You are on page 1of 11

European Journal of Medicinal Chemistry 40 (2005) 991–1001

www.elsevier.com/locate/ejmech

Original article

Design, synthesis and in vitro and in vivo antitumor activities


of novel b-carboline derivatives
R. Cao, H. Chen, W. Peng, Y. Ma, X. Hou, H. Guan, X. Liu, A. Xu *
Department of Biochemistry and Center for Biopharmaceutical Research, College of Life Sciences, Sun Yat-sen (Zhongshan) University,
135 Xin Gang Xi Road, Guangzhou 510275, China

Received 4 December 2004; received in revised form 18 April 2005; accepted 22 April 2005
Available online 13 June 2005

Abstract
To further our SAR study on the chemistry and antitumor activity/neurotoxicity of b-carboline alkaloids, several series of b-carboline
derivatives with various substituents were designed and synthesized from the starting material L-tryptophan on the basis of harmine chemical
structure. Cytotoxic activities of these compounds were investigated in vitro. The results showed that some b-carboline derivatives had
significant cytotoxic activities against human tumor cell lines. Among all the synthesized b-carboline derivatives, the compounds 27, 28 and
32, having a benzyl substituent at both position-2 and 9, respectively, were found to be the most potent compounds with IC50 value lower than
50 µM against all human tumor cell lines examined. Acute toxicities and antitumor activities of the selected b-carboline derivatives in mice
were also evaluated. The results demonstrated that a benzyl substituent at position-2 increased the antitumor activity as well as acute toxicity
significantly. However an (ethoxycarbonyl)amino substituent at position-3 reduced the acute toxicity as well as antitumor activity remarkedly.
These data suggested that (1) the antitumor potencies of b-carboline derivatives were enhanced by the introduction of benzyl substituent into
the position-2; (2) the acute toxicity of b-carboline derivatives reduced dramatically by the introduction of an appropriate substituent into the
position-3 and 9; (3) the b-carboline structure might be an important basis for the design and synthesis of new antitumor drugs with significant
antitumor activity and low toxicity.
© 2005 Elsevier SAS. All rights reserved.
Keywords: b-Carboline; Synthesis; Cytotoxic; Acute toxicity; Antitumor; SAR

1. Introduction activities against human tumor cell lines. Our previous inves-
tigation [22] also showed that harmine and its derivatives had
The malignant tumor has long been one of the serious dis- significant antitumor activities in mice bearing both Lewis
eases threatening human health. To discover and develop novel lung cancer and Sarcoma 180, while they exhibited remark-
therapeutic agents for the treatment of malignancy has a vital able neurotoxic effects including tremor, twitch and jumping
importance. One of the successful and effective approaches in experimental animal models. Furthermore we previously
in the search for new antitumor agents from natural products reported [22] that the introduction of appropriate substitu-
is to synthesize novel compounds by simple chemical modi- ents into the position-3 and 9 in b-carboline nucleus could
fication on the basis of natural leading compounds. contribute to their enhancing antitumor activities and decreas-
The b-carboline alkaloids, containing planar tricyclic sys- ing acute toxicity besides their loss of neurotoxic effect com-
tem, are a large group of naturally-occurring and synthetic pletely.
alkaloids [1–9]. Recently these compounds attracted a con- Inspired by these results, in the present investigation, we
siderable attention due to their biological and pharmaceuti- designed and synthesized numerous b-carboline derivatives
cal properties [9–17]. Harmine is the most representative with various substituents at different position in b-carboline
b-carboline alkaloid, which was originally isolated from seeds nucleus on the basis of harmine chemical structure, it is pos-
of Pegannum harmala L. [4]. Recent work [18–21] demon- sible to discover and obtain new compounds with compa-
strated that harmine and its derivatives were highly cytotoxic rable activity and lower toxicity than Harmine. Moreover this
study will further elucidate the structure–activity relation-
* Corresponding author. Tel.: +86 20 8411 3655; fax: +86 20 8403 8377. ships (SAR) of these compounds with regard to antitumor
E-mail address: ls36@zsu.edu.cn (A. Xu). activity and neurotoxicity/acute toxicity. We report here the
0223-5234/$ - see front matter © 2005 Elsevier SAS. All rights reserved.
doi:10.1016/j.ejmech.2005.04.008
992 R. Cao et al. / European Journal of Medicinal Chemistry 40 (2005) 991–1001

preparation of novel b-carboline derivatives, their antitumor in anhydrous methanol or ethanol to give the methyl and ethyl
activities and toxicological effects. carbamates 8 and 9 [9], respectively (see Scheme 1).
Following the same procedures as given above, the ethyl
9-ethyl-b-carboline-3-carboxylate 10 and ethyl 9-benzyl-b-
2. Chemistry carboline-3-carboxylate 11, prepared by N9-ethylation and
N9-benzylation through reacting with ethyl or benzyl bro-
The preparation of the 1,2,3,4-tetrahydro-b-carboline mide in the presence of sodium hydride in DMF and THF as
derivatives employed in this investigation was accomplished previously reported [22], were reacted with 85% hydrazine
via the well-known Pictet–Spengler reaction [6,23,24] utiliz- hydrate in ethanol gave the hydrazide 12 and 13, respec-
ing the appropriate aldehyde in either alkaline [25,26] or acid tively, which after treatment with sodium nitrite, yielded the
media [23–27]. In general, conversion of the 1,2,3,4- corresponding acyl azide 14 and 15. When compounds 14
tetrahydro-b-carboline derivatives to the corresponding and 15 were refluxed in anhydrous methanol or ethanol to
b-carboline derivatives was carried out either by oxidation of produce the methyl and ethyl carbamates 16–18, respec-
the tetrahydro derivatives with sulfur in refluxing xylene [28] tively (see Scheme 2).
or with chloranil in refluxing s-tetrachloroethane [24] or with Previous studies [22] with Harmine (7-methoxy-1-methyl-
lead tetraacetate in glacial acetic acid [25]. b-carboline) and Norharman (b-carboline) derivatives have
The 1,2,3,4-tetrahydro-b-carboline-3-carboxylic acid 2 was shown that substitution of alkyl or benzyl groups at the
prepared by the condensation of L-tryptophan with formalde- N9-position of b-carboline ring system frequently promotes
hyde according to the method of Lippke et al. [25] and was the cytotoxic activity of these compounds. For this reason, a
subsequently converted to the ethyl 1,2,3,4-tetrahydro-b- series of 9-substituted 1-methyl-b-carboline (Harman) deriva-
carboline-3-carboxylate 3 by heating ethanolic hydrogen chlo- tives were designed and synthesized with substituents rang-
ride solution. The compound 3 was then subjected to the oxi- ing in size from methyl to phenylpropyl group. The 1-methyl-
dation with sulfur in refluxing xylene to provide ethyl b-carboline 20, prepared by the condensation of L-tryptophan
b-carboline-3-carboxylate 4 (Scheme 1). This sequence of with acetaldehyde in acid solution and followed by aromati-
reactions can be easily scaled up to give enough material 4 zation, oxidation and decarboxylation in a single step through
for further transformations. the action of potassium dichromate according to the method
In order to examine the effect of amino substitutents at posi- previously described by Synder et al. [29], were alkylated or
tion-3 on the antitumor activities of the compounds, 3-amino- benzylated by the action of sodium hydride in anhydrous DMF
b-carboline 7 was prepared by a Curtius-type reaction start- followed by the addition of the relevant appropriate alkylat-
ing with compound 4 [7,9,25]. Refluxing of compound 4 with ing and benzylating agents to afford compounds 21–26. Acci-
85% hydrazine hydrate in ethanol gave the hydrazide 5 [9], dentally, we found that both the N9 and N2-position of the
and then was treated with sodium nitrite, yielding the corre- compound 20 were benzylated simultaneously at 50–60 °C
sponding acyl azide 6 [9]. Compound 6 was further refluxed
in acetic acid, and rearranged to afford the amine 7 [9] in
good yield. Alternatively, the compound 6 could be refluxed

Scheme 1. Scheme 2.
R. Cao et al. / European Journal of Medicinal Chemistry 40 (2005) 991–1001 993

for 2 h led to novel compound 27 in good yield (see Scheme


3). This reaction condition can be employed to prepare other
2,9-bisubstitued b-carboline derivatives 28 and 30–32 (see
Scheme 4).
The b-carboline-3-carboxylic acid 33 [25], prepared by
hydrolyzation the compound 4, was reacted with benzyl bro-
mide in anhydrous DMF solution in the presence of sodium
hydride to yield compound 34 in a good yield. Compound 4
was reacted with ethyl diamine according to the method
described by Xiao et al. [30] to provided the 3-carboxamide
derivative 35 (see Scheme 5). The conversion of ethyl
b-carboline-3-carboxylate 4 and ethyl 9-benzyl-b-carboline-
3-carboxylatate 11 to the 3-hydroxymethyl derivatives 36 and
38, respectively, can be accomplished easily by the treatment
of 4 and 11 with lithium borohydride according to the method
reported by Cain et al. [7], which after refluxing in acetic
acid solution, yielded the corresponding derivatives 37 and
Scheme 3. 39 (see Schemes 5 and 6).
The 1-methyl-1,2,3,4-tetrahydro-b-carboline-3-carboxylic
acid 19 was prepared by condensation of L-tryptophan with
acetaldehyde according to the method of Synder et al. [24].
Esterification of 19 with ethanol in the presence of SOCl2
yielded the corresponding ester 40, followed by dehydroge-
nation with sulfur in xylene to give ethyl 1-methyl-b-
carboline-3-carboxylate 41. The N9-position of 41 was fur-
ther n-butylated by the action of sodium hydride in anhydrous
DMF gave derivatives 42, followed by hydrolyzation in alka-
line solution to provide the corresponding 3-carboxylic acid
derivative 43 (see Scheme 7). The chemical structures of all
the synthesized novel compounds were confirmed by FAB-
MS, UV, IR, 1H-NMR and elemental analyses data.

3. Results and discussion

3.1. Cytotoxicity in vitro

The cytotoxic potential of all synthesized b-carboline


derivatives was evaluated in vitro against a panel of human
Scheme 4. tumor cell lines. Compound 43 was converted into its water-
soluble sodium salts, and the other compounds (except 27,

Scheme 5.
994 R. Cao et al. / European Journal of Medicinal Chemistry 40 (2005) 991–1001

Scheme 6.

Scheme 7.

28 and 30–32) examined were all prepared in the form of position-2 and 9 simultaneously led to the compounds 30,
hydrochloride by the usual methods before use. The results which failed to displayed significant cytotoxic activities
were summarized in Table 1. against PLA-801 and Lovo cell lines. Furthermore, the com-
As shown in Table 1, the compounds 27, 28 and 32 dem- pound 31 having an ethyl group at position-2 and 9 simulta-
onstrated the highest cytotoxic activity with all IC50 values neously was inactive as cytotoxic agent. It is noteworthy that
lower than 50 µM against human tumor cell lines tested. The incorporation of benzyl substituent at position-2 and 9 in
compounds 8, 20–23, 35 and 38 displayed moderate cyto- b-carboline simultaneously afforded the compound 32, which
toxic activities, and other compounds only had marginal or had prominent cytotoxic activities against all the screened
no cytotoxic effect in any cell lines. tumor cell lines. In order to further corroborate the influence
Among all the compounds screened, 3-amino-b-carboline of benzyl group at position-2 on cytotoxic activity of
7 and its derivatives 9 and 16–18 (except 8) only showed mar- b-carboline derivatives, we designed and synthesized the com-
ginal or no cytotoxic activity, suggesting that the amino sub- pound 28. As we predicted, the compound 28 exhibited sig-
stituent at position-3 was unfavorable for their cytotoxic nificant cytotoxic activities against all six human tumor cell
activities. lines with all IC50 values lower than 40 µM. These results,
Of all 1-methyl-b-carboline derivatives, the compound 20 together with the cytotoxic activity of the compound 27, fur-
having no substituent at position-9 in b-carboline showed ther confirmed that the benzyl substituent at position-2 might
moderate cytotoxic activity. Introducing a short chain alkyl play a vital role in determining significant cytotoxic activi-
substituent into position-9 in b-carboline led to compound ties of these compounds.
21 (methyl) and 22 (ethyl) and 23 (butyl), which all exhibited The cytotoxic activity decreased greatly after converting
more potent cytotoxic activities than the parent compound the compounds 4 and 11 into its 3-hydroxymethyl derivatives
20. The compound 24 with a benzyl group at position- 36–37 and 38–39, respectively, whereas the cytotoxic activi-
9 displayed selective cytotoxic activities against HepG2 and ties increased significantly after converting the compound 4
Hela cell lines. Whereas introducing a pentafluorobenzyl or into its 3-carboxamide derivative 35. The benzyl at position-
phenylpropyl into position-9 led to the compounds 25 and 9 in b-carboline, the compound in which the ethoxycarbonyl
26, respectively, which all demonstrated decreased cytotoxic at position-3 was converted into the corresponding benzy-
activities remarkedly. Interestingly, further introducing a ben- loxycarbonyl (34), led to complete loss of cytotoxic activity.
zyl into position-2 in b-carboline afforded the compound 27, These results indicated that the introduction of an appropri-
which displayed the prominent and broader spectrum of cyto- ate carboxamide substituent into 3-position of b-carboline ring
toxic activities against all six human tumor cell lines tested system facilitates the increase of their cytotoxic activities,
with all IC50 values lower than 50 µM. while 3-hydroxymethyl group was unfavorable.
The compound 29 [22] having no substituent in b-carboline An overview of the cytotoxic activities data of all new syn-
nucleus showed significant cytotoxic activity against PLA- thesized b-carboline derivatives and of the earlier report [22]
801 and Lovo cell lines. However, introducing a methyl into clearly indicate two results: (i) the position and nature of sub-
R. Cao et al. / European Journal of Medicinal Chemistry 40 (2005) 991–1001 995

Table 1
Cytotoxicity of b-carboline derivatives in vitro c (µM a)
Compound PLA-801b HepG2 b Bel-7402 b BGC-823 b Hela b Lovo b
Harmined 45 46 54 68 60 66
4d 295 241 293 278 100 160
5 741 365 >1000 741 395 310
7 572 337 >1000 856 312 242
8 199 233 134 211 170 134
9 >1000 >1000 >1000 >1000 >1000 >1000
11 d 78 38 45 38 13 122
12 512 262 371 113 238 191
13 516 139 60 267 247 250
16 >1000 >1000 >1000 >1000 >1000 >1000
17 >1000 >1000 >1000 >1000 >1000 >1000
18 >1000 >1000 >1000 >1000 >1000 >1000
20 317 115 151 215 186 275
21 163 100 96 160 159 128
22 271 148 203 282 165 213
23 150 122 145 364 102 152
24 324 84 126 >1000 78 324
25 >1000 >1000 370 251 477 >1000
26 297 948 449 987 >1000 297
27 11 7 43 15 13 40
28 14 20 36 40 34 31
29 d 17 228 379 274 353 35
30 275 197 371 263 72 >1000
31 >1000 425 >1000 >1000 857 >1000
32 32 35 45 39 32 20
34 >1000 >1000 >1000 >1000 >1000 >1000
35 138 80 164 84 86 102
36 >1000 579 739 783 617 485
37 775 >1000 588 638 779 248
38 161 89 112 111 105 101
39 254 298 128 412 497 123
41 262 86 227 111 97 83
42 199 104 229 288 224 89
43 316 264 449 487 >1000 634
a
Cytotoxicity as IC50 for each cell line, is the concentration of compound that causes a 50% growth inhibition to untreated cells using the MTT assay.
b
Cell lines include non-small cell lung carcinoma (PLA-801), liver carcinoma (HepG2 and Bel-7402), gastric carcinoma (BGC-823), cervical carcinoma
(Hela), colon carcinoma (Lovo).
c
Data represent the mean values of three independent determinations.
d
See Ref. [22].

stituents on the b-carboline nucleus seem to modulate cyto-


toxic activity and (ii) the nature of substituents specially at
position-2, 3 and 9 significantly contribute to the cytotoxic Table 2
efficacy. Acute toxic effects of b-carboline derivatives in mice
Compound Acute toxicity
3.2. Assessment of acute toxicity LD50 (mg kg–1, 95% CL) Neurotoxic effect
Harmine b 59.00 (43.50–110.00) +a
The LD50 values and scores for neurotoxicity of the selected 18 >500 _
b-carboline derivatives in mice after administration by i.p. route 20 32.63 (28.79–36.99) _
were summarized in Table 2. All the tested compounds resulted 24 68.63 (60.96–77.27) _
in acute toxic manifestation but caused no obvious neuro- 25 348.44 (303.43–400.12) _
toxic reaction including tremor, twitch, jumping, tetanus and 27 5.82 (4.97–6.82) _
28 65.73 (58.25–74.11) _
supination just like Harmine. Animals were drowsy and exhib-
38 247.13 (213.51–286.03) _
ited a decrease in locomotor activity after the administration
43 65.30 (57.65–73.96) _
of the compounds. Of all the tested compounds, the com- a
Acute neurotoxic manifestation were denoted by “+” and “–”. A “+”
pound 27 displayed the highest acute toxicity with LD50 value represents toxic reactions including tremble, twitch, jumping, tetanus and
of 5.82 mg kg–1. The compounds 20, 24, 28 and 43 also exhib- supination, while “–” means no such reaction.
ited remarkable acute toxicity with LD50 value of 32.63, b
See Ref. [22].
996 R. Cao et al. / European Journal of Medicinal Chemistry 40 (2005) 991–1001

68.63, 65.73 and 65.30 mg kg–1, respectively, while for the substituent into position-2 of b-carboline nucleus improved
compounds 25 and 38, acute toxicities were much less with their antitumor activities significantly, while incorporation of
LD50 value of 348.44 and 247.13 mg kg–1, respectively. Espe- hydroxymethyl or ethoxycarbonylamino group at position-
cially, the compound 18 caused no death at tested dosage, 3 in b-carboline nucleus impaired their antitumor activities.
except for partial contortion at injection point at the highest
dosage used. Autopsy of the animals that died in the course
of experiment and the necropsy findings in surviving animals 4. Conclusion
at the end of experimental period (14 days) revealed no appar-
ent changes in any organs. Our current investigation corroborated the previous obser-
A total analysis to the acute toxicity and neurotoxic effect vations [22] that the antitumor activity and acute toxicity as
of b-carboline derivatives examined above and of the previ- well as neurotoxic effect of b-carboline derivatives are
ous investigation [22] clearly suggested that (1) the acute tox- substituent-dependant. An overview of the present study and
icity and neurotoxic effects of b-carboline derivatives de- of the previous report [22], we arrived at the following con-
pended upon the presence, location and nature of the clusions: (1) the methoxyl group at position-7 in b-carboline
substituent introduced into the b-carboline nucleus; (2) the nucleus plays a very crucial role in determining their remark-
acute toxicity increased greatly by introducing a benzyl into able neurotoxic effects including tremor, twitch, jumping, teta-
position-2 while it decreased dramatically by introducing nus and supination; (2) introducing appropriate substituents
ethoxycarbonylamino into position-3; (3) the methoxy group into position-9 and 2 of b-carboline derivatives increases
at position-7 in b-carboline nucleus might be play a very cru- greatly their antitumor activities; (3) the nature of substitu-
cial role in determining their remarkable neurotoxic effects. ents specially at position-3 and 1 significantly contributes to
the decreased acute toxicity of b-carboline derivatives; (4)
3.3. Evaluation of antitumor activity b-carboline framework is an important pharmacophore for
the design and synthesis of novel antitumor agents with sig-
Eight b-carboline derivatives were selected for evaluation nificant pharmacological activity and low toxicity.
in vivo against mice bearing Lewis lung cancer and Sar- In this paper we described a preliminary SAR between
coma180. The tumor inhibition rates of these compounds were b-carboline derivatives and antitumor activities, while the anti-
summarized in Table 3. All the tested compounds exhibited tumor activities of the studied b-carboline derivatives pre-
potent antitumor activities. Compounds 24, 27, 28 and 43 sented here remained relatively moderate. It is necessary to
demonstrated remarkable antitumor activities with the tumor further design and synthesize more potent antitumor agents
inhibition rate of over 40% against mice bearing Lewis lung together with low neurotoxicities by simple chemical struc-
cancer at dose 22.0, 2.0, 20.0 and 22.0 mg kg–1, respectively. tural modification on the basis of the present as well as the
Furthermore, compound 24 showed the highest antitumor previous investigation. Further investigation to elucidate the
effect with the tumor inhibition rate of 47.3% against mice pharmacological mechanisms leading to the improved anti-
bearing Lewis lung cancer, and compound 28 exhibited almost tumor effects are underway in our laboratory. We hope that
equal antitumor activity against mice both bearing Lewis lung these easy-to-synthesize b-carboline derivatives should serve
cancer and Sarcoma 180. The other compounds tested showed as the basic system for searching for more efficient antitumor
moderate antitumor activity with tumor inhibition rates rang- drugs.
ing from 28.1% to 38.6% at dose ranging from 10.0 to
100.0 mg kg–1. It is interesting to note that the Lewis lung
cancer was more susceptible to all tested compounds than the 5. Experimental
Sarcoma 180. The results indicated that introducing a benzyl
Table 3 5.1. Materials
Antitumor effects of b-carboline derivatives against mice bearing Sarcoma
180 and Lewis lung carcinoma Melting points were determined in capillary tubes on an
Compound Dose Tumor inhibition rate (%) electrothermal PIF YRT-3 apparatus and without correction.
(mg kg–1) Lewis lung carcinoma Sarcoma180 UV spectra were measured on Shimadzu UV 2501PC Spec-
Harminea 7.5 34.1 15.3 trometer. FAB-MS spectra were obtained from VG ZAB-HS
18 100.0 30.9 29.1 spectrometer. FT-IR spectra were run on a Bruker Equinox
20 10.0 36.3 35.2
55 Fourier transformation infrared spectrometer. 1H-NMR
24 22.0 47.3 35.5
25 100.0 38.6 31.5
spectra were recorded on a Varian INOVA 500NB spectrom-
27 2.0 43.5 39.0 eter. Elemental analyses (C, H, N) were carried out on an
28 20.0 41.6 41.1 Elementar Vario EL CHNS Elemental Analyzer. Silica gel
38 100.0 34.5 28.1 F254 was used in analytical thin-layer chromatography (TLC)
43 22.0 41.3 36.0 and silica gel was used in column chromatography.
CTX 30.0 86.4 87.3 All chemicals were purchased from commercial suppliers
a
See Ref. [22]. and were dried and purified when necessary. 1,2,3,4-tetra-
R. Cao et al. / European Journal of Medicinal Chemistry 40 (2005) 991–1001 997

hydro-b-carboline-3-carboxylic acid (2) [25], ethyl 1,2,3,4- H-8), 7.56–7.59 (1H, m, H-5), 7.40–7.42 (1H, d, J = 8.0 Hz,
tetrahydro-b-carboline-3-carboxylate (3) [25], ethyl b-carbo- H-6), 7.23–7.26 (1H, m, H-7), 4.37–4.41 (2H, m, NCH2CH3),
line-3-carboxylate (4) [25], b-carboline-3-carbohydrazide (5) 3.87 (3H, s, OCH3), 1.45–1.48 (3H, m, NCH2CH3). Anal.
[9], 3-azidocarbonyl-b-carboline (6) [9], 3-amino-b-carboline Calc. for C15H15N3O2: C, 66.91; H, 5.58; N, 15.61. Found:
(7) [9], 3-[(methoxycarbonyl)amino]-b-carboline (8) [9], C, 66.78; H, 5.79; N, 15.52.
3-[(ethoxycarbonyl)amino] b-carboline (9) [9], ethyl 9-ehtyl-
b-carboline-3-carboxylate (10) [22], ethyl 9-benzyl-b- 5.1.4. 3-[(Ethoxycarbonyl)amino]-9-ethyl-b-carboline (17)
carboline-3-carboxylate(11) [22], 1-methyl-1,2,3,4-tetra- The same conditions as for the preparation of 16 were used
hydro-b-carboline-3-carboxylic acid (19) [29], 1-methyl-b- for the preparation of 17 except that ethanol was the reaction
carboline (20) [29], b-carboline (29) [8], b-carboline-3- solvent. White crystals of 17 were obtained (0.8 g, 56%); m.p.
carboxylic acid (33) [25], b-carboline-3-carboxamide (35) 217–218 °C; FAB-MS m/z (M + 1) 284; UV kmax 376, 364,
[30], 3-hydroxymethyl-b-carboline (36) [7] and ethyl 296, 240, 202 nm IR (KBr) 3420, 3202, 2974, 1721, 1627,
1-methyl-b-carboline-3-carboxylate 41 [32] are known com- 1585, 1532, 1471, 1281, 1219 cm–1; 1H-NMR (500 MHz,
pounds. CDCl3) d 8.68 (1H, s, H-4), 8.59 (1H, s, H-1), 8.15–8.17 (1H,
d, J = 8.0 Hz, H-8), 7.55–7.59 (1H, m, H-5), 7.39–7.40 (1H,
5.1.1. 3-Carbohydrazide-9-ethyl-b-carboline (12) d, J = 8.5 Hz, H-6), 7.23–7.26 (1H, m, H-7), 4.32–4.38 (4H,
A solution of compound 10 (2.68 g, 10 mmol) in ethanol m, NCH2CH3, OCH2CH3), 1.41–1.47 (6H, m, NCH2CH3,
(100 ml) containing 85% hydrazine hydrate (15 ml) was OCH2CH3). Anal. Calc. for C16H17N3O2: C, 67.84; H, 6.00;
refluxed for 8 h. The resulting mixture was cooled and the N, 14.84. Found: C, 67.73; H, 6.16; N, 14.75.
precipitate was collected by filtration, washed with ethanol,
and then air-dried. The solid was recrystallized from ethanol, 5.1.5. 3-Carbohydrazide-9-benzyl-b-carboline (13)
giving white crystals 12 (2.0 g, 83%), m.p. 195–196 °C; A solution of compound 11 (3.3 g, 10 mmol) in ethanol
FAB-MS m/z (M + 1) 255; UV kmax 359, 343, 303, 272, 238, (100 ml) containing 85% hydrazine hydrate (20 ml) was
221 nm IR (KBr) 3298, 3202, 2955, 1627, 1590, 1531, 1458, refluxed for 10 h. Then the resulting mixture was treated in a
1331, 1261, 1128 cm–1; 1H-NMR (500 MHz, DMSO-d6) d manner similar to that described for 12 to afford white crys-
9.61 (1H, s, NH), 9.03 (1H, s, H-4), 8.82 (1H, s, H-1), 8.40– tals 13 (2.8 g, 87%), m.p. 209–211 °C; FAB-MS m/z (M + 1)
8.42 (1H, d, J = 7.5 Hz, H-8), 7.75–7.76 (1H, d, J = 8.5 Hz, 317; UV kmax 356, 342, 272, 238,207 nm IR (KBr) 3349,
H-5), 7.64–7.67 (1H, m, H-6), 7.32–7.35 (1H, m, H-7), 4.58– 3300, 3201, 3059, 1619, 1556, 1496, 1461, 1336, 1200 cm–1;
1
4.62 (2H, m, NCH2CH3), 4.54 (2H, s, NH2), 1.37–1.40 (3H, H-NMR (500 MHz, DMSO-d6) d 9.62 (1H, s, NH), 9.04
m, NCH2CH3). Anal. Calc. for C14H14N4O: C, 66.14; H, 5.51; (1H, s, H-4), 8.85 (1H, s, H-1), 8.43–8.45 (1H, d, J = 8.0 Hz,
N, 22.05. Found: C, 65.92; H, 5.76; N, 22.14. H-8), 7.77–7.79 (1H, d, J = 8.5 Hz, H-5), 7.61–7.65 (1H, m,
H-6), 7.33–7.36 (1H, m, H-7), 7.21–7.30 (5H, m, ArH), 5.83
5.1.2. 3-Azidocarbonyl-9-ethyl-b-carboline (14) (2H, s, NCH2Ar), 4.54 (2H, s, NH2), 1.37–1.40 (3H, m,
A suspension of compound 12 (2.54 g, 10 mmol) in water NCH2CH3). Anal. Calc. for C19H16N4O: C, 72.15; H, 5.06;
(200 ml) was dissolved by the addition of concentrated hydro- N, 17.72. Found: C, 72.06; H, 5.21; N, 17.77.
chloric acid (20 ml). The yellow solution was cooled to 0 °C
before the addition of a solution of sodium nitrite (2.1 g, 5.1.6. 3-Azidocarbonyl-9-benzyl-b-carboline (15)
30 mmol) in water (30 ml). After 40 min at 0 °C, the mixture A suspension of compound 13 (3.16 g, 10 mmol) in water
was made basic with saturated aqueous sodium hydrogen car- (500 ml) was dissolved by the addition of concentrated hydro-
bonate, and then the precipitate was collected by filtration, chloric acid (20 ml). The yellow solution was cooled to
washed well with water, and dried in a desiccator under 0 before the addition of a solution of sodium nitrite (2.1 g,
vacuum, yielding yellowish solid 14 (2.3 g, 87%) with a ten- 30 mmol) in water (30 ml). After 40 min at 0 °C, the resulting
dency to decompose. The material was used without further mixture was treated in a manner similar to that described for
purification for the following steps. 14, yielding yellowish solid 15 (2.9 g, 89%) with a tendency
to decompose. The material was used without further purifi-
5.1.3. 3-[(Methoxycarbonyl)amino]-9-ethyl-b-carboline (16) cation for the following steps.
A mixture of the azide 14 (1.32 g, 10 mmol) in methanol
(100 ml) was refluxed for 10 h. The reaction mixture was 5.1.7. 3-[(Ethoxycarbonyl)amino]-9-benzyl-b-carboline (18)
concentrated in vacuum, whereupon the product 16 precipi- A mixture of the azide 15 (2.9 g, 8.86 mmol) in ethanol
tated. Then the precipitate was collected by filtration, washed (150 ml) was refluxed for 10 h. The reaction mixture was
with cooled ethanol. The solid was recrystallized from etha- concentrated in vacuum, whereupon the product 18 precipi-
nol, giving white crystals 16 (0.8 g, 59%), m.p. 213–214 °C; tated. Then the precipitate was collected by filtration, washed
FAB-MS m/z (M + 1) 270; UV kmax 376, 365, 296, 251, 241, with cooled ethanol. The solid was recrystallized from ethyl
203 nm IR (KBr) 3435, 3207, 2984, 1727, 1630, 1595, 1472, acetate, giving white crystals 18 (1.8 g, 57%), m.p. 218–
1282, 1228, 1079 cm–1; 1H-NMR (500 MHz, CDCl3) d 8.67 219 °C; FAB-MS m/z (M + 1) 346; UV kmax 373, 362, 295,
(1H, s, H-4), 8.55 (1H, s, H-1), 8.16–8.17 (1H, d, J = 8.0 Hz, 251 nm IR (KBr) 3248, 3200, 2981, 1722, 1629, 1534, 1467,
998 R. Cao et al. / European Journal of Medicinal Chemistry 40 (2005) 991–1001

1281, 1217 cm–1; 1H-NMR (500 MHz, CDCl3) d 8.88 (1H, s, 1


H-NMR (500 MHz, CDCl3) d 8.31–8.32 (1H, d, J = 5.5 Hz,
H-4), 8.78 (1H, s, H-1), 8.21–8.22 (1H, d, J = 7.5 Hz, H-8), H-4), 8.11–8.13 (1H, d, J = 7.5 Hz, H-8), 7.84–7.85 (1H, d,
8.06–8.08 (2H, d, J = 8.0 Hz, H-5, H-6), 7.67–7.70 (1H, m, J = 5.5 Hz, H-3), 7.56–7.59 (1H, m, H-5), 7.45–7.47 (1H, d,
H-7), 7.54–7.60 (3H, m, ArH), 7.35–7.38 (1H, m, ArH), 7.31– J = 8.5 Hz, H-6), 7.25–7.28 (1H, m, H-7), 4.53–4.56 (2H, m,
7.32 (1H, d, J = 8.0 Hz, ArH), 5.79 (2H, s, NCH2Ar), 4.51– NCH2CH2CH2CH3), 3.06 (3H, s, CH3), 1.80–1.86 (2H, m,
4.55 (2H, m, OCH2CH3), 1.47–1.50 (3H, m, OCH2CH3). NCH2CH2CH2CH3), 1.42–1.49 (2H, m, NCH2CH2CH2CH3),
Anal. Calc. for C21H19N3O2: C, 73.04; H, 5.51; N, 12.17. 0.97–1.00 (3H, m, NCH2CH2CH2CH3). Anal. Calc. for
Found: C, 72.94; H, 5.67; N, 12.09. C16H18N2: C, 80.67; H, 7.56; N, 11.77. Found: C, 80.58; H,
7.77; N, 11.63.
5.1.8. 1,9-Dimethyl-b-carboline (21)
A mixture of 20 (1.82 g, 10 mmol) and anhydrous DMF 5.1.11. 9-Benzyl-1-methyl-b-carboline (24)
(50 ml) was stirred at RT until clear, and then 60% NaH (0.6 g, A mixture of 20 (1.82 g, 10 mmol) and anhydrous DMF
15 mmol) and iodomethane (2 ml, 30 mmol) were added. (60 ml) was stirred at RT until clear, and then 60% NaH (0.6 g,
The mixture was stirred at RT for 1 h. The resulting mixture 15 mmol) and benzyl bromide (5 ml, 40 mmol) were added
was poured into H2O (100 ml), and extracted with ethyl and heated at reflux for 3 h, and then treated in a manner
acetate. The organic phase was washed with water and brine, similar to that described for 21 to afford white crystals 24
then dried over anhydrous sodium sulfate, filtered and evapo- (2.3 g, 85%), m.p. 111–112 °C (from ether); FAB-MS m/z
rated. The oil obtained was purified by silica column chro- (M + 1) 273; UV kmax 355, 341, 287, 242, 237 nm IR (KBr)
matography with ethyl acetate as the eluent. Upon recrystal- 3029, 1615, 1561, 1446, 1356, 1238, 1131 cm–1; 1H-NMR
lization, white crystals of 21 were obtained (1.6 g, 82%), m.p. (500 MHz, CDCl3) d 8.33–8.34 (1H, d, J = 5.5 Hz, H-4),
103–104 °C (from ether); FAB-MS m/z (M + 1) 197; UV 8.12–8.14 (1H, d, J = 8.0 Hz, H-8), 7.84–7.86 (1H, d,
kmax 358, 343, 288, 243, 236 nm IR (KBr) 3050, 2920, 1614, J = 5.5 Hz, H-3), 7.48–7.51 (1H, m, H-5), 7.32–7.33 (1H, d,
1560, 1446, 1288, 1231, 1134 cm–1; 1H-NMR (500 MHz, J = 8.0 Hz, H-6), 7.21–7.28 (4H, m, H-7, ArH), 6.96–6.97
CDCl3) d 8.28–8.29 (1H, d, J = 5.0 Hz, H-4), 8.08–8.09 (1H, (2H, m, ArH), 5.74 (2H, s, CH2Ar), 2.85 (3H, s, CH3). Anal.
d, J = 8.0 Hz, H-8), 7.79–7.80 (1H, d, J = 5.5 Hz, H-3), 7.55– Calc. for C19H16N2: C, 83.82; H, 5.88; N, 10.29. Found: C,
7.59 (1H, m, H-5), 7.41–7.42 (1H, d, J = 8.5 Hz, H-6), 7.24– 83.67; H, 6.07; N, 10.22.
7.27 (1H, m, H-7), 4.11 (3H, s, NCH3), 3.07 (3H, s, CH3).
Anal. Calc. for C13H12N2: C, 79.59; H, 6.12; N, 14.29. Found: 5.1.12. 9-(2′,3′,4′,5′,6′-Pentafluoro)benzyl-1-methyl-b-car-
C, 79.39; H, 6.35; N, 14.20. boline (25)
A mixture of 20 (1.82 g, 10 mmol) and anhydrous DMF
5.1.9. 9-Ethyl-1-methyl-b-carboline (22) (60 ml) was stirred at RT until clear, and 60% NaH (0.6 g,
A mixture of 20 (1.82 g, 10 mmol) and anhydrous DMF 15 mmol) and aa-bromo-2,3,4,5,6-pentafluorotoluene (3 ml,
(60 ml) was stirred at RT until clear, and then 60% NaH (0.6 g, 20 mmol) were added, and then reacted and treated in a man-
15 mmol) and iodoethane (2.5 ml, 30 mmol) were added. ner similar to that described for 21 to afford white crystals 25
The mixture was stirred at RT for 2 h, and then treated in a (2.8 g, 77%), m.p. 195–196 °C (from ether); FAB-MS m/z
manner similar to that described for 21 to afford yellow oil (M + 1) 363; UV kmax 349, 335, 286, 235 nm IR (KBr) 3056,
22 (1.7 g, 81%); FAB-MS m/z (M + 1) 211; UV kmax 358, 1618, 1564, 1505, 1445, 1347, 1207, 1120 cm–1; 1H-NMR
344, 289, 243, 237 nm IR (KBr) 3046, 2980, 2512, 1623, (500 MHz, CDCl3) d 8.36–8.38 (1H, d, J = 5.5 Hz, H-4),
1537, 1462, 1334, 1231, 1139 cm–1; 1H-NMR (500 MHz, 8.10–8.12 (1H, d, J = 8.0 Hz, H-8), 7.84–7.85 (1H, d,
CDCl3) d 8.21–8.22 (1H, d, J = 5.5 Hz, H-4), 8.01–8.02 (1H, J = 5.0 Hz, H-3), 7.51–7.55 (1H, m, H-5), 7.28–7.32 (2H, m,
d, J = 8.0 Hz, H-8), 7.69–7.70 (1H, d, J = 5.0 Hz, H-3), 7.47– H-6, H-7), 5.93 (2H, s, CH2Ar), 3.07 (3H, s, CH3). Anal.
7.49 (1H, m, H-5), 7.34–7.36 (1H, d, J = 8.5 Hz, H-6), 7.15– Calc. for C19F5H11N2: C, 62.98; H, 3.04; N, 7.73. Found: C,
7.17 (1H, m, H-7), 4.24–4.27 (2H, m, NCH2CH3), 3.03 (3H, 62.75; H, 3.25; N, 7.67.
s, CH3), 1.37–1.40 (3H, m, NCH2CH3). Anal. Calc. for
C14H14N2: C, 80.00; H, 6.67; N, 13.33. Found: C, 79.78; H, 5.1.13. 9-Phenylpropyl-1-methyl-b-carboline (26)
6.96; N, 13.18. A mixture of 20 (1.82 g, 10 mmol) and anhydrous DMF
(60 ml) was stirred at RT until clear, and then 60% NaH (0.6 g,
5.1.10. 9-n-Butyl-1-methyl-b-carboline (23) 15 mmol) and 1-bromo-3-phenylpropane (6 ml, 40 mmol)
A mixture of 20 (1.82 g, 10 mmol) and anhydrous DMF were added and refluxed for 2 h, and then treated in a manner
(60 ml) was stirred at RT until clear, and then 60% NaH (0.6 g, similar to that described for 21 to afford white crystals 26
15 mmol) and n-butyl iodide (6 ml, 50 mmol) were added. (2.4 g, 80%), m.p. 119–120 °C (from ether); FAB-MS m/z
The mixture was stirred at RT for 2 h, and then treated in a (M + 1) 301; UV kmax 358, 344, 289, 243, 237 nm IR (KBr)
manner similar to that described for 21 to afford white crys- 3028, 2931, 1617, 1560, 1447, 1363, 1234, 1118 cm–1;
1
tals 23 (1.8 g, 76%), m.p. 124–126 °C (from ether); FAB-MS H-NMR (500 MHz, CDCl3) d 8.29–8.30 (1H, d, J = 5.0 Hz,
m/z (M + 1) 239; UV kmax 358, 344, 289, 242, 236 nm IR H-4), 8.07–8.09 (1H, d, J = 8.0 Hz, H-8), 7.79–7.80 (1H, d,
(KBr) 2962, 2589, 1618, 1564, 1472, 1357, 1255, 1119 cm–1; J = 5.0 Hz, H-3), 7.51–7.54 (1H, m, H-5), 7.17–7.31 (7H, m,
R. Cao et al. / European Journal of Medicinal Chemistry 40 (2005) 991–1001 999

H-6, H-7, ArH), 4.49–4.52 (2H, m, NCH2CH2CH2Ar), 2.89 C13H13IN2: C, 48.15; H, 4.01; N, 8.64. Found: C, 48.03; H,
(3H, s, CH3), 2.72–2.75 (2H, m, NCH2CH2CH2Ar), 2.12– 4.25; N, 8.52.
2.18 (2H, m, NCH2CH2CH2Ar). Anal. Calc. for C21H20N2:
C, 84.00; H, 6.67; N, 9.33. Found: C, 79.86; H, 6.87; N, 9.25. 5.1.17. 2,9-Diethyl-b-carbolinium iodate (31)
A mixture of 29 (0.84 g, 5 mmol) and anhydrous DMF
5.1.14. 2,9-Dibenzyl-1-methyl-b-carbolinium bromate (27) (30 ml) was stirred at RT until clear, and then 60% NaH (0.6 g,
A mixture of 20 (1.82 g, 10 mmol) and anhydrous DMF 15 mmol) and iodoethane (2.5 ml, 30 mmol) were added.
(50 ml) was stirred at RT until clear, and then 60% NaH (0.6 g, The mixture was stirred at 50–60 °C for 2 h, and then follow-
15 mmol) and benzyl bromide (5 ml, 40 mmol) were added. ing the method described for 27 to afford yellow crystals 31
The mixture was stirred at 50–60 °C for 2 h. The resulting (1.1 g, 63%), m.p. 214–215 °C. FAB-MS m/z 225; UV kmax
mixture was poured into H2O, and extracted with ethyl acetate. 389, 310, 261, 257, 220, 210 nm IR (KBr) 3437, 2977, 1815,
The organic phase was washed with water and brine, and then 1639, 1509, 1458, 1243,1158 cm–1; 1H-NMR (CDCl3) d 9.61
dried over anhydrous sodium sulfate, filtered and evaporated. (1H, s, H-4), 8.68–8.69 (1H, d, J = 6.5 Hz, H-1), 8.61–8.63
The yellow solid was recrystallized from ethanol, and golden (1H, d, J = 6.5 Hz, H-8), 8.42–8.44 (1H, d, J = 8.0 Hz, H-3),
crystals of 27 were obtained (3.6 g, 72%), m.p. > 270 °C; 7.84–7.89 (2H, m, H-5, H-6), 7.48–7.51 (1H, m, H-7), 4.83–
FAB-MS m/z 363; UV kmax 383, 312, 260 nm IR (KBr) 3032, 4.87 (2H, m, N+CH2CH3), 4.67–4.72 (2H, m, NCH2CH3),
3004, 1623, 1573, 1456, 1336, 1247, 1221 cm–1; 1H-NMR 1.75–1.78 (3H, m, N + CH 2 CH 3 ), 1.51–1.54 (3H, m,
(500 MHz, DMSO-d6) d 8.95–8.96 (1H, d, J = 6.5 Hz, H-4), NCH2CH3). Anal. Calc. for C15H17IN2: C, 51.14; H, 4.83; N,
8.89–8.91 (1H, d, J = 6.5 Hz, H-8), 8.61–8.62 (1H, d, 7.95. Found: C, 50.97; H, 5.03; N, 7.86.
J = 8.0 Hz, H-3), 7.89–7.91 (1H, d, J = 9.0 Hz, H-5), 7.83–
7.87 (1H, m, H-6), 7.53–7.56 (1H, m, H-7), 7.24–7.40 (6H, 5.1.18. 2,9-Dibenzyl-b-carbolinium bromate (32)
m, ArH), 7.15–7.17 (2H, d, J = 8.0 Hz, ArH), 7.01–7.02 (2H, A mixture of 29 (1.68 g, 10 mmol) and anhydrous DMF
d, J = 7.0 Hz, ArH), 6.08 (2H, s, N+CH2Ar), 6.05 (2H, s, (60 ml) was stirred at RT until clear, and then 60% NaH (1.2 g,
NCH2Ar), 2.94 (3H, s, CH3). Anal. Calc. for C26H23BrN2: C, 30 mmol) and benzyl bromide (5 ml, 40 mmol) were added.
70.43; H, 5.19; N, 6.32. Found: C, 70.32; H, 5.43; N, 6.42. The mixture was stirred at 50–60 °C for 2 h, and then follow-
ing the method described for 27 to afford golden crystals 32
5.1.15. 2,9-Dibenzyl-3-ethoxycarbonyl-1-methyl-b-carbo- (3.6 g, 76%), m.p. > 270 °C. FAB-MS m/z 349; UV kmax 390,
linium bromate (28) 313, 262, 235, 205 nm IR (KBr) 3409, 2982, 1644, 1511,
A mixture of 4 (1.2 g, 5 mmol) and anhydrous DMF (30 ml) 1453, 1337, 1211, 1134 cm–1; 1H-NMR (CDCl3) d 9.57 (1H,
was stirred at RT until clear, and then 60% NaH (1.2 g, s, H-4), 8.71 (2H, s, H-1, H-8), 8.42–8.44 (1H, d, J = 8.5 Hz,
30 mmol) and benzyl bromide (2.5 ml, 20 mmol) were added. H-3), 7.81–7.82 (2H, m, H-5, H-6), 7.40–7.50 (6H, m, H-7,
The mixture was stirred at 50–60 °C for 2 h, and following ArH), 7.19–7.28 (5H, m, ArH), 5.95 (2H, s, N+CH2Ar), 5.86
the method described for 27 to afford golden crystals 28 (2.3 g, (2H, s, N CH2Ar). Anal. Calc. for C25H21BrN2: C, 69.93; H,
84%), m.p. > 270 °C. FAB-MS m/z 421; UV kmax 397, 317, 4.90; N, 6.53. Found: C, 69.84; H, 5.13; N, 6.46.
284, 243 nm IR (KBr) 3421, 2976, 1726, 1630, 1457, 1367,
1257, 1096 cm–1; 1H-NMR (500 MHz, CDCl3) d 9.88 (1H, s, 5.1.19. Benzyl 9-benzyl-b-carboline-3-carboxylate (34)
H-4), 9.30 (1H, s, H-1), 8.55–8.57 (1H, d, J = 7.5 Hz, H-8), A mixture of compound 33 (2.12 g, 10 mmol) and anhy-
7.87–7.92 (2H, m, H-5, H-6), 7.55–7.59 (1H, m, H-7), 7.20– drous DMF (50 ml) was stirred at RT till clear, and then 60%
7.36 (10H, m, ArH), 6.39 (2H, m, N+CH2Ar), 5.98 (2H, s, NaH (1.2 g, 30 mmol) and benzyl bromide (5 ml, 40 mmol)
NCH2Ar), 4.44–4.48 (2H, m, OCH2CH3), 1.34–1.37 (3H, m, were added and stirred at RT for 2 h. The resulting solution
OCH2CH3). Anal. Calc. for C28H25BrN2O2: C, 67.07; H, 4.99; was poured into H2O (100 ml), and extracted with ethyl
N, 5.59. Found: C, 66.89; H, 5.23; N, 5.47. acetate (3 × 200 ml). The organic phase was washed with
water and brine, then dried over anhydrous sodium sulfate,
5.1.16. 2,9-Dimethyl-b-carbolinium iodate (30) filtered and evaporated. The oil obtained was purified by silica
A mixture of 29 (0.84 g, 5 mmol) and anhydrous DMF column chromatography with ethyl acetate as the eluent. Upon
(30 ml) was stirred at RT until clear, and then 60% NaH (0.6 g, recrystallization, white crystals of 34 were obtained (2.3 g,
15 mmol) and iodomethane (2 ml, 30 mmol) were added. 57%), m.p. 169–170 °C. FAB-MS m/z (M + 1) 393; UV kmax
The mixture was stirred at 50–60 °C for 1 h, and then follow-
356, 341, 305, 273, 236 nm IR (KBr) 3400, 3064, 2935, 1709,
ing the method described for 27 to afford yellow crystals 30
1623,1584, 1497, 1461, 1336, 1244, 1109 cm–1; 1H-NMR
(1.2 g, 73%), m.p. > 270 °C. FAB-MS m/z 197; UV kmax 390,
(CDCl3) d 8.96 (1H, s, H-4), 8.91 (1H, s, H-1), 8.22–8.24
309, 261, 257, 220, 210 nm IR (KBr) 3447, 2985, 1642, 1517,
(1H, m, H-8), 7.60–7.64 (1H, m, H-5), 7.49–7.55 (3H, m,
1467, 1376, 1335, 1262, 1153 cm–1; 1H-NMR (CDCl3) d 9.42
H-6, H-7, ArH), 7.31–7.40 (4H, m, ArH), 7.25–7.29 (3H, m,
(1H, s, H-4), 8.66–8.67 (1H, d, J = 6.5 Hz, H-1), 8.51–8.52
ArH), 7.12–7.14 (2H, m, ArH), 5.63 (2H, s, OCH2Ar), 5.52
(1H, d, J = 6.0 Hz, H-8), 8.43–8.45 (1H, d, J = 8.0 Hz, H-3),
(2H, s, NCH2Ar). Anal. Calc. for C26H20N2O2: C, 79.59; H,
7.83–7.91 (2H, m, H-5, H-6), 7.50–7.53 (1H, m, H-7), 4.57
5.10; N, 7.14. Found: C, 79.48; H, 5.26; N, 7.19.
(3H, m, N+CH3), 4.13 (3H, m, NCH3). Anal. Calc. for
1000 R. Cao et al. / European Journal of Medicinal Chemistry 40 (2005) 991–1001

5.1.20. 3-Acetyloxymethyl-b-carboline (37) H-6), 7.19–7.31 (6H, m, H-7, ArH), 5.76 (2H, s, NCH2Ar),
A mixture of compound 36 (1.98 g, 10 mmol) and acetic 5.28 (2H, s, CH 2 ), 2.11 (3H, s, CH 3 ). Anal. Calc. for
acid (50 ml) was refluxed for 2 h. The solvent was evaporated C21H18N2O2: C, 76.36; H, 5.45; N, 8.48. Found: C, 76.24; H,
in vacuum, and water (50 ml) was added. The aqueous sus- 5.68; N, 8.39.
pension was extracted with ethyl acetate (3 × 200 ml). The
organic extracts were combined, and the solvent was evapo- 5.1.23. Ethyl 9-n-butyl-1-methyl-b-carboline-3-carboxylate
rated in vacuum. The residue was chromatographed on silica (42)
gel with petroleum ether/acetone (2:1) as the eluent. Upon A mixture of 41 (2.54 g, 10 mmol) and anhydrous DMF
recrystallization, white crystals of 37 were obtained (2.2 g, (60 ml) was stirred at RT until clear, and then 60% NaH (0.6 g,
92%), m.p. 131–132 °C (from ether); FAB-MS m/z (M + 1) 15 mmol) and n-butyl iodide (6 ml, 50 mmol) were added.
241; UV kmax 326, 315, 284, 259, 230, 207 nm IR (KBr) The mixture was stirred at RT for 1 h. The resulting mixture
3371, 2938, 1743, 1692, 1616, 1450, 1372, 1238 cm–1; was poured into H2O, and extracted with ethyl acetate. The
1 organic phase was washed with water and brine, then dried
H-NMR (500 MHz, CDCl3) d 9.47 (1H, s, NH), 9.03 (1H, s,
H-4), 8.32–8.34 (1H, d, J = 8.0 Hz, H-1), 8.26–8.28 (1H, d, over anhydrous sodium sulfate, filtered and evaporated. The
J = 8.5 Hz, H-8), 8.19–8.20 (1H, m, H-5), 7.67–7.70 (1H, m, oil obtained was purified by silica column chromatography
H-6), 7.47–7.51 (1H, m, H-7), 5.29 (2H, s, CH2), 2.16 (3H, s, with ethyl acetate as the eluent. Upon recrystallization, white
CH3). Anal. Calc. for C14H12N2O2: C, 70.00; H, 5.00; N, crystals of 42 were obtained (2.3 g, 74%), m.p. 83–84 °C
11.67. Found: C, 69.91; H, 5.19; N, 11.58. (from ether); FAB-MS m/z (M + 1) 311; UV kmax 355, 338,
308, 273, 240 nm IR (KBr) 3059, 2964, 2868, 1680, 1620,
5.1.21. 9-Benzyl-3-hydroxymethyl-b-carboline (38) 1557, 1456, 1342, 1270, 1138 cm–1; 1H-NMR (500 MHz,
A suspension of compound 11 (3.3 g, 10 mmol) in THF CDCl3) d 8.72 (1H, s, H-4), 8.14–8.16 (1H, d, J = 8.0 Hz,
(400 ml) was treated with LiAlH4 (1.2 g, 30 mmol), and the H-8), 7.58–7.61 (1H, m, H-5), 7.46–7.48 (1H, d, J = 8.5 Hz,
mixture was refluxed for 10 h. The reaction mixture was H-6), 7.30–7.33 (1H, m, H-7), 4.50–4.55 (4H,
cooled and treated with H2O and stirred overnight. The sol- NCH2CH2CH2CH3, OCH2CH3), 3.11 (3H, s, CH3), 1.79–
vent was evaporated in vacuum and H2O was added. The aque- 1.85 (2H, m, NCH 2 CH 2 CH 2 CH 3 ), 1.42–1.50 (5H, m,
ous suspension was extracted with ethyl acetate. The com- NCH 2 CH 2 CH 2 CH 3 , OCH 2 CH 3 ), 0.96–0.99 (3H, m,
bined organic extracts was washed with water and brine, then NCH2CH2CH2CH3). Anal. Calc. for C19H22N2O2: C, 73.55;
dried over anhydrous sodium sulfate, filtered and evaporated. H, 7.10; N, 9.03. Found: C, 73.43; H, 7.29; N, 8.96.
The oil obtained was purified by silica column chromatogra-
phy with ethyl acetate as the eluent. Upon recrystallization, 5.1.24. 9-n-Butyl-1-methyl-b-carboline-3-carboxylic acid (43)
white crystals of 2a were obtained (1.8 g, 80%), m.p. 120– A mixture of compound 42 (1.55 g, 5 mmol), NaOH (0.8 g,
122 °C (from ethyl acetate); FAB-MS m/z (M + 1) 289; UV 20 mmol), ethanol (50 ml) and H2O (100 ml) was refluxed
kmax 361, 347, 291, 284, 239, 215 nm IR (KBr) 3170, 2938, for 1 h, and the ethanol was removed on the rotary evapora-
1627, 1559, 1467, 1363, 1264 cm–1; 1H-NMR (500 MHz, tor. The mixture was neutralized (pH 5) with 5 M HCl and
CDCl3) d 8.73 (1H, s, H-4), 8.13–8.15 (1H, d, J = 8.0 Hz, cooled. The precipitate was collected, washed well with H2O
H-1), 7.96 (1H, s, H-8), 7.54–7.58 (1H, m, H-5), 7.41–7.43 and dried in vacuum, yellow solid was obtained (1.4 g, 99%).
(1H, d, J = 8.5 Hz, H-6), 7.28–7.31 (1H, m, H-7), 7.23–7.27 M.p. 187–188 °C; FAB-MS m/z (M + 1) 283; UV kmax 356,
(3H, m, ArH), 7.11–7.12 (2H, m, ArH), 5.51 (2H, s, CH2Ar), 341, 271, 240 nm IR (KBr) 3250–2100, 1745, 1622, 1586,
4.94 (2H, s, CH2OH), 4.01 (1H, s, CH2OH). Anal. Calc. for 1365, 1205 cm–1; 1H-NMR (500 MHz, DMSO-d6) d 8.76 (1H,
C19H16N2O: C, 79.17; H, 5.56; N, 9.72. Found: C, 79.10; H, s, H-4), 8.35–8.37 (1H, d, J = 7.5 Hz, H-8), 7.74–7.76 (1H, d,
5.72; N, 9.63. J = 8.5 Hz, H-5), 7.63–7.66 (1H, m, H-6), 7.32–7.35 (1H, m,
H-7), 4.60–4.64 (2H, m, NCH2CH2CH2CH3), 3.04 (3H, s,
5.1.22. 9-Benzyl-3-acetyloxymethyl-b-carboline (39) CH3), 1.73–1.79 (2H, m, NCH2CH2CH2CH3), 1.35–1.43
A mixture of compound 38 (1.44 g, 5 mmol) and acetic acid (2H, m, NCH 2 CH 2 CH 2 CH 3 ), 0.91–0.94 (3H, m,
(30 ml) was refluxed for 2 h. The solvent was evaporated in NCH2CH2CH2CH3). Anal. Calc. for C17H18N2O2: C, 72.34;
vacuum, and water (30 ml) was added. The aqueous suspen- H, 6.38; N, 9.93. Found: C, 72.23; H, 6.57; N, 9.97.
sion was extracted with ethyl acetate (3 × 100 ml). The organic
extracts were combined, and the solvent was evaporated in 5.2. Pharmacology
vacuum. The residue was chromatographed on silica gel with 5.2.1. Cytotoxicity assays in vitro
ethyl acetate as the eluent. Upon recrystallization, white crys- Cytotoxicity assays in vitro were carried out using 96 mi-
tals of 39 were obtained (1.5 g, 94%), m.p. 141–142 °C (from crotitre plate cultures and MTT staining according to the pro-
ether); FAB-MS m/z (M + 1) 331; UV kmax 360, 346, 291, cedures described by Al-Allaf and Rashan [18]. Briefly, cells
284, 239, 214 nm IR (KBr) 3428, 3027, 2942, 1726, 1622, were grown in RPMI-1640 medium containing 10% (v/v) fetal
1452, 1352, 1245 cm–1; 1H-NMR (500 MHz, CDCl3) d 9.01 bovine serum and 100 ug ml–1 penicillin and 100 ug ml–1
(1H, s, H-4), 8.30–8.31 (1H, d, J = 8.0 Hz, H-1), 8.20 (1H, s, streptomycin. Cultures were propagated at 37 °C in a humi-
H-8), 7.73–7.75 (1H, d, J = 8.5 Hz, H-5), 7.58–7.61 (1H, m, fied atmosphere containing 5% CO2. Drug stock solutions
R. Cao et al. / European Journal of Medicinal Chemistry 40 (2005) 991–1001 1001

were prepared in DMSO. The final concentration of DMSO Acknowledgements


in the growth medium was 2% (v/v) or lower, the concentra- This work was supported by Xinjiang Huashidan Pharmaceu-
tion without effect on cell replication. The tumor cell line tical Co. Ltd. and Guangzhou Commission of Science and Tech-
panel consisted of non-small cell lung carcinoma (PLA- nology (97-Z-12-01). We also thank Center for Analysis and Test
801), liver carcinoma (HepG2 and Bel-7402), gastric carci- of SunYat-sen (Zhongshan) University for UV, FAB-MS, IR and
noma (BGC-823), cervical carcinoma (Hela), colon carci- 1
H-NMR spectra as well as elemental analyses data. We are also
noma (Lovo). In all of these experiments, three replicate wells grateful to Professor Huifang Yan and co-workers for perfor-
were used to determine each point. mance of the antitumor and acute toxic assays in mice.

5.2.2. Assay of acute toxicity References


Acute toxicity assay was performed according to the
method described by Cao et al. [22]. Briefly, healthy [1] M.M. Airaksinen, I. Kari, Med. Biol. 59 (1981) 21–34.
[2] T.S. Zhou, W.C. Ye, Z.T. Wang, Phytochemistry 49 (1998) 1807–1809.
C57BL/6 mice (9–12 weeks) weighing 18–22 g were housed [3] G.M. Carbrera, A.M. Seldes, J. Nat. Prod. 62 (1999) 759–760.
in rooms where the temperature was approximately 24 ± 2 °C [4] S. Budavari, in: The Merck Index, Merck and Co. Inc, 11th edn.,
with a relative humidity 60–70%, and in 12 h light–dark cycle. Rahway, NJ, 1989.
The sterile food and water were provided according to insti- [5] R.S. Kusurkar, S.K. Goswami, S.M. Vyas, Tetrahedron Lett. 44
(2003) 4761–4763.
tutional guidelines. Prior to each experiment, mice were fas- [6] K.P. Lippke, W.G. Schunack, W. Wenning, W.E. Muller, J. Med.
tened overnight and allowed free access to water. Various Chem. 26 (1983) 499–503.
doses of the b-carboline derivatives ranging from 10.0– [7] M. Cain, R.W. Weber, F. Guzman, J.M. Cook, S.A. Barker, K.C. Rice,
500.0 mg kg–1 dissolved in 0.5% carboxymethyl cellulose et al., J. Med. Chem. 25 (1982) 1081–1091.
sodium (CMC-Na) salt solution were given via intraperito- [8] T.J. Hagen, P. Skolnick, J.M. Cook, J. Med. Chem. 30 (1987) 750–753.
[9] R.H. Dodd, C. Ouannes, L.P. Carvalho, A. Valin, P. Venault,
neal (i.p.) to different groups of healthy C57BL/6 mice, and G. Chapouthier, et al., J. Med. Chem. 28 (1985) 824–828.
each group contained 10 mice (five males and five females). [10] C. Braestrup, M. Nielsen, C.E. Olsen, Proc. Natl. Acad. Sci. USA 77
After the administration of the compounds, mice were (1980) 2288–2292.
observed continuously for the first 2 h for any gross behav- [11] W. Schlecker, A. Huth, E. Ottow, J. Mulzer, Synthesis (Mass.) 131
(1995) 1225–1227.
ioral changes and deaths, then intermittently for the next 24 h
[12] A. Batch, R.H. Dodd, J. Org. Chem. 63 (1998) 872–877.
and occasionally thereafter for 14 days, and for the onset of [13] P. Molina, P.M. Fresneda, S. Garcia-Zafra, P. Almendros, Tetrahedron
any delayed effects. All animals were sacrificed at the 14th Lett. 35 (1994) 8851–8853.
day after drug administration and checked macroscopically [14] N. Lin, M. Zhao, C. Wang, S. Peng, Bioorg. Med. Chem. Lett. 12
for possible damage to the heart, liver and kidneys. Mice of (2002) 585–587.
[15] P. Rivas, B.K. Cassels, A. Morello, Y. Repetto, Comp. Biochem.
immediate death following drug administration were also Physiol. 122 (1999) 27–31.
examined for any possible organ damage. LD50 values were [16] K. Hayashi, M. Nagao, T. Sugimura, Nucleic Acids Res. 4 (1977)
calculated graphically as described [31]. 3679–3685.
[17] D. Csanyi, G. Hajos, Z. Riedl, Bioorg. Med. Chem. Lett. 10 (2000)
1767–1769.
5.2.3. Assay of antitumor activity [18] T.A.K. Al-Allaf, L.J. Rashan, Eur. J. Med. Chem. 33 (1998) 817–820.
Antitumor activity against Lewis lung cancer and Sar- [19] J. Ishida, H.-K. Wang, K.F. Bastow, C.-Q. Hu, K.-H. Lee, Bioorg.
coma 180 was performed as described by Cao et al. [22] with Med. Chem. 9 (1999) 3319–3324.
a slightly modification. Briefly, Lewis lung cancer and [20] P. Molina, P.M. Fresneda, S. Garcia-Zafra, P. Almendros, Tetrahedron
S180 sarcoma cell lines were provided by Shanghai Institute Lett. 36 (1995) 3581–3582.
[21] S.G. Abdel-Moty, S. Sakai, N. Aimi, H. Takayama, M. Kitajima,
of Pharmaceutical Industry. Tumor cells of Lewis lung can- A. E1-Shorbagi, A.N. Ahmed, N.M. Omar, Eur. J. Med. Chem. 32
cer and S180 sarcoma were inoculated to mice. After 7 days, (1997) 1009–1017.
tumors were taken out and cells harvested. Viable tumor cells [22] R. Cao, Q. Chen, X. Hou, H. Chen, H. Guan, Y. Ma, et al., Bioorg.
(2 × 106 cells per mouse) were inoculated to the armpit of Med. Chem. 12 (2004) 4613–4623.
[23] A. Brossi, A. Focella, S. Teitel, J. Med. Chem. 16 (1973) 418–420.
mice by subcutaneous injection. Each compound was injected
[24] H.R. Synder, C.H. Hansch, L. Katz, S.M. Parmerter, E.C. Spaeth, J.
by intraperitoneal (i.p.) to different group mice (each group Am. Chem. Soc. 70 (1948) 219–221.
containing 10 female mice) 24 h after the inoculation at dose [25] P. Lippke, G. Schunack, W. Wenning, W.E. Muller, J. Med. Chem. 26
about one third of LD50 value once a day for consecutive (1983) 499–503.
7 days. Cytophosphane (CTX) at 30.0 mg kg–1 was used as a [26] D.G. Harvey, E.J. Miller, W. Robson, J. Chem. Soc. (1941) 153–158.
[27] W.A. Jacobs, L.C. Craig, J. Biol. Chem. 113 (1936) 759–765.
positive control and vehicle as negative control. The weights [28] S.K. Srivastava, A. Agarwal, P.M.S. Chauhan, S.K. Agarwal, A.P. Bha-
of animals were recorded every 3 days. All animals were sac- duri, S.N. Singh, et al., Bioorg. Med. Chem. 7 (1999) 1223–1236.
rificed at the 21st day after tumor inoculation and the tumors [29] H.R. Snyder, S.M. Parmerter, L. Katz, J. Am. Chem. Soc. 70 (1948)
were excised and weighed. The inhibition rate was calcu- 222–225.
[30] S. Xiao, W. Lin, C. Wang, M. Yang, Bioorg. Med. Chem. Lett. 11
lated as follows:共 C–T 兲⁄C×100 (2001) 437–441.
T: average tumor weight of treated group; C: average tumor [31] J. Kassa, J. Vachek, Toxicology 177 (2002) 179–185.
weight of negative control group. [32] G. Gauzzo, G. Jori, J. Org. Chem. 37 (1972) 1429–1433.

You might also like