Title of Invention

"TRICYCLIC DERIVATIVES OR PHARMACEUTICALLY ACCEPTABLE SALTS THEREOF, THEIR PREPARATIONS AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM"

Abstract The present invention relates to tricyclic derivatives or pharmaceutically acceptable salts thereof, their preparations and pharmaceutical compositions containing them. More precisely, the present invention relates to tricyclic derivatives as colchicine derivatives, pharmaceutically acceptable salts thereof, their preparations and pharmaceutical compositions containing them. Tricyclic derivatives of the present invention show very powerful cytotoxicity to cancer cell lines but were much less toxic than colchicine or taxol, confirmed through animal toxicity test. Tricyclic derivatives of the invention also decrease the volume and weight of a tumor and have a strong angiogenesis inhibiting activity in HUVEC cells. Thus, tricyclic derivatives of the present invention can effectively be used as an anticancer agent, anti-proliferation agent and an angiogenesis inhibitor.
Full Text TRICYCLIC DERIVATIVES OR PHARMACEUTICALLY
ACCEPTABLE SALTS THEREOF, THEIR PREPARATIONS
AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
FIELD OF THE INVENTION
The present invention relates to tricyclic
derivatives represented by following , or
pharmaceutically acceptable salts thereof, their
preparations and pharmaceutical compositions containing
them.

(Wherein; RI, R2/
the description. )
-i and X are as defined in
BACKGROUND
One of pseudo-alkaloid compounds, colchicine has
an anti-inflammation action, making it a therapeutic
agent for rheumatoid arthritis [Internal Medicine, 86,
1
Mo.2, 342-345, 2000] . Colchicine and thiocolchicine
derivatives have functions of muscle relaxation and
anti-inflammation (USP 5 973 204, EP 0870761 Al) .
Thiocolchicoside has been used for the treatment of
contracture and inflammation in skeletal muscles. Also,
»
colchicine inhibits infiltration of monocytes and Tcells
in a transplanted organ in animal experiment and
at the same time restrains the production of TNF-a, IL-
1 and IL-6, inflammatory cytokines, suggesting an
inhibiting effect on immune response [ Nephrol., 4(6), 1294-1299, 1993; Transplantation
Proceedings, 32, 2091-2092, 2002] . Thus, colchicine is
very attractive candidate for the development of an
immune response inhibitor (WO 02/100824) .
Colchicine inhibits a inicrotubule assembly by the
interaction with tubulin, resulting in the suppression
of cell division [The Alkaloids, 1991, 41, 125-176; USP
4 533 675] . Such colchicine has been used for the
treatment of gout and other inflammatory diseases
related to gout. However, the • use of colchicine is
limited to an acute inflammatory .disease because of the
limitation in therapeutic index and toxicity to
gastrointestinal tract [Pharmacotherapy, 11, 3, 196-211,
1991].
All the endeavors to develop colchiciue
derivatives as an anbicancer drug have not been
successful so far [USP 3 222 253; USP 00/6080739; WO
97/01570] , and only demecolcine has been used for the
treatment of leukemia. However, toxicity to
gastrointestinal tract and limitation in therapeutic
index are still problems of demecolcine.
The present inventors have completed this
invention by developing colchicine derivatives having
excellent activities of anticancer, anti-proliferation
and angiogenesis inhibition that have now stable
therapeutic index resulted from decreased toxicity.
SUMMARY OF THE INVENTION
It is an object of the present invention to
provide tricyclic derivatives or pharmaceutically
acceptable salts thereof having excellent activities of
anti-cancer, anti-proliferation and angiogenesis
inhibition with stable therapeutic index by reduced
toxicity.
It is also an object of this invention to provide
a preparation method for- tricyclic derivatives or
pharmaceutically acceptable salts thereof.
It is a further object of this invention to
provide a pharmaceutical composition containing
tricyclic derivatives or pharmaceutically acceptable
salts thereof as an effective ingredient.
BRIEF DESCRIPTION OF THE DRAWINGS
PIG. 1 is a graph showing the changes of the
volume of a tumor in a BALB/c nude mouse transplanted
with human lung cancer cell line NCI-H460 after the
administration of tricyclic derivatives of the present
invention (Example 8),
FIG. 2 is a graph showing the changes of the body
weight of a BALB/c nude mouse transplanted with human
lung cancer cell line NCI-H4GO after the administration
of tricyclic derivatives of the present invention
(Example 8),
FIG. 3 is a graph showing the changes of the
volume of a tumor in a BALB/c nude mouse transplanted
with human lung cancer cell line NCI-H460 after the
administration of tricyclic derivatives of the present
invention (Example 12) by different concentrations (1,
3, 10 ing/kg) ,
v. FIG. 4 is a graph showing the changes of the body
#
weight of a BALB/c nude mouse transplanted with human
lung cancer cell line NCI-H460 after the administration
of tricyclic derivatives of the present invention
(Example 12) by different concentrations (1, 3, 10
rag/kg) ,
FIG. 5 is a set of photographs showing the volume
of a tumor growing in a BALB/c nude mouse transplanted
with human lung cancer cell line NCI-H460, which was
separated on the 14th day after the administration of
tricyclic derivatives of the present invention,
PIG. 6 is a set. of photographs showing the
activity of tricyclic derivatives of the present
invention to inhibit angiogenesis in HUVEC cells.
DETAILED DESCRIPTION OF PREFERRED EMBODIMENTS
The present invention relates to tricyclic
derivatives represented by following , or
pharmaceutically acceptable salts '.thereof.

(Wherein,
(1) R1 is -T1-B1;
in which T1 is -X1-, -X1-C(X2)-, -N(R5)-/ -N(R5)C (X3) -, -
N(R5)S(0)n1-, -N(R5)C(0) -X1- or -N(RS) C (X1)NH-, in that
X1 and X2 are each 0 or S, R5 is each H or C1-C5 alkyl
group, nx is an integer of 1-2; and Bx is selected from
a group consisting of following (a) ~ (j) ,
(a)
&
(CH2)n3-R7
— (CH2)n2

~(
CHj)n2
-ZfR7
(b)
(c)
(CH2)n3-T2-B2
-CCH-CH—o)n4-R7 (d)
Z2 Z3
-(CH2)n5-CH-(CH2)n6-R7
^(
CH2)n2
ZfT2-B2
—(CH-CH-0)n4-T2-B2
(g)
(h)
z, z3
~-(CII2)n5-CII-(CH2)n6-T2-B2
Wherein, Rs and Ra are each H, halogen, hydroxy,
Ci~C3 alkoxy, amino, nitro, cyano or Ci^Ca lower alkyl
group; R7 and R9 are each independently halogen, hydroxy,
mercapto, -ONO, -ON02 or SNO, in which R7 and R9 are
same or different;
is C5-C6 membered saturated or
unsaturated heterocyclic ring containing 1~2 of hetero
atom, in which the hetero atom is selected from a group
S and N, preferably,
v vj -v~ ~v Y Y
Cl C! C3 01 cs C6
/r-i N
H
C7
_jr-N
^ ~y~
H
Cti
/ — ^
fi
C9
/— NH
fi
CIO Cll C12
more preferably, Cl (pyridyl group) substituted at
position 2 and 6 or position 2 and 5, C7 (pyrrolyl
group) substituted at position 2 and 5 or position 2
and 4, Cll (thiophenyl group) or C12 (furanyl group) ;
Z1 is C1 - C10 straight -chain or branched-chain alkyl
group, preferably C2-Cs straight-chain, or branchedchain
alkyl group or cycloalkyl group having
substituent; Z2 and Z3 are each independently H or
methyl group, in which Z3 is H when Z2 is methyl group,
Z2 is H when Z3 is methyl group; T2 is -X1- or -X1-C(X2)-,
in that X1 and X2 are each independently O or S; B2 is
selected from a group consisting of said (a) , (b) , (c) ,
(d) or (e) ; n2 is an integer of 0-3, n3 is an integer of
0~5, n4 is an integer of 1-5, ns and n6 are each
independently an integer of 1-6;
(2) R2 and R3 are each independently H, -P03H2,
phosphonate, sulfate, C3~C7 cycloalkyl, C2 ~ C7 alkenyl,
C2~C7 alkynyl,, Cx ~ C7 alkanoyl , GI ~ C7 straight-chain or
branched-chain alkyl or sugar, in which sugar is a
8
monosaccharide such as glucuronyl, glucosyl or
galactosyl;
(3) R4 is OCH3, SCH3 or NR10R11, in which R10 and R1X
are each independently H or C1-5 alkyl;
(4) X is 0 or S.)
Preferably in the compound of ,
(1) R1 is -T1-B1;
in which TO. is -N (R5) C (X2) - , -N(RS) C (0) -Xx- or -
N(R5)C(X1)NH-, in that X1 and X2 are each O, R5 is each
H or C1~C5 alkyl group; and B1 is selected from a group
consisting of following (a) ~ (j) ,
(a)
(Cl I2)n3-T2-B2
— (CH2)n2
(CH2)n3-R7
CH2)n2
-ZfR7
(b)
(c)
-(CH-CH-0)n4-R7 (d)
22 Z3
(CH2)n3-T2-B2
(8)
CH2)n2
ZfT2-B2
-(CH-CH—O)n4-T2-B2
Z2 Z3
00
-(CH2)n5— CH-(CH2)n6-R7 (CH2)n5—CH— (CH2)n6-T2-B2
Wherein, R6 and RB are each H, halogen, hydroxy,
C1~C3 alkoxy, amino, nitro, cyano or C1~C3 lower alkyl
group; R7 and R9 are each independently halogen, hydroxy,
mercapto(thiol) , -ONO, -ON02 or SNO, in which R7 and R9
are same or different}
is C5-C6 rnembeied saturated or
unsaturated heterocyclic ring containing 1-2 of hetero
atom, in which the hetero atom is selected from a group
consisting of 0, S and N, preferably, •
N N ^N'
Cl C2 C3
/r-N ,—v
V*
C4
/— NH V
~V V
H H
C5 C6
/ ^ "V" "V
C7 C8 C9 CIO Cll C12
more preferably, Cl (pyridyl group) substituted at
position 2 and 6 or position 2 and 5, C7 (pyrrolyl
group) substituted at position 2 and 5 or position 2
and 4, Cll (thiophenyl group) or C12 (furanyl group),a
bond of substituents may be at symmetrical or
asymmetrical position; Za is C1~C10 straight-chain or
branched-chain alkyl group, preferably C2-CS straightchain
or branched-chain alkyl group or cycloalkyl group
10
having substituent; Z2 and Z3 are each independently H
or methyl group, in which Z3 is H when Z2 is methyl
group, Z2 is H when Z3 is methyl group; T2 is -X1- or -
V
X1-C(X2)-, in that X1 and X2 are each O or S; B2 is
selected from a group consisting of said (a), (b), (c),
(d) or (e) ; n2 is an integer of 0-3,. n3 is an integer of
0-5, n4 is an integer of l~3, ns and n6 are each
independently an integer of 1-3;
(2) R2 and R3 are each independently C3 ~ C7
cycloalkyl or C1~C7 alkyl;
(3) R4 is SCH3 or OCH3;
(4) X is 0 or S.
Preferably, the compounds of
comprise:
1) 6-nitrooxymethyl-N-[(7S)-1,2,3-trimethoxy-10-
methylsulfanyl-9-oxo-5,6,7,9-tetrahydrobenzo[
a]heptalen -7-yl]-nicotineamide; "
2) 5-nitrooxymethyl-furan-2-carboxylic acid-
[ (7S) -l^^-trimethoxy-lO-methylsulfanyl-g-oxo-SfS,?, 9-
tetrahydro-benzo[a]heptalen-7-yl]-amide;
3) N-[(7S)-3-isopropoxy-l,2-dimethoxy-10-methylsulfanyl-
9-oxo-5, 6,7, 9-tetrahydro-benzo [a] heptalen-7-
yl] -3-nitrooxymethyl-benzamide;
11
4) N-[(7S)-3-ethoxy-l,2-dimethoxy-10-methylaulfanyl-
9-oxo-5,6,7,9-tetrahydro-benzo[a]heptalen-7-
yl]-3-nitrooxymethyl-benzamide;
5) 6-nitrooxymethyl-pyridine-2-carboxylic acid-
[ (7S) -1, 2 , 3-trimethoxy-10~rnethylsulf anyl-9-oxo-5, 6,7,9-
*
tetrahydro-benzo[a]heptalen-7-yl]-amide;
6) 5-nitrooxymethyl-thiophene-2-carboxylic acid-
[(7S)-l,2,3-trimethoxy-10-methylsulfanyl-9-oxo-5,6,7,9-
tetrahydro-benzo[a]heptalen-7-yl] -amide;
7) N- [ (7S) -3-cycloperityloxy-l, 2-dimethoxy-10-
methylsulfanyl-9-oxo-5,6,7,9-tetrahydrobenzo
[a] heptalen -7-yl]-3-nitrooxymethyl-benzamide;
8) N-[(73)-3-ethoxy-l,2-dimethoxy-10-
methylsulfanyl-9-oxo-5,6,7,9-tetrahydrobenzo
[a]heptalen-7-yl]-2-fluoro-3-nitrooxymethylbenzamide;
9) 2-fluoro-N-[(7S)-3-isopropoxy-l,2-dimethoxy-
10-methylsulfanyl-9-oxo-5,6,7, 9-tetrahydrobenzo
[a]heptalen-7-yl]-3-nitrooxymethyl-benzamide;
10) 2-fluoro-3-nitrooxymethyl-N-[(7S)-1,2,3-
trimethoxy-10-methylsulfanyl-9-oxo-5,6,7,9-tetrahydrobenzo
[a] heptalen-7-yl ] -benzatnide ;
11) N-[(7S)-3-cyclopentyloxy-l,2-dimethoxy-10-
methylsulfanyl-9-oxo-5,6,7,9-tetrahydro-
12
benzo [a]heptalen-7-yl] -2-fluoro-3-nitrooxytnethylbensamide
,-
12) 3-fluoro-5-nitrooxymethyl-N-[(7S)-1,2,3-
trimethoxy-10-methylsulfanyl-9-oxo-5,6,7,9-tetrahydrobenzo[
a]heptalen-7-yl]-benzamide ;•
13) N- [ ( I S ) -3-ethoxy-l,2-ditnethoxy-10-
methylsulfanyl-9-oxo-5, 6,7,9-tetrahydrobenzo[
a]heptalen-7-yl]-3-fluoro-5-nitrooxymethylbenzamide;
14) 3-fluoro-N-[(7S) -3-isopropoxy-l,2-dimethoxy-
10-methylsulfanyl-9-oxo-5, 6,7, 9-tetrahydrobenzo
[a] heptalen-7-yl] -5-nitrooxymethyl-benzamide;
15) N- [ (7S) -3-cyclopentyloxy-l, 2-dimethoxy-10-
methylsulfanyl-9-oxo-5,6,7,9-tetrahydrobenzo[
a]heptalen-7-yl] -3-fluoro-5-nitrooxymethylbenzamide
;
16) 4-fluoro-3-nitrooxymethyl-N-[(7S)-1,2,3-
trimethoxy-10-methylsulfanyl-9-oxo-5,6,7,9-tetrahydrobenzo[
a]heptalen-7-yl]-benzamide;
17) 2-fluoro-5-nitrooxymethyl-N-[(7S)-1,2,3-
trimethoxy-10-methylsulfanyl-9-oxo-5,6,7,9-tetrahydrobenzo[
a]heptalen-7-yl]-benzamide;
18) 3-hydroxy-5-nitrooxymethyl-N-[(7S)-1,2,3-
trimethoxy-10-methylsulfanyl-9-oxo-5,6,7,9-tetrahydrobenzo[
a]heptalen-7-yl]-benzamide;
13
19) 3,5-Jbis-nitrooxymethyl-N- [ (IS) -1,2,3-
trimethoxy-10-methylsulfanyl-9-oxo-5,6,7,9-tetrahydrobenzo[
a]heptalen-7-yl]-benzamide;
20) 2-hydroxy-4-nitrooxymethyl-N- [ (IS)-1,2,3-
trimethoxy-10-methylsulfanyl-9-oxo-5,*6,7, 9-tetrahydro-
*
benzo[a]heptalen-7-yl]-benzamide;
21) 4-nitrooxymethyl-thiophene-2-carboxylic acid
[(IS)-l,2,3-trimethpxy-10-methylsulfanyl-9-oxo-5,6,7,2-
tetrahydro-benzo[a]heptalen-7-yl]-amide;
22) 3-nitrooxymethyl-thiophene-2-carboxylic acid
[(IS)-1,2,3-trimethoxy-10-methylsulfanyl-9-oxo-5,6,7,9-
tetrahydro-benzo[a]heptalen-7-yl]-amide;
23) 2-(3-nitrooxymethyl-phenyl)~N- [ (7S)-1,2,3-
trimethoxy-10-methylsulfanyl-9-oxo-5,6,7,9-tetrahydrobenzo[
a]heptalen-7-yl]-acetamide;
24) 3-(2-nitrooxy-ethyl)-N-[(7S)-1,2,3-
trimethoxy-10-methylsulfanyl-9-oxo-5,6,7,9-tetrahydrobenzo[
a]heptalen-7-yl]-benzamide;
25) 3-nitrooxybenzoic acid-5-[(7S)-1,2,3-
tritnethoxy-10-methylsulfanyl-9-oxo-5, 6,7, 9-tetrahydrobenzo[
a]heptalen-7-yl-carbamoyl]-pyridine-2-ylmethylester;
;
26) 4-nitrooxybutyric acid-5-[ (7S)-1,2,3-
trimethoxy-10-methylsulfanyl-9-oxo-5,6,7,9-tetrahydro-
14
benzo [a]heptalen-7-yl-carbamoyl]-pyridine-2-ylmethylester;
27) 3-nitrooxymethyl-benzole acid-6-[(7S)-1,2,3-
trimethoxy-10-methylsulfanyl-9-oxo-5,6,7,9-tetrahydrobenzo[
a]heptalen-7-yl-carbamoyl]-pyridine-2-yl-
»
methylester;
28) 4-nitrooxybutyric acid-6-[(7S)-1,2,3-
trimethoxy-10-methylsulfanyl-9-oxo-5,6,7,9-tetrahydrobenzo[
a]heptalen-7-yl-carbamoyl]-pyridine-2-yltnethylester;
29) 3-nitrooxymethyl-benzole acid-2-[(7S)-1,2,3-
trimethoxy-10-methylsulfanyl-9-oxo-5,6,7,9-tetrahydrobenzo[
a]heptalen-7-yl-carbamoyl]-phenylester;
30) 4-nitrooxybutyric acid-2-[(7S)-1,2,3-
trimethoxy-10-methylsulfanyl-9-oxo-5,6,7,9-tetrahydrobenzo[
a]heptalen-7-yl-carbamoyl]-phenylester;
31) 3-nitrooxymethyl-benzole acid-3-[(7S)-1,2,3-
trimethoxy-10-methylsulfanyl-9-oxo-5,6,7,9-tetrahydrobenzo
[a]heptalen-7-yl-carbamoyl]-phenylester;
32) 4-nitrooxybutyric acid-3-[(7S)-1,2,3-
trimethoxy-10-methylsulfanyl-9-oxo-5,6,7,9-tetrahydrobenzo[
a]heptalen-7-yl-carbamoyl]-phenyleater;
33) 3-nitrooxymethyl-benzoic acid-3-[(7S)-1,2,3-
trimethoxy-10-methylsulfanyl-9-oxo-5,6,7,9-tetrahydrobenzo[
a]heptalen-7-yl-carbamoyl]-benzylester;
15
34) 4-nitrooxybutyric acid-3-[(7S)-1,2,3-
trimethoxy-10-methylsulfanyl-9-oxo-5,6,7, 9-tetrahydrobenzo
[a]heptalen-7-yl-carbamoyl] -benzylester;
35) 2-nitrosothio-N-[(7S)-1,2,3-trimethoxy-10-
Tnethylsulfanyl-9-oxo-5, 6 , 7 , 9-tetrahydro- " '

benzo[a]heptalen-7-yl]-benzamide;
36) 3-nitrosooxymethyl-N-[(7S)-1,2,3-trimethoxy-
10-methylsulfanyl-9-oxo-5,6,7,9-tetrahydrobenzo[
a]heptalen-7-yl]-benzamide;
37) 3-fluoro-5-nitrosooxymethyl-N-[(73)-1,2,3-
trimethoxy-10-methylsulfanyl-9-oxo-5,6,7,9-tetrahydrobenzo[
a]heptalen-7-yl]-benzamide;
38) 3-nitroaothiomethyl-N-[(7S)-1,2,3-trimethoxy-
10-methylsulfanyl-9-oxo-5,6,7,9-tetrahydrobenzo
[a] heptalen-7 -yl] -beirzam.ide ;
39) 3-fluoro-5-nitrosothiomethyl-N- [ (7S) -1,2,3;-
triraethoxy-10-methylsulfanyl-9-oxo-5,6,7,9-tetrahydrobenzo[
a]heptalen-7-yl]-benzamide;
. 40) 3-fluoro-5-nitrooxymethyl-N-[(7S)-1,2,3,10-
tetramethoxy-9-oxo-5,6,7,9-tetrahydro-benzo[a]heptalen-
7-yl] -benzamide;
41) 3-nitrooxymethyl-N-methyl-N- [(7S)-1,2,3-
trimethoxy-10-methylsulfanyl-9-oxo-5,6,7,9-tetrahydrobenzo
[a] heptalen-7-yl] -benzamide;
16
42) 3-fluoro-N-methyl-5-nitrooxymethyl-N-[(7S)-
1,2,3-trimethoxy-10-methylsulfanyl-9-oxo-5,6,7,9-
tetrahydro-benzo[a]heptalen-7-yl]-benzamide;
43) 2- (3-fluoro-5-nitrooxymethyl-phenyl)-N-[(7S)-
1,2, 3 -trimethoxy-10-methylsulf anyl -9 -oxo-5,6,7,9-
*
tetrahydro-benzo[a]heptalen-7-yl]-acetamide; or
44) 2- (2-fluoro-5-nitrooxymethyl-phenyl)-N-[(7S)-
l,2,3-trimethoxy-10-methylsulfanyl-9-oxo-5,6, 7,9-
tetrahydro-benzo[a]heptalen-7-yl]-acetamide.
The present invention also provides
pharmaceutically acceptable salts of the compound
represented by . Pharmaceutically acceptable
salts of the present invention can include acid
addition salt of a compound according to the invention
when the compound is fully basic. Such acid addition
salt includes salts holding inorganic acid providing
pharmaceutically acceptable anion such as hydrogen
halide, or organic acid, or salts holding sulfuric acid
or phosphoric acid, or salts holding trifluoroacetic
acid, citric acid or maleic acid. And, it include for
example hydrochlorides, hydrobromides, phosphonates,
sulfates, alkylsulfonates, arylsulfonates, acetates,
benzoates, citrates, maleates, fumarates, succinates,
lactates and tartarates by suitable salts. When a
17
compound of the present invention . is fully acidic,
pharmaceutically acceptable salts can include inorganic
salts or organic salts providing pharmaceutically
acceptable cation. Said inorganic salts include sodium
salts, potassium salts, calcium salts or magnesium
salts, etc., said organic salts include methylamine
salts, dimethylamine salts, trimethylamine salts,
piperidine salts or morpholine salts, etc.
The present invention also provides a preparation
method for tricyciic derivatives represented by the
. The preparation method for tricyclic
derivatives of the present invention is described in
the below Scheme 1 ~ Scheme 8. Precisely, in the
, when RX is -T1-B1 and BI is one. of said (a) ,
(b) , (c) , (d) and (e), the derivatives are prepared
according to the method of Scheme 1 ~ Scheme 6. In the
meantime, in the , when Rx is -T1-B1 and Bj.
is one of said (f) , (g) , (h) , (i) and (j), the
derivatives are prepared by the method of Scheme 7 and
Scheme 8. And a concrete compound of the
is represented by general formulas (IIa), (IIb), (IIe),
(IId), (IIe), (IIf), (IIg), (IIh), (IIi), (II j), (IIk),
(II1) , (IIm) , (IIn) , (IIo) and (IIp) in Scheme 1 ~
Scheme 8.
18
VIII
,ON(0)n7
lla

Hal—Y-OH
XI
XII
RS
,^SNOn
d

19
D-X2H
Hal
XIV
VI1
,OP
XVII
rv 1 JL
XVI
> D-A xvm
,JCON(0)n:
lie
JL.
XIX
Xv:; ^vY'SNO
nr

D-NHR5
m
XX
(0)n,
II
XXI
(0)ns
•^'S"Y ON(0)n7

(OK
xxn IIli
20
D-NHR,
0 nrV OP
ra ii) deprotecting
o
D-NAxr
XXIV
XXV
o
Hi
Uj

m ii) deprotecting
xxvn
R5 xxvm
1
Ilk
ni

21
XXXIII
•'?«
j- Y'^°
Ilk

In the above Scheme, E is following El ~ E6
respectively;
22
x
El E2 E3
(O)n7 O ?M
-N Y ~N X, ^ N
E4 E5 E6
Wherein, Xi( X2 and X3 are each 0 or S.
In the above Scheme, D i s , a n d R2,R3, R4
and X are same as defined in the ;
R5 is H or lower alkyl; X1, X2 and X3 are each
independently 0 or S; Hal1 and Hal2 are halogens; Hal1
and Hal2 of general formula (IV) and (IX) are each same
or different halogens, for example F, Cl, Br or I; P is
conventional protecting group of hydroxy such as
methoxymethyl, t-butyldimethylsilyl or benzyl; Y and Y1
are same or different, and indicate following general
formula (a1), (b1); (c1), (d1) and (e1) respectively,
23
-(CH2)n2 (a1)
-(CH-CH—0)n4-
Z, Z3
(d')
zf~
(c1)
-(CH2)n5-CH~(CH2)n6-
(e')
Wherein, R6, R8, R9, Z1, Z2, Z3, n2, n3,
n4, n5 and n6 are same as defined in the , n?
and na are integers of 1-2.
The preparation method for tricyclic derivatives
of the present invention is illustrated more precisely
hereinafter.

According to method 1 of the present invention
for the preparation of compounds of formula (lla) and
(l1b), the compound represented by formula (V) is
prepared by amidation reaction making amine compound of
formula (III) be reacted with halogen compound of
formula (IV), which is step 1. In step 1, a base might
be excluded, but the reaction is generally performed
with a solvent such as dichloromethane, chloroform,
24
tetrahydrofuran, diethylether, toluene or
dimethylformamide etc., which have no influence on
amidation reaction, in the presence of pyridine,
triethylamine, diethylisopropylamine or Nmethylmorpholine
etc., a base that can be acceptable
»
for amidation reaction in general. Reaction temperature
is not limited in particular, but generally reaction
can performed under cold temperature or elevated
temperature, is performed preferably at room
temperature.
In step 2, conversion of the compound of formula
(V) prepared in the above step 1 to nitrooxy compound
(n7 = 2) of formula (IIa) and to nitrosooxy compound
(n7 = 1 ) of formula (IIa) was accomplished by nitration
reaction and nitrosation reaction, respectively.
Nitration reaction needs a compound that is able to
convert halogen into nitrate, arid is performed using
silver nitrate (AgN03) , t-butylammonium nitrate
(Bu4NN03) , etc., in the presence of chloroform,
acetonitrile, a mixture of acetonit.rile and aqueous
solution, or dichloromethane, which are all solvents
not affecting the reaction. Nitrosation reaction might
use a compound that is able to convert halogen into
nitrosate, too, and is performed preferably using
silver nitrite (AgN02) or sodium nitrite (NaN02) in the
25
presence of chloroform, acetonitrile, a mixture of
acetonitrile and aqueous solution, aqueous solution, or
dichloromethane, which are also solvents not affecting
the reaction. Reaction temperature is not limited in
particular, but generally reaction can performed under
»
cold temperature or elevated temperature, is performed
preferably at room temperature.
Another way to give the compound of formula (IIa)
is as follows; reaction of the compound of formula
(III) with the compound of formula (VI) is performed to
give the compound of formula (VII) , and then,
conversion of the compound of formula (VII) to the
compound of formula (Ila) is accomplished. The
reaction of the compound of formula (III) and the
compound of formula (VI) is performed in the presence
of a coupling agent such as 1-(3-ditnethylaminopropyl) -
3-ethylcarbodiimide(EDGI), 1-hydroxybenzotriazole
hydrate(HOBT) or 1,3-dicyclohexyl carbodiimide(DCC).
This reaction might be performed without a base, but
generally with a base such as 4-dimethylaminopyridine,
pyridine, triethylamine, diethylisopropylamine, Nmethylmorpholine
or dimethylphenylamine etc., which can
be used in amidation reaction, in a solvent having no
negative effect on the reaction, for example
acetonitrile, dimethylformamide, dichloromethane, etc.
26
Reaction temperature is not limited in particular, but
generally reaction can performed under cold temperature
or elevated temperature, is performed preferably at
room temperature. Direct conversion of the compound of
formula (VII) to the compound of formula (Ila) is
»
accomplished by the reaction of alcohol with
triphenylphosphin (PPh3) , N-bromosuccineimide (NBS) and
silver nitrate, or silver nitrite. The reaction is
performed in a solvent having no effect on the reaction
such as chloroform, acetonitrile, dichloromethane, a
mixture of acetonitrile and dichloromethane, etc.
Reaction temperature is not limited in particular, but
generally reaction is performed under cold temperature
or at room temperature. Another way for conversion of
the compound of formula (VII) to the compound of
formula (Ila) is as follows; conversion of the compound
of formula (VII) to halogen compound of formula (V) is
accomplished first, and then conversion thereof to the
compound of formula (Ila) is accomplished again. At
this time, the conversion into halogen compound is
performed by using a reagent that generally converts
hydroxy group to halogen, for example tribromophosphin,
tetrabromomethane etc., in the presence of chloroform*
acetonitrile, dichloromethane etc., which are solvents
having no negative effect on the reaction. Reaction
27
temperature is not limited in particular, but generally
reaction is performed under cold temperature or at room
temperature.
Processes for preparing the compound of formula
(IIb) of the method 1 of the present invention are as
follows; conversion of hydrogen included in alcohol of
formula (VII) to a leaving group such as mesylate,
tosylate or triplate is accomplished, followed by
reaction with potassium thioacetate, to give
thioacetate ester compound. Hydrolysis of the compound
in the presence of a base is accomplished to give the
compound of formula (VIII) . At this time, a base is
selected among general bases that are able to hydrolyze
an ester compound, for example sodium hydroxide,
potassium hydroxide or sodium thiomethoxide. And an
alcohol solution such as methanol or ethanol is
preferred as a solvent for the reaction. Reaction
temperature is not limited in particular, but generally
reaction can performed under cold temperature or
elevated temperature, is performed preferably at room
temperature. Reaction of the compound of formula (VIII)
with sodium nitrite under an acidic condition, leads to
the conversion of the compound to nitrosothio compound
of formula (IIb) . A solvent for the reaction is
selected from a group consisting of methanol, ethanol,
28
acetonitrile, a mixture of acetonitrile and aqueous
solution, or dichloromethane etc., which is not to
affect the reaction. Reaction temperature is not
limited in particular, but generally reaction can
performed under cold temperature "or elevated
»
temperature, is performed preferably at room
temperature.

According to method 2 of the present invention,
compounds of formula (IIe) and (IId) are prepared.
Particularly, in step 1, reaction of the compound of
formula (III) with the compound of formula (IX) is
accomplished to give the compound of formula (X) . This
reaction is performed in analogy to the procedure
described in method 1 in which conversion of the
compound of formula (III) to the compound of formula
(V) was accomplished by amidation reaction.
In step 2, conversion of the compound of formula
(X) prepared in step 1 to the compound of formula (IIe)
is accomplished by nitration reaction along with
nitrosation reaction. This reaction is performed in
analogy to the procedure described in method 1 in which
conversion of the compound of formula (V) to the
compound of formula (IIa) was accomplished.
29
Another way to give the compound of formula (IIe)
is as follows; reaction of the compound of formula
(III) with the compound of formula (XI.) is performed to
give the compound of formula (XII) , and then,
conversion of . the compound of formula (XII) to the
compound of formula (IIe) is accomplished. Reaction of
the compound of formula (XI) with the compound of
formula (III) is performed in analogy to the procedure
described in method 1 in which conversion of the
compound of formula (III) to the compound of formula
(V) was accomplished by amidation reaction. Conversion
of the compound of formula (XII) to the compound of
formula (IIe) is accomplished under the same condition
as provided for the conversion of the compound of
formula (VII) to the compound of formula (IIa) in
method 1.
Conversion of the compound of formula (XII) to
the compound of formula (IId) is accomplished under the
same condition as provided for the conversion of the
compound of formula (VII) to the compound of formula
(IIb) in method 1.

According to method 3 of the present invention,
compounds of formula (IIe) and (IIf) are prepared.
30
Particularly, in step 1, reaction of the compound of
formula (XIV) with the compound of formula (IV) is
accomplished to give the compound of formula (XV). The
reaction in this method is esterification reaction of
.alcohol (X2=0) or thioalcohol (X2=S) with acyl or
thioacyl halide, which is performed in the presence of
a base that is generally acceptable for esterification
reaction. Preferable bases are pyridine, 4-
dimethylaminopyridine, triethylamine,
diethylisopropylamine, 2,6-lutidine, sodium hydride
(NaH) , cesium carbonate, or sodium hydroxide and can be
used along with a phase transfer catalyst such as
benzyltriethylammoniumchloride. Also, above reaction is
preferably performed in a solvent having no negative
effect on the reaction, for example dichloromethane,
chloroform, tetrahydrofuran, diethylether, toluene,
dimethylformamide, acetonitrile or aqueous solution.
Reaction temperature is not limited in particular, but
generally reaction can performed under cold temperature.
or elevated temperature, is performed preferably at
room temperature.
In step 2, conversion of the compound of formula
(XV) prepared in step 1 to the compound of formula
(IIe) is accomplished by nitration reaction along with
nitrosation reaction. This reaction is performed in
31
analogy to the procedure described in method 1 in which
conversion of the compound of formula (V) to the
compound of formula (IIa) was accomplished.
Another way to give the compound of formula (IIe)
is as follows; reaction of the compound of formula
(XIV) with the compound of formula (VI} having a
protective group in alcohol group is performed to give
the compound of formula (XVII), followed by
deprotection reaction to give the compound of formula
(XVIII). Conversion of the resultant compound to the
compound of formula (lie) is accomplished. The
reaction of the compound of formula (XIV) with the
compound of formula (VI') is processed by
esterification reaction of alcohol (X2=O) or
thioalcohol (X2=S) and carboxylic acid or
thiocarboxylic acid. The reaction is performed either
in an aqueous solution supplemented with an acid such
as hydrochloric acid, sulfuric acid, dodecylbenzene
sulfonic acid or p-toluenesulfonic acid, at room
temperature or under elevated temperature, or under the
same condition provided for conversion of the compound
of formula (III) to the compound of formula (VII) in
method 1. Another esterification reaction is performed
by Misunobu reaction using triphenylphosphine and
diethyl azodicarboxylate in a solvent not affecting the
32
reaction. And the solvent is preferably selected from
a group consisting of dichloromethane, chloroform,
tetrahydrofuran, diethylether, toluene or acetonitrile.
Reaction temperature is not limited in particular, but
general-ly reaction is performed under cold temperature
or at room temperature. Protecting and deprotecting
reaction of alcohol group is performed by known method
in general organic synthesis.
Reaction of the compound of formula (XIV) with
the compound of formula (XVI) is performed to give the
compound of formula (XVIII) in analogy to the procedure
described in method 3 in which conversion of the
compound of formula (XIV) to the compound of formula
(XV) was accomplished.
Conversion of the compound of formula (XVIII) to
the compound of formula (Ie) is performed under the
same condition provided for conversion of the compound
of formula (VII) to the compound ' of formula (IIa) in
method 1.
Conversion of the compound of formula (XVIII) to
the compound of formula (IIf) is performed under the
same condition provided for conversion, of the compound
of formula (VII) to the compound of formula (IIb) in
method 1.
33

According to method 4 of the present invention,
compounds of formula (IIg) and (IIh) are prepared.
Particularly, in step 1, reaction of the compound of
formula (III) with the compound of formula (XX) is
accomplished to give the compound of formula (XXI).
When the compound of formula (IIg) is
sulfinylamide (na=l) , the reaction of the compound of
formula (III) with sulfinylhalide of formula (XX) is
performed without a base or with a base that is
applicable to amidation reaction, for example pyridine,
triethylamine, diethylisopropylamine, Nmethylmorpholine
or dimethylphenylamine, in a solvent
having no negative effect on the reaction such ns
dichloromethane, chloroform, tetrahydrofuran,
diethylether, toluene or dimethylformamide. Reaction
temperature is not limited in particular, but generally
reaction can performed under cold temperature or
elevated temperature, is performed preferably at room
temperature.
When the compound of formula (Ilg) is
sulfonylamide (nB=2) , the reaction of the compound of
formula (III) with sulfonylhalide of formula (XX) is
performed either without a base or with a base that is
applicable to amidation reaction in general, for
34
example pyridine, triethylamine, diethylisopropylamine,
N-methylmorpholine, sodium hydroxide, sodium carbonate
or potassium carbonate, in a solvent having no negative
effect on the reaction such as dichloromethane,
chloroform, tstrahydrofuran, diethylether, toluene or
dimethylformaraide. Reaction temperature is not limited
in particular, but generally reaction can performed
under cold temperature or elevated temperature, is
performed preferably at room temperature.
In step 2, conversion of the compound of formula
(XXI) prepared in step 1 to the compound of formula
(IIg) is performed under the same condition provided
for conversion of the compound of formula (VII) to the
• compound of formula (IIa) in method 1.
Conversion of the compound of formula (XXI) to
the compound of formula (IIh) is performed under the
same condition provided for conversion of the compound
of formula (VII) to the compound of formula (IIb) in
method l.

According to method 5 of the present invention,
compounds of formula (Hi) and formula (IIj) are
prepared as follows. In step 1, reaction of the
compound of formula (III) with the compound of formula
35
(XXIII) having a protecting group to alcohol group is
performed, followed by deprotection reaction.
Conversion of the compound of formula (XXIV) resulted
from the above reaction to the compound of formula
(Hi) is accomplished. At this time, the reaction of
the compound of formula (III) with the compound of
formula (XXIII) is performed by using a coupling
reagent such as carbonyl dichloride, triphosgen, di-tbutyl
dicarbonate or 1,1'-carbonyl diimidazole etc.
The reaction can be performed either without a base or
with a base that is generally acceptable for amidation
reaction, for example pyridine, triethylamine,
diethylisopropylamine, N-methylmorpholine or
dimethylphenylamine, in a solvent having no negative
effect on the reaction such as dichloromethane,
chloroform, tetrahydrofuran, diethylether, ethanol or
dimethylformamide. Reaction temperature is not limited
in particular, but generally reaction is performed
under cold temperature or at room temperature. And
deprotection reaction is performed by known method in
general organic synthesis.
In step 2, conversion of the compound of formula
(XXIV) prepared in step 1 to the compound of formula
(Hi) is performed under the same condition provided
36
for conversion of the compound of formula (VII) to the
compound o£ formula (IIa) in method 1.
Conversion of the compound of formula (XXIV) to
the compound of formula (IIj) is performed under the
same condition provided for conversion of the compound
of formula (VII) to the compound of formula (IIb) in
method 1.

According to method 6 of the present invention,
compounds of formula (IIk) and formula (II1) are
prepared as follows. In step 1, reaction of the
compound of formula (III) with the compound of formula
(XXVI) having a protecting group to alcohol group is
performed, followed by deprotection reaction. Then,
conversion of the resultant compound of formula (XXVII)
to the compound of formula (IIk) is accomplished. At
this time, the reaction of the compound of formula
(III) with the compound of formula (XXVI) is performed
either without a base or with a base that is acceptable
for amidation reaction, for example pyridine,
triethylamine, diethylisopropylamine or Nmethylmorpholine
etc, in a solvent having no negative
effect on the reaction such as dichloromethane,
chloroform, tetrahydrofuran, diethylether, benzene,
37
acetonitrile, etc. Reaction temperature is not limited,
in particular, but generally reaction is performed
under cold temperature or at room temperature.
Protecting and deprotecting reaction of alcohol group
is performed by known method in general organic
synthesis.
In step 2, conversion of the compound of formula
(XXVII) prepared in step l to the compound of formula
(IIk) is performed under the same condition provided
for conversion of the compound of formula (VII) to the
compound of formula (IIa) in method 1.
Conversion of the compound of formula (XXVII) to
the compound of formula (II1) is performed under the
same condition provided for conversion of the compound
of formula (VII) to the compound of formula (IIb) in
method 1.

According to method 7 of the present invention,
compounds of formula (IIm) and formula (IIn) are
prepared from the compound of formula (XXIX) in analogy
to the procedure described in method 3.

38
According to method 8 of the present invention,
compounds of formula (IIo) and formula (IIp) are
prepared as follows. In step 1, reaction of the
compound of formula (XXIX) with the compound of formula
(IX) is performed to give a compound of formula
(XXXIV). This reaction is performed by etherification
reaction of alcohol (X2=0) or thioalcohol (X2=S) with
alkylhalide in the presence of a base acceptable for
etherification reaction. As a base suitable for that
purpose, sodium hydride (NaH), t-potassium butoxide (t-
BuOK), n-BuLi, sodium hydroxide, potassium hydroxide
and a phase transfer catalyst such as
benzyltriethylammoniumchloride etc, or crown ether is
preferred. The reaction is performed preferably in a
solvent having no negative effect on the reaction, for
example dichloromethane, chloroform, tetrahydrofuran,
diethylether, toluene, dimethylformamide, aqueous
solution, dimethylsufoxide or benzene, etc. Reaction
temperature is not limited in particular, but generally
reaction can performed under cold temperature or
elevated temperature, is performed under cold
temperature or at room temperature.
In step 2, conversion of the compound of formula
(XXXIV), prepared in the above step 1, to the compound
of formula (IIo) is accomplished by nitration reaction
39
or nitrosation reaction. This reaction is performed in
analogy to the procedure described in method 1 in which,
conversion of the compound of formula (V) to the
compound of formula (IIa) was accomplished.
Another way to prepare the compound of formula
(IIo) is as follows. Reaction of the compound of
formula (XXIX) with the compound of formula (XI')
having a protecting group to alcohol group is performed,
followed by deprotection reaction to give the compound
of. formula (XXXV) . Conversion of the compound of
formula (XXXV) to the compound of formula (IIo) is
accomplished. Reaction of the compound of formula
(XXIX) with the compound of formula (XI') is performed
under the same condition given for conversion of the
compound of formula (XXIX) to the compound of formula
(XXXIV) accomplished by etherification reaction in
method 8.
Conversion of the compound of formula (XXXV) to
the compound of formula (IIo) is performed under the
same condition provided for conversion of the compound
i
of formula (VII) to the compound of formula (Ila) in
method 1.
Conversion of the compound of formula (XXXV) to
the compound of formula (IIp) is performed under the
same condition provided for conversion of the compound
40
of formula (VII) to the compound of formula (IIb) in
method 1.
The target compounds given by the above reactions
can be separated and purified by general methods such
as column chromatography, recrystallisation, etc.
The present invention provides also a
pharmaceutical composition containing tricyclic
derivatives represented by the or
pharmaceutically acceptable salts thereof as an
effective ingredient.
Tricyclic derivatives according to the present
invention or pharmaceutically acceptable salts thereof
show very strong cytotoxicity to cancer cell lines but
have much less toxicity 'to test animals than colchicine
or taxol injection has.
When tricyclic derivatives of the present
invention were administered to a BALB/c nude mouse
transplanted with human lung cancer cell line NCI-H460,
\the size and the weight of a tumor were remarkably
decreased in proportion to the dosage.
Tricyclic derivatives of the present invention
also have a strong activity of antiangiogenesis in
HUVEC cells.
41
Therefore, tricyclic derivatives of the present
invention or pharmaceutically acceptable salts thereof
can be effectively used as an anticancer agent, an
anti-proliferation agent and an angiogenesis inhibitor.
The composition of the present invention might
additionally include, in addition to tricyclic
derivatives or pharmaceutically acceptable salts
thereof, at least one of active ingredients having the
same or similar function to the mentioned tricyclic
derivatives or pharmaceutically acceptable salts
thereof.
The said tricyclic derivatives or
pharmaceutically acceptable salts thereof can be
administered orally or parenterally and be prepared in
general forms of pharmaceutical formulation. The
tricyclic derivatives of the present invention or
pharmaceutically acceptable salts thereof can be
prepared for oral or parenteral administration by
mixing with generally used fillers, extenders, binders,
wetting agents, disintegrant, diluents such as
surfactants, or excipients. Solid formulations for
oral administration are tablets, pills, powders,
granules and capsules. These solid formulations are
prepared by mixing one or more suitable excipients such
42
as starch, calcium carbonate, sucrose, lactose and
galatin, etc. Except for the simple excipients,
lubricants, for example magnesium stearate, talc, etc,
can be used. Liquid formulations for oral
administrations are suspensions, solutions, emulsions
and syrups, and the above mentioned formulations can
contain various excipients such as wetting agents,
sweeteners, aromatics and preservatives in addition to
generally used simple diluents such as water and liquid
paraffin. Formulations for parenteral administration
are sterilized aqueous solutions, water-insoluble
excipients, suspensions, emulsions, lyophilized agent
and suppositories. Water insoluble excipients and
suspensions can contain, in addition to the active
compound or compounds, propylene glycol, polyethylene
glycol, vegetable oil like olive oil, injectable ester
like ethylolate, etc. Suppositories can contain
witepsol, macrogol, tween 61, cacao butter, laurin
butter, glycerol and gelatin.
The composition of the present invention can be
prepared for either oral or parenteral administration
(for example, intravenous, subcutaneous,
intraperitoneal or local injection), and dosage is
determined by weight, age, gender, condition of health
and diet of a patient and administration method,
43
excretion rate and severity of a disease. The
preferable effective dosage of the tricyclic
derivatives of the present invention is 3-300 nig/kg
(body weight), and administration times are once or
several times per day.
EXAMPLES
Practical and presently preferred embodiments of
the present invention are illustrative as shown in the
following Examples.
However, the present invention is not limited by
following examples.
7-Deacetylcolchicine used in the below examples
was prepared by the method described in [EP 0493064;
Synthetic Communications 1997, 27(2), 293-296].
7-Amino-l,2,3-trimethoxy-10-methylsulfanyl-6,7-
dihydro-5H-benzo[a]-heptalen-9-one was prepared by the
method described in (WO 9421598; Bioorganic & Medicinal
Chemistry, Vol 5, No. 12, pp 2277-2282, 1997).
Thiodemecolcine was prepared by the method
described in (J. Med. Chem, 1985, 28, 1204-1208).
(7S)-7-Amino-3-cyclopentyloxy-l,2-dimethoxy-10-
methylsulfanyl-6,7-dihydro-5H-benzo[a]heptalen-9-one,
44
(7S)-7-amino-3-isopropoxy-l,2-dimethoxy-lOmethylsulfanyl-
6,7-dihydro-5H-benzo[a]heptalen-9-one,
(7S)_7_amino-3-ethoxy-l,2-dimethoxy-10-methylsulfanyl-
6,7-dihydro-5H-benzo[a]heptalen-9-one were prepared by
the method described in (WO 9611184).
Example 1: Preparation of 6-n:.trooxymethyl-,N- [ (7S) -
l,2,3-trimethoxy-10-methyleulfanyl-9-oxo--5, 6,7,9-
tetrahydro-benzo[a]heptalen-7-yl]-nicotineamide
Preparation of 6-hydroxymethyl-Jr- [ (7S) -1,2,3-
tritnethoxy-10-tnethylsulfanyl-9-oxo-5, 6,7, 9-tetrahydrobenzo
[a]heptalen-7-yl3-nicotineamide
CO2H
EDCI, DMAP
MeCN
MeS ° MeS
6-Hydroxymethyl-nicotinic acid was synthesized by
the method described in (Bioorg. Med. Chem. Lett, 1996,
6, 3025-3028) .
45
To a solution of 7-amino-l,2,3-trimethoxy-10-
methylsulfanyl-6, 7-dihydro-5H-benzo [a] -heptalen-9-one
(300 mg, 0.80 tnmol) , 6-hydroxymethylnicotinic acid (135
mg, 0.88 tnmol) and DMAP (60 rag, 0.48 tnmol) in 10 mfi of
acetonitrile was added EDCI (308 mg, 1.60 mmol) at 0°C.
*
The reaction mixture was stirred at room temperature
for 2 hours. Water was added to quench the reaction,
and aqueous layer was extracted with ethyl acetate.
Combined organic layer was dried over anhydrous, sodium
sulfate, filtered and concentrated under reduced
pressure. The residue was purified by column
chroma tography (ethyl acetate: methanol = 8:1), to give
244 mg (yield: 60%, solid having yellow color) of the
target compound.
1H'NMR (400MHz, CDC13) : 52.07-2.15(m, 1H) , 2.31-
2.44(m, 2H) , 2.45(s, 3H) , 2.56-2.59(m, 1H) , 3.75(s, 3H)
3.91(s, 3H) , 3.97(s, 3H) , 4.66(q, J=10.2Hz, 2H) , 4.90-
4.93(m, 1H) , 6.56(s, 1H) , 7.13(t, J=9.1Hz, 2H) , 7.40(d,
J=10.2Hz, 1H) , 7.52(8, 1H) , B.15(dd, J=2.2, 5.8Hz,
8.80(d, J=6.9Hz, 1H) , 8.96(s,
46
Preparation of 6-nitrooxymethyl-JJ~ [ (7S) -1_, 2/3-
trimethoxy-10-methylsulf anyl-9-oxo-S ,6,7 , 9~tetrahydrobenzo[
a]heptalen-7-yl]-nicotineamide
MeQ
ON02
The compound (100 mg, 0.19 mmol) prepared in the
step 1 of Example 1 and triphenylphosphine (57 rag, 0.21
mmol) were dissolved in acetonitrile/dichloromethane
(1.25 ml/0.5 ml), therein NBS (42 mg, 0.23 mmol) was
added at -35°C. The reaction mixture was stirred for
20 minutes. Thereafter, therein silver nitrate (40 mg,
0.23 mmol) was slowly added dropwise at room
temperature , the reaction mixture was stirred at room
temperature for 18 hours. Water was added to quench
the reaction, and aqueous layer was extracted with
chloroform. Combined organic layer was dried over
anhydrous sodium sulfate, filtered and concentrated
under reduced pressure. The residue was purified by
column chroma tography ( chloroform rmethanol = 10:1), to
47
give 18 mg (yield: 35%, solid having yellow color) of
the target compound.
NMR (400MHz, CDC13): 52. 09-2 .13.(m, 1H) , 2.31-
2.43(m, 2H) , 2.46(s, 3H) , 2 .55-2 . 64 (m, 1H) , 3.75(s, 3H) ,
3.91(s, 3H) , 3.97(s, 311), 4.93-4.98 (m, *1H) , 5.50(s,
2H) , S.56(s, 1H) , 7.16(t, J=10.9Hz, 1H) , 7.26(d,
J=8.8Hz, 1H), 7.40(d, J=10.6Hz, 1H), 7.59(s,
8.27(dd, J=2.2, 5.8Hz, 1H), 8.77(d, J=7.3Hz,
9.08(s,
Example 2-4
Compounds of Example 2 - Example 4 were
synthesized in analogy to the procedure as described in
Example 1, and intermediates were prepared by the
method described as follows.
Preparation of 5-hydroxymethyl-furan-
2-carboxylic acid
5 r-Hydroxymethyl-f uran-2 -carboxylic acid was
synthesized by the method described in (Helv. Chim.
Acta, 1926, 9, 10S8) .
48
Preparation of 3-hydroxymethylbenzoic
acid
nw 2NNaOH
OH
EtOH
Isophthalic acid diethylester (9.100 g, 40.95
mmol) was dissolved in tetrahydrofuran (20ml).
Thereafter, therein lithiumborohydride. (11.26 ml, 22.52
mmol, 2M tetrahydrofuran solution) was slowly added
dropwise, and the reaction mixture was refluxed for 3
hours. Water was added to quench the reaction, and
aqueous layer was extracted with ethyl acetate.
Combined organic layer was dried over anhydrous
magnesium sulfate, filtered and concentrated under
reduced pressure. The residue was purified by column
chromatography (hexane:ethyl acetate = 2:1), to give
5.76 g (yield: 77.1%, colorless liquid) of 3-
hydroxymethylbenzoic acid ethylester.
Ester compound (1.317 g, 7.311 mmol) prepared
above was dissolved in ethanol (6 ml) . Thereafter,
therein 2N NaOH aqueous solution (11.0 ml, 21.93 mmol)
was slowly added dropwise, and the reaction mixture was
stirred at room temperature for 1 hour. The reaction
mixture was neutralized with 1% HCl aqueous solution,
49
extracted with ethyl acetate, and washed with saturated
NaCl solution. Combined organic layer was dried over
anhydrous magnesium sulfate, filtered and concentrated
under reduced pressure. The residue was purified by
column chromatography (ethyl acetate:methyl alcohol =
5:1), to give 1.03 g (yield: 99.2%, white solid) of the
target compound.
H NMR (400MHz, CD3OD) : 54.66(s, 2H) , 7 . 4 4 ( t ,
J=7.7HZ, 1H) , 7.5G(d, J=7.7Hz, 1H) , 7.92(d, J-=7.7Hz,
1H) , 8.04(3, 1H)
* Example 2; 5-nitrooxymethyl-furan-2-carboxylic acid-
[ (7S) -1,2, 3-trimethoxy-10-methylsulfanyl-9-oxo-5, 6, 7,_9-
tetrahydro-benzo [a] heptalen-7-yl] -amide
Me.O;-
-QM02
aH NMR (400MHz, CDG13) : 52. 37-2 . 51 (m, 3H) , 2.46(s,
3H) , 2.61-2.92(m, 1H) , 3.74(s, 3H) , 3.93(s, 3H), 3.98(s,
3H) , 4. 82-4. 85 (m, 1H) , 4.85(d, J=13.2Hz, 1H) , 4.90(d,
50
J=13.2Hz, 1H), 6.39(3, 1H) , 6.58(s, 1H) , 6.64(8, 1H) ,
7.16(d, J=10.6Hz, 1H) , 7.42(d, J=10.2Hz, 1H) , 7.72 (s,
1H) , 8.99(s, 1H)
Example _ 3j _ N- [ (7S) -3-isopropoxy-l, 2-^dimethoxy-10-
methylsulf anyl-9-oxo-5 , 6 ,1 , 9-tetrahydrobenzo
[a] heptalen-7-yl] -3 -nitrooxymethyl-benzamlde
fylpO?
Q,N'Q
lll NMR (400MHz, CDC13) : 61,42 (t, ; J=6.6Hz, 6H),
2 . 2 2 - 2 . 2 7 ( m , 1H) , 2 . 3 4 - 2 . 4 9 ( s , 2H) , 2.54-2.57(m, 1H) ,
3.75(s, 3H) , 3.96(3, 3H) , 4 . 59-4 . 63 (m, 1H) , 4.93-4.97(01,
1H) , 5.23(q, J=13.0H2, 2H) , 6.56(s, 1H) , 7.15(d,
J=10.6Hz, Hi), 7. 18-7.25 (m, 1H) , 7.33(cl, J=O.OHz, 2H) ,
7.46(d, J=10.2Hz, 1H) , 7.64(s, lH),7.71.(d, J=8.0Hz,
7.88 (s,. 1H) , 8.40 (s, 1H)
51
Example 4 : N- [ (_7_s) -3 -ethgxy_-1, 2 -dimethoxy-1 Qjmethylsulf
anyl- Q-oxo- 5,6,7, 9 -tetrarahydrobenzo
[a] heptalen-7-yl]-3-nitr-oxymethyl-benzamide
•0"
XH NMR (400MH2, CDC13) : 61.49(t, J=6.9Hz, 3H) ,
2.17-2. 24. (m, 1H) , 2. 34-2. 47 (in, 211) , 2.45(s, 3H) , 2.49-
2.58(m, 1H) , 3.75(3, 3H) , U.iT/U, 311), 4 .12-4 .15 (m, 2H) ,
4.89-4.96 (tn, 1H) , 5.24(q, J=12.0Hz, 2H) , 6.56(s, III),
7.15(d/ J=10.2Hz, 1H) , 7.21-7.25(m/ III), 7.31-7.35(01,
1H) , 7.41(d, J=10.6Hz, 1H) , 7.60(s, 1H) , 7.71(d,
J=7.3Hz, 1H) , 7.78(s, 111), 0.23(3, 1H)
52
Example 5; Preparation of __6-nitrooxymethyl_-pyridine-2-
carboxylic acid- [(75) -1,2,3-trimethoxy-lQmethylsulfanyl-
9-oxo-5, 6,7,9-tetrahydrpbenzolfa]
heptal en - 7 -yl ] - amide
Preparation of 6-hydroxymethyl-pyridine-2-
carboxylic acid- [ (75) -l,2,3-trimethoxy-10-
methylsulfanyl-9-oxo-5,6,7,9-tetrahydrobenzo[
a]heptalen -7-yl]-amide
EDCI.DMAP
MeCN
MeS
According to the similar procedure in the step l
of Example 1l, by using 6-hydroxymethyl pyridine-2-
carboxylic acid (23 mg, 0.17 mmol), 35 mg (yield: 53%,
yellow solid) of the target compound was obtained.
XH NMR (400MHz, CDC13) : 52.45(s, 3H), 2.33-2.50(m,
3H) 2.52-2.73 (m, 1H) , 3.75(s, 3H) , 3.92(s, 3H) , 3.97(s",
53
3H) , 4.28(d, J-14Hz., Ill), 4 , 4 4 ( d , J-l-lHz, 1H) 4 . f - ; G -
- l . D 2 ( m , HI), C . b V ; , , , : : . . , . •, . 1 ' ( a , J- : 1 0 . 4 z . , 1.H) , V . . 7 -
7.48(m, 4H) , 7.73(3, 111), 9 . 80 (d, J^CHK, 111)
Preparation oi: 6-b/romomethyl-pyrji.diiv.- 2
carboxylic acid-1 l-iLLzi-JL-r': trimethoxy - I 0 -
methylsul£anyl-9-oxo-5, 6 , 7 , 9 -
tetrahydroObenzo [a] heptalen -7 - vlj -
MeS
PBl'3
CH-.CI,
MeO—V/ --r
t'Tf
MeS
Br
A compound (50,45,mmol prepared in the
step 1 was dissolved in dichlorometharie (6 ml) .
Thereafter, therein tribromophosphine (PBr3, 0.005 ml,
0.05 mmol) was slowly added dropwise at 0°C, and the
reaction mixture was stirred at room temperature for 3
hours. Methanol was added to quenen the reaction.
Combined organic layer was dried over anhydrous
magnesium sulfate, filtered arid and concentrated under
reduced pressure. The residence was purified by column
chromatography (chloroform:methanol = 99:1), to give 40
mg (yield: 71%, yellow solid) of the target compound.
XH NMR (400MHz, cnc!3): 52 . 06- 2 . 14 (in, 1H) , 2.33-
2.41(m, 1H). , 2.4l(s, 3H) , 2. 46-2. 54 (m, 1H) 2 . 58-2 . 63 (m,
1H) , 3.72(s, 3H), 3.90(3, 3H) , 3.95(S,*3H), 4.55(d,
J=2.8Hz, 2H) , 4. 77-4, 83 (m, 1H) , 6.57(s, 1H) , 7.04(d,
•7-10. 4Hz, 1H) , 7.29(s, 1H) , 7.31(d, J-10.4HZ, III),
7.58(d, J=7.6Hs, 1H) , 7.78(t, J = 7 . GHz , • HI), 7.90(d,
i7=»8.0Hz, 1H) , 8.53(d, J"=7.$Hz, 1H)
Preparation of 6-nibrooxyniet:hyl-pyridine-2-
carboxylic acid- [ (7S) -1, 2 , 3 -r.rimethoxy-lQmethylsulfanyl-
9-oxo-5,6,7,9-tetrahydrobenzo
[a]heptalen -7-yl]-amide
AgNOa
MeCN
MeS
'ON02
MeS
A compound (40 mg, 0.070 utmol) prepared in the
step 2 was dissolved in acetonitrile (3rnl) . Thereafter,
therein silver nitrate (23 mg, 0.14 mmol) was slowly
55
added dropwise, and the reaction mixture was stirred at
room temperature for 12 hours. The reaction mixture
was washed with water and concentrated under reduced
pressure. The residue was purified by column
chromatography ( chloroform: met: hano 1 = 99:1), to give 17
mg (yield: 44.7%, yellow solid) of the target compound.
XH NMR (400MHz, CDC13) : 52. 03-2.10 (m, 1H) , 2.33-
2.41(m, 1H) , 2.43(s, 3H) , 2.46-2.54(m, 1H) 2.58-2.65(m,
1H) , 3.73(s, 3H) , 3.92(3, 3H) , 3.95(S, 3H) , 4.77-4.83(m,
1H) , 5.63(d, i7=3.2Hz:, 2H) , 6.58(s, 1H) , 7.06(d,
J=10.4Hz, 1H) , 1.21(3, 1H) , 7.31(d, J=»10.4Hz, 1H) ,
7.53(d, J=7.6Hz, 1H) , 7.88(t, J=B.OHz, 1H) , 8.02(d,
J=7.2Hz, 1H) , 8.39(d, J-7.2Hz,
Example 6-24
Compounds of Example 6 - Example 24 were
synthesized in analogy to the procedure as described in
Example 5, and intermediates were prepared by the
method described as follows.
56
Preparation of 6-hydroxymethylpyridine-
2-carboxylic acid
NaOH
MeOH
6- Hydroxyme thyl- pyr idine-2-carboxy lie acid
ethylester (200 mg, 1.1 rtimol) ( 104, 2251-2257) was dissolved in methanol (1 ml) .
Thereafter, therein 2N NaOH aqueous solution (1 ml) was
slowly added dropwise, and the reaction mixture was
stirred at room temperature for 1 hour. The reaction
mixture was acidified (pH=3) with 2N HC1 . Solvent was
concentrated under reduced pressure, then, dissolved inmethanol
and filtered. The filtrate was concentrated
under reduced pressure to give 150 mg (yield: 89%,
white solid) of the target compound.
XH NMR (400MHz, CD3OD) : 55.05(s, 2H) , 8.34(d,
J=8.0Hz, 1H) , 8.47(d, J=8.0Hz, 1H) 8.73(d, J=8.0Hz,
57
Preparation of 5-hydroxymethylthiophene-
2-carboxylic acid
MeOH Me° )Me THF
o
2N-NaOH,
HO'
MeOH
Thiophene-2 , 5-dicarboxylic acid (4 g, 23.3 mmol)
was dissolved in methanol (300ml) . Catcilytic amount of
sulfuric acid was slowly added therein. The reaction
mixture was re fluxed to give 3.8 g (yield: 81.7%, white
solid) of thiophene-2 , 5-dicarboxylic acid dimethylester .
The thiophene-2, 5-dicarboxylic acid dimethylester (3.7
g, 18.84 mmol) was dissolved in anhydrous
tetrahydrofuran (50 ml) at room temperature under a
nitrogen atmosphere. 2 . OM Lithiutnborohydrn.de
tetrahydrofuran solution (5.5 ml, 11 mmol) was slowly
added therein at 0°C. The reaction mixture was
refluxed for 3 hours to give 2.1 g (yield: 64. 7 r , white
solid) of 5-hydroxymethyl-thiophene-2-carboxy ic acid
methylester . 5 -Hydroxymethyl -thiophene-2 -corboxylic
acid methyl ester (2.1 g, 12.2 mmol) was dissolved in
methanol (20 ml) . 2N NaOH aqueous solution (15ml) was
slowly added therein. The reaction mixture was stirred
58
at room temperature for l hour to give 1.75 g (yield:
89%, white solid) of the target compound.
1H NMR (400MHz, CDC13): 53.90(Br, 1H) , 4.79(d,
J-O.BHz,, 2H) , 6.97(d, J=4Hz, 1H) , 7 .66 (d, J=4Hz, 1H)J
Preparation of 2-fluoro-3-
hydroxymethyl-benzoic acid
XT F O: OF' 1 nU ^. urn'-^1. T.r
2-Fluoroisophthalic acid (3 g, 16.3 inmol) (J".
Amer. Chem. Soc. , 1943, 65, 2308) was dissolved in
methanol (150 ml) . Catalytic amount of sulfuric acid
was slowly added therein. The reaction mixture was
refluxed. The resultant was concentrated under reduced
pressure to remove solvent, and dissolved in ethyl
acetate. Combined organic layer was washed with
saturated sodium carbonate, dried over anhydrous sodium
sulfate and concentrated under reduced pressure. The
residue was purified by column chrolmatography (ethyl
acetaterhexane = 1:2), to give 3.1 g (yield: 08%, white
solid), of 2-f luoro-isophthalic acid dimethylester.
59
Thiophene-2,5-dicarboxylic acid dimethylester (3.1 g,
14.6 mmol) was dissolved in anhydrous tetrahydrofuran
(50 ml) at room temperature under a nitrogen atmosphere.
2.0M Lithiumborohydride tetrahydrofuran solution (4.4
ml, 8.7 mmol) • was slowly added therein at 0°C. The
reaction mixture was refluxed for 3 hours. The reaction
mixture was acidified with IN HCl aqueous solution,
concentrated under reduced pressure to remove solvent,
and extracted with chloroform. Combined organic layer
was dried over anhydrous sodium sulfate, filtered, and
solvent was concentrated under reduced pressure. The
residue was purified by column chromatography
(dichloromethane: methanol = 99:1}, to give 1.5 g
(yield: 58%, white solid) of 2-f luor o -3- hydr oxyme thy 1-
benzoic acid methylester. The 2-fluoro-3~
hydroxymethyl-benzpic acid methylester (1.3 g, 7i6
mmol) was dissolved in methanol (20 ml) . 2N NaOH
aqueous solution (14 ml) was slowly added therein. The
reaction mixture was stirred at room temperature for 1
hour. The reaction mixture was acidified (pH=3) with
2N HCl. Solvent was concentrated under reduced
pressure, then, dissolved in methanol and filtered.
The filtrate was concentrated under reduced pressure to
give 1.15 g (yield: 88%, white solid) of the target
compound.
60
MMR (400MHz, DMSO-d6-) : 54.58(d, J=5.2Hz,
2H),5.37(t, J-=5.2HZ, 1H) 7.28(t, J=SHz, 1H) , 7.68(t,
, 7.74(t, J=8Hz,
Preparation __ of 3-£luoro-5-
hydroxymethyl -benzole acid
Preparation of 3-f luoro- 5 -hydroxyme thylbenzoic
acid methyleater
5-Fluoroisophthalic acid dimetliylester (1.6 g,
7.54 mmol) (J. Org. Chem; 1969, 34, 1960-1961) was
dissolved in tetrahydrofuran (15 ml) . Thereafter,
therein 2.0 M lithiutnborohydr ide tetrahydrofuran
solution (2.6 ml, 5.27 mmol) was slowly added dropwise
at 0°C, and the reaction mixture was refluxed for 3
hours. The reaction mixture was acidified with IN HC1
aqueous solution, concentrated under reduced pressure
to remove solvent, and extracted with chloroform.
61
Combined organic layer was dried over anhydrous sodium
sulfate, filtered, and concentrated under reduced
pressure. The residue was purified by column
chromatography (dichloromethane : methanol = 99:1), to
give 800 mg (yield: 57%, white solid) of the target
compound .
XH NMR (400MHz, CDC13): 63.92(d, J"=1.6Hz, 3H) ,
4.75(d, J=4Hz, 2H) , 7.29-7.32(m, 1H) , 7.61(dd, J=9.2,
I
1.6Hz, 1H) , 7.8(d, iJ=0.8ITz,
Preparation of 3-fluoro-5-hydroxymethy1-
benzoic acid
MeO.
NaOH
MeOH
According to the similar procedure in the method
of intermediate 3, by using a compound prepared in the
step 1, 1.6 g (yield: 94%, white solid) of the target
compound was obtained.
XH NMR(400MHz, CD3OD) : 5-1 . 66 (d, J=O.QHs, 211),
7.33-7.36(m/ 1H), 7.56-7.59(m, 1H), 7.03-7.84(ra/
62
Preparation of 4-£luoro-3-
hydroxymethyl-benzole acid
Preparation of 4-fluoro-isophthalic acid
KMn04
HO
H20, reflux
4-Fluoro-3-methyl-benzole acid (2.52 g, 16.346
tnmol) and potassium permanganate (10.33 g, 65.382 mmol)
were dissolved in aqueous solution (300 ml) , and the
mixture was refluxed for 1 day. The reaction mixture
was filtered and the resultant solution was cooled down
at room temperature, then cone. HC1 solution was added
thereto. The produced solid was heated until it was
completely melted. The temperature was lowered again
into room temperature and then the solid was filtered,
to give 2.08 g (yield: 69.1%, white solid) of the
target compound.
H NMR (400MHz, CD3OD) : 57.32(dd, 17=10.4, 8. GHz,
1H), 8.21-8.25(m, 1H) , 8.59(dd, J=7 . 0, .2 . 4Hz, 1H)
63
Preparation of 4-fluoro-isophthalic acid
dimethylester
O
cat. H2SO4
OH MeO.
MeOH, reflux
,OMe
O
A compound prepared in the step 1 (2.08 g, 11.29
mmol) was dissolved in methanol (30 ml) . Then, 10
drops of cone, sulfuric acid were added therein. The
reaction mixture was refluxed for 1 day, neutralized
with saturated sodium hydrogen carbonate aqueous
solution, and extracted with chloroform. Combined
organic layer was dried over anhydrous sodium sulfate,
filtered, and concentrated under reduced pressure, to
give 2.21 g (yield: 92.2%, white solid) of the target
compound .
XH NMR (400MHz, CDC13) : 53.96(s, 3H) , 3.97(s, 3H) ,
7.22(dd, J-10.3, 8.8Hz, 1H) , 8. 20-8. 23 (m, 1H) , 8.64(dd,
J=7.0, 2.2Hz,
64
_grgparation_ _of 4- l: 1 uoro-3 -hydrpxymethyl -
benzoic acid methylester and 2-fluoro-5-hydroxymethylbenzoic
acid methylester
LiBH4
MeO
THF. reflux
A compound prepared in the step 2 (104.5 mg,
0.493 mmol) was dissolved in tetrahydrofuran solution
(4 ml) . Then, 2M lithiumborohydride tetrahydrofuran
solution (0.123 ml, 0.246 mmol) was slowly added
therein. The reaction mixture was refluxed for 1 day.
The reaction was quenched by water. Then, pH was
adjusted to 5 with 1M HGl solution at 0°C. Extraction
with ethyl acetate was performed. Combined organic
layer was dried over anhydrous sodium sulfate, filtered,
and concentrated under reduced pressure. The residue
was purified by column chromatography
(hexane:ethylacetate = 2:1), to give 4-fluoro-3-
hydroxymethyl-benzoic acid methylester (45.4 mg, yield:
50.1%, colorless liquid) and 2-fluoro-5-hydroxymethylbenzoic
acid methylester (15.4 mg, yield: 17.0%,
colorless liquid).
65
4-fluoro-3-hydroxymethyl-benzoic acid
methylester:
XH NMR (400MHz, CDC13) : 52.73(t, J-S.lHz, 1H) ,
3.90(0, 3H) , 4.78(d, J-5.1HZ, 2H) , 7.07(dd, JW9.2,
9.2Hz, 1H) , 7.93-7-.97(m, 1H) , 8.14(dd, J=7.1, 2.2Hz,
1H)
2-fluoro-5-hydroxymethyl-benzoic acid
methylester:
1H NMR (4 0 0MHz , CDCl3) : 51.96(1;,. J*=4 . 4Hz , 1H} ,
3.94(S, 3H) , 4.70(d, J-4.4Hz, 2H) , 7.13(dd, J=10.6,
8.4Hz, 1H) , 7.52-7.56(m, 1H) , 7.92(dd, J=7.0, 2.2Hz,
1H)
Preparation of 4-fluoro-3-hydroxymethylbenzoic
acid
2N NaOl-l / MeOl-l
- i-io
According to the similar procedure in the method
of intermediate 3, by using a compound prepared in the
step 3 (1.074 g, 5.380 mmol), 0.906 g (yield: 91.3%,
white solid) of the target compound was obtained.
66
XH NMR (400MHz, CD3OD) : 54.69(s, 2H) , 7.13
J=9.9, 8.8Hz, 1H) , 7. 90-7. 97 (m, 1H) , 8.16(dd, J=7.3,
2.2Hz,
_ Pi-eparation _ of _ 2-fluoro-5-
hydroxymethyl -benzole acid
F
2N NaOH / MeOH
'Ma HO
According to the similar procedure in the method
of intermediate 3, by using 2-fluoro-5-hydroxymethylbenzole
acid methylester prepared in the step 3 of
intermediate 7, a target compound was obtained.
XH NMR (400 MHz, CD3OD) : 54.61(s, 2H) , 7.17(dd,
J-11.0, 8.4Hz, 1H) , 7.55-7.59(m, 1H) , 7.93(dd, J=7.l;
2.4Hz,
67
Preparation of 3-hydroxy-5-
hydr oxyme t hy 1 - ben z ojic_
o
NaOH / MeOH H|
OH
5-Hydroxy-isophthalic acid raethylester (300 mg,
1.42 mmol) was dissolved in tetrahydrofuran (20 ml) at
O°C under a nitrogen atmosphere. Lithium aluminium
hydride (30 mg, 0.7 mmol) was added therein. The
reaction mixture was stirred at room temperature for 3
hours. Water was added to quench the reaction, and
aqueous layer was extracted with ethyl acetate.
Combined organic layer was dried over anhydrous sodium
sulfate, filtered and concentrated under reduced
pressure. The residue was purified by column
chromatography (dichloromethane:methanol = 20:1).
ABbove-obtained 3-hydroxy-5-hydroxymethyl-benzoic acid
methyleater (170 mg, 0.93 mmol) was dissolved in
methanol (1 ml) . IN NaOH aqueous solution (1 ml) was
added therein. The reaction mixture was stirred at
room temperature for 1 hour. The reaction mixtux'e was
acidified (pH=3) with 2N HC1. Solvent was concentrated
under reduced pressure, and then, dissolved in methanol
68
and filtered. The filtrate was concentrated under
reduced pressure to give 150 mg (yield: 96%, white
solid) of the target compound.
XH NMR (400MHz, CD3OD) : 53.87(3, 3H),, 4.57(s, 2H) ,
7.02(s, 1H) , 7.31(s, 1H) , 7.48(s,
^Intermediate 10> Preparation of 3,5-Ms-hydroxymethylbenzoic
acid
Preparation of 3,S-^is-hydroxymethyl-benzoic
acid methylester
OMe 1 eq. LAH HO'
THF
OMe
Benzene-1 , 3 , 5-tricarboxylic acid trimethylester
(1.010 g, 4.003 mmol) was dissolved in tetrahydrofuran
(15ml) . Thereafter, the temperature was lowered into
0°C, and therein lithium aluminium hydride (0.160 g,
4.003 mmol) was slowly added, and the reaction mixture
was stirred at room temperature for 3 hours. Water
69
(0.15 ml) and 15% NaOH aqueous solution (0.15 ml) were
elowly added to quench the reaction. Aqueous solution
(0.45 ml) was added again. Solvent was concentrated
under reduced pressure. The residue was purified by
column chromatography (hexane:ethyl acetate = 1:2), to

give 0.34 g (yield: 43.1%, colorless liquid) of the
target compound.
XH NMR (400MHz, CD3OD) : 53.91(s, 3H) , 4.66(s, 4H),
7.59(3, 1H) , 7.93(3, 2H)
r Preparation of 3,5-bis-hydroxvmethyl-benzoic
acid
According to the similar procedure in the method
of intermediate 3, by using a compound prepared in the
Step 1 (1.50 g, 7.65 mmol) , 0.617 g (yijfld: 44.3%,
white solid) of the target compound was obtained.
H NMR (400MHz, CD3OD) : 55.21(s, 4H) , 7.55(s,
7.92(3, 2H)
70
Preparation of 2-hydroxy-4-
hydroxymethyl-benzole acid
Preparation of 2 -hydroxy-4 -bydroxymethy1-
benzoic acid methylester
HO.
i) NBS, Benzoyl peroxide, CGI
2) H2O, Dioxane
MeO.
2-Hydroxy-4-methyl-benzole acid methylester (166
mg, 1 mmol) was dissolved in CC14 (1.5ml). NBS (177 mg,
1 mmol) and benzoyl peroxide (5 mg, 0.02 mmol) were
added therein. The reaction mixture was stirred at
70°C for 12 hours. Then, the reaction mixture was
washed with water, dried over anhydrous sodium sulfate,
and filtered. Solvent was concentrated under reduced
pressure. The residue was purified by column
chromatography (dichloromethane:hexane =1:4), to give
4-bromomethyl-2-hydroxy-benzoic acid methylester (130
mg, yield: 53%, white solid). The resultant compound
i1
(130 mg, 0.53 mmol) was dissolved in aqueous solution
(1.5. ml) and 1,4-dioxane (1.5 ml), and the mixture was
stirred at 90°C for 12 hours. The reaction mixture was
71
extracted with chloroform. Combined organic layer was
dried over anhydrous sodium sulfate, filtered and
concentrated under reduced pressure. The residue was
purified by column chromatography (ethyl acetate:hexane
- 1:4), to give 55 nig (yield: 57%, white Solid) of the
barget compound.
H NMR (400MHz, CDC13): 53.95(s, 311) , 4.71(d,
J=6Hz, 2H) , 6.88(d, J=8Hz, III), 6.99 (a, IE), 7.82(d,
J=8Hz, 1H) , 10.79(3, 1H)
Preparation of 2_- hydroxy-_4 -hydroxyme thy.1 -
benzoic acid
x--^ ^"^
NaOH
MeOH/HoO
According to the similar procedure in the method
of intermediate 3, by using a compound prepared in the
Step 1, 45 mg (yield: 90%, white solid) of the target
compound was obtained..
72
Preparation of 4-hydroxymethylthiophene-
2-carboxcylic acid
A: 4-methyl-thiophene-2-carboxylic acid.
B; Preparation of 3-methyl-thiophene-_2-
carboxylic acid
i)n-Bu Li/Ether
2.5M n-Butyllithium (12.2 ml/ 30.55 nunol) was
V,
dissolved in diethyl ether (1 ml) at room temperature
under a nitrogen atmosphere. 3-Methyl-thiophene (3 g,
30.55 mmol) dissolved in cliethyl ether was slowly added
therein. The mixture was refluxed for 2 hours.
Reaction chamber was cooled to 0°C and dry-ice was
slowly added therein. Reaction was quenched with 45 ml
of water, Diethyl ether layer was extracted and
removed. Water layer was acidified with IN HCl
solution and extracted with ethylacetate. Combined
organic layer was dried over anhydrous sodium sulfate,
filtered and concentrated under reduced pressure. The
residue was purified by column chroma tography
73
(hexane:ethyl acetate = 9:1), to give the target
compound A (900 mg, white solid) and B (650 mg, white
solid).
Preparation of 4 -bromgrnethyl- thiophene- 2-
carboxylic acid methyl ester
4-Methyl-thiophene-2-carboxylic acid (900 mg,
6.33 mmol) prepared in the step 1 was dissolved in
methanol (15 ml) . Catalytic amount of sulfuric acid was
slowly added therein. The reaction mixture was refluxed
to .give 4 -methyl -thiophene -2 -carboxylic acid
methylester (890 mg, yield: 90%, white solid) . 4-
methyl -thiophene -2 -carboxylic acid methyl ester (200 mg,
1.28 mmol) and NBS (215 mg, 1.216 mmol) , and benzoyl
peroxide of catalytic amount were dissolved in
tetrachloromethaiie solution (5 ml) . The reaction
mixture was refluxed (70°C) for 3 hours to give 165 mg
(yield: 55%, white solid) of the target compound.
T3. NMR(400MHz, CDC13) ; 53.89(s, 3H) , 4.46(s, 2H) ,
7.49(S, 1H) , 7.80(s, 1H)
74
Preparation of 4-hydroxymethyl-thiophene-2-
carboxylic acid
fit • /—.Oil ' " H2Q/M-pio-xariB" Jr.' $ MeO.H/HizO
A compound (150 mg, 0.638 mmol) prepared in the
step 2 was dissolved in 1,4-dioxane (1.5 ml) and water
(1.5 ml). Silver nitrate (130 mg, 0.765 mmol) was
slowly added therein. The reaction mixture was stirred
at room temperature for 12 hours to give 4-
hydroxymethyl-thiophene-2-carboxylic acid methyl ester
(60 mg, yield: 55%, white solid). This compound (60 mg,
0.348 mmol) was dissolved in methanol (1 ml) at room
temperature. IN NaOH aqueous solution (1 ml) was slowly
added therein, and the reaction mixture was stirred at
room temperature for 1 hour, to give 50 mg (yield: 95%,
white solid) of the target compound.
75
intermediate 13 > Preparation of 3-hydroxytnethylthiophene-
2-carboxylic acid
Preparation of 3 -bromomethyl-thiophene-2-
carboxylic acid methyl ester
E>f
NBS,-Den?pyl peroxide;-
, r - . - " H3CO .
S Me.OH If S' CCL,; 1
Q .U Q
According to the similar procedure in the method
of step 2 of intermediate 12, by using 3-methylthiophene
-2 -carboxyl ic acid (650 mg, 4.57 mmol)
prepared in the step 1 of intermediate 12, 750 mg
(yield: 79%, white solid) of the target compound was
obtained.
H NMR (400MHz, CDC13); 53.90(s, 3H) , 4.91(s, 2H) ,
7.18(d, J=5.2Hz, 1H) , 7 . 4 6 ( d , J=5.2Hz,
Preparation of 3 -hydroxyme thyl - thiophene -2-
carboxylic acid
76
Br
HUO/'M-Dfoxane: IT ':SX 'MeOH/.H2-0
' •
According to the similar procedure in the method
of step 3 of intermediate 12, by using a compound
prepared in the step 1 (750 mg, 3.19 mmol) , 210 mg
(yield: 92%, white solid) of the target compound was
obtained.
Preparation of (3-hydroxymethylphenyl)-
acetic acid
CaC.03;
H20/Me.QH.-
m-Tolyl acetic acid ethyl ester (1 g, 5.6 mmol),
NBS (948 mg, 5.33 mmol) and benzoyl peroxide of
catalytic amount were dissolved in tetrachloromethane
(15 ml) . The reaction mixture was refluxed (70°C) for
3 hours to give (3-bromomethyl-phenyl)-acetic acid
ethyl ester (GOO mg, yield: 42%, white solid). And (3-
bromomethyl-phenyl)-acetic acid ethyl ester (130 mg,
0.50 mmol) and calcium carbonate (300 mg, 3 mmol) were
77
dissolved in water (2 ml) and 1,4-dioxane (2 ml). The
reaction mixture was refluxed to give (3-hydroxymethylphenyl)-
acetic acid ethyl ester (85 mg, yield: 87%,
white solid). (3-Hydroxymethyl-phenyl)-acetic acid
ethyl ester (85 mg, 0.43 mmol) was dissolved in
methanol (1 ml) at room temperature. IN NaOH aqueous
solution (1 ml) was added therein. The reaction mixture
was stirred at room temperature for 1 hour, to give 65
mg (yield: 91%, white solid) of the target compound.
H NMR(400MHz, CDC13) ; S3.66(s, 2H) , 4.57{s, 2H) ,
7.21-7. 32 (m, 4H)
•^Intermediate 15> Preparation of 3- (2-hydroxy-ethyl) -
benzoic acid
pec
THF.
NaOK
M^a-LAlr * ^NX^k^W^^h
|"|pf_J
•f'NaHMDS.
«)• (Metivoxyrnelhyl)triphe hyl
rphbsplidniurh Chlafide"
TftF
BOH
OMe
M00
78
Isophthalic acid (5 g, 30 mmol) was dissolved in
methanol (50 ml) . Catalytic amount of sulfuric acid
was added therein. The reaction mixture was stirred at
reflux for 12 hours to give isophthalic acid dimethyl
ester (5.2 g, yield: 90%, white solid). Isophthalic
acid dimethyl ester (5.2 g, 26.7rnmol) was dissolved in
tetrahydrofuran (30 ml) . 2M Li thiumborohydr ide
tetrahydrofuran (13 ml, 26.7 mmol) was added therein.
The reaction mixture was refluxed to give 3-
hydroxymethyl'-benzoic acid methyl ester (2.7 g, yield:
63%, colorless liquid), and 3-hydroxymethyl-benzoic
acid methyl ester (200 mg, 1.20 mmol) was dissolved in
dichloromethane (5 ml) . PCC (388 mg, 1.8 mmol) was
added therein. The reaction mixture was stirred at
room temperature for 3 hours to give 3-formyl-bensbic
acid methyl ester (140 mg, yield: 72%, white solid).
(Methoxymethyl)triphenylphosphonium chloride (770 mg,
2.24 mmol) was dissolved in tetrahydrofuran (5ml) under
a nitrogen atmosphere. 1M NaHMDS tetrahydrofuran (2 ml,
2.04 mmol) was slowly added therein at -78"C, and the
reaction mixture was stirred for 1 hour. 3-Formylbenzoic
acid methyl ester (160 mg, 0.975 mmol)
dissolved in tetrahydrofuran (2 ml) was slowly added
therein. The reaction mixture was stirred at room
79
temperature for 24 hours to give 3- (2-methoxyvinyl) -
benzole acid methyl ester (121 mg, yield: 65%, white
solid). 3- (2-Methoxyvinyl) -benzole acid methyl ester
(120 mg, 0.63 mmol) was dissolved in tetrahydrofuran (3
ml) . 4M HCl (2 ml) was added therein, and the mixture
was stirred at room temperature for 24 hours, to give
3- (2-oxo-ethyl) -benzole acid methyl eater (60 mg,
yield: 54%, white solid). 3- (2-Oxo-ethyl) -benzole acid
methyl ester (60 .mg, 0.036 mmol) was dissolved in
ethanol (2 ml). MaBIli (25 ing, 0 , G7 mmol) was added
therein at 0°C, and the mixture was stirred at1 hour,
to give 3- (2-hydroxyethyl) -benzole acid methyl ester
(50 mg, yield: 84%, white solid). 3 - (2-Hydroxyethyl) -
benzole acid methyl ester (50 mg, 0.28 mmol) was
dissolved in methanol (1 ml) . IN NaOH aqueous solution
(1 ml) was added therein. The reaction mixture was
stirred at room temperature for 1 hour to give 43 mg
(yield: 95%, white solid) of the target compound.
1II NMR(400MH2, CDC13) ; 62 . 32 (br, 1H) , 2.89(t,
J=6.4HZ, 2H) , 3.85(t, J=6.4Hz, 2H) , 7.36(t, i7=7.6Hz,
1H) , 7.42(d, ,J=5.6Hz, 1H) , 7.88(d, .7=8. 8Hz, 1H) , 7. 89 (a,
1H)
80
Example 6: 5-nitrooxymethyl-thiophene-2-carboxylic
acid- [ (7S) -1, 2 , 3-trimethoxy-lO-methylsxilf anyl-9-oxo-
5,6,7,9-tetrahydro~benzo[a]heptalen-7-yl]-amide
MeS
ZH NMR(400MHz, CDC13) ; 62 . 07-2 .11 (m, 1H) , 2.32-
2.45(01, 2H) , 2.45(s, 3H) , 2.53-2.56(m, 1H) , 3.71(s, 3H) ,
3.91(3, 3H) , 3.96(S, 3H) , 4. 84-4. 91 (m, 1H) , 5.46(s, 2H) ,
6.56(s, 1H) , 6.95(d, J=3.6Hs, 1H) , 7.13(d, J-10.8Hzf
1H), 7.37(d, J=10.8Hz, 1H)/ 7.58(d, J«4.0Hz,
7.61(S, 1H), 8.50(d, J=6.SHz,
81
Example 7 : N- [ (7S) -3-cyclopentyjoxy-1-2-dlmethoxy-10
methylsulfanyl-9-oxo-5,6,7,9-tetrahydrobenzo
[a]heptalen-7-yl] -S-nitrooxymethyl-benzamlde
fvleS
^•H NMR(400MHz, CDC13) ; 61. 61-1.73 (m, 2H) , 1.85-
2.00(m, 6H) , 2.21-2.65(m, 4H) , 2.44(s, 3H) , 3.75(s,
3H), 3.93(S, 3H) , 4 . 82-4.88 (m, 1H) , 4.?2-5.00(m, 1H) ,
5.16(dd, J=12, 31.2H2, 2H) , 6.55(s, 1H) , 7.13-7.17 (m,
2H) , 7.28(d, J=6.4Hz, 1H), 7.41(d, J=10.8Hz, 1H) , 7.68-
7.70{m, 1H) , 7.72(s, 1H) , 7.78(s, 1H), 8.78(d, J»7.2Hz,
1H)
Example 8: N- [ (75)-3-ethoxy-l,2-dimethoxy-lQmethylsulfanyl-
9-oxo-5,6,7,9-tetrahydro-
82
benzo[a]heptalen-7-yl3 -2-fluoro-3-nitrooxyTnethylbenzamide
XH NMR(400MHz, CDC13) ; 51.49(t, J=6.9Hz, 3H) ,
1.93-1.98 (m, 1H) , 2.31-2.39(m, 1H) , 2.43(e, 3H) , 2.46-
2.49(m, 1H) , 2.51-2.59 (m, l.H) , 3.73(s, 3H) , 3.96(s, 3H) ,
4.11-4.14(111, 2H) , 4.83-4.86(01, 1H) , 5.53(d, J=12.8Hz,
1H) , 5.59(d, ,7=12.4Hz, 1H) , G.56(s, 1H) , 7.07(d,
j=10.2Hz, 1H) , 7.10-7.15(m, III), 7.20-7.28(m, 1H) ,
7.31(d, J-=10.2Hz, 1H) , 7.57(t, J"=6.4Hz7 1H) , 7.97(t,
J=6.9Ez, IE)
83
Example 9; 2-fluoro-N- [ (7S) -3-isopropoxy-1,2-dimethoxy-
10-methylBulf anyl-9-oxo-5 , 6 , 7 , 9-tetrahydro—
benzo[a]heptalen-7-yl]-3-nitrooxymethyl-benzamide
Me:a
*H NMR (400MHz, CDC13) ; 51.41(q, J=6.0Hz, 6H) ,
2-1.99(m, 1H), 2.31-2.40(m, 1H) , 2.43(s, 3H) , 2.46-
2,52(m, 1H) , 2.55-2.60(m, 1H) , 3 . 7 2 ( s ; 3H) , 3.94(s, 3H)
4.57-4.63(m1, IE), 4.83-4.89 (m, Hi), 5.53(d, J=12.8Hz,
1H), ' 5.59(d, J=12.4Hz, 1H), 6.57(s, 1H) , 7.07(d,
JV.10.6Hz, 1H) , • 7.15-7.19(m, 1H) , 7.23-7.27(m, 1H) ,
7.34(d,. J=10.2Hz, 1H) , 7.56(t, J=7.1Hz, 1H) , 7 . 7 ( t ,
84
Example 10: 2 -f luoro-g-i-i t-CKjxymgLh 1-- [(75) -l:,j,
tritnethoxy- 10 -me thylsulf anyl-9-oxo_--_5, _6 , 7 , 9-tetrahydro
benzo[a]heptalen-7-yl]-benzamide
- NMR (400MHz, GDC13) ; 61. 95-2 . 02 (m, 1H) , 2.31-
2.54(m, 2H) , 2.43(s, 3H) , 2.59-2.63(m, 1H) , 3.73(s,
3H), 3.87(s, 3H) , 3.92(s, 3H) , 4.82-4.88(^1, 1H) ,
5.51(d, J-=12.8Hz, 1H) , 5.60(d, .J=12 . QIIz, 1H) , 6.50(s,
1H) , 7.06(d, J=10.6Hz, 1H) , 7.22-7.27'(m, 2H) , 7.28(s,
1H) , 7.32(d, J=10.6Hz, 1H) , 7.54-7.58(m, 1H) , 7.93-
7.98(m,
Example 11; N- [ (7S) -3-cycloperityloxy-1, 2 -dlmethoxy-10-
methylsulfanyl-9-oxo-5,6,7,9-betrahydro-
85
benzo [a]heptalen-7'yl]-2-fluoro-3-nitrooxymethylbenzamide
TH NMR(400MHz, CDC13) ; 51. 61-1. 73 (m, 2H) , 1.85-
2.00(10, 6H) , 2.35-2.52 (m, . 4H) , 2.42(s, 3H) , 3.72(s,
3H) , 3.93(3, 3H) , 4. 82-4. 88 (m, 2H) , 5.57(dd, J"=12 . 8 ,
37.6HZ, 2H), 6.55(s, 1H), 7.05(d, J=10.4Hz, 1H), 7.09-
7.13(m, 1H) , 7.26(d, i7=7.2Hz, 2I-I) , 7.32(d, .7=10.4PIz,
1H), 7.55-7.59(m, 1H), 7.96-8.00(m, 1H)
Example 12; S-fluoro-S-nitrooxymethyl-Jf-[ (7S)-1,2 , 3-
trimethoxy-10-methylsul£anyl-9-oxo-5,6,7,9-tetrahydrobenzo
[a]heptalen-7-yl] -benzamlclf
86
•fvleS
F
XH NMR(400MHz, CDC13); 62.31-2.45(m, 3H) , 2.46(3,
3H) , 2.59-2.63(m, 1H) , 3.75(g, 3H) , 3.93(s, 3H) , 3.98(S,
3H) , 4.90-4.95(m, 1H) , 5.11(dd, J=12.8, 45.6Hz, 2H) ,
6.57(s, 1H) , 7.03(d, iJ=7 . 6Hz , Hi) , 7.19(d, iJ=8.8Hz, Hi) ,
7.30-7.33(111, 1H) , 7.42(d, J=10.0Hz, 1H) , 7.55(s, 1H) ,
7.68(s, 1H), 8.7S(d, J=7.2Hz,
Example 13: N- [ (75) -3 - e L hqxy--1, 2-dimethoxy-10-
methylsulfanyl-9-oxo-5, 6, 7, 9-tetrahydrobenzo[
a]heptalen-7-yl) -3-£luoro-5-nitrooxymethylbenzamide
'MeS
87
XH NMR(400MHz, CDC13) ; 51.50(t, J=6.9, 3H) ,
2.33-2.42 (m, 3H) , 2.46(s, 311), 2 . 51-2 . 59 (m, 1H) , 3.75(s,
3H) , 3.98(s, 3H) , 4.11-4.16(m, 2H) , 4.90-4.93(m, 1H) ,
5.05{d,. J=12.4, 1H) , 5.17 (d, J=12.4, 1H) , 6.56(s, 1H) /
7.03(d, J=7.3, 1H) , 7.20(d, J=10.6, 1H), 7.31(d, J=9.1,
»
1H) , 7.45(d, J=10.6, 1H) , 7.55(s, 1H) , 7.69(s,
8.88(d, J=7.3, 1PI)
Example 14: 3-fluoro-N-[(75)-3-isopropoxy-l,2-
dimethoxy-lO-methylsulfanyl-g-oxo-S,6,7,9-tetrahydrobenzo[
a]heptalen-7-yl]-5-nitrooxymethyl-benzamide
LH NMR(400MHz, CDC13) ; 61. 37-1. 44 (m, 6H) ,
2.34-2.41(m, 3H), 2.46(s/3H), 2.51-2.58(m, 1H), 3.75(s,
3H) , 3.96(3, 3H) , 4. 57-4. 62 (m, III), 4. 91-4. 95 (m, 1H) ,
5.05(d, J=12.4, 1H), 5.17(d, J=12.4, 1H), 6.56(s, 1H),
7.03(d, J=8.4, 1H), 7.20(d, J=10.6, 1H) , 7.31(d, J=7.3,
1H) , 7.45(d, J=10.6, III) , 7 . 5 : 5 ( 3 , 1H) , 7 . 7 0 ( s , 1I-I) ,
8.91(d, J-7.3, III)
Example 15; N- [ (75) -3-cyclopentyloxy-l,2-dimethoxy-10-
methyleulfanyl-9-oxo-5,6,7, 9-tetrahycIrobenzo[
a]heptalen-7-yl3 -3-fluoro-5-nitrooxyjnethylbenzamide
WeS 0. •QNO>>
XH NMR(400MHz, CDC13) ; 61.64-1.69(m, 2H) ,
1.83-1.98(m, 6H) , 2.35-2.43(m7 3H) , 2.46(s, 3H) ,
2.51-2.58(m, 1H) , 3.74(s, 3H) , 3 . 9 4 ( s , 3H) , 4 . 8 2 ~ 4 . 8 4 ( m ,
1H), 4.91-4. 94 (m, III) ,. 5.04(d, ,7=12.4, III), 5.16(d,
J-12.4, 1H) , 6.55(S j III), 7 . 0 2 ( d , J=8.4, 1H) , 7.20(d,
J=10.6, 1H) , 7.31(d, J=7.3f 1H) , 7.45(d, J=10.2,
7.55(S, 1H) , 7.70(s, III), 8.93(d, J=6.9, 1H)
89
Example 16: 4-fluoro-3-nitrooxymethyl-N- [ (75) -1, 2, 3-
trimethoxy-lO-methylsulfanyl-9-oxo-5,6,7,9-tetrahydro-
*beh2oIa]heptalen.-7-yl] -benzamide
H NMR (400MHz, CDC13) ; 62 . 22-2 .51 (m, 3H) , 2.46(s,
3H)2.55-2.63(m, 1H) , 3.76(s, 3H) , 3 . 9 2 ( s , 3H) , 3 . 9 8 ( s ,
3H)4.90-4.97 (m, 1H) , 5.10(d, J=12.1Hz, 1H) , 5.35(d,
J=12.1Hz, 1H) , 6 . 5 7 ( s , 1H), 6 . 8 6 ( d d , J=9.2, 8.8Hz, 1H),
7.18(d, J=10.3Hz, 1H) , 7.43(d, J=10.3B2, 1H) , 7.72(s,
1H) , 7.76-7.80 (m, 1H) , 7.91(dd, J=6.80, 2.2Hz, HI),
a.93(d, J=7.2Hz,
Example 17: 2-fluoro-5-nitrooxymethyl-JJ'- [ (7S) -1,2,3-
tritnethoxy-lO-methylsulfanyl-g-oxo-S, 6,7,9-tetrahydrobenzo
[a]heptalen-7-yl]-benzamide
90
•f
XH NMR(400MHz, CDC13) ; 51.94-2.01(m, 1H) , 2.31-
2.54{m, 2H) , 2.43(s, 3H) , 2 . 50-2 . 63 (m, 1H) , 3.73(s, 3H) ,
3.92(S, 3H) , 3.97(s, 3H) , 4.81-4.87 (m, 1H) , 5.36(s, 2H) ,
6.5.7(8, 1H) , 7.06(d, J=10.3Hz, HI), 7.16-7.28 (m, 2H) ,
7.26(s, 1H) , 7.33(d, J=10.3H-z, I I I ) , 7 . 51-7.55 (m, I I I ) ,
7.99(dd, j=7.3, 2.6Hz, II!)
Example 18: 3-hydroxy-5-nitrooxymethyl-J7- [ (75) -1, 2, 3-
triTnetho3cy-10-methylsulfanyl-9-oxo-5, 6 , 7 , 9-tetrahydrobenzo
[a]heptalen-7-yl] -benzamide
MeS ON.09'
91
4H NMR(400MHz, CDC13) ; 52 . 30-2 . 43 (m, 6H) , 2.56-
2.57(m, 1H) , 3.51(s, 3H) , 3 . 9 0 ( s , 311) , 3.92(s, 3H) ,
4.82-4.85(m, 1H), 5.00(dd, J=30.8Hz, 12.8Hz, 2H) ,
6.56(s, 1H) , 6.62(s, 1H) , 7.11(s, 2H) , 7.16-7.19(m, 2H) ,
7.41(d, J=10.8Hz, 1H), 7 . 6 6 ( s , 1H), 8.57(brs, 1H, NH),
8.78(brs, 1H, OH)
Example 19: 3,5-bia-nitrooxymebhyl-Jf- [ (7S) -1,2,3-
trimethoxy-10-methylsulfanyl-9-oxo-5, 6,7, 9-tetrahydrobenzo[
a]heptalen-7-yl]-benzamide
ON.O->
XH NMR (400MHz, CDC13) ; 52 . 29-2 . 51 {m, 3H) , 2.46(3,
3H), 2.60-2.62(m, 1H), 3.77(s, 3H), 3.93{s, 3H), 3.99(s,
3H) , 4.94-4.98 (m, 1H) , 5.18(s, 411), 6.58(s, 1H) , 7 . 2 0 ( d ,
J=10.6Hz, 1H) , 7.26(s, III), 7.45(d, J=10.6Hz, 1H) ,
7-.77(s, 2H) , 7.81(8, 1H) , 9.12(d, J=7.0Hz, 1H)
Example 20 : 2-hydroxy-4-riitrooxymethyl-N- [ (7S)-1, 2, 3 -
trimethoxy-10-methyl3ulfanYl-9-oxo-5 , 6 , 7 , g-tetrahydrobenzo[
a]heptalen-7-yl]-benzamide
MeS
a'H NMR (400MHz, CDC13); 52 . 25-2 . 63 (m, 4H) , 2.47(s,
3H), 3.74(s, 3H) , 3.91(s, 3H) , 3.97{S, 3H), 4.85-4.92(111,
, 5.27{d, J=4.8Hz, 2H), 6.53(d, .6.61(0, 1H) , 7.20(d, J=10.4Hz, 1H) , 7.42(d, iJ=10Hz,
, 7.63(s, IE) , 7.72(d, J=8Hz, Hi), 9.13(d, ^7=6.8112,
1H) , 12.29(3, 1H)
Example 21: 4-nitrooxyrnethyl-t hiophene - 2 - carboxyl i c
acid [ (7S) -1,2, 3-trimethoxy-l0-methylsulf ariyl-9-oxo-
5,6,7,9-tetrahydro-benzo[a]heptalen-7-yl]-amide
93
Hi NMR(400MHz, CDC13); 52.21-2.29(m, 1H) , 2.34-
2.46(m, 5H), 2.51-2.58(m, 1H), 3.71(s, 3H) , 3.91(s, 3H),
3.96(5, 3H), 4.88-4.95(m, 1H) , 5.18(dd, J-12.8, 20Hz,
2H), 6.56(s, 1H) , 7.17(d, J=10.4Hz, 1H) , 7.30(s, 1H) ,
j7.41(d, J=10.4Hz, 1H) , 7.61 (a, III), 7.74(s, 1H) , 8.81(d,
j=6.4Hz, 1H)
Example 22; 3-nitrooxymethyl-thiophene-2-carboxylic
acid [(7S)-1,2,3-trimethoxy-10-methylsulfanyl-9-oxo-
5,6,7,9-tetrahydro-benzq[a]heptalen-7-yl]-amide
94
XH NMR (400MHz, CDC13) ; 52 . 04-2 .17 (m, 1H) , 2.30-
2.45(m, 5H) , 2.57-2.64(m, 1H) , 3.73(s, 3H) , 3.91(s, 1H) ,
3.96(S, 3H) , 4.67-4.88(m, 1H) , 5.66(d, J-=»13.6Hz, 1H) ,
i
5.80(d, J=13.6Hz, 1H) , 6.56(s, 1H) , 7 . 02 (d, .7=4.4112,
1H) , 7.08(d, J=l0.4Hs, Hi), 7.26(d, iJ=4.4Hz, 1H) ,
*
7.29(d, i7=10.4Hz, 1H) , 7.43(s, 1H) , 7.45(d, J=7.2H2,
1H)
Example 23 ; 2- (3-nitrooxymethyl-phenyl)-N-[(7S) -1,2,3-
trinlethoxy-lQ-Tnethylsulfanyl-9-oxo-5, 6, 7, 9~tetrahydrobenzo
[a] heptalen-7-yl-] -acetamide
NMR(400MHz, CDC13) ; 51.82-1.95(m, 1H) , 2.21-
2.28(m, 1H) , 2.31-2.42(01, 1H) , 2.45(s/3H), 2,47-2.53(m,
1H), 3.51(d, J=14.0Hz, 1H) , 3.64(s, 3H) , 3.66(d,
J=14.0Hz, 1H) , 3.88(s, 3H) , 3.93(s, 3H) , 4.66-4.72(m,
5.40(s, 2H) , 6.51(s, 1H) , 7.11(d, J=10.4Hz,
7.24-7.38(tn, 5H) , 7.48(3, 1H) , 7.90(3, J=7.2Hz, 1H)
95
Example 24: 3-(2-nitrooxy-ethyl)-N-[(75)-1,2,3-
trimethoxy-lO-methylsulfanyl-9-oxo-5,6,7,9-tetrahydrobenzo[
a]heptalen-7-yl] -benzamide
MeS
NMR (400MHz, CDC13) ; 52 . 05-2 .11 (m, 1H) , 2.17-
, 2H) , 2.46(s, 3H), 2.56-2.60(m, 1H), 2.81-2.91(m,
2H), 3.69(s, 3H) , 3.91(s, '3H), 3.97(s, 3H) , 4.45(t,
J=6.a,Hz, 2H) , 4.89-4.95(m, 1H) , 6.56(s, 1H) , 7.12-
7.24(m, 3H), 7.39(d, J=10.4Hz, 1H), 7.61-7.64(m, 2H),
7.69(8, 1H) , 8.22(d, J-=6.8Hz, 1H)
96
Example 25: Preparation of 3 -nihrooxy- benzole acid-S-
[(7S) -1,2, 3-trimethoxy-10-Tnethylsulfanyl-9-oxo-5, 6,7,9-
tetrahydro-benzo [a] heptalen-7-yl-carbamoyl] -pyridine-2 -
l -Methyles t er
Preparation of 3 -chlorome.thyl -benzole acid- (5-
_[_{7S) - 1 ,2 , 3 - trime thoxy- 1 0 -me t hyJLsu- If a n.yl- 9 -oxo- 5 ,6,7, 9 ^
tetrahydro-benzo [a] heptalen-7 -yl-carbamonyl] -pyridine-
2~yl } -methylester
MeO
WeO
MeO
A compotond prepared in step 1 of the Example, i
(100 mg, 0.19 mmol) was dissolved in 3 uifi of
dlchlororaethane. 3-(Chloromethyl) benzoylchloride
(0.030 ml, 0.21 mmol) and triethylamine (0.082 ml, 0.59
mmol) were slowly added therein, and the mixture was
reacted at room temperature for 10 minutes. Water was.
added to quench the reaction, arid aqueous layer was
97
extracted with dichlorornethane. Combined organic layer
was dried over anhydrous sodium sulfate, filtered and
concentrated under reduced pressure. The residue was
purified by column chromatoqraphy (ethyl acetate) , to
give 99 mg (yield: 79%, yellow solid) of the target
compound .
aH NMR (400MHz, CDC13) : 62 . 14-2 .16 (m, 1H) , 2.30-
2.39(m, 1H) , 2. 43-2. 45 (m, 1H) , 2. 48 (a, 3H) , 2. 68-2. 96 (m,
1H) , 3.67(s, 3H) , 3.90(sf 3H) , 3.91(s, 3H) , 4.83-4.93(m,
1H), 4.88(8, 2H) , 5.51(8, 2H) , 6.77(s, 1H) , 7.27(s, 1H) ,
7.52(t, J=7.7Hz, 1H) , 7. 65-7. 70 (m, 2H) , 8.05(d, J=7.7Hz,
8.14(8, 1H) , 8.28(d, J=10.6Hz, l H ) , - 9 . 0 2 ( s ,
Preparation of 3 ~ ji itrooxy-benzoic acid- { 5 -
[(7S) -1 ,2,3 -trimethoxy- 10 -methylsulfanyl-g-oxo-S, 6,7,9 -
fcetrahydro-benzo [a] heptalen-7-yl-car-bamoyl] -pyridine-2-
yl } -methylester
// 1)Nal, acetone
2)AgNO3, MeCN
98
A compound prepared in the step 1 (90 mg, 0.13
mmol) and sodium iodide (31 mg, 0.20 mmol) were
dissolved in 3 ml of acetone, and the mixture was
reacted at room temperature for 1 day. Water was added
to quench the reaction, and aqueous layer was extracted
with ethyl acetate. Solvent was concentrated under
reduced pressure. The reaction concentrate and silver
nitrate (3 Omg, 0.045 mmol) were dissolved in 5 ml of

acetonitrile, and the mixture was reacted at room
temperature for l hour. Water was added to quench the
reaction, and aqueous layer war; extracted with ethyl
acetate. Solvent was concentrated under reduced
pressure. The residue was purified by short column
nhromatography (ethyl acetate) and PLC, to give 26 mg
[yield: 29%, yellow solid) of the target compound.
XH NMR (400MH2, CDC13) : 52 . 09-2 .17 (m, 1H) , 2.35-
2.41(m, 2H) , 2.44(s, 3H) , 2.55-2.58(m, 1H) , 3.74(s, 3H) ,
43.91(8, 3H) , 3.96(3, 3H) , 4.92-4.95 (m, 1H) , 5.45(8,
2H) , 5.47(s, 2H) , 6.56(s, 1H) , 7.13(d, J=10.2Hz, 1H) ,
7.35(t, J-9.1HZ, 2H), 7.50(t, J=Q.5Hz, 1H), 7.54(s, 1H),
7.62(d, J=7.3Hz, 1H) , 8.14(d, J=B.3Hz, 2H) , 8.24(dd,
J«2.2, 6.2Hz, 1H, ArH), 8.36(d, J-6.9Hz, 1H) , 9.09(s,
1H)
99
Example 26~34
Compounds of Example 26 - Example 34 were
synthesized in analogy to the procedure as described in
Example 25, and intermediates were prepared by the
method described as follows.
intermediate 16> Preparation of 2-hyebco3cy-N- [ (7S) -
1,2/3-trimsthoxy-10-methylsulfanyl-9-oxo-5,6,7,9-
tetrahydro-benzo [a]I heptalen-7 - yl] - be nz amide
MeC .. ,, x
EDCI, HOBt, Et3N J—4 ] n I
OH
DMF
MeS ° MeS °
2-Hydroxy-benzoic acid (203 mg, 1.47 mmol) , EDCI
(385 mg/ 2.01 mmol) and HOBt (271 mg, 2.01 mmol) were
dissolved in dimethylforniamide (10 ml) . Triethylamine
(0.37 ml, 2.67 mmol) was added therein, and the mixture
was stirred at room temperature for 1 day. 7-Aminol,
2,3-trimethoxy-10-methylsulfonyl-6,7-dihydro-5Hbenzo[
al-heptalen-9-one (500 mg, 1.34 mmol) was added
100
therein, and the mixture was stirred at room
temperature for l day. Water was added to quench the
reaction, and aqueous layer was extracted with diethyl
ether. Combined organic layer was dried over anhydrous
sodium sulfate, filtered and concentrated under reduced
pressure. The residue was purified by column
chromatography (hexane: ethyl acetate = 1:5) and
recrystallized in methanol, to give 516 mg (yield: 78%,
yellow solid) of the target compound.
XH NMR (400MHz, CDCl3) : 52 . 14-2 . 17 (m, 1H) , 2.31-
2.37(m, 1H) , 2.40-2.44(3, 1H) , 2.45(s, 3H) , 2.56-2.60(m,
ev 3.74(S, 3H) , 3.91(8, 3H) , 3.97(s, 3H) , 4.85-
, 6.56(8, 1H) , 6.64(d, J=7.7Hz, 1H) , 6.77(d,
, 7.15(d, J=10.6Hz, 1H) , 7.22(d, J-7.3H3,
1H) , 7.38(d, J=10.6Hz, III), 7.57(s, 1H) , 7.71(d,
J=8.0Hz,
Preparation of 3-hydroxy-Jf- [ (7S) -
1, 2,3-trimethoxy-lO-methylsulf anyl-9-oxo-5, 6,7,9-
tetrahydro-benzo [a] heptalen-7-yl 3 -benzamide
101
MeQ
EDCI, HOST
•OH / DMF
MeS Me!
According to the similar procedure in the
preparation method of intermediate 16, by using 3-
hydroxybenzoicacid (497 mg, 1.33 mmol), 558 mg (yield:
85%, yellow solid) of the target compound was obtained.
XH NMR (400MHz, DMSO-d6): 62.07-2.15 (m, 1H) , 2.10-
2.35(m, 6H) , 3.55(s, 3H) , 3.80(s, 3H) , 3.84(s, 3H) ,
4.53-4.56(m, 1H) , 6.81(s, 1H) , 6.92 (d, J-6.8HZ, 1H) ,
7.10(8, 1H), 7.15-7.31(m, 5H) , 8.98(d, J-7.6H2, 1H, -
NH), 9.70(s, 1H, -OH)
Example 26; 4-nitrooxybutyric acid-5- [ (_7S) -1,2,3-
trimethoxy-lO-methylsulfanyl-9-oxo-5,6,7,9-tetrahydrobenzo
[a] heptalen-7-Yl-cairbainoyl ] -- p y r i d .1 tie - 2 - y 1 -
methylester
102
aH NMR (400MHz, CDC13) : 52 . 06-2 . 16 (m, 1H) , 2.08-
2.11(m, 2H) , 2.31-2.49{m, 1H) , 2.45(3, 3H) , 2.57(t,
J-7.1HZ, 2H), 3.75(s, 3H), 3.91(s, 3H) , 3.96(s,
3H) , 4.52(t, J=6.4Hz, 2H) , 4.91-4.94(m, 1H) , 5.21(s,
2H) , -6.56(8, 1H) , 7.13(d, J=i0.y][-., 1H) , 7.21-7.25 ( m ,
1H) , 7.38(01, J=10.6Hz, 111} , 7.52(s, 1H) , 8.22 (a.,
J-8.0HZ, 1H), 9.05(s, 1H)
27 : 3 -nitrooxymethyl -benzoicacid- 6- [ (7S) -1,2,3-
triraethoxy-10-methylsulfanyl-9-oxo-5, 6,1, 9-tetrahydrobenzo[
a]heptalen-7-yl-carbamoyl]-pyridine-2-ylmethylester
XH NMR (400MH2, CDCla) : SI. 84-1.92 (m, 1H) , 2.33-
2.60(m, 1H), 2.43(s, 3H), 2.46-2.54(m, 1H) 2.55-2.60(m,
1H), 3.73(S, 3H) , 3.92(3, 3H) , 3.96(3, 3H) , 4.74-4 .81 (m,
1H), 5.51(s, 2H) 5.57(d, J=4.0Hz, 1H) , 6.56(s, HI),
»
7.04(d, J=10.4Hz, 1H) , 7.25(d, J=10.4Hz, 1H) , 7.54-
7.60(m, 2H), 7.65(d, J=7.GHz, 1H), 7.87(t, J-8.0HZ, 1H),
7.99(d, J-=7.6Hz, 1H) , 8.21(s, 2H) , 8.22(8, 1H) , 8.41(d,
J«7.2Hz,
Example 28; 4-nitrooxybutyric acid-6- [ (7S)-1,2,3-
trimethoxy-10-methylsul£anyl-9-oxo-5,6,7,9~tetrahydrobenzo[
a]heptalen-7-yl-carbamoyl]-pyridine-2-ylme,
thyl ester
MeS
XH NMR (400MHz, CDC13) : 52 . 02-2 . 09 (tn, 1H) , 2.11-
2.18(m, 2H), 2.21-2.40(m, 2H), 2 . 4 3 ( s , 3H) , 2.47-2.56(m,
1H) . 2.63(t, J=7.2Hz, 2H) , 3.73(s, 311), 3 . 9 2 ( s , 311),
104
3.96(S, 3H) , 4.57(t, J=6.4Hz, 2H) , 4.77-4.83(m,
5.30(d, J-A.8llz, 2H) , 6.58(s, 1H) , 7.05(d, J=10.4Hz,
1H), 7.25(S, 1H) , 7.32(d, J=10.4Hz, 1H) , 7.52(d,
J-8.0HZ, 1H) , 7.84(t, J=a.OHz, 1H) , 7.98(d, J=8.0Hz,
1H) , 8.45(d, i7=6.8Hz, IB)
Example 29; 3-nitrooxymethyl-benzole acid-2-[(7S)-
1,2,3-trimethoxy-10-methylsu1fanyl-9-oxo-5,6,7,9-
tetrahydro-benzo[a]heptalen-7-yl-carbamoyl]-phenylester
•Me
XH NMR (400MHz, CDC13): 51. 48-1.55 (m, 1H) , 1.83-
1.89(m, 1H) , 2.22 -2.3 9 (m, 211), 2.42(s, 3H) f 3.62(s, 3H) ,
3.87(8, 3H) , 3.92(g, 3H) , 4 .63-4 . 70 (m, 1H) , 5.54(s, 2H) ,
6.47(s, 1H) , 6.89(d, J=7.4Hz, 1H) , 7.03(d/ J=10.2Hz,
1H), 7.17(s, 1H) , 7.23(d, J=10.1Hz, 1H) , 7.35(t,
J-7.3HZ, 1H) , 7.54(t, J=7.4Hz, III), 7.58(t, J=7.6I!::,
1H) , 7.72(d, J=8.0I-I2, III) ,7.03 (cl, J=7.7Hz, 1H) , 8.26(m,
2H)
105
Example 30: 4-nitrooxybutyric acid-2-[(7S)-1,2,3-
trimethoxy-10-methylsulfanyl-9-oxo-5,6,7,9-tetrahydrobenzo[
a]heptalen-7-yl-carbamoyl] -phenylesfrer
XH NMR (400MHz, CDG13) : 51.60-1.90 (m, 1H) , 2.10-
2.14(m, 2H) , 2.26-2.39(m, 1H) , 2.44(s, 3H) , 2.46-2.51(m,
1H) .2.56-2.61(01, 1H) , 2.80(t, J=7.1Hz, 2H) , 3.70(s,
3H), 3.92(s, 3H) , 3.96(s, 3H) , 4.55(t, J=6.2Hz, 2H) ,
4.74-4.81 (m, 1H) , G.57(s, 1I-I) , 6 . 82 (d, J=6.9Hz, IK),
7.08(d, J=10.6Hz, 1H) , 7.13(d, J=9.1Hz, 1H) , 7.23(s,
1H), 7.28-7.36 (m, III), 7.47 ( t , J=9.9Hz, 1H) , 7.68(d,
J=9.5Hz,
106
Example 31: 3-nitrogoxyrnebhyl-benzole acid-3- [ (7S) -
1,2,3-trimethoxy-10-methylsulfanyl-9-oxo-5,6,7,9-
tetrahydro-benzo [a]heptalen-7-yl-carbamoyl]-phenylesber
XH NMR (400MHz, CDC13) : 62 . 05-2 .15 (m, 1H) , 2.23-
2.59(m, 6H) , 3.75(s, 3H) , 3.91(3', 3H) , 3.97(s, 3H) ,
4,90-5.00(01, 1H), 5.49(8, 2H) , 6.56(3, 1H) , 7.03(d,
J-10.4HZ, 1H) , 7.27(s, 2H) , 7.34(d, J-8.8HZ, 2H) 7.53-
7.65(m, 2H), 7.76(3, 1H) , 7 . 7 7 ( d , J-6.4HZ, 1H), 8.13(m,
3H)
Example 32; 4-nitrooxybutyric acid-3-[(7S)-1,2,3-
trimethoxy-lO-methylsulfanyl-S-oxo-S,6,7,9-tetrahydrobenzo[
a]heptalen-7-yl-carbamoyl]-phenylester
107
XH NMR (400MHz, CDC13) : 62.04-2.18 (m, 3H) , 2.25-
2.60(m, 6H) , 2.65(t, iT=6.8Hs, 211), 3.74 (a, 3H) , 3.91(5,
3 H ) , - 3 . 9 6 ( s , 3H) , 4 . 5 6 ( t , J-6.4HZ, 2H) , 4 . 8 7 - 4 . 9 4 ( m ,
1H), .6.56(3, 1H) , 7.08-7.12 (m, 2H) , 7.26-7.35(m, 2H) ,
7.48(S7 1H) , 7.59(8, 1H) , 7.7l(d/ J=8.0Hz, 1H), 7.75 (d,
J=7.6Hz, 1H, -NH)
Example 33; 3-nitrooxymethyl-benzole acid-3-[ (7S)-
1/2,3-triTnethoxy-lO-methyl-sulfanyl-9-oxo-5y 6,7, 9-
tietrahydro-benzo [a] heptalen-7-yl-carbamoyl] -benzylester
"0-
108
XH NMR (400MHz, CDC13) : 52 . 05-2 .18 (m, 1H) , 2.31-
2H) , 2.40(s, 3H) , 2.55-2.59 (m, 1H) , 3.75(8, 3H) ,
3.91(s, 3H) , 3.97(s, 3H) , 4 . 90--1 . 96 (m, 1H) , 5.24(s, 211} ,
5.44(s, 2H) , 6.56(s, 1H) , 7.09(d, J=10.6Hz,
7.27(dd, J=7.S, 7.6Hz, 1H), 7.36(d, J=10.6Hz,
»
7.44(d, J"=7-.6Hz, 1H) , 7.45(dd, J=8..0, 7.6HZ,
7.58(d, J=8.0Hz, 1H) , 7.59(s, 1H) , 7.79(d, J=7.6Hz, 1H)
7.91(3, 1H) , 8.05(cl, J=7.6IIa, I I I ) , 0 . 0 6 ( 3 , 1H) , 8.10(d,
J=7.2Hz,
Example 34; 4-nitrooxybutyric acid-3- [(7S)-1,2,3-
trimethO3cy-10-methylsulfa.nyl-9-oxp-5, 6 , 7, 9-tetrahydrobenzo[
a]heptalen-7-yl-carbamoyl]-benzylester
.MeO
'B;
JH NMR (400MHz, CDC13) : 52 . 00-2 .14 (m, 3H) , 2.31-
2.51(01, 4H) , 2 . 4 4 ( s , 3H) , 2 . 5 6 - 2 . 6 0 ( m , 1H) , 3.75(s, 3PI) ,
3.92(8, 3H) , 3.97(s, 3H) , 4.49(t, J=6.2HZ, 2H) , 4.89-
4.95(m, 1H) , 4.99(d, J=12.4Hz, 1H) , 5.04(d, J-12.4HZ,
109
6.56(s,'lH), 7.11(d, J=10.6Hz, 1H) , 7.26(dd, J=7.6,
7.6H2, 1H) , 7. 35-7. 37 (m, 211), 7.54(s, 1H) ; 7.76(d,
J-B.OHz, 1H) , 7.81(S, 1H) , 8.00(d. J=7.6Hz, 1H)
Example 35 ; Preparation of 2 -nitrosothio-N- [ (7S) -1 , 2 , 3 -
trimethoxy- 10 -me thyl sul f anyl-9-oxo-5, 6,7, 9-tetrahydrobengo
[a] heptalen- 7 -yl] -benzamide
Preparation of 2 -mercapto-N- [ (7S) -1,2 , 3 -
trimethoxy-10 -me thyl sul fanyl - 9 - oxo -5,6,7,9 -1 e trahydro -
benzo[a]heptalen-7-yl]-benzamide
M
OH SH , 2) Et3N, CH2CI2
Excess thionylchloride was added in thiosalycilic
acid (115 mg, 0.75 mmol) , and the mixture was stirred
with heating for 1 day. The reaction mixture was
concentrated under reduced pressure to remove
thionylchloride, and to give a chloride compound. 7-
110
amino-l,2,3-trimethoxy-10-methylGulfonyl-6,7-dihydro-
Eff-benzo [a]-hepcalen-9-one (243 mg, 0.62 BBlol) was
dissolved in purified dichloromethane. Triethylamine
(0.26 ml, 1.86 mmol) was slowly added therein.
Thiosalycilic chloride dissolved in dichloromethane was
also added therein at 0°C, and the mixture was stirred
for 30 minutes. Water was added to quench t&ft reaction,
and aqueous layer was extracted with chloroform.
Combined organic layer was dried over anhydrous sodium
sulfate, filtered and concentrated under reduced
pressure. The residue was purified by column
chromatography (chloroform: me thanol = 12:1), to give
210 mg (yield: 66%, white solid) of the target compound.
XH NMR (400MH2, CDCl3) : 52. 06-2 .17 (m, 1H) , 2.33-
2.49(m, 2H) , 2.42(s, 3H) , 2.54-2.59(m, 1H) , 3.72(s, 3H) ,
3.92(S, 3H) , 3.96(8, 3H) , 4.86-4.93 (in, 1H) , 6.56(s,
1H), 7.06(d, J=10.6Hz, 1H) , 7.14(t, J=7.5Hz,
!7.24(d/ J«=7.7Hz, 1H) , 7.31(d, J=10.6Hz,
7.51(s, 1H) , 7.58(d, J=7.3Hz, Hi), 7.63(t, J=8
2H)
111
Preparation of 2-nitrosothio-W- ( (78) -1, 2, 3-
trimetho3cy-10-Tnethylsulfanyl-9-oxo-5,6,7,9-tetrahydrobenzo
[a] heptalen-7-yl] -benzamide
r~\ I
MeO ^^' 1A/-HCI. NaN02
Me°H
MeS MeS
Compound prepared in step 1 (40 mg/ 0.078 mmol)
was dissolved in methanol. 1 N HC1 aqueous solution (3
ml) was added therein. Sodium nitrite (NalNO2, 6.5 mg,
0.094 mmol) dissolved in water (1.5 ml) was also added
therein; and the mixture was stirred at room
temperature for 1 hour. Sodium hydrogen carbonate was
added to quench the reaction, and aqueous layer was
extracted with chloroform. Combined, organic layer was
dried over anhydrous sodium sulfate, filtered and
concentrated under reduced pressure. The residue was
purified by column chromatography (chlorbform:methanol
12:1) and recrystallized in methanol, to give 34.1 mg
(yield: 81%, yellow solid) of the target compound.
112
XH NMR (400MHz, CDC13) : S2 . 07-2 .14 (m, 1H) , 2.31-
2.37(m, 1H), 2 . 4 2 ( s , 3H), 2 . 4 5 - 2 . 4 8 ( m , 1H), 2.54-2.59(ra,
1H) , 3.72(s, 311), 3.91(s, 311), 3.96(s, 3H) . 4.88-
4.94(m, 1H) , 6.56(s, 1H) , 7.06(d, J=10.6Hz, 1H) , 7.10(t,
J=7.5Hz, 1H) , 7.21(t, J=6.9Hz, 1H) , 7.31(d, J»10.6Hz,
»
1H), 7.54(8, 1H) , 7.64(t, J=8.6Hz, 2H) , 7.72(d,
J-7.3HZ, 1H)
Example 36; Preparation of 3-nitroBooxytnatfcarly-[ (7S)-
l, 2,3-trimethoxy-10-methylsulf anyl-9-oxo-5, 6,7,9-
tetrahydro-benzo [a] heptalen -7- yl ] - ben garni de
Me
-M-x
1) Nal, Acetone
2) AgNO2, MeCN
MeS
3-Chloromethyl-W- [(7S)-1,2,3-trimethoxy-10-
methylsulfanyl-S-oxo-S, 6, 7, 9-tet.rahydrobenzo [a]heptalen
-7-yl] -benzamide (119.8 mg, 0.22.8 mmol) and sodium
iodide (136,5 mg, 0.911 mmol) were dissolved in acetone
(15 ml) , and the mixture was stirred at 55°C for 1 day.
The reaction mixture was extracted with chloroform and
113
washed with saturated sodium chloride aqueous solution.
Combined organic layer was dried over anhydrous sodium
sulfate, filtered and concentrated under reduced
pressure. The residue and silver nitrite (125.5 mg,
0.812 mmol) were dissolved in acetonitrile (5 ml) , and
the mixture was stirred at room temperature for 1 day.
The reaction mixture was extracted with chloroform.
Combined organic layer was dried over anhydrous sodium
sulfate, filtered and concentrated under reduced
pressure. The residue was purified by column
chromatography (ethyl acetate:chloroform = 4:1), to
give 35.4 mg (yield: 32.4%, yellow solid) of the target
compound.
XH NMR (400MHz, CDC13) : 52 . 35-2 . 55 (m, 3H) , 2.48{s,
3H) , 2.62-2.66 (m, 1H) , 3.76(s, 311) , 3.84(d, •J=12.ailz,
1H), 3.92(s, 3H) , 3.98(3, 3H) , 4.26{d, J=»12.8Hz, 1H) ,
4.91-4.97(m, 1H) , 6.58(a, 1H) , 6.96(dd, J^7.7, 7.7Hz,
1H), 7.22(d, J=10.6Hz, 1H) , 7.26-7.30 (m, 2H) , 7.44-
7.47(m, 2H) , 7.86(s, 1H) , 9.30(rl, J=6
114
Example 37 ; 3 -f luoro-S-iiitrosooxymethyl-jy- [ (75) -1, 2 ,.3 -
t r ime thoxy-10 - me thy 1 sul f anyl - 9 - oxo -5,6,7,9- tetrahydr o -
benzo[a]heptalen-7-yl] -benzamide
A target compound was synthesized in analogy to
the procedure as described in the Example 36.
•.Meo;
Me a
H NMR (400 MHz, CDC13) : 52 . 04-2 . 20 (m, 1H) , 2.47(s,
3H), 2. 52-2. 62 (m, 2H) , 2. 67-2. 72 (m, 1H) , 3.81(s, 3H) ,
3.91(S, 3H), 3.97(s, 3H), 4.71(s, 2H) , 4.93-4.96(m, 1H),
6.42(d, J=6.2, 1H), 6.58(s, III), 7.14-7.21 (m, 2H) ,
7.30(d, J=8.4, 1H), 7.42(s, 1H), 7.53(d, J=10.6,
115
Example 38; Preparation of 3-nitrosothiomethyl-jy- [ (75) -
1,2,3-trimethoxy-10-methylsulfanyl- 9-oxo-5,6,7,9-
tetrahydro-benzo[a]heptalen-7-yl]-benzamide
Preparation of methanesulfonic acid-3- [ (7SJ-
1,2,3-trimethoxy-10-methylsulfanyl- 9 -oxo-5,6,7,9-
tetrahydro-benzo[a]heptalen-7-yl-carbamoyl]-benzyl
ester
M
MeS
MeQ
MfiO
O
MeO
MeS MsO
3-Hydroxymethyl-N-[(7S)-1,2,3 -trimethoxy-10-
methylsul£anyl-9-oxo-5,6,7,9-tetrahydrobenzo
[a]heptalen-7-yl]-benzamide (252.5 rag, 0.497 mmol)
was dissolved in dichloromethane (10 ml), and the
temperature was lowered into o°C.
Methanesulfonylchloride (42.4 ìl 0.547 mmol) and
triethylamine (0.104 ml, 0.745 mmol) were added therein,
and the mixture was stirred at room temperature for 2
116
hours. The reaction mixture was extracted with
chloroform. combined organic layer was dried over
anhydrous sodium sulfate, filtered and concentrated
under reduced pressure. The residue was purified by
column chromatography (ethyl acetate: chloroform a 3:2),
to give 182.3 mg (yield: 62.6%, yellow solid) of the
target compound.
XH NMR (400 MHz, CDC13) : 62 . 14-2 .21 (m, 1H) , 2.31-
2.50(m, 2H) , 2.46(3, 3H) , 2. 57-2. 62 (m, 1H) , 2.85(s, 3H) ,
3.76(S, 3H) , 3.92(s, 3H) , 3. 97 (a, 3H) , 4.87-4.94(m, 1H) ,
5.06(d, i7=12.7Hz, III), 5.10(d, J-.i2.7Hz, 1H) , 6.57(s,
1H) , 7.14(d, J-10.6HZ, HI), 7.28(dd, J=7 .7 , 7.7HZ, 1H) ,
1.38-7. 42 (m, 2H) , 7.56(s, 1H) , 7.'74(d, J=8HZ1H) ,
df J=7.7Hz,
Preparation of thioacetic acid-_S-3-_[ (7S) -
1, 2, 3-trimethoxy- 10 -methylsulfanyl- 9-oxo-5, 6, 7,9-
tetrahydro-benzp [a] heptalen-7-yl-carbamoyl] -benzyl
ester
117
MeQ
A compound prepared in the step 1 (162.3 mg,
0.311 mol) was dissolved in acetone (6 ml) , and the
temperature was lowered into 0°C. Potassium thioacetate
(53.2 mg, 0.467 mmol) was slowly added therein at 0°C,
and the mixture was stirred for 1 hour. The reaction
mixture was extracted with chloroform. Combined
organic layer was dried over anhydrous sodium sulfate,
filtered and concentrated under reduced pressure, to
give 173.6 mg (yield: 90.6%, yellow solid) of the
target compound.
*H NMR (400MHz, CDC13) : 52 . 05-2 . 14 (m, 1H) , 2.30(s,
2H) , 2.32-2.49(m, 211), 2.44(0, 311), 2 . 52-2 . 61 (m, III),
3.74(s, 3H), 3.92(s, 3H) , 3.97(s, 3H) , 4.0l(d, 47=13. 9Hz,
1H) , 4.05(d, JXL3.9HZ, 1H) , 4. 87-4. 93 (m, |£) , 6.56(s,
7.10(d, J=10.3Hz, 1H) , 7.21(dd, J=7.7, 7.7H2,
.36(m, 2H) , 7.49(s, 1H) , 7. 64 -7. 71 (m, 3H)
118
Preparation of 3-mercaptomethylfsjpi-(7S) -1,2,3-
trimethoxy-10-methylsulfanyl-9-oxo-5,6,7, 9-tetrahydrobenzo
[a] heptalen-7-yl] -benzaml.de
MeS
HS
A compound prepared in the step 2 (165.2 mg,
0.292 mol) was dissolved in methanol (6 ml) , and the
temperature was lowered into o"0. Sodium thiomethoxide
(21.5 mg, 0.307 mmol) was slowly added therein at 0°C,
and the mixture was stirred at room temperature for 30
minutes. Reaction was quenched by adding 0.1N HCl
aqueous solution. The reaction mixture was extracted
with chloroform and washed with saturated sodium
chloride aqueous solution. Combined organic layer was
dried over anhydrous sodium sulfate, filtered and
concentrated under reduced pressure. The residue was
purified by column chromatography (ethyl
acetate:chloroform = 1:1), to give 182.3 mg (yield:
62.6%, yellow solid) of the target: compound.
119
H NMR (400MHz, CDC13): 51.70(t, J-7.3HZ, HI),
2.12-2.19(m, 1H) , 2.31-2.50(m, 2H) , 2.18(s, 3H) , 2.57-
2.61(m, 1H) , 3.58(d, J-=7.3Hz, 1H) , 3.'74(s, 3H) , 3.92(s,
3H), 3. 97 (a, 3H) , 4 .89-4 .95 (m, 1H) , 6.57(s/ 1H|, 7.12(d,
|7-10.3HZ, 1H) , 7.19(dd, J"=7.7, 7.7Hz, 1H) , 7.34(d,
r»7.7H2, 1H), 7.37(d, J-10.3II7., Ill), 7.55(s, I I I ) ,
7.59(d, J=7.7Hz, 1H) , 7 . 7 0 ( s , 1H) , 7,93(d, J=7.3Hz, HI)
Preparation of 3-nitrosothiomethyl-JV- [ (7S) -
1,2,3-trimethoxy-lO-methylsulf any 1 - 9 - oxo -5,6,7,9-
tetrahydro-benzo [a] heptalen-7-yl] -benzamide
MeQ
1N-HCI/NaN02
MeOH
O
MeS MeS
A compound prepared in the step 3 (101.2 mg,
0.193 tnol) was dissolved in rnethanol (3 ml) and
dimethylformamide (3ml) , and the temperature was
lowered into 0°C. 0.1N HCl aqueous solution (3 ml) and
120
sodium nitrite (16.0 mg) were slowly added therein at
0°C, and the mixture was stirred at room temperature
for 2 hours. The reaction mixture was extracted with
Chloroform and washed with saturated sodium carbonate
solution. Combined organic layer was dried over
anhydrous sodium sulfate, filtered and concentrated
under reduced pressure. The residue was purified by
column chromatography (ethyl acetate:chloroform:hexane
3:2:1.5), to give 19.5 mg (yield: 18.3%, yellow
solid) of the target compound.
aH NMR (400MHz, CDC13) : 52.08-2 .16 (mf 1H) , 2.31-
2.51(m, 2H) , 2.45(8, 3H), 2.57-2.61(m, 1H) , 3.57(s, 3H),
3.80(s, 2H), 3.92(s, 3H) , 3.97(s, 3H) , 4.89-4.96(m, 1H) ,
6.57(S, 1H), 7.10(d, J=10.3Hz, 1H) , 7.25(dd, J=7.7,
7.7HZ, 1H) , 7.36(d, J=10.3I-Iz, 1.11), 7.38(d, J=7.7Hz, III),
7.53(3, 1H) , 7.68(d, J=7.7Il2'./ 111), 7.77(8, 1H) , 7.79(d;
i7«7.0Hz,
121
Example 39; 3-fluoro-5-nitrosothiomethyl-N- f (78) -1,2,3 -
trimethoxy- 1 0 -me thy 1 sul f anyl - 9 - oxo -5,6,7,9- te trahydro -
benzo [a] heptalen-7-yl] -benzamide
A target compound of Example 39 was , synthesized
in analogy to the procedure as described iti^trHBi Example
38.
McD-
•.Me
H NMR (400MHz, CDC13): 62.30-2.48(m, 3H) , 2.44(s,
3H), 2.56-2. 59 (m, 1H) , 3.57(q, J=13.9, 16.5, 2H) ,
3.73(s, 3H), 3.91(3, 3H) , 3.97(s, 3H) , 4.97-5.03(m, 1H)
6.57(8, 1H), 7.02(d, J=8.4, 1H) , 7.15(d, J«10.6, 1H) ,
7.39(d, J=10.2, 1H) , 7.43(s, 1H) , 7.64 (d, J=9.1, HI),
7.73(s, 1H) , B.96(d, J=7.3, III)
122
Example 40; Preparation of 3-fluorg-j-nitrooxymethyl-M-
[(7S) -l,2,3,10-tetrametho:Ky-9-o:x:o-5, 6,7, 9-tetrahydrobenzo[
a]heptalen-7-yl]-benzamide
[ (78) -1,2,3,10-tetramethoxy-9-0X0-5, 6,1, 9-tetrahydrobenzo[
a]heptalen-7-yl]-benzamide
DH
:DMF
Jpeacetylcolchicine (150 mg, 0.42 mmol), 3-fluoroithyl-
benzoic acid (85 mg, 0.50 nvmol) and
HOBt (67 mg, 0.50 mmol) were dissolved in
dimethylformamide solution (2 ml) , and the temperature
was lowered into 0"C. EDCI (95 mg, 0.50 mmol) was
slowly added therein at 0°C, and the mixture was
Stirred at room temperature. Water was added to quench
*^ ' •
the reaction, and aqueous layer was extracted with
ethyl acetate. Combined organic layer was dried over
anhydrous sodium sulfate, filtered and
123
under reduced pressure. The residue was purified by
column chromatography (chloroform: ethyl acetate = 2:1),
to give 110 mg (yield: 52%, yellow solid) of the target
compound.
1H NMR (400MHz, CDC13) : 52 . 40-2 . 58 (m, 3H) , 2.62-
2.69(tn, 1H) , 3.64-3.70 (in, 1H) , 3.74(s, 3H) , 3.92(s, 311),
3.98(8, 3H) , 4.07(S, 311), 4 . 23-4 . 20 (m, 1H) , 4 .81-4 . 88 (m,
1H), 6.58(a, 1H) , 6.87(d, J=9.2Hz, 1H), 6.97(d, J=9.2Hz,
1H), 7.03(d, i7=10.4Hz, 1H) , 7.38(8, 1H) , 7.50(d,
•7-10.4Hz, 1H) , 7.96(s, 1H) , 9.64 (A, J=6.0Hz, 1H)
Preparation of 3-fluoro-5-nitrooxyreethyl-N-
[ (78) -1,2,3,10-tetramethoxy-9_-oxo-5_/ 6,_7 / 9-tetrahydrobenzo[
a]heptalen-7-yl]-benzamidc
MeO •^QFf MeO
According to the similar procedure in the step 2
and 3 of Example 5, by using a compound prepare in the
124
step 1 (90 mg, 0.10 mmol), 30 mg (yield: 30%, yellow
solid) of the target compound was obtained.
H NMR (400MHz, CDC13) : 52.30-2.49(m, 3H) , 2.58-
2.59(m, 1H) , 3.75(8, 3H) , 3.92(s, 311)3.98(8, 311),
4.05(S, 3H) , 4.89-4.93(m, 1H) , 4.94(d, J=12.8Hz, 1H) ,
5.10(d, ,J=12.eHz:, 1H) , 6.58(s, 1H) , 6.96 (d, J-8.4H2,
1H) , 7.01(d, J=10.4Hz, 1H) , 7.17(d, J=8.4Hz, 1H) ,
,7.45(8, 1H) , 7.48(d, J=10.4Hz, 1H) , 7.89{s, 1H) , 9.25(d,
Example 41; Preparation of 3-nitrooxymethyl-i^i^thyl-N-
[ (75) -l^^-trimethoxy-lO-methylsulfanyl-g-oxo-S^,?, 9-
tetrahydro-benzo [a] heptal «3n~_7_-yl_] - be viz amide
Preparation of 3-chloromethy|^i^|ethyl-N-
*-l,2,3-brimethpxy-10-methylsulfanyl-9-oxo-5<.6> tetrahydro-benzo[a] heptalen-7-yl]-benzamide
PJridih.il-
MC
Ma
Mei
125
Thiodemecolcine (50 mg, 0.129 mmol) and pyridine
(0.012 ml, 0.154 mmol) were dissolved in
dichloromethane, and the temperature was lowered into
O°C. 3-(Chloromethyl)benzoyl chloride (0.022 ml, 0.154
mmol) was slowly added therein at 0°C, and the mixture
was stirred at room temperature. Water was added to
guench the reaction, and aqueous layer was extracted
with ethyl acetate. Combined organic layer was dried
over anhydrous sodium sulfate, filtered and
concentrated under reduced pressure. The residue was
purified by column chromatography (chloroform:ethyl
-acetate solid) of the target compound.
aH NMR (400MHz, CDC13) : 52.24-2.38 (m, 2H), 2.44(s,
3H),2.45-2.58(m, 1H) , 2.61-2.74(m, 1H) , 3.25{s, 3H) ,
3.72(8, 3H) , 3.91(s, 3H) , 3.92(s, 3H) , 4.54(s, 2H) ,
5.05(br, 1H) , 6.57(3, 1H) , 7.06(3, J=10.0Hz, 1PI) ,
7.09(S, 1H) , 7.22-7.41(m, 511)
126
Preparation of 3-nitrooxymethyl-N-methyl-N-
[(7S)-1,2,3-trimethoxy-lQ-methylsulfanyl-9-oxo-5y6,7,9-
tetrahydro-benzo[a3heptalen-7-yl3-benzamide
iJNal/Acetone
•ii)AaN03/;c:H3CN
According to the similar procedure in the step 2
of Example 25, by using a compound prepared in the step
1 (45 mg, 0.085 mmol), 40 mg (yield: 83%, yellow solid)
pf the target compound was obtained.
XH NMR {400MHz, CDC13): 52.24-2.38(m, 2H), 2.44(a;
3H) ,2.45-2.58 (m, 1H) , 2 . 61-2 . 74 (in, Hi), 3.24(s, 311),
3.71(8, 3H) , 3.91(s, 311), 3.92(s, 3H) , 4.77(br, 1H) ,
5.58(s, 2H), 6.85(s, 2H), 7.21(d, J=10.0Hz, 1H), 7.29{d,
J-10.4HZ, 1H), 7.40-7.53(m, 4H)
Example 42: Preparation of 3 -fluoro-N-methyl-5-
taltrooxymethyl -N- [ (75) -1,2, 3-l:rimethoxy-10-
127
mebhylsulfanyl-9-oxo-5,6,7,9-tetrahydrobenzo
[a]heptalen-7-yl]-benzamide
Preparation of 3-fluoro-5-hydroxytnethyl-Nmethyl-
N- [(75)-1,2,3-trimethoxy-10-methylsulfanyl-9-
oxo-5,6,1, 9-tetrahydro-ben2O[a]heptalen-7-yl}-benzamide
MoO
HO
>=0 i)Bhyl diloroforma(e,.TEA
' lOPjfriiljha
.MP
HO'
3-Pluoro-5-hydroxymel:hyl-benzoic acid (52 mg,
0.309 mmol) was dissolved in dichloromethane (3 ml)
under a nitrogen atmosphere. Ethylchloroformate (0.022
ml, 0.231 mmol) and TEA (0.042 ml, 0.309 mmol) were
Slowly added therein at 0°C, and the mixture was
Stirred at 0°C for 30 minutes. Pyridine (0.012 ml,
0.154 mmol) and thiodemecolcine (60 mg, 0.154 mmol)
were also added therein, and the mixture was stirred at
room temperature f or 3 hours. Water was added to
guench the reaction, and aqueous layer was extracted
with ethyl acetate. Combined organic layer was dried
over anhydrous sodium sulfate, filtered and
128
concentrated under reduced pressure. The residue was
purified by column chromatography (chloroform: ethyl
acetate = 2:1), to give 30 rag (yield: 21%, yellow
solid) of the target compound.
aH NMR (400MHz, CDC13) : 51.76(br, 1H) , 2.24-2.38(m,
2H) , 2.44(s, 3H) ,2.45-2.58 (m, III), 2 . 61-2 . 74 (m, Hi),
3.23(8, 3H) , 3.72(s, 311), 3.93(s, 3H) , 3.97(s, 3H) ,
4.65(s, 2H) , 5.02(br, 1H) , 6.57(8, 1H) , 6.92(s, 1H) ,
6.99-7.24 (m, 4H) , 7.34(s, III)
Preparation o£ 3-f luoro-N-methyl- 5 -
nitrooxymethyl-N- [ (7S) -1,2,3-trimethoxy-lOmethylsuIf
anyl-9-oxo-5,6,7, 9-1etrahydrobenzo
[a] heptalen-7 -yl] -benzamicle
^CH-
0'.
According to the similar procedure in the step 2
and 3 of Example 5, by using a compound prepared in the
129
step 1 (55 mg, 0.101 mmol) , 20 mg (yield: 37%, yellow
solid) of the target compound was obtained.
XH NMR (400MHz, CDC13) : 52. 22-2 .41 (m, 2H) , 2.45(s,
3H) , 2.45-2. 58 (m, 1H) , 2 . 61-2 . 74 (in, 1H) , 3.24(s, 3H) ,
»
3.75(s, 3H) , 3.91(8, 311), 3.95 ( a , 3H) , 5.02(br, 1H) ,
5.39(s, 2H) , 6.57(s, 1H) , 7 . 0 5 ~ 7 . 2 4 ( m , 5H) , 7.34(s,
Example 43 : _Prepcirati_o_n. of 2- (3-£luoro:1 bnitrooxymethyl-
phenyl) -N- [ (7S) -l, 2, S-trimethQanr-lOmethylsulfanyl-
9-oxo-5,6, 7, 9-tetrahydrobenzo[
a]heptalen-7 -y1]- acetamide
Preparation of (3-fluoro-5-hydpq«ymethylphenyl)-
acetic acid
Matt
THF
KCM, iQ-'Crovvn-B.-
Br — —- - MeO:
BOH/:-H2o; H0_, OH
A compound prepared in the step 1 of intermediate
6 (2.5 g, 13.57 mmol) was dissolved in dichloromethane
130
(30 ml). PBr3 (1.15 ml, 12.21 mmol) was slowly added
therein, and the m.ixture wan stirred at room
temperature for 3 hours to give 3-bromomethyl-5-fluorobenzoic
acid methyl ester (1.6 g, yield: 50%, white
solid). 3-Bromomethyl-5-fluoro-benzoic acid methyl
»
ester (1.5 g, 6.1 mmol), KCM (l.G g, 24.4 mmol) and 18-
crown-6 (820 mg, 3.1 rmnol) were dissolved In
acetonitrile (10 nil) . The re si CM. ion mixture was stirred
at room temperature ior 10 houra to give 3-cyanomethyl-
5-fluoro-benzoic acid methyl ester (900 mg, yield: 77%,
white solid). 3-Cyanomethyl-5-fluoro-benzoic acid
methyl ester (900 mg, 4.65 mmol) was dissolved in
tetrahydrofuran (10ml) . 2M Lithiutnborohydride
tetrahydrofuran (2.3 ml, 4.6 mmol) was slowly added
therein, and the mixture was re fluxed, to give (3-
fluoro-5-hydroxymethyl-phenyl)-acetonitrile (430 mg,
yield: 56%, white Kc.licl) . (3 1'luoro-5-hydroxymethylphenyl)
-acetonitrile (400 ny, 2.42 mmol) and
potassiumhydroxide (1.34 g, 23.8 mmol) were dissolved
in ethanol (10 ml) and water (5 ml) . The reaction
mixture was re fluxed for 24 hour;:: to give 356 mq
(yield: 80%, white uolid) of the target compound.
1H NMR(400MHz, CD3OD); 53.62(s, 2H), 4.59(3, 2H),
6.94(d, J=9.6Hz, 1H), 7.04(d, J=9.6Hz, 1H), 7.08(s,
131
Preparation of 2- (3-fluoro-5-hydroxytneth>lphenyl)-
N-[(7S)-1,2,3-trimethoxy-lO-methylsulfanyl-9-
oxo-5,6,7,g-tetrahydro-benzo[a]heptalen-7-yl3-acetamide
Me
OH
>+ V fr
HO^
:0
EDCI,.H0Bt
DMF
MeS
According to the similar procedure in the step 1
of Example 40, by using a compound prepared in the step
1 (30 mg, 0.16 mmol) , 58 mg (yield: 70%, yellow solid)
of the target compound was obtained.
:H NMR (400MHz, CDC13) : 51 . 92-2 . 01 (m, 1H) , 2.19-
2.40(m, 2H) , 2.4l(s, 3H) , 2 . 46-2 . 52 (m, 1H) , 3.4S(d,
J=14.0,Hz, 1H) , 3.52(d, J=14.0,Hz, 1H) , 3.63(s, 3H) ,
3.89(s, 3H) , 3.91(0, 3H) , 4 . 64-4 . 74 (m, 1H) , 4.75(s, 2H) ,
6.53(s, 1H) , 6.86-6.94(m, 3H) , 7.07(d, J-=10.8Hz, III),
7.13(s, 1H) , 7.30(d, J=10.8Hz, 1H) , 7.39(s,
132
Preparation of methanesulfonic acid 3-fluoro-
5-[(7S)-(l,2,3-trimethoxy-lO-methylsul£anyl-9-oxo-
5,6,7, 9 - tet r ahydro - ben20 [a ]lie;p t: ci1 on ~ 7-ylcarbamoyl) -
methyl]-benzyl ester
MSCI;TEA
MC.
'Mi
According to the similar procedure in the step 1
of Example 38, by using a compound prepared in the step
2 (58 mg, 0.107 mmol) , GO mg (yield: 90%, yellow solid)
of the target compound was obtained.
Preparation of 2-(3-flu o r o - 5 - n i t rooxytne t hy 1 -
phenyl) -N- [ (7S) -.1 ,_2 , 3 - triiuethoxy-10 - me thy 1 sul f anyl - 9 -
oxo-5,6,7,9-tetrahydro-benzo[a] heptalen-7-yl]-acetamide
133
Me'p
'i)|\lal/Acetone- MeQ-if

According to the similar procedure in the step 2
of Example 25, by using a compound prepared in the step
3 (60 mg, 0.097 mmol) , 30 nig (y:u-:ld: 83%, yellow solid)
of the target compound was obtained.
H NMR (400MHz, CDC13) : 51. 92-1.98 {m, 1H) , 2.22-
2.42(m, 2H) , 2.46(s, 311), 2.49-2.52(m, 1H) , 3.50(d,
J=14.0,Hz, 1H) , 3.651::;, 3ii:, 3 . b6 (d, iJ=14 . 0 , Hz
1H) , 3.89(s, 311), 3.93(s, 311), 4. 64-4. 74 (m, 111),
5.37(s, 2H) , 6. 52 (a, 1H) , 6.97(d, Jr=8.8Hz/ 1H) , 7.11-
7.17(m, 3H) , 7.35(d, J=10.8Hz, 1H) , 7. 50 (a, 1H) ,
8.04(d, J=7
Example 44 ; 2- (2-f luoro-5-niti-ooxymethyl-phenyl) -N-
[{7S) -l/Z/B-trimethoxy-lO-methylsulfanyl-g-oxp-S, 6, 7, 9-
tetrahydro-benzo [a ] hep I; a 1 e 1 1 - 7 - y 1 ]_-acetaamide
A compound of Example 44 was synthesized in
t
analogy to the procedure as described in Example 43,
134
and an intermediate was prepared by the
described as follows.
Me
XH NMR (400MHz, CDC13) : 61. 09-1. 93 (m, 1H) , 2.26-
2.29(m, 1H), 2.34-2.40(m, Hi), 2.43(8, 3H), 2.49-2.54(m,
1H) , 3.61(s, 311), 3.G2(d, .T. l-;, . lil, h i 1 H ) , 3.72(0,
J=15.2Hz, 1H) , 3.89(s, 311), 3.92(0, 311), 4.70-4.75(m,
1H) , 5.49(s, 2H) , 6.53(s, 1H) , 7.06-7.11(01, 2H) , 7.25-
7.31(01, 2H) , 7.35(t, J=7.6Hz/ 1H) , 7.44(s, 1H) , 7.57(d,
J«7.2Hz,
Preparation of (2-£luoro-5-
hydroxymethyl-phenyl)-acetic acid
MeO
XOH
135
According to the similar procedure in the step 1
of Example 43, by using a compound prepared in the
intermediate 5 (2.2 g, 11.94 mmol), 400 mg (yield: 90%,
white solid) of the target compound was obtained.
Experimental Example 1: Cytotoxicity test to cancer
cell lines
Cytotoxicity to A549 (Korea Research Institute of
Chemical Technology) , SK-OV-3 (Korea Research Institute
of Chemical Technology), SK-MEL-2 (Korea Research
Institute of Chemical Technology), HCT-15 (Korea
Research Institute of Chemical Technology) and MCF7
(Korean Cell Line Bank, Seoul National University
School of medicine} cells was measured by
Sulforhodamin-B (SRB) method (1909, National Cancer
Institute (NCI)) which was developed for the
measurement of in vitro anticancer activity of a drug.
Cells were separated using 0.25% trypsin-EDTA solution,
leading to the preparation of a cell suspension by
5xl03 - 2xl04 cells/well. Then, the suspension was
distributed into a 96 well plate by 100 ìl/well, which
Was cultured in a 37°C, 5% CO-, incubator for 24 hours.
Compounds prepared in examples of the present invention
were used as a sample. Precisely, the compound way
136
dissolved in dimethylsulfoxide and diluted with RPMI
1640 medium before being uso.d a.s a sample. The final
concentration of the sample used varied ranging 1 ìM ~
0.00001 ìM. The medium was removed from the 96 well
plate and then diluted sample solution was added by 100
ìl/well, followed by further culture in a 37 C, 5% C02
incubator for 48 hours. Tz (time zero) plate was
collected from the point of adding the sample. Upon
completing the culture, the medium was removed from
each well along with Tz plate, then 10%
trichloroacetic acid (TCA) was added by 100 ìl/well.
The plate was left at 4 C for 1 hour to let cells be
fixed on the floor of the plate. After cells were
fixed completely, the plate was washed with water 5-6
times to remove the remaining trichloroacetic acid
solution completely and moisture was completely dried
at room temperature. 0.4% Sulforhodamine-B was
dissolved in 1% acetic acid solution to prepare a
staining solution. Cell.s were stained for 30 minutes
with a dye added by 100 ^ to each well of the
completely dried plate. Then, the plate was washed
with 1% acetic acid solution 5-6 times to remove
sulforhodamine-B remained uncombined with cells. Then,
the plate was dried at room temperuture. 10 mM Tris
solution was added by 100 ìl/well thereto to dissolve
137
the dye, and optical density (OD) was measured ' at 520
nm with a micro plate reader.
ED50 (concentration that inhibits cancer cell
growth 50%, 50% effective dose., nM/ml) of the sample to
cancer cell was calculated aa follows. OD value at the
point of beginning the culture with the sample was
determined as Tz (time zero) value. OD value of a well
to be cultured without the cample was determined as a
control value (C) . OD vaJue of a well pretreated with
the sample was determined as experimental value (T) .
After calculating T?. , C and T, cytotoxicity of the
sample was measured by the: below ^Mathematical 'Formula

T£ Tz, (T-Tz) / (C-Tz) .x 100
T > Tz., (T-T2.) /Tz -A 100
ED50, the concentration that can inhibit cancer
cell growth by 50%, wan calculated by using, a
regression analysis of .lotus program based on the
degree of cytotoxicity obtained by the Formula 1>.
138
Each EDSO of paclitaxed, doxorubicin and
colchicine, which were used as controls, was also
calculated by the same way as described in the above.
Results were presented in Table 1.
Cytotoxicity to cancer cell lines (NT: Not tested)
Example
Paclitaxel
Doxorub i c i n
Colchicine
Example 1
Example 2
'Example 3
Example 4
Example 5
Example 6
Example 7
Example 8
Example 9
Example 10
Example 11
Example 12
Example 13
Example 14
Example 15
Cytotoxicity [ED50 : nM]
A549
0.3
13 .0
21.0
1.8
15.8
2.4
0. 01
.1. . 0
0.70
>50
0.02
0.49
0 .20
>50
0.05
0.13
28 .2
30.2
SK-OV-3
1.2
47.0
18.0
1.0
9.3
4.0
0 . 01
1 . U
0.37
>50
0.04
0.34
0.23
>50
0.27
0.10
30.6
>50
SK-MEL-2
0.1
16.0
6.0
0.4 '
5.4
1.0
0 . 0 1
.1 . 1
0 .08
>50
0.01
0 .22
0 .12
>50
0.11
0.04
32. 0
22.5
HCT15
0.1
25.0
9.0
0.6
2.3
4.8
0.03
4.4
0.27
>50
0.03
0.64
0.39
>50
0.05
0.11
9.9
23.4
MCF-7
0.9
50.0
NT
NT
2.9
2.3
0.01
NT
0.16
>50
0.01
0.13
0.09
39.0
0.03
0.02
11.2
18.8
139
OH
os e-Z.fr
S'OI
O'S
.IN
e foi
•IN
L'TrZ
T O ' O
,IN
•LN
,IN
UN
iN
IN
JIN
£N
£N
e-e
T Z 'O
O T ' O
S T ' T
2'8
J.JST
80'0
IM
OS >'9e
E ' f r S
6'6
0' E
S'Z,
/ L ' O
6*02
T O ' O
O ' T
80 '0
8*0
Z'S
8*0
o- e
0' Z
0'OS OVSI
fr-e
S6**0
ei'o
E O ' T
T'OI
£ 'S
S I 'O
O ' T
os os L' 09
OG E' E
T > ' 6
T ' O
6 'T17
TO' 0
n i ,
5 0 ' 0
6 • 0
. 9 ' £
6 ' 0
0' t'
0 ' /.
Q'LT?
O ' T r T
S'e
/L8'0
62 - 0
L I- • Z
0 ' T E
0 ' 1
LI' 0
0 ' T
os os 9'S!>
Z ' K Z
I' :l7
8'8E
T ' T
OS TO' 0
9 ' D
O ' T
0' E
Z ' B
O ' E
0 ' S'l
( ) ' 9 't:
0'OS fr'S
9 'S
T^'T
G S ' O
E O ' T
O ' ^ E
8' e
G T ' O
O ' T
os os 6 ' £ ^
^,'01>
17' 2
os 6'Z
05 9 0 ' 0
» ' E
Z'S
O T ' T
S ' E
T ' T
0 ' 9
o - g
O ' T C
8'8
fr'9
T T ' T
L G ' O
E 6 ' E
S'ES
T ' 8
T S ' O
9 ' T
Zf. stduiBxg
Tfr siduiBxg
0^ s"[duiBxg
6E sjdujBxg
8£ aiduiBXg
^e STduiBxa
9E ajduiBXH
5E siduiHxa
^e 3T.durexg
EE ajduiHxg
SE aiduiexg
IE s-[durexa
oe siduiexg
62 9-[duiBX3
82 sfduiexg
Z^S 9-[duiexg
92 afdurexg
92 atduiexg
E2 aiduiHxg
ZZ ai;duiexg
is ajduiexg
OZ aidurexg
6T sidui^xa
8T STduiBxg
LI 9-[duiBxg
9T aiduiexg
As shown in Table 1, tricyclic derivatives
according to the present invention showed very strong
cytotoxicity to cancer cell lines.
Experimental Example 2: Inhibiting effect of tricyclic
derivatives of the present invention on tumor growth
In order to investigate inhibiting effect of
tricyclic derivatives of the invention on tumor growth,
following experiments were performed.
Samples for the experiment were stored in a
refrigerator all the time. Different dosages of
compounds were administered to a test animal; dosage of
a compound prepared in the Example 8 was 10 mg/kg,
dosage of a compound prepared in the Example 12 was 1,
3, 10 mg/kg respectively and dosage of a positive
control was 2.5 mg/kg (Taxol) and 2 mg/kg (Adriamycin).
The compound of the Example 12 was dissolved in 4%
tween 80, and the positive control Taxol was dissolved
in a mixed solvent of 5% athanol + 25% cremophor + 75%
PBS. The prepared sample is subject to be precipitated,
so that tip sonication was performed right before the
administration to disperse it well.
7 week-old female S.P.F. BALB/c nude mice,
provided by Charles Rivet: Co., Japan, were used as teat
141
animals. The test animals were adapted in a Hepafilter
room over a week before the test. The
temperature was 21±2°C, humidity was 55±5% and 12-hour
light and dark cycle was automatically repeated in that
lab. Solid feed (CheilJedang) was sterilized by
radioactive rays and drinking water was also sterilized
by autoclave. Feed and water were taken by the animal
freely. Cancer cell line used .in this experiment was
NCI-H460 (human lung tumor cell line) provided by Korea
Research Institute of Bioscience and Biotechnology.
The tumor cell line, stored in liquid nitrogen,
was thawed and cultured in a 37 °C, 5% C02 incubator for
a required time. Upon completing the culture, all the
cells were recovered and cell concentration of the
culture fluid was adjusted using PBS to 3xl07 cells/ml.
The adjusted cell culture solution was injected
hypodermically into armpit between right shoulder
girdle and chest wall by 0.3 ml per mouse. From the
next day of grafting, NCI-H460 xenografted nude mice
were administered intraperitoneal everyday with the
sample solution by 0.2 ml/20 g of weight, once a day.
After the grafting of tumor cells, the volume of
a tumor in each individual was measured in three
dimensions by using a vernier caliper, which was
represented in the below Mathematical Formula 2>.
142
Mathematical Formula 2>
Tumor volume = (lengUixwidthxhoight) /2
Body weight changes of animal were measured three
times a week. Each xenografted nude mouse was
sacrificed to separate a tumor, which was then weighed.
All the test results of experimental groups were
compared with those of control groups by t-Test to see
if there is any significant difference between the two
groups.
Changes of the volume and the weight of a tumor
were shown in Table 2 and in FIG. 1, 3, and 5, and
changes of the body weight of a mouse were shown in
Table 3, in FIG. 2 and in FIG. 4.

Experime
ntal
Group
V.C-1
V.C-2
Example
8
Example
12
Dosage
(mg/ks)
0
0
10
1
3
Tumor volume (mm1)
Day 0
0.0±
0.0
0.0 +
0.0
o.o±
0.0
0.0±
0.0
o.o±
0.0
Day 4
28.3 +
8.1
26.7 +
3.8
18. 0±
3.7*
20. 7±
3.1
16 . 9±
5 . 1 *
Day 7
53. 5±
13 .7
•iy .6+
12.3
25. 2±
12.8**
49.2+
12.2
31.2 +
1 1 . 7 *
Day 9
90.3 +
2 _S . 5
00 . !) +
26.7
29.6 +
13.0**
*
78.1 +
13 . 0
49.7 +
1 r^ . 0 A *
Day 11
167. 5±
43.8
130.2±
54.9
35. 2±
14.0**
*
146.S±
n .B
9S.1 +
2n . 1**
Day 14
295. 2±
62.2
289. 0±
53.1
44. 5±
15.0***
299. 3±
41.5
206. 8±
48. 0*
Tumor
Weight
(mg)
Day 14
1171.2
±303 .5
1087 .9
±300.5
143.4+
57.8**
*
950 . 9+
174.8
714.5+
103 . B *
Taxol
Adriamyc
in
10
2.5
2
0 . 0±
0 . 0
o.o±
0 . 0
0.0±
0.0
.'LS.l-J;
6.7*
17. 5±
4.6***
21. 8±
8.3
2 9.0* *
32. 7±
3.8**
33.7+
8.7*
3 b . 2 ±
12.9 * *
*
47.2±0
47. 6±
16.2**
G9.4±
26.9**
*
~*
81. 6±
24.5**
103. 0±0
~
138. 1±
39.9***
390 .9:1;
0
"
534. 1±
87.9
* Significance test (t-Test) : *(p ***(p V.C-1 : 4% tween 80,
V.C-2 : 5% ethanol + 25% cremophor -i- 75% PBS

Experime
Til- nc 9a 1x
Group
V.C-1
V.C-2
Example
8
Example
12
Taxol
Adriamyc
in
Dosage
(nig/ kg)
0
0
10
1
3
10
2 .5
2
Aniuun 1
(n)
6
6
5
6
6
6
(Day 14-
2}
c>
(Day :)-
9
Day 11,
Day 14-
1)
6
Day
1
100. 0±
0.0
100.0+
0 . 0
100. 0±
0 . 0
100. 0±
0.0
100 .0+
0.0
100. 0±
o . d
100 . 0:i
0 . 0
100. 0±
0 . 0
Hocly
Day
3
105. 4±
2.5
106. 9±
2 . 4
103 .5±
2 9
105.0+
1.4
102.8+
2 . 0
103.9+
0 . 9
ll)o . 0-i
.1. . ' •
105.0+
4.1
w Day
7
108.7
±
2.4
110. 5±
1 . 5
99. 0 +
4 . 3 * *
104 , 2±
5.3
101. 0±
4.2**
92. 2±
~\ 15 * * *
9 - ' . !!•!;
.' ] . 1 •'• * 'A'
103 . 9+
5.5
Change
Day
9
107. 2±
3.7
108. 5±
2 .6
92. 3±
4 _ 1***
99. 6±
4.4**
99. 3±
3.3*
89. 4±
2 .0**
y v . i ± o
100. 5±
4.6*
1 (%)
Day
11
107. 8±
4.6
109. 0±
52.6
89. 3±
3.7***
97. S±
3.6**
99. 1±
2.8**
92. 3±
2.9***
96. 4±
3 .5***
Day
14
107. 8±
3.2
108.9+
3.6
89.4 +
5.3***
98.3-1
3.2***
94. 5±
3 _ 9 * * *
93.4±0
85.1 +
1.8***
)K Significance test (t-Test)
***(p (p 144
V.C-1 : 4% tween 8O,
V.C-2 : 5% ethanol + 25% cremophor + 75% PBS
As shown in Table 2, and FIG. 1, 3 and 5, the
size and the weight of a tumor of NCI-H460 xenografted
BALB/c nude mouse were remarkably decreased when it was
administered with tricyclic derivatives of the present
invention (prepared in Example 8 and Example 12),
comparing to when being administered with a solvent
only (V.C-1, V.C-2) or with a positive control (taxol,
adriamycin) . In particular, in the case of the
compound of the Example 12, the volume and the weight
of a tumor were much decreased with dosage of 10 mg/kg
than with the dosage of 1. mg/kg. And in the case of the
compound of the Example 8, inhibition rate of the tumor
volume showed 85% when it was administered with 10 mg/kg.
Therefore, decreasing rate of the volume and the weight
of a tumor can appreciate in proportion to dosage of
tricyclic derivatives of the present invention.
As shown in Table 3 and in FIG. 2 and 4, the
weight of NCI-H460 xenografted BALB/c nude mouse was
decreased about 10% when it was administered with
tricyclic of the present invention (prepared in Example
8 and Example 12), comparing to when being administered
145
With a solvent only (V.C-1, V.C-2) or with a positive
control (taxol, adriamycin).
Therefore, tricyclic derivatives of the present
invention make the volume and the weight of a tumor
smaller and lighter dose-dependently, and also show
excellent anticancer effect. So, tricyclic derivatives
of the invention can be effectively used as an
anticancer agent and as a anti-proliferation agent.
Experimental Example 3; Inhibitory affect of tricyclic
derivatives of the present invention on capillary-like
tube formation in HUVEG cells (Capillary-like tube
formation assay)
Matrigel used in this experiment was a product of
BioCoat. Matrigel was thawed in. a refrigerator for 24
hours before use. Thawing matrigel, 96 well plate and
yellow tip were put on ice. Then, matrigel was
distributed into each well of the plate by 40 (d. The
polymerization of the plate was performed in a 37C
incubator for 30 minutes. Each well of the plate was
inoculated with 180 ìl of HUVEC cell solution (2xl04
cells/ml) along with 20 ìl of the compound of the
Example 12 in serum-free media (0.3, l, 3, 10 and 30 /;g
146
/ml), followed by further culture for 24 hours.
Tube formation was observed under a microscope to
investigate-inhibition activity of angiogenesis.
Fumagilin and doxorubicin were used as a positive
control.
The results were shown in PIG. 6.
As shown in FIG. 6, tricyclic derivatives of the
present invention (prepared in the Example 12) had
angiogenesis inhibition activity with dosage over 0.3
ìg/ml, which was as good effect a a that of fumagilin, a
positive control, with the dosage of. 10 ìg/ml. Further,
tricyclic derivatives of the invention (prepared in the
Example 12) totally inhibited angiogenesis with the
dosage over 10 ìg/ml.
Thus, tricyclic derivatives of the present
invention can be effectively lined as an angiogenesifj
inhibitor.
Experimental Example 4: Acute toxicity test
5-week-old ICR thrice having the weight of 25-35 g
(SPF, SLC Co. Japan) were: m;ed ur. uei:;t animals. A pair
of female and male of the mice was given for the test
of each compound. Group T2, T3, T4 and T5 were
arranged (10 animals per group) for the test of acute
147
toxicity of the compound prepared in the Example 12.
The compound of the Example 12 was dissolved in a
Solvent [5% DMSO; 20% tween 80; 75% PBS(-)], which was
injected into the abdominal cavity of the mouse (dosage
was shown in Table 4), followed by observation for 7
days. Control group (Tl) was administered with only a
solvent without the compound of (-he Example 12. Arul
the results were shown in Table 1 .
For comparison, colchicine was injected in the
test animals by the same method as described in the
above. The group treated with colchicine was
composed of 6 mice. The test results were shown in
Table 5.
Taxol produced by Bristol Myers Sqibb Co. was
also used for comparison, and the injection and the
test of acute toxicity were performed by the same
method as described in the. above. The results were
shown in Table 6.

Acute toxicity test with a compound prepared in Example
12
Group Animal
Number
rDosage
(mg/kg)
Test Day (Death)
1 2 M4!5 6 7 Total
148
Tl
T2
T3
T4
T5
10
10
10
10
10
Solvent
30
40
50
60
-
-
-
-
-
-
-
-
-
-
-
-
-
_
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
0/10
0/10
0/10
0/10
0/10

Acute toxicity test with colchicine
Group
Tl
T2
T3
T4
T5
Animal
Number
6
6
6
6
5
Dosage
(mg/kg)
Solvent
0.5
1.0
2.0
5.0
Test Day (Death)
1
-
-
-
-
2
-
-
-
3
3
-
1
-
4
-
1
5
-
-
-
1
6
-
-
-
2
-
7
-
-
-
-
-
Total
0/6
0/6
0/6
3/6
5/5

Acute toxicity test with Taxol
Group
Tl
T2
T3
T4
T5
Animal
Number
5
5
5
5
5
Dosage
(mg/kg)
4.0
6.0
9.0
13.0
20.0
Test Day (Death)
1
-
-
-
2
5
2
-
1
-
2
-
3
-
-
-
-
-
4
-
-
-
-
-
5
-
-
2
-
-
6
-
-
-
-
-
7
-
-
-
-
-
Total
0/5
1/5
2/5
4/5
5/5
As shown in Table 4, 5 and 6, toxicity of natural
colchicine to a mouse was confirmed to be LD50 = 2mg/kg,
which was very similar to earlier reported value of
LDSO = 1.6mg/kg [Medicinal Research Reviews, Vol.8,
No.l, 77-94 (1988)]. Toxicity of taxol injection,
»
paclitaxel, was LDSO = 9 - 13 mg/kg. (i.v.
administration) . But toxicity of the compound of the
Example 12 of the present invention was LDso = 60mg/kg,
which was 30 times as week toxicity as that of
colchicine and also weaker than v.hat of taxol injection.
Thus, the compound of the present invention was proved
to have less toxicity to normal cells than colchicine
or Taxol injection.
INDUSTRIAL APPL1 CA'RILITY
Tricyclic derivatives of the present invention
have very strong cytotoxicity to cancer cell lines but
less toxicity to animals themselves than colchicine or
Taxol injection has. Tricylic: derivatives of the
invention further decrease the volume and the weight of:
a tumor and inhibit angiogenesis in HUVEC cells
excellently. Therefore, the derivatives can b
effectively used as an anticancer agent, antiproliferation
agent and an angiogenesis inhibitor as
150
e
well. In addition, tricyclic derivatives of the
present invention can be obtained with ease and be
formulated easily for oral. administration or for
injection owing to its water-solubility.






We claim:
1. A 5,6,7,9-tetrahydro-benzo[a]heptalen derivative of general Formula 1 Rj-O
(Formula Removed)
Wherein,
(l)R,is-T,-B,;
in which Ti is -N(Rs)C(0)-, in that R5 is H or Ci~Cs alkyl group; and Bi is selected from a group consisting of following (a)-(c);
(Formula Removed)
wherein,
(Formula Removed)
R6 is H or halogen;
R7 is hydroxy or -ONO2, with the proviso that when R6 is H, R7 is -ONO2;
T2 is -O-C(O)-;
B2 is said (a) or — (CH2)n3-R7.
n1 is an integer of 0-1;
n2 is an integer of 0-5; and
n3 is an integer of 2~5;
(2) R2 is CH3;
(3) R3 is C1~C4 straight-chain or branched-chain alkyl or C3~C7cycloalkyl, with the proviso that when B1 is (a) and R6 is H, R3 is C2H5;

(4) R4 is OCH3, SCH3 or NR10R11, in which R10 and R11 are each independently H or C1-5 alkyl;
(5) X is O
or a pharmaceutically acceptable salt thereof. 2. The compound as claimed in claim 1, selected from the group consisting of:
1) 6-nitrooxymethyl-N-[(7S)-l,2,3-trimethoxy-10-methylsulfanyl-9-oxo-5,6,7,9-
tetrahydro-benzo[a]heptalen-7-yl]-nicotineamide;
2) N-[(7S)-3-ethoxy-l,2-dimethoxy-10-methyl-sulfanyl-9-oxo-5,6,7,9-
tetrahydro-benzo[a]heptalen-7-yl]-3-nitrooxymethyl-benzamide;
3) 6-nitrooxymethyl-pyridine-2-carboxylic acid-[(7S)-l,2,3-trimethoxy-10-
methylsulfanyl-9-oxo-5,6,7,9-tetrahydro-benzo[a]heptalen-7-yl]-amide;
4) 5-nitrooxymethyl-thiophene-2-carboxylic acid-[(7S)-1,2,3-trimethoxy-10-
methylsulfanyl-9-oxo-5,6,7,9-tetrahydro-benzo[a]heptalen-7-yl]-amide;
5) N-[(7S)-3-ethoxy-l,2-dimethoxy-10-methylsulfanyl-9-oxo-5,6,7,9-tetrahydro-
benzo[a]heptalen-7-yl]-2-fluoro-3-nitrooxymethyl-benzamide;
6) 2-fluoro-N-[(7S)-3-isopropoxy-l,2-dimethoxy-10-methylsulfanyl-9-oxo-
5,6,7,9-tetrahydro-benzo[a]heptalen-7-yl]-3-nitrooxymethyl-benzamide;
7) 2-fluoro-3-nitrooxymethyl-N-[(7S)-l,2,3-trimethoxy-10-methylsulfanyl-9-oxo-5,6,7,9-tetrahydro-benzo[a]heptalen-7-yl]-benzamide;
8) 3-fIuoro-5-nitrooxymethyl-N-[(7S)-l,2,3-trimethoxy-10-methylsulfanyl-9-oxo-5,6,7,9-tetrahydro-benzo[a]heptalen-7-yl]-benzamide;
9) N-[(7S)-3-ethoxy-l,2-dimethoxy-10-methylsulfanyl-9-oxo-5,6,7,9-tetrahydro-
benzo[a]heptalen-7-yl]-3-fluoro-5-nitrooxymethyl-benzamide;
10) 3-fluoro-N-[(7S)-3-isopropoxy-l,2-dimethoxy-10-methylsulfanyl-9-oxo-
5,6,7,9-tetrahydro-benzo[a]heptalen-7-yl]-5-nitrooxymethyl-benzamide;
11) N-[(7S)-3-cyclopentyloxy-l,2-dimethoxy-10-methylsulfanyl-9-oxo-5,6,7,9-
tetrahydro-benzo[a]heptalen-7-yl]-3-fluoro-5-nitrooxymethyl-benzamide;
12) 2-fluoro-5-nitrooxymethyl-N-[(7S)-l,2,3-trimethoxy-10-methylsulfanyl-9-
oxo-5,6,7,9-tetrahydro-benzo[a]heptalen-7-yl]-benzamide;
13) 4-nitrooxymethyl-thiophene-2-carboxylic acid [(7S)-l,2,3-trimethoxy-10-methylsulfanyl-9-oxo-5,6,7,9-tetrahydro-benzo[a]heptalen-7-yl]-amide;
14) 3-nitrooxymethyl-thiophene-2-carboxylic acid [(7S)-l,2,3-trimethoxy-10-methylsulfanyl-9-oxo-5,6,7,9-tetrahydro-benzo[a]heptalen-7-yl]-amide;
15) 3-nitrooxymethyl-benzoic acid-2-[(7S)-l,2,3-trimethoxy-10-methylsulfanyl-
9-oxo-5,6,7,9-tetrahydro-benzo[a]heptalen-7-yl-carbamoyl]-phenylester;
16) 4-nitrooxybutyric acid-2-[(7S)-1,2,3-trimethoxy-10-methylsulfanyl-9-oxo-
5,6,7,9-tetrahydro-benzo[a]heptalen-7-yl-carbamoyl]-phenylester;
17) 3-nitrooxymethyl-benzoic acid-3-[(7S)-l,2,3-trimethoxy-10-rnethylsulfanyl-
9-oxo-5,6,7,9-tetrahydro-benzo[a]heptalen-7-yl-carbamoyl]-phenylester;
18) 4-nitrooxybutyric acid-3-[(7S)-1,2,3-trimethoxy-10-methylsulfanyl-9-oxo-
5,6,7,9-tetrahydro-benzo[a]heptalen-7-yl-carbamoyl]-phenylester;
19) 3-nitrooxymethyl-benzoic acid-3-[(7S)-l,2,3-trimethoxy-10-methylsulfanyl-
9-oxo-5,6,7,9-tetrahydro-benzo[a]heptalen-7-yl-carbamoyl]-benzylester;
20) 4-nitrooxybutyric acid-3-[(7S)-1,2,3-trimethoxy-10-methylsulfanyl-9-oxo-
5,6,7,9-tetrahydro-benzo[a]heptalen-7-yl-carbamoyl]-benzylester;
3. A method for preparing the compound as claimed in claim 1 comprising the following steps:
(Formula Removed)
respectively at room temperature, or; reaction of the resultant compound of formula VII with the compound of formula IV to give the compound of formula V at room temperature (Step 1); and
(2) Nitration or nitrosation of the compound of formula (V) prepared by step 1 or

(Formula Removed)
the compound of formula (VII) to give compound at room
temperature (Step 2).
(Formula Removed)

(Wherein, D is , and R2, R3, R4 and X are same as defined in the
of claim 1;
R5 is H or C1~ C5 alkyl group;
X1 is O;
Hal1 and Hal2 are halogens;
Hal1 and Hal2 of formula (IV) are each same or different halogens, for example F, C1,
Br or I; Y is selected from the group consisting of formula (a') and (b'),
(Formula Removed)
wherein, , R6, n1, and n2 are same as defined in the of claim
1).
4. The compound as claimed in claim 1 as and when used in the preparation of a pharmaceutical composition for treatment or prevention of cancer and/or angiogenesis related disorders.

Best View in Resolution of 1024x768 or later. Enable Javascript for Better Performance.

Documents:

384-DELNP-2006-Abstract-(16-02-2009).pdf

384-delnp-2006-abstract.pdf

384-delnp-2006-assignment.pdf

384-DELNP-2006-Claims-(16-02-2009).pdf

384-delnp-2006-claims-(19-03-2009).pdf

384-delnp-2006-claims-(24-03-2009).pdf

384-delnp-2006-complete specification (granted).pdf

384-DELNP-2006-Correspondence-Others-(16-02-2009).pdf

384-delnp-2006-correspondence-others-(19-03-2009).pdf

384-DELNP-2006-Correspondence-Others-(20-10-2008).pdf

384-delnp-2006-correspondence-others-(24-03-2009).pdf

384-delnp-2006-correspondence-others-1.pdf

384-delnp-2006-correspondence-others.pdf

384-delnp-2006-description (complete)-(19-03-2009).pdf

384-delnp-2006-description (complete).pdf

384-delnp-2006-drawings.pdf

384-DELNP-2006-Form-1-(16-02-2009).pdf

384-delnp-2006-form-1.pdf

384-delnp-2006-form-13-(16-02-2009).pdf

384-delnp-2006-form-13-(20-10-2008).pdf

384-delnp-2006-form-18.pdf

384-DELNP-2006-Form-2-(16-02-2009).pdf

384-delnp-2006-form-2.pdf

384-DELNP-2006-Form-3-(16-02-2009).pdf

384-delnp-2006-form-3.pdf

384-delnp-2006-form-5.pdf

384-delnp-2006-gpa-(19-03-2009).pdf

384-DELNP-2006-Others-Document-(16-02-2009).pdf

384-delnp-2006-pct-101.pdf

384-delnp-2006-pct-210.pdf

384-delnp-2006-pct-237.pdf

384-delnp-2006-pct-301.pdf

384-delnp-2006-pct-304.pdf

384-delnp-2006-pct-308.pdf

384-delnp-2006-pct-311.pdf

384-delnp-2006-pct-401.pdf

384-delnp-2006-pct-402.pdf

384-delnp-2006-pct-409.pdf

384-delnp-2006-pct-416.pdf

384-DELNP-2006-Petition-137-(16-02-2009).pdf

384-DELNP-2006-Petition-138-(16-02-2009).pdf


Patent Number 233339
Indian Patent Application Number 384/DELNP/2006
PG Journal Number 13/2009
Publication Date 27-Mar-2009
Grant Date 28-Mar-2009
Date of Filing 20-Jan-2006
Name of Patentee JE IL PHARMACEUTICAL CO., LTD
Applicant Address 745-4 BANPO-I DONG, SEOCHO-GU, SEOUL 137-810, REPUBLIC OF KOREA.
Inventors:
# Inventor's Name Inventor's Address
1 JOE, BO-YOUNG #101-813 INJUNG-MELODY APT., DUNJEON-RI, POGOK-MYUN, YONGIN CITY, KYUNGGI-DO 449-812, REPUBLIC OF KOREA
2 PARK, SANG-WOO 13/3 205 SUNGWON JUTAEK, 366-I YULJEON-DONG, JANGAN-GU, SUWON CITY, KYUNGGI-DO 440-827, REPUBLIC OF KOREA
3 KIM, KWANG HEE #112-035 HANSUNG APT., 380, GUGAL-RI, KIHUNG-EUB, YONGIN CITY, KYUNGGI-DO 449-735, REPUBLIC OF KOREA
4 OH, BYUNG-KYU B-301 JIN-HEUNG PARK, 74-16 GIMNYANGJANG-DONG, YONGIN CITY, KYUNGGI-DO 449-924, REPUBLIC OF KOREA
5 CHOI, JONG-HEE 16/2 1029-13 GWONSEON-DONG, GWONSEON-GU, SUWON CITY, KYUNGGI-DO 441-822, REPUBLIC OF KOREA
6 KIM, MYUNG-HWA #304-1202 SSANG-YONG APT., 621, SANGHA-RI, GUSUNG-MYUN, YONGIN CITY, KYUNGGI-DO 449-942, REPUBLIC OF KOREA
7 CHUN, KWANGWOO #102-1502 JINWOO APT., 265, SAMGA-DONG, YONGIN CITY, KYUNGGI-DO 449-718, REPUBLIC OF KOREA
8 CHOI, JAE-WON 2/9 29-53 SUSAK-DONG, EUNPYUNG-GU, SEOUL 122-090, REPUBLIC OF KOREA
PCT International Classification Number C07C 317/30
PCT International Application Number PCT/KR2004/001518
PCT International Filing date 2004-06-23
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 10-2003-0041547 2003-06-25 Republic of Korea