, 3H), 1.24 (s, 1H), 1.07 (s, 3H), 1.06 (s, 3H), 0.91 (s, 3H), 0.90-0.85 (m, SH), 0.56-0.52 (m , 1H), 0.34 (s, 3H).
Compound 9":
ESI-MS (m/z): 864.6 [M+H]'; LC-MS retention time RT = 1.87 min. HPLC retention time RT = 13.84 min.
Example 10 (1S,2S)-N-((63S,4S,Z)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(((R)-1- methylpyrrolidin-2- yl)ethynyl)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)- pyridazinacyclonecaphane-4-yl)-2-methylcyclopropane-1-carboxamide
O
Mis) >
HN—N © (S) 9
IS n= HN (S) s “(Sg
N UN >
NI
—, Bi 4 \
NAAR)
Example 10 was prepared by the following steps: 92
BL-5758
LU505117
Boc-N.,
Rr)
I 00 00
Ss As ! wai Fo mas { oO No o= —
Nds Br N N / 2, % INT-9 ° N78 ° NS e/ © or | — Cs,CO, Et
Oo oO _—
N= HN 5 dan ten a | oun >% a8 J = Dean 70°C, 12h NTN NSN oN A #7
N
INT-3 |] ll pg n-Boc ©, N-Boc 10a 10b
Oy io ° 0 SN
Ss p © (s) N Ny, > lod 7 os a Ÿ
NTS HCHO, /
TFA | = NaBH3CN N— 10 +
DCM OS) MeOH, rt
NS Ly 0 lo °°
J + Heol le
Jd NA
Il ©) N=
SS
DH NA
10c = N =
R)
Ne 10°
Step 1: Dissolve intermediate Int-3 (55 mg, 0.08 mmol) and intermediate Int-15 (30 mg, 0.08 mmol) in a mixed solvent of 1,4-dioxane (3 mL) and water (0.2 mL), add 1,1-bis(diphenylphosphine)ironium dichloride palladium (6 mg, 0.01 mmol) and potassium phosphate (47 mg, 0.02 mmol) thereto. The reaction system was replaced with nitrogen and then heated to 70 °C and stirred for 12 hours. After the reaction solution was cooled to room temperature, the reaction solution was filtered through celite, and the filtrate was concentrated. The residue was purified by preparative thin-layer chromatography (dichloromethane/methanol=30/1) to obtain compound 10a (62 mg, yield 96%). ESI-MS (m/z): 879.2 [M+H]".
Step 2: Dissolve compound 10a (62 mg, 0.07 mmol) in DMF (2 mL), add cesium carbonate (47 mg, 0.14 mmol), then add iodoethane (23 mg, 0.14 mmol) dropwise to the reaction solution. The reaction solution was stirred at room temperature for 6 hours. 93
BL-5758
LCMS detected that the reaction was complete. Saturated brine was added to the 12505117 reaction system, the reaction solution was extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to obtain the crude compound 10b (64 mg, yield 99%).
ESI-MS (m/z): 908.3 [M+H]*.
Step 3: Compound 10b (64 mg, 0.07 mmol) was dissolved in dichloromethane (1 mL), and trifluoroacetic acid (41 mg, 0.36 mmol) was added dropwise to the reaction solution at 0 °C. The reaction solution was stirred at 0 °C for 1 hour. LCMS detected that the reaction was complete. Add saturated aqueous sodium bicarbonate solution to the reaction system to quench the reaction, the reaction solution was extracted with dichloromethane, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to obtain compound 10¢ (57 mg, yield 99%) crude products. ESI-MS (m/z): 807.6 [M+H]".
Step 4: The crude compound 10c (57 mg, 0.07 mmol) was dissolved in methanol (2 mL), and aqueous formaldehyde (0.05 mL) was added dropwise to the reaction solution at room temperature, and the reaction solution was stirred at room temperature for 10 min. Then sodium cyanoborohydride (14 mg, 0.21 mmol) was slowly added to the reaction solution, and the reaction solution was stirred at room temperature for 3 hours. LCMS detected that the reaction was complete. Add saturated ammonium chloride aqueous solution to the reaction system to quench the reaction. The reaction solution was extracted with dichloromethane, the organic phases were combined and concentrated, and the residue was purified by preparative liquid chromatography to obtain white solid compound 10 (3 mg, yield 5.2%) and the isomer 10' (4 mg, yield 6.9%). The absolute configurations drawn by the two compounds are based on empirical assumptions. In the existing analytical methods, 10 is a compound with relatively low polarity and relatively long retention time in LC-MS and HPLC, and 10' is compound with relatively high polarity and relatively short retention time in LC-MS and HPLC.
Compound 10: 94
BL-5758
ESI-MS (m/z): 820.0 [M+H]. LC-MS retention time RT = 1.97 min. HPLC 505717 retention time RT = 14.55 min. "HNMR (500 MHz, DMSO) 5 8.79 (d, J = 2.1 Hz, 1H), 8.55 — 8.47 (m, 2H), 7.82 (d, J= 2.1 Hz, 1H), 7.81 (s, 1H), 7.75 (dd, J = 8.7, 1.5 Hz, 1H), 7.58 (d, J= 8.7 Hz, 1H), 5.56 (t, J= 9.3 Hz, 1H), 5.10 — 5.05 (m, 1H), 4.36 — 4.15 ( m, 4H), 4.12 — 4.03 (m, 1H), 3.57 (s, 2H), 3.37 (t, J= 6.8 Hz, 1H), 3.25 (s, 3H), 3.17 — 3.11 (m, 1H), 3.00 — 2.94 (m, 1H), 2.83 - 2.72 (m, 2H), 2.41 — 2.31 (m, SH), 2.19 — 2.11 (m, 1H), 2.08 (d, J = 7.5 Hz, 1H), 1.94 — 1.68 (m, 6H), 1.56 — 1.46 (m, 2H), 1.35 (d, J = 6.1 Hz, 3H), 1.23 (s, 1H), 1.06 (s, 3H), 0.91 (s, 3H), 0.87 (t, J= 7.0 Hz, 4H), 0.57 - 0.52 (m, 1H), 0.34 (s, 3H).
Compound 10":
ESI-MS (m/z): 820.0 [M+H]. LC-MS retention time RT = 1.90 min. HPLC retention time RT = 13.87 min. "H NMR (500 MHz, DMSO) 8 8.80 (d, J =2.1 Hz, 1H), 8.53 (dd, J = 9.3, 2.3 Hz, 2H), 7.96 (d,J=2.1 Hz, 1H), 7.81 (s, 1H), 7.74 (dd, J = 8.7, 1.5 Hz, 1H), 7.54 (d, J = 8.7 Hz, 1H), 5.54 (t, J = 9.4 Hz, 1H), 5.07 — 5.02 (m, 1H) , 4.27 — 4.18 (m, 2H), 3.99 — 3.90 (m, 2H), 3.85 — 3.80 (m, 1H), 3.70 — 3.66 (m, 1H), 3.57 — 3.52 (m, 1H), 3.42 — 3.37 (m, 1H), 3.31 (s, 2H), 3.19 — 3.01 (m, SH), 2.80 — 2.75 (m, 2H), 2.40 — 2.28 (m,
SH), 2.18 — 2.12 (m, 2H), 1.92 — 1.69 (m, SH), 1.57 — 1.47 (m, 2H), 1.21 (d, J = 6.3 Hz, 3H), 1.10 (t, J = 7.1 Hz, 3H), 1.07 (s, 3H), 0.93 (s , 3H), 0.90 — 0.85 (m, 1H), 0.56 (dd,
J=7.7, 5.5 Hz, 1H), 0.50 (s, 3H).
Example 11 (1r,2R,3S)-N-((63S,4S,Z)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(((S)-1- methylpyrrolidin- 2-yl)ethynyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo- 61,62,63,64,65,66-hexahydro-11 H-8-oxa-2(4,2 )-thiazola-1(5,3)-indola-6(1,3)- pyridazinacyclonecaphane-4-yl)-2,3-dimethylcyclopropane-1-carboxamide 95
BL-5758
LU505117
O
Mods)
HN—N © (S) 9 = N= HN {r)
N © ~ S DB (S) —_ N \ 5
Example 11 is prepared by the following steps:
Boc-N (s)
Il 00 00 278 SA oO Nu o= O0=
Sov. (lg Br TX N / © % INT-16 ° NTS © NTS
PH | Cato © A Pddepioh, KıPOs, OC DWF, R 09
N= s = Os) dioxane, H20, | N | N 9, = 70°C, 12h NTN N sul EE 7
H
INT4 Il Il (s) Bo (S) n-Boc 11a 11b os 99 / o "we um 0 Wl ff
SS
Co N 9 0 (s) N \ A
Cho NO =/ N ox / — oO
NTS HCHO, ©
TFA | — NaBH3CN N— no,
DCM OS) MeOH, rt ee}
NTN Oy NNO 6
U 7 / o "© nue
Ill Ow N= H ff
SS
ZH NA
110 = N = (s) vu a
Step 1: Dissolve intermediate Int-4 (93 mg, 0.14 mmol) and intermediate Int-16 (50 mg, 0.12 mmol) in a mixed solvent of 1,4-dioxane (3 mL) and water (0.2 mL), add 96
BL-5758 1,1-bis(diphenylphosphine)ironium dichloride palladium (9 mg, 0.01 mmol) and 505717 potassium phosphate (78 mg, 0.37 mmol) thereto. After the system was replaced with nitrogen, it was heated to 70 °C and stirred for 12 hours. After the reaction solution was cooled to room temperature, the reaction solution was filtered through celite, and the filtrate was concentrated. The residue was purified by preparative thin-layer chromatography (dichloromethane/methanol=30/1) to obtain compound 11a (105 mg, yield 96%). ESI-MS (m/z): 892.4 [M+H]".
Step 2: Dissolve compound 11a (105 mg, 0.12 mmol) in DMF (2 mL), add cesium carbonate (115 mg, 0.35 mmol), then add iodoethane (55 mg, 0.35 mmol) dropwise to the reaction solution. The reaction solution was stirred at room temperature for 6 hours. LCMS detected that the reaction was complete. Saturated brine was added to the reaction system, the reaction solution was extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to obtain the crude compound 11b (108 mg, yield 99%). ESI-MS (m/z): 920.5 [M+H]".
Step 3: Compound 11b (108 mg, 0.12 mmol) was dissolved in dichloromethane (1 mL), and trifluoroacetic acid (67 mg, 0.59 mmol) was added dropwise to the reaction solution at 0 °C. The reaction solution was stirred at 0 °C for 1 hour. LCMS detected that the reaction was complete. Saturated aqueous sodium bicarbonate solution was added to the reaction system to quench the reaction, the reaction solution was extracted with dichloromethane, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to obtain compound 11¢ (96 mg, yield 99%) crude products. ESI-MS (m/z): 820.4 [M+H]".
Step 4: The crude compound 11¢ (96 mg, 0.12 mmol) was dissolved in methanol (2 mL), and aqueous formaldehyde (0.05 mL) was added dropwise to the reaction solution at room temperature, and the reaction solution was stirred at room temperature for 10 min. Then sodium cyanoborohydride (22 mg, 0.35 mmol) was slowly added to 97
BL-5758 the reaction solution, and the reaction solution was stirred at room temperature for 3 LUSOS117 hours. LCMS detected that the reaction was complete. Add saturated ammonium chloride aqueous solution to the reaction system to quench the reaction, extract the reaction solution with dichloromethane, combine and concentrate the organic phase, and the residue was purified by preparative liquid chromatography to obtain white solid compound 11 (3 mg, yield 3%) and epimer 11' (5 mg, yield 5%). The absolute configurations drawn by the two compounds are based on empirical assumptions. In the existing analytical methods, 11 is a compound with relatively low polarity and relatively long retention time in LC-MS and HPLC, and 11' is compound with relatively high polarity and relatively short retention time in LC-MS and HPLC.
Compound 11:
ESI-MS (m/z): 834.5 [M+H]. LC-MS retention time RT = 2.07 min. HPLC retention time RT = 15.25 min. 'H NMR (500 MHz, DMSO) 5 8.79 (d, J = 2.0 Hz, 1H), 8.50 (s, 1H), 8.39 (d, J = 8.9 Hz, 1H), 7.82 (d, J = 2.1 Hz, 1H), 7.81 (s, 1H), 7.75 (dd, J = 8.6, 1.3 Hz, 1H), 7.58 (d, J= 8.7 Hz, 1H), 5.55 (t, J = 9.2 Hz, 1H), 5.09 — 5.02 (m, 1H), 4.37 - 4.13 (m, 5H), 4.12 - 4.02 (m, 1H), 3.57 (s, 2H), 3.24 (s, 3H), 3.18 - 3.10 (m, 1H), 2.99 — 2.92 (m, 1H), 2.82 — 2.77 (m, 1H), 2.76 — 2.71 (m, 1H), 2.36 (s, 3H), 2.35 — 2.31 (m, 1H), 2.19 — 2.12 (m, 1H), 2.10 - 2.05 (m, 1H), 1.94 — 1.72 (m, 6H), 1.55 - 1.46 (m, 1H), 1.35 (d, J = 6.1
Hz, 3H), 1.23 (s, 1H), 1.16 (t, J = 5.5 Hz, 2H), 1.08 (d, J = 5.9 Hz, 3H), 1.06 (d, J = 5.5
Hz, 3H), 0.91 (s, 3H), 0.88 (t, J = 7.1 Hz, 3H), 0.35 (s, 3H).
Compound 11"
ESI-MS (m/z): 834.5 [M+H]. LC-MS retention time RT = 2.01 min. HPLC retention time RT = 14.61 min.
Example 12 (1S,25)-N-((63S,4S,Z)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-((1- methylazetidin-3-yl)ethynyl) 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)- pyridazinacyclonecaphane-4-yl)-2-methylcyclopropane-1-carboxamide 98
BL-5758
O LU505117
Sr ((S) )
HN—N
O
O s ° 9 HN
N= S %(S)
S
N x Xo, y NI —, N
N
/
Example 12 is prepared by the following steps:
Boc
N
! 00 00 y A® SA 4 i f i f oO, No o= o=
Norris Br TX N / 2, & INT-12 ° NTS ° NTS
O | = Cs,CO;, Etl ° 9 — = 2500» EY
A5 Pd(dppiOl, KPO, | OC DMF. OY
N= s “Os dioxane, H,0, °o J = [— 70°C, 12h NTN NTN
B = H P= J oO N
N
INT-3 Il il doc 12a os 125 dy L y a (S) N=
SS
(gn p uf NA
NH (5) =NH MN os a Ÿ
NTS HCHO,
TFA | NaBH3CN N 12 +
DCM OS) MeOH, rt
Hos 5 7 8, y A
I © N=
N =
SA
N 12¢ = N / ‘ 12"
N
/
Replace INT-15 in Example 10 with INT-12, and use similar methods and reaction steps to obtain compound 12 and epimer 12'. The absolute configurations drawn by the 99
BL-5758 two compounds are based on empirical assumptions. In the existing analytical methods, LUSOS117 12 is a compound with relatively low polarity and relatively long retention time in LC-
MS and HPLC, and 12' is compound with relatively high polarity and relatively short retention time in LC-MS and HPLC.
Compound 12:
ESI-MS (m/z): 806.5 [M+H]'; LC-MS retention time RT = 1.88 min. HPLC retention time RT = 13.59 min. 'H NMR (500 MHz, DMSO-d6) 8 8.77 (d, J = 2.1 Hz, 1H), 8.55 — 8.46 (m, 2H), 7.83 — 7.77 (m, 2H), 7.75 (dd, J = 8.7, 1.7 Hz, 1H), 7.58 (d, J = 8.7 Hz, 1H), 5.56 (t, J =9.1 Hz, 1H), 5.10 — 5.05 (m, 1H), 4.36 — 4.16 (m, 4H), 4.07 (d, J = 7.5 Hz, 1H), 3.61 — 3.52 (m, 4H), 3.45 (p, J = 7.3 Hz, 1H), 3.25 (s, 3H), 3.18 — 3.06 (m, 3H), 2.99 — 2.94 (m, 1H), 2.79 — 2.72 (m, 1H), 2.40 — 2.35 (m, 1H), 2.22 (s, 3H), 2.12 — 2.05 (m, 1H), 1.84 — 1.75 (m, 2H), 1.54 — 1.46 (m, 2H), 1.35 (d, J = 6.0 Hz, 3H), 1.09 — 1.05 (m, 4H), 0.93 — 0.85 (m, 7H), 0.58 — 0.52 (m, 1H), 0.33 (s, 3H).
Compound 12"
ESI-MS (m/z): 806.5 [M+H]'; LC-MS retention time RT = 1.83 min. HPLC retention time RT = 12.92 min. 'H NMR (500 MHz, DMSO-de) & 8.77 (d, J = 2.1 Hz, 1H), 8.55 — 8.46 (m, 2H), 7.83 — 7.77 (m, 2H), 7.75 (dd, J = 8.7, 1.7 Hz, 1H), 7.58 (d, J = 8.7 Hz, 1H), 5.56 (t, J =9.1 Hz, 1H), 5.10 — 5.05 (m, 1H), 4.36 — 4.16 (m, 4H), 4.07 (d, J = 7.5 Hz, 1H), 3.61 — 3.52 (m, 4H), 3.45 (p, J = 7.3 Hz, 1H), 3.25 (s, 3H), 3.18 — 3.06 (m, 3H), 2.99 — 2.94 (m, 1H), 2.79 — 2.72 (m, 1H), 2.40 — 2.35 (m, 1H), 2.22 (s, 3H), 2.12 — 2.05 (m, 1H), 1.84 — 1.75 (m, 2H), 1.54 — 1.46 (m, 2H), 1.35 (d, J = 6.0 Hz, 3H), 1.09 — 1.05 (m, 4H), 0.93 — 0.85 (m, 7H), 0.58 — 0.52 (m, 1H), 0.33 (s, 3H).
Example 13 (1S,28)-N-((63S,4S,Z)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(((S)-1- methylazetidin-2- yl)ethynyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo- 61,62,63,64,65,66-hexahydro-11H-8-oxa-2(4,2)- Thiazola-1(5,3)-indola-6(1,3)- 100
BL-5758 . . . LU505117 pyridazinacyclonecaphane-4-yl)-2-methylcyclopropane-1-carboxamide
O
San (5) )
HN—N
O
© (S) O 79 HN—
N= %(s) >
N IN y NL
SN BE
Example 13 is prepared by the following steps:
ON-Boc 26 00 00 \ SA N SA > wii + i f oO No o= = v s \ Br N / HNN Ce INT43 0 Ws ° N°5
O | = Cs,CO;, Etl o © 79 -_
Ge J Pd(dppf)Cla, K3POy, 98 DMF, rt og
N= 5 “85 dioxane, H,O, °o À > = 70°C, 12h NST NSN a N A 7 /
N
INT-3 Il I (s) (s)
N-Boc N—Boc 13a 13b
O © Ls d A Vo (S) N=
SS
Gaal dent a
NH (s)~=NH _ os + J
NTS HCHO, (s)
TFA | — NaBH3CN N 13 +
DCM Os), MeOH, rt o
N > N Oy NO © = Hs) J 5
J, [WE © w= (S) N =
NH X N \ 13c gy — (s)
NL 13
Replace INT-15 in Example 10 with INT-13, and use similar methods and reaction 101
BL-5758 steps to obtain compound 13 and epimer 13'. The absolute configurations drawn by the LUSOS117 two compounds are based on empirical assumptions. In the existing analytical methods, 13 is a compound with relatively low polarity and relatively long retention time in LC-
MS and HPLC, and 13' is compound with relatively high polarity and relatively short retention time in LC-MS and HPLC.
Compound 13:
ESI-MS (m/z): 806.5 [M+H]"; LC-MS retention time RT = 1.88 min. HPLC retention time RT = 13.92 min.
Compound 13":
ESI-MS (m/z): 806.5 [M+H]'; LC-MS retention time RT = 1.82 min. HPLC retention time RT = 13.32 min.
Example 14 (1r,2R,3S)-N-((638,4S,Z)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(((S)-1- methylpyrrolidin- 3-yl)ethynyl)pyridin-3-yl)-10,10-dimethyl-58,7-dioxo- 61,62,63,64,65,66-hexahydro-11 H-8-oxa-2(4,2 )-thiazola-1(5,3)-indola-6(1,3)- pyridazinacyclonecaphane-4-yl)-2,3-dimethylcyclopropane-1-carboxamide
O
Mods)
HN—N 9 (S) 9 = NA HN (S) s 2 5)
N > — Bi 4
A
\(S)
ND
Example 14 is prepared by the following steps: 102
BL-5758
Boe, LU5051 17 (s)
I 00 00 9 . ar ox = / 2 5 INTa7 ° Ws ° N°5 of % ——— | Cato © N PP Pd(dppf)Cl2, K3PO4, os DMF, it © ° NS “05 done, HO, DE Ls A Ly 7 ;
INT4 (S) (s)
M oc 14a N oc 14b
Os GA ° 0e / o He yo
Qe nd # ff
SS
Ce N 9 RW (8) N NZ iE SNH (s) r=NH P — AN 0 Ÿ
NTS HCHO, 9
TFA | = NaBH3CN N— 14 +
DCM OS MeOH, rt
NS Ly 0. lah ° 4 os , 3 he
Il Ow N= H YF 9 A >
Noe + AN (s) 14'
N
Replace INT-15 in Example 10 with INT-17, and use similar methods and reaction steps to obtain compound 14 and epimer 14'. The absolute configurations drawn by the two compounds are based on empirical assumptions. In the existing analytical methods, 14 is a compound with relatively low polarity and relatively long retention time in LC-
MS and HPLC, and 14' is compound with relatively high polarity and relatively short retention time in LC-MS and HPLC.
Compound 14:
ESI-MS (m/z): 834.5 [M+H]”; LC-MS retention time RT = 2.02 min. HPLC retention time RT = 14.80 min. 'H NMR (500 MHz, DMSO) 8 8.74 (d, J = 2.1 Hz, 1H), 8.49 (d, J = 1.3 Hz, 1H), 8.39 (d, J = 9.0 Hz, 1H), 7.81 (s, 1H ), 7.76 (d, J = 2.1 Hz, 1H), 7.75 (dd, J = 8.7, 1.6 103
BL-5758
Hz, 1H), 7.57 (d, J= 8.7 Hz, 1H), 5.55 (t, J = 9.4 Hz, 1H ), 5.10 — 5.05 (m, 1H), 4.35 — 505717 4.29 (m, 1H), 4.25 (d, J= 6.1 Hz, 1H), 4.24 — 4.20 (m, 1H), 4.20 — 4.15 (m, 1H), 4.09 - 4.03 (m, 1H), 3.57 (s, 2H), 3.24 (s, 3H), 3.17 - 3.11 (m, 2H), 2.98 - 2.92 (m, 1H), 2.83 (t J= 8.2 Hz, 1H), 2.76 — 2.72 (m, 1H), 2.48 — 2.45 (m, 1H), 2.40 — 5.35 (m, 1H), 2.26 (s, 3H), 2.24 — 2.20 (m, 1H), 2.10 — 2.05 (m, 1H), 2.03 — 1.99 (m, 1H), 1.91 — 1.83 (m, 2H), 1.80 — 1.78 (m, 1H), 1.34 (d, J= 6.1 Hz, 3H), 1.23 (s, 3H), 1.16 (t, J = 5.4 Hz, 3H), 1.08 (d, /=5.9 Hz, 3H), 1.06 (d, J = 5.7 Hz, 3H), 0.91 (s, 3H), 0.88 (d, J = 7.0 Hz, 3H), 0.34 (s, 3H).
Compound 14":
ESI-MS (m/z): 834.5 [M+H]'; LC-MS retention time RT = 1.95 min. HPLC retention time RT = 14.11 min. "H NMR (500 MHz, DMSO) 5 8.76 (d, J = 2.0 Hz, 1H), 8.52 (s, 1H), 8.40 (d, J = 9.0 Hz, 1H), 7.90 (d, J = 2.0 Hz, 1H), 7.82 (s, 1H), 7.73 (dd, J = 8.6, 1.3 Hz, 1H), 7.54 (d,J = 8.7 Hz, 1H), 5.53 (t, J= 9.3 Hz, 1H), 5.10 — 5.05 (m, 1H), 4.27 — 4.20 (m, 2H), 3.97 — 3.89 (m, 2H), 3.85 — 3.77 (m, 1H), 3.70 — 3.65 (m, 1H), 3.56 — 3.51 (m, 1H) , 3.26 — 3.21 (m, 2H), 3.19 — 3.12 (m, 2H), 3.08 (s, 3H), 3.07 — 3.02 (m, 1H), 2.86 — 2.82 (m, 1H), 2.81 — 2.71 (m, 2H), 2.56 — 2.53 (m, 1H), 2.45 (d, J= 7.2 Hz, 1H), 2.34 — 2.29 (m, 1H), 2.26 (s, 3H), 2.25 — 2.19 (m, 1H), 2.14 — 2.10 (m, 1H), 2.03 — 1.96 (m, 1H), 1.88 — 1.84 (m, 1H), 1.81 — 1.79 (m, 1H), 1.56 — 1.43 (m, 3H), 1.23 (s, 3H), 1.20 (d, J = 6.2 Hz, 3H), 1.18 (s, 1H), 1.11 (s, 1H), 1.09 (d, J= 5.7 Hz, 3H), 1.07 (d, J= 5.7 Hz, 3H), 0.93 (s, 3H), 0.85 (t, J= 6.2 Hz, 1H).
Example 15 (1S,2S)-N-((63S,4S,Z)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(3-((R)-3- methylmorpholino) prop-1-yn-1-yl)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)- pyridazinacyclonecaphane-4-yl)-2-methylcyclopropane-1-carboxamide 104
BL-5758 ©) LU505117
LS)
Oy NO à o Hel Le
J N "76 (S) N=
S
N x y NO] — N 2 (R)
O N
\_/
Example 15 is prepared by the following steps: , #3 7 Le g NINE © N= "44 = 7
Hol Ms y de O A, Ze A WHO A 15 = NN +
NA À = DIEA, DCM, 50°C f — J À S J ©
INT-19 Jd N “No
OMs ©) =
A4 > — N + y - a "MN 15"
Step 1: Dissolve compound INT-19 (20 mg, 0.02 mmol) in dichloromethane (5 mL), add thereto (R)-3 methylmorpholine hydrochloride (10 mg, 0.07 mmol) and diisopropylethylamine (18 mg, 0.14 mmol). The reaction mixture was stirred at 50 °C for 16 hours. After the reaction was complete, the reaction solution was concentrated, and the residue was purified by preparative liquid chromatography to obtain white solid compound 15 (3 mg, yield 15.0%) and epimer 15' (5 mg, yield 25.0%). The absolute configurations drawn by the two compounds are based on empirical assumptions. In the existing analytical methods, 15 is a compound with relatively low polarity and relatively long retention time in LC-MS and HPLC, and 15' is compound with relatively high polarity and relatively short retention time in LC-MS and HPLC. 105
BL-5758
Compound 15: LUSOS117
ESI-MS (m/z): 850.6 [M+H]'; LC-MS retention time RT = 1.90 min. HPLC retention time RT=13.50 min. "HNMR (500 MHz, DMSO-d6) 5 8.80 (d, J = 2.1 Hz, 1H), 8.54 — 8.48 (m, 2H), 7.85 (d, J = 2.1 Hz, 1H), 7.81 (s, 1H), 7.75 (dd, J = 8.6, 1.6 Hz, 1H), 7.58 (d, J = 8.7
Hz, 1H), 5.56 (t, J = 9.1 Hz, 1H), 5.10 — 5.05 (m, 1H), 4.38 — 4.15 (m, 4H), 4.12 — 4.04 (m, 1H), 3.78 — 3.61 (m, 4H), 3.58 (s, 2H), 3.52 — 3.47 (m, 1H), 3.25 (s, 3H), 3.19 — 3.02 (m, 3H), 3.00 — 2.95 (m, 1H), 2.80 — 2.69 (m, 2H), 2.41 — 2.36 (m, 1H), 2.11 — 2.05 (m, 1H), 1.84 — 1.74 (m, 2H ), 1.57 — 1.46 (m, 2H), 1.35 (d, J = 6.0 Hz, 3H), 1.07 (s, 3H), 0.94 — 0.91 (m, 6H), 0.88 (t, J = 7.0 Hz, 3H), 0.58 — 0.52 (m, 1H), 0.34 (s, 3H).
Compound 15"
ESI-MS (m/z): 850.6 [M+H]'; LC-MS retention time RT = 1.86 min. HPLC retention time RT=13.14 min. "HNMR (500 MHz, DMSO-d6) 5 8.82 (d, J = 2.1 Hz, 1H), 8.58 — 8.48 (m, 2H), 7.99 (d, J = 2.1 Hz, 1H), 7.81 (s, 1H), 7.74 (dd, J = 8.6, 1.7 Hz, 1H), 7.54 (d, J = 8.6
Hz, 1H), 5.55 (t, J = 9.1 Hz, 1H), 5.10 — 5.05 (m, 1H), 4.28 — 4.16 (m, 2H), 4.01 — 3.89 (m, 2H), 3.87 — 3.73 (m, 3H), 3.71 — 3.60 (m, 3H), 3.57 — 3.45 (m, 2H), 3.18 — 3.12 (m, 1H ), 3.09 (s, 3H), 3.07 — 3.02 (m, 2H), 2.83 — 2.67 (m, 2H), 2.63 — 2.53 (m, 2H), 2.35 — 2.30 (m, 1H), 2.15 — 2.0 (m , 1H), 1.85 — 1.74 (m, 2H), 1.58 — 1.45 (m, 2H), 1.22 (d,
J= 6.2 Hz, 3H), 1.11 (t, J= 7.1 Hz, 3H), 1.07 (s, 3H), 0.95 — 0.91 (m, 6H), 0.90 — 0.84 (m, 1H), 0.58 — 0.53 (m, 1H), 0.50 (s, 3H).
Example 16 (1S,2S)-N-((63S,4S,Z)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(pyridin-2- ylethynyl)pyridin-3-yl )-10,10-dimethyl-5,7-dioxo-61,62,63,64,65,66-hexahydro- 11H-8-oxa-2(4,2)-thiazola-1(5,3)- indola-6(1,3)-pyridazinacyclonecaphane-4-yl)- 2-methylcyclopropane-1-carboxamide 106
BL-5758
LU505117
O © O
NUON 0 o He Jus / N°
O H (S) (S) N=
S
N NS
NN]
X
\ AN
Example 16 is prepared by the following steps: 1 >
NL
I 5 Q d
N LSA)
SN a dE 0, NZ o=
J ) © Br » >
HN—N © \NT-20 NZ S
Da | = Cs,CO;, Etl o 9 — | SoCs FA
N uN—/ Pd(dppf)Cl,, K3PO4, os, ] DMF, rt = “© di , H,0, mo” B= ste AA
B | J H oO N
H
INT-3 Ill
= 16a
GA Co og oh 9 o Hel Js o Hol le
Jd N°76 Jd N°76 © N= © N=
S «LS
NI > + PN —, N — N dl — J —A
À 16 A 16
Replace INT-15 in Example 10 with INT-20, and use similar methods and reaction steps to obtain compound 16 and epimer 16'. The absolute configurations drawn by the two compounds are based on empirical assumptions. In the existing analytical methods, 16 is a compound with relatively low polarity and relatively long retention time in LC-
MS and HPLC, and 16' is compound with relatively high polarity and relatively short 107
BL-5758 retention time in LC-MS and HPLC. LUS0S117
Compound 16:
ESI-MS (m/z): 814.5 [M+H]"; LC-MS retention time RT = 1.89 min. HPLC retention time RT = 14.05 min. "H NMR (500 MHz, DMSO-d6) 5 8.92 (dd, J = 7.1, 2.1 Hz, 1H), 8.63 — 8.54 (m, 1H), 8.50 — 8.39 (m, 2H), 7.99 (d, J = 2.1 Hz, 1H), 7.83 (tt, J = 7.7, 2.1 Hz, 1H), 7.74 (s, 1H), 7.71 — 7.65 (m, 2H), 7.53 (d, J = 8.8 Hz, 1H), 7.42 — 7.37 (m, 1H), 5.49 (1, J = 9.1 Hz, 1H), 5.10 — 5.05 (m, 1H), 4.33 — 4.21 (m, 2H), 4.20 — 4.09 (m, 2H), 4.07 — 4.02 (m , 1H), 3.52 (s, 2H), 3.26 — 3.23 (m, 2H), 3.21 (s, 1H), 3.10 — 3.05 (m, 1H), 2.95 — 2.88 (m, 1H), 2.73 — 2.65 (m , 1H), 2.40 — 2.35 (m, 1H), 2.05 — 2.00 (m, 1H), 1.72 (s, 3H), 1.51 — 1.40 (m, 2H), 1.32 (t, J = 6.4 Hz, 3H), 1.17 (s, 2H), 1.02 — 0.99 (m, 3H), 0.89 — 0.80 (m, 6H), 0.52 — 0.45 (m, 1H), 0.29 (s, 3H).
Compound 16":
ESI-MS (m/z): 814.5 [M+H]"; LC-MS retention time RT = 1.86 min. HPLC retention time RT = 13.61 min. "H NMR (500 MHz, DMSO-de) 5 9.00 (d, J = 2.0 Hz, 1H), 8.64 (dt, J = 4.7, 1.4
Hz, 1H), 8.57 — 8.48 (m, 2H), 8.20 (d, J = 2.1 Hz, 1H), 7.90 (td, J = 7.7, 1.8 Hz, 1H), 7.82 (s, 1H), 7.77 — 7.70 (m, 2H), 7.56 (d, J = 8.7 Hz, 1H), 7.50 — 7.45 (m, 1H),5.55 (t J= 9.2 Hz, 1H), 5.10 — 5.05 (m, 1H), 4.25 — 4.20 (m, 2H), 4.03 — 3.93 (m, 2H), 3.89 — 3.84 (m, 1H), 3.70 — 3.65 (m, 1H), 3.60 — 3.55 (m, 1H), 3.33 (s, 1H), 3.19 — 3.14 (m, 1H), 3.12 (s, 3H), 3.05 ( s, 1H), 2.77 (s, 1H), 2.38 — 2.32 (m, 1H), 2.15 — 2.10 (m, 1H), 1.80 (s, 2H), 1.57 — 1.45 (m, 2H), 1.24 (d, J = 6,3 Hz, 3H), 1.14 (t, J = 7.1 Hz, 3H), 1.09 — 1.06 (m, 4H), 0.94 (s, 3H), 0.88 (d, J= 6.4 Hz, 1H), 0.57 (t,J = 4.2 Hz, 1H), 0.53 (s, 3H).
Example 17 (1S,2S)-N-((63S,4S,Z)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(pyridin-3- ylethynyl)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)-pyridazinacyclonecaphane-4-yl)- 108
BL-5758 2-methylcyclopropane-1-carboxamide LUSOS117
O
Mui) )
HN—N
O (S) 9
Ts n= HN (S)
S NS)
N x >>
NN] —, À / N — N
Example 17 is prepared by the following steps:
N°S =
I 00
N pas 1 S pur oO A 0
HN-N O INT-21 N 0 | = CszCO,, Etl
J © To - | ST,
N HN— Pd(dppf)Clz, KsPO4, og DMF, rt = Zs) dioxane, Hz0, ny 7 7 >= 70°C, 12h NTN
B LU H
OÖ N
H
INT-3 Il
ZN
> 17a © Ge
Oxy y" ° Ox y" °
O (S) 1, (S) O (S) bi (5)
Jd NN J NN (5) N= (5) N= s s
NA > + NA = =/ N — N ya ya a 17 a 17 y y
Replace INT-15 in Example 10 with INT-21, and use similar methods and reaction steps to obtain compound 17 and epimer 17'. The absolute configurations drawn by the two compounds are based on empirical assumptions. In the existing analytical methods, 17 is a compound with relatively low polarity and relatively long retention time in LC- 109
BL-5758
MS and HPLC, and 17' is compound with relatively high polarity and relatively short LUSOS117 retention time in LC-MS and HPLC.
Compound 17:
ESI-MS (m/z): 814.5 [M+H]"; LC-MS retention time RT = 1.91 min. HPLC retention time RT = 14.20 min. "H NMR (500 MHz, DMSO-de) 5 8.97 (d, J = 2.1 Hz, 1H), 8.83 (dd, J = 2.2, 0.9
Hz, 1H), 8.63 (dd, J = 4.9, 1.7 Hz, 1H) , 8.56 — 8.48 (m, 2H), 8.05 (dd, J = 7.6, 2.0 Hz, 2H), 7.82 (s, 1H), 7.77 (dd, J = 8.7, 1.6 Hz, 1H), 7.60 (d, J = 8.7 Hz, 1H), 7.53 — 7.48 (m, 1H), 5.56 (t, J= 9.1 Hz, 1H), 5.10 — 5.05 (m, 1H), 4.40 — 4.29 (m, 2H), 4.28 — 4.14 (m, 2H), 4.12 — 4.07 (m, 1H), 3.59 (s, 2H), 3.32 (s, 1H), 3.28 (s, 3H), 3.18 — 3.13 (m, 1H), 3.00 — 2.95 (m, 1H), 2.80 — 2.74 (m, 1H), 2.45 — 2.40 (m, 1H), 2.12 — 2.07 (m, 1H), 1.79 (s, 2H), 1.56 — 1.47 (m, 2H), 1.38 (d, J= 6.1 Hz, 3H), 1.11 — 1.05 (m, 4H), 0.93 (s, 3H), 0.93 — 0.88 (m, SH), 0.57 — 0.51 (m, 1H), 0.36 (s, 3H).
Compound 17"
ESI-MS (m/z): 814.5 [M+H]"; LC-MS retention time RT = 1.86 min. HPLC retention time RT = 13.67 min. "H NMR (500 MHz, DMSO-de) 5 8.98 (d, J = 2.0 Hz, 1H), 8.82 (dd, J = 2.1, 0.9
Hz, 1H), 8.63 (dd, J = 4.9, 1.7 Hz, 1H), 8.58 — 8.50 (m, 2H), 8.18 (d, J = 2.1 Hz, 1H), 8.05 (dt, J = 8.0, 1.9 Hz, 1H), 7.82 (s, 1H), 7.75 (dd, J= 8.7 , 1.6 Hz, 1H), 7.56 (d, J = 8.7 Hz, 1H), 7.53 — 7.47 (m, 1H), 5.55 (t, J= 9.2 Hz, 1H), 5.10 — 5.05 (m, 1H), 4.29 — 4.13 (m, 2H), 4.00 — 3.95 (m, 2H), 3.90 — 3.85 (m, 1H), 3.70 — 3.65 (m, 1H), 3.58 — 3.53 (m, 1H), 3.32 (s, 1H), 3.19 — 3.14 (m, 1H), 3.12 (s, 3H), 3.10 — 3.05 (m, 1H), 2.81 — 2.74 (m, 1H), 2.40 — 2.35 (m, 1H), 2.16 — 2.11 (m, 1H), 1.84 — 1.79 (m, 2H), 1.58 — 1.42 (m, 2H), 1.24 (d, J= 6.3 Hz, 3H), 1.13 (t J= 7.2 Hz, 3H), 1.07 (d, J= 1.7
Hz, 4H), 0.94 (s, 3H), 0.88 (h, J = 3.7 Hz, 1H), 0.60 — 0.55 (m, 1H), 0.52 (s, 3H).
Biological screening and results of RAS inhibitors 110
BL-5758
Test Example 1: In vitro cell proliferation inhibition test 505717
Due to the diversity of RAS mutations and in order to evaluate the activity of compounds in different RAS mutant cell lines, we selected KRASWT, KRASC!2C,
KRASS!P, KRASS!?Y and BRAF mutant cell lines (see the table below) for in vitro activity evaluation and screening of compounds .
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% CO». 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 of the 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 111
BL-5758
LU505117 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
Exam ICso (nM) ple
NCI- | MIA | LS513 | AsPC- | HCCIS | SW480 | NCI- NCI- HT-29
H358 | PaCa- 1 88 H727 | H520 2
BEE EEE on Tow [ae [on [on [or [oe [om wn po er ew [em we see on [en Lo [on [ow [on [wo [am | we ve Lo Lo [oo [oom] wo | vw | on 112
BL-5758
LU505117 ow [om [se [wr [oo [oo [ow [on [ om os [eo em ve |e [ve wn Lo [oe [oe ow | an | me | om | or pee [ee er [on ow [55 [10 [ra [res pee [on [wo [or [ae | [oe [res pere La [en [on oe |e [re
Co [ee [om en |e ee zum os [x [oe [on [ww |e [vn * NT means not tested 113

Claims (2)

BL-5758 LU505117 Claims
1. A compound with a structure of formula (I) or a pharmaceutically acceptable salt, isotopic derivative, or stereoisomer thereof: oy NT O o (R) R O vx 4), N= Rs! R, s N SN Re / \ J a J Re’ Ry Ra m R7 R7' © wherein: Ri represents C1-C6 alkyl, preferably C1-C3 alkyl, o— o— Ro represents represents = , preferably À ; Rj represents -O(C1-C6) alkyl , -O (Co-Cs alkylene) (C3-Cg) cycloalkyl, -O (Co-Cs alkylene) (3-8 membered) heterocycloalkyl, -O (C,-Cs alkylene )R1 or hydrogen; -Cy1-(R4) in formula (I) is selected from the following: 114
BL-5758 vt Fr ; : 5 win LU505117 meld N uf F +f9 5,05) “NR NT he ve Y EE ge RE A WS OO > EL A *, A + A yd Ra } X AS 0 © CO © LUNG (© 10 EN, (S) N° x (SON, N
*.(s) (R) *. (8) (R) #,(S) (R) * „ À He. ZN rR WF s) O ©) N (S) (S) (S) (R) N N N _ y AN ! oe) IN (5) - “er #45) NT =,(S) A(S) 76 oH rm OH Vo © / x HQ | © o— OH NT = X Oo +,
*. # = x. R) > 0 N Le “Nn a Ki 5) NA 0 = 9 N —0 07 9 N _ 0 0 * N= , "a we ao x, = | Er EY (S) " (No Lo wherein, * represents the site where -Cy1-(R4)p is linked to the linking site in formula (I); Rs represents-Cy2-(R5)q Or-NRyRo', wherein, -Cy2-(Rs)q in formula (I) is selected from the following: 115
BL-5758 ; ; * LU505117 * # 3 # R) GS) ch \S (R) \(S) = di due de GG FG / / \ " " F F /\ > ~~ ~ "ve Dre qd} 0 N-* Oo N=+ ON A N Lu — Yo jo oo a ha GE SN © (S) (BR) + ONT SS U 4 Cs Ce NT Cx IN x S N—= I IN . OMR) qe qe So) Se N Le Ay) Des OH 9 HO Ho— OH nd _ HO,(s) HOMR oO N= IN HEN * HN Cones Th An IN _ x x _ x _ = À _ x 0s/ 7 = = Ch GS N hel PNT NN NN \_+ MON Xl CS Ch 2 Co N-N NH, N N wherein, * represents the site where -Cy2-(Rs)q is linked to the linking site in formula (I);
Ro and Ro each independently represent Ci-Cs alkyl which may be optionally substituted by q Rs;
Rs represents hydrogen, halogen, Ci-Cs alkyl , -(Co-Cs alkylene) ORa, -(Co-Cs alkylene) NRaRa'; or Rs on the two C atoms of Cy2 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 can contain 0, 1, 2 or 3 heteroatoms selected from N, O or S; or two Rs on the same C atom of Cy; is and the C atom connected thereto can form a 3-8 membered ring, and the 3-8 membered ring optionally can contain 0, 1, 2 or 3 selected from N, O or S of heteroatoms;
Rs and Re each independently represent hydrogen, halogen, C1-C6 alkyl, C3-Cs cycloalkyl or -(Co-Cs alkylene)CN;
116
BL-5758 R ‘each i LU505117 7 and R7 each independently represent hydrogen, C1 -Cs alkyl; or R7, R7 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; more preferably, R7 represents hydrogen; wherein, p and q independently represent 0, 1 or 2; m represents 0, 1, 2 or 3; Ra and Ra' independently represent hydrogen, C1-Cs alkyl, and Cs-Cs cycloalkyl; wherein, if Ra and Ra' are connected to the same N atom, the Ra and Ra' can be connected to the common N atom to form a 4-8 membered ring, which optionally may contain 0, 1, 2 or 3 heteroatoms selected from N, O or S; each of the alkyl, cycloalkyl, heterocycloalkyl and alkylene can be independently substituted by 0, 1, 2, 3, 4, 5 or 6 halogen atoms.
2. A compound having the structure: or ed 00 oe 09 oy GA 9 oa 90 FE SR 02e SN nn IN 4 = TX 7 À amy 4 = am y = we 99 N le O9 N oe 99 oO ol °° oO / 3 Fler / A la J 9 ha J 9 A Er EL SS fa fa {fans + / — — / — + À — MH — oy So, oy Gi 04 à oy 00 + oy 4 ws 7 voor da L 7 vs 4, L 2 vu a L 7 je) rer ST 2220 as: = SN ÿ = SN 4 J AN ÿ = SN 4 = 117
LU505117A 2023-09-15 2023-09-15 A pan-KRAS inhibitor compound LU505117B1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
LU505117A LU505117B1 (en) 2023-09-15 2023-09-15 A pan-KRAS inhibitor compound

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
LU505117A LU505117B1 (en) 2023-09-15 2023-09-15 A pan-KRAS inhibitor compound

Publications (1)

Publication Number Publication Date
LU505117B1 true LU505117B1 (en) 2024-03-15

Family

ID=90300600

Family Applications (1)

Application Number Title Priority Date Filing Date
LU505117A LU505117B1 (en) 2023-09-15 2023-09-15 A pan-KRAS inhibitor compound

Country Status (1)

Country Link
LU (1) LU505117B1 (en)

Similar Documents

Publication Publication Date Title
TWI723511B (en) Highly active sting protein agonist compound
WO2019158070A1 (en) A2a and/or a2b receptor antagonist
WO2022089454A1 (en) High-activity wnt pathway inhibitor compound
CN111548343B (en) Preparation method of high-activity CSF1R inhibitor compound
WO2020038387A1 (en) High activity sting protein agonist
US20230399327A1 (en) High activity hpk1 kinase inhibitor
JP7281834B2 (en) PD-L1 antagonist compounds
CA3160606A1 (en) Pd-l1 antagonist compound
EP4006033A1 (en) Adenosine receptor antagonist
LU505117B1 (en) A pan-KRAS inhibitor compound
JP7299411B2 (en) adenosine receptor antagonist
CN111440148B (en) Preparation method of adenosine receptor antagonist
TW202144347A (en) TRANSFORMING GROWTH FACTOR-β RECEPTOR INHIBITOR
WO2024022365A1 (en) Wnt pathway inhibitor compound
WO2022199561A1 (en) Hpk1 kinase inhibitor compound
WO2023005894A1 (en) Inhibitor compound for wnt pathway
CN117903169A (en) Pan-KRAS inhibitor compound
WO2023143546A1 (en) Wnt pathway inhibitor compounds
CN117396482A (en) Wnt pathway inhibitor compound
WO2022214008A1 (en) Highly active hpk1 kinase inhibitor
CN117534685A (en) pan-KRAS inhibitor compound

Legal Events

Date Code Title Description
FG Patent granted

Effective date: 20240315