Next Article in Journal
Carotenoids in Marine Animals
Previous Article in Journal
The New Carotenoid Pigment Moraxanthin Is Associated with Toxic Microalgae
Previous Article in Special Issue
Recent N-Atom Containing Compounds from Indo-Pacific Invertebrates
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Synthesis of 1-Substituted Carbazolyl-1,2,3,4-tetrahydro- and Carbazolyl-3,4-dihydro-β-carboline Analogs as Potential Antitumor Agents

1
School of Pharmacy, National Taiwan University, Taipei 100, Taiwan
2
National Research Institute of Chinese Medicine, Taipei 112, Taiwan
3
Institute of Biochemistry, National Yang-Ming University, Taipei 112, Taiwan
*
Author to whom correspondence should be addressed.
Mar. Drugs 2011, 9(2), 256-277; https://doi.org/10.3390/md9020256
Submission received: 30 December 2010 / Revised: 31 January 2011 / Accepted: 7 February 2011 / Published: 10 February 2011
(This article belongs to the Special Issue Marine Alkaloids)

Abstract

:
A series of 1-substituted carbazolyl-1,2,3,4-tetrahydro- and carbazolyl-3,4-dihydro-β-carboline analogs have been synthesized and evaluated for antitumor activity against human tumor cells including KB, DLD, NCI-H661, Hepa, and HepG2/A2 cell lines. Among these, compounds 2, 6, 7, and 9 exhibited the most potent and selective activity against the tested tumor cells. As for inhibition of topoisomerase II, compounds 114 and 18 showed better activity than etoposide. Among them, compounds 3, 4, 7, 9, and 10 exhibited potent activity. The structure and activity relationship (SAR) study revealed correlation between carbon numbers of the side chain and biological activities. The molecular complex with DNA for compound 2 was proposed.

1. Introduction

Marine invertebrates are rich in β-carboline alkaloids [13]. These natural β-carboline metabolites have been found to possess interesting antitumor and antiviral activities [46]. Eudistomins [7,8] and manzamines [9,10], which were isolated from tunicates and sponges, respectively, are of particular interest. The antiviral eudistomines C and E were active against HSV-2, Vaccinia virus and RNA viruses [11]. In addition, the novel manzamines exhibited potent antitumor, antibacterial, antifungal and anti-HIV activities [1214]. The major compound, manzamine A, showed most potent activity against murine P-388 cells [15]. Our previous paper reported that manzamines A–D and H showed significant cytotoxicities against human KB-16, A-549 and HT-29 tumor cells [16]. The β-carboline and 3,4-dihydro-β-carboline moieties in manzamines appear to be essential for the biological activity. In order to investigate the structure and activity relationship (SAR) and bioactive center of manzamine A, the synthesis of 1-substituted carbazolyl-1,2,3,4-tetrahydro- and carbazolyl-3,4-dihydro-β-carboline derivatives were initiated [17]. The limited source of manzamine A make this current investigation more urgent and important. The purpose of this study does not include all SAR studies of manzamines and β-carboline analogs. However, a combination of β-carboline and various carbazole analogs is addressed. An attempt to realize the possible active center of manzamine A and the importance of the alkyl substituents on the nitrogen atom of carbazole ring was conducted. Thus, a facile synthetic method by the application of N-alkylation, Duff reaction [18], Pictet-Spengler reaction [19,20] and DDQ oxidation [21] succeeded in the production of compounds 118. In this communication, we wish to report the preparation, structural elucidation and bioactivities of 1-substituted carbazolyl-1,2,3,4-tetrahydro- and carbazolyl-3,4-dihydro-β-carboline analogs.

2. Results and Discussion

2.1. Analog Design and Chemistry

The basic strategy for the synthesis of the target substances involved molecular modeling and SAR studies of manzamine analogs. To improve our knowledge of the main structural requirements needed for high antitumor activity, we synthesized two new series of 1-substituted carbazolyl-1,2,3,4-tetrahydro-β-carboline and carbazolyl-3,4-dihydro-β-carboline derivatives based on the analog design of manzamine A. These new compounds bear an N-alkyl carbazole conjugated with a β-carboline-like nucleus. The main part of manzamine A illustrated in Figure 1 is similar in both shape and size to our target compounds. The distances between N atom on carbazole and N atoms on carboline-nucleus are almost the same as those in the main part. With the aim of studying the SAR, we focused on the lengh of the N-alkyl side chain on the carbazole ring. Compounds 1926 were prepared by N-alkylation of carbazole with the appropriate alkyl bromide as depicted in Scheme 1. Subsequent synthesis of compounds 2734 was achieved by Duff reaction, which required hexamethylenetetramine/trifloroacetic acid and the appropriate N-alkyl carbazole. Compounds 1, 3, 5, 7, 9, 11, 13, 15, and 17 (A series) were furnished from tryptamine and series of N-substituted 3-carbazolyl carboxyaldehydes (2734, and N-ethyl-3-carboxyaldehyde, which was purchased from Sigma-Aldrich Co.) by application of Pictet-Spengler cyclization. Subsequent oxidation of 1,2,3,4-tetrahydro-β-carbolines (A) by DDQ yielded 3,4-dihydro-β-carboline derivatives (B series: 2, 4, 6, 8, 10, 12, 14, 16, 18). The preparation and spectral data for 134 are described in the Experimental Section.

2.2. Biological Activity

Cytotoxicity of new products 118 was tested against KB (human mouth epidermoid carcinoma); DLD (human colon adenocarcinoma), NCI-H661 (human lung large cell carcinoma) and Hepa (human hepatoma), HepG2/A2 (human hepatoma) tumor cells in vitro. The IC50 values of these compounds are summarized in Table 1. Table 1 shows that compounds 112 exhibited significant and/or selective cytotoxic activities. Among them, compounds 2 and 6 are most potent against KB tumor cells selectively. Compound 7 is more potent than 2 against NCI-H661 tumor cells although compound 2 shows most promising activity against all tumor cells. On the other hand, compounds 1418 are inactive toward four tumor cells while compounds 1113 exhibit weak, marginal or no activity. Table 1 also shows that these compounds have selective cytotoxicity against HepG2/A2 tumor cells. Compounds 24 and 8 are active while others are inactive. In this assay, compound 2 exhibits most potent activity against the HepG2/A2 system.
The SAR study revealed that there was not a linear relationship between the carbon number of the side chain at N atom in carbazole and cytotoxicity. Nevertheless, we observed that, in general, elongation of the alkyl chain resulted in a decrease in activity. In fact, compounds 1118 showed very weak or no activity toward HepG2/A2, KB and NCI-H661 cells even though compound 11 was active in DLD assay.
Inhibition activity of compounds 118 was evaluated against human DNA topoisomerases I and II. Table 2 indicates that compounds 114 and 18 are more potent inhibitors of human DNA topoisomerase II than etoposide. Among the tested compounds, 3, 4, 7, 9 and 10 were most potent. On the other hand, all compounds were inactive or showed mild activity against DNA topoisomerase I. Compounds 3, 4, 9 and 10 were 10- to 15-fold more potent against topoisomerase II (compared to etoposide) and superior to compounds 1 and 2. On the other hand, compound 18 showed great activity while compounds 1517 did not.
Compound 7 showed much better activity than that of 2 suggesting that the different degrees of activity might be explained by the differences in binding affinity or bioavailability such as drug uptake and fate of metabolism.

2.3. Molecular Modeling

To rationalize the biological activity results obtained for this novel series of carbazolyl-3,4-dihydro-β-carbolines, we carried out a molecular modeling study of compound 2. The molecular modeling was performed by making use of two double strand DNA fragments (CGCTAGGG)2 and (CGCGAATTCGCGG)2. Figure 2 shows the result of d(CGCATGGG)2-compound 2 complex. It was found that the binding conformation of this complex was formed by intercalation. Unlike the usual planar intercalators, compound 2 inserts into base pairs of DNA in a scissor-like conformation. We observed that the carbazole moiety of compound 2 was parallel with the thymidine base. However, the conformation of d(CGCGAATTGCG)2-compound 2 complex, as illustrated in Figure 3, was formed by minor groove binding. In this case, compound 2 interacts with DNA by putting the carbazole moiety in the minor groove leaving the β-carboline chromophore outside. The estimated binding energy values for these two interaction forms are −16 and −186 kcal/mole, respectively. In these experiments, the occurrence of hydrogen bonding was observed in both conformations and resulted in the formation of a stable drug-DNA complex. The hydrogen bonding between the N-9 of β-carboline and the oxygen atom of C-2 in thymidine had a bonding length 1.86 and 2.02 Ǻ, respectively [22].

3. Experimental Section

3.1. General Experimental Procedures

All of melting points were taken on a Buchi mp B-540 apparatus and were uncorrected. UV and IR spectra were recorded on Hitachi U-3210 and JASCO A-100 IR spectrophotometers, respectively. EIMS spectra were obtained on a MAT 112S-JMS D300 spectrometer, using direct inlet systems. HRMS data were taken on a JMX 110 mass spectrometer. 1H- and 13C-NMR spectra were recorded on a Bruker FT-300 spectrometer. Analytical thin-layer chromatography (TLC) was carried out on Kiesel gel GF254 plates and detection was made under UV light. EM Kieselgel 60 (230–400 mesh ASTM) was used for column chromatography.

3.2. Synthesis of Compounds 1, 3, 5, 7, 9, 11, 13, 15, and 17

To a stirred solution of tryptamine (160 mg, 0.1 mmol), the appropriate substituted aldehyde (2734, and N-ethyl-3-carbazolyl carboxyaldehyde; 0.1 mmol) in acetic acid (30 mL) was heated to 100 °C. The reaction mixture was maintained at 100 °C for 24 h. After cooling, the reaction mixture was neutralized with NH4OH solution and extracted with CHCl3. The CHCl3 layer was evaporated under vacuum, and the residue was chromatographed on a Silica gel column (10 g) and eluted with solvent mixture of CHCl3/MeOH using the following ratios and volumes (70:1, 50:1, 30:1 and 10:1; each 30 mL), to afford compounds 1, 3, 5, 7, 9, 11, 13, 15, and 17 with a yield in the range of 80–85%.

3.2.1. 1-(9′-Ethyl-3′-carbazolyl)-1,2,3,4-tetrahydro-β-carboline (1)

Yellow solid; mp 240–242 °C; UV λmax 249, 269, 296, 332, 346 nm; IR (KBr) νmax 3164, 2973, 1600, 1471, 1334, 806 cm−1; 1H NMR (CDCl3) δH 5.36 (1H, s, H-1), 3.37 (1H, m, H-3a), 3.13 (1H, m, H-3b), 2.87 (1H, m, H-4a), 2.97 (1H, m, H-4b), 7.64 (1H, d, overlap, H-5), 7.17 (1H, m, H-6), 7.14 (1H, m, H-7), 7.11 (1H, d, overlap, H-8), 7.29 (1H, d, overlap, H-1′), 7.36 (1H, d, overlap, H-2′), 8.04 (1H, s, H-4′), 7.44 (1H, d, overlap, H-5′), 7.52 (1H, m, H-6′), 7.25 (1H, m, H-7′), 8.00 (1H, d, overlap, H-8′), 4.37 (2H, q, J = 7.1 Hz, H-1″), 1.45 (3H, t, J = 7.1 Hz, H-2″); 13C NMR (CDCl3) δC 58.4 (d, C-1), 43.0 (t, C-3), 43.0 (t, C-4), 110.0 (s, C-4a), 127.6 (s, C-4b), 118.2 (d, C-5), 119.3 (d, C-6), 121.6 (d, C-7), 111.1 (d, C-8), 136.1 (s, C-8a), 135.5 (s, C-9a), 108.7 (d, C-1′), 126.5 (d, C-2′), 132.3 (s, C-3′), 120.7 (d, C-4′), 122.8 (s, C-4′a), 123.1 (s, C-4′b), 108.7 (d, C-5′), 126.0 (d, C-6′), 119.1 (d, C-7′), 120.5 (d, C-8′), 139.8 (s, C-8′a), 140.4 (s, C-9′a), 37.6 (t, C-1″), 13.9 (q, C-2″); EIMS m/z 365 (100, M+), 364 (82), 336 (46), 335 (63), 306 (31), 183 (17), 171 (51), 160 (16), 143 (18), 115 (17), 77 (5); HREIMS m/z 365.1890 ([M]+, calcd. for C25H23N3, 365.1892).

3.2.2. 1-(9′-Propyl-3′-carbazolyl)-1,2,3,4-tetrahydro-β-carboline (3)

Yellow solid; mp 110–112 °C; UV λmax 269, 296, 332, 346 nm; IR (KBr) νmax 3168, 3050, 2964, 1598, 1469, 1371, 808 cm−1; 1H NMR (CDCl3) δH 5.37 (1H, s, H-1), 3.18 (1H, m, H-3a), 3.43 (1H, m, H-3b), 2.85 (1H, m, H-4a), 2.99 (1H, m, H-4b), 7.58 (1H, d, overlap, H-5), 7.19 (1H, m, H-6), 7.17 (1H, m, H-7), 7.16 (1H, d, overlap, H-8), 7.34 (1H, d, overlap, H-1′), 7.41 (1H, d, overlap, H-2′), 8.02 (1H, s, H-4′), 7.43 (1H, d, overlap, H-5′), 7.48 (1H, dd, overlap, H-6′), 7.23 (1H, dd, overlap, H-7′), 7.98 (1H, d, overlap, H-8′), 4.24 (2H, t, J = 7.0 Hz, H-1″), 1.89 (2H, m, H-2″), 0.96 (3H, t, J = 7.3 Hz, H-3″);13C NMR (CDCl3) δC 58.3 (d, C-1), 42.8 (t, C-3), 22.4 (t, C-4), 109.9 (s, C-4a), 127.5 (s, C-4b), 118.2 (d, C-5), 119.3 (d, C-6), 121.6 (d, C-7), 111.1 (d, C-8), 136.1 (s, C-8a), 135.0 (s, C-9a), 109.7 (d, C-1′), 126.4 (d, C-2′), 131.7 (s, C-3′), 120.6 (d, C-4′), 122.6 (s, C-4′a), 122.9 (s, C-4′b), 108.9 (d, C-5′), 125.9 (d, C-6′), 119.1 (d, C-7′), 120.4 (d, C-8′), 140.4 (s, C-8′a), 140.9 (s, C-9′a), 44.7 (t, C-1″), 22.4 (t, C-2″)11.8 (q, C-3″); EIMS m/z 379 (89, M+), 378 (100, M-1+),350 (65), 306 (31), 190 (22), 180 (22), 171 (58), 159 (43), 115 (13), 77 (4).

3.2.3. 1-(9′-[1″-Methyl]propyl-3′-carbazolyl)-1,2,3,4-tetrahydro-β-carboline (5)

Yellow solid; mp 127–130 °C; UV λmax 222, 297, 332, 346 nm; IR (KBr) νmax 3454, 2969, 2929, 1458, 1335, 742 cm−1; 1H NMR (CDCl3) δH 5.31 (1H, s, H-1), 3.16 (1H, m, H-3a), 3.42 (1H, m, H-3b), 2.87 (1H, m, H-4a), 2.97 (1H, m, H-4b), 7.64 (1H, d, overlap, H-5), 7.15 (1H, m, H-6), 7.15 (1H, m, H-7), 7.15 (1H, d, overlap, H-8), 7.53 (1H, d, overlap, H-1′), 7.33(1H, d, overlap, H-2′), 8.04 (1H, s, H-4′), 7.45 (1H, d, overlap, H-5′), 7.45 (1H, dd, overlap, H-6′), 7.21 (1H, dd, overlap, H-7′), 8.02 (1H, d, overlap, H-8′), 4.67 (2H, m, H-1″), 1.68 (2H, d, J = 6.9 Hz, H-2″), 2.03 (1H, m, H-3a″), 2.15 (1H, m, H-3b″), 0.96 (3H, t, J = 7.3 Hz, H-4″); 13C NMR (CDCl3) δC 58.3 (d, C-1), 43.0 (t, C-3), 19.1 (t, C-4), 110.0 (s, C-4a), 127.5 (s, C-4b), 118.2 (d, C-5), 109.3 (d, C-6), 121.5 (d, C-7), 110.1 (d, C-8), 135.9 (s, C-8a), 135.1 (s, C-9a), 110.3 (d, C-1′), 126.0 (d, C-2′), 131.6 (s, C-3′), 120.5 (d, C-4′), 122.9 (s, C-4′a), 123.8 (s, C-4′b), 110.2 (d, C-5′), 125.6 (d, C-6′), 118.7 (d, C-7′), 120.3 (d, C-8′), 140.5 (s, C-8′a), 139.7 (s, C-9′a), 53.0 (t, C-1″), 14.1 (t, C-2″), 28.0 (t, C-3″), 11.5 (t, C-4″); EIMS m/z 393 (100, M+), 364 (36), 336 (17), 306 (34), 193 (20), 180 (11), 171 (44), 144 (20), 115 (19), 57 (42).

3.2.4. 1-(9′-Pentyl-3′-carbazolyl)-1,2,3,4-tetrahydro-β-carboline (7)

Yellow solid; mp 129–130 °C; UV λmax 243, 266, 296, 332, 347 nm; IR (KBr) νmax 3405, 3164, 3050, 2929, 1600, 1467, 1338, 806 cm−1; 1H NMR (CDCl3) δH 5.28 (1H, s, H-1), 3.14 (1H, m, H-3a), 3.41 (1H, m, H-3b), 2.85 (1H, m, H-4a), 2.99 (1H, m, H-4b), 7.63 (1H, d, overlap, H-5), 7.19 (1H, m, H-6), 7.18 (1H, m, H-7), 7.16 (1H, d, overlap, H-8), 7.34 (1H, d, overlap, H-1′), 7.37 (1H, d, overlap, H-2′), 8.03 (1H, s, H-4′), 7.42 (1H, d, overlap, H-5′), 7.49 (1H, dd, overlap, H-6′), 7.25 (1H, dd, overlap, H-7′), 7.99 (1H, d, overlap, H-8′), 4.27 (2H, t, J = 7.1 Hz, H-1″), 1.91 (2H, m, H-2″), 1.41 (2H, m, H-3″), 1.41 (2H, m, H-4″), 0.96 (3H, t, J = 7.3 Hz, H-2″); 13C NMR (CDCl3) δC 58.4 (d, C-1), 43.1 (t, C-3), 22.6 (t, C-4), 110.0 (s, C-4a), 127.6 (s, C-4b), 118.2 (d, C-5), 119.3 (d, C-6), 121.6 (d, C-7), 111.0 (d, C-8), 136.0 (s, C-8a), 135.4 (s, C-9a), 109.0 (d, C-1′), 126.4 (d, C-2′), 132.1 (s, C-3′), 120.6 (d, C-4′), 122.6 (s, C-4′a), 123.0 (s, C-4′b), 108.9 (d, C-5′), 125.9 (d, C-6′), 119.0 (d, C-7′), 120.4 (d, C-8′), 140.3 (s, C-8′a), 140.9 (s, C-9′a), 43.2 (t, C-1″), 28.7 (t, C-2″), 29.4 (t, C-3″), 22.5 (t, C-4″), 14.0 (q, C-5″); EIMS m/z 407 (90, M+), 406 (100, M − H+), 378 (55), 306 (31), 204 (13), 179 (19), 171 (46), 159 (27), 115 (15), 55 (8).

3.2.5. 1-(9′-[3″-Methylbutyl]-3′-carbazolyl)-1,2,3,4-tetrahydro-β-carboline (9)

Yellow solid; mp 99–101 °C; UV λmax 222, 296 nm; IR (KBr) νmax 3404, 3192, 3050, 2922, 1468, 1331, 742 cm−1; 1H NMR (CDCl3) δH 5.34 (1H, s, H-1), 3.18 (1H, m, H-3a), 3.46 (1H, m, H-3b), 2.87 (1H, m, H-4a), 3.00 (1H, m, H-4b), 7.64 (1H, d, overlap, H-5), 7.17 (1H, dd, overlap, H-6), 7.15 (1H, dd, overlap, H-7), 7.14 (1H, d, overlap, H-8), 7.36 (1H, d, overlap, H-1′), 7.40 (1H, d, overlap, H-2′), 8.04 (1H, s, H-4′), 7.47 (1H, d, overlap, H-5′), 7.56 (1H, dd, overlap, H-6′), 7.22 (1H, dd, overlap, H-7′), 8.01 (1H, d, overlap, H-8′), 4.29 (2H, t, J = 7.2 Hz, H-1″), 1.73 (2H, m, H-2″), 1.73 (2H, m, H-3″), 1.02 (2H, d, J = 6.0 Hz, H-4″), 1.02 (3H, d, J = 6.0 Hz, H-5″); 13C NMR (CDCl3) δC 58.1 (d, C-1), 42.6 (t, C-3), 22.3 (t, C-4), 109.7 (s, C-4a), 127.3 (s, C-4b), 117.9 (d, C-5), 119.0 (d, C-6), 121.3 (d, C-7), 110.8 (d, C-8), 135.8 (s, C-8a), 135.2 (s, C-9a), 108.1 (d, C-1′), 126.2 (d, C-2′), 132.0 (s, C-3′), 120.4 (d, C-4′), 122.5 (s, C-4′a), 122.8 (s, C-4′b), 108.6 (d, C-5′), 125.7 (d, C-6′), 118.8 (d, C-7′), 120.2 (d, C-8′), 140.5 (s, C-8′a), 139.9 (s, C-9′a), 41.2 (t, C-1″), 37.3 (t, C-2″), 25.9 (t, C-3″), 22.4 (t, C-4″), 22.4 (q, C-5″); EIMS m/z 407 (90, M+), 378 (48), 319 (15), 306 (25), 204 (29), 180 (23), 171 (64), 160 (51), 55 (81), 43 (57).

3.2.6. 1-(9′-Octyl-3′-carbazolyl)-1,2,3,4-tetrahydro-β-carboline (11)

Yellow solid; mp 87–89 °C; UV λmax 247, 266, 296, 332, 347 nm; IR (KBr) νmax 3160, 3056, 2927, 1600, 1469, 1332, 806, 742 cm−1; 1H NMR (CDCl3) δH 5.32 (1H, s, H-1), 3.18 (1H, m, H-3a), 3.44 (1H, m, H-3b), 2.88 (1H, m, H-4a), 2.97 (1H, m, H-4b), 7.64 (1H, d, overlap, H-5), 7.19 (1H, m, H-6), 7.17 (1H, m, H-7), 7.15 (1H, d, overlap, H-8), 7.33 (1H, d, overlap, H-1′), 7.39 (1H, d, overlap, H-2′), 8.04 (1H, s, H-4′), 7.47 (1H, d, overlap, H-5′), 7.54 (1H, dd, overlap, H-6′), 7.27 (1H, dd, overlap, H-7′), 8.01 (1H, d, overlap, H-8′), 4.27 (2H, t, J = 7.0 Hz, H-1″), 1.86 (2H, m, H-2″), 1.26–1.35 (10H, overlap, H-3″∼H-7″), 0.88 (3H, t, J = 6.7 Hz, H-8″); 13C NMR (CDCl3) δC 58.4 (d, C-1), 42.9 (t, C-3), 22.5 (t, C-4), 110.0 (s, C-4a), 127.6 (s, C-4b), 118.2 (d, C-5), 119.1 (d, C-6), 121.6 (d, C-7), 111.1 (d, C-8), 136.1 (s, C-8a), 135.3 (s, C-9a), 109.0 (d, C-1′), 126.4 (d, C-2′), 132.1 (s, C-3′), 120.6 (d, C-4′), 122.6 (s, C-4′a), 123.0 (s, C-4′b), 108.9 (d, C-5′), 125.9 (d, C-6′), 118.7 (d, C-7′), 120.4 (d, C-8′), 140.3 (s, C-8′a), 140.9 (s, C-9′a), 43.2 (t, C-1″), 29.1 (t, C-2″), 27.4 (t, C-3″), 29.5 (t, C-4″), 29.3 (t, C-5″), 31.8 (t, C-6″), 22.7 (t, C-7″), 14.2 (q, C-8″); EIMS m/z 449 (83, M+), 448 (100, M − H+), 420 (57), 405 (7), 319 (20), 306 (25), 225 (9), 180 (17), 171 (37), 160 (15), 57 (11).

3.2.7. 1-(9′-Decyl-3′-carbazolyl)-1,2,3,4-tetrahydro-β-carboline (13)

Yellow solid; mp 102–104 °C; UV λmax 240, 266, 296, 332, 347 nm; IR (KBr) vmax 3048, 2925, 1598, 1467, 1332, 806 cm−1; 1H NMR (CDCl3) δH 5.37 (1H, s, H-1), 3.18 (1H, m, H-3a), 3.43 (1H, m, H-3b), 2.88 (1H, m, H-4a), 2.99 (1H, m, H-4b), 7.55 (1H, d, overlap, H-5), 7.15 (1H, m, H-6), 7.14 (1H, dd, m), 7.16 (1H, d, overlap, H-8), 7.33 (1H, d, overlap, H-1′), 7.40 (1H, d, overlap, H-2′), 8.02 (1H, s, H-4′), 7.38 (1H, d, overlap, H-5′), 7.47 (1H, dd, overlap, H-6′), 7.20 (1H, dd, overlap, H-7′), 8.00 (1H, d, overlap, H-8′), 4.25 (2H, t, J = 7.0 Hz, H-1″), 1.83 (2H, m, H-2″), 1.24–1.32 (14H, overlap, H-3″∼H-9″), 0.88 (3H, t, J =6.3 Hz, H-10″); 13C NMR (CDCl3) δC 58.0 (d, C-1), 42.4 (t, C-3), 21.6 (t, C-4), 109.8 (s, C-4a), 127.2 (s, C-4b), 118.2 (d, C-5), 119.4 (d, C-6), 121.8 (d, C-7), 110.9 (d, C-8), 135.9 (s, C-8a), 133.7 (s, C-9a), 108.9 (d, C-1′), 126.4 (d, C-2′), 130.1 (s, C-3′), 120.7 (d, C-4′), 122.4 (s, C-4′a), 122.9 (s, C-4′b), 108.7 (d, C-5′), 125.9 (d, C-6′), 119.0 (d, C-7′), 120.6 (d, C-8′), 140.4 (s, C-8′a), 140.7 (s, C-9′a), 43.1 (t, C-1″), 28.9 (t, C-2″), 27.2 (t, C-3″), 29.2–29.5 (t, C-4″∼C-7″), 31.8 (t, C-5″), 31.8 (t, C-8″), 22.6 (t, C-9″), 14.0 (q, C-10″); EIMS m/z 477 (87, M+), 476 (100, M − H+), 448 (46), 306 (21), 239 (15), 180 (25), 171 (56), 160 (43), 144 (13), 69 (12), 57 (36).

3.2.8. 1-(9′-Hexadecyl-3′-carbazolyl)-1,2,3,4-tetrahydro-β-carboline (15)

Yellow solid; mp 111–113 °C; UV λmax 240, 270 nm; IR (KBr) νmax 2952, 2923, 1467 cm−1; 1H NMR (CDCl3) δH 5.34 (1H, s, H-1), 3.15 (1H, m, H-3a), 3.39 (1H, m, H-3b), 2.86 (1H, m, H-4a), 3.00 (1H, m, H-4b), 7.60 (1H, d, overlap, H-5), 7.14 (1H, dd, overlap, H-6), 7.16 (1H, dd, overlap, m), 7.17 (1H, d, overlap, H-8), 7.36 (1H, d, overlap, H-1′), 7.39 (1H, d, overlap, H-2′), 8.01 (1H, s, H-4′), 7.34 (1H, d, overlap, H-5′), 7.48 (1H, dd, overlap, H-6′), 7.21 (1H, dd, overlap, H-7′), 8.03 (1H, d, overlap, H-8′), 4.24 (2H, t, J = 7.2 Hz, H-1″), 1.83 (2H, m, H-2″), 1.26–1.33 (14H, overlap, H-3″∼H-15″), 0.90 (3H, t, J = 6.6 Hz, H-16″); 13C NMR (CDCl3) δC 58.4 (d, C-1), 43.2 (t, C-3), 22.7 (t, C-4), 109.9 (s, C-4a), 127.4 (s, C-4b), 118.2 (d, C-5), 119.4 (d, C-6), 121.7 (d, C-7), 110.9 (d, C-8), 134.9 (s, C-8a), 135.9 (s, C-9a), 109.0 (d, C-1′), 126.3 (d, C-2′), 131.3 (s, C-3′), 120.4 (d, C-4′), 122.5 (s, C-4′a), 122.9 (s, C-4′b), 108.8 (d, C-5′), 125.8 (d, C-6′), 119.0 (d, C-7′), 120.6 (d, C-8′), 140.8 (s, C-8′a), 140.4 (s, C-9′a), 43.2 (t, C-1″), 22.7–31.9 (t, C-4″∼C-15″), 14.1 (q, C-16″) ; EIMS m/z 561 (4, M+), 368 (1), 313 (1), 236 (3), 180 (2), 97 (6), 83 (11), 69 (17), 57 (28), 44 (100).

3.2.9. 1-(9′-Eicosyl-3′-carbazolyl)-1,2,3,4-tetrahydro-β-carboline (17)

Yellow amorphous powder; UV λmax 221, 263 nm; IR (KBr) νmax 3048, 2925, 1598, 1467, 1332, 806 cm−1; 1H NMR (CDCl3) δH 5.38 (1H, s, H-1), 3.18 (1H, m, H-3a), 3.41 (1H, m, H-3b), 2.88 (1H, m, H-4a), 3.01 (1H, m, H-4b), 7.59 (1H, d, overlap, H-5), 7.14 (1H, dd, overlap, H-6), 7.16 (1H, dd, overlap, m), 7.17 (1H, d, overlap, H-8), 7.36 (1H, d, overlap, H-1′), 7.39 (1H, d, overlap, H-2′), 8.01 (1H, s, H-4′), 7.34 (1H, d, overlap, H-5′), 7.46 (1H, dd, overlap, H-6′), 7.21 (1H, dd, overlap, H-7′), 8.03 (1H, d, overlap, H-8′), 4.24 (2H, t, J = 7.2 Hz, H-1″), 1.83 (2H, m, H-2″), 1.26–1.33 (14H, overlap, H-3″∼H-19″), 0.91 (3H, t, J = 6.7 Hz, H-20″); 13C NMR (CDCl3) δC 57.2 (d, C-1), 40.8 (t, C-3), 20.0 (t, C-4), 108.5 (s, C-4a), 127.3 (s, C-4b), 117.9 (d, C-5), 119.1 (d, C-6), 121.9 (d, C-7), 111.0 (d, C-8), 130.8 (s, C-8a), 136.2 (s, C-9a), 108.8 (d, C-1′), 126.3 (d, C-2′), 131.8 (s, C-3′), 120.2 (d, C-4′), 122.1 (s, C-4′a), 122.8 (s, C-4′b), 108.7 (d, C-5′), 125.8 (d, C-6′), 118.9 (d, C-7′), 120.8 (d, C-8′), 140.6 (s, C-8′a), 140.5 (s, C-9′a), 42.9 (t, C-1″), 22.4–31.6 (t, C-2″∼C-19″), 13.7 (q, C-20″); EIMS m/z 617 (4, M+), 588 (1), 179 (6), 171 (26), 160 (43), 144 (4), 91 (4), 71 (15), 69 (10), 57 (59), 43 (100).

3.3. Synthesis of Compounds 2, 4, 6, 8, 10, 12, 14, 16, and 18

To a stirred solution of compound A (1, 3, 5, 7, 9, 11, 13, 15, 17, 0.68 mmol) in EtOH (2 mL) and CHCl3 (6 mL) at room temperature, 2,3-dichloro-5,6-dicyanobezoquinone (DDQ, 160 mg) was added. The reaction mixture was stirred for 30 mins. After concentration, the residue was applied on a prep. TLC and developed with CHCl3/MeOH (10:1) to yield a series of B (2, 4, 6, 8, 10, 12, 14, 16, and 18) (23–64% yield).

3.3.1. 1-(9′-Ethyl-3′-carbazolyl)-3,4-dihydro-β-carboline (2)

Yellow solid; mp 175–177 °C; UV λmax 236, 288, 323, 346 nm; IR (KBr) νmax 3164, 2973, 1600, 1471, 1334, 806 cm−1; 1H NMR (CDCl3) δH 3.70 (2H, t, J = 7.5 Hz, H-3), 2.93 (2H, t, J = 7.5 Hz, H-4), 7.64 (1H, d, overlap, H-5), 7.19 (1H, m, H-6), 7.25 (1H, m, H-7), 7.43 (1H, d, overlap, H-8), 7.32 (1H, d, overlap,H-1′), 7.87 (1H, d, overlap, H-2′), 8.52 (1H, s, H-4′), 7.30 (1H, d, overlap, H-5′), 7.46 (1H, m, H-6′), 7.22 (1H, m, H-7′), 8.01 (1H, d, overlap, H-8′), 4.14 (2H, q, J = 7.0 Hz, H-1″), 1.34 (3H, t, J = 7.0 Hz, H-2″); 13C NMR (CDCl3) δC 160.5 (s, C-1), 46.4 (t, C-3), 19.4 (t, C-4), 112.8 (s, C-4a), 127.7 (s, C-4b), 120.1 (d, C-5), 120.6 (d, C-6), 125.2 (d, C-7), 112.8 (d, C-8), 138.1 (s, C-8a), 127.7 (s, C-9a), 108.7 (d, C-1′), 126.8 (d, C-2′), 141.4 (s, C-3′), 121.4 (d, C-4′), 122.9 (s, C-4′a), 123.1 (s, C-4′b), 108.5 (d, C-5′), 126.1 (d, C-6′), 119.5 (d, C-7′), 121.1 (d, C-8′), 126.1 (s, C-8′a), 140.3 (s, C-9′a), 37.4 (t, C-1″), 13.7 (q, C-2″); EIMS m/z 363 (100, M+), 362 (90), 346 (16), 335 (20), 319 (9), 306 (10), 174 (15), 160 (19), 77 (4), 55 (11); HREIMS m/z 363.1724 ([M]+, calcd. for C25H21N3, 363.1735).

3.3.2. 1-(9′-Propyl-3′-carbazolyl)-3,4-dihydro-β-carboline (4)

Yellow solid; mp 126–128 °C; UV λmax 239, 288, 323 nm; IR (KBr) νmax 3064, 2929, 1596, 1467, 1336, 808, 746 cm−1; 1H NMR (CDCl3) δH 3.96 (2H, t, J = 8.2 Hz, H-3), 2.98 (2H, t, J = 8.3 Hz, H-4), 7.64 (1H, d, overlap, H-5), 7.17 (1H, m, H-6), 7.26 (1H, m, H-7), 7.44 (1H, d, overlap, H-8), 7.38 (1H, d, overlap, H-1′), 7.92 (1H, d, overlap, H-2′), 8.58 (1H, s, H-4′), 7.34 (1H, d, overlap, H-5′), 7.47 (1H, m, H-6′), 7.21 (1H, m, H-7′), 8.09 (1H, d, 7.6, H-8′), 4.21 (2H, t, J = 6.9 Hz, H-1″), 1.88 (2H, m, H-2″), 0.95(3H, t, 7.4); 13C NMR (CDCl3) δH 160.2 (s, C-1), 47.3 (t, C-3), 19.4 (t, C-4), 119.0 (s, C-4a), 125.4 (s, C-4b), 120.1 (d, C-5), 120.5 (d, C-6), 125.0 (d, C-7), 112.5 (d, C-8), 137.5 (s, C-8a), 127.8 (s, C-9a), 109.1 (d, C-1′), 126.5 (d, C-2′), 141.9 (s, C-3′), 120.9 (d, C-4′), 122.8 (s, C-4′a), 123.0 (s, C-4′b), 109.0 (d, C-5′), 126.3 (d, C-6′), 119.5 (d, C-7′), 120.8 (d, C-8′), 126.2 (s, C-8′a), 141.0 (s, C-9′a), 44.7 (t, C-1″), 22.3(t, C-2″), 11.7 (q, C-3″); EIMS m/z 377 (98, M+), 376 (100, M − H+), 346 (55), 319 (15), 306 (15), 205 (14), 180 (8), 174 (47), 159 (16), 115 (11), 77 (9), 57 (21); HREIMS m/z 377.1880 ([M]+, calcd. for C26H23N3, 377.1882).

3.3.3. 1-(9′-[1″-Methyl]propyl-3′-carbazolyl)-3,4-dihydro-β-carboline (6)

Yellow solid; mp 151–154 °C; UV λmax 237, 288 nm; IR (KBr) νmax 3051, 2921, 1468, 1335, 743 cm−1; 1H NMR (CDCl3) δH 3.67 (2H, t, J = 8.1 Hz, H-3), 2.88 (2H, t, J = 8.1 Hz, H-4), 7.68 (1H, d, overlap, H-5), 7.19 (1H, m, H-6), 7.37 (1H, m, H-7), 7.56 (1H, d, overlap, H-8), 7.43 (1H, d, overlap,H-1′), 8.05 (1H, d, overlap, H-2′), 8.83 (1H, s, H-4′), 7.45 (1H, d, overlap, H-5′), 7.34 (1H, m, H-6′), 7.08 (1H, m, H-7′), 8.08 (1H, d, 7.6, H-8′), 4.56 (2H, t, J = 6.9 Hz, H-1″), 1.56 (2H, m, H-2″), 1.95 (3H, m, H-3″), 0.70 (3H, m, H-4″); 13C NMR (CDCl3) δC 160.2 (s, C-1), 43.0 (t, C-3), 19.4 (t, C-4), 113.9 (s, C-4a), 125.8 (s, C-4b), 120.9 (d, C-5), 121.6 (d, C-6), 126.8 (d, C-7), 113.9 (d, C-8), 140.9 (s, C-8a), 126.2 (s, C-9a), 111.0 (d, C-1′), 127.9 (d, C-2′), 140.8 (s, C-3′), 123.7 (d, C-4′), 123.2 (s, C-4′a), 123.7 (s, C-4′b), 111.0 (d, C-5′), 127.9 (d, C-6′), 120.5 (d, C-7′), 121.8 (d, C-8′), 124.0 (s, C-8′a), 140.1 (s, C-9′a), 53.7 (t, C-1″), 19.1 (q, C-2″), 28.1 (t, C-3″), 11.6 (q, C-4″); EIMS m/z 391 (72, M+), 360 (100), 332 (45), 306 (33), 180 (74), 167 (40), 140 (20), 115 (31), 57 (35), 41 (88); HRESIMS m/z 392.2125 ([M + H]+, calcd. for C27H26N3, 392.2127).

3.3.4. 1-(9′-Pentyl-3′-carbazolyl)-3,4-dihydro-β-carboline (8)

Yellow solid; mp 97–98 °C; UV λmax 236, 288, 324 nm; IR (KBr) νmax 3064, 2929, 1601, 1467, 1334, 808 cm−1; 1H NMR (CDCl3) δH 3.40 (2H, t, J = 7.0 Hz, H-3), 2.91 (2H, t, J = 7.8 Hz, H-4), 7.62 (1H, d, overlap, H-5), 7.21 (1H, m, H-6), 7.34 (1H, m, H-7), 7.57 (1H, d, overlap, H-8), 7.27 (1H, d, overlap,H-1′), 7.97 (1H, d, overlap, H-2′), 8.64 (1H, s, H-4′), 7.31 (1H, d, overlap, H-5′), 7.48 (1H, m, H-6′), 7.25 (1H, m, H-7′), 8.06 (1H, d, 7.6, H-8′), 3.90 (2H, t, J = 6.9 Hz, H-1″), 1.71 (2H, m, H-2″), 1.28 (3H, m, H-3″), 1.34 (3H, m, H-4″), 0.84 (t, 6.6, H-5″); 13C NMR (CDCl3) δC 160.5 (s, C-1), 44.9 (t, C-3), 19.3 (t, C-4), 119.4 (s, C-4a), 124.2 (s, C-4b), 120.1 (d, C-5), 120.6 (d, C-6), 125.7 (d, C-7), 113.0 (d, C-8), 138.8 (s, C-8a), 127.2 (s, C-9a), 108.7 (d, C-1′), 127.0 (d, C-2′), 142.0 (s, C-3′), 121.7 (d, C-4′), 122.6 (s, C-4′a), 123.0 (s, C-4′b), 108.6 (d, C-5′), 126.0 (d, C-6′), 119.4 (d, C-7′), 121.1 (d, C-8′), 124.8 (s, C-8′a), 140.5 (s, C-9′a), 42.6 (t, C-1″), 28.3 (t, C-2″), 29.1 (t, C-3″), 22.2 (t, C-4″), 13.8 (q, C-5″); EIMS m/z 405 (83, M+), 404 (100, M − H+), 377 (25), 346 (29), 332 (9), 306 (13), 203 (10), 174 (55), 159 (25), 57 (16); HREIMS m/z 405.2199 ([M]+, calcd. for C28H27N3, 405.2205).

3.3.5. 1-(9′-[3″-Methylbutyl]-3′-carbazolyl)-3,4-dihydro-β-carboline (10)

Yellow solid; mp 135–137 °C; UV λmax 236, 289 nm; IR (KBr) νmax 3068, 2927, 1587, 1465, 1332, 741 cm−1; 1H NMR (CDCl3) δH 3.56 (2H, t, J = 8.0 Hz, H-3), 2.76 (2H, t, J = 8.0 Hz, H-4), 7.41 (1H, d, overlap, H-5), 7.16 (1H, m, H-6), 7.41 (1H, m, H-7), 7.79 (1H, d, overlap, H-8), 7.24 (1H, d, overlap,H-1′), 8.03 (1H, d, overlap, H-2′), 8.86 (1H, s, H-4′), 7.16 (1H, d, overlap, H-5′), 7.36 (1H, m, H-6′), 7.14 (1H, m, H-7′), 8.12 (1H, d, J = 7.6 Hz, H-8′), 3.74 (2H, t, J = 6.9 Hz, H-1″), 1.44 (2H, m, H-2″), 1.54 (2H, m, H-3″), 0.90 (6H, d, J = 7.6 Hz, H-4″, 5″); 13C NMR (CDCl3) δC 160.9 (s, C-1), 41.2 (t, C-3), 19.3 (t, C-4), 114.3 (s, C-4a), 126.8 (s, C-4b), 120.6 (d, C-5), 121.5 (d, C-6), 126.8 (d, C-7), 114.3 (d, C-8), 140.6 (s, C-8a), 126.8 (s, C-9a), 109.3 (d, C-1′), 128.8 (d, C-2′), 143.1 (s, C-3′), 123.4 (d, C-4′), 121.7 (s, C-4′a), 122.5 (s, C-4′b), 108.9 (d, C-5′), 128.0 (d, C-6′), 120.4 (d, C-7′), 121.6 (d, C-8′), 124.2 (s, C-8′a), 140.6 (s, C-9′a), 41.2 (t, C-1″), 37.1 (q, C-2″), 26.1 (t, C-3″), 22.4 (q, C-4″, 5″); EIMS m/z 405 (22, M+), 346 (13), 332 (3), 180 (2), 174 (10), 160 (5), 83 (7), 69 (12), 55 (20), 44 (100); HRESIMS m/z 406.2284 ([M + H]+, calcd. for C28H28N3, 406.2283).

3.3.6. 1-(9′-Octyl-3′-carbazolyl)-3,4-dihydro-β-carboline (12)

Yellow solid; mp 91–92 °C; UV λmax 226, 287, 324 nm; IR (KBr) νmax 3058, 2927, 1596, 1467, 1336, 744 cm−1; 1H NMR (CDCl3) δH 3.77 (2H, t, J = 8.3 Hz, H-3), 2.88 (2H, t, J = 8.7 Hz, H-4), 7.62 (1H, d, overlap, H-5), 7.17 (1H, m, H-6), 7.30 (1H, m, H-7), 7.45 (1H, d, overlap, H-8), 7.33 (1H, d, overlap,H-1′), 7.87 (1H, d, overlap, H-2′), 8.53 (1H, s, H-4′), 7.36 (1H, d, overlap, H-5′), 7.51 (1H, m, H-6′), 7.19 (1H, m, H-7′), 8.03 (1H, d, 7.6, H-8′), 4.08 (2H, t, J = 6.9 Hz, H-1″), 1.76 (2H, m, H-2″), 1.30 (3H, m, H-3″), 1.27–1.29 (8H, m, H-4″∼H-7″), 0.92 (t, 6.1, H-8″); 13C NMR (CDCl3) δC 160.1 (s, C-1), 44.8 (t, C-3), 19.4 (t, C-4), 118.1 (s, C-4a), 125.6 (s, C-4b), 120.0 (d, C-5), 120.3 (d, C-6), 124.5 (d, C-7), 112.2 (d, C-8), 136.8 (s, C-8a), 128.3 (s, C-9a), 109.0 (d, C-1′), 126.5 (d, C-2′), 141.5 (s, C-3′), 120.7 (d, C-4′), 122.9 (s, C-4′a), 122.9 (s, C-4′b), 108.9 (d, C-5′), 125.8 (d, C-6′), 119.3 (d, C-7′), 120.4 (d, C-8′), 128.0 (s, C-8′a), 141.0 (s, C-9′a), 43.2 (t, C-1″), 29.0 (t, C-2″), 27.3 (t, C-3″), 29.4 (t, C-4″), 29.2 (t, C-5″), 31.8 (t, C-6″), 22.6 (t, C-7″), 14.2 (q, C-8″); EIMS m/z 447 (81, M+), 446 (100, M − H+), 432 (6), 419 (20), 404 (8), 362 (10), 346 (29), 319 (15), 174 (67), 160 (9), 69 (23), 57 (46); HREIMS m/z 447.2673 ([M]+, calcd. for C31H33N3, 447.2674).

3.3.7. 1-(9′-Decyl-3′-carbazolyl)-3,4-dihydro-β-carboline (14)

Yellow solid; mp 78–80 °C; UV λmax 233, 289, 323 nm; IR (KBr) νmax 3058, 2927, 1598, 1467, 1336, 808 cm−1; 1H NMR (CDCl3) δH 3.96 (2H, t, J = 7.9 Hz, H-3), 2.94 (2H, t, J = 8.3 Hz, H-4), 7.67 (1H, d, overlap, H-5), 7.20 (1H, m, H-6), 7.28 (1H, m, H-7), 7.38 (1H, d, overlap, H-8), 7.43 (1H, d, overlap,H-1′), 7.85 (1H, d, overlap, H-2′), 8.47 (1H, s, H-4′), 7.41 (1H, d, overlap, H-5′), 7.50 (1H, m, H-6′), 7.22 (1H, m, H-7′), 8.02 (1H, d, 7.7, H-8′), 4.23 (2H, t, J = 7.0 Hz, H-1″), 1.83 (2H, m, H-2″), 1.28–1.33 (14H, m, H-4″∼H-9″), 0.93 (t, 6.3, H-10″); 13C NMR (CDCl3) δC 159.9 (s, C-1), 48.1 (t, C-3), 19.2 (t, C-4), 117.9 (s, C-4a), 125.4 (s, C-4b), 119.8 (d, C-5), 120.1 (d, C-6), 124.3 (d, C-7), 112.1 (d, C-8), 136.7 (s, C-8a), 128.2 (s, C-9a), 108.8 (d, C-1′), 125.7 (d, C-2′), 141.3 (s, C-3′), 120.6 (d, C-4′), 122.7 (s, C-4′a), 122.7 (s, C-4′b), 108.7 (d, C-5′), 125.9 (d, C-6′), 119.1 (d, C-7′), 120.3 (d, C-8′), 127.8 (s, C-8′a), 140.7 (s, C-9′a), 43.0 (t, C-1″), 28.7 (t, C-2″), 27.1 (t, C-3″), 29.1–29.6 (t, C-4″∼C-7″), 31.7 (t, C-8″), 22.5 (t, C-9″), 14.0 (q, C-10″); EIMS m/z 475 (39, M+), 460 (4), 349 (27), 335 (25), 335 (63), 319 (10), 173 (76), 159 (22), 97 (19), 83 (23), 69 (38), 57 (79); HREIMS m/z 475.2987 ([M]+, calcd. for C33H37N3, 475.2988).

3.3.8. 1-(9′-Hexadecyl-3′-carbazolyl)-3,4-dihydro-β-carboline (16)

Yellow solid; mp 90–92 °C; UV λmax 238, 288 nm; IR (KBr) νmax 3060, 2925, 1597, 1468, 1338, 744 cm−1; 1H NMR (CDCl3) δH 3.93 (2H, t, J = 8.1 Hz, H-3), 2.93 (2H, t, J = 8.1 Hz, H-4), 7.64 (1H, d, overlap, H-5), 7.26 (1H, m, H-6), 7.32 (1H, m, H-7), 7.52 (1H, d, overlap, H-8), 7.38 (1H, d, overlap, H-1′), 7.86 (1H, d, overlap, H-2′), 8.49 (1H, s, H-4′), 7.41 (1H, d, overlap, H-5′), 7.45 (1H, m, H-6′), 7.23 (1H, m, H-7′), 8.04 (1H, d, J = 7.6 Hz, H-8′), 4.19 (2H, t, J = 7.2 Hz, H-1″), 1.80 (2H, m, H-2″), 1.12–1.46 (26H, m, H-3″∼15″), 0.90 (3H, t, J = 6.6 Hz, H-16″); 13C NMR (CDCl3) δC 160.1 (s, C-1), 47.5 (t, C-3), 19.4 (t, C-4), 112.9 (s, C-4a), 125.4 (s, C-4b), 120.0 (d, C-5), 120.4 (d, C-6), 125.4 (d, C-7), 112.4 (d, C-8), 137.3 (s, C-8a), 127.8 (s, C-9a), 108.9 (d, C-1′), 126.1 (d, C-2′), 141.7 (s, C-3′), 120.7 (d, C-4′), 122.7 (s, C-4′a), 122.9 (s, C-4′b), 109.0 (d, C-5′), 126.1 (d, C-6′), 119.4 (d, C-7′), 120.4 (d, C-8′), 124.9 (s, C-8′a), 140.7 (s, C-9′a), 43.1 (t, C-1″), 22.6–31.9 (C-2″∼15″), 14.1 (q, C-16″); EIMS m/z 559 (20, M+), 532 (3), 418 (2), 346 (20), 180 (6), 174 (30), 83 (5), 69 (11), 57 (41), 44 (100); HRESIMS m/z 560.4008 ([M + H]+, calcd. for C39H50N3, 560.4005).

3.3.9. 1-(9′-Eicosyl-3′-carbazolyl)-3,4-dihydro-β-carboline (18)

Yellow solid; mp 91–93 °C; UV λmax 222, 239, 260, 288 nm; IR (KBr) νmax 3062, 2921, 1465, 1338, 744 cm−1; 1H NMR (CDCl3) δH 3.87 (2H, t, J = 8.0 Hz, H-3), 2.93 (2H, t, J = 8.10 Hz, H-4), 7.61 (1H, d, overlap, H-5), 7.16 (1H, m, H-6), 7.33 (1H, m, H-7), 7.53 (1H, d, overlap, H-8), 7.29 (1H, d, overlap,H-1′), 7.99 (1H, d, overlap, H-2′), 8.70 (1H, s, H-4′), 7.38 (1H, d, overlap, H-5′), 7.44 (1H, m, H-6′), 7.18 (1H, m, H-7′), 8.08 (1H, d, J = 7.3 Hz, H-8′), 4.07 (2H, t, J = 7.3 Hz, H-1″), 1.75 (2H, m, H-2″), 1.25–1.37 (34H, m, H-3″∼19″), 0.88 (3H, t, J = 6.7 Hz, H-20″); 13C NMR (CDCl3) δC 160.6 (s, C-1), 44.8 (t, C-3), 19.2 (t, C-4), 113.2 (s, C-4a), 127.3 (s, C-4b), 120.3 (d, C-5), 120.9 (d, C-6), 126.4 (d, C-7), 113.2 (d, C-8), 139.2 (s, C-8a), 127.3 (s, C-9a), 109.1 (d, C-1′), 127.3 (d, C-2′), 142.4 (s, C-3′), 122.1 (d, C-4′), 122.6 (s, C-4′a), 123.0 (s, C-4′b), 109.1 (d, C-5′), 126.4 (d, C-6′), 119.9 (d, C-7′), 121.1 (d, C-8′), 126.4 (s, C-8′a), 140.8 (s, C-9′a), 43.0 (t, C-1″), 22.6–31.9 (C-2″∼19″), 14.1 (q, C-20″); EIMS m/z 615 (17, M+), 588 (4), 364 (20), 246 (14), 174 (15), 121 (35), 91 (41), 71 (45), 69 (64), 57 (100); HRESIMS m/z 616.4627 ([M + H]+, calcd. for C43H48N3, 616.4631).

3.4. Synthesis of Compounds 1922

To a stirred solution of carbazole (2.0 g, 1.2 mmol) and K2CO3 (1.7 g) in acetone (30 mL), 1-bromopropane, 1-bromopentane, 1-bromooctane and 1-bromodecane were slowly added, respectively (each 5 mL). The reaction mixture was stirred at 50 °C for 24 hours. After filtration and evaporation of the solvent under vacuum, the residue was chromatographed on a silicagel column (60 g) and eluted with n-hexane to afford compounds 1922 with a yield which varied in a range of 23–25%.

3.4.1. N-Propylcarbazole (19)

White solid; UV λmax 244, 261, 293, 330, 344 nm; IR (CH2Cl2) νmax 3021, 1596, 1484, 1344, 721 cm−1; 1H NMR (CDCl3) δH 8.16 (1H, d, J = 7.7 Hz, H-2), 7.31 (1H, dd, J = 7.7, 6.8 Hz, H-3), 7.52 (1H, dd, J = 7.7, 6.8 Hz, H-4), 7.45 (1H, d, J = 7.9 Hz, H-5), 4.32 (2H, t, J = 7.1 Hz, H-1′), 1.97 (2H, m, H-2′), 1.03 (3H, t, J = 7.4 Hz, H-3′); 13C NMR (CDCl3) δC 140.3 (s, C-1), 120.1 (d, C-2), 118.8 (d, C-3), 126.5 (d, C-4), 108.9 (d, C-5), 122.6 (s, C-6), 44.1 (t, C-1′), 22.3 (t, C-2′), 11.5 (q, C-3′); EIMS m/z 209 (85, M+), 180 (100), 166 (30), 152 (56), 140 (16), 127 (5), 90 (8), 84 (8), 77 (7), 49 (11).

3.4.2. N-Pentylcarbazole (20)

White solid; UV λmax 254, 261, 293, 330, 344 nm; IR (CH2Cl2) νmax 3064, 1596, 1484, 1346, 721 cm−1; 1H NMR (CDCl3) δH 8.14 (1H, d, J = 7.7 Hz, H-2), 7.26 (1H, dd, J = 7.7, 6.8 Hz, H-3), 7.50 (1H, dd, J = 7.7, 6.8 Hz, H-4), 7.43 (1H, d, J = 7.9 Hz, H-5), 4.33 (2H, t, J = 7.1 Hz, H-1′), 1.94 (2H, m, H-2′), 1.38–1.43 (4H, overlap, H-3′, 4′), 0.91 (3H, t, J = 6.8 Hz, H-5′; 13C NMR (CDCl3) δC 140.4 (s, C-1), 120.3 (d, C-2), 118.6 (d, C-3), 125.5 (d, C-4), 108.6 (d, C-5), 122.8 (s, C-6), 43.0 (t, C-1′), 28.6 (t, C-2′), 29.4 (t, C-3′), 22.4 (t, C-4′), 13.9 (q, C-5′); EIMS m/z 237 (27, M+), 180 (100), 166 (7), 152 (19), 140 (5), 127 (2), 84 (2), 77 (2), 49 (2).

3.4.3. N-Octylcarbazole (21)

White solid; UV λmax 264, 263, 330, 344 nm; IR (CH2Cl2) νmax 3040, 1596, 1484, 1346, 748, 721 cm−1; 1H NMR (CDCl3) δH 8.23 (1H, d, J = 7.7 Hz, H-2), 7.33 (1H, dd, J = 7.7, 6.8 Hz, H-3), 7.58 (1H, dd, J = 7.7, 6.8 Hz, H-4), 7.51 (1H, d, J = 7.9 Hz, H-5), 4.37 (2H, t, J = 7.2 Hz, H-1′), 1.97 (2H, m, H-2′), 1.37–1.44 (10H, overlap, H-3′∼7′), 1.01 (3H, t, J = 6.2 Hz, H-8′; 13C NMR (CDCl3) δC 140.6 (s, C-1), 120.5 (d, C-2), 118.8 (d, C-3), 125.7 (d, C-4), 108.8 (d, C-5), 123.0 (s, C-6), 43.2 (t, C-1′), 29.1 (t, C-2′), 27.4 (t, C-3′), 29.5 (t, C-4′), 29.3 (t, C-5′), 31.9 (t, C-6′), 22.8 (t, C-7′), 14.2 (q, C-8′); EIMS m/z 279 (84, M+), 180 (100), 166 (17), 152 (31), 140 (7), 81 (8), 77 (4), 69 (29), 55 (33).

3.4.4. N-Decylcarbazole (22)

White solid; UV λmax 263, 330, 344 nm; IR (CH2Cl2) νmax 2925, 2852, 1484, 1342, 748, 721 cm−1; 1H NMR (CDCl3) δH 7.96 (1H, d, J = 7.7 Hz, H-2), 7.09 (1H, dd, J = 7.7, 6.9 Hz, H-3), 7.34 (1H, dd, J = 7.7, 6.9 Hz, H-4), 7.24 (1H, d, J = 8.0 Hz, H-5), 4.12 (2H, t, J = 7.2 Hz, H-1′), 1.71 (2H, m, H-2′), 1.11–1.19 (14H, overlap, H-3′∼9′), 0.76 (3H, t, J = 6.4 Hz, H-10′; 13C NMR (CDCl3) δC 140.3 (s, C-1), 120.2 (d, C-2), 118.6 (d, C-3), 125.4 (d, C-4), 108.5 (d, C-5), 122.8 (s, C-6), 42.9 (t, C-1′), 28.8 (t, C-2′), 27.2 (t, C-3′), 29.2–29.5 (t, C-4′∼7′), 31.8 (t, C-8′), 22.6 (t, C-9′), 14.0 (q, C-10′); EIMS m/z 307 (84, M+), 194 (7), 180 (100), 166 (14), 152 (22), 140 (5), 81 (5), 69 (23), 55 (32).

3.5. Synthesis of Compounds 2326

To a stirred solution of carbazole (1.0 g, 0.6 mmol) and KOH (0.5 g) in EtOH (20 mL), 1-bromo-3-methylbuane, 2-bromopentane, 1-bromohexadecane, 1-bromoeicosane were slowly added, respectively (each 1 mL or 1 g). The reaction mixture was stirred at 50 °C for 24 h. After filtration and evaporation of the solvent under vacuum, the residue was chromatographed on a silica gel column (30 g) and eluted with n-hexane to afford compounds 2326 with a yield which varied in a range of 50–75%.

3.5.1. N-3′-Methylbutylcarbazole (23)

White solid; UV λmax 221, 294, 331, 345 nm; IR (CH2Cl2) νmax 3053, 2956, 1452, 748, 721 cm−1; 1H NMR (CDCl3) δH 8.14 (1H, d, J = 7.7 Hz, H-2), 7.26 (1H, dd, J = 7.7, 7.0 Hz, H-3), 7.49 (1H, dd, J = 7.9, 7.0 Hz, H-4), 7.43 (1H, d, J = 7.9 Hz, H-5), 4.34 (2H, t, J = 7.4 Hz, H-1′), 1.77 (4H, m, H-2′, 3′), 1.06 (4H, d, J = 6.2 Hz, H-4′, 5′); 13C NMR (CDCl3) δC 140.3 (s, C-1), 120.3 (d, C-2), 118.7 (d, C-3), 125.5 (d, C-4), 108.5 (d, C-5), 122.9 (s, C-6), 41.3 (t, C-1′), 26.1 (t, C-2′), 37.5 (d, C-3′), 22.6 (q, C-4′), 22.6 (q, C-5′); EIMS m/z 237 (94, M+), 180 (100), 166 (20), 152 (39), 140 (15), 127 (4), 69 (5), 41 (69).

3.5.2. N-1′-Methylpropylcarbazole (24)

White solid; UV λmax 221, 294, 330, 344 nm; IR (CH2Cl2) νmax 2962, 1454, 748, 721 cm−1; 1H NMR (CDCl3) δH 8.19 (1H, d, J = 7.7 Hz, H-2), 7.29 (1H, dd, J = 7.7, 6.9 Hz, H-3), 7.50 (1H, dd, J = 8.0, 6.9 Hz, H-4), 7.58 (1H, d, J = 8.0 Hz, H-5), 4.74 (2H, m, H-1′), 1.74 (2H, d, J = 7.0 Hz, H-2′), 2.07, (2H, m, H-3′), 0.86 (3H, t, J = 7.4 Hz, H-4′); 13C NMR (CDCl3) δC 139.9 (s, C-1), 120.2 (d, C-2), 118.5 (d, C-3), 125.3 (d, C-4), 110.0 (d, C-5), 123.2 (s, C-6), 52.8 (d, C-1′), 19.0 (q, C-2′), 27.9 (t, C-3′), 11.4 (q, C-4′); EIMS m/z 223 (36, M+), 194 (100), 180 (4), 166 (36), 152 (6), 140 (21), 115 (7), 57 (13), 49 (8).

3.5.3. N-Hexadecylcarbazole (25)

White solid; UV λmax 229, 236, 261, 293 nm; IR (CH2Cl2) νmax 3049, 2943, 1452, 746, 719 cm−1; 1H NMR (CDCl3) δH 8.11 (1H, d, J = 7.8 Hz, H-2), 7.24 (1H, dd, J = 7.8, 6.9 Hz, H-3), 7.47 (1H, dd, J = 8.0, 6.9 Hz, H-4), 7.42 (1H, d, J = 8.0 Hz, H-5), 4.31 (2H, t, J = 7.2 Hz, H-1′), 1.88 (2H, m, H-2′), 1.25–1.27 (26H, overlap, H-3′∼15′), 0.89 (3H, t, J = 6.3 Hz, H-16′); 13C NMR (CDCl3) δC 140.2 (s, C-1), 120.3 (d, C-2), 118.6 (d, C-3), 125.6 (d, C-4), 108.7 (d, C-5), 122.8 (s, C-6), 43.2 (t, C-1′), 22.8–32.0 (t, C-2′∼15′), 14.2 (q, C-16′); EIMS m/z 391 (82, M+), 194 (7), 180 (100), 167 (9), 152 (10), 97 (11), 83 (18), 69 (30), 57 (51), 43 (68).

3.5.4. N-Eicosylcarbazole (26)

White solid; UV λmax 333, 346 nm; IR (CH2Cl2) νmax 3050, 2956, 1450, 746, 719 cm−1; 1H NMR (CDCl3) δH 8.25 (1H, d, J = 7.7 Hz, H-2), 7.37 (1H, t, J = 7.4 Hz, H-3), 7.60 (1H, t, J = 7.4 Hz, H-4), 7.52 (1H, d, J = 8.5 Hz, H-5), 4.38 (2H, t, J = 7.2 Hz, H-1′), 1.98 (2H, m, H-2′), 1.40–1.44 (34H, overlap, H-3′∼19′), 1.07 (3H, t, J = 6.4 Hz, H-16′); 13C NMR (CDCl3) δC 140.4 (s, C-1), 120.3 (d, C-2), 118.7 (d, C-3), 125.5 (d, C-4), 108.6 (d, C-5), 122.9 (s, C-6), 43.0 (t, C-1′), 22.7–32.0 (t, C-2′∼19′), 14.1 (q, C-20′); EIMS m/z 447 (69, M+), 194 (5), 180 (100), 167 (8), 152 (7), 97 (3), 83 (5), 69 (9), 57 (33), 43 (60).

3.6. Synthesis of Compounds 2730

To a stirred solution of N-propylcarbazole (19, 620 mg, 2.9 mmol), N-pentylcarbazole (20, 680 mg, 2.8 mmol), N-octylcarbazole (21, 1 g, 3.5 mmol), or N-decylcarbazole (22, 1 g, 3.2 mmol), with hexamethylenetetramine (420–500 mg, 3–3.5 mmol) in tetrahydrofuran (3–10 mL), trifluroacetic acid (1–3.5 mL) was slowly added. The reaction mixtures were refluxed at 80 °C for 3.5 h. After cooling, the solution was partitioned between H2O and CHCl3. The CHCl3-soluble layer was evaporated under vacuum, the residue was chromatographed on a silica gel column (10 g) and eluted with N-hexane/CHCl3 mixture with increasing polarity to yield compounds 2730 with a yield 30–34%.

3.6.1. N-Propyl-3-carbazolyl Carboxyaldehyde (27)

White solid; UV λmax 234, 271, 329 nm; IR (CH2Cl2) νmax 2964, 2875, 1685, 1592, 1469, 1349, 1338, 806 cm−1; 1H NMR (CDCl3) δH 7.45 (1H, d, J = 8.4 Hz, H-1), 7.98 (1H, d, J = 8.5 Hz, H-2), 8.60 (1H, s, H-4), 7.47 (1H, d, J = 6.2 Hz, H-5), 7.53 (1H, dd, J = 7.1, 6.2 Hz, H-6), 7.33 (1H, dd, J = 7.7, 7.1 Hz, H-7), 8.15 (1H, d, J = 7.7 Hz, H-8), 10.09 (s, CHO), 4.29 (2H, t, J = 7.1 Hz, H-1′), 1.93 (2H, m, H-2′), 0.98 (3H, t, J = 7.4 Hz, H-3′); 13C NMR (CDCl3) δC 109.0 (d, C-1), 127.2 (d, C-2), 128.5 (s, C-3), 124.0 (d, C-4), 123.1 (s, C-4a), 123.0 (s, C-4b), 109.5 (d, C-5), 126.7 (d, C-6), 120.3 (d, C-7), 120.7 (d, C-8), 141.3 (s, C-8a), 144.2 (s, C-9a), 191.8 (d, CHO), 44.9 (t, C-1′), 22.3 (t, C-2′), 11.8 (q, C-3′); EIMS m/z 237 (84, M+), 208 (100), 180 (24), 166 (12), 152 (21), 139 (7), 84 (7), 77 (3), 69 (5), 49 (15).

3.6.2. N-Pentyl-3-carbazolyl Carboxyaldehyde (28)

White solid; UV λmax 234, 271, 329 nm; IR (CH2Cl2) νmax 2954, 2929, 1685, 1592, 1467, 1349, 1326, 748 cm−1; 1H NMR (CDCl3) δH 7.47 (1H, d, J = 8.4 Hz, H-1), 8.01 (1H, d, J = 8.5 Hz, H-2), 8.61 (1H, s, H-4), 7.50 (1H, d, J = 6.0 Hz, H-5), 7.55 (1H, dd, J = 7.0, 6.0 Hz, H-6), 7.33 (1H, dd, J = 7.6, 7.0 Hz, H-7), 8.16 (1H, d, J = 7.6 Hz, H-8), 10.10 (s, CHO), 4.34 (2H, t, J = 7.3 Hz, H-1′), 1.89 (2H, m, H-2′), 1.26–1.37 (2H, m, H-3′), 1.26–1.37 (2H, m, H-4′), 0.98 (3H, t, J = 6.3 Hz, H-5′); 13C NMR (CDCl3) δC 109.3 (d, C-1), 127.1 (d, C-2), 128.4 (s, C-3), 123.9 (d, C-4), 122.9 (s, C-4a), 123.0 (s, C-4b), 108.9 (d, C-5), 126.6 (d, C-6), 120.2 (d, C-7), 120.7 (d, C-8), 141.1 (s, C-8a), 144.0 (s, C-9a), 191.7 (d, CHO), 43.3 (t, C-1′), 28.5 (t, C-2′), 29.3 (t, C-3′), 22.3 (t, C-4′), 13.8 (q, C-5′); EIMS m/z 266 (99, M + 1+), 265 (76, M+), 208 (34), 180 (14), 167 (12), 154 (100), 136 (73), 107 (25), 89 (21), 77 (25), 55 (26).

3.6.3. N-Octyl-3-carbazolyl Carboxyaldehyde (29)

White solid; UV λmax 234, 276, 290, 331 nm; IR (CH2Cl2) νmax 2950, 2927, 2875, 1687, 1592, 1467, 1351, 1338 cm−1; 1H NMR (CDCl3) δH 7.46 (1H, d, J = 8.4 Hz, H-1), 8.02 (1H, d, J = 8.5 Hz, H-2), 8.62 (1H, s, H-4), 7.48 (1H, d, J = 7.7 Hz, H-5), 7.54 (1H, dd, J = 7.1, 7.7 Hz, H-6), 7.33 (1H, dd, J = 7.6, 7.1 Hz, H-7), 8.16 (1H, d, J = 7.6 Hz, H-8), 10.10 (s, CHO), 4.34 (2H, t, J = 7.1 Hz, H-1′), 1.88 (2H, m, H-2′), 1.25–1.36 (2H, m, H-3′), 1.25–1.30 (2H, m, H-4′), 1.26–1.36 (2H, m, H-5′), 1.25–1.36 (2H, m, H-6′), 1.25–1.36 (2H, m, H-7′), 0.86 (3H, t, J = 7.4 Hz, H-8′); 13C NMR (CDCl3) δC 109.0 (d, C-1), 127.2 (d, C-2), 128.5 (s, C-3), 124.0 (d, C-4), 123.1 (s, C-4a), 123.0 (s, C-4b), 109.4 (d, C-5), 126.7 (d, C-6), 120.3 (d, C-7), 120.8 (d, C-8), 141.2 (s, C-8a), 144.1 (s, C-9a), 191.8 (d, CHO), 43.5 (t, C-1′), 29.1 (t, C-2′), 27.3 (t, C-3′), 29.7 (t, C-4′), 29.3 (t, C-5′), 31.8 (t, C-6′), 22.6 (t, C-7′), 14.1 (q, C-8′); EIMS m/z 307 (71, M+), 242 (12), 224 (56), 208 (100), 180 (34), 166 (13), 152 (25), 139 (6), 77 (4), 69 (26), 55 (43).

3.6.4. N-Decyl-3-carbazolyl Carboxyaldehyde (30)

White solid; UV λmax 234, 274, 290, 331 nm; IR (CH2Cl2) νmax 2952, 2925, 1687, 1592, 1467, 1351, 1338, 806 cm−1; 1H NMR (CDCl3) δH 7.47 (1H, d, J = 8.4 Hz, H-1), 8.02 (1H, d, J = 8.4 Hz, H-2), 8.62 (1H, s, H-4), 7.49 (1H, d, J = 7.5 Hz, H-5), 7.54 (1H, dd, J = 6.9, 7.5 Hz, H-6), 7.35 (1H, dd, J = 7.6, 6.9 Hz, H-7), 8.17 (1H, d, J = 7.6 Hz, H-8), 10.11 (s, CHO), 4.31 (2H, t, J = 7.2 Hz, H-1′), 1.91 (2H, m, H-2′), 1.26–1.37 (2H, m, H-3′), 1.26–1.37 (2H, m, H-4′), 1.26–1.37 (2H, m, H-5′), 1.26–1.37 (2H, m, H-6′), 1.26–1.37 (2H, m, H-7′), 1.26–1.37 (2H, m, H-8′), 1.26–1.37 (2H, m, H-9′), 0.89 (3H, t, J = 6.2 Hz, H-10′); 13C NMR (CDCl3) δC 109.0 (d, C-1), 127.2 (d, C-2), 128.5 (s, C-3), 124.0 (d, C-4), 123.1 (s, C-4a), 123.0 (s, C-4b), 109.4 (d, C-5), 126.7 (d, C-6), 120.3 (d, C-7), 120.8 (d, C-8), 141.2 (s, C-8a), 144.1 (s, C-9a), 191.8 (d, CHO), 43.5 (t, C-1′), 28.9 (t, C-2′), 27.3 (t, C-3′), 29.3–29.5 (t, C-4′), 29.3–29.5 (t, C-5′), 29.3–29.5 (t, C-6′), 29.3–29.5 (t, C-7′), 31.9 (t, C-8′). 22.7 (t, C-9′), 14.1 (q, C-10′); EIMS m/z 335 (67, M+), 224 (7), 208 (100), 194 (5), 180 (23), 166 (7), 77 (1), 69 (9), 55 (3), 43 (43).

3.7. Synthesis of Compounds 3134

Trifluroacetic acid (2.5–5 mL) was slowly added to a stirred solution of N-3′-methylbutylcarbazole (23, 950 mg, 5.1 mmol), N-1′-methylpropylcarbazole (24, 950 mg, 4.3 mmol), N-hexadecylcarbazole (25, 1.2 g, 3.0 mmol), or N-decylcarbazole (26, 950 mg, 2.1 mmol), with hexamethylenetetramine (810 mg–1 g, 5.1–6.3 mmol) in tetrahydrofuran (2.5–5 mL). The reaction mixtures were refluxed at 80 °C for 3.5 hours. After cooling, the solution was partitioned between H2O and CHCl3. The CHCl3-soluble layer was evaporated under vacuum, the residue was chromatographed on a silica gel column (10 g) and eluted with N-hexane/CHCl3 mixture with increasing polarity to yield compounds 3134 with a yield of 21–25%.

3.7.1. N-3′-Methylbutyl-3-carbazolyl Carboxyaldehyde (31)

White solid; UV λmax 232, 271, 294, 331 nm; IR (CH2Cl2) νmax 2956, 1685, 1593, 748 cm−1; 1H NMR (CDCl3) δH 7.45 (1H, d, J = 8.3 Hz, H-1), 8.01 (1H, d, J = 8.5 Hz, H-2), 8.60 (1H, s, H-4), 7.45 (1H, d, J = 8.0 Hz, H-5), 7.54 (1H, dd, J = 8.0Hz, H-6), 7.33 (1H, dd, J = 7.2, 7.7 Hz, H-7), 8.15 (1H, d, J = 7.7 Hz, H-8), 10.10 (s, CHO), 4.33 (2H, t, J = 6.6 Hz, H-1′), 1.76 (2H, m, H-2′), 1.76 (3H, m, H-3′), 1.04 (2H, d, J = 5.0 Hz, H-4′), 1.04 (2H, d, J = 5.0 Hz, H-3′); 13C NMR (CDCl3) δC 108.8 (d, C-1), 127.1 (d, C-2), 128.5 (s, C-3), 123.9 (d, C-4), 123.0 (s, C-4a), 123.1 (s, C-4b), 109.2 (d, C-5), 126.7 (d, C-6), 120.3 (d, C-7), 120.7 (d, C-8), 141.0 (s, C-8a), 143.9 (s, C-9a), 191.7 (d, CHO), 41.7 (t, C-1′), 26.1 (t, C-2′), 37.4 (d, C-3′), 22.5 (q, C-4′), 22.5 (q, C-5′); EIMS m/z 265 (81, M+), 208 (100), 195 (9), 180 (34), 166 (16), 152 (33), 139 (9), 84 (8), 69 (10), 41 (33).

3.7.2. N-1′-Methylpropyl-3-carbazolyl Carboxyaldehyde (32)

White solid; UV λmax 232, 276, 294, 332 nm; IR (CH2Cl2) νmax 2962, 1685, 1684, 1591, 1489, 1338, 748 cm−1; 1H NMR (CDCl3) δH 7.58 (1H, d, J = 8.7 Hz, H-1), 8.00 (1H, d, J = 8.7 Hz, H-2), 8.64 (1H, s, H-4), 7.58 (1H, d, J = 8.2 Hz, H-5), 7.51 (1H, dd, J = 8.2, 7.1 Hz, H-6), 7.32 (1H, dd, J = 7.61, 7.7 Hz, H-7), 8.18 (1H, d, J = 7.7 Hz, H-8), 10.10 (s, CHO), 4.73 (2H, m, H-1′), 1.71 (2H, d, J = 7.0 Hz, H-2′), 2.50–2.30 (2H, m, H-3′), 0.80 (2H, m, H-4′); 13C NMR (CDCl3) δC 110.1 (d, C-1), 126.8 (d, C-2), 128.2 (s, C-3), 123.7 (d, C-4), 123.4 (s, C-4a), 123.4 (s, C-4b), 110.8 (d, C-5), 126.4 (d, C-6), 120.0 (d, C-7), 120.6 (d, C-8), 141.0 (s, C-8a), 144.0 (s, C-9a), 191.6 (d, CHO), 53.5 (d, C-1′), 19.1 (q, C-2′), 28.0 (t, C-3′), 11.4 (q, C-4′); EIMS m/z 251 (80, M+), 222 (100), 194 (78), 180 (5), 166 (54), 152 (10), 139 (31), 113 (6), 69 (11), 57 (29).

3.7.3. N-Hexadecyl-3-carbazolyl Carboxyaldehyde (33)

White solid; UV λmax 234, 294, 332 nm; IR (CH2Cl2) νmax 2920, 1689, 1593, 1462, 1352, 806 cm−1; 1H NMR (CDCl3) δH 7.48 (1H, d, J = 8.5 Hz, H-1), 8.01 (1H, d, J = 8.5 Hz, H-2), 8.62 (1H, s, H-4), 7.46 (1H, d, J = 8.1 Hz, H-5), 7.54 (1H, dd, J = 8.1, 7.1 Hz, H-6), 7.33 (1H, dd, J = 7.1, 7.7 Hz, H-7), 8.17 (1H, d, J = 7.7 Hz, H-8), 10.10 (s, CHO), 4.34 (2H, t, J = 7.2 Hz, H-1′), 1.90 (2H, m, H-2′), 1.24–1.26 (2H, m, H-3′), 1.24–1.26 (2H, m, H-4′), 1.24–1.26 (2H, m, H-5′), 1.24–1.26 (2H, m, H-6′∼15′), 0.89 (2H, m, H-16′); 13C NMR (CDCl3) δC 109.0 (d, C-1), 127.2 (d, C-2), 128.6 (s, C-3), 126.8 (d, C-4), 123.1 (s, C-4a), 123.1 (s, C-4b), 109.5 (d, C-5), 126.8 (d, C-6), 120.3 (d, C-7), 120.8 (d, C-8), 141.2 (s, C-8a), 144.1 (s, C-9a), 191.8 (d, CHO), 43.5 (t, C-1′), 22.8–32.0 (m, C-2′), 22.8–32.0 (m, C-3′), 22.8–32.0 (m, C-4′), 22.8–32.0 (m, C-5′), 22.8–32.0 (m, C-6′∼15′), 14.2 (q, C-16′); EIMS m/z 419 (53, M+), 208 (100), 194 (4), 180 (20), 166 (6), 152 (6), 83 (2), 69 (7), 57 (31), 43 (91).

3.7.4. N-Eicosyl-3-carbazolyl Carboxyaldehyde (34)

White solid; UV λmax 235, 275, 290 nm; IR (CH2Cl2) νmax 2918, 1687, 752 cm−1; 1H NMR (CDCl3) δH 7.46 (1H, d, J = 8.5 Hz, H-1), 8.01 (1H, d, J = 8.5 Hz, H-2), 8.60 (1H, s, H-4), 7.46 (1H, d, J = 8.1 Hz, H-5), 7.54 (1H, dd, J = 8.1, 7.1 Hz, H-6), 7.33 (1H, dd, J = 7.1, 7.7 Hz, H-7), 8.17 (1H, d, J = 7.7 Hz, H-8), 10.10 (s, CHO), 4.31 (2H, t, J = 7.2 Hz, H-1′), 1.89 (2H, m, H-2′), 1.25–1.27 (2H, m, H-3′), 1.25–1.27 (2H, m, H-4′), 1.25–1.27 (2H, m, H-5′), 1.25–1.27 (2H, m, H-6′∼15′), 1.25–1.27 (2H, m, H-16′), 1.25–1.27 (2H, m, H-17′∼19′), 0.90 (3H, t, J = 7.7 Hz, H-20′); 13C NMR (CDCl3) δC 108.9 (d, C-1), 127.1 (d, C-2), 128.8 (s, C-3), 123.9 (d, C-4), 123.0 (s, C-4a), 123.0 (s, C-4b), 109.3 (d, C-5), 126.6 (d, C-6), 120.2 (d, C-7), 120.7 (d, C-8), 140.6 (s, C-8a), 143.6 (s, C-9a), 191.6 (d, CHO), 43.4 (t, C-1′), 22.7–31.9 (t, C-2′), 22.7–31.9 (t, C-3′), 22.7–31.9 (t, C-4′), 22.7–31.9 (t, C-5′), 22.7–31.9 (t, C-6′∼15′), 22.7–31.9 (t, C-16′), 22.7–31.9 (t, C-17′∼19′), 14.1 (q, C-20′).; EIMS m/z 475 (71, M+), 208 (100), 194 (5), 180 (28), 166 (7), 152 (8), 83 (7), 69 (17), 57 (57), 43 (88)

3.8. Cytotoxicity Assay

The cytotoxic activities of compounds against KB (human mouth epidermoid carcinoma), DLD (human colon adenocarcinoma), NCI-H661 (human lung large cell carcinoma), Hepa (human hepatoma), and HepG2/A2 (human hepatoblastoma) cells were assayed by the MTT{3-(4,5-dimethylthiazole-2-yl)-2,5-diphenyltetrazolium bromide} colorimetric assay as previously described [23]. The cells for assay were cultured in RPMI-1640 medium supplemented with 5% CO2 in an incubator at 37 °C. The cytotoxicity assay depends on the binding of methylene blue to fixed monolayers of cells at pH 8.5, washing the monolayer, and releasing the dye by lowering the pH value. Samples and control standard drugs were prepared at a concentration of 1, 10, 40, and 100 μg/mL. After seeding 2880 cells/well in a 96-well microplate for 3 h, 20 μL of sample or standard agent was placed in each well and incubated at 37 °C for 3 days. After removing the medium from the microplates, the cells were fixed with 10% formaldehyde in 0.9% saline for 30 min, then dyed with 1% (w/v) methylene blue in 0.01 M borate-buffer (100 μL/well) for 30 min. The 96-well plate was dipped into a 0.01 M borate-buffer solution four times in order to remove the dye. Then, 100 μL/well of EtOH-0.1 M HCl (1:1) was added as a dye eluting solvent, and the absorbance was measured on a microtiter plate reader (Dynatech, MR 7000) at a wavelength of 650 nm. The ED50 value was defined by a comparison with the untreated cells as the concentration of test sample resulting in 50% reduction of absorbance. Doxorubicin was used as a standard compound.

3.9. Relaxation Assay of Topoisomerases I and II

Topoisomerases I and II (topo I and II) assays were measured by assessing relaxation of supercoiled pBR322 plasmid DNA according to [24]. Using camptothecin (CPT) as topo I and etoposide (VP-16) as topo II positive controls, test samples were dissolved in 5% (v/v) DMSO and then diluted to appropriate concentrations. In summary, topo I (TopoGen) was mixed with the test sample and 10× volume of assay buffer (100 mM Tris-HCl, 10 mM EDTA, 1.5 M NACl, 1.0% BSA, 1 M spermidine, and 50% glycerol), and then supercoiled DNA (pBR322) was added. In topo II assay, the mixture contained test sample and buffer including 50 mM Tris-HCl, 120 mM KCl, 10 mM MgCl2, 0.5 mM ATP, 0.5 mM dithiothreitol, 2 μg BSA, pBR322 plasmid DNA (0.25 μg), and 3U of topo II (TopoGen) in final volume of 20 μL. After incubation of topo I or topo II mixture for 30 min at 37 °C, 2 λ 10% SDS and 2.5 λ proteinase K were added for 1 h. The reaction mixtures were electrophoresed on a 2% agarose gel (50 V, 20 min; 100 V, 30 min; 110 V, 30 min) and stained with ethidium bromide. Finally, by a densitometer of ImageMaster® (Fujifilm thermal imaging system, FTI-500), the gels were directly scanned and the area representing supercoiled DNA was calculated. Concentrations for 50% inhibition (IC50) were determined by interpolation from plots of topoisomerases I or II activity versus inhibitor concentration. Etoposide was used as a standard.

3.10. Molecular Modeling

The three dimensional structures of DNA duplexes were obtained from the Brookhaven Protein Databank (PDB code 108d or 264d). The 3D structures of compound 2 were constructed as a protonated form and were assigned Gasteiger-Huckel partial charges using Sybyl 7.0 (Tripos Associates; St. Louis, MO, USA) as previously described [25]. The AutoTors module was used to specify two rotatable bonds in 2 for AutoDock. The initial complex structure was then optimized by energy minimization with the Tripos force field, employing the Powell method with an energy-gradient-convergence criterion of 0.05 kcal/(mol Ǻ) and a distance-dependent dielectric constant of 4 r. To carry out AutoDock simulations, a grid box was defined to enclose the interaction cavity with dimensions of 18.8 × 18.8 × 18.8 Ǻ. The grid maps for energy scoring were calculated with Autogrid using a grid-point spacing of 0.375 Ǻ, and 100 × 76 × 76 grid points for 108 d, or 70 × 70 × 110 grid points for 264 d. 200 independent docking runs were carried out using the Lamarckian genetic algorithm (LGA) with a maximum number of 5,000,000 energy evaluations and a population of 50 randomly initiated individuals. The lowest-energy docked conformations proposed the energetically favorable binding modes of compound 2 to DNA.

4. Conclusions

In summary, a series of 1-substituted carbazolyl-1,2,3,4-tetrahydro- and carbazolyl-3,4-dihydro-β-carboline analogs have been synthesized and evaluated as potential antitumor agents. Among them, compound 7 and 6 showed the most potent and selective activity against NCI-H661 and KB tumor cells, respectively. Compound 9 possessed most potent activity against DLD tumor cells. Compound 2 exhibited most promising activity against KB, NCI-H661 and Hepa (or HepG2/A2) tumor cell lines. Inhibition of human DNA topoisomerase II revealed that compounds 3 and 4 are quite promising for further development of enzyme inhibitors. The SAR revealed that there was a lack of complete correlation between carbon numbers of the side chain and biological activities. However, the negative correlation was present between the alkyl side chain length in cytotoxicity tests. The optimal chain length is between 2 and 5 carbons. The optimal chain length for anti-topoisomerase II may be 3 and 5 carbons. The DNA binding capacity, which favored minor groove binding, was thus influenced by the alkyl substitution on carbazole chromophore and proton donor on β-carboline chromophore. On the basis of the SAR study, synthesis of analogs of lead compounds 2, 3, 6, 7 and 9, in order to search for more potent activity, is currently in progress.

Acknowledgments

We thank Ho Chao Lein and Yu Shiu Ching of the NSC Southern NMR and MS Instrument Center for measurement of NMR an MS spectral data. This research was supported by grants from the National Science Council, Republic of China (NSC 98-2113-M-002-002-MY3).
  • Samples Availability: Available from the corresponding author.

References

  1. Blackman, AJ; Matthews, DJ; Narkowicz, CK. β-Carboline alkaloids from the marine bryozoan Costaticella hastata. J Nat Prod 1987, 50, 494–496. [Google Scholar]
  2. Kearns, PS; Coll, JC; Rideout, JA. A beta-carboline dimer from an ascidian, Didemnum sp. J Nat Prod 1995, 58, 1075–1076. [Google Scholar]
  3. Kobayashi, J; Tsuda, M; Kawasaki, N; Sasaki, T; Mikami, Y. 6-Hydroxymanzamine A and 3,4-Dihydromanzamine A, New Alkaloids from the Okinawan Marine Sponge Amphimedon sp. J Nat Prod 1994, 57, 1737–1740. [Google Scholar]
  4. Mc Nulty, J; Still, IWJ. Selective methylation and Stereoselective Reduction of a β-Carboline to a N(2)-Methyl-1,2,3,4-tetrahydro-β-carboline. Tetrahedron Lett 1995, 36, 7965–7966. [Google Scholar]
  5. Still, IWJ; Mc Nulty, J. The Synthesis of Eudistomins S and T: β-carbolines from the Tunicate Eudistoma olivaceum. Heterocycles 1989, 29, 2057–2059. [Google Scholar]
  6. Nowak, W; Gerlach, H. Synthesis of Manzamine C, Infractine and 6-Hydroxyinfractine. Liebigs Ann Chem 1993, 1993, 153–159. [Google Scholar]
  7. Badre, A; Boulanger, A; Mansom, AE; Banaigs, B; Combaut, G; Francisco, C. Eudistomin U and Isoeudistomin U, New Alkaloids from the Carribean Ascidian Lissoclinum fragile. J Nat Prod 1994, 57, 528–533. [Google Scholar]
  8. Rinehart, KL, Jr; Kobajashi, J; Harbour, GC; Gilmore, J; Mascal, M; Holt, TG; Shield, LS; Lafargue, F. Eudistomins A–Q, β-carbolines from the antiviral Caribbean tunicate Eudistoma olivaceum. J Am Chem Soc 1987, 109, 3378–3387. [Google Scholar]
  9. Crews, P; Cheng, XC; Adamczeski, M; Rodriguez, J; Jaspars, M; Schmitz, FJ; Traeger, SC; Pordesimo, EO. 1,2,3,4-tetrahydro-8-hydroxymanzamines, alkaloids from two different haplosclerid sponges. Tetrahedron 1994, 50, 13567–13574. [Google Scholar]
  10. Sakai, R; Kohmoto, S; Higa, T; Jefford, CW; Bernardinelli, G. Manzamine B and C, two novel alkaloids from the sponge haliclona sp. Tetrahedron Lett 1987, 28, 5493–5496. [Google Scholar]
  11. Munro, MHG; Luibrand, RT; Blunt, JW. The search for antiviral and anticancer compounds from marine organisms. In Bioorganic Marine Chemistry; Scheuer, PJ, Ed.; Springer-Verlag: New York, NY, USA, 1987; Volume 1; pp. 103–105. [Google Scholar]
  12. Ichiba, T; Sakai, R; Kohmoto, S; Saucy, G; Higa, T. New manzamine alkaloids from a sponge of the genus xestospongia. Tetrahedron Lett 1988, 29, 3083–3086. [Google Scholar]
  13. Higa, T. Structure elucidation. In Studies in Natural Product Chemistry; Atta-Ur-Rahman, Ed.; Elsevier: New York, NY, USA, 1989; Volume 5; pp. 346–353. [Google Scholar]
  14. Yousaf, M; Hammond, NL; Peng, J; Wahyuono, S; McIntosh, KA; Charman, WN; Mayer, AMS; Hamann, MT. New Manzamine Alkaloids from an Indo-Pacific Sponge. Pharmacokinetics, Oral Availability, and the Significant Activity of Several Manzamines against HIV-I, AIDS Opportunistic Infections, and Inflammatory Diseases. J Med Chem 2004, 47, 3512–3517. [Google Scholar]
  15. Sakai, R; Higa, T; Jefford, CW; Bernardinelli, G. Manzamine A, a novel antitumor alkaloid from a sponge. J Am Chem Soc 1986, 108, 6404–6405. [Google Scholar]
  16. Shen, YC; Tai, HR; Duh, CY. Bioactive constituents from Heliclona sp., a Formosan marine sponge. Chin Pharm J 1996, 48, 1–10. [Google Scholar]
  17. Yeh, SF; Shen, YC. 1-Substituted 1,2,3,4-Tetrahydro-β-Carboline and 3,4-Dihydro-β-Carboline and Analogs as Antitumor Agents. US Patent 6,720,331 2004. [Google Scholar]
  18. Duff, JC. A new method for the preparation of p-dialkylaminobenzaldehydes. J Chem Soc 1945, 276–277. [Google Scholar] [CrossRef]
  19. Valentine, D, Jr; Scot, JW. Asymmetric synthesis. Synthesis 1978, 1978, 329–356. [Google Scholar]
  20. Kawashima, Y; Horiguchi, A; Taguchi, M; Tuyuki, Y; Karasawa, Y; Araki, H; Hatayama, K. Synthesis and Pharmacological Evaluation of 1,2,3,4-Tetrahydro-β-carboline Derivatives. Chem Pharm Bull 1995, 43, 783–787. [Google Scholar]
  21. Kondo, K; Shigemori, H; Kikuchi, Y; Ishibashi, M; Sasaki, T; Kobajashi, J. Ircinals A and B from the Okinawan marine sponge Ircinia sp.: plausible biogenetic precursors of manzamine alkaloids. J Org Chem 1992, 57, 2480–2483. [Google Scholar]
  22. Pezzuto, JM; Che, CT; Mc Pherson, DD; Zhu, JP; Topcu, G; Erdelmeir, CAJ; Cordell, GA. DNA as an Affinity Probe Useful in the Detection and Isolation of Biologically Active Natural Products. J Nat Prod 1991, 54, 1522–1530. [Google Scholar]
  23. Shen, Y-C; Wang, S-S; Pan, Y-L; Lo, K-L; Chakraborty, R; Chien, CT; Kuo, YH; Lin, Y-C. New Taxane Diterpenoids from the Leaves and Twigs of Taxus sumatrana. J Nat Prod 2002, 65, 1848–1852. [Google Scholar]
  24. Yang Kuo, LM; Chen, KY; Hwang, SY; Chen, JL; Liu, YY; Liaw, CC; Ye, PH; Chou, CJ; Shen, CC; Kuo, YH. DNA Topoisomerase I Inhibitor, Ergosterol Peroxide from Penicillium oxalicum. Planta Med 2005, 71, 77–79. [Google Scholar]
  25. Tu, L-C; Chen, C-S; Hsiao, I-C; Chern, J-W; Lin, C-H; Shen, Y-C; Yeh, S-F. The β-Carboline Analog Mana-Hox Causes Mitotic Aberration by Interacting with DNA. Chem Biol 2005, 12, 1317–1324. [Google Scholar]
Figure 1. The main part of manzamine A.
Figure 1. The main part of manzamine A.
Marinedrugs 09 00256f1
Figure 2. Plot of the d(CGCATGGG)2-compound 2 complex. (a) Line plot: Three-dimensional structural model of 2 with DNA receptor binding site; (b) Space-filling plot: Compound 2, red; DNA duplex, blue.
Figure 2. Plot of the d(CGCATGGG)2-compound 2 complex. (a) Line plot: Three-dimensional structural model of 2 with DNA receptor binding site; (b) Space-filling plot: Compound 2, red; DNA duplex, blue.
Marinedrugs 09 00256f2
Figure 3. Plot of the d(CGCGAATTGCG)2-compound 2 complex. (a) Line plot: Three-dimensional structural model of 2 with DNA receptor binding site; (b) Space-filling plot: Compound 2, red; DNA duplex, blue.
Figure 3. Plot of the d(CGCGAATTGCG)2-compound 2 complex. (a) Line plot: Three-dimensional structural model of 2 with DNA receptor binding site; (b) Space-filling plot: Compound 2, red; DNA duplex, blue.
Marinedrugs 09 00256f3
Scheme 1. Preparation of compounds 136.
Scheme 1. Preparation of compounds 136.
Marinedrugs 09 00256f4
Table 1. Cytotoxicity of Compounds 118 against Human Cancer Cells (IC50, μg/mL) a.
Table 1. Cytotoxicity of Compounds 118 against Human Cancer Cells (IC50, μg/mL) a.
Compound/Cell lineKBDLDNCI-H661HepaHepG2/A2
13.471.691.82NT bNT
20.711.090.84NT0.60
31.841.582.14NT6.10
42.722.165.02NT5.10
53.00NTNT2.25NT
60.48NTNT2.67NT
73.453.750.22NT20.50
8>201.293.39NT5.40
92.96NTNT1.12NT
103.13NTNT4.40NT
11>201.57>20NT20.70
129.132.555.28NT30.10
136.3610.4013.90NT>60
14>20>20>20>20>60
15>20>20>20>20>60
16>20>20>20>20>60
17>20>20>20>20>60
18>20>20>20>20>60
aThe concentration of compound which inhibited 50% (IC50) of the growth of tumor cell lines (KB, human mouth epidermoid carcinoma; DLD, human colon adenocarcinoma; NCI-H661, human lung large cell carcinoma; Hepa and HepG2/A2, human hepatoma); All data estimated by interpolation method; Doxorubicin was used as a positive control (IC50, 0.1 μg/mL);
bNot tested.
Table 2. Inhibition of DNA topoisomerases I and II (IC50, μg/mL) a by compounds 118.
Table 2. Inhibition of DNA topoisomerases I and II (IC50, μg/mL) a by compounds 118.
CompoundDNA topoisomerase IDNA topoisomerase II
1>1009.5
2>1008.5
347.01.6
4>1002.1
5>10010.0
6>10026.0
789.02.7
858.022.0
996.02.8
1067.02.7
11>10030.0
12>10029.0
13>10028.0
14>10019.0
15>100>100
16>100>100
17>100>100
18>1005.6
Camptothecin17.0NT
EtoposideNT31.0
aEach compound was examined with five concentrations at 5, 10, 25, 50 and 100 μg/mL; The IC50 value was measured based on the degree of inhibition at these five concentrations; NT: Not tested.

Share and Cite

MDPI and ACS Style

Shen, Y.-C.; Chang, Y.-T.; Lin, C.-L.; Liaw, C.-C.; Kuo, Y.H.; Tu, L.-C.; Yeh, S.F.; Chern, J.-W. Synthesis of 1-Substituted Carbazolyl-1,2,3,4-tetrahydro- and Carbazolyl-3,4-dihydro-β-carboline Analogs as Potential Antitumor Agents. Mar. Drugs 2011, 9, 256-277. https://doi.org/10.3390/md9020256

AMA Style

Shen Y-C, Chang Y-T, Lin C-L, Liaw C-C, Kuo YH, Tu L-C, Yeh SF, Chern J-W. Synthesis of 1-Substituted Carbazolyl-1,2,3,4-tetrahydro- and Carbazolyl-3,4-dihydro-β-carboline Analogs as Potential Antitumor Agents. Marine Drugs. 2011; 9(2):256-277. https://doi.org/10.3390/md9020256

Chicago/Turabian Style

Shen, Ya-Ching, Yao-To Chang, Chun-Ling Lin, Chia-Ching Liaw, Yao Haur Kuo, Lan-Chun Tu, Sheau Farn Yeh, and Ji-Wang Chern. 2011. "Synthesis of 1-Substituted Carbazolyl-1,2,3,4-tetrahydro- and Carbazolyl-3,4-dihydro-β-carboline Analogs as Potential Antitumor Agents" Marine Drugs 9, no. 2: 256-277. https://doi.org/10.3390/md9020256

Article Metrics

Back to TopTop